51
|
Punt A, Pinckaers N, Peijnenburg A, Louisse J. Development of a Web-Based Toolbox to Support Quantitative In-Vitro-to-In-Vivo Extrapolations (QIVIVE) within Nonanimal Testing Strategies. Chem Res Toxicol 2021; 34:460-472. [PMID: 33382582 PMCID: PMC7887804 DOI: 10.1021/acs.chemrestox.0c00307] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Indexed: 12/25/2022]
Abstract
The goal of the present study was to develop an online web-based toolbox that contains generic physiologically based kinetic (PBK) models for rats and humans, including underlying calculation tools to predict plasma protein binding and tissue:plasma distribution, to be used for quantitative in-vitro-to-in-vivo extrapolations (QIVIVE). The PBK models within the toolbox allow first estimations of internal plasma and tissue concentrations of chemicals to be made, based on the logP and pKa of the chemicals and values for intestinal uptake and intrinsic hepatic clearance. As a case study, the toolbox was used to predict oral equivalent doses of in vitro ToxCast bioactivity data for the food additives methylparaben, propyl gallate, octyl gallate, and dodecyl gallate. These oral equivalent doses were subsequently compared with human exposure estimates, as a low tier assessment allowing prioritization for further assessment. The results revealed that daily intake levels of especially propyl gallate can lead to internal plasma concentrations that are close to in vitro biological effect concentrations, particularly with respect to the inhibition of human thyroid peroxidase (TPO). Estrogenic effects were not considered likely to be induced by the food additives, as daily exposure levels of the different compounds remained 2 orders of magnitude below the oral equivalent doses for in vitro estrogen receptor activation. Overall, the results of the study show how the toolbox, which is freely accessible through www.qivivetools.wur.nl, can be used to obtain initial internal dose estimates of chemicals and to prioritize chemicals for further assessment, based on the comparison of oral equivalent doses of in vitro biological activity data with human exposure levels.
Collapse
Affiliation(s)
- Ans Punt
- Wageningen
Food Safety Research, Akkermaalsbos 2, 6708 WB Wageningen, The Netherlands
| | - Nicole Pinckaers
- Wageningen
Food Safety Research, Akkermaalsbos 2, 6708 WB Wageningen, The Netherlands
| | - Ad Peijnenburg
- Wageningen
Food Safety Research, Akkermaalsbos 2, 6708 WB Wageningen, The Netherlands
| | - Jochem Louisse
- Wageningen
Food Safety Research, Akkermaalsbos 2, 6708 WB Wageningen, The Netherlands
| |
Collapse
|
52
|
Abstract
Currently, peptide-nanoparticle (NP) conjugates have been demonstrated to be efficient and powerful tools for the treatment and the diagnosis of various diseases as well as in the bioimaging application. Several bioconjugation strategies have been adopted to formulate the peptide-NP conjugates. In this review, we discuss the exciting applications of peptide-gold (Au) NP conjugates in the area of drug delivery, targeting, cancer therapy, brain diseases, vaccines, immune modulation, biosensor, colorimetric detection of heavy metals, and bio-labeling in vitro and in vivo models. Within this framework, various approaches such as radiotherapy, photothermal therapy, photodynamic therapy and chemo-photothermal therapy have been demonstrated for the treatment of several diseases. Moreover, we highlight how the morphology, size, density of peptide and the protein corona influence the biological activity, biodistribution and biological fate of peptide-AuNP conjugates. In the end, we discuss the future outlook and the challenges being faced in the clinical translation of the peptide-AuNP conjugates. Overall, this review emphasizes that the peptide-AuNP conjugates might be used as potential theranostic agents for the treatment of life-threatening diseases in an economical fashion in the future.
Collapse
Affiliation(s)
- Akhilesh Rai
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
53
|
Stow R, Kendrick J, Ibbotson N, Adjin-Tettey G, Murphy B, Trojca R, Miller J, Konradsen G, Øvlisen K, Helleberg H, Hansen JJ. A new group housing approach for non-human primate metabolism studies. J Pharmacol Toxicol Methods 2021; 107:106947. [PMID: 33429039 DOI: 10.1016/j.vascn.2020.106947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 11/26/2022]
Abstract
Understanding the absorption, distribution, metabolism and excretion (ADME) of candidate drugs in preclinical species is an integral part of the safety and efficacy evaluation in drug development. For this purpose, the housing of single animals in metabolism cages has historically been common practice for ADME studies. Whilst mini-pigs and dogs are selected wherever possible, non-human primates (NHPs) are used where there is no suitable scientific alternative. Having undergone only minimal revisions over the past 30 years, the traditional single-housing metabolism cage design for NHPs significantly limits normal vertical movement and social behaviours in primates. Minimising animal suffering and improving welfare is an important aspect of working with animals in research and Novo Nordisk A/S, together with collaborators, has focused on this area for many years. A novel metabolism cage for group housing of NHPs has been designed in a joint collaboration between Novo Nordisk A/S and Covance Inc. The advantages of this novel cage are extensive, including a significantly increased cage volume and ability for socialisation, as well as improvements to alleviate stress and boredom. The excretion balance data from six male NHPs housed in single or group metabolism cages were compared using the radiolabelled test compound [14C]-quetiapine. Welfare, in terms of stress and behaviour, when animals were single or group housed was also assessed. Mean recoveries of radioactivity were shown to be comparable irrespective of housing design (83.2% for group-housed animals vs. 87.1% for single-housed animals), supporting the potential suitability of NHP group housing for future metabolism ADME studies.
Collapse
Affiliation(s)
- Ruth Stow
- Covance Laboratories Ltd., Harrogate, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Maková B, Mik V, Lišková B, Gonzalez G, Vítek D, Medvedíková M, Monfort B, Ručilová V, Kadlecová A, Khirsariya P, Gándara Barreiro Z, Havlíček L, Zatloukal M, Soural M, Paruch K, D'Autréaux B, Hajdúch M, Strnad M, Voller J. Cytoprotective activities of kinetin purine isosteres. Bioorg Med Chem 2021; 33:115993. [PMID: 33497938 DOI: 10.1016/j.bmc.2021.115993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/31/2020] [Indexed: 01/23/2023]
Abstract
Kinetin (N6-furfuryladenine), a plant growth substance of the cytokinin family, has been shown to modulate aging and various age-related conditions in animal models. Here we report the synthesis of kinetin isosteres with the purine ring replaced by other bicyclic heterocycles, and the biological evaluation of their activity in several in vitro models related to neurodegenerative diseases. Our findings indicate that kinetin isosteres protect Friedreich́s ataxia patient-derived fibroblasts against glutathione depletion, protect neuron-like SH-SY5Y cells from glutamate-induced oxidative damage, and correct aberrant splicing of the ELP1 gene in fibroblasts derived from a familial dysautonomia patient. Although the mechanism of action of kinetin derivatives remains unclear, our data suggest that the cytoprotective activity of some purine isosteres is mediated by their ability to reduce oxidative stress. Further, the studies of permeation across artificial membrane and model gut and blood-brain barriers indicate that the compounds are orally available and can reach central nervous system. Overall, our data demonstrate that isosteric replacement of the kinetin purine scaffold is a fruitful strategy for improving known biological activities of kinetin and discovering novel therapeutic opportunities.
Collapse
Affiliation(s)
- Barbara Maková
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Václav Mik
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Barbora Lišková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, Olomouc CZ-77515, Czech Republic
| | - Gabriel Gonzalez
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic; Department of Neurology, Palacký University Olomouc, Faculty of Medicine and Dentistry and University Hospital, Olomouc, Czech Republic
| | - Dominik Vítek
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, Olomouc CZ-77515, Czech Republic
| | - Martina Medvedíková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, Olomouc CZ-77515, Czech Republic
| | - Beata Monfort
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Veronika Ručilová
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. listopadu 1192/12, Olomouc CZ-783-71, Czech Republic
| | - Alena Kadlecová
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Prashant Khirsariya
- Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Zoila Gándara Barreiro
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Libor Havlíček
- Isotope Laboratory, The Czech Academy of Science, Institute of Experimental Botany, Vídeňská 1083, Praha 4 CZ-14220, Czech Republic
| | - Marek Zatloukal
- Department of Chemical Biolology and Genetics, Centre of the Region Hana for Biotechnological and Agricultural Research, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Miroslav Soural
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. listopadu 1192/12, Olomouc CZ-783-71, Czech Republic
| | - Kamil Paruch
- Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Benoit D'Autréaux
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, Olomouc CZ-77515, Czech Republic
| | - Miroslav Strnad
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic
| | - Jiří Voller
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, Olomouc CZ-77515, Czech Republic; Laboratory of Growth Regulators, Palacký University & Institute of Experimental Botany AS CR, Šlechtitelů 27, Olomouc CZ-78371, Czech Republic.
| |
Collapse
|
55
|
Peneda Pacheco D, Suárez Vargas N, Visentin S, Petrini P. From tissue engineering to engineering tissues: the role and application of in vitro models. Biomater Sci 2021; 9:70-83. [DOI: 10.1039/d0bm01097a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review defines and explores the engineering process and the multifaceted potential and limitations of models within the biomedical field.
Collapse
Affiliation(s)
- Daniela Peneda Pacheco
- Department of Chemistry
- Materials and Chemical Engineering “Giulio Natta” – Politecnico di Milano
- Italy
| | - Natalia Suárez Vargas
- Department of Chemistry
- Materials and Chemical Engineering “Giulio Natta” – Politecnico di Milano
- Italy
| | - Sonja Visentin
- Molecular Biotechnology and Health Sciences Department
- University of Torino
- Torino
- Italy
| | - Paola Petrini
- Department of Chemistry
- Materials and Chemical Engineering “Giulio Natta” – Politecnico di Milano
- Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R)
- Politecnico di Milano Unit
| |
Collapse
|
56
|
Whole blood or plasma: what is the ideal matrix for pharmacokinetic-driven drug candidate selection? Future Med Chem 2020; 13:157-171. [PMID: 33275044 DOI: 10.4155/fmc-2020-0187] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In the present era of drug development, quantification of drug concentrations following pharmacokinetic studies has preferentially been performed using plasma as a matrix rather than whole blood. However, it is critical to realize the difference between measuring drug concentrations in blood versus plasma and the consequences thereof. Pharmacokinetics using plasma data may be misleading if concentrations differ between plasma and red blood cells (RBCs) because of differential binding in blood. In this review, factors modulating the partitioning of drugs into RBCs are discussed and the importance of determining RBC uptake of drugs for drug candidate selection is explored. In summary, the choice of matrix (plasma vs whole blood) is an important consideration to be factored in during drug discovery.
Collapse
|
57
|
Mignani S, Shi X, Ceña V, Shcharbin D, Bryszewska M, Majoral JP. In vivo therapeutic applications of phosphorus dendrimers: state of the art. Drug Discov Today 2020; 26:677-689. [PMID: 33285297 DOI: 10.1016/j.drudis.2020.11.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/27/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, rue des Saints Peres, 75006 Paris, France; CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China.
| | - Valentin Ceña
- CIBERNED, ISCII, Madrid, Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, Avda. Almansa, 14, 02006 Albacete, Spain
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, Minsk, Belarus
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, 31077, Toulouse Cedex 4, France; Université Toulouse 118 route de Narbonne, 31077, Toulouse Cedex 4, France.
| |
Collapse
|
58
|
Kropf C, Begnaud F, Gimeno S, Berthaud F, Debonneville C, Segner H. In Vitro Biotransformation Assays Using Liver S9 Fractions and Hepatocytes from Rainbow Trout (Oncorhynchus mykiss): Overcoming Challenges with Difficult to Test Fragrance Chemicals. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2020; 39:2396-2408. [PMID: 32915480 DOI: 10.1002/etc.4872] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/02/2020] [Accepted: 09/08/2020] [Indexed: 06/11/2023]
Abstract
In vitro metabolic stability assays using rainbow trout (Oncorhynchus mykiss) isolated hepatocytes (RT-HEP) or hepatic S9 fractions (RT-S9) were introduced to provide biotransformation rate data for the assessment of chemical bioaccumulation in fish. The present study explored the suitability of the RT-HEP and RT-S9 assays for difficult test chemicals, and the in vitro-based predictions were compared to in silico-based predictions and in vivo-measured bioconcentration factors (BCFs). The results show that volatile or reactive chemicals can be tested with minor modifications of the in vitro protocols. For hydrophobic chemicals, a passive dosing technique was developed. Finally, a design-of-experiment approach was used to identify optimal in vitro assay conditions. The modified assay protocols were applied to 10 fragrances with diverse physicochemical properties. The in vitro intrinsic clearance rates were higher in the S9 than in the hepatocyte assay, but the in vitro-in vivo (IVIV) predictions were comparable between the 2 assays. The IVIV predictions classified the test chemicals as nonbioaccumulative (BCF < 2000), which was in agreement with the in vivo data but in contrast to the in silico-based predictions. The findings from the present study provide strong evidence that the RT-HEP and RT-S9 assays can provide reliable estimates of in vivo biotransformation rates for test chemicals with difficult physicochemical properties. Environ Toxicol Chem 2020;39:2396-2408. © 2020 SETAC.
Collapse
Affiliation(s)
- Christian Kropf
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Frédéric Begnaud
- Corporate R&D Division/Analytical Innovation, Firmenich International, Geneva, Switzerland
| | - Sylvia Gimeno
- Legal and Compliance, Firmenich Belgium, Louvain-La-Neuve, Belgium
| | - Fabienne Berthaud
- Corporate R&D Division/Analytical Innovation, Firmenich International, Geneva, Switzerland
| | - Christian Debonneville
- Corporate R&D Division/Analytical Innovation, Firmenich International, Geneva, Switzerland
| | - Helmut Segner
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
59
|
Kerr WJ, Knox GJ, Reid M, Tuttle T, Bergare J, Bragg RA. Computationally-Guided Development of a Chelated NHC-P Iridium(I) Complex for the Directed Hydrogen Isotope Exchange of Aryl Sulfones. ACS Catal 2020; 10:11120-11126. [PMID: 33123410 PMCID: PMC7587147 DOI: 10.1021/acscatal.0c03031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/27/2020] [Indexed: 02/05/2023]
Abstract
Herein, we report the rational, computationally-guided design of an iridium(I) catalyst system capable of enabling directed hydrogen isotope exchange (HIE) with the challenging sulfone directing group. Substrate binding energy was used as a parameter to guide rational ligand design via an in silico catalyst screen, resulting in a lead series of chelated iridium(I) NHC-phosphine complexes. Subsequent preparative studies show that the optimal catalyst system displays high levels of activity in HIE, and we demonstrate the labeling of a broad scope of substituted aryl sulfones. We also show that the activity of the catalyst is maintained at low pressures of deuterium gas and apply these conditions to tritium radiolabeling, including the expedient synthesis of a tritium-labeled drug molecule.
Collapse
Affiliation(s)
- William J. Kerr
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XL, Scotland, U.K
| | - Gary J. Knox
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XL, Scotland, U.K
| | - Marc Reid
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XL, Scotland, U.K
| | - Tell Tuttle
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XL, Scotland, U.K
| | - Jonas Bergare
- Early Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Ryan A. Bragg
- Early Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| |
Collapse
|
60
|
Kola-Mustapha AT, Yohanna KA, Ghazali YO, Ayotunde HT. Design, formulation and evaluation of Chasmanthera dependens Hochst and Chenopodium ambrosioides Linn based gel for its analgesic and anti-inflammatory activities. Heliyon 2020; 6:e04894. [PMID: 32984602 PMCID: PMC7492848 DOI: 10.1016/j.heliyon.2020.e04894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/04/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022] Open
Abstract
Background The incidence of pain and inflammation in West Africa and in fact globally, continues to increase at an alarming rate. This research was conducted to investigate the analgesic and anti-inflammatory activities of leaf extracts of Chasmanthera dependens and Chenopodium ambrosioides; formulate and evaluate polyherbal gels from their combination in a bid to providing topical therapeutic solutions to pain and inflammation. Methods Pre-formulation studies (phytochemical analysis, in vitro analgesic and anti-inflammatory activities) were conducted on the methanol leaf extracts of Chasmanthera dependens and Chenopodium ambrosioides. Individual and polyherbal gels were prepared using polymer carbopol 940 (1%) at combination ratios of 0:100, 25:75, 50:50, 75:25 and 100:0 Chasmanthera:Chenopodium. These herbal gels were evaluated for physical parameters, pH, viscosity, extrudability and spreadability. Analgesic and anti-inflammatory activities of herbal gels were evaluated by their inhibitory activities (percentage inhibition) against COX-2, TNF-α, IL-10, PGE-2 and compared with commercial diclofenac gel. Results The phytochemicals of the two extracts detected gave varied contents of major classes of secondary metabolites. The pre formulation inhibitory studies of the two extracts exhibited dose dependent inhibitory activities against COX-2, TNF-α, IL-10, PGE-2. The physical appearance, homogeneity, and consistency of the herbal formulations were good. The herbal gels were spreadable with good extrudability. The pH of the herbal gels ranged from 4.5 ± 0.4 to 5.2 ± 0.4. The viscosity of the herbal gels ranged between 4.3 ± 0.2 and 4.7 ± 0.4 Pas. The herbal gels exhibited significant differences in inhibitory activities against COX-2, TNF-α, IL-10, PGE-2 when compared with control commercial diclofenac gel. Conclusion The outcomes, including the inhibition of mediators COX-2, TNF-α, IL-10, PGE-2, confirm the use of the plant extracts under study, the individual and polyherbal gels formulated for the potential topical therapeutic treatment of pain and inflammation.
Collapse
Affiliation(s)
- A T Kola-Mustapha
- Department of Pharmaceutics and Industrial Pharmacy, University of Ilorin, Ilorin, Nigeria
| | - K A Yohanna
- Department of Pharmaceutics and Industrial Pharmacy, University of Ilorin, Ilorin, Nigeria
| | - Y O Ghazali
- Department of Pharmaceutics and Industrial Pharmacy, University of Ilorin, Ilorin, Nigeria
| | - H T Ayotunde
- Department of Pharmaceutics and Industrial Pharmacy, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
61
|
Sudsakorn S, Bahadduri P, Fretland J, Lu C. 2020 FDA Drug-drug Interaction Guidance: A Comparison Analysis and Action Plan by Pharmaceutical Industrial Scientists. Curr Drug Metab 2020; 21:403-426. [DOI: 10.2174/1389200221666200620210522] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/28/2020] [Accepted: 05/28/2020] [Indexed: 12/26/2022]
Abstract
Background:
In January 2020, the US FDA published two final guidelines, one entitled “In vitro Drug
Interaction Studies - Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions Guidance for Industry”
and the other entitled “Clinical Drug Interaction Studies - Cytochrome P450 Enzyme- and Transporter-Mediated
Drug Interactions Guidance for Industry”. These were updated from the 2017 draft in vitro and clinical DDI
guidance.
Methods:
This study is aimed to provide an analysis of the updates along with a comparison of the DDI guidelines
published by the European Medicines Agency (EMA) and Japanese Pharmaceuticals and Medical Devices Agency
(PMDA) along with the current literature.
Results:
The updates were provided in the final FDA DDI guidelines and explained the rationale of those changes
based on the understanding from research and literature. Furthermore, a comparison among the FDA, EMA, and
PMDA DDI guidelines are presented in Tables 1, 2 and 3.
Conclusion:
The new 2020 clinical DDI guidance from the FDA now has even higher harmonization with the
guidance (or guidelines) from the EMA and PMDA. A comparison of DDI guidance from the FDA 2017, 2020,
EMA, and PMDA on CYP and transporter based DDI, mathematical models, PBPK, and clinical evaluation of DDI
is presented in this review.
Collapse
Affiliation(s)
- Sirimas Sudsakorn
- Department of Drug Metabolism and Pharmacokinetics, Sanofi-Genzyme, Waltham, MA 02451, United States
| | - Praveen Bahadduri
- Department of Drug Metabolism and Pharmacokinetics, Sanofi-Genzyme, Waltham, MA 02451, United States
| | - Jennifer Fretland
- Department of Drug Metabolism and Pharmacokinetics, Sanofi-Genzyme, Waltham, MA 02451, United States
| | - Chuang Lu
- Department of Drug Metabolism and Pharmacokinetics, Sanofi-Genzyme, Waltham, MA 02451, United States
| |
Collapse
|
62
|
Kakutani N, Iwai T, Ohno Y, Kobayashi S, Nomura Y. Evaluation of covalent binding of flutamide and its risk assessment using 19F-NMR. Xenobiotica 2020; 51:88-94. [PMID: 32876521 DOI: 10.1080/00498254.2020.1817626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The formation of reactive metabolites (RMs) is a problem in drug development that sometimes results in severe hepatotoxicity. As detecting RMs themselves is difficult, a covalent binding assay using expensive radiolabelled tracers is usually performed for candidate selection. This study aimed to provide a practical approach toward the risk assessment of hepatotoxicity induced by covalent binding before candidate selection. We focused on flutamide because it contains a trifluoromethyl group that shows a strong singlet peak by 19F nuclear magnetic resonance (NMR) spectrometry. The covalent binding of flutamide was evaluated using quantitative NMR and its risk for hepatotoxicity was assessed by estimating the RM burden, an index that reflects the body burden associated with RM exposure by determining the extent of covalent binding, clinical dose and in vivo clearance. The extent of covalent binding and RM burden was 296 pmol/mg/h and 37.9 mg/day, respectively. Flutamide was categorised as high risk with an RM burden >10 mg/day consistent with its clinical hepatotoxicity. These results indicate that a combination of covalent binding assay using 19F-NMR and RM burden is useful for the risk assessment of RMs without using radiolabelled compounds.
Collapse
Affiliation(s)
- Nobuyuki Kakutani
- Drug Metabolism and Pharmacokinetics Research Laboratories, Japan Tobacco Inc, Takatsuki, Japan
| | - Takahiro Iwai
- Product Development Laboratories, Japan Tobacco Inc, Takatsuki, Japan
| | - Yasushi Ohno
- Product Development Laboratories, Japan Tobacco Inc, Takatsuki, Japan
| | - Satoru Kobayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Japan Tobacco Inc, Takatsuki, Japan
| | - Yukihiro Nomura
- Drug Metabolism and Pharmacokinetics Research Laboratories, Japan Tobacco Inc, Takatsuki, Japan
| |
Collapse
|
63
|
Mulder T, Bobba S, Johnson K, Grandner JM, Wang W, Zhang C, Cai J, Choo EF, Khojasteh SC, Zhang D. Bioactivation of α, β-Unsaturated Carboxylic Acids Through Acyl Glucuronidation. Drug Metab Dispos 2020; 48:819-829. [PMID: 32616543 DOI: 10.1124/dmd.120.000096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/23/2020] [Indexed: 02/13/2025] Open
Abstract
After oral administration to monkeys of [14C]GDC-0810, an α,β-unsaturated carboxylic acid, unchanged parent and its acyl glucuronide metabolite, M6, were the major circulating drug-related components. In addition, greater than 50% of circulating radioactivity in plasma was found to be nonextractable 12 hours post-dose, suggesting possible covalent binding to plasma proteins. In the same study, one of the minor metabolites was a cysteine conjugate of M6 (M11) that was detected in plasma and excreta (urine and bile). The potential mechanism for the covalent binding to proteins was further investigated using in vitro methods. In incubations with glutathione (GSH) or cysteine (5 mM), GSH and cysteine conjugates of M6 were identified, respectively. The cysteine reaction was efficient with a half-life of 58.6 minutes (k react = 0.04 1/M per second). Loss of 176 Da (glucuronic acid) followed by 129 Da (glutamate) in mass fragmentation analysis of the GSH adduct of M6 (M13) suggested the glucuronic acid moiety was not modified. The conjugation of N-glucuronide M4 with cysteine in buffer was >1000-fold slower than with M6. Incubations of GDC-0810, M4, or M6 with monkey or human liver microsomes in the presence of NADPH and GSH did not produce any oxidative GSH adducts, and the respective substrates were qualitatively recovered. In silico analysis quantified the inherent reactivity differences between the glucuronide and its acid precursor. Collectively, these results show that acyl glucuronidation of α,β-unsaturated carboxylic acids can activate the compound toward reactivity with GSH, cysteine, or other biologically occurring thiols and should be considered during the course of drug discovery. SIGNIFICANCE STATEMENT: Acyl glucuronidation of the α,β-unsaturated carboxylic acid in GDC-0810 activates the conjugated alkene toward nucleophilic addition by glutathione or other reactive thiols. This is the first example that a bioactivation mechanism could lead to protein covalent binding to α,β-unsaturated carboxylic acid compounds.
Collapse
Affiliation(s)
- Teresa Mulder
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Sudheer Bobba
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Kevin Johnson
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Jessica M Grandner
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Wei Wang
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Chenghong Zhang
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Jingwei Cai
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Edna F Choo
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - S Cyrus Khojasteh
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| | - Donglu Zhang
- Drug Metabolism and Disposition (T.M., S.B., K.J., W.W., C.Z., J.C., E.F.C., S.C.K., D.Z.) and Discovery Chemistry (J.M.G.), Genentech, South San Francisco, California
| |
Collapse
|
64
|
Are pigs a suitable animal model for in vivo metabolism studies of new psychoactive substances? A comparison study using different in vitro/in vivo tools and U-47700 as model drug. Toxicol Lett 2020; 329:12-19. [PMID: 32380122 DOI: 10.1016/j.toxlet.2020.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 02/04/2023]
|
65
|
AlSheikh HMA, Sultan I, Kumar V, Rather IA, Al-Sheikh H, Tasleem Jan A, Haq QMR. Plant-Based Phytochemicals as Possible Alternative to Antibiotics in Combating Bacterial Drug Resistance. Antibiotics (Basel) 2020; 9:E480. [PMID: 32759771 PMCID: PMC7460449 DOI: 10.3390/antibiotics9080480] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/26/2020] [Accepted: 08/01/2020] [Indexed: 12/30/2022] Open
Abstract
The unprecedented use of antibiotics that led to development of resistance affect human health worldwide. Prescription of antibiotics imprudently and irrationally in different diseases progressed with the acquisition and as such development of antibiotic resistant microbes that led to the resurgence of pathogenic strains harboring enhanced armors against existing therapeutics. Compromised the treatment regime of a broad range of antibiotics, rise in resistance has threatened human health and increased the treatment cost of diseases. Diverse on metabolic, genetic and physiological fronts, rapid progression of resistant microbes and the lack of a strategic management plan have led researchers to consider plant-derived substances (PDS) as alternative or in complementing antibiotics against the diseases. Considering the quantitative characteristics of plant constituents that attribute health beneficial effects, analytical procedures for their isolation, characterization and phytochemical testing for elucidating ethnopharmacological effects has being worked out for employment in the treatment of different diseases. With an immense potential to combat bacterial infections, PDSs such as polyphenols, alkaloids and tannins, present a great potential for use, either as antimicrobials or as antibiotic resistance modifiers. The present study focuses on the mechanisms by which PDSs help overcome the surge in resistance, approaches for screening different phytochemicals, methods employed in the identification of bioactive components and their testing and strategies that could be adopted for counteracting the lethal consequences of multidrug resistance.
Collapse
Affiliation(s)
- Hana Mohammed Al AlSheikh
- Department of Prosthetic Dental Sciences, College of Dentistry, Kind Saud University, Riyadh P.O. BOX 145111, Saudi Arabia;
| | - Insha Sultan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdul Aziz University, Jeddah P.O. BOX 80200, Saudi Arabia;
| | - Hashem Al-Sheikh
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India
| | | |
Collapse
|
66
|
Simulation of the environmental degradation of diuron (herbicide) using electrochemistry coupled to high resolution mass spectrometry. Electrochim Acta 2020. [DOI: 10.1016/j.electacta.2020.136485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
67
|
Miller RR, Madeira M, Wood HB, Geissler WM, Raab CE, Martin IJ. Integrating the Impact of Lipophilicity on Potency and Pharmacokinetic Parameters Enables the Use of Diverse Chemical Space during Small Molecule Drug Optimization. J Med Chem 2020; 63:12156-12170. [DOI: 10.1021/acs.jmedchem.9b01813] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Randy R. Miller
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Maria Madeira
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Harold B. Wood
- Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Wayne M. Geissler
- Business Development & Licensing, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Conrad E. Raab
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Iain J. Martin
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
68
|
Temerdashev AZ, Dmitrieva EV. Methods for the Determination of Selective Androgen Receptor Modulators. JOURNAL OF ANALYTICAL CHEMISTRY 2020. [DOI: 10.1134/s1061934820070187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
69
|
Stevens LJ, Donkers JM, Dubbeld J, Vaes WHJ, Knibbe CAJ, Alwayn IPJ, van de Steeg E. Towards human ex vivo organ perfusion models to elucidate drug pharmacokinetics in health and disease. Drug Metab Rev 2020; 52:438-454. [DOI: 10.1080/03602532.2020.1772280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Lianne J. Stevens
- Department of Surgery, Division of Transplantation, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- The Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Joanne M. Donkers
- The Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Jeroen Dubbeld
- Department of Surgery, Division of Transplantation, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Wouter H. J. Vaes
- The Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Catherijne A. J. Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands
| | - Ian P. J. Alwayn
- Department of Surgery, Division of Transplantation, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Evita van de Steeg
- The Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
| |
Collapse
|
70
|
Valcourt DM, Kapadia CH, Scully MA, Dang MN, Day ES. Best Practices for Preclinical In Vivo Testing of Cancer Nanomedicines. Adv Healthc Mater 2020; 9:e2000110. [PMID: 32367687 PMCID: PMC7473451 DOI: 10.1002/adhm.202000110] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/17/2020] [Indexed: 01/06/2023]
Abstract
Significant advances have been made in the development of nanoparticles for cancer treatment in recent years. Despite promising results in preclinical animal models, cancer nanomedicines often fail in clinical trials. This failure rate could be reduced by defining stringent criteria for testing and quality control during the design and development stages, and by performing carefully planned preclinical studies in relevant animal models. This article discusses best practices for the evaluation of nanomedicines in murine tumor models. First, a recommended set of experiments to perform is introduced, including discussion of the types of data to collect during these studies. This is followed by an outline of various tumor models and their clinical relevance. Next, different routes of nanoparticle administration are overviewed, followed by a summary of important controls to include in in vivo studies of nanomedicine. Finally, animal welfare considerations are discussed, and an overview of the steps involved in achieving US Food and Drug Administration approval after animal studies are completed is provided. Researchers should use this report as a guideline for effective preclinical evaluation of cancer nanomedicine. As the community adopts best practices for in vivo testing, the rate of clinical translation of cancer nanomedicines is likely to improve.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Chintan H Kapadia
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Mackenzie A Scully
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, DE, 19713, USA
| |
Collapse
|
71
|
Lau J, Rousseau E, Kwon D, Lin KS, Bénard F, Chen X. Insight into the Development of PET Radiopharmaceuticals for Oncology. Cancers (Basel) 2020; 12:E1312. [PMID: 32455729 PMCID: PMC7281377 DOI: 10.3390/cancers12051312] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022] Open
Abstract
While the development of positron emission tomography (PET) radiopharmaceuticals closely follows that of traditional drug development, there are several key considerations in the chemical and radiochemical synthesis, preclinical assessment, and clinical translation of PET radiotracers. As such, we outline the fundamentals of radiotracer design, with respect to the selection of an appropriate pharmacophore. These concepts will be reinforced by exemplary cases of PET radiotracer development, both with respect to their preclinical and clinical evaluation. We also provide a guideline for the proper selection of a radionuclide and the appropriate labeling strategy to access a tracer with optimal imaging qualities. Finally, we summarize the methodology of their evaluation in in vitro and animal models and the road to clinical translation. This review is intended to be a primer for newcomers to the field and give insight into the workflow of developing radiopharmaceuticals.
Collapse
Affiliation(s)
- Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Etienne Rousseau
- Department of Nuclear Medicine and Radiobiology, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Daniel Kwon
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
72
|
|
73
|
Yong YF, Tan SC, Liew MWO, Yaacob NS. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) method development for screening of potential tamoxifen-drug/herb interaction via in vitro cytochrome P450 inhibition assay. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122148. [PMID: 32416571 DOI: 10.1016/j.jchromb.2020.122148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/19/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022]
Abstract
Screening for potential drug-drug interaction (DDI) or herb-drug interaction (HDI) using in vitro cytochrome P450 inhibition (IVCI) assays requires robust analytical methods with high sensitivity and reproducibility. Utilization of liquid chromatography-mass spectrometry (LC-MS) for analyte quantification is often hampered by the presence of non-volatile IVCI sample buffer constituents that often results in ion suppression. In this study, to enable screening of drug interactions involving tamoxifen (TAM) metabolism using IVCI-LC-MS/MS, a liquid-liquid extraction (LLE) method was developed and optimized for sample clean-up. Utilization of chloroform as extraction solvent and adjustment of sample pH to 11 was found to result in satisfactory recovery (>70%) and low ion suppression (<19%). A LC-MS/MS method was subsequently developed and validated for simultaneous quantification of major TAM metabolites, such as N-desmethyltamoxifen (NDT), endoxifen (EDF) and 4-hydroxytamoxifen (HTF) to enable IVCI sample analysis. Satisfactory separation of E-/Z-isomers of endoxifen with peak resolution (Rs) of 1.9 was achieved. Accuracy and precision of the method was verified within the linear range of 0-50 ng/mL for NDT, 0-25 ng/mL for HTF and 0-25 ng/mL for EDF (E/Z isomers). Inhibitory potency (IC50, Ki and mode of inhibition) of known CYP inhibitors and Strobilanthes crispus extract was then evaluated using the validated method. In summary, the results demonstrated applicability of the developed LLE and validated LC-MS/MS method for in vitro screening of DDI and HDI involving TAM metabolism.
Collapse
Affiliation(s)
- Y F Yong
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - S C Tan
- Usains Biomics Laboratory Testing Services Sdn. Bhd., Universiti Sains Malaysia, Minden 11800, Penang, Malaysia
| | - Mervyn W O Liew
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - N S Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
74
|
Ramaiahgari SC, Auerbach SS, Saddler TO, Rice JR, Dunlap PE, Sipes NS, DeVito MJ, Shah RR, Bushel PR, Merrick BA, Paules RS, Ferguson SS. The Power of Resolution: Contextualized Understanding of Biological Responses to Liver Injury Chemicals Using High-throughput Transcriptomics and Benchmark Concentration Modeling. Toxicol Sci 2020; 169:553-566. [PMID: 30850835 DOI: 10.1093/toxsci/kfz065] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prediction of human response to chemical exposures is a major challenge in both pharmaceutical and toxicological research. Transcriptomics has been a powerful tool to explore chemical-biological interactions, however, limited throughput, high-costs, and complexity of transcriptomic interpretations have yielded numerous studies lacking sufficient experimental context for predictive application. To address these challenges, we have utilized a novel high-throughput transcriptomics (HTT) platform, TempO-Seq, to apply the interpretive power of concentration-response modeling with exposures to 24 reference compounds in both differentiated and non-differentiated human HepaRG cell cultures. Our goals were to (1) explore transcriptomic characteristics distinguishing liver injury compounds, (2) assess impacts of differentiation state of HepaRG cells on baseline and compound-induced responses (eg, metabolically-activated), and (3) identify and resolve reference biological-response pathways through benchmark concentration (BMC) modeling. Study data revealed the predictive utility of this approach to identify human liver injury compounds by their respective BMCs in relation to human internal exposure plasma concentrations, and effectively distinguished drug analogs with varied associations of human liver injury (eg, withdrawn therapeutics trovafloxacin and troglitazone). Impacts of cellular differentiation state (proliferated vs differentiated) were revealed on baseline drug metabolizing enzyme expression, hepatic receptor signaling, and responsiveness to metabolically-activated toxicants (eg, cyclophosphamide, benzo(a)pyrene, and aflatoxin B1). Finally, concentration-response modeling enabled efficient identification and resolution of plausibly-relevant biological-response pathways through their respective pathway-level BMCs. Taken together, these findings revealed HTT paired with differentiated in vitro liver models as an effective tool to model, explore, and interpret toxicological and pharmacological interactions.
Collapse
Affiliation(s)
- Sreenivasa C Ramaiahgari
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Scott S Auerbach
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Trey O Saddler
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Julie R Rice
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Paul E Dunlap
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Nisha S Sipes
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Michael J DeVito
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Ruchir R Shah
- Sciome, LLC, Research Triangle Park, Durham, North Carolina 27709
| | - Pierre R Bushel
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709.,Division of Intramural Research, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Bruce A Merrick
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Richard S Paules
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Stephen S Ferguson
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| |
Collapse
|
75
|
Manier SK, Wagmann L, Flockerzi V, Meyer MR. Toxicometabolomics of the new psychoactive substances α-PBP and α-PEP studied in HepaRG cell incubates by means of untargeted metabolomics revealed unexpected amino acid adducts. Arch Toxicol 2020; 94:2047-2059. [PMID: 32313995 PMCID: PMC7303098 DOI: 10.1007/s00204-020-02742-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/06/2020] [Indexed: 01/06/2023]
Abstract
Toxicometabolomics, essentially applying metabolomics to toxicology of endogenous compounds such as drugs of abuse or new psychoactive substances (NPS), can be investigated by using different in vitro models and dedicated metabolomics techniques to enhance the number of relevant findings. The present study aimed to study the toxicometabolomics of the two NPS α-pyrrolidinobutiophenone (1-phenyl-2-(pyrrolidin-1-yl)butan-1-one, α-PBP) and α-pyrrolidinoheptaphenone (1-phenyl-2-(pyrrolidin-1-yl)heptan-1-one, α-PEP, PV8) in HepaRG cell line incubates. Evaluation was performed using reversed-phase and normal-phase liquid chromatography coupled with high-resolution mass spectrometry in positive and negative ionization mode, respectively, to analyze cells and cell media. Statistical evaluation was performed using one-way ANOVA, principal component discriminant function analysis, as well as hierarchical clustering. In general, the analysis of cells did not mainly reveal any features, but the parent compounds of the drugs of abuse. For α-PBP an increase in N-methylnicotinamide was found, which may indicate hepatotoxic potential of the substance. After analysis of cell media, significant features led to the identification of several metabolites of both compounds. Amino acid adducts with glycine and alanine were found, and these have not been described in any study before and are likely to appear in vivo. Additionally, significant changes in the metabolism of cholesterol were revealed after incubation with α-PEP. In summary, the application of metabolomics techniques after HepaRG cells exposure to NPS did not lead to an increased number of identified drug metabolites compared to previously published studies, but gave a wider perspective on the physiological effect of the investigated compounds on human liver cells.
Collapse
Affiliation(s)
- Sascha K Manier
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421, Homburg, Germany
| | - Lea Wagmann
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421, Homburg, Germany
| | - Veit Flockerzi
- Department of Experimental and Clinical Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421, Homburg, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
76
|
Manier SK, Niedermeier S, Schäper J, Meyer MR. Use of UPLC-HRMS/MS for In Vitro and In Vivo Metabolite Identification of Three Methylphenidate-derived New Psychoactive Substances. J Anal Toxicol 2020; 44:156-162. [PMID: 31355413 DOI: 10.1093/jat/bkz052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/15/2019] [Accepted: 06/09/2019] [Indexed: 11/14/2022] Open
Abstract
The distribution of so-called new psychoactive substances (NPS) as substitute for common drug of abuse was steadily increasing in the last years, but knowledge about their toxicodynamic and toxicokinetic properties is lacking. However, a comprehensive knowledge of their toxicokinetics, particularly their metabolism, is crucial for developing reliable screening procedures and to verify their intake, e.g., in case of intoxications. The aim of this study was therefore to tentatively identify the metabolites of the methylphenidate-derived NPS isopropylphenidate (isopropyl 2-phenyl-2-(2-piperidyl) acetate, IPH), 4-fluoromethylphenidate (methyl 2-(4-fluorophenyl)-2-(piperidin-2-yl) acetate, 4-FMPH) and 3,4-dichloromethylphenidate (methyl 2-(3,4-dichlorophenyl)-2-(piperidin-2-yl) acetate, 3,4-CTMP) using different in vivo and in vitro techniques and ultra-high performance liquid chromatography-high-resolution mass spectrometry (UHPLC-HRMS/MS). Urine samples of male rats were analyzed, and the transfer to human metabolism was done by using pooled human S9 fraction (pS9), which contains the microsomal fraction of liver homogenisate as well as its cytosol. UHPLC-HRMS/MS analysis of rat urine revealed 17 metabolites for IPH (14 phase I and 3 phase II metabolites), 13 metabolites were found for 4-FMPH (12 phase I metabolites and 1 phase II metabolite) and 7 phase I metabolites and no phase II metabolites were found for 3,4-CTMP. pS9 incubations additionally indicated that all investigated substances were primarily hydrolyzed, resulting in the corresponding carboxy metabolites. Finally, these carboxy metabolites should be used as additional analytical targets besides the parent compounds for comprehensive mass spectrometry-based screening procedures.
Collapse
Affiliation(s)
- Sascha K Manier
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Sophia Niedermeier
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Jan Schäper
- State Bureau of Criminal Investigation Bavaria, 80636 München, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| |
Collapse
|
77
|
Zhu LJ, Chen L, Bai CF, Wu AG, Liang SC, Huang FH, Cao SS, Yang L, Zou WJ, Wu JM. A rapid and sensitive UHPLC-MS/MS method for the determination of ziyuglycoside I and its application in a preliminary pharmacokinetic study in healthy and leukopenic rats. Biomed Pharmacother 2020; 123:109756. [DOI: 10.1016/j.biopha.2019.109756] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/26/2022] Open
|
78
|
Kendrick J, Stow R, Ibbotson N, Adjin-Tettey G, Murphy B, Bailey G, Miller J, Helleberg H, Finderup Grove M, Øvlisen K, Hansen JJ. A novel welfare and scientific approach to conducting dog metabolism studies allowing dogs to be pair housed. Lab Anim 2020; 54:588-598. [PMID: 32063096 DOI: 10.1177/0023677220905330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolism cages are designed to conduct absorption, distribution, metabolism and excretion (ADME) studies, enabling an 'excretion balance' scientific objective to be met. Historically, the design of dog metabolism cages has involved single housing. This type of housing has limitations for normal social behaviours and has been largely unchanged for 25-30 years. Improving animal welfare is a focus area for the authorities as well as the industry throughout the European Union. A collaboration was developed between Novo Nordisk and Covance to enhance the design of metabolism cages, allowing dogs to be pair housed. The purpose of the study was to compare excretion balance data from pair-housed and singly housed dogs in order to demonstrate that conducting excretion balance studies with a pair-housing design improves animal welfare without compromising the scientific integrity of the study. A radiolabelled test compound, [14C]-Quetiapine, was selected for this investigation based on its excretion profile. The assessment of the dogs' stress levels was investigated by measuring the levels of serum cortisol as an indicative biomarker. Results were inconclusive due to large variations in cortisol levels. However, dogs appeared calmer in the pair-housing setting. The overall mean recovery (±standard deviation) for pair-housed animals (94.0 ± 0.66% of the dose) was equivalent to that from singly housed dogs (93.0 ± 2.29%). Based on these data, we conclude that pair housing of dogs for future metabolism ADME studies does not compromise the scientific integrity, and therefore is a major progression in the design of these studies, enhancing welfare.
Collapse
Affiliation(s)
| | - Ruth Stow
- Covance Laboratories Ltd, North Yorkshire, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Liu H, Bolleddula J, Nichols A, Tang L, Zhao Z, Prakash C. Metabolism of bioconjugate therapeutics: why, when, and how? Drug Metab Rev 2020; 52:66-124. [PMID: 32045530 DOI: 10.1080/03602532.2020.1716784] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioconjugation of therapeutic agents has been used as a selective drug delivery platform for many therapeutic areas. Bioconjugates are prepared by the covalent linkage of active compounds (small or large molecule) to a carrier molecule (lipids, proteins, peptides, carbohydrates, and polymers) through a chemical linker. The linkage of the active component to a carrier molecule enhances the therapeutic window through a targeted delivery and by reducing toxicity. Bioconjugates also possess improved pharmacokinetic properties such as a long half-life, increased stability, and cleavage by intracellular enzymes/environment. However, premature cleavage of the bioconjugates and the resulting metabolites/catabolites may produce undesirable toxic effects and, hence, it is critical to understand cleavage mechanisms, metabolism of bioconjugates, and translatability to human in the discovery stages. This article provides a comprehensive overview of linker cleavage pathways and catabolism/metabolism of antibody-drug conjugates, glycoconjugates, polymer-drug conjugates, lipid-drug conjugates, folate-targeted small molecule-drug conjugates, and drug-drug conjugates.
Collapse
Affiliation(s)
- Hanlan Liu
- KSQ Therapeutics Inc., Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
80
|
Shojaeimehr T, Tasbihi M, Acharjya A, Thomas A, Schomäcker R, Schwarze M. Impact of operating conditions for the continuous-flow degradation of diclofenac with immobilized carbon nitride photocatalysts. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2019.112182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
81
|
Assirey E, Alsaggaf A, Naqvi A, Moussa Z, Okasha RM, Afifi TH, Abd-El-Aziz AS. Synthesis, Biological Assessment, and Structure Activity Relationship Studies of New Flavanones Embodying Chromene Moieties. Molecules 2020; 25:molecules25030544. [PMID: 32012737 PMCID: PMC7037824 DOI: 10.3390/molecules25030544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/15/2020] [Accepted: 01/26/2020] [Indexed: 02/04/2023] Open
Abstract
Novel flavanones that incorporate chromene motifs are synthesized via a one-step multicomponent reaction. The structures of the new chromenes are elucidated by using IR, 1H-NMR, 13C-NMR, 1H-1H COSY, HSQC, HMBC, and elemental analysis. The new compounds are screened for their in vitro antimicrobial and cytotoxic activities. The antimicrobial properties are investigated and established against seven human pathogens, employing the agar well diffusion method and the minimum inhibitory concentrations. A majority of the assessed derivatives are found to exhibit significant antimicrobial activities against most bacterial strains, in comparison to standard reference drugs. Moreover, their cytotoxicity is appraised against four different human carcinoma cell lines: human colon carcinoma (HCT-116), human hepatocellular carcinoma (HepG-2), human breast adenocarcinoma (MCF-7), and adenocarcinoma human alveolar basal epithelial cell (A-549). All the desired compounds are subjected to in-silico studies, forecasting their drug likeness, bioactivity, and the absorption, distribution, metabolism, and excretion (ADME) properties prior to their synthetic assembly. The in-silico molecular docking evaluation of all the targeted derivatives is undertaken on gyrase B and the cyclin-dependent kinase. The in-silico predicted outcomes were endorsed by the in vitro studies.
Collapse
Affiliation(s)
- Eman Assirey
- Department of Chemistry, Taibah University, Madinah 30002, Saudi Arabia; (E.A.); (A.A.); (A.N.); (R.M.O.)
- Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PEI C1A 4P3, Canada
| | - Azhaar Alsaggaf
- Department of Chemistry, Taibah University, Madinah 30002, Saudi Arabia; (E.A.); (A.A.); (A.N.); (R.M.O.)
- Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PEI C1A 4P3, Canada
| | - Arshi Naqvi
- Department of Chemistry, Taibah University, Madinah 30002, Saudi Arabia; (E.A.); (A.A.); (A.N.); (R.M.O.)
| | - Ziad Moussa
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain 15551, UAE;
| | - Rawda M. Okasha
- Department of Chemistry, Taibah University, Madinah 30002, Saudi Arabia; (E.A.); (A.A.); (A.N.); (R.M.O.)
| | - Tarek H. Afifi
- Department of Chemistry, Taibah University, Madinah 30002, Saudi Arabia; (E.A.); (A.A.); (A.N.); (R.M.O.)
| | - Alaa S. Abd-El-Aziz
- Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PEI C1A 4P3, Canada
- Correspondence: ; Tel.: +1-(902)-566-0400
| |
Collapse
|
82
|
Oxidative metabolism of typical phenolic compounds of Danshen by electrochemistry coupled to quadrupole time-of-flight tandem mass spectrometry. Food Chem 2020; 315:126270. [PMID: 32028199 DOI: 10.1016/j.foodchem.2020.126270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/16/2020] [Accepted: 01/19/2020] [Indexed: 11/23/2022]
Abstract
An electrochemistry coupled to online quadrupole time-of-flight tandem mass spectrometry (EC/Q-TOF/MS) was applied to investigate the oxidative transformation and metabolic pathway of five phenolic acids in Danshen sample. Simulation of the phase I oxidative metabolism was carried out in an electrochemical reactor equipped with a glassy carbon working electrode. The phase II reactivity of the generated oxidative products towards biomolecules (such as glutathione) was investigated by ways of covalent adduct formation experiments. The results obtained by EC/MS were compared with well-known in vitro studies by conducting rat liver microsome incubations. Structures of the electrochemically produced metabolites were identified by accurate mass measurement and previously results in vivo metabolites. It was indicated that the electrochemical oxidation was in good accordance with similar products found in vivo experiments. In conclusion, this work confirmed that EC/Q-TOF/MS was a promising analytical tool in the prediction of metabolic transformations of functional foods.
Collapse
|
83
|
Lu C, Di L. In vitro
and
in vivo
methods to assess pharmacokinetic drug– drug interactions in drug discovery and development. Biopharm Drug Dispos 2020; 41:3-31. [DOI: 10.1002/bdd.2212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Chuang Lu
- Department of DMPKSanofi Company Waltham MA 02451
| | - Li Di
- Pharmacokinetics, Dynamics and MetabolismPfizer Worldwide Research & Development Groton CT 06340
| |
Collapse
|
84
|
Bulutoglu B, Mert S, Rey-Bedón C, Deng SL, Yarmush ML, Usta OB. Rapid maturation of the hepatic cell line Huh7 via CDK inhibition for PXR dependent CYP450 metabolism and induction. Sci Rep 2019; 9:15848. [PMID: 31676845 PMCID: PMC6825149 DOI: 10.1038/s41598-019-52174-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/12/2019] [Indexed: 12/26/2022] Open
Abstract
CYP3A4, a cytochrome P450 enzyme regulated by the nuclear receptor PXR, is involved in most of the drug metabolizing pathways. Studying the regulation/induction of CYP3A4 and PXR is critical in toxicology and drug-drug interaction (DDI) studies. Primary human hepatocytes constitute the preferred in vitro platform for drug development efforts. However, they are expensive, scarce and heterogeneous. Hepatic cell lines, such as Huh7, could provide a cost-effective alternative, however, they express negligible amounts of CYP450s and PXR. In this study, we show that dinaciclib, a potent cyclin dependent kinase inhibitor, significantly increases the basal CYP3A4 and PXR levels in 24 hours. We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. More importantly, through a direct demonstration using amiodarone and rifampicin as model drugs, we showed that matured Huh7s present a suitable platform for DDI studies.
Collapse
Affiliation(s)
- Beyza Bulutoglu
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Safak Mert
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Camilo Rey-Bedón
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Sarah L Deng
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA.
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| | - O Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA.
| |
Collapse
|
85
|
Sibinovska N, Žakelj S, Kristan K. Suitability of RPMI 2650 cell models for nasal drug permeability prediction. Eur J Pharm Biopharm 2019; 145:85-95. [PMID: 31639418 DOI: 10.1016/j.ejpb.2019.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/01/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022]
Abstract
The RPMI 2650 cell line has been a subject of evaluation as a physiological and pharmacological model of the nasal epithelial barrier. However, its suitability for drug permeability assays has not yet been established on a sufficiently large set of model drugs. We investigated two RPMI 2650 cell models (air-liquid and liquid-liquid) for nasal drug permeability determination by adopting the most recent regulatory guidelines on showing suitability of in vitro permeability methods for drug permeability classification. The permeability of 23 model drugs and several zero permeability markers across the cell models was assessed. The functional expression of two efflux transporters P-glycoprotein (P-gp) and Breast Cancer Resistant Protein (BCRP) was shown to be negligible by bidirectional transport studies using appropriate transporter substrates and inhibitors. The model drug permeability determined in the two RPMI 2650 cell models was correlated with the fully differentiated nasal epithelial model (MucilAir™). Additionally, correlations between the drug permeability in the investigated cell models and the ones determined in the Caco-2 cells and isolated rat jejunum were established. In conclusion, the air-liquid RPMI 2650 cell model is a promising pharmacological model of the nasal epithelial barrier and is much more suitable than the liquid-liquid model for nasal drug permeability prediction.
Collapse
Affiliation(s)
- Nadica Sibinovska
- University of Ljubljana, Faculty of Pharmacy, Chair of Biopharmaceutics and Pharmacokinetics, Aškerčeva c. 7, SI- 1000 Ljubljana, Slovenia
| | - Simon Žakelj
- University of Ljubljana, Faculty of Pharmacy, Chair of Biopharmaceutics and Pharmacokinetics, Aškerčeva c. 7, SI- 1000 Ljubljana, Slovenia
| | - Katja Kristan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; Lek Pharmaceuticals, d.d., Sandoz Development Center Slovenia, Verovškova 57, 1526 Ljubljana, Slovenia.
| |
Collapse
|
86
|
Chen Z, Huang J, Li L. Recent advances in mass spectrometry (MS)-based glycoproteomics in complex biological samples. Trends Analyt Chem 2019; 118:880-892. [PMID: 31579312 PMCID: PMC6774629 DOI: 10.1016/j.trac.2018.10.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Protein glycosylation plays a key role in various biological processes and disease-related pathological progression. Mass spectrometry (MS)-based glycoproteomics is a powerful approach that provides a system-wide profiling of the glycoproteome in a high-throughput manner. There have been numerous significant technological advances in this field, including improved glycopeptide enrichment, hybrid fragmentation techniques, emerging specialized software packages, and effective quantitation strategies, as well as more dedicated workflows. With increasingly sophisticated glycoproteomics tools on hand, researchers have extensively adapted this approach to explore different biological systems both in terms of in-depth glycoproteome profiling and comparative glycoproteome analysis. Quantitative glycoproteomics enables researchers to discover novel glycosylation-based biomarkers in various diseases with potential to offer better sensitivity and specificity for disease diagnosis. In this review, we present recent methodological developments in MS-based glycoproteomics and highlight its utility and applications in answering various questions in complex biological systems.
Collapse
Affiliation(s)
- Zhengwei Chen
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Junfeng Huang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53705, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
87
|
Metabolic stability and its role in the discovery of new chemical entities. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2019; 69:345-361. [PMID: 31259741 DOI: 10.2478/acph-2019-0024] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/29/2018] [Indexed: 01/19/2023]
Abstract
Determination of metabolic profiles of new chemical entities is a key step in the process of drug discovery, since it influences pharmacokinetic characteristics of therapeutic compounds. One of the main challenges of medicinal chemistry is not only to design compounds demonstrating beneficial activity, but also molecules exhibiting favourable pharmacokinetic parameters. Chemical compounds can be divided into those which are metabolized relatively fast and those which undergo slow biotransformation. Rapid biotransformation reduces exposure to the maternal compound and may lead to the generation of active, non-active or toxic metabolites. In contrast, high metabolic stability may promote interactions between drugs and lead to parent compound toxicity. In the present paper, issues of compound metabolic stability will be discussed, with special emphasis on its significance, in vitro metabolic stability testing, dilemmas regarding in vitro-in vivo extrapolation of the results and some aspects relating to different preclinical species used in in vitro metabolic stability assessment of compounds.
Collapse
|
88
|
Kumari J, Teotia AK, Karande AA, Kumar A. A minimally-invasive cryogel based approach for the development of human ectopic liver in a mouse model. J Biomed Mater Res B Appl Biomater 2019; 108:1022-1032. [PMID: 31397074 DOI: 10.1002/jbm.b.34454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/10/2019] [Accepted: 07/09/2019] [Indexed: 11/09/2022]
Abstract
Human liver tissue is preferable over nonhuman liver tissue for preclinical drug screening, as the former can better predict side effects specific to humans. However, due to limited supply and ethical issues with human liver tissue, it is desirable to develop an animal model having functional human liver tissue. In this study, we have established an ectopic functional human liver tissue in a mouse model, using a minimally-invasive method. Firstly, a human liver tissue mass using HepG2 cells and poly(N-isopropylacrylamide) (PNIPAAm) incorporated poly(ethylene glycol)-alginate-gelatin (PAG) cryogel matrix was developed in vitro. It was later implanted in mouse peritoneal cavity using a 16 G needle. Viscoelastic nature along with low Young's modulus provided injectable properties to the cryogel. We confirmed minimal cell loss/death while injecting. Further, by in vivo study efficacy of both injectable and surgical implantation approaches were compared. No significant difference in terms of cell infiltration, human serum albumin (HSA) secretion and enzyme activity confirmed efficacy. This model developed using a minimally-invasive approach can overcome the limitations of surgical implantation due to its cost effective and user friendly nature.
Collapse
Affiliation(s)
- Jyoti Kumari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| | - Arun K Teotia
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| | - Anjali A Karande
- Department of Biochemistry, Indian Institute of Sciences, Bangalore, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| |
Collapse
|
89
|
Kotthoff L, Lisec J, Schwerdtle T, Koch M. Prediction of Transformation Products of Monensin by Electrochemistry Compared to Microsomal Assay and Hydrolysis. Molecules 2019; 24:molecules24152732. [PMID: 31357593 PMCID: PMC6696283 DOI: 10.3390/molecules24152732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 11/16/2022] Open
Abstract
The knowledge of transformation pathways and identification of transformation products (TPs) of veterinary drugs is important for animal health, food, and environmental matters. The active agent Monensin (MON) belongs to the ionophore antibiotics and is widely used as a veterinary drug against coccidiosis in broiler farming. However, no electrochemically (EC) generated TPs of MON have been described so far. In this study, the online coupling of EC and mass spectrometry (MS) was used for the generation of oxidative TPs. EC-conditions were optimized with respect to working electrode material, solvent, modifier, and potential polarity. Subsequent LC/HRMS (liquid chromatography/high resolution mass spectrometry) and MS/MS experiments were performed to identify the structures of derived TPs by a suspected target analysis. The obtained EC-results were compared to TPs observed in metabolism tests with microsomes and hydrolysis experiments of MON. Five previously undescribed TPs of MON were identified in our EC/MS based study and one TP, which was already known from literature and found by a microsomal assay, could be confirmed. Two and three further TPs were found as products in microsomal tests and following hydrolysis, respectively. We found decarboxylation, O-demethylation and acid-catalyzed ring-opening reactions to be the major mechanisms of MON transformation.
Collapse
Affiliation(s)
- Lisa Kotthoff
- Department of Analytical Chemistry and Reference Materials, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Potsdam, Germany
| | - Jan Lisec
- Department of Analytical Chemistry and Reference Materials, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Potsdam, Germany
| | - Matthias Koch
- Department of Analytical Chemistry and Reference Materials, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany.
| |
Collapse
|
90
|
Characterization of Porcine Hepatic and Intestinal Drug Metabolizing CYP450: Comparison with Human Orthologues from A Quantitative, Activity and Selectivity Perspective. Sci Rep 2019; 9:9233. [PMID: 31239454 PMCID: PMC6592956 DOI: 10.1038/s41598-019-45212-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Over the past two decades, the pig has gained attention as a potential model for human drug metabolism. Cytochrome P450 enzymes (CYP450), a superfamily of biotransformation enzymes, are pivotal in drug metabolism. Porcine CYP450 has been demonstrated to convert typical substrates of human CYP450. Nevertheless, knowledge and insight into porcine CYP450 quantity and substrate selectivity is scant, especially regarding intestinal CYP450. The current study aimed to map the quantities of hepatic and intestinal CYP450 in the conventional pig by using a proteomic approach. Moreover, the selectivity of the six most common used probe substrates (phenacetin, coumarin, midazolam, tolbutamide, dextromethorphan, and chlorzoxazone) for drug metabolizing enzyme subfamilies (CYP1A, CYP2A, CYP3A, CYP2C, CYP2D and CYP2E respectively), was investigated. Hepatic relative quantities were 4% (CYP1A), 31% (CYP2A), 14% (CYP3A), 10% (CYP2C), 28% (CYP2D) and 13% (CYP2E), whereas for the intestine only duodenal CYP450 could be determined with 88% for CYP3A and 12% for CYP2C. Furthermore, the results indicate that coumarin (CYP2A), midazolam (CYP3A), tolbutamide (CYP2C), and dextromethorphan (CYP2D) are as selective for porcine as for human CYP450. However, phenacetin (CYP1A2) and chlorzoxazone (CYP2E1) are less selective for the specific enzyme, despite similarities in selectivity towards the different enzymes involved compared to humans.
Collapse
|
91
|
Schneider D, Oskamp A, Holschbach M, Neumaier B, Bauer A, Bier D. Relevance of In Vitro Metabolism Models to PET Radiotracer Development: Prediction of In Vivo Clearance in Rats from Microsomal Stability Data. Pharmaceuticals (Basel) 2019; 12:ph12020057. [PMID: 31013984 PMCID: PMC6631687 DOI: 10.3390/ph12020057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 01/03/2023] Open
Abstract
The prediction of in vivo clearance from in vitro metabolism models such as liver microsomes is an established procedure in drug discovery. The potentials and limitations of this approach have been extensively evaluated in the pharmaceutical sector; however, this is not the case for the field of positron emission tomography (PET) radiotracer development. The application of PET radiotracers and classical drugs differs greatly with regard to the amount of substance administered. In typical PET imaging sessions, subnanomolar quantities of the radiotracer are injected, resulting in body concentrations that cannot be readily simulated in analytical assays. This raises concerns regarding the predictability of radiotracer clearance from in vitro data. We assessed the accuracy of clearance prediction for three prototypical PET radiotracers developed for imaging the A1 adenosine receptor (A1AR). Using the half-life (t1/2) approach and physiologically based scaling, in vivo clearance in the rat model was predicted from microsomal stability data. Actual clearance could be accurately predicted with an average fold error (AFE) of 0.78 and a root mean square error (RMSE) of 1.6. The observed slight underprediction (1.3-fold) is in accordance with the prediction accuracy reported for classical drugs. This result indicates that the prediction of radiotracer clearance is possible despite concentration differences of more than three orders of magnitude between in vitro and in vivo conditions. Consequently, in vitro metabolism models represent a valuable tool for PET radiotracer development.
Collapse
Affiliation(s)
- Daniela Schneider
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Angela Oskamp
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Marcus Holschbach
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Andreas Bauer
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
- Neurological Department, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany.
| | - Dirk Bier
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| |
Collapse
|
92
|
Mercier C, Jacqueroux E, He Z, Hodin S, Constant S, Perek N, Boudard D, Delavenne X. Pharmacological characterization of the 3D MucilAir™ nasal model. Eur J Pharm Biopharm 2019; 139:186-196. [PMID: 30951820 DOI: 10.1016/j.ejpb.2019.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/18/2019] [Accepted: 04/02/2019] [Indexed: 01/01/2023]
Abstract
The preclinical evaluation of nasally administered drug candidates requires screening studies based on in vitro models of the nasal mucosa. The aim of this study was to evaluate the morpho-functional characteristics of the 3D MucilAir™ nasal model with a pharmacological focus on [ATP]-binding cassette (ABC) efflux transporters. We initially performed a phenotypic characterization of the MucilAir™ model and assessed its barrier properties by immunofluorescence (IF), protein mass spectrometry and examination of histological sections. We then focused on the functional expression of the ABC transporters P-glycoprotein (P-gp), multidrug resistance associated protein (MRP)1, MRP2 and breast cancer resistance protein (BCRP) in bidirectional transport experiments. The MucilAir™ model comprises a tight, polarized, pseudo-stratified nasal epithelium composed of fully differentiated ciliated, goblet and basal cells. These ABC transporters were all expressed by the cell membranes. P-gp and BCRP were both functional and capable of actively effluxing substrates. The MucilAir™ model could consequently represent a potent tool for evaluating the interaction of nasally administered drugs with ABC transporters.
Collapse
Affiliation(s)
- Clément Mercier
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France.
| | - Elodie Jacqueroux
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France.
| | - Zhiguo He
- Université de Lyon, Saint-Etienne F-42023, France; Laboratoire de biologie, d'ingénierie et d'imagerie de la greffe de cornée, BiiGC, EA2521 Saint-Etienne, France.
| | - Sophie Hodin
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France.
| | - Samuel Constant
- Epithelix Sàrl, 14 chemin des aulx, CH-1228 Plan-les-Ouates, Geneva, Switzerland.
| | - Nathalie Perek
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France.
| | - Delphine Boudard
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France; UF6725 Cytologie et Histologie Rénale, CHU de Saint-Etienne, Saint-Etienne, France.
| | - Xavier Delavenne
- INSERM, U1059, Dysfonction Vasculaire et Hémostase, Saint-Etienne, France; Université de Lyon, Saint-Etienne F-42023, France; Laboratoire de Pharmacologie Toxicologie Gaz du sang, CHU de Saint-Etienne, Saint-Etienne, France.
| |
Collapse
|
93
|
Li Z, Di L, Maurer TS. Theoretical Considerations for Direct Translation of Unbound Liver-to-Plasma Partition Coefficient from In Vitro to In Vivo. AAPS JOURNAL 2019; 21:43. [DOI: 10.1208/s12248-019-0314-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/26/2019] [Indexed: 12/25/2022]
|
94
|
Physiologically-Based Pharmacokinetic Modeling for Drug-Drug Interactions of Procainamide and N-Acetylprocainamide with Cimetidine, an Inhibitor of rOCT2 and rMATE1, in Rats. Pharmaceutics 2019; 11:pharmaceutics11030108. [PMID: 30845766 PMCID: PMC6470842 DOI: 10.3390/pharmaceutics11030108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 01/11/2023] Open
Abstract
Previous observations demonstrated that cimetidine decreased the clearance of procainamide (PA) and/or N-acetylprocainamide (NAPA; the primary metabolite of PA) resulting in the increased systemic exposure and the decrease of urinary excretion. Despite an abundance of in vitro and in vivo data regarding pharmacokinetic interactions between PA/NAPA and cimetidine, however, a mechanistic approach to elucidate these interactions has not been reported yet. The primary objective of this study was to construct a physiological model that describes pharmacokinetic interactions between PA/NAPA and cimetidine, an inhibitor of rat organic cation transporter 2 (rOCT2) and rat multidrug and toxin extrusion proteins (rMATE1), by performing extensive in vivo and in vitro pharmacokinetic studies for PA and NAPA performed in the absence or presence of cimetidine in rats. When a single intravenous injection of PA HCl (10 mg/kg) was administered to rats, co-administration of cimetidine (100 mg/kg) significantly increased systemic exposure and decreased the systemic (CL) and renal (CLR) clearance of PA, and reduced its tissue distribution. Similarly, cimetidine significantly decreased the CLR of NAPA formed by the metabolism of PA and increased the AUC of NAPA. Considering that these drugs could share similar renal secretory pathways (e.g., via rOCT2 and rMATE1), a physiologically-based pharmacokinetic (PBPK) model incorporating semi-mechanistic kidney compartments was devised to predict drug-drug interactions (DDIs). Using our proposed PBPK model, DDIs between PA/NAPA and cimetidine were successfully predicted for the plasma concentrations and urinary excretion profiles of PA and NAPA observed in rats. Moreover, sensitivity analyses of the pharmacokinetics of PA and NAPA showed the inhibitory effects of cimetidine via rMATE1 were probably important for the renal elimination of PA and NAPA in rats. The proposed PBPK model may be useful for understanding the mechanisms of interactions between PA/NAPA and cimetidine in vivo.
Collapse
|
95
|
Docci L, Parrott N, Krähenbühl S, Fowler S. Application of New Cellular and Microphysiological Systems to Drug Metabolism Optimization and Their Positioning Respective to In Silico Tools. SLAS DISCOVERY 2019; 24:523-536. [PMID: 30817893 DOI: 10.1177/2472555219831407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New cellular model systems for drug metabolism applications, such as advanced 2D liver co-cultures, spheroids, and microphysiological systems (MPSs), offer exciting opportunities to reproduce human biology more closely in vitro with the aim of improving predictions of pharmacokinetics, drug-drug interactions, and efficacy. These advanced cellular systems have quickly become established for human intrinsic clearance determination and have been validated for several other absorption, distribution, metabolism, and excretion (ADME) applications. Adoption will be driven through the demonstration of clear added value, for instance, by more accurate and precise clearance predictions and by more reliable extrapolation of drug interaction potential leading to faster progression to pivotal proof-of-concept studies. New experimental systems are attractive when they can (1) increase experimental capacity, removing optimization bottlenecks; (2) improve measurement quality of ADME properties that impact pharmacokinetics; and (3) enable measurements to be made that were not previously possible, reducing risk in ADME prediction and candidate selection. As new systems become established, they will find their place in the repository of tools used at different stages of the research and development process, depending on the balance of value, throughput, and cost. In this article, we give a perspective on the integration of these new methodologies into ADME optimization during drug discovery, and the likely applications and impacts on drug development.
Collapse
Affiliation(s)
- Luca Docci
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland.,2 Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Neil Parrott
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | | | - Stephen Fowler
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
96
|
Chi Q, Wang L, Xie D, Wang X. Characterization of in vitro metabolism of focal adhesion kinase inhibitors by LC/MS/MS. J Pharm Biomed Anal 2019; 168:163-173. [PMID: 30807921 DOI: 10.1016/j.jpba.2019.02.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/14/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is critically involved in cell migration, spreading and proliferation at the early step of various cancers. Small molecule inhibitors of FAK are effective to inhibit its activation in the process of tumor formation in cell. To better understand biotransformation of FAK inhibitors, this work has investigated in vitro phase I metabolism of inhibitors (namely PF-573228, PF-562271 and PF-03814735) by rat liver microsomes model. Using liquid chromatography - quadrupole time of flight mass spectrometry and tandem mass spectrometry (LC/Q-TOF/MS and MS/MS), three metabolites of PF-573228 and PF-562271 were observed and characterized, respectively. These in vitro metabolites were reported for the first time. The structures and fragmentation patterns of these metabolites were elucidated, and phase I metabolic pathways for FAK inhibitors were proposed. The main metabolic pathways of PF-573228 were hydroxylation, dehydrogenation and N-dealkylation. For PF-562271, they were hydroxylation and dehydrogenation. Hydroxylation was observed as the primary metabolism for PF-0381473.
Collapse
Affiliation(s)
- Quan Chi
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Ling Wang
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Dong Xie
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Xian Wang
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China.
| |
Collapse
|
97
|
Transformation Products of Organic Contaminants and Residues-Overview of Current Simulation Methods. Molecules 2019; 24:molecules24040753. [PMID: 30791496 PMCID: PMC6413221 DOI: 10.3390/molecules24040753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 01/27/2023] Open
Abstract
The formation of transformation products (TPs) from contaminants and residues is becoming an increasing focus of scientific community. All organic compounds can form different TPs, thus demonstrating the complexity and interdisciplinarity of this topic. The properties of TPs could stand in relation to the unchanged substance or be more harmful and persistent. To get important information about the generated TPs, methods are needed to simulate natural and manmade transformation processes. Current tools are based on metabolism studies, photochemical methods, electrochemical methods, and Fenton’s reagent. Finally, most transformation processes are based on redox reactions. This review aims to compare these methods for structurally different compounds. The groups of pesticides, pharmaceuticals, brominated flame retardants, and mycotoxins were selected as important residues/contaminants relating to their worldwide occurrence and impact to health, food, and environmental safety issues. Thus, there is an increasing need for investigation of transformation processes and identification of TPs by fast and reliable methods.
Collapse
|
98
|
Marin TM, de Carvalho Indolfo N, Rocco SA, Basei FL, de Carvalho M, de Almeida Gonçalves K, Pagani E. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact 2019; 299:59-76. [DOI: 10.1016/j.cbi.2018.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 11/10/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022]
|
99
|
Dobson THW, Tao RH, Swaminathan J, Maegawa S, Shaik S, Bravo-Alegria J, Sharma A, Kennis B, Yang Y, Callegari K, Haltom AR, Taylor P, Kogiso M, Qi L, Khatua S, Goldman S, Lulla RR, Fangusaro J, MacDonald TJ, Li XN, Hawkins C, Rajaram V, Gopalakrishnan V. Transcriptional repressor REST drives lineage stage-specific chromatin compaction at Ptch1 and increases AKT activation in a mouse model of medulloblastoma. Sci Signal 2019; 12:12/565/eaan8680. [PMID: 30670636 DOI: 10.1126/scisignal.aan8680] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In medulloblastomas (MBs), the expression and activity of RE1-silencing transcription factor (REST) is increased in tumors driven by the sonic hedgehog (SHH) pathway, specifically the SHH-α (children 3 to 16 years) and SHH-β (infants) subgroups. Neuronal maturation is greater in SHH-β than SHH-α tumors, but both correlate with poor overall patient survival. We studied the contribution of REST to MB using a transgenic mouse model (RESTTG ) wherein conditional NeuroD2-controlled REST transgene expression in lineage-committed Ptch1 +/- cerebellar granule neuron progenitors (CGNPs) accelerated tumorigenesis and increased penetrance and infiltrative disease. This model revealed a neuronal maturation context-specific antagonistic interplay between the transcriptional repressor REST and the activator GLI1 at Ptch1 Expression of Arrb1, which encodes β-arrestin1 (a GLI1 inhibitor), was substantially reduced in proliferating and, to a lesser extent, lineage-committed RESTTG cells compared with wild-type proliferating CGNPs. Lineage-committed RESTTG cells also had decreased GLI1 activity and increased histone H3K9 methylation at the Ptch1 locus, which correlated with premature silencing of Ptch1 These cells also had decreased expression of Pten, which encodes a negative regulator of the kinase AKT. Expression of PTCH1 and GLI1 were less, and ARRB1 was somewhat greater, in patient SHH-β than SHH-α MBs, whereas that of PTEN was similarly lower in both subtypes than in others. Inhibition of histone modifiers or AKT reduced proliferation and induced apoptosis, respectively, in cultured REST-high MB cells. Our findings linking REST to differentiation-specific chromatin remodeling, PTCH1 silencing, and AKT activation in MB tissues reveal potential subgroup-specific therapeutic targets for MB patients.
Collapse
Affiliation(s)
- Tara H W Dobson
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rong-Hua Tao
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Shinji Maegawa
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shavali Shaik
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Javiera Bravo-Alegria
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ajay Sharma
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bridget Kennis
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanwen Yang
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keri Callegari
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amanda R Haltom
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pete Taylor
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mari Kogiso
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lin Qi
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Soumen Khatua
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stewart Goldman
- Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA
| | - Rishi R Lulla
- Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA
| | - Jason Fangusaro
- Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA
| | | | