1
|
Mu H, Ye L, Wang B. Detailed resume of S-methyltransferases: Categories, structures, biological functions and research advancements in related pathophysiology and pharmacotherapy. Biochem Pharmacol 2024; 226:116361. [PMID: 38876259 DOI: 10.1016/j.bcp.2024.116361] [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: 03/05/2024] [Revised: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Methylation is a vital chemical reaction in the metabolism of many drugs, neurotransmitters, hormones, and exogenous compounds. Among them, S-methylation plays a significant role in the biotransformation of sulfur-containing compounds, particularly chemicals with sulfhydryl groups. Currently, only three S-methyltransferases have been reported: thiopurine methyltransferase (TPMT), thiol methyltransferase (TMT), and thioether methyltransferase (TEMT). These enzymes are involved in various biological processes such as gene regulation, signal transduction, protein repair, tumor progression, and biosynthesis and degradation reactions in animals, plants, and microorganisms. Furthermore, they play pivotal roles in the metabolic pathways of essential drugs and contribute to the advancement of diseases such as tumors. This paper reviews the research progress on relevant structural features, metabolic mechanisms, inhibitor development, and influencing factors (gene polymorphism, S-adenosylmethionine level, race, sex, age, and disease) of S-methyltransferases. We hope that a better comprehension of S-methyltransferases will help to provide a reference for the development of novel strategies for related disorders and improve long-term efficacy.
Collapse
Affiliation(s)
- Hongfei Mu
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Lisha Ye
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Baolian Wang
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
2
|
Izat N, Bolleddula J, Abbasi A, Cheruzel L, Jones RS, Moss D, Ortega-Muro F, Parmentier Y, Peterkin VC, Tian DD, Venkatakrishnan K, Zientek MA, Barber J, Houston JB, Galetin A, Scotcher D. Challenges and Opportunities for In Vitro-In Vivo Extrapolation of Aldehyde Oxidase-Mediated Clearance: Toward a Roadmap for Quantitative Translation. Drug Metab Dispos 2023; 51:1591-1606. [PMID: 37751998 DOI: 10.1124/dmd.123.001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.
Collapse
Affiliation(s)
- Nihan Izat
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jayaprakasam Bolleddula
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Armina Abbasi
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Lionel Cheruzel
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Robert S Jones
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Darren Moss
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Fatima Ortega-Muro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Yannick Parmentier
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Vincent C Peterkin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Dan-Dan Tian
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Karthik Venkatakrishnan
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Michael A Zientek
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| |
Collapse
|
3
|
Russell DA, Chau MK, Shi Y, Levasseur IN, Maldonato BJ, Totah RA. METTL7A (TMT1A) and METTL7B (TMT1B) Are Responsible for Alkyl S-Thiol Methyl Transferase Activity in Liver. Drug Metab Dispos 2023; 51:1024-1034. [PMID: 37137720 PMCID: PMC10353073 DOI: 10.1124/dmd.123.001268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023] Open
Abstract
S-methylation of drugs containing thiol-moieties often alters their activity and results in detoxification. Historically, scientists attributed methylation of exogenous aliphatic and phenolic thiols to a putative S-adenosyl-L-methionine (SAM)-dependent membrane-associated enzyme referred to as thiol methyltransferase (TMT). This putative TMT appeared to have a broad substrate specificity and methylated the thiol metabolite of spironolactone, mertansine, ziprasidone, captopril, and the active metabolites of the thienopyridine prodrugs, clopidogrel, and prasugrel. Despite TMT's role in the S-methylation of clinically relevant drugs, the enzyme(s) responsible for this activity remained unknown. We recently identified methyltransferase-like protein 7B (METTL7B) as an alkyl thiol methyltransferase. METTL7B is an endoplasmic reticulum-associated protein with similar biochemical properties and substrate specificity to the putative TMT. Yet, the historic TMT inhibitor 2,3-dichloro-α-methylbenzylamine (DCMB) did not inhibit METTL7B, indicating that multiple enzymes contribute to TMT activity. Here we report that methyltransferase-like protein 7A (METTL7A), an uncharacterized member of the METTL7 family, is also a SAM-dependent thiol methyltransferase. METTL7A exhibits similar biochemical properties to METTL7B and putative TMT, including inhibition by DCMB (IC50 = 1.17 μM). Applying quantitative proteomics to human liver microsomes and gene modulation experiments in HepG2 and HeLa cells, we determined that TMT activity correlates closely with METTL7A and METTL7B protein levels. Furthermore, purification of a novel His-GST-tagged recombinant protein and subsequent activity experiments prove that METTL7A can selectively methylate exogenous thiol-containing substrates, including 7α-thiospironolactone, dithiothreitol, 4-chlorothiophenol, and mertansine. We conclude that the METTL7 family encodes for two enzymes, METTL7A and METTL7B, which are now renamed thiol methyltransferase 1A (TMT1A) and thiol methyltransferase 1B (TMT1B), respectively, that are responsible for thiol methylation activity in human liver microsomes. SIGNIFICANCE STATEMENT: We identified methyltransferase-like protein 7A (thiol methyltransferase 1A) and methyltransferase-like protein 7B (thiol methyltransferase 1B) as the enzymes responsible for the microsomal alkyl thiol methyltransferase (TMT) activity. These are the first two enzymes directly associated with microsomal TMT activity. S-methylation of commonly prescribed thiol-containing drugs alters their pharmacological activity and/or toxicity, and identifying the enzymes responsible for this activity will improve our understanding of the drug metabolism and pharmacokinetic (DMPK) properties of alkyl- or phenolic thiol-containing therapeutics.
Collapse
Affiliation(s)
- Drake A Russell
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| | - Marvin K Chau
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| | - Yuanyuan Shi
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| | - Ian N Levasseur
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| | - Benjamin J Maldonato
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| | - Rheem A Totah
- University of Washington, Department of Medicinal Chemistry, Seattle, Washington
| |
Collapse
|
4
|
Ma S, Cho S, Sahasranaman S, Zhao W, Pang J, Ding X, Dean B, Wang B, Hsu JY, Ware J, Salphati L. Absorption, Metabolism, and Excretion of Taselisib (GDC-0032), a Potent β-Sparing PI3K Inhibitor in Rats, Dogs, and Humans. Drug Metab Dispos 2023; 51:436-450. [PMID: 36623882 DOI: 10.1124/dmd.122.001096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Taselisib (also known as GDC-0032) is a potent and selective phosphoinositide 3-kinase (PI3K) inhibitor that displays greater selectivity for mutant PI3Kα than wild-type PI3Kα To better understand the absorption, distribution, metabolism, and excretion properties of taselisib, mass balance studies were conducted following single oral doses of [14C]taselisib in rats, dogs, and humans. Absolute bioavailability (ABA) of taselisib in humans was determined by oral administration of taselisib at the therapeutic dose followed by intravenous dosing of [14C]taselisib as a microtracer. The ABA in humans was 57.4%. Absorption of taselisib was rapid in rats and dogs and moderately slow in humans. The recovery of radioactivity in excreta was high (>96%) in the three species where feces was the major route of excretion. Taselisib was the major circulating component in the three species with no metabolite accounting for >10% of the total drug-derived material. The fraction absorbed of taselisib was 35.9% in rats and 71.4% in dogs. In rats, absorbed drug underwent moderate to extensive metabolism and biliary excretion of taselisib was minor. In dog, biliary excretion and metabolism were major clearance pathways. In humans, 84.2% of the dose was recovered as the parent drug in excreta indicating that metabolism played a minor role in the drug's clearance. Major metabolism pathways were oxidation and amide hydrolysis in the three species while methylation was another prominent metabolism pathway in dogs. The site of methylation was identified on the triazole moiety. In vitro experiments characterized that the N-methylation was dog-specific and likely mediated by a thiol methyltransferase. SIGNIFICANCE STATEMENT: This study provides a comprehensive description of the absorption, distribution, and metabolism and pharmacokinetic properties of taselisib in preclinical species and humans. This study demonstrated the importance of oral bioavailability results for understanding taselisib's clearance pathways. The study also describes the identification and characterization of a unique dog-specific N-methylation metabolite of taselisib and the enzyme mediating N-methylation in vitro.
