1
|
Byer-Alcorace A, Thomas C, Taub ME, Piekos S. Improved clearance predictions for aldehyde oxidase substrates using a novel triculture human hepatocyte model. Drug Metab Dispos 2025; 53:100051. [PMID: 40147225 DOI: 10.1016/j.dmd.2025.100051] [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: 11/23/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/29/2025] Open
Abstract
Over the last several decades, efforts in medicinal chemistry have aimed to reduce the extent of CYP metabolism of new chemical entities. This approach, however, has led to increased susceptibility to metabolism by non-CYP-mediated pathways, particularly involving other phase I enzymes such as aldehyde oxidase (AO). Commonly used in vitro models, such as suspended or cocultured primary human hepatocytes, have limitations in evaluating the disposition of compounds metabolized by AO due to low or variable levels of enzyme activity. Thus, an in vitro model that exhibits high to moderate levels of AO activity that can better predict the contribution of AO to drug metabolism and its impact on drug clearance is needed. A novel, 2D+ primary human hepatocyte model, TruVivo, was evaluated for its potential utility to improve hepatic clearance (CLh) predictions and determine the contribution of AO to drug metabolism in humans. TruVivo demonstrated stable levels of AO activity for at least 2 weeks that were higher than levels in other hepatocyte models. CLh predictions generated using TruVivo for the reference compounds carbazeran, zoniporide, zaleplon, and O6-benzylguanine were within 2-fold of reported in vivo CL values. Furthermore, the estimated fraction metabolized by AO for zaleplon and zoniporide was within 25% of reported in vivo values, whereas that for carbazeran and O6-benzylguanine was similar to those generated in other systems. These findings suggest TruVivo may offer a novel means to assess CLh of AO substrates more accurately, even over extended incubation times for low clearance compounds. SIGNIFICANCE STATEMENT: The TruVivo in vitro primary human hepatocyte model maintains high levels of aldehyde oxidase (AO) activity for at least 14 days in culture, making the system suitable for evaluating slowly metabolized compounds, particularly those metabolized by AO. This novel system may therefore be useful for improving human clearance predictions for AO substrates.
Collapse
Affiliation(s)
- Alexander Byer-Alcorace
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Cody Thomas
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Stephanie Piekos
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut.
| |
Collapse
|
2
|
Lo Iacono M, Corrao S, Alberti G, Amico G, Timoneri F, Russo E, Cucina A, Indelicato S, Rappa F, Corsello T, Saieva S, Di Stefano A, Di Gaudio F, Conaldi PG, La Rocca G. Characterization and Proteomic Profiling of Hepatocyte-like Cells Derived from Human Wharton's Jelly Mesenchymal Stromal Cells: De Novo Expression of Liver-Specific Enzymes. BIOLOGY 2025; 14:124. [PMID: 40001892 PMCID: PMC11851833 DOI: 10.3390/biology14020124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025]
Abstract
End-stage liver disease (ESLD), affecting millions worldwide, represents a challenging issue for clinical research and global public health. Liver transplantation is the gold standard therapeutic approach but shows some drawbacks. Hepatocyte transplantation could be a reliable alternative for patient treatment. Mesenchymal stromal cells derived from Wharton's jelly of the umbilical cord (WJ-MSCs) can differentiate into hepatocyte-like cells (HLCs) and show immunomodulatory functions. Due to the increasing demand for fully characterized cell therapy vehicles warranting both the safety and efficacy of treatments, in this work, we extensively characterized WJ-MSCs before and after the application of a hepatocyte-directed differentiation protocol. HLCs exhibited a morphology resembling that of hepatocytes, expressed early and late hepatic markers (α-fetoprotein, albumin, CK18, HNF4-α), and acquired hepatic functions (glycogen synthesis, xenobiotics detoxification), as also revealed by the shotgun proteomics approach. HLCs maintained the same pattern of immunomodulatory molecule expression and mesenchymal markers, other than displaying specific enzymes, suggesting these cells as promising candidates for cellular therapy of ESLD. Our work shed new light on the basic biology of HLCs, suggesting new therapeutic approaches to treat ESLD.
Collapse
Affiliation(s)
- Melania Lo Iacono
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (F.R.)
| | - Simona Corrao
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy;
| | - Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (F.R.)
| | - Giandomenico Amico
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (G.A.); (F.T.); (P.G.C.)
- Unit of Regenerative Medicine and Immunotherapy, Ri.MED Foundation, 90133 Palermo, Italy
| | - Francesca Timoneri
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (G.A.); (F.T.); (P.G.C.)
- Unit of Regenerative Medicine and Immunotherapy, Ri.MED Foundation, 90133 Palermo, Italy
| | - Eleonora Russo
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Annamaria Cucina
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) University of Palermo, 90127 Palermo, Italy; (A.C.); (S.I.); (F.D.G.)
| | - Sergio Indelicato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) University of Palermo, 90127 Palermo, Italy; (A.C.); (S.I.); (F.D.G.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (F.R.)
- The Institute of Translational Pharmacology, National Research Council of Italy (CNR), 90146 Palermo, Italy
| | - Tiziana Corsello
- Department of Pediatrics, Division of Clinical and Experimental Immunology and Infectious Diseases (CEIID), University of Texas Medical Branch, Galveston, TX 77550, USA;
| | - Salvatore Saieva
- Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Antonino Di Stefano
- Laboratory of Cardio-Respiratory Apparatus Cytoimmunopathology, “S. Maugeri” Foundation, IRCCS, Medical Center of Veruno, 281010 Novara, Italy;
| | - Francesca Di Gaudio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) University of Palermo, 90127 Palermo, Italy; (A.C.); (S.I.); (F.D.G.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (G.A.); (F.T.); (P.G.C.)
| | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (F.R.)
| |
Collapse
|
3
|
Himstedt A, Rapp H, Stopfer P, Lotz R, Scheuerer S, Arnhold T, Sauer A, Borghardt JM. Beyond CL and V SS: A comprehensive approach to human pharmacokinetic predictions. Drug Discov Today 2024; 29:104238. [PMID: 39521329 DOI: 10.1016/j.drudis.2024.104238] [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: 08/03/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
This article presents a comprehensive examination of processes related to the prediction of human pharmacokinetics (PK), a crucial task of clinical drug candidate selection. By systematically incorporating in vitro absorption, distribution, metabolism and excretion (ADME) and in vivo PK data with expert judgement, the study achieves high-quality human PK predictions for 40 orally administered compounds from Boehringer Ingelheim's new chemical entity (NCE) portfolio. Overall, the article provides a detailed evaluation of and guidance for a structured process to predict full concentration-time profiles beyond single-parameter predictions, using state-of-the-art methodologies. Furthermore, it discusses future challenges and improvements, and aims to provide valuable insights for scientists working in drug metabolism and PK (DMPK) or PK/pharmacodynamics (PK/PD) modelling.
