1
|
Li W, Vazvaei-Smith F, Dear G, Boer J, Cuyckens F, Fraier D, Liang Y, Lu D, Mangus H, Moliner P, Pedersen ML, Romeo AA, Spracklin DK, Wagner DS, Winter S, Xu XS. Metabolite Bioanalysis in Drug Development: Recommendations from the IQ Consortium Metabolite Bioanalysis Working Group. Clin Pharmacol Ther 2024; 115:939-953. [PMID: 38073140 DOI: 10.1002/cpt.3144] [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: 09/19/2023] [Accepted: 12/05/2023] [Indexed: 03/13/2024]
Abstract
The intent of this perspective is to share the recommendations of the International Consortium for Innovation and Quality in Pharmaceutical Development Metabolite Bioanalysis Working Group on the fit-for-purpose metabolite bioanalysis in support of drug development and registration. This report summarizes the considerations for the trigger, timing, and rigor of bioanalysis in the various assessments to address unique challenges due to metabolites, with respect to efficacy and safety, which may arise during drug development from investigational new drug (IND) enabling studies, and phase I, phase II, and phase III clinical trials to regulatory submission. The recommended approaches ensure that important drug metabolites are identified in a timely manner and properly characterized for efficient drug development.
Collapse
Affiliation(s)
- Wenkui Li
- Pharmacokinetic Sciences, Novartis Biomedical Research, East Hanover, New Jersey, USA
| | - Faye Vazvaei-Smith
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Gordon Dear
- Drug Metabolism and Pharmacokinetics, GSK, Ware, UK
| | - Jason Boer
- Drug Metabolism and Pharmacokinetics, Incyte Corporation, Wilmington, Delaware, USA
| | - Filip Cuyckens
- Drug Metabolism and Pharmacokinetics, Janssen R & D, Beerse, Belgium
| | - Daniela Fraier
- Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yuexia Liang
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Ding Lu
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts, USA
| | - Heidi Mangus
- Drug Metabolism and Pharmacokinetics, Agios Pharmaceuticals Inc., Cambridge, Massachusetts, USA
| | - Patricia Moliner
- Enzymology and Metabolism, Department of Translational Medicine and Early Development, Sanofi, Montpellier, Occitanie, France
| | - Mette Lund Pedersen
- DMPK, Research and Early Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrea A Romeo
- Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Douglas K Spracklin
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut, USA
| | - David S Wagner
- Drug Metabolism and Disposition, AbbVie, North Chicago, Illinois, USA
| | - Serge Winter
- Pharmacokinetic Sciences, Novartis Biomedical Research, Basel, Switzerland
| | - Xiaohui Sophia Xu
- Clinical Bioanalysis, Translation Medicine, Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| |
Collapse
|
2
|
Cuyckens F, Hvenegaard MG, Cassidy KC, Spracklin DK, James AD, Pedersen ML, Scarfe G, Wagner DS, Georgi K, Schulz SI, Schieferstein H, Bjornsdottir I, Romeo AA, Da Violante G, Blech S, Moliner P, Young GC. Recommendations on the Use of Multiple Labels in Human Mass Balance Studies. Drug Metab Dispos 2024; 52:153-158. [PMID: 38216306 DOI: 10.1124/dmd.123.001429] [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: 09/18/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 01/14/2024] Open
Abstract
The administration of radiolabeled drug candidates is considered the gold standard in absorption, distribution, metabolism, and excretion studies for small-molecule drugs since it allows facile and accurate quantification of parent drug, metabolites, and total drug-related material independent of the compound structure. The choice of the position of the radiolabel, typically 14C or 3H, is critical to obtain relevant information. Sometimes, a biotransformation reaction may lead to cleavage of a part of the molecule. As a result, only the radiolabeled portion can be followed, and information on the fate of the nonlabeled metabolite may be lost. Synthesis and administration of two or more radiolabeled versions of the parent drug as a mixture or in separate studies may resolve this issue but comes with additional challenges. In this paper, we address the questions that may be considered to help make the right choice whether to use a single or multiple radiolabel approach and discuss the pros and cons of different multiple-labeling strategies that can be taken as well as alternative methods that allow the nonlabeled part of the molecule to be followed. SIGNIFICANCE STATEMENT: Radiolabeled studies are the gold standard in drug metabolism research, but molecules can undergo cleavage with loss of the label. This often results in discussions around potential use of multiple labels, which seem to be occurring with increased frequency since an increasing proportion of the small-molecule drugs are tending towards larger molecular weights. This review provides insight and decision criteria in considering a multiple-label approach as well as pros and cons of different strategies that can be followed.
Collapse
Affiliation(s)
- Filip Cuyckens
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Mette G Hvenegaard
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Kenneth C Cassidy
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Douglas K Spracklin
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Alexander D James
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Mette L Pedersen
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Graeme Scarfe
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - David S Wagner
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Katrin Georgi
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Simone I Schulz
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Hanno Schieferstein
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Inga Bjornsdottir
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Andrea A Romeo
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Georges Da Violante
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Stefan Blech
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Patricia Moliner
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| | - Graeme C Young
- Janssen R&D, Beerse, Belgium (F.C.); H. Lundbeck A/S, Copenhagen, Denmark (M.G.H.); Eli Lilly and Company, Indianapolis, Indiana (K.C.C.); Pfizer Inc., Groton, Connecticut (D.K.S.); Novartis, Basel, Switzerland (A.D.J.); Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (M.L.P.); Sosei Heptares, Cambridge, United Kingdom (G.S.); AbbVie, North Chicago, Illinois (D.S.W.); Bayer AG, Wuppertal, Germany (K.G., S.I.S.); The Healthcare Business of Merck KGaA, Darmstadt, Germany (H.S.); Novo Nordisk, Maaloev, Denmark (I.B.); Roche Pharma Research and Early Development, Basel, Switzerland (A.A.R.); Servier, Gif-sur-Yvette, France (G.Da.V.); Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany (S.B.); Sanofi, Montpellier, France (P.M.); and GSK Research & Development Ltd., Stevenage (G.C.Y.)
| |
Collapse
|
3
|
Alanazi IM, R Alzahrani A, Zughaibi TA, Al-Asmari AI, Tabrez S, Henderson C, Watson D, Grant MH. Metabolomics Analysis as a Tool to Measure Cobalt Neurotoxicity: An In Vitro Validation. Metabolites 2023; 13:698. [PMID: 37367855 DOI: 10.3390/metabo13060698] [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: 03/14/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
In this study, cobalt neurotoxicity was investigated in human astrocytoma and neuroblastoma (SH-SY5Y) cells using proliferation assays coupled with LC-MS-based metabolomics and transcriptomics techniques. Cells were treated with a range of cobalt concentrations between 0 and 200 µM. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay revealed cobalt cytotoxicity and decreased cell metabolism in a dose and time-dependent manner was observed by metabolomics analysis, in both cell lines. Metabolomic analysis also revealed several altered metabolites particularly those related to DNA deamination and methylation pathways. One of the increased metabolites was uracil which can be generated from DNA deamination or fragmentation of RNA. To investigate the origin of uracil, genomic DNA was isolated and analyzed by LC-MS. Interestingly, the source of uracil, which is uridine, increased significantly in the DNA of both cell lines. Additionally, the results of the qRT-PCR showed an increase in the expression of five genes Mlh1, Sirt2, MeCP2, UNG, and TDG in both cell lines. These genes are related to DNA strand breakage, hypoxia, methylation, and base excision repair. Overall, metabolomic analysis helped reveal the changes induced by cobalt in human neuronal-derived cell lines. These findings could unravel the effect of cobalt on the human brain.
Collapse
Affiliation(s)
- Ibrahim M Alanazi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, Makkah 21955, Saudi Arabia
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, Makkah 21955, Saudi Arabia
| | - Torki A Zughaibi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed I Al-Asmari
- Laboratory Department, King Abdul-Aziz Hospital, Ministry of Health, Jeddah 22421, Saudi Arabia
- Toxicology and Forensic Science Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Catherine Henderson
- Department of Biomedical Engineering, University of Strathclyde, Glasgow G4 0NW, UK
| | - David Watson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Mary Helen Grant
- Department of Biomedical Engineering, University of Strathclyde, Glasgow G4 0NW, UK
| |
Collapse
|
4
|
Li W, Picard F. Toxicokinetics in preclinical drug development of small-molecule new chemical entities. Biomed Chromatogr 2022:e5553. [PMID: 36415962 DOI: 10.1002/bmc.5553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Toxicokinetics (TK) is an integral part of nonclinical (preclinical) safety assessment of small-molecule new chemical entities in drug development. It is employed to describe the systemic exposure of a drug candidate and/or its important metabolite(s) achieved in study animals and elucidate the relationship (proportional, over-proportional, or under-proportional) between systemic exposure and dose administered and the associated differences/similarities between male and female animals along with the possible accumulation/induction. TK data and the derived parameters are employed to propose safe starting doses for clinical use of the new drug candidate through proper extrapolation of findings in study animals to humans. This review has attempted to highlight the health authority expectations on TK assessment in supporting preclinical safety profiling of new chemical entities. A robust TK assessment requires good understanding of absorption, distribution, metabolism, and elimination processes of drug candidate, adequate TK sampling (e.g., controls where relevant), implementation of fit-for-purpose bioanalytical methods (validated or scientifically qualified) along with necessary measures to prevent mis-dosing or ex vivo contamination, and establishment of stability of the drug candidate and/or its metabolite(s) in the intended species matrix to ensure the reliability of bioanalytical and TK data. The latter provides a vital link between animal experiments and human safety.
Collapse
Affiliation(s)
- Wenkui Li
- Pharmacokinetic Sciences-Drug Disposition, Novartis Institutes for BioMedical Research, East Hanover, New Jersey, USA
| | - Franck Picard
- Pharmacokinetic Sciences-Drug Disposition, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
5
|
Bauman JN, Doran AC, King-Ahmad A, Sharma R, Walker GS, Lin J, Lin TH, Telliez JB, Tripathy S, Goosen TC, Banfield C, Malhotra BK, Dowty ME. The Pharmacokinetics, Metabolism, and Clearance Mechanisms of Abrocitinib, a Selective Janus Kinase Inhibitor, in Humans. Drug Metab Dispos 2022; 50:1106-1118. [PMID: 35701182 DOI: 10.1124/dmd.122.000829] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022] Open
Abstract
Abrocitinib is an oral once-daily Janus kinase 1 selective inhibitor being developed for the treatment of moderate-to-severe atopic dermatitis. This study examined the disposition of abrocitinib in male participants following oral and intravenous administration using accelerator mass spectroscopy methodology to estimate pharmacokinetic parameters and characterize metabolite profiles. The results indicated abrocitinib had a systemic clearance of 64.2 L/h, a steady state volume of distribution of 100 L, extent of absorption >90%, time to maximum plasma concentration of ≈0.5 hour, and absolute oral bioavailability of 60%. The half-life of both abrocitinib and total radioactivity was similar with no indication of metabolite accumulation. Abrocitinib was the main circulating drug species in plasma (≈26%) with 3 major mono-hydroxylated metabolites (M1, M2, and M4) at >10%. Oxidative metabolism was the primary route of elimination for abrocitinib with the greatest disposition of radioactivity shown in the urine (≈85%). In vitro phenotyping indicated abrocitinib cytochrome P450 fraction of metabolism assignments of 0.53 for CYP2C19, 0.30 for CYP2C9, 0.11 for CYP3A4, and ≈0.06 for CYP2B6. The principal systemic metabolites M1, M2, and M4 were primarily cleared renally. Abrocitinib, M1, and M2 showed pharmacology with similar Janus kinase 1 selectivity, whereas M4 was inactive. Significance Statement This study provides a detailed understanding of the disposition and metabolism of abrocitinib, a JAK inhibitor for atopic dermatitis, in humans, as well as characterization of clearance pathways and pharmacokinetics of abrocitinib and its metabolites.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tsung H Lin
- Inflammation and Immunology, Pfizer Inc, United States
| | | | | | - Theunis C Goosen
- Pharmacokinetics, Dynamics & Metabolism, Pfizer, Inc, United States
| | | | | | | |
Collapse
|
6
|
Wang X, Dowty ME, Wouters A, Tatulych S, Connell CA, Le VH, Tripathy S, O'Gorman MT, Winton JA, Yin N, Valdez H, Malhotra BK. Assessment of the Effects of Inhibition or Induction of CYP2C19 and CYP2C9 Enzymes, or Inhibition of OAT3, on the Pharmacokinetics of Abrocitinib and Its Metabolites in Healthy Individuals. Eur J Drug Metab Pharmacokinet 2022; 47:419-429. [PMID: 35226304 PMCID: PMC9050788 DOI: 10.1007/s13318-021-00745-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/11/2022]
Abstract
Background and Objective Abrocitinib is a Janus kinase 1-selective inhibitor for the treatment of moderate-to-severe atopic dermatitis. Abrocitinib is eliminated primarily by metabolism involving cytochrome P450 (CYP) enzymes. Abrocitinib pharmacologic activity is attributable to the unbound concentrations of the parent molecule and 2 active metabolites, which are substrates of organic anion transporter 3 (OAT3). The sum of potency-adjusted unbound exposures of abrocitinib and its 2 active metabolites is termed the abrocitinib active moiety. We evaluated effects of CYP inhibition, CYP induction, and OAT3 inhibition on the pharmacokinetics of abrocitinib, its metabolites, and active moiety. Methods Three fixed-sequence, open-label, phase I studies in healthy adult volunteers examined the drug–drug interactions (DDIs) of oral abrocitinib with fluvoxamine and fluconazole, rifampin, and probenecid. Results Co-administration of abrocitinib with fluvoxamine or fluconazole increased the area under the plasma concentration–time curve from time 0 to infinity (AUCinf) of the unbound active moiety of abrocitinib by 91% and 155%, respectively. Co-administration with rifampin decreased the unbound active moiety AUCinf by 56%. The OAT3 inhibitor probenecid increased the AUCinf of the unbound active moiety by 66%. Conclusions It is important to consider the effects of DDIs on the abrocitinib active moiety when making dosing recommendations. Co-administration of strong CYP2C19/2C9 inhibitors or CYP inducers impacted exposure to the abrocitinib active moiety. A dose reduction by half is recommended if abrocitinib is co-administered with strong CYP2C19 inhibitors, whereas co-administration with strong CYP2C19/2C9 inducers is not recommended. No dose adjustment is required when abrocitinib is administered with OAT3 inhibitors. Clinical Trials Registration IDs NCT03634345, NCT03637790, NCT03937258 Supplementary Information The online version contains supplementary material available at 10.1007/s13318-021-00745-6.
Collapse
Affiliation(s)
| | | | | | | | | | - Vu H Le
- Pfizer Inc., New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Quantification of abemaciclib and metabolites: evolution of bioanalytical methods supporting a novel oncolytic agent. Bioanalysis 2021; 13:711-724. [PMID: 33870730 DOI: 10.4155/bio-2021-0039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Bioanalytical methods undergo many revisions and modifications throughout drug development to meet the objectives of the study and development program. Results: Validated LC-MS/MS methodology used to quantify abemaciclib and four metabolites in human plasma is described. The method, initially validated to support the first-in-human study, was successfully modified to include additional metabolites as in vitro and in vivo information about the activity and abundance of human metabolites became available. Consistent performance of the method over time was demonstrated by an incurred sample reanalysis passing rate exceeding 95%, across clinical studies. An overview of the numerous methods involved during the development of abemaciclib, including the quantification of drugs evaluated as combination regimens and used as substrates during drug-drug interaction studies, is presented. Conclusion: Robust bioanalytical methods need to be designed with the flexibility required to support the evolving study objectives associated with registration and post-registration trials.
Collapse
|
8
|
Wang EQ, Le V, O'Gorman M, Tripathy S, Dowty ME, Wang L, Malhotra BK. Effects of Hepatic Impairment on the Pharmacokinetics of Abrocitinib and Its Metabolites. J Clin Pharmacol 2021; 61:1311-1323. [PMID: 33749838 PMCID: PMC8518898 DOI: 10.1002/jcph.1858] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/18/2021] [Indexed: 11/07/2022]
Abstract
Abrocitinib, an oral once-daily Janus kinase 1 selective inhibitor, is under development for treatment of atopic dermatitis. This phase 1, nonrandomized, open-label, single-dose study (NCT03626415) investigated the effect of hepatic impairment on pharmacokinetics (PK), safety, and tolerability of abrocitinib and its metabolites after a 200-mg oral dose. Twenty-four subjects with varying degrees of hepatic function (normal, mild, and moderate impairment) were enrolled (N = 8/group). Active moiety PK parameters were calculated as the sum of unbound PK parameters for abrocitinib and its active metabolites. For abrocitinib, the ratios (percentages) of adjusted geometric means for area under the concentration-time curve from time 0 extrapolated to infinite time (AUCinf ) and maximum plasma concentration (Cmax ) were 133.33 (90% confidence interval [CI], 86.17-206.28) and 94.40 (90%CI, 62.96-141.55), respectively, for subjects with mild hepatic impairment vs normal hepatic function. The corresponding comparisons of ratios (percentages) for AUCinf and Cmax were 153.99 (90%CI, 99.52-238.25) and 105.53 (90%CI, 70.38-158.24), respectively, for subjects with moderate hepatic impairment. Exposures of the metabolites were generally lower in subjects with hepatic impairment. For abrocitinib active moiety, the ratios (percentages) of adjusted geometric means of unbound AUCinf were 95.74 (90%CI, 72.71-126.08) and 114.82 (90%CI, 87.19-151.20) in subjects with mild and moderate impairment vs normal hepatic function, respectively. Abrocitinib was generally safe and well tolerated. Hepatic impairment had no clinically relevant effect on the PK and safety of abrocitinib and the exposure of abrocitinib active moiety. These results support the use of abrocitinib without dose adjustment in subjects with mild or moderate hepatic impairment.
Collapse
Affiliation(s)
- Ellen Q. Wang
- Clinical Pharmacology, Global Product DevelopmentPfizer Inc.New YorkNew YorkUSA
| | - Vu Le
- Pfizer Inc.GrotonConnecticutUSA
| | | | | | - Martin E. Dowty
- Pharmacokinetics Dynamics and Metabolism, Medicine DesignPfizer Inc.CambridgeMassachusettsUSA
| | - Lisy Wang
- Clinical Development and Operation, Global Product DevelopmentPfizer Inc.GrotonConnecticutUSA
| | - Bimal K. Malhotra
- Clinical Pharmacology, Global Product DevelopmentPfizer Inc.New YorkNew YorkUSA
| |
Collapse
|
9
|
van Groen BD, van Duijn E, de Vries A, Mooij MG, Tibboel D, Vaes WHJ, de Wildt SN. Proof of Concept: First Pediatric [ 14 C]microtracer Study to Create Metabolite Profiles of Midazolam. Clin Pharmacol Ther 2020; 108:1003-1009. [PMID: 32386327 PMCID: PMC7689753 DOI: 10.1002/cpt.1884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022]
Abstract
Growth and development affect drug-metabolizing enzyme activity thus could alter the metabolic profile of a drug. Traditional studies to create metabolite profiles and study the routes of excretion are unethical in children due to the high radioactive burden. To overcome this challenge, we aimed to show the feasibility of an absorption, distribution, metabolism, and excretion (ADME) study using a [14 C]midazolam microtracer as proof of concept in children. Twelve stable, critically ill children received an oral [14 C]midazolam microtracer (20 ng/kg; 60 Bq/kg) while receiving intravenous therapeutic midazolam. Blood was sampled up to 24 hours after dosing. A time-averaged plasma pool per patient was prepared reflecting the mean area under the curve plasma level, and subsequently one pool for each age group (0-1 month, 1-6 months, 0.5-2 years, and 2-6 years). For each pool [14 C]levels were quantified by accelerator mass spectrometry, and metabolites identified by high resolution mass spectrometry. Urine and feces (n = 4) were collected up to 72 hours. The approach resulted in sufficient sensitivity to quantify individual metabolites in chromatograms. [14 C]1-OH-midazolam-glucuronide was most abundant in all but one age group, followed by unchanged [14 C]midazolam and [14 C]1-OH-midazolam. The small proportion of unspecified metabolites most probably includes [14 C]midazolam-glucuronide and [14 C]4-OH-midazolam. Excretion was mainly in urine; the total recovery in urine and feces was 77-94%. This first pediatric pilot study makes clear that using a [14 C]midazolam microtracer is feasible and safe to generate metabolite profiles and study recovery in children. This approach is promising for first-in-child studies to delineate age-related variation in drug metabolite profiles.
Collapse
Affiliation(s)
- Bianca D. van Groen
- Intensive Care and Department of Pediatric SurgeryErasmus Medical Center – Sophia Children’s HospitalRotterdamThe Netherlands
| | | | | | - Miriam G. Mooij
- Intensive Care and Department of Pediatric SurgeryErasmus Medical Center – Sophia Children’s HospitalRotterdamThe Netherlands
- Department of PediatricsLeiden University Medical CenterLeidenThe Netherlands
- Department of Pharmacology and ToxicologyRadboud UniversityNijmegenThe Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric SurgeryErasmus Medical Center – Sophia Children’s HospitalRotterdamThe Netherlands
| | | | - Saskia N. de Wildt
- Intensive Care and Department of Pediatric SurgeryErasmus Medical Center – Sophia Children’s HospitalRotterdamThe Netherlands
- Department of Pharmacology and ToxicologyRadboud UniversityNijmegenThe Netherlands
| |
Collapse
|
10
|
Zeller A, Brigo A, Brink A, Guerard M, Lang D, Muster W, Runge F, Sutter A, Vock E, Wichard J, Schadt S. Genotoxicity Assessment of Drug Metabolites in the Context of MIST and Beyond. Chem Res Toxicol 2019; 33:10-19. [DOI: 10.1021/acs.chemrestox.9b00348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Andreas Zeller
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Alessandro Brigo
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Andreas Brink
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Melanie Guerard
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Dieter Lang
- Bayer AG, Pharmaceuticals, Research Center Aprath, 42096 Wuppertal, Germany
| | - Wolfgang Muster
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Frank Runge
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400 Biberach, Germany
| | - Andreas Sutter
- Bayer AG, Pharmaceuticals, Research Center Aprath, 42096 Wuppertal, Germany
| | - Esther Vock
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88400 Biberach, Germany
| | - Jörg Wichard
- Bayer AG, Pharmaceuticals, Research Center Aprath, 42096 Wuppertal, Germany
| | - Simone Schadt
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| |
Collapse
|
11
|
Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY. Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Xenografts. Mol Cancer Ther 2018; 17:2481-2489. [PMID: 30206106 DOI: 10.1158/1535-7163.mct-18-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/10/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Min Tong
- Poseida Therapeutics Inc., San Diego, California
| | | | - Wei Zhang
- Hera BioLabs Inc., Lexington, Kentucky
| | | | | | - Eric Ostertag
- Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Sahar Mazhar
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Jack Crawford
- Hera BioLabs Inc., Lexington, Kentucky.,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Goutham Narla
- Hera BioLabs Inc., Lexington, Kentucky.,The University of Michigan, Ann Arbor, Michigan
| | - Tseten Y Jamling
- Hera BioLabs Inc., Lexington, Kentucky. .,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| |
Collapse
|
12
|
Fraser K, Bruckner DM, Dordick JS. Advancing Predictive Hepatotoxicity at the Intersection of Experimental, in Silico, and Artificial Intelligence Technologies. Chem Res Toxicol 2018; 31:412-430. [PMID: 29722533 DOI: 10.1021/acs.chemrestox.8b00054] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adverse drug reactions, particularly those that result in drug-induced liver injury (DILI), are a major cause of drug failure in clinical trials and drug withdrawals. Hepatotoxicity-mediated drug attrition occurs despite substantial investments of time and money in developing cellular assays, animal models, and computational models to predict its occurrence in humans. Underperformance in predicting hepatotoxicity associated with drugs and drug candidates has been attributed to existing gaps in our understanding of the mechanisms involved in driving hepatic injury after these compounds perfuse and are metabolized by the liver. Herein we assess in vitro, in vivo (animal), and in silico strategies used to develop predictive DILI models. We address the effectiveness of several two- and three-dimensional in vitro cellular methods that are frequently employed in hepatotoxicity screens and how they can be used to predict DILI in humans. We also explore how humanized animal models can recapitulate human drug metabolic profiles and associated liver injury. Finally, we highlight the maturation of computational methods for predicting hepatotoxicity, the untapped potential of artificial intelligence for improving in silico DILI screens, and how knowledge acquired from these predictions can shape the refinement of experimental methods.
Collapse
Affiliation(s)
- Keith Fraser
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Dylan M Bruckner
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Jonathan S Dordick
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| |
Collapse
|
13
|
Campos ML, Cerqueira LB, Silva BCU, Franchin TB, Galdino-Pitta MR, Pitta IR, Peccinini RG, Pontarolo R. New Pioglitazone Metabolites and Absence of Opened-Ring Metabolites in New N-Substituted Thiazolidinedione. Drug Metab Dispos 2018; 46:879-887. [PMID: 29618574 DOI: 10.1124/dmd.117.079012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 03/30/2018] [Indexed: 12/21/2022] Open
Abstract
Thiazolidinediones (TZDs) are drugs used to treat type 2 diabetes mellitus; however, several safety concerns remain regarding the available drugs in this class. Therefore, the search for new TZD candidates is ongoing; metabolism studies play a crucial step in the development of new candidates. Pioglitazone, one of the most commonly used TZDs, and GQ-11, a new N-substituted TZD, were investigated in terms of their metabolic activity in rat and human liver microsomes to assess their metabolic stability and investigate their metabolites. Methods for preparation of samples were based on liquid-liquid extraction and protein precipitation. Quantitation was performed using liquid chromatography (LC)-tandem mass spectrometry, and the metabolite investigation was performed using ultraperformance LC coupled to a hybrid quadrupole-time of flight mass spectrometer. The predicted intrinsic clearance of GQ-11 was 70.3 and 46.1 ml/kg per minute for rats and humans, respectively. The predicted intrinsic clearance of pioglitazone was 24.1 and 15.9 ml/kg per minute for rats and humans, respectively. The pioglitazone metabolite investigation revealed two unpublished metabolites (M-D and M-A). M-A is a hydration product and may be related to the mechanism of ring opening and the toxicity of pioglitazone. The metabolites of GQ-11 are products of oxidation; no ring-opening metabolite was observed for GQ-11. In conclusion, under the same experimental conditions, a ring-opening metabolite was observed only for pioglitazone. The resistance of GQ-11 to the ring opening is probably related to N-substitution in the TZD ring.
Collapse
Affiliation(s)
- Michel Leandro Campos
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Letícia Bonancio Cerqueira
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Bruna Cristina Ulian Silva
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Taísa Busaranho Franchin
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Marina Rocha Galdino-Pitta
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Ivan Rocha Pitta
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Rosângela Gonçalves Peccinini
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| | - Roberto Pontarolo
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil (M.L.C., L.B.C., R.P.); Department of Natural Active Principles and Toxicology, Faculdade de Ciências Farmacêuticas, São Paulo University (UNESP), Araraquara, São Paulo, Brazil (B.C.U.S., T.B.F., R.G.P.); and Laboratory of Design and Drug Synthesis, Universidade Federal de Pernambuco, Pernambuco, Brazil (M.R.G.-P., I.R.P.)
| |
Collapse
|
14
|
Schadt S, Bister B, Chowdhury SK, Funk C, Hop CECA, Humphreys WG, Igarashi F, James AD, Kagan M, Khojasteh SC, Nedderman ANR, Prakash C, Runge F, Scheible H, Spracklin DK, Swart P, Tse S, Yuan J, Obach RS. A Decade in the MIST: Learnings from Investigations of Drug Metabolites in Drug Development under the “Metabolites in Safety Testing” Regulatory Guidance. Drug Metab Dispos 2018; 46:865-878. [DOI: 10.1124/dmd.117.079848] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/21/2018] [Indexed: 11/22/2022] Open
|
15
|
Vishwakarma SK, Bardia A, Lakkireddy C, Nagarapu R, Habeeb MA, Khan AA. Bioengineered humanized livers as better three-dimensional drug testing model system. World J Hepatol 2018; 10:22-33. [PMID: 29399275 PMCID: PMC5787681 DOI: 10.4254/wjh.v10.i1.22] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/28/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To develop appropriate humanized three-dimensional ex-vivo model system for drug testing.
METHODS Bioengineered humanized livers were developed in this study using human hepatic stem cells repopulation within the acellularized liver scaffolds which mimics with the natural organ anatomy and physiology. Six cytochrome P-450 probes were used to enable efficient identification of drug metabolism in bioengineered humanized livers. The drug metabolism study in bioengineered livers was evaluated to identify the absorption, distribution, metabolism, excretion and toxicity responses.
RESULTS The bioengineered humanized livers showed cellular and molecular characteristics of human livers. The bioengineered liver showed three-dimensional natural architecture with intact vasculature and extra-cellular matrix. Human hepatic cells were engrafted similar to the human liver. Drug metabolism studies provided a suitable platform alternative to available ex-vivo and in vivo models for identifying cellular and molecular dynamics of pharmacological drugs.
CONCLUSION The present study paves a way towards the development of suitable humanized preclinical model systems for pharmacological testing. This approach may reduce the cost and time duration of preclinical drug testing and further overcomes on the anatomical and physiological variations in xenogeneic systems.
Collapse
Affiliation(s)
- Sandeep Kumar Vishwakarma
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Avinash Bardia
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Chandrakala Lakkireddy
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Raju Nagarapu
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Md Aejaz Habeeb
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| |
Collapse
|
16
|
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: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [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
|
17
|
Luffer-Atlas D, Atrakchi A. A decade of drug metabolite safety testing: industry and regulatory shared learning. Expert Opin Drug Metab Toxicol 2017; 13:897-900. [DOI: 10.1080/17425255.2017.1364362] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Debra Luffer-Atlas
- Drug Disposition and Toxicology, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Aisar Atrakchi
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
18
|
Sato-Nakai M, Kawashima K, Nakagawa T, Tachibana Y, Yoshida M, Takanashi K, Morcos PN, Binder M, Moore DJ, Yu L. Metabolites of alectinib in human: their identification and pharmacological activity. Heliyon 2017; 3:e00354. [PMID: 28725874 PMCID: PMC5506877 DOI: 10.1016/j.heliyon.2017.e00354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/21/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023] Open
Abstract
Two metabolites (M4 and M1b) in plasma and four metabolites (M4, M6, M1a and M1b) in faeces were detected through the human ADME study following a single oral administration of [14C]alectinib, a small-molecule anaplastic lymphoma kinase inhibitor, to healthy subjects. In the present study, M1a and M1b, which chemical structures had not been identified prior to the human ADME study, were identified as isomers of a carboxylate metabolite oxidatively cleaved at the morpholine ring. In faeces, M4 and M1b were the main metabolites, which shows that the biotransformation to M4 and M1b represents two main metabolic pathways for alectinib. In plasma, M4 was a major metabolite and M1b was a minor metabolite. The contribution to in vivo pharmacological activity of these circulating metabolites was assessed from their in vitro pharmacological activity and plasma protein binding. M4 had a similar cancer cell growth inhibitory activity and plasma protein binding to that of alectinib, suggesting its contribution to the antitumor activity of alectinib, whereas the pharmacological activity of M1b was insignificant.
Collapse
Affiliation(s)
- Mika Sato-Nakai
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kosuke Kawashima
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Toshito Nakagawa
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Yukako Tachibana
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Miyuki Yoshida
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kenji Takanashi
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Peter N Morcos
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| | - Martin Binder
- Roche Innovation Center Basel, Knozern-Hauptsitz, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - David J Moore
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| | - Li Yu
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| |
Collapse
|
19
|
Towards a Humanized Mouse Model of Liver Stage Malaria Using Ectopic Artificial Livers. Sci Rep 2017; 7:45424. [PMID: 28361899 PMCID: PMC5374446 DOI: 10.1038/srep45424] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
The malaria liver stage is an attractive target for antimalarial development, and preclinical malaria models are essential for testing such candidates. Given ethical concerns and costs associated with non-human primate models, humanized mouse models containing chimeric human livers offer a valuable alternative as small animal models of liver stage human malaria. The best available human liver chimeric mice rely on cellular transplantation into mice with genetically engineered liver injury, but these systems involve a long and variable humanization process, are expensive, and require the use of breeding-challenged mouse strains which are not widely accessible. We previously incorporated primary human hepatocytes into engineered polyethylene glycol (PEG)-based nanoporous human ectopic artificial livers (HEALs), implanted them in mice without liver injury, and rapidly generated human liver chimeric mice in a reproducible and scalable fashion. By re-designing the PEG scaffold to be macroporous, we demonstrate the facile fabrication of implantable porous HEALs that support liver stage human malaria (P. falciparum) infection in vitro, and also after implantation in mice with normal liver function, 60% of the time. This proof-of-concept study demonstrates the feasibility of applying a tissue engineering strategy towards the development of scalable preclinical models of liver stage malaria infection for future applications.
Collapse
|
20
|
Kamimura H, Ito S, Chijiwa H, Okuzono T, Ishiguro T, Yamamoto Y, Nishinoaki S, Ninomiya SI, Mitsui M, Kalgutkar AS, Yamazaki H, Suemizu H. Simulation of human plasma concentration-time profiles of the partial glucokinase activator PF-04937319 and its disproportionate N-demethylated metabolite using humanized chimeric mice and semi-physiological pharmacokinetic modeling. Xenobiotica 2016; 47:382-393. [PMID: 27389028 DOI: 10.1080/00498254.2016.1199063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
1. The partial glucokinase activator N,N-dimethyl-5-((2-methyl-6-((5-methylpyrazin-2-yl)carbamoyl)benzofuran-4-yl)oxy)pyrimidine-2-carboxamide (PF-04937319) is biotransformed in humans to N-methyl-5-((2-methyl-6-((5-methylpyrazin-2-yl)carbamoyl)benzofuran-4-yl)oxy)pyrimidine-2-carboxamide (M1), accounting for ∼65% of total exposure at steady state. 2. As the disproportionately abundant nature of M1 could not be reliably predicted from in vitro metabolism studies, we evaluated a chimeric mouse model with humanized liver on TK-NOG background for its ability to retrospectively predict human disposition of PF-04937319. Since livers of chimeric mice were enlarged by hyperplasia and contained remnant mouse hepatocytes, hepatic intrinsic clearances normalized for liver weight, metabolite formation and liver to plasma concentration ratios were plotted against the replacement index by human hepatocytes and extrapolated to those in the virtual chimeric mouse with 100% humanized liver. 3. Semi-physiological pharmacokinetic analyses using the above parameters revealed that simulated concentration curves of PF-04937319 and M1 were approximately superimposed with the observed clinical data in humans. 4. Finally, qualitative profiling of circulating metabolites in humanized chimeric mice dosed with PF-04937319 or M1 also revealed the presence of a carbinolamide metabolite, identified in the clinical study as a human-specific metabolite. The case study demonstrates that humanized chimeric mice may be potentially useful in preclinical discovery towards studying disproportionate or human-specific metabolism of drug candidates.