Collapse
Affiliation(s)
- Shuguang Ma
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Srikumar Sahasranaman
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Weiping Zhao
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Jodie Pang
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Xiao Ding
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Brian Dean
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Bin Wang
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Jerry Y Hsu
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Joseph Ware
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| | - Laurent Salphati
- Department of Drug Metabolism and Pharmacokinetics (S.M., S.C., W.Z., J.P., X.D., B.D., L.S.) and Department of Clinical Pharmacology (S.S., J.Y.H., J.W.), Genentech, Inc., South San Francisco, California; and XenoBiotic Laboratories (B.W.), Inc., Plainsboro, New Jersey
| |
Collapse
|
5
|
Tian Q, Jiang J, Yin H, Zhang Y, Li Y, Wu P, Peng C, Wang Z, Zhou J, Zeng H, Zhong D. Investigating the Metabolic Mechanisms of Butaselen, An Ebselen Analog. Curr Drug Metab 2022; 23:928-939. [PMID: 35619304 DOI: 10.2174/1389200223666220520115014] [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: 01/21/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Butaselen is an ebselen analog that is under clinical trials for treating hepatic and pulmonary fibrosis. Our previous studies showed that butaselen is mainly present in human plasma in the form of M2, a free Se-methylated metabolite. OBJECTIVE This study aimed to investigate the metabolic mechanisms of butaselen. METHODS AND RESULTS Butaselen was incubated with human plasma. Butaselen immediately disappeared, and the butaselen-HSA (human serum albumin) adduct was detected by HPLC-HRMS, showing that butaselen covalently binds to HSA. The butaselen-HSA adduct was precipitated using acetonitrile and then incubated with PBS, Cys, and GSH for 1 hour. The product was M1, a reduced form of butaselen. The results indicated that HSA, Cys, and GSH can reduce the butaselen-HSA covalent bond. The binding site for butaselen could be the cysteine-34 residue of HSA through pronase and trypsin hydrolysis. Incubating butaselen with cysteine, butaselen-Cys, butaselen-2Cys, and M1 were generated, indicating the covalent binding and reduction of butaselen by cysteine. We incubated liver microsomes and cytosol with butaselen, 6.22 and 246 nM M2 were generated, respectively. The results demonstrated that cytosolic enzymes are mainly involved in M2 production. The amount of M2 in the liver cytosol decreased from 246 nM to 2.21 nM when 10 mM m-anisic acid (a specific TPMT enzyme inhibitor) was added, showing that TPMT is responsible for M2 formation. CONCLUSION Butaselen was covalently bound to HSA, and the binding site was the cysteine-34 residue of HSA. The butaselen-HSA adduct was reduced by free thiol compounds to generate M1. M1 was further metabolized to M2 by cytosolic TPMT. This study provides a basis for studying the pharmacokinetics of selenium-containing drugs.
Collapse
Affiliation(s)
- Qianqian Tian
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Hanwei Yin
- Shanghai Yuanxi Medicine Corp, Shanghai 201203, China
| | - Yifan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yilin Li
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Zhijie Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Jialan Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Huihui Zeng
- Shanghai Yuanxi Medicine Corp, Shanghai 201203, China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Maldonato BJ, Russell DA, Totah RA. Human METTL7B is an alkyl thiol methyltransferase that metabolizes hydrogen sulfide and captopril. Sci Rep 2021; 11:4857. [PMID: 33649426 PMCID: PMC7921093 DOI: 10.1038/s41598-021-84218-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/12/2021] [Indexed: 11/15/2022] Open
Abstract
Methylation of alkyl thiols is a biotransformation pathway designed to reduce thiol reactivity and potential toxicity, yet the gene and protein responsible for human alkyl thiol methyltransferase (TMT) activity remain unknown. Here we demonstrate with a range of experimental approaches using cell lines, in vitro systems, and recombinantly expressed enzyme, that human methyltransferase-like protein 7B (METTL7B) catalyzes the transfer of a methyl group from S-adenosyl-L-methionine (AdoMet) to hydrogen sulfide (H2S) and other exogenous thiol small molecules. METTL7B gene modulation experiments, including knockdown in HepG2 cells and overexpression in HeLa cells, directly alter the methylation of the drug captopril, a historic probe substrate for TMT activity. Furthermore, recombinantly expressed and purified wild-type METTL7B methylates several thiol compounds, including H2S, 7α-thiospironolactone, L-penicillamine, and captopril, in a time- and concentration-dependent manner. Typical for AdoMet-dependent small molecule methyltransferases, S-adenosyl-L-homocysteine (AdoHcy) inhibited METTL7B activity in a competitive fashion. Similarly, mutating a conserved aspartate residue, proposed to anchor AdoMet into the active site, to an alanine (D98A) abolished methylation activity. Endogenous thiols such as glutathione and cysteine, or classic substrates for other known small molecule S-, N-, and O-methyltransferases, were not substrates for METTL7B. Our results confirm, for the first time, that METTL7B, a gene implicated in multiple disease states including rheumatoid arthritis and breast cancer, encodes a protein that methylates small molecule alkyl thiols. Identifying the catalytic function of METTL7B will enable future pharmacological research in disease pathophysiology where altered METTL7B expression and, potentially H2S levels, can disrupt cell growth and redox state.