Collapse
Affiliation(s)
- Anneke Himstedt
- Global Research DMPK, Global Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Hermann Rapp
- Global Research DMPK, Global Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Peter Stopfer
- Clinical Pharmacology, Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ralf Lotz
- Nonclinical Pharmacokinetics, Global Nonclinical Safety and DMPK, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Stefan Scheuerer
- Nonclinical Pharmacokinetics, Global Nonclinical Safety and DMPK, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Thomas Arnhold
- Clinical Pharmacology, Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Achim Sauer
- Global Research DMPK, Global Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| | - Jens Markus Borghardt
- Global Research DMPK, Global Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Barnes JP, Yang SM, Thompson TS, Guevarra CM, Pleasant SS, Do TQ, Crouch RD. Mechanistic Investigation of the Time-Dependent Aldehyde Oxidase Inhibitor Hydralazine. Drug Metab Dispos 2023; 51:782-791. [PMID: 36921993 DOI: 10.1124/dmd.123.001257] [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: 01/11/2023] [Revised: 02/15/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
The anti-hypertensive agent hydralazine is a time-dependent inhibitor of the cytosolic drug-metabolizing enzyme aldehyde oxidase (AO). Glutathione (GSH) was found to suppress the inhibition of AO by hydralazine in multiple enzyme sources (human liver and kidney cytosol, human liver S9, rat liver S9, and recombinant human AO) and with different AO substrates (zoniporide, O6 -benzylguanine, and dantrolene). Hydralazine-induced AO inactivation was unaffected when GSH was added to the incubation mixture after pre-incubation of hydralazine with AO (rather than during the pre-incubation), suggesting that GSH traps a hydralazine reactive intermediate prior to enzyme inactivation. Consistent with previous reports of 1-phthalazylmercapturic acid formation when hydralazine was incubated with N-acetylcysteine, we detected a metabolite producing an MS/MS spectrum consistent with a 1-phthalazyl-GSH conjugate. O6 -Benzylguanine, an AO substrate, did not protect against hydralazine-induced AO inactivation, implying that hydralazine does not compete with O6 -benzylguanine for binding to the AO active site. Catalase also failed to protect AO from hydralazine-induced inactivation, suggesting that hydrogen peroxide is not involved. However, an allosteric AO inhibitor (thioridazine) offered some protection, indicating a catalytic role for AO in the bioactivation of hydralazine. AO inhibition by phthalazine (a substrate and inhibitor of AO and a metabolite of hydralazine) was unaffected by the presence of GSH. GSH also prevented hydralazine from inhibiting the nitro-reduction of dantrolene by AO. Furthermore, the GSH-hydralazine combination stimulated dantrolene reduction. Phthalazine inhibited only oxidation reactions, not reduction of dantrolene. Together, these results support the hypothesis that hydralazine is converted to a reactive intermediate that inactivates AO. SIGNIFICANCE STATEMENT: These studies suggest that a reactive intermediate of hydralazine plays a primary role in the mechanism of aldehyde oxidase (AO) inactivation. Inactivation was attenuated by glutathione and unaffected by catalase. Phthalazine (hydralazine metabolite) inhibited AO regardless of the presence of glutathione; however, phthalazine inhibited only oxidation reactions, while hydralazine inhibited both oxidation and reduction reactions. This report advances our mechanistic understanding of hydralazine as an AO inhibitor and provides information to facilitate appropriate use of hydralazine when probing AO metabolism.
Collapse
Affiliation(s)
- J Paige Barnes
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Shaoyun M Yang
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Taylor S Thompson
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Caithlyne M Guevarra
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Shaniya S Pleasant
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Tri Q Do
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| | - Rachel D Crouch
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee (J.P.B., S.M.Y., T.S.T., C.M.G., S.S.P., T.Q.D., R.D.C.); and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee (R.D.C.)
| |
Collapse
|
7
|
Esmaeeli M, Nimtz M, Jänsch L, Ruddock LW, Leimkühler S. Inactivation of Human Aldehyde Oxidase by Small Sulfhydryl-Containing Reducing Agents. Drug Metab Dispos 2023; 51:764-770. [PMID: 37012073 DOI: 10.1124/dmd.122.001244] [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: 12/27/2022] [Revised: 02/13/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023] Open
Abstract
Human aldehyde oxidase (hAOX1) is a molybdoflavoenzyme that belongs to the xanthine oxidase (XO) family. hAOX1 is involved in phase I drug metabolism, but its physiologic role is not fully understood to date, and preclinical studies consistently underestimated hAOX1 clearance. In the present work, we report an unexpected effect of the common sulfhydryl-containing reducing agents, e.g., dithiothreitol (DTT), on the activity of hAOX1 and mouse aldehyde oxidases. We demonstrate that this effect is due to the reactivity of the sulfido ligand bound at the molybdenum cofactor with the sulfhydryl groups. The sulfido ligand coordinated to the Mo atom in the XO family of enzymes plays a crucial role in the catalytic cycle and its removal results in the total inactivation of these enzymes. Because liver cytosols, S9 fractions, and hepatocytes are commonly used to screen the drug candidates for hAOX1, our study suggests that DTT treatment of these samples should be avoided, otherwise false negative results by an inactivated hAOX1 might be obtained. SIGNIFICANCE STATEMENT: This work characterizes the inactivation of human aldehyde oxidase (hAOX1) by sulfhydryl-containing agents and identifies the site of inactivation. The role of dithiothreitol in the inhibition of hAOX1 should be considered for the preparation of hAOX1-containing fractions for pharmacological studies on drug metabolism and drug clearance.
Collapse
Affiliation(s)
- Mariam Esmaeeli
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Manfred Nimtz
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Lothar Jänsch
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Lloyd W Ruddock
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Silke Leimkühler
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Non-cytochrome P450 enzymes involved in the oxidative metabolism of xenobiotics: Focus on the regulation of gene expression and enzyme activity. Pharmacol Ther 2021; 233:108020. [PMID: 34637840 DOI: 10.1016/j.pharmthera.2021.108020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Oxidative metabolism is one of the major biotransformation reactions that regulates the exposure of xenobiotics and their metabolites in the circulatory system and local tissues and organs, and influences their efficacy and toxicity. Although cytochrome (CY)P450s play critical roles in the oxidative reaction, extensive CYP450-independent oxidative metabolism also occurs in some xenobiotics, such as aldehyde oxidase, xanthine oxidoreductase, flavin-containing monooxygenase, monoamine oxidase, alcohol dehydrogenase, or aldehyde dehydrogenase-dependent oxidative metabolism. Drugs form a large portion of xenobiotics and are the primary target of this review. The common reaction mechanisms and roles of non-CYP450 enzymes in metabolism, factors affecting the expression and activity of non-CYP450 enzymes in terms of inhibition, induction, regulation, and species differences in pharmaceutical research and development have been summarized. These non-CYP450 enzymes are detoxifying enzymes, although sometimes they mediate severe toxicity. Synthetic or natural chemicals serve as inhibitors for these non-CYP450 enzymes. However, pharmacokinetic-based drug interactions through these inhibitors have rarely been reported in vivo. Although multiple mechanisms participate in the basal expression and regulation of non-CYP450 enzymes, only a limited number of inducers upregulate their expression. Therefore, these enzymes are considered non-inducible or less inducible. Overall, this review focuses on the potential xenobiotic factors that contribute to variations in gene expression levels and the activities of non-CYP450 enzymes.
Collapse
|
13
|
Uehara S, Yoneda N, Higuchi Y, Yamazaki H, Suemizu H. Oxidative metabolism and pharmacokinetics of the EGFR inhibitor BIBX1382 in chimeric NOG-TKm30 mice transplanted with human hepatocytes. Drug Metab Pharmacokinet 2021; 41:100419. [PMID: 34624627 DOI: 10.1016/j.dmpk.2021.100419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022]
Abstract
The epidermal growth factor receptor inhibitor BIBX1382 has failed in drug development because of poor oral exposure and low bioavailability associated with its extensive metabolism by aldehyde oxidase (AOX) in humans. In this study, we investigated the metabolic profiles and pharmacokinetics of BIBX1382 in chimeric NOG-TKm30 mice with humanized liver (humanized liver mice). After intravenous and oral BIBX1382 administration, increased plasma clearance and decreased oral exposure together with high production of the predominant oxidative metabolite (M1, BIBU1476) and secondary oxidized metabolite (M2) were observed in humanized liver mice. Extensive oxidation rates of BIBX1382 were observed in hepatocytes from humanized liver mice and were suppressed by the typical human AOX1 inhibitors raloxifene and hydralazine. Liver cytosolic fractions from humans, humanized liver mice, cynomolgus monkeys, minipigs, and guinea pigs, but not fractions from dogs, rabbits, rats, and mice, displayed high BIBX1382 clearance and resulted in oxidative metabolite production. These results indicate that humanized liver mice have human-type AOX activity based on the transplanted human liver AOX1 function. Humanized liver mice can be considered an important animal model for understanding the metabolism and pharmacokinetics of AOX drug substrates.
Collapse
Affiliation(s)
- Shotaro Uehara
- Central Institute for Experimental Animals, Kawasaki, Japan.
| | - Nao Yoneda
- Central Institute for Experimental Animals, Kawasaki, Japan
| | | | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan
| | | |
Collapse
|
14
|
Paragas EM, Choughule K, Jones JP, Barr JT. Enzyme Kinetics, Pharmacokinetics, and Inhibition of Aldehyde Oxidase. Methods Mol Biol 2021; 2342:257-284. [PMID: 34272698 DOI: 10.1007/978-1-0716-1554-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aldehyde oxidase (AO) has emerged as an important drug metabolizing enzyme over the last decade. Several compounds have failed in the clinic because the clearance or toxicity was underestimated by preclinical species. Human AO is much more active than rodent AO, and dogs do not have functional AO. Metabolic products from AO-catalyzed oxidation are generally nonreactive and often they have much lower solubility. AO metabolism is not limited to oxidation as AO can also catalyze reduction of oxygen and nitrite. Reduction of oxygen leads to the reactive oxygen species (ROS) superoxide radical anion and hydrogen peroxide. Reduction of nitrite leads to the formation of nitric oxide with potential pharmacological implications. AO is also reported to catalyze the reductive metabolism of nitro-compounds, N-oxides, sulfoxides, isoxazoles, isothiazoles, nitrite, and hydroxamic acids. These reductive transformations may cause toxicity due to the formation of reactive metabolites. Moreover, the inhibition kinetics are complex, and multiple probe substrates should be used when assessing the potential for DDIs. Finally, AO appears to be amenable to computational predictions of both regioselectivity and rates of reaction, which holds promise for virtual screening.
Collapse
Affiliation(s)
- Erickson M Paragas
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Kanika Choughule
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, Boston, MA, USA
| | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - John T Barr
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, South San Francisco, CA, USA.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Cheshmazar N, Dastmalchi S, Terao M, Garattini E, Hamzeh-Mivehroud M. Aldehyde oxidase at the crossroad of metabolism and preclinical screening. Drug Metab Rev 2019; 51:428-452. [DOI: 10.1080/03602532.2019.1667379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Narges Cheshmazar
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mineko Terao
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
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
|
20
|
Affiliation(s)
- Christine Beedham
- Honorary Senior Lecturer, Faculty of Life Sciences, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
21
|
Abbasi A, Paragas EM, Joswig-Jones CA, Rodgers JT, Jones JP. Time Course of Aldehyde Oxidase and Why It Is Nonlinear. Drug Metab Dispos 2019; 47:473-483. [PMID: 30787100 PMCID: PMC6439458 DOI: 10.1124/dmd.118.085787] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
Abstract
Many promising drug candidates metabolized by aldehyde oxidase (AOX) fail during clinical trial owing to underestimation of their clearance. AOX is species-specific, which makes traditional allometric studies a poor choice for estimating human clearance. Other studies have suggested using half-life calculated by measuring substrate depletion to measure clearance. In this study, we proposed using numerical fitting to enzymatic pathways other than Michaelis-Menten (MM) to avoid missing the initial high turnover rate of product formation. Here, product formation over a 240-minute time course of six AOX substrates-O6-benzylguanine, N-(2-dimethylamino)ethyl)acridine-4-carboxamide, zaleplon, phthalazine, BIBX1382 [N8-(3-Chloro-4-fluorophenyl)-N2-(1-methyl-4-piperidinyl)-pyrimido[5,4-d]pyrimidine-2,8-diamine dihydrochloride], and zoniporide-have been provided to illustrate enzyme deactivation over time to help better understand why MM kinetics sometimes leads to underestimation of rate constants. Based on the data provided in this article, the total velocity for substrates becomes slower than the initial velocity by 3.1-, 6.5-, 2.9-, 32.2-, 2.7-, and 0.2-fold, respectively, in human expressed purified enzyme, whereas the K m remains constant. Also, our studies on the role of reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, show that ROS did not significantly alter the change in enzyme activity over time. Providing a new electron acceptor, 5-nitroquinoline, did, however, alter the change in rate over time for mumerous compounds. The data also illustrate the difficulties in using substrate disappearance to estimate intrinsic clearance.
Collapse
Affiliation(s)
- Armina Abbasi
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Erickson M Paragas
- Department of Chemistry, Washington State University, Pullman, Washington
| | | | - John T Rodgers
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, Washington
| |
Collapse
|
22
|
Xie J, Saburulla NF, Chen S, Wong SY, Yap ZP, Zhang LH, Lau AJ. Evaluation of Carbazeran 4-Oxidation and O 6-Benzylguanine 8-Oxidation as Catalytic Markers of Human Aldehyde Oxidase: Impact of Cytosolic Contamination of Liver Microsomes. Drug Metab Dispos 2019; 47:26-37. [PMID: 30337443 DOI: 10.1124/dmd.118.082099] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/15/2018] [Indexed: 02/13/2025] Open
Abstract
The present study investigated the contribution of microsomal cytochrome P450 and cytosolic aldehyde oxidase-1 (AOX-1) to carbazeran 4-oxidation and O 6-benzylguanine 8-oxidation in human liver microsomal, cytosolic, and S9 fractions. Incubations containing carbazeran and human liver microsomes with or without exogenously added NADPH yielded comparable levels of 4-oxo-carbazeran. O 6-Benzylguanine 8-oxidation occurred in microsomal incubations, and the extent was increased by NADPH. Human recombinant CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5 did not catalyze carbazeran 4-oxidation, whereas CYP1A2 was highly active in O 6-benzylguanine 8-oxidation. 1-Aminobenzotriazole, a pan-cytochrome P450 inhibitor, decreased O 6-benzylguanine 8-oxidation, but not carbazeran 4-oxidation, in microsomal incubations, whereas 1-aminobenzotriazole and furafylline (a CYP1A2-selective inhibitor) did not inhibit carbazeran 4-oxidation or O 6-benzylguanine 8-oxidation in human liver S9 fraction. Carbazeran 4-oxidation in incubations containing human liver microsomes (from multiple donors and commercial suppliers) was attributed to microsomal preparations contaminated with AOX-1, as suggested by liver microsomal experiments indicating a decrease in carbazeran 4-oxidation by an AOX-1 inhibitor (hydralazine), and to detection of AOX-1 protein (at one-third the level of that in liver cytosol). Cytosolic contamination of liver microsomes was further demonstrated by the formation of dehydroepiandrosterone sulfate (catalyzed by cytosolic sulfotransferases) in liver microsomal incubations containing dehydroepiandrosterone. In conclusion, carbazeran 4-oxidation and O 6-benzylguanine 8-oxidation are enzyme-selective catalytic markers of human AOX-1, as shown in human liver S9 fraction expressing cytochrome P450 and AOX-1. This study highlights the negative impact of cytosolic contamination of liver microsomes on the interpretation of reaction phenotyping data collected in an in vitro study performed in microsomal fractions.
Collapse
Affiliation(s)
- Jiarong Xie
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Nur Fazilah Saburulla
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Shiyan Chen
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Siew Ying Wong
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Ze Ping Yap
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Linghua Harris Zhang
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| | - Aik Jiang Lau
- Department of Pharmacy, Faculty of Science (J.X., N.F.S., S.C., S.Y.W., Z.P.Y., A.J.L.) and Department of Pharmacology, Yong Loo Lin School of Medicine (A.J.L.), National University of Singapore, Singapore; and NANO BIOTEC, LLC., Whippany, New Jersey (L.H.Z.)
| |
Collapse
|
23
|
Yilmaz Y, Williams G, Walles M, Manevski N, Krähenbühl S, Camenisch G. Comparison of Rat and Human Pulmonary Metabolism Using Precision-cut Lung Slices (PCLS). Drug Metab Lett 2019; 13:53-63. [PMID: 30345935 DOI: 10.2174/1872312812666181022114622] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/28/2018] [Accepted: 10/08/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Although the liver is the primary organ of drug metabolism, the lungs also contain drug-metabolizing enzymes and may, therefore, contribute to the elimination of drugs. In this investigation, the Precision-cut Lung Slice (PCLS) technique was standardized with the aims of characterizing and comparing rat and human pulmonary drug metabolizing activity. METHOD Due to the limited availability of human lung tissue, standardization of the PCLS method was performed with rat lung tissue. Pulmonary enzymatic activity was found to vary significantly with rat age and rat strain. The Dynamic Organ Culture (DOC) system was superior to well-plates for tissue incubations, while oxygen supply appeared to have a limited impact within the 4h incubation period used here. RESULTS The metabolism of a range of phase I and phase II probe substrates was assessed in rat and human lung preparations. Cytochrome P450 (CYP) activity was relatively low in both species, whereas phase II activity appeared to be more significant. CONCLUSION PCLS is a promising tool for the investigation of pulmonary drug metabolism. The data indicates that pulmonary CYP activity is relatively low and that there are significant differences in enzyme activity between rat and human lung.