Collapse
Affiliation(s)
- Hidetaka Kamimura
- a Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Tokyo , Japan.,b Laboratory Animal Research Department , Central Institute for Experimental Animals, Kawasaki , Kanagawa , Japan
| | - Satoshi Ito
- a Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Tokyo , Japan
| | - Hiroyuki Chijiwa
- c Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Ibaraki , Japan
| | - Takeshi Okuzono
- c Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Ibaraki , Japan
| | - Tomohiro Ishiguro
- c Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Ibaraki , Japan
| | - Yosuke Yamamoto
- c Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Ibaraki , Japan
| | - Sho Nishinoaki
- c Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Ibaraki , Japan
| | - Shin-Ichi Ninomiya
- a Drug Development Solutions Division, Sekisui Medical Co., Ltd. , Tokyo , Japan
| | - Marina Mitsui
- d Showa Pharmaceutical University, Machida , Tokyo , Japan , and
| | | | - Hiroshi Yamazaki
- d Showa Pharmaceutical University, Machida , Tokyo , Japan , and
| | - Hiroshi Suemizu
- b Laboratory Animal Research Department , Central Institute for Experimental Animals, Kawasaki , Kanagawa , Japan
| |
Collapse
|
21
|
Martin IJ, Hill SE, Baker JA, Deshmukh SV, Mulrooney EF. A Pharmacokinetic Modeling Approach to Predict the Contribution of Active Metabolites to Human Efficacious Dose. ACTA ACUST UNITED AC 2016; 44:1435-40. [PMID: 27260151 DOI: 10.1124/dmd.116.070391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/02/2016] [Indexed: 02/03/2023]
Abstract
A preclinical drug candidate, MRK-1 (Merck candidate drug parent compound), was found to elicit tumor regression in a mouse xenograft model. Analysis of samples from these studies revealed significant levels of two circulating metabolites, whose identities were confirmed by comparison with authentic standards using liquid chromatography-tandem mass spectrometry. These metabolites were found to have an in vitro potency similar to that of MRK-1 against the pharmacological target and were therefore thought to contribute to the observed efficacy. To predict this contribution in humans, a pharmacokinetic (PK) modeling approach was developed. At the mouse efficacious dose, the areas under the plasma concentration time curves (AUCs) of the active metabolites were normalized by their in vitro potency compared with MRK-1. These normalized metabolite AUCs were added to that of MRK-1 to yield a composite efficacious unbound AUC, expressed as "parent drug equivalents," which was used as the target AUC for predictions of the human efficacious dose. In vitro and preclinical PK studies afforded predictions of the PK of MRK-1 and the two active metabolites in human as well as the relative pathway flux to each metabolite. These were used to construct a PK model (Berkeley Madonna, version 8.3.18; Berkeley Madonna Inc., University of California, Berkeley, CA) and to predict the human dose required to achieve the target parent equivalent exposure. These predictions were used to inform on the feasibility of the human dose in terms of size, frequency, formulation, and likely safety margins, as well as to aid in the design of preclinical safety studies.
Collapse
Affiliation(s)
- Iain J Martin
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Susan E Hill
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - James A Baker
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Sujal V Deshmukh
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Erin F Mulrooney
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| |
Collapse
|
22
|
Best practices for metabolite quantification in drug development: updated recommendation from the European Bioanalysis Forum. Bioanalysis 2016; 8:1297-305. [DOI: 10.4155/bio-2016-0103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Metabolite quantification and profiling continues to grow in importance in today's drug development. The guidance provided by the 2008 FDA Metabolites in Safety Testing Guidance and the subsequent ICH M3(R2) Guidance (2009) has led to a more streamlined process to assess metabolite exposures in preclinical and clinical studies in industry. In addition, the European Bioanalysis Forum (EBF) identified an opportunity to refine the strategies on metabolite quantification considering the experience to date with their recommendation paper on the subject dating from 2010 and integrating the recent discussions on the tiered approach to bioanalytical method validation with focus on metabolite quantification. The current manuscript summarizes the discussion and recommendations from a recent EBF Focus Workshop into an updated recommendation for metabolite quantification in drug development.
Collapse
|
23
|
Klencsár B, Bolea-Fernandez E, Flórez MR, Balcaen L, Cuyckens F, Lynen F, Vanhaecke F. Determination of the total drug-related chlorine and bromine contents in human blood plasma using high performance liquid chromatography–tandem ICP-mass spectrometry (HPLC–ICP-MS/MS). J Pharm Biomed Anal 2016; 124:112-119. [DOI: 10.1016/j.jpba.2016.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/14/2016] [Accepted: 02/15/2016] [Indexed: 02/03/2023]
|
24
|
Iegre J, Hayes MA, Thompson RA, Weidolf L, Isin EM. Database Extraction of Metabolite Information of Drug Candidates: Analysis of 27 AstraZeneca Compounds with Human Absorption, Distribution, Metabolism, and Excretion Data. Drug Metab Dispos 2016; 44:732-40. [DOI: 10.1124/dmd.115.067850] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/10/2016] [Indexed: 01/20/2023] Open
|
25
|
Thompson RA, Isin EM, Ogese MO, Mettetal JT, Williams DP. Reactive Metabolites: Current and Emerging Risk and Hazard Assessments. Chem Res Toxicol 2016; 29:505-33. [DOI: 10.1021/acs.chemrestox.5b00410] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Richard A. Thompson
- DMPK, Respiratory, Inflammation & Autoimmunity iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Emre M. Isin
- DMPK, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Monday O. Ogese
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| | - Jerome T. Mettetal
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, 35 Gatehouse Dr, Waltham, Massachusetts 02451, United States
| | - Dominic P. Williams
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| |
Collapse
|
26
|
March S, Ramanan V, Trehan K, Ng S, Galstian A, Gural N, Scull MA, Shlomai A, Mota MM, Fleming HE, Khetani SR, Rice CM, Bhatia SN. Micropatterned coculture of primary human hepatocytes and supportive cells for the study of hepatotropic pathogens. Nat Protoc 2015; 10:2027-53. [PMID: 26584444 PMCID: PMC5867906 DOI: 10.1038/nprot.2015.128] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of therapies and vaccines for human hepatropic pathogens requires robust model systems that enable the study of host-pathogen interactions. However, in vitro liver models of infection typically use either hepatoma cell lines that exhibit aberrant physiology or primary human hepatocytes in culture conditions in which they rapidly lose their hepatic phenotype. To achieve stable and robust in vitro primary human hepatocyte models, we developed micropatterned cocultures (MPCCs), which consist of primary human hepatocytes organized into 2D islands that are surrounded by supportive fibroblast cells. By using this system, which can be established over a period of days, and maintained over multiple weeks, we demonstrate how to recapitulate in vitro hepatic life cycles for the hepatitis B and C viruses and the Plasmodium pathogens P. falciparum and P. vivax. The MPCC platform can be used to uncover aspects of host-pathogen interactions, and it has the potential to be used for drug and vaccine development.
Collapse
Affiliation(s)
- Sandra March
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Vyas Ramanan
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kartik Trehan
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shengyong Ng
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ani Galstian
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nil Gural
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Margaret A Scull
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Amir Shlomai
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Maria M Mota
- Unidade de Malaria, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Heather E Fleming
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Salman R Khetani
- Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Sangeeta N Bhatia
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
27
|
Abstract
Most of what we know about a drug prior to human clinical studies is derived from animal testing. Because animals and humans have substantial differences in their physiology and in their drug metabolism pathways, we do not know very much about the pharmacokinetic and pharmacodynamic behavior of a drug in humans until after it is administered to many people. Hence, drug-induced liver injury has become a significant public health problem, and we have a very inefficient drug development process with a high failure rate. Because the human liver is at the heart of these problems, chimeric mice with humanized livers could be used to address these issues. We examine recent evidence indicating that drug testing in chimeric mice could provide better information about a drug's metabolism, disposition, and toxicity (i.e., its "behavior") in humans and could aid in developing personalized medicine strategies, which would improve drug efficacy and safety.
Collapse
Affiliation(s)
- Dan Xu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305;
| | - Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305;
| |
Collapse
|
28
|
Hutzler JM, Ring BJ, Anderson SR. Low-Turnover Drug Molecules: A Current Challenge for Drug Metabolism Scientists. Drug Metab Dispos 2015; 43:1917-28. [PMID: 26363026 DOI: 10.1124/dmd.115.066431] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/10/2015] [Indexed: 01/12/2023] Open
Abstract
In vitro assays using liver subcellular fractions or suspended hepatocytes for characterizing the metabolism of drug candidates play an integral role in the optimization strategy employed by medicinal chemists. However, conventional in vitro assays have limitations in their ability to predict clearance and generate metabolites for low-turnover (slowly metabolized) drug molecules. Due to a rapid loss in the activity of the drug-metabolizing enzymes, in vitro incubations are typically performed for a maximum of 1 hour with liver microsomes to 4 hours with suspended hepatocytes. Such incubations are insufficient to generate a robust metabolic response for compounds that are slowly metabolized. Thus, the challenge of accurately estimating low human clearance with confidence has emerged to be among the top challenges that drug metabolism scientists are confronted with today. In response, investigators have evaluated novel methodologies to extend incubation times and more sufficiently measure metabolism of low-turnover drugs. These methods include plated human hepatocytes in monoculture, and a novel in vitro methodology using a relay of sequential incubations with suspended cryopreserved hepatocytes. In addition, more complex in vitro cellular models, such as HepatoPac (Hepregen, Medford, MA), a micropatterned hepatocyte-fibroblast coculture system, and the HµREL (Beverley Hills, CA) hepatic coculture system, have been developed and characterized that demonstrate prolonged enzyme activity. In this review, the advantages and disadvantages of each of these in vitro methodologies as it relates to the prediction of clearance and metabolite identification will be described in an effort to provide drug metabolism scientists with the most up-to-date experimental options for dealing with the complex issue of low-turnover drug candidates.
Collapse
Affiliation(s)
- J Matthew Hutzler
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Barbara J Ring
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Shelby R Anderson
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| |
Collapse
|
29
|
Kiebist J, Holla W, Heidrich J, Poraj-Kobielska M, Sandvoss M, Simonis R, Gröbe G, Atzrodt J, Hofrichter M, Scheibner K. One-pot synthesis of human metabolites of SAR548304 by fungal peroxygenases. Bioorg Med Chem 2015; 23:4324-4332. [PMID: 26142319 DOI: 10.1016/j.bmc.2015.06.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/03/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022]
Abstract
Unspecific peroxygenases (UPOs, EC 1.11.2.1) have proved to be stable oxygen-transferring biocatalysts for H2O2-dependent transformation of pharmaceuticals. We have applied UPOs in a drug development program and consider the enzymatic approach in parallel to a conventional chemical synthesis of the human metabolites of the bile acid reabsorption inhibitor SAR548304. Chemical preparation of N,N-di-desmethyl metabolite was realized by a seven-step synthesis starting from a late precursor of SAR548304 and included among others palladium catalysis and laborious chromatographic purification with an overall yield of 27%. The enzymatic approach revealed that the UPO of Marasmius rotula is particularly suitable for selective N-dealkylation of the drug and enabled us to prepare both human metabolites via one-pot conversion with an overall yield of 66% N,N-di-desmethyl metabolite and 49% of N-mono-desmethylated compound in two separated kinetic-controlled reactions.
Collapse
Affiliation(s)
- Jan Kiebist
- Enzymtechnology, Faculty of Science, BTU Cottbus-Senftenberg, Großenhainer Str. 57, 01968 Senftenberg, Germany
| | - Wolfgang Holla
- Isotope Chemistry & Metabolite Synthesis, DSAR-DD, Sanofi-Aventis, Industriepark Höchst G876, 65926 Frankfurt am Main, Germany.
| | - Johannes Heidrich
- Isotope Chemistry & Metabolite Synthesis, DSAR-DD, Sanofi-Aventis, Industriepark Höchst G876, 65926 Frankfurt am Main, Germany
| | - Marzena Poraj-Kobielska
- Department of Bio- and Environmental Sciences, TU Dresden-IHI Zittau, Markt 23, 02763 Zittau, Germany
| | - Martin Sandvoss
- Isotope Chemistry & Metabolite Synthesis, DSAR-DD, Sanofi-Aventis, Industriepark Höchst G876, 65926 Frankfurt am Main, Germany
| | - Reiner Simonis
- Isotope Chemistry & Metabolite Synthesis, DSAR-DD, Sanofi-Aventis, Industriepark Höchst G876, 65926 Frankfurt am Main, Germany
| | - Glenn Gröbe
- Enzymtechnology, Faculty of Science, BTU Cottbus-Senftenberg, Großenhainer Str. 57, 01968 Senftenberg, Germany
| | - Jens Atzrodt
- Isotope Chemistry & Metabolite Synthesis, DSAR-DD, Sanofi-Aventis, Industriepark Höchst G876, 65926 Frankfurt am Main, Germany
| | - Martin Hofrichter
- Department of Bio- and Environmental Sciences, TU Dresden-IHI Zittau, Markt 23, 02763 Zittau, Germany
| | - Katrin Scheibner
- Enzymtechnology, Faculty of Science, BTU Cottbus-Senftenberg, Großenhainer Str. 57, 01968 Senftenberg, Germany
| |
Collapse
|
30
|
Analytical challenges for conducting rapid metabolism characterization for QIVIVE. Toxicology 2015; 332:20-9. [DOI: 10.1016/j.tox.2013.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 12/22/2022]
|
31
|
Tiered approach to metabolite quantification: regional practices reviewed by Japan Bioanalysis Forum discussion group. Bioanalysis 2015; 7:935-8. [DOI: 10.4155/bio.15.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
32
|
Li P, Li Z, Beck WD, Callahan PM, Terry AV, Bar-Peled M, Bartlett MG. Bio-generation of stable isotope-labeled internal standards for absolute and relative quantitation of phase II drug metabolites in plasma samples using LC–MS/MS. Anal Bioanal Chem 2015; 407:4053-63. [DOI: 10.1007/s00216-015-8614-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 01/24/2023]
|
33
|
Xu D, Wu M, Nishimura S, Nishimura T, Michie SA, Zheng M, Yang Z, Yates AJ, Day JS, Hillgren KM, Takeda ST, Guan Y, Guo Y, Peltz G. Chimeric TK-NOG mice: a predictive model for cholestatic human liver toxicity. J Pharmacol Exp Ther 2014; 352:274-80. [PMID: 25424997 DOI: 10.1124/jpet.114.220798] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the substantial interspecies differences in drug metabolism and disposition, drug-induced liver injury (DILI) in humans is often not predicted by studies performed in animal species. For example, a drug (bosentan) used to treat pulmonary artery hypertension caused unexpected cholestatic liver toxicity in humans, which was not predicted by preclinical toxicology studies in multiple animal species. In this study, we demonstrate that NOG mice expressing a thymidine kinase transgene (TK-NOG) with humanized livers have a humanized profile of biliary excretion of a test (cefmetazole) drug, which was shown by an in situ perfusion study to result from interspecies differences in the rate of biliary transport and in liver retention of this drug. We also found that readily detectable cholestatic liver injury develops in TK-NOG mice with humanized livers after 1 week of treatment with bosentan (160, 32, or 6 mg/kg per day by mouth), whereas liver toxicity did not develop in control mice after 1 month of treatment. The laboratory and histologic features of bosentan-induced liver toxicity in humanized mice mirrored that of human subjects. Because DILI has become a significant public health problem, drug safety could be improved if preclinical toxicology studies were performed using humanized TK-NOG.
Collapse
Affiliation(s)
- Dan Xu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Manhong Wu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Sachiko Nishimura
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Toshihiko Nishimura
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Sara A Michie
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Ming Zheng
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Zicheng Yang
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Alexander John Yates
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Jeffrey S Day
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Kathleen M Hillgren
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Saori Takedai Takeda
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Yuan Guan
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Yingying Guo
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| | - Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California (D.X., M.W., T.N., M.Z., Yu.G., G.P.); Center for the Advancement of Health and Bioscience, Sunnyvale, California (S.N., T.N.); Central Institute for Experimental Animals, Kawasaki, Japan (T.N.); Department of Pathology, Stanford University, Stanford, California (S.A.M.); Bruker CAM & LSC7, Fremont, California (Z.Y., A.J.Y.); Department of Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.S.D., K.M.H., Yi.G.); and In Vivo Sciences International, Sunnyvale, California (S.T.T.)
| |
Collapse
|
34
|
Sharma R, Litchfield J, Atkinson K, Eng H, Amin NB, Denney WS, Pettersen JC, Goosen TC, Di L, Lee E, Pfefferkorn JA, Dalvie DK, Kalgutkar AS. Metabolites in Safety Testing Assessment in Early Clinical Development: A Case Study with a Glucokinase Activator. Drug Metab Dispos 2014; 42:1926-39. [DOI: 10.1124/dmd.114.060087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
35
|
Johnson TR, Tan W, Goulet L, Smith EB, Yamazaki S, Walker GS, O’Gorman MT, Bedarida G, Zou HY, Christensen JG, Nguyen LN, Shen Z, Dalvie D, Bello A, Smith BJ. Metabolism, excretion and pharmacokinetics of [14C]crizotinib following oral administration to healthy subjects. Xenobiotica 2014; 45:45-59. [DOI: 10.3109/00498254.2014.941964] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
36
|
Haglund J, Halldin MM, Brunnström Å, Eklund G, Kautiainen A, Sandholm A, Iverson SL. Pragmatic Approaches to Determine the Exposures of Drug Metabolites in Preclinical and Clinical Subjects in the MIST Evaluation of the Clinical Development Phase. Chem Res Toxicol 2014; 27:601-10. [DOI: 10.1021/tx400449z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Johanna Haglund
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | | - Åsa Brunnström
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | - Göran Eklund
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | | - Anna Sandholm
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | |
Collapse
|
37
|
A reflection on fit-for-purpose metabolite investigation at different stages of drug development. Bioanalysis 2014; 6:591-4. [DOI: 10.4155/bio.14.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
38
|
Hendrikx JJMA, Dubbelman AC, Rosing H, Schinkel AH, Schellens JHM, Beijnen JH. Quantification of docetaxel and its metabolites in human plasma by liquid chromatography/tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2013; 27:1925-1934. [PMID: 23939959 DOI: 10.1002/rcm.6654] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 06/09/2013] [Accepted: 06/09/2013] [Indexed: 06/02/2023]
Abstract
RATIONALE During drug development accurate quantification of metabolites in biological samples using mass spectrometry is often hampered by the lack of metabolites of chemically pure quality. However, quantification of metabolites can be useful for assessment and interpretation of (pre)clinical data. We now describe an approach to quantify docetaxel metabolites in human plasma by liquid chromatography/tandem mass spectrometry (LC/MS/MS) using docetaxel calibration standards. METHODS Metabolites (M1/M3, M2 and M4) were generated using microsomal incubations. Retention times of docetaxel and its metabolites were assessed using an LC/UV assay and peak identification was performed by LC/MS(n). Samples containing isolated metabolites from human faeces were quantified by LC/UV and used as references for spiking human plasma samples. LC/MS/MS was applied to sensitively quantify docetaxel and its metabolites in human plasma using docetaxel calibration standards in a range of 0.25-500 ng/mL. RESULTS Because ionisation of docetaxel and its metabolites differed, correction factors were established to quantify the metabolites using docetaxel calibration samples. During method validation, accuracy and precision of the metabolites were within ±7.7% and ≤17.6%, respectively, and within ±14.3% and ≤10.1%, respectively, for docetaxel. Metabolites were found to be unstable in human plasma at ambient temperature. After storage up to 1 year at -20 °C, recovered metabolite concentrations were within ±25%. CONCLUSIONS Development and validation of an LC/MS/MS assay for the quantification of docetaxel and its metabolites M1/M3, M2 and M4 using docetaxel calibration standards is described. The same approach may be used for quantification of metabolites of other drugs by LC/MS/MS when chemically pure reference substances are unavailable.
Collapse
Affiliation(s)
- J J M A Hendrikx
- Department of Pharmacy and Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
39
|
Poraj-Kobielska M, Atzrodt J, Holla W, Sandvoss M, Gröbe G, Scheibner K, Hofrichter M. Preparation of labeled human drug metabolites and drug-drug interaction-probes with fungal peroxygenases. J Labelled Comp Radiopharm 2013; 56:513-9. [PMID: 24285530 DOI: 10.1002/jlcr.3103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/28/2013] [Accepted: 07/01/2013] [Indexed: 01/15/2023]
Abstract
Enzymatic conversion of a drug can be an efficient alternative for the preparation of a complex metabolite compared with a multi-step chemical synthesis approach. Limitations exist for chemical methods for direct oxygen incorporation into organic molecules often suffering from low yields and unspecific oxidation and also for alternative whole-cell biotransformation processes, which require specific fermentation know-how. Stable oxygen-transferring biocatalysts such as unspecific peroxygenases (UPOs) could be an alternative for the synthesis of human drug metabolites and related stable isotope-labeled analogues. This work shows that UPOs can be used in combination with hydrogen/deuterium exchange for an efficient one-step process for the preparation of 4'-OH-diclofenac-d6. The scope of the reaction was investigated by screening of different peroxygenase subtypes for the transformation of selected deuterium-labeled substrates such as phenacetin-d3 or lidocaine-d3. Experiments with diclofenac-d7 revealed that the deuterium-labeling does not affect the kinetic parameters. By using the latter substrate and H2 (18) O2 as cosubstrate, it was possible to prepare a doubly isotope-labeled metabolite (4'-(18) OH-diclofenac-d6). UPOs offer certain practical advantages compared with P450 enzyme systems in terms of stability and ease of handling. Given these advantages, future work will expand the existing 'monooxygenation toolbox' of different fungal peroxygenases that mimic P450 in vitro reactions.
Collapse
Affiliation(s)
- Marzena Poraj-Kobielska
- Department of Biological and Environmental Sciences, TU Dresden - International Institute Zittau, Markt 23, 02763, Zittau, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Callegari E, Kalgutkar AS, Leung L, Obach RS, Plowchalk DR, Tse S. Drug metabolites as cytochrome p450 inhibitors: a retrospective analysis and proposed algorithm for evaluation of the pharmacokinetic interaction potential of metabolites in drug discovery and development. Drug Metab Dispos 2013; 41:2047-55. [PMID: 23792812 DOI: 10.1124/dmd.113.052241] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Understanding drug-drug interactions (DDIs) is a key component of clinical practice ensuring patient safety and efficacy of medicines. The role of drug metabolites in DDIs is a developing area of science, and has been recently highlighted in a draft regulatory guidance. The guidance states that metabolites representing ≥25% of the parent drug's area under the plasma concentration/time curve and/or >10% of exposure of total drug-related material should trigger in vitro characterization of metabolites for cytochrome P450 inhibition and propensity for DDIs. The relationship between in vitro cytochrome P450 inhibitory potency, systemic exposure, and DDI potential of drug metabolites was examined using the Pfizer development database to identify compounds with pre-existing in vivo biotransformation data, where circulating metabolites were identified in humans. The database yielded 33 structurally diverse compounds with collectively 115 distinct circulating metabolites. Of these, 52% (60/115) achieved exposures >25% of parent drug levels as judged from mass balance/metabolite identification studies. It was noted that 14 metabolite standards for 12 parent drugs had been synthesized, monitored in clinical studies, and examined for cytochrome P450 inhibition. For the 14 metabolite/parent drug pairs, no clinically relevant DDIs were expected to occur against the major human cytochrome P450 isoforms. A review of the literature for parent/metabolite DDI information was also conducted to examine trends using a larger data set. Leveraging the analysis of both internal and literature-based data sets, an algorithm was devised for use in drug discovery/early development to assess cytochrome P450 inhibitory potential of drug metabolites and the propensity to cause a clinically relevant DDI.
Collapse
Affiliation(s)
- Ernesto Callegari
- Pharmacokinetics, Dynamics and Metabolism-New Chemical Entities (E.C., L.L., R.S.O., S.T.) and Clinical Pharmacology (D.R.P.), Pfizer Inc., Groton, Connecticut; and Pharmacokinetics, Dynamics and Metabolism-New Chemical Entities, Pfizer Inc., Cambridge, Massachusetts (A.S.K.)
| | | | | | | | | | | |
Collapse
|
41
|
Peltz G. Can 'humanized' mice improve drug development in the 21st century? Trends Pharmacol Sci 2013; 34:255-60. [PMID: 23602782 PMCID: PMC3682766 DOI: 10.1016/j.tips.2013.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 02/08/2023]
Abstract
Chimeric mice, which have human hepatocytes engrafted in their liver, have been used to study human drug metabolism and pharmacodynamic responses for nearly 20 years. However, there are very few examples where their use has prospectively impacted the development of a candidate medication. Here, three different chimeric mouse models and their utility for pharmacology studies are evaluated. Several recent studies indicate that using these chimeric mouse models could help to overcome traditional (predicting human-specific metabolites and toxicities) and 21st century problems (strategies for personalized medicine and selection of optimal combination therapies) in drug development. These examples suggest that there are many opportunities in which the use of chimeric mice could significantly improve the quality of preclinical drug assessment.
Collapse
Affiliation(s)
- Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| |
Collapse
|
42
|
Li P, Gong Y, Lim HK, Jian W, Edom RW, Salter R, Silva J, Weng N. Bio-generation of stable isotope labeled internal standards for absolute and relative quantitation of drug metabolites in plasma samples by LC–MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 926:92-100. [DOI: 10.1016/j.jchromb.2013.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/06/2013] [Accepted: 03/08/2013] [Indexed: 01/23/2023]
|
43
|
Ding J, Chen X, Gao Z, Dai X, Li L, Xie C, Jiang H, Zhang L, Zhong D. Metabolism and pharmacokinetics of novel selective vascular endothelial growth factor receptor-2 inhibitor apatinib in humans. Drug Metab Dispos 2013; 41:1195-210. [PMID: 23509226 DOI: 10.1124/dmd.112.050310] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Apatinib is a new oral antiangiogenic molecule that inhibits vascular endothelial growth factor receptor-2. The present study aimed to determine the metabolism, pharmacokinetics, and excretion of apatinib in humans and to identify the enzymes responsible for its metabolism. The primary routes of apatinib biotransformation included E- and Z-cyclopentyl-3-hydroxylation, N-dealkylation, pyridyl-25-N-oxidation, 16-hydroxylation, dioxygenation, and O-glucuronidation after 3-hydroxylation. Nine major metabolites were confirmed by comparison with reference standards. The total recovery of the administered dose was 76.8% within 96 hours postdose, with 69.8 and 7.02% of the administered dose excreted in feces and urine, respectively. About 59.0% of the administered dose was excreted unchanged via feces. Unchanged apatinib was detected in negligible quantities in urine, indicating that systemically available apatinib was extensively metabolized. The major circulating metabolite was the pharmacologically inactive E-3-hydroxy-apatinib-O-glucuronide (M9-2), the steady-state exposure of which was 125% that of the apatinib. The steady-state exposures of E-3-hydroxy-apatinib (M1-1), Z-3-hydroxy-apatinib (M1-2), and apatinib-25-N-oxide (M1-6) were 56, 22, and 32% of parent drug exposure, respectively. Calculated as pharmacological activity index values, the contribution of M1-1 to the pharmacology of the drug was 5.42 to 19.3% that of the parent drug. The contribution of M1-2 and M1-6 to the pharmacology of the drug was less than 1%. Therefore, apatinib was a major contributor to the overall pharmacological activity in humans. Apatinib was metabolized primarily by CYP3A4/5 and, to a lesser extent, by CYP2D6, CYP2C9, and CYP2E1. UGT2B7 was the main enzyme responsible for M9-2 formation. Both UGT1A4 and UGT2B7 were responsible for Z-3-hydroxy-apatinib-O-glucuronide (M9-1) formation.