Collapse
Affiliation(s)
- Benjamin J Maldonato
- Department of Medicinal Chemistry, University of Washington, 1959 NE Pacific Ave, Box 357610, Seattle, WA, 98195, USA
| | - Drake A Russell
- Department of Medicinal Chemistry, University of Washington, 1959 NE Pacific Ave, Box 357610, Seattle, WA, 98195, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, 1959 NE Pacific Ave, Box 357610, Seattle, WA, 98195, USA.
| |
Collapse
|
9
|
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. [DOI: 10.1124/dmd.120.000082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022] Open
|
10
|
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
|
11
|
Maw HH, Zeng X, Campbell S, Taub ME, Teitelbaum AM. N-Methylation of BI 187004 by Thiol S-Methyltransferase. Drug Metab Dispos 2018. [PMID: 29514826 DOI: 10.1124/dmd.117.079764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BI 187004, an 11β-hydroxysteroid dehydrogenase 1 inhibitor, was administered once daily for 14 days to eight patients with type 2 diabetes mellitus. N-methylation was identified as a major biotransformation pathway. In four patients treated with BI 187004, the plasma exposure of an N-methylbenzimidazole metabolite [N-methylbenzimidazole regioisomer 1 (M1)] was 7-fold higher than the remaining four patients, indicating a substantial degree of metabolic variation. To identify the methyltransferase enzymes responsible for N-methylation, BI 187004 was incubated with human liver microsomes (HLM), human kidney microsomes (HKM), and their respective cytosolic preparations in the presence and absence of isoform-selective chemical inhibitors. Additionally, BI 187004 was incubated with several human recombinant methyltransferases: catechol O-methyltransferase (rhCOMT), histamine N-methyltransferase (rhHNMT), nicotinamide N-methyltransferase (rhNNMT), glycine N-methyltransferase (rhGNMT), and thiopurine S-methyltransferase (rhTPMT). M1 was principally observed in HLM and HKM incubations, minimally formed in liver and kidney cytosol, and not formed during incubations with recombinant methyltransferase enzymes. In all microsomal and cytosolic incubations, the formation of M1 was inhibited only by 2,3-dichloro-α-methylbenzylamine (DCMB), an inhibitor of thiol S-methyltransferase (TMT), providing evidence that TMT catalyzed the formation of M1. Interestingly, the N-methylbenzimidazole regioisomer (M14) was only observed in vitro, predominantly during incubations with human kidney cytosol and rhHNMT. The formation of M14 was inhibited by amodiaquine (an HNMT inhibitor) and DCMB, providing additional evidence that both HNMT and TMT catalyzed M14 formation. Overall, using BI 187004 as a substrate, this study demonstrates a novel TMT-mediated N-methylation biotransformation and an HNMT-mediated regioselective N-methylation.