Collapse
Affiliation(s)
- Yildiz Yilmaz
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Gareth Williams
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Nenad Manevski
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Stephan Krähenbühl
- Clinical Pharmacology and Toxicology, University Hospital, Basel, Switzerland
| | - Gian Camenisch
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
24
|
Zhang JW, Xiao W, Gao ZT, Yu ZT, Zhang JYJ. Metabolism of c-Met Kinase Inhibitors Containing Quinoline by Aldehyde Oxidase, Electron Donating, and Steric Hindrance Effect. Drug Metab Dispos 2018; 46:1847-1855. [PMID: 30209037 DOI: 10.1124/dmd.118.081919] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/10/2018] [Indexed: 11/22/2022] Open
Abstract
Some quinoline-containing c-Met kinase inhibitors are aldehyde oxidase (AO) substrates. 3-Substituted quinoline triazolopyridine analogs were synthesized to understand the electron-donating and steric hindrance effects on AO-mediated metabolism. Metabolic stability studies for these quinoline analogs were carried out in liver cytosol from mice, rats, cynomolgus monkeys, and humans. Several 3-N-substituted analogs were found to be unstable in monkey liver cytosolic incubations (half-life, <10 minutes), and five of them (63, 53, 51, 11, and 71) were chosen for additional mechanistic studies. Mono-oxygenation on the quinoline ring was identified by liquid chromatography tandem mass spectrometry. Metabolite formation was inhibited by the AO inhibitors menadione and raloxifene, but not by the xanthine oxidase inhibitor allopurinol. It was found that small electron-donating groups at the 3-quinoline moiety made the analogs more susceptible to AO metabolism, whereas large 3-substituents could reverse the trend. Although species differences were observed, this trend was applicable to all species tested. Small electron-donating substituents at the 3-quinoline moiety increased both affinity (decreased Michaelis constant) and V max maximum velocity toward AO in kinetic studies, whereas large substituents decreased both parameters probably as a result of steric hindrance. Based on our analysis, a common structural feature with high AO liability was proposed. Our finding could provide useful information for chemists to minimize potential AO liability when designing quinoline analogs.
Collapse
Affiliation(s)
- Jiang Wei Zhang
- China Novartis Institutes for BioMedical Research, Shanghai, People's Republic of China
| | - Wen Xiao
- China Novartis Institutes for BioMedical Research, Shanghai, People's Republic of China
| | - Zhen Ting Gao
- China Novartis Institutes for BioMedical Research, Shanghai, People's Republic of China
| | - Zheng Tian Yu
- China Novartis Institutes for BioMedical Research, Shanghai, People's Republic of China
| | - Ji Yue Jeff Zhang
- China Novartis Institutes for BioMedical Research, Shanghai, People's Republic of China
| |
Collapse
|
25
|
Substrate selectivity of human aldehyde oxidase 1 in reduction of nitroaromatic drugs. Arch Biochem Biophys 2018; 659:85-92. [DOI: 10.1016/j.abb.2018.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/27/2018] [Accepted: 10/23/2018] [Indexed: 11/17/2022]
|
26
|
Burton RD, Hieronymus T, Chamem T, Heim D, Anderson S, Zhu X, Hutzler JM. Assessment of the Biotransformation of Low-Turnover Drugs in the H µREL Human Hepatocyte Coculture Model. Drug Metab Dispos 2018; 46:1617-1625. [PMID: 30135244 DOI: 10.1124/dmd.118.082867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/15/2018] [Indexed: 01/24/2023] Open
Abstract
Metabolic profiles of four drugs possessing diverse metabolic pathways (timolol, meloxicam, linezolid, and XK469) were compared following incubations in both suspended cryopreserved human hepatocytes and the HμREL hepatocyte coculture model. In general, minimal metabolism was observed following 4-hour incubations in both suspended hepatocytes and the HμREL model, whereas incubations conducted up to 7 days in the HμREL coculture model resulted in more robust metabolic turnover. In the case of timolol, in vivo human data suggest that 22% of the dose is transformed via multistep oxidative opening of the morpholine moiety. Only the first-step oxidation was detected in suspended hepatocytes, whereas the relevant downstream metabolites were produced in the HµREL model. For meloxicam, both the hydroxymethyl and subsequent carboxylic acid metabolites were abundant following incubation in the HμREL model, while only a trace amount of the hydroxymethyl metabolite was observed in suspension. Similar to timolol, linezolid generated substantially higher levels of morpholine ring-opened carboxylic acid metabolites in the HμREL model. Finally, while the major aldehyde oxidase-mediated mono-oxidative metabolite of XK469 was minimally produced in hepatocyte suspension, the HμREL model robustly produced this metabolite, consistent with a pathway reported to account for 54% of the total urinary excretion in human. In addition, low-level taurine and glycine conjugates were identified in the HµREL model. In summary, continuous metabolite production was observed for up to 7 days of incubation in the HµREL model, covering cytochrome P450, aldehyde oxidase, and numerous conjugative pathways, while predominant metabolites correlated with relevant metabolites reported in human in vivo studies.
Collapse
Affiliation(s)
- Richard D Burton
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Todd Hieronymus
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Taysir Chamem
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - David Heim
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Shelby Anderson
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Xiaochun Zhu
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - J Matthew Hutzler
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| |
Collapse
|
27
|
Huang S, Adams E, Van Schepdael A. Study of aldehyde oxidase by micellar electrokinetic chromatography separation of O 6 -benzylguanine and 8-oxo-O 6 -benzylguanine. Electrophoresis 2018; 40:330-335. [PMID: 30221782 DOI: 10.1002/elps.201800279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/21/2018] [Accepted: 09/07/2018] [Indexed: 12/21/2022]
Abstract
A separation method for O6 -benzylguanine (O6 -BG) and 8-oxo-O6 -benzylguanine (8-oxo-O6 -BG) is developed by using MEKC. This study includes the optimization of separation and incubation parameters for both off-line and on-line procedures. The BGE consisted of 25 mM sodium phosphate buffer-methanol (70:30, v/v), apparent pH 7.4, in which SDS and methyl-β-cyclodextrin were dissolved yielding final concentrations of 50 and 15 mM, respectively. Separations were performed at 15 kV using an untreated fused-silica capillary (40 cm length, effective length is 30 cm) with the detection wavelength at 195 nm. The capillary was kept at 15°C. Good performances were demonstrated for the repeatability and linearity. The LOQ was determined to be 14 μM for 8-oxo-O6 -BG (S/N = 10). The accuracy values showed a bias of +7.9% for 50 μM and -7.0% for 100 μM. Premix and transverse diffusion of laminar flow profiles (TDLFP) methods were used for on-line mixing and reaction of the substrate O6 -BG with aldehyde oxidase. Both procedures were successful in mixing as well as subsequent separation of the substrate and the metabolite, while the repeatability of TDLFP (14.7% (n = 3)) was much better than the premix technique.
Collapse
Affiliation(s)
- Shengyun Huang
- KU Leuven-University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, Leuven, Belgium
| | - Erwin Adams
- KU Leuven-University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, Leuven, Belgium
| | - Ann Van Schepdael
- KU Leuven-University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, Leuven, Belgium
| |
Collapse
|
28
|
Zheng J, Xin Y, Zhang J, Subramanian R, Murray BP, Whitney JA, Warr MR, Ling J, Moorehead L, Kwan E, Hemenway J, Smith BJ, Silverman JA. Pharmacokinetics and Disposition of Momelotinib Revealed a Disproportionate Human Metabolite-Resolution for Clinical Development. Drug Metab Dispos 2018; 46:237-247. [PMID: 29311136 DOI: 10.1124/dmd.117.078899] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022] Open
Abstract
Momelotinib (MMB), a small-molecule inhibitor of Janus kinase (JAK)1/2 and of activin A receptor type 1 (ACVR1), is in clinical development for the treatment of myeloproliferative neoplasms. The pharmacokinetics and disposition of [14C]MMB were characterized in a single-dose, human mass-balance study. Metabolism and the pharmacologic activity of key metabolites were elucidated in multiple in vitro and in vivo experiments. MMB was rapidly absorbed following oral dosing with approximately 97% of the radioactivity recovered, primarily in feces with urine as a secondary route. Mean blood-to-plasma [14C] area under the plasma concentration-time curve ratio was 0.72, suggesting low association of MMB and metabolites with blood cells. [14C]MMB-derived radioactivity was detectable in blood for ≤48 hours, suggesting no irreversible binding of MMB or its metabolites. The major circulating human metabolite, M21 (a morpholino lactam), is a potent inhibitor of JAK1/2 and ACVR1 in vitro. Estimation of pharmacological activity index suggests M21 contributes significantly to the pharmacological activity of MMB for the inhibition of both JAK1/2 and ACVR1. M21 was observed in disproportionately higher amounts in human plasma than in rat or dog, the rodent and nonrodent species used for the general nonclinical safety assessment of this molecule. This discrepancy was resolved with additional nonclinical studies wherein the circulating metabolites and drug-drug interactions were further characterized. The human metabolism of MMB was mediated primarily by multiple cytochrome P450 enzymes, whereas M21 formation involved initial P450 oxidation of the morpholine ring followed by metabolism via aldehyde oxidase.
Collapse
Affiliation(s)
- Jim Zheng
- Gilead Sciences, Inc., Foster City, California
| | - Yan Xin
- Gilead Sciences, Inc., Foster City, California
| | | | | | | | | | | | - John Ling
- Gilead Sciences, Inc., Foster City, California
| | | | - Ellen Kwan
- Gilead Sciences, Inc., Foster City, California
| | | | | | | |
Collapse
|
29
|
Offline derivatization LC–MS/MS method for simultaneous estimation of vanillin and vanillic acid in guinea pig plasma. Bioanalysis 2018; 10:131-142. [DOI: 10.4155/bio-2017-0213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Vanillin used as a positive control substrate of aldehyde oxidase activity gets metabolized to vanillic acid. Low MW and low sensitivity in negative ion mode are challenges with these analytes. Our objective was to develop a simple offline derivatization LC–MS/MS method to address these challenges. Methodology/results: A simple dansyl chloride derivatization of the phenolic groups on vanillin and vanillic acid was adopted to enable easy ionization in commonly used acidic mobile phases. Calibration curves were linear over the concentrations of 4.88–1250 nM with an LLOQ of 0.64 fmoles on column for both analytes. Conclusion: The qualified method was successfully applied to simultaneously measure vanillin and vanillic acid in plasma and urine from a guinea pig pharmacokinetic study.
Collapse
|
30
|
Cruciani G, Milani N, Benedetti P, Lepri S, Cesarini L, Baroni M, Spyrakis F, Tortorella S, Mosconi E, Goracci L. From Experiments to a Fast Easy-to-Use Computational Methodology to Predict Human Aldehyde Oxidase Selectivity and Metabolic Reactions. J Med Chem 2017; 61:360-371. [DOI: 10.1021/acs.jmedchem.7b01552] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Gabriele Cruciani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| | - Nicolò Milani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Paolo Benedetti
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| | - Susan Lepri
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Lucia Cesarini
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Massimo Baroni
- Molecular Discovery Ltd, Centennial
Park, Borehamwood, Hertfordshire, United Kingdom
| | - Francesca Spyrakis
- Department
of Drug Science and Technology, University of Turin, via P. Giuria
9, 10125 Turin, Italy
| | - Sara Tortorella
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
- Molecular Horizon srl, via Montelino
32, 06084 Bettona, Italy
| | - Edoardo Mosconi
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
- Computational
Laboratory for Hybrid/Organic Photovoltaics, National Research Council−Institute of Molecular Science and Technologies, Via Elce
di Sotto 8, I-06123 Perugia, Italy
| | - Laura Goracci
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
31
|
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
|
32
|
Paragas E, Humphreys SC, Min J, Joswig-Jones CA, Leimkühler S, Jones JP. ecoAO: A Simple System for the Study of Human Aldehyde Oxidases Role in Drug Metabolism. ACS OMEGA 2017; 2:4820-4827. [PMID: 28884164 PMCID: PMC5579547 DOI: 10.1021/acsomega.7b01054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/09/2017] [Indexed: 06/07/2023]
Abstract
Although aldehyde oxidase (AO) is an important hepatic drug-metabolizing enzyme, it remains understudied and is consequently often overlooked in preclinical studies, an oversight that has resulted in the failure of multiple clinical trials. AO's preclusion to investigation stems from the following: (1) difficulties synthesizing metabolic standards due to the chemospecificity and regiospecificity of the enzyme and (2) significant inherent variability across existing in vitro systems including liver cytosol, S9 fractions, and primary hepatocytes, which lack specificity and generate discordant expression and activity profiles. Here, we describe a practical bacterial biotransformation system, ecoAO, addressing both issues simultaneously. ecoAO is a cell paste of MoCo-producing Escherichia coli strain TP1017 expressing human AO. It exhibits specific activity toward known substrates, zoniporide, 4-trans-(N,N-dimethylamino)cinnamaldehyde, O6-benzylguanine, and zaleplon; it also has utility as a biocatalyst, yielding milligram quantities of synthetically challenging metabolite standards such as 2-oxo-zoniporide. Moreover, ecoAO enables routine determination of kcat and V/K, which are essential parameters for accurate in vivo clearance predictions. Furthermore, ecoAO has potential as a preclinical in vitro screening tool for AO activity, as demonstrated by its metabolism of 3-aminoquinoline, a previously uncharacterized substrate. ecoAO promises to provide easy access to metabolites with the potential to improve pharmacokinetic clearance predictions and guide drug development.
Collapse
Affiliation(s)
- Erickson
M. Paragas
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Sara C. Humphreys
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Joshua Min
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Carolyn A. Joswig-Jones
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Silke Leimkühler
- Department
of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Jeffrey P. Jones
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| |
Collapse
|
33
|
Kumar R, Joshi G, Kler H, Kalra S, Kaur M, Arya R. Toward an Understanding of Structural Insights of Xanthine and Aldehyde Oxidases: An Overview of their Inhibitors and Role in Various Diseases. Med Res Rev 2017; 38:1073-1125. [DOI: 10.1002/med.21457] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/05/2017] [Accepted: 06/13/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Raj Kumar
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
| | - Harveen Kler
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
| | - Sourav Kalra
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
- Centre for Human Genetics and Molecular Medicine
| | - Manpreet Kaur
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
| | - Ramandeep Arya
- Laboratory for Drug Design and Synthesis, Centre for Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences; Central University of Punjab; Bathinda 151001 India
| |
Collapse
|
34
|
Felts A, Rodriguez AL, Blobaum AL, Morrison RD, Bates BS, Thompson Gray A, Rook JM, Tantawy MN, Byers FW, Chang S, Venable DF, Luscombe VB, Tamagnan GD, Niswender CM, Daniels JS, Jones CK, Conn PJ, Lindsley CW, Emmitte KA. Discovery of N-(5-Fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (VU0424238): A Novel Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 5 Selected for Clinical Evaluation. J Med Chem 2017; 60:5072-5085. [PMID: 28530802 PMCID: PMC5484149 DOI: 10.1021/acs.jmedchem.7b00410] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Indexed: 12/22/2022]
Abstract
Preclinical evidence in support of the potential utility of mGlu5 NAMs for the treatment of a variety of psychiatric and neurodegenerative disorders is extensive, and multiple such molecules have entered clinical trials. Despite some promising results from clinical studies, no small molecule mGlu5 NAM has yet to reach market. Here we present the discovery and evaluation of N-(5-fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (27, VU0424238), a compound selected for clinical evaluation. Compound 27 is more than 900-fold selective for mGlu5 versus the other mGlu receptors, and binding studies established a Ki value of 4.4 nM at a known allosteric binding site. Compound 27 had a clearance of 19.3 and 15.5 mL/min/kg in rats and cynomolgus monkeys, respectively. Imaging studies using a known mGlu5 PET ligand demonstrated 50% receptor occupancy at an oral dose of 0.8 mg/kg in rats and an intravenous dose of 0.06 mg/kg in baboons.
Collapse
Affiliation(s)
- Andrew
S. Felts
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Alice L. Rodriguez
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Anna L. Blobaum
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Ryan D. Morrison
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Brittney S. Bates
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Analisa Thompson Gray
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jerri M. Rook
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mohammed N. Tantawy
- Department
of Radiology and Radiological Sciences, Vanderbilt University Institute
of Imaging Science, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
| | - Frank W. Byers
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sichen Chang
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Daryl F. Venable
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Vincent B. Luscombe
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Gilles D. Tamagnan
- Molecular
NeuroImaging, a Division of inviCRO, New Haven, Connecticut 06510, United States
| | - Colleen M. Niswender
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - J. Scott Daniels
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Carrie K. Jones
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Vanderbilt
Center for Neuroscience Drug Discovery, Department of
Pharmacology and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
35
|
Wilkinson DJ, Southall RL, Li M, Wright LM, Corfield LJ, Heeley TA, Bratby B, Mannu R, Johnson SL, Shaw V, Friett HL, Blakeburn LA, Kendrick JS, Otteneder MB. Minipig and Human Metabolism of Aldehyde Oxidase Substrates: In Vitro–In Vivo Comparisons. AAPS JOURNAL 2017; 19:1163-1174. [DOI: 10.1208/s12248-017-0087-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/10/2017] [Indexed: 12/19/2022]
|
36
|
Crouch RD, Hutzler JM, Daniels JS. A novel in vitro allometric scaling methodology for aldehyde oxidase substrates to enable selection of appropriate species for traditional allometry. Xenobiotica 2017; 48:219-231. [PMID: 28281401 DOI: 10.1080/00498254.2017.1296208] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
1. Failure to predict human pharmacokinetics of aldehyde oxidase (AO) substrates using traditional allometry has been attributed to species differences in AO metabolism. 2. To identify appropriate species for predicting human in vivo clearance by single-species scaling (SSS) or multispecies allometry (MA), we scaled in vitro intrinsic clearance (CLint) of five AO substrates obtained from hepatic S9 of mouse, rat, guinea pig, monkey and minipig to human in vitro CLint. 3. When predicting human in vitro CLint, average absolute fold-error was ≤2.0 by SSS with monkey, minipig and guinea pig (rat/mouse >3.0) and was <3.0 by most MA species combinations (including rat/mouse combinations). 4. Interspecies variables, including fraction metabolized by AO (Fm,AO) and hepatic extraction ratios (E) were estimated in vitro. SSS prediction fold-errors correlated with the animal:human ratio of E (r2 = 0.6488), but not Fm,AO (r2 = 0.0051). 5. Using plasma clearance (CLp) from the literature, SSS with monkey was superior to rat or mouse at predicting human CLp of BIBX1382 and zoniporide, consistent with in vitro SSS assessments. 6. Evaluation of in vitro allometry, Fm,AO and E may prove useful to guide selection of suitable species for traditional allometry and prediction of human pharmacokinetics of AO substrates.
Collapse
Affiliation(s)
- Rachel D Crouch
- a Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA and
| | - J Matthew Hutzler
- b Q2 Solutions, Bioanalytical and ADME Labs , Indianapolis , IN , USA
| | - J Scott Daniels
- a Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA and
| |
Collapse
|
37
|
Miyamoto M, Iwasaki S, Chisaki I, Nakagawa S, Amano N, Hirabayashi H. Comparison of predictability for human pharmacokinetics parameters among monkeys, rats, and chimeric mice with humanised liver. Xenobiotica 2017; 47:1052-1063. [DOI: 10.1080/00498254.2016.1265160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Maki Miyamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Sayaka Nakagawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
38
|
Jensen KG, Jacobsen AM, Bundgaard C, Nilausen DØ, Thale Z, Chandrasena G, Jørgensen M. Lack of Exposure in a First-in-Man Study Due to Aldehyde Oxidase Metabolism: Investigated by Use of 14C-microdose, Humanized Mice, Monkey Pharmacokinetics, and In Vitro Methods. Drug Metab Dispos 2017; 45:68-75. [PMID: 27737930 DOI: 10.1124/dmd.116.072793] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/12/2016] [Indexed: 11/22/2022] Open
Abstract
Inclusion of a microdose of 14C-labeled drug in the first-in-man study of new investigational drugs and subsequent analysis by accelerator mass spectrometry has become an integrated part of drug development at Lundbeck. It has been found to be highly informative with regard to investigations of the routes and rates of excretion of the drug and the human metabolite profiles according to metabolites in safety testing guidance and also when additional metabolism-related issues needed to be addressed. In the first-in-man study with the NCE Lu AF09535, contrary to anticipated, surprisingly low exposure was observed when measuring the parent compound using conventional bioanalysis. Parallel accelerator mass spectrometry analysis revealed that the low exposure was almost exclusively attributable to extensive metabolism. The metabolism observed in humans was mediated via a human specific metabolic pathway, whereas an equivalent extent of metabolism was not observed in preclinical species. In vitro, incubation studies in human liver cytosol revealed involvement of aldehyde oxidase (AO) in the biotransformation of Lu AF09535. In vivo, substantially lower plasma exposure of Lu AF09535 was observed in chimeric mice with humanized livers compared with control animals. In addition, Lu AF09535 exhibited very low oral bioavailability in monkeys despite relatively low clearance after intravenous administration in contrast to the pharmacokinetics in rats and dogs, both showing low clearance and high bioavailability. The in vitro and in vivo methods applied were proved useful for identifying and evaluating AO-dependent metabolism. Different strategies to integrate these methods for prediction of in vivo human clearance of AO substrates were evaluated.
Collapse
Affiliation(s)
- Klaus Gjervig Jensen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Anne-Marie Jacobsen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Christoffer Bundgaard
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Dorrit Østergaard Nilausen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Zia Thale
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Gamini Chandrasena
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Martin Jørgensen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| |
Collapse
|
39
|
Argikar UA, Potter PM, Hutzler JM, Marathe PH. Challenges and Opportunities with Non-CYP Enzymes Aldehyde Oxidase, Carboxylesterase, and UDP-Glucuronosyltransferase: Focus on Reaction Phenotyping and Prediction of Human Clearance. AAPS JOURNAL 2016; 18:1391-1405. [PMID: 27495117 DOI: 10.1208/s12248-016-9962-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/13/2016] [Indexed: 01/28/2023]
Abstract
Over the years, significant progress has been made in reducing metabolic instability due to cytochrome P450-mediated oxidation. High-throughput metabolic stability screening has enabled the advancement of compounds with little to no oxidative metabolism. Furthermore, high lipophilicity and low aqueous solubility of presently pursued chemotypes reduces the probability of renal excretion. As such, these low microsomal turnover compounds are often substrates for non-CYP-mediated metabolism. UGTs, esterases, and aldehyde oxidase are major enzymes involved in catalyzing such metabolism. Hepatocytes provide an excellent tool to identify such pathways including elucidation of major metabolites. To predict human PK parameters for P450-mediated metabolism, in vitro-in vivo extrapolation using hepatic microsomes, hepatocytes, and intestinal microsomes has been actively investigated. However, such methods have not been sufficiently evaluated for non-P450 enzymes. In addition to the involvement of the liver, extrahepatic enzymes (intestine, kidney, lung) are also likely to contribute to these pathways. While there has been considerable progress in predicting metabolic pathways and clearance primarily mediated by the liver, progress in characterizing extrahepatic metabolism and prediction of clearance has been slow. Well-characterized in vitro systems or in vivo animal models to assess drug-drug interaction potential and intersubject variability due to polymorphism are not available. Here we focus on the utility of appropriate in vitro studies to characterize non-CYP-mediated metabolism and to understand the enzymes involved followed by pharmacokinetic studies in the appropriately characterized surrogate species. The review will highlight progress made in establishing in vitro-in vivo correlation, predicting human clearance and avoiding costly clinical failures when non-CYP-mediated metabolic pathways are predominant.
Collapse
Affiliation(s)
- Upendra A Argikar
- Analytical Sciences and Imaging, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts, USA
| | - Philip M Potter
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - J Matthew Hutzler
- Q2 Solutions, Bioanalytical and ADME Labs, Indianapolis, Indiana, USA
| | - Punit H Marathe
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, New Jersey, USA.
| |
Collapse
|
40
|
Cerny MA. Prevalence of Non-Cytochrome P450-Mediated Metabolism in Food and Drug Administration-Approved Oral and Intravenous Drugs: 2006-2015. Drug Metab Dispos 2016; 44:1246-52. [PMID: 27084892 DOI: 10.1124/dmd.116.070763] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/14/2016] [Indexed: 02/13/2025] Open
Abstract
In recent years, claims of increased involvement of non-cytochrome P450 (non-P450) enzymes in the metabolism of drugs have appeared in the literature. However, no temporal summaries of the contribution of non-P450 enzymes to the metabolism of drugs have been published. Using data from human radiolabeled absorption, distribution, metabolism, and excretion studies available for a set of 125 orally or intravenously administered small-molecule drugs approved by the United States Food and Drug Administration from 2006 to 2015, the contributions of P450 and non-P450 enzymes to the formation of major metabolites (≥10% of dose) were assessed and tabulated. Over this time frame, the involvement of P450 versus non-P450 enzymes in the formation of major metabolites is compared, and the individual non-P450 enzymes responsible are described. This analysis indicates that non-P450 enzymes contribute significantly to the metabolism of the 125 drugs analyzed. Approximately 30% of the metabolism of these drugs is carried out by non-P450 enzymes, with the predominant non-P450 enzymes identified being glucuronosyltransferases (11.7%), hydrolases (10.8%), carbonyl reductases (2.4%), and aldehyde oxidase (1.1%). Although significant, the relative contribution of non-P450 enzymes to drug metabolism does not appear to have increased dramatically over the last 10 years. As the current evaluation involves drugs which emerged from the discovery phase >10 years ago, this evaluation may not reflect the current or evolving situation in some research organizations; therefore, additional monitoring and assessment of the involvement of non-P450 enzymes in the metabolism of drugs will be conducted in the future.
Collapse
Affiliation(s)
- Matthew A Cerny
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Pfizer, Inc., Groton, Connecticut
| |
Collapse
|
41
|
Foti RS, Dalvie DK. Cytochrome P450 and Non-Cytochrome P450 Oxidative Metabolism: Contributions to the Pharmacokinetics, Safety, and Efficacy of Xenobiotics. Drug Metab Dispos 2016; 44:1229-45. [PMID: 27298339 DOI: 10.1124/dmd.116.071753] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022] Open
Abstract
The drug-metabolizing enzymes that contribute to the metabolism or bioactivation of a drug play a crucial role in defining the absorption, distribution, metabolism, and excretion properties of that drug. Although the overall effect of the cytochrome P450 (P450) family of drug-metabolizing enzymes in this capacity cannot be understated, advancements in the field of non-P450-mediated metabolism have garnered increasing attention in recent years. This is perhaps a direct result of our ability to systematically avoid P450 liabilities by introducing chemical moieties that are not susceptible to P450 metabolism but, as a result, may introduce key pharmacophores for other drug-metabolizing enzymes. Furthermore, the effects of both P450 and non-P450 metabolism at a drug's site of therapeutic action have also been subject to increased scrutiny. To this end, this Special Section on Emerging Novel Enzyme Pathways in Drug Metabolism will highlight a number of advancements that have recently been reported. The included articles support the important role of non-P450 enzymes in the clearance pathways of U.S. Food and Drug Administration-approved drugs over the past 10 years. Specific examples will detail recent reports of aldehyde oxidase, flavin-containing monooxygenase, and other non-P450 pathways that contribute to the metabolic, pharmacokinetic, or pharmacodynamic properties of xenobiotic compounds. Collectively, this series of articles provides additional support for the role of non-P450-mediated metabolic pathways that contribute to the absorption, distribution, metabolism, and excretion properties of current xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| | - Deepak K Dalvie
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| |
Collapse
|
42
|
Zetterberg C, Maltais F, Laitinen L, Liao S, Tsao H, Chakilam A, Hariparsad N. VX-509 (Decernotinib)-Mediated CYP3A Time-Dependent Inhibition: An Aldehyde Oxidase Metabolite as a Perpetrator of Drug-Drug Interactions. Drug Metab Dispos 2016; 44:1286-95. [PMID: 27298338 DOI: 10.1124/dmd.116.071100] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/10/2016] [Indexed: 02/13/2025] Open
Abstract
(R)-2-((2-(1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-4-yl)amino)-2-methyl-N-(2,2,2-trifluoroethyl)butanamide (VX-509, decernotinib) is an oral Janus kinase 3 inhibitor that has been studied in patients with rheumatoid arthritis. Patients with rheumatoid arthritis often receive multiple medications, such as statins and steroids, to manage the signs and symptoms of comorbidities, which increases the chances of drug-drug interactions (DDIs). Mechanism-based inhibition is a subset of time-dependent inhibition (TDI) and occurs when a molecule forms a reactive metabolite which irreversibly binds and inactivates drug-metabolizing enzymes, potentially increasing the systemic load to toxic concentrations. Traditionally, perpetrating compounds are screened using human liver microsomes (HLMs); however, this system may be inadequate when the precipitant is activated by a non-cytochrome P450 (P450)-mediated pathway. Even though studies assessing competitive inhibition and TDI using HLM suggested a low risk for CYP3A4-mediated DDI in the clinic, VX-509 increased the area under the curve of midazolam, atorvastatin, and methyl-prednisolone by approximately 12.0-, 2.7-, and 4.3-fold, respectively. Metabolite identification studies using human liver cytosol indicated that VX-509 is converted to an oxidative metabolite, which is the perpetrator of the DDIs observed in the clinic. As opposed to HLM, hepatocytes contain the full complement of drug-metabolizing enzymes and transporters and can be used to assess TDI arising from non-P450-mediated metabolic pathways. In the current study, we highlight the role of aldehyde oxidase in the formation of the hydroxyl-metabolite of VX-509, which is involved in clinically significant TDI-based DDIs and represents an additional example in which a system-dependent prediction of TDI would be evident.
Collapse
Affiliation(s)
- Craig Zetterberg
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Francois Maltais
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Leena Laitinen
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Shengkai Liao
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Hong Tsao
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Ananthsrinivas Chakilam
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| |
Collapse
|
43
|
Upregulation of the growth arrest-specific-2 in recurrent colorectal cancers, and its susceptibility to chemotherapy in a model cell system. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1345-53. [PMID: 27085973 DOI: 10.1016/j.bbadis.2016.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/04/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is one of the most common life-threatening malignances worldwide. CRC relapse markedly decreases the 5-year survival of patients following surgery. Aberrant expression of genes involved in pathways regulating the cell cycle, cell proliferation, or cell death are frequently reported in CRC tumorigenesis. We hypothesized that genes involved in CRC relapse might serve as prognostic indicators. We first evaluated the significance of gene sequences in the feces of patients with CRC relapse by consulting a public database. Tumorigenesis of target tissues was tested through tumor cell growth, cell cycle regulation, and chemotherapeutic efficacy. We found a highly significant correlation between CRC relapse and growth arrest-specific 2 (GAS2) gene expression. Based on cell models, the overexpressed GAS2 was associated with cellular growth rate, cell cycle regulation, and with chemotherapeutic sensitivity. Cell division was impaired by treating cells with 2-[4-(7-chloro-2-quinoxalinyloxy)phenoxy]-propionic acid (XK469), even when the cells were overexpressing GAS2. Thus, downregulation of GAS2 expression might control CRC relapse after curative resection. GAS2 could serve as a noninvasive marker from the feces of patients with prediagnosed CRC. Our findings suggest that GAS2 could have potential clinical applications for predicting early CRC relapse after radical resection, and that XK469 might impair tumor cell division by reducing GAS2 expression or blocking its cellular translocation. This will help in selecting the best therapeutic option, 5-fluorouracil in combination with XK469, for patients overexpressing GAS2 in CRC cells. Thus, GAS2 might act as a prognostic biomolecule and potential therapeutic target in patients with CRC relapse.
Collapse
|
44
|
Isobe T, Ohta M, Kaneko Y, Kawai H. Species differences in metabolism of ripasudil (K-115) are attributed to aldehyde oxidase. Xenobiotica 2015; 46:579-590. [DOI: 10.3109/00498254.2015.1096981] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
45
|
Sodhi JK, Wong S, Kirkpatrick DS, Liu L, Khojasteh SC, Hop CECA, Barr JT, Jones JP, Halladay JS. A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos 2015; 43:908-15. [PMID: 25845827 PMCID: PMC4429680 DOI: 10.1124/dmd.114.061804] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/06/2015] [Indexed: 12/31/2022] Open
Abstract
GDC-0834, a Bruton's tyrosine kinase inhibitor investigated as a potential treatment of rheumatoid arthritis, was previously reported to be extensively metabolized by amide hydrolysis such that no measurable levels of this compound were detected in human circulation after oral administration. In vitro studies in human liver cytosol determined that GDC-0834 (R)-N-(3-(6-(4-(1,4-dimethyl-3-oxopiperazin-2-yl)phenylamino)-4-methyl-5-oxo- 4,5-dihydropyrazin-2-yl)-2-methylphenyl)-4,5,6,7-tetrahydrobenzo[b] thiophene-2-carboxamide) was rapidly hydrolyzed with a CLint of 0.511 ml/min per milligram of protein. Aldehyde oxidase (AO) and carboxylesterase (CES) were putatively identified as the enzymes responsible after cytosolic fractionation and mass spectrometry-proteomics analysis of the enzymatically active fractions. Results were confirmed by a series of kinetic experiments with inhibitors of AO, CES, and xanthine oxidase (XO), which implicated AO and CES, but not XO, as mediating GDC-0834 amide hydrolysis. Further supporting the interaction between GDC-0834 and AO, GDC-0834 was shown to be a potent reversible inhibitor of six known AO substrates with IC50 values ranging from 0.86 to 1.87 μM. Additionally, in silico modeling studies suggest that GDC-0834 is capable of binding in the active site of AO with the amide bond of GDC-0834 near the molybdenum cofactor (MoCo), orientated in such a way to enable potential nucleophilic attack on the carbonyl of the amide bond by the hydroxyl of MoCo. Together, the in vitro and in silico results suggest the involvement of AO in the amide hydrolysis of GDC-0834.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Susan Wong
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Donald S Kirkpatrick
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Lichuan Liu
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - S Cyrus Khojasteh
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Cornelis E C A Hop
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - John T Barr
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Jeffrey P Jones
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Jason S Halladay
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| |
Collapse
|
46
|
Affiliation(s)
- Deepak Dalvie
- Pfizer Global Research and Development, LaJolla Laboratories San Diego
| | - Michael Zientek
- Pfizer Global Research and Development, LaJolla Laboratories San Diego
| |
Collapse
|
47
|
Sanoh S, Tayama Y, Sugihara K, Kitamura S, Ohta S. Significance of aldehyde oxidase during drug development: Effects on drug metabolism, pharmacokinetics, toxicity, and efficacy. Drug Metab Pharmacokinet 2015; 30:52-63. [DOI: 10.1016/j.dmpk.2014.10.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 12/28/2022]
|
48
|
Zientek MA, Youdim K. Reaction phenotyping: advances in the experimental strategies used to characterize the contribution of drug-metabolizing enzymes. Drug Metab Dispos 2015; 43:163-81. [PMID: 25297949 DOI: 10.1124/dmd.114.058750] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
During the process of drug discovery, the pharmaceutical industry is faced with numerous challenges. One challenge is the successful prediction of the major routes of human clearance of new medications. For compounds cleared by metabolism, accurate predictions help provide an early risk assessment of their potential to exhibit significant interpatient differences in pharmacokinetics via routes of metabolism catalyzed by functionally polymorphic enzymes and/or clinically significant metabolic drug-drug interactions. This review details the most recent and emerging in vitro strategies used by drug metabolism and pharmacokinetic scientists to better determine rates and routes of metabolic clearance and how to translate these parameters to estimate the amount these routes contribute to overall clearance, commonly referred to as fraction metabolized. The enzymes covered in this review include cytochrome P450s together with other enzymatic pathways whose involvement in metabolic clearance has become increasingly important as efforts to mitigate cytochrome P450 clearance are successful. Advances in the prediction of the fraction metabolized include newly developed methods to differentiate CYP3A4 from the polymorphic enzyme CYP3A5, scaling tools for UDP-glucuronosyltranferase, and estimation of fraction metabolized for substrates of aldehyde oxidase.
Collapse
Affiliation(s)
- Michael A Zientek
- Worldwide Research and Development, Pharmacokinetics, Pharmacodynamics, and Metabolism, Pfizer Inc., San Diego, California (M.A.Z.); and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel, Switzerland (K.Y.)
| | - Kuresh Youdim
- Worldwide Research and Development, Pharmacokinetics, Pharmacodynamics, and Metabolism, Pfizer Inc., San Diego, California (M.A.Z.); and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel, Switzerland (K.Y.)
| |
Collapse
|
49
|
Manevski N, Balavenkatraman KK, Bertschi B, Swart P, Walles M, Camenisch G, Schiller H, Kretz O, Ling B, Wettstein R, Schaefer DJ, Pognan F, Wolf A, Litherland K. Aldehyde oxidase activity in fresh human skin. Drug Metab Dispos 2014; 42:2049-57. [PMID: 25249692 DOI: 10.1124/dmd.114.060368] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Human aldehyde oxidase (AO) is a molybdoflavoenzyme that commonly oxidizes azaheterocycles in therapeutic drugs. Although high metabolic clearance by AO resulted in several drug failures, existing in vitro-in vivo correlations are often poor and the extrahepatic role of AO practically unknown. This study investigated enzymatic activity of AO in fresh human skin, the largest organ of the body, frequently exposed to therapeutic drugs and xenobiotics. Fresh, full-thickness human skin was obtained from 13 individual donors and assayed with two specific AO substrates: carbazeran and zoniporide. Human skin explants from all donors metabolized carbazeran to 4-hydroxycarbazeran and zoniporide to 2-oxo-zoniporide. Average rates of carbazeran and zoniporide hydroxylations were 1.301 and 0.164 pmol⋅mg skin(-1)⋅h(-1), resulting in 13 and 2% substrate turnover, respectively, after 24 hours of incubation with 10 μM substrate. Hydroxylation activities for the two substrates were significantly correlated (r(2) = 0.769), with interindividual variability ranging from 3-fold (zoniporide) to 6-fold (carbazeran). Inclusion of hydralazine, an irreversible inhibitor of AO, resulted in concentration-dependent decrease of hydroxylation activities, exceeding 90% inhibition of carbazeran 4-hydroxylation at 100 μM inhibitor. Reaction rates were linear up to 4 hours and well described by Michaelis-Menten enzyme kinetics. Comparison of carbazeran and zoniporide hydroxylation with rates of triclosan glucuronidation and sulfation and p-toluidine N-acetylation showed that cutaneous AO activity is comparable to tested phase II metabolic reactions, indicating a significant role of AO in cutaneous drug metabolism. To our best knowledge, this is the first report of AO enzymatic activity in human skin.
Collapse
Affiliation(s)
- Nenad Manevski
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Kamal Kumar Balavenkatraman
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Barbara Bertschi
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Piet Swart
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Markus Walles
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Gian Camenisch
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Hilmar Schiller
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Olivier Kretz
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Barbara Ling
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Reto Wettstein
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Dirk J Schaefer
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Francois Pognan
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Armin Wolf
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| | - Karine Litherland
- Drug Metabolism and Pharmacokinetics (N.M., P.S., M.W., G.C., H.S., O.K., K.L.) and Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland (B.L., R.W., D.J.S.)
| |
Collapse
|
50
|
Hutzler JM, Cerny MA, Yang YS, Asher C, Wong D, Frederick K, Gilpin K. Cynomolgus monkey as a surrogate for human aldehyde oxidase metabolism of the EGFR inhibitor BIBX1382. Drug Metab Dispos 2014; 42:1751-60. [PMID: 25035284 DOI: 10.1124/dmd.114.059030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
BIBX1382 was an epidermal growth factor receptor inhibitor under clinical investigation for treatment of cancer. This candidate possessed an attractive preclinical absorption, distribution, metabolism, and excretion profile, yet failed in clinical studies due in part to poor oral exposure, resulting from extensive metabolism by aldehyde oxidase (AO). In vitro metabolism studies were performed in liver cytosol and cryopreserved hepatocytes from multiple species. In addition, a pharmacokinetic study was performed in cynomolgus monkey for comparison with the reported human pharmacokinetics of BIBX1382. Estimated hepatic clearance of BIBX1382 in rhesus (42 ml/min per kg) and cynomolgus monkey (43 ml/min per kg) liver cytosol was comparable to human (≥93% of liver blood flow). Metabolite identification after incubation of BIBX1382 in liver cytosol fortified with the AO inhibitor raloxifene confirmed that AO is involved in the formation of the predominant metabolite (BIBU1476, M1) in cynomolgus monkey. After intravenous and oral administration of BIBX1382 to cynomolgus monkeys, high plasma clearance (118 ml/min per kg) and low oral exposure (C(max) = 12.7 nM and 6% oral bioavailability) was observed, with the exposure of M1 exceeding BIBX1382 after oral dosing. This pharmacokinetic profile compared favorably with the human clinical data of BIBX1382 (plasma clearance 25-55 ml/min per kg and 5% oral bioavailability). Thus, it appears that cynomolgus monkey represents a suitable surrogate for the observed human AO metabolism of BIBX1382. To circumvent clinical failures due to uncharacterized metabolism by AO, in vitro studies in the appropriate subcellular fraction, followed by pharmacokinetic and toxicokinetic studies in the appropriately characterized surrogate species should be conducted for substrates of AO.
Collapse
Affiliation(s)
- J Matthew Hutzler
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Matthew A Cerny
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Young-Sun Yang
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Constance Asher
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Diane Wong
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Kosea Frederick
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| | - Kyle Gilpin
- Boehringer-Ingelheim Pharmaceuticals Inc., Medicinal Chemistry, Drug Discovery Support (DMPK), Ridgefield, Connecticut
| |
Collapse
|