Collapse
Affiliation(s)
- Juefang Ding
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Milne SB, Mathews TP, Myers DS, Ivanova PT, Brown HA. Sum of the parts: mass spectrometry-based metabolomics. Biochemistry 2013; 52:3829-40. [PMID: 23442130 DOI: 10.1021/bi400060e] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolomics is a rapidly growing field of research used in the identification and quantification of the small molecule metabolites within an organism, thereby providing insights into cell metabolism and bioenergetics as well as processes important in clinical medicine, such as disposition of pharmaceutical compounds. It offers comprehensive information about thousands of low-molecular mass compounds (<1500 Da) that represent a wide range of pathways and intermediary metabolism. Because of its vast expansion in the past two decades, mass spectrometry has become an indispensable tool in "omic" analyses. The use of different ionization techniques such as the more traditional electrospray and matrix-assisted laser desorption, as well as recently popular desorption electrospray ionization, has allowed the analysis of a wide range of biomolecules (e.g., peptides, proteins, lipids, and sugars), and their imaging and analysis in the original sample environment in a workup free fashion. An overview of the current state of the methodology is given, as well as examples of application.
Collapse
Affiliation(s)
- Stephen B Milne
- Departments of Pharmacology, Chemistry, and Biochemistry, The Vanderbilt Institute of Chemical Biology, Vanderbilt University , Nashville, Tennessee 37240, United States
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Stepan AF, Mascitti V, Beaumont K, Kalgutkar AS. Metabolism-guided drug design. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20317k] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Minagawa T, Nakano K, Furuta S, Iwasa T, Takekawa K, Minato K, Koga T, Sato T, Kawashima K, Kurahashi Y, Onodera H, Naito S, Nakamura K. Perspectives on non-clinical safety evaluation of drug metabolites through the JSOT workshop. J Toxicol Sci 2012; 37:667-73. [PMID: 22863847 DOI: 10.2131/jts.37.667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The prompt and appropriate safety assessment of drug metabolite(s) was mentioned in regulatory guidances such as an International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) guidance, entitled "Guidance on Non-clinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals" (ICH M3(R2)) implemented in January 1 of 2011 in Japan, and has become a significant issue in the drug development. Upon release of ICH M3(R2) Step 4, a survey was conducted between March and April 2010 on the safety assessment of drug metabolites in 63 member companies of the Japan Pharmaceutical Manufacturers Association (JPMA). The Pharmacokinetics Team in the Non-Clinical Evaluation Expert Committee in JPMA conducted a questionnaire survey and compiled the results to comprehend how safety of drug metabolites are currently assessed at research-based pharmaceutical companies in Japan. The assessment of "Metabolites in Safety Testing" (MIST) can be divided into three stages based on the research purpose as follows: MIST 1 is a stage of estimating human drug metabolites and predicting their potential risks, MIST 2 is a stage of deciding the necessity for non-clinical safety studies, and MIST 3 is a stage of conducting non-clinical safety studies. In this paper, we propose typical approaches on safety assessment of metabolites that meet the purpose of each stage, considering the current level of scientific technology. Our proposals are based on the results from our survey and a symposium about the safety assessment of drug metabolites at the 37th annual meeting of the Japanese Society of Toxicology held in June 2010.
Collapse
Affiliation(s)
- Toshiya Minagawa
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Acikgöz A, Giri S, Bader A. Detection of nanolevel drug metabolites in an organotypic culture of primary human hepatocytes and porcine hepatocytes with special reference to a two-compartment model. Int J Nanomedicine 2012; 7:5859-72. [PMID: 23226017 PMCID: PMC3512542 DOI: 10.2147/ijn.s29651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The quantification of drug metabolites produced during drug metabolism is a growing concern for the pharmaceutical industry, regulatory agencies such as the US Food and Drug Administration, the European Medicines Agency, and others. As 70% of drugs are known reactive metabolites and have black box warnings, they are a major cause of drug-induced injury and lead to drug attrition in early or late clinical stages. According to a 2006 survey report of pharmaceutical companies, drug-induced liver injury was ranked first in terms of adverse events, and it remains the most common reason for restriction or withdrawal of a drug from the market by the Food and Drug Administration. Although there are many reasons underlying drug-induced liver injury, one of the most important is liver failure induced by drug metabolites. Generally, a drug produces metabolites that may bind to cellular molecules and trigger a toxicological effect, cause serious adverse drug reactions, or alter cellular functions. Experimental cellular models that attempt to qualify drug metabolites from cell cultures rely on human plasma and urine obtained from clinical trials and supernatant during early in vitro experiments. However, there is a lack of information about the quantification of drug metabolites inside human hepatocytes, where the drug is extensively metabolized. To overcome this limitation, we used the highly accepted, gold standard organotypic cellular model of primary human hepatocytes to investigate and quantify the parent drug, as well as drug metabolites inside human hepatocytes and outside human hepatocytes to evaluate the quantity of drug metabolites, which are assumed to have remained inside the primary human hepatocytes. We refer to this as a two-compartment model, where one compartment is supernatant compared with in vivo hepatic blood circulation, and the other is inside the hepatocyte cell compared with the inside of in vivo human liver. We detected the nanoconcentrations of all major metabolites (desmethyldiazepam, temazepam, and oxazepam) of the diazepam drug, both inside the cells (matrix) and outside the hepatocyte cells (supernatant) at different time points (primary human hepatocytes: 0, 1, 2, 4, 8, and 24 hours; primary porcine hepatocytes: 0, 1, 2, 5, and 24 hours) during biotransformation in an organotypic sandwich cellular model. Although it is difficult to detect tissue distribution of metabolites in humans, we strongly recommend testing in a two-compartment model of primary human hepatocytes, as nonhuman models may not reflect human metabolism. Preclinical drug screening assessment tests that use this two-compartment strategy may facilitate safer registration of new drug candidates.
Collapse
Affiliation(s)
- Ali Acikgöz
- Center for Biotechnology and Biomedicine, Cell Techniques and Applied Stem Cell Biology, Universität Leipzig, Germany
| | | | | |
Collapse
|
49
|
Nishimura T, Nishimura T, Hu Y, Wu M, Pham E, Suemizu H, Elazar M, Liu M, Idilman R, Yurdaydin C, Angus P, Stedman C, Murphy B, Glenn J, Nakamura M, Nomura T, Chen Y, Zheng M, Fitch WL, Peltz G. Using chimeric mice with humanized livers to predict human drug metabolism and a drug-drug interaction. J Pharmacol Exp Ther 2012; 344:388-96. [PMID: 23143674 DOI: 10.1124/jpet.112.198697] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Interspecies differences in drug metabolism have made it difficult to use preclinical animal testing data to predict the drug metabolites or potential drug-drug interactions (DDIs) that will occur in humans. Although chimeric mice with humanized livers can produce known human metabolites for test substrates, we do not know whether chimeric mice can be used to prospectively predict human drug metabolism or a possible DDI. Therefore, we investigated whether they could provide a more predictive assessment for clemizole, a drug in clinical development for the treatment of hepatitis C virus (HCV) infection. Our results demonstrate, for the first time, that analyses performed in chimeric mice can correctly identify the predominant human drug metabolite before human testing. The differences in the rodent and human pathways for clemizole metabolism were of importance, because the predominant human metabolite was found to have synergistic anti-HCV activity. Moreover, studies in chimeric mice also correctly predicted that a DDI would occur in humans when clemizole was coadministered with a CYP3A4 inhibitor. These results demonstrate that using chimeric mice can improve the quality of preclinical drug assessment.
Collapse
Affiliation(s)
- Toshihiko Nishimura
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Howe PWA, Ament Z, Knowles K, Griffin JL, Wright J. Combined use of filtered and edited 1 H NMR spectroscopy to detect 13 C-enriched compounds in complex mixtures. NMR IN BIOMEDICINE 2012; 25:1217-1223. [PMID: 22407896 DOI: 10.1002/nbm.2791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 01/20/2012] [Accepted: 01/27/2012] [Indexed: 05/31/2023]
Abstract
In conventional metabolism and pharmacokinetic studies, radioactive isotopes are used to identify and quantify the breakdown products of xenobiotics. However, the stable isotope (13) C provides a cheaper and less hazardous alternative. Metabolites of (13) C-enriched xenobiotics can be detected, quantified and identified by (13) C-filtered NMR spectroscopy. However, one obstacle to using (13) C is its 1.1% natural abundance that produces a background signal in (13) C-filtered NMR spectra of crude biological extracts. The signal makes it difficult to distinguish between (13) C-enriched xenobiotics resonances from endogenous metabolites unrelated to the xenobiotic. This study proposes that the (13) C background signal can be distinguished from resonances of (13) C-enriched xenobiotics by the absence of a (12) C component in the xenobiotic. This is detected by combined analysis of (13) C-filtered and -edited NMR spectra. The theory underlying the approach is described and the method is demonstrated by the detection of sub-microgram amounts of (13) C-enriched phenacetin in crude extracts of hepatocyte microsomes.
Collapse
Affiliation(s)
- P W A Howe
- Syngenta, Jealott's Hill Research Centre, Bracknell, RG42 6EY, UK.
| | | | | | | | | |
Collapse
|