Collapse
Affiliation(s)
- Hlaing H Maw
- Drug Metabolism and Pharmacokinetics (H.H.M., M.E.T., A.M.T.), Chemical Development (X.Z.), and Material and Analytical Sciences (S.C.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Xingzhong Zeng
- Drug Metabolism and Pharmacokinetics (H.H.M., M.E.T., A.M.T.), Chemical Development (X.Z.), and Material and Analytical Sciences (S.C.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Scot Campbell
- Drug Metabolism and Pharmacokinetics (H.H.M., M.E.T., A.M.T.), Chemical Development (X.Z.), and Material and Analytical Sciences (S.C.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Mitchell E Taub
- Drug Metabolism and Pharmacokinetics (H.H.M., M.E.T., A.M.T.), Chemical Development (X.Z.), and Material and Analytical Sciences (S.C.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Aaron M Teitelbaum
- Drug Metabolism and Pharmacokinetics (H.H.M., M.E.T., A.M.T.), Chemical Development (X.Z.), and Material and Analytical Sciences (S.C.), Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
12
|
Lv D, Zhao M, Chen L, Yu D, Yun X, Yang Q, Huang X. An Inter-Ethnic Comparison Study of Ziprasidone Plasma Levels, Dosage and Clinical Response in Patients with Schizophrenia. Psychiatry Investig 2017; 14:360-367. [PMID: 28539955 PMCID: PMC5440439 DOI: 10.4306/pi.2017.14.3.360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 04/14/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate ziprasidone plasma concentration, daily dose and clinical efficacy and safety in Han Chinese and Mongolian patients with first-episode schizophrenia. METHODS A total of 123 inpatients affected by schizophrenia were recruited from the Mental Health Center of Inner Mongolia in China. Ziprasidone plasma concentration, clinical efficacy and side effects were systematically evaluated at baseline, and at 1, 2, 4, and 6 weeks. Metabolic measures such as changes in weight, body mass index (BMI), fasting blood glucose (FBG), triglycerides, and cholesterol, were also recorded. RESULTS 90 patients completed the study. Compared with Han patients, on average, Mongolian patients received a significantly higher ziprasidone dosage for adequate symptom control during the 6-week period and had a lower plasma concentration-to-dose ratio. The Mongolian patients also experienced greater increases in weight and BMI. No significant differences between the two ethnic groups were found in the rate of reduction in the Positive and Negative Syndrome Scale (PANSS) score, Treatment Emergent Symptom Scale (TESS) total score, FBG, triglycerides, cholesterol or Q-Tc interval. CONCLUSION Compared to Han Chinese patients, Mongolian patients appeared to have increased ziprasidone clearance and require higher doses to achieve effective treatment for schizophrenia.
Collapse
Affiliation(s)
- Dongsheng Lv
- Department of Psychiatry, Mental Health Center of Inner Mongolia, Inner Mongolia, Hohhot, China
- Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, China
| | - Meirong Zhao
- Department of Psychiatry, Mental Health Center of Inner Mongolia, Inner Mongolia, Hohhot, China
- Department of Psychiatry, the First Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, Hohhot, China
| | - Lixia Chen
- Department of Psychiatry, Mental Health Center of Inner Mongolia, Inner Mongolia, Hohhot, China
| | - Dongsheng Yu
- Department of Psychiatry, Mental Health Center of Inner Mongolia, Inner Mongolia, Hohhot, China
| | - Xiaobin Yun
- Department of Psychiatry, Mental Health Center of Inner Mongolia, Inner Mongolia, Hohhot, China
| | - Qing Yang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Xiaojun Huang
- Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
13
|
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: 65] [Impact Index Per Article: 7.2] [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
|
14
|
Structure and function of mammalian aldehyde oxidases. Arch Toxicol 2016; 90:753-80. [DOI: 10.1007/s00204-016-1683-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/16/2016] [Indexed: 12/12/2022]
|
15
|
Mandrioli R, Protti M, Mercolini L. Evaluation of the pharmacokinetics, safety and clinical efficacy of ziprasidone for the treatment of schizophrenia and bipolar disorder. Expert Opin Drug Metab Toxicol 2014; 11:149-74. [PMID: 25483358 DOI: 10.1517/17425255.2015.991713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Multiple strategies exist for the pharmacological treatment of schizophrenia and related disorders. In the last 20 years, several 'new' compounds have been introduced, called 'atypical antipsychotics', which have higher efficacy and better tolerability than first-generation neuroleptics. Among them, ziprasidone (ZPR) is currently finding widespread use, and it has also been shown to be active as an augmenter in bipolar disorder therapy. AREAS COVERED This review aims to provide the latest information on ZPR, an 'atypical' agent for the pharmacological therapy of schizophrenia and bipolar disorder. A literature search has been carried out with the keywords 'ziprasidone', 'schizophrenia', 'psychosis', 'bipolar', 'pharmacokinetics' and 'clinical trials'. In this process, particular attention has been paid to the drug pharmacokinetic characteristics and its safety in clinical use. EXPERT OPINION ZPR shares most advantages and disadvantages with other atypical antipsychotics. However, it can be useful for its low tendency to cause metabolic syndrome and hyperprolactinaemia, especially in patients suffering from excess weight, hyperlipidaemia, diabetes or who have suffered from hyperprolactinaemia when using other antipsychotics. However, there are serious doubts as to whether ZPR should be administered to patients suffering from arrhythmias or QTc prolongation, and even more for administration to bipolar patients undergoing polypharmacy with antidepressants.
Collapse
Affiliation(s)
- Roberto Mandrioli
- Alma Mater Studiorum - University of Bologna, Department for Life Quality Studies (QuVi) , Corso d'Augusto 237, 47921 Rimini , Italy +39 0541 434624 ; +39 0541 434608 ;
| | | | | |
Collapse
|
16
|
Kazui M, Hagihara K, Izumi T, Ikeda T, Kurihara A. Hepatic microsomal thiol methyltransferase is involved in stereoselective methylation of pharmacologically active metabolite of prasugrel. Drug Metab Dispos 2014; 42:1138-45. [PMID: 24733788 DOI: 10.1124/dmd.114.057661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Prasugrel, a thienopyridine antiplatelet drug, is converted in animals and humans to the pharmacologically active metabolite R-138727 [(2Z)-{1-[(1RS)-2-cyclopropyl-1-(2-fluorophenyl)-2-oxoethyl]-4-sulfanylpiperidin-3-ylidene}ethanoic acid], which has two chiral centers, occurring as a mixture of four isomers. The RS and RR isomers are more active than the SS and SR isomers (RS > RR > > SR = SS). The pharmacologically active metabolite is further metabolized to an S-methylated metabolite that is the major identified inactive metabolite in humans. In rat, dog, and human liver microsomes supplemented with S-adenosyl methione, the SS and SR isomers of the active metabolite were extensively S-methylated while the RS and RR isomers were not. Addition of 2,3-dichloromethyl benzylamine (50 µM) completely inhibited the S-methylation reaction, indicating that the microsomal and cytosolic thiol methyltransferase but not the cytosolic thiopurine S-methyltransferase is involved in the methylation. The hepatic intrinsic clearance values for methylation of the RS, RR, SS, and SR isomers (ml/min/kg) were 0, 0, 40.4, and 37.6, respectively, in rat liver microsomes, 0, 0, 11.6, and 2.5, respectively, in dog liver microsomes, and 0, 0, 17.3, and 17.7, respectively, in human liver microsomes, indicating that the RS and RR isomers are not methylated in vitro and that the methylation of SS and SR isomers is high with rat > human > dog. This finding in vitro agreed well with the in vivo observation in rats and dogs, where the S-methylated SS and SR isomers were the major metabolites in the plasma whereas negligible amounts of S-methylated RS and RR isomers were detected after intravenous administration of the pharmacologically active metabolites.
Collapse
Affiliation(s)
- Miho Kazui
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo (M.K., K.H., T.Iz., A.K.); Yokohama College of Pharmacy, Yokohama (T.Ik.), Japan
| | - Katsunobu Hagihara
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo (M.K., K.H., T.Iz., A.K.); Yokohama College of Pharmacy, Yokohama (T.Ik.), Japan
| | - Takashi Izumi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo (M.K., K.H., T.Iz., A.K.); Yokohama College of Pharmacy, Yokohama (T.Ik.), Japan
| | - Toshihiko Ikeda
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo (M.K., K.H., T.Iz., A.K.); Yokohama College of Pharmacy, Yokohama (T.Ik.), Japan
| | - Atsushi Kurihara
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo (M.K., K.H., T.Iz., A.K.); Yokohama College of Pharmacy, Yokohama (T.Ik.), Japan
| |
Collapse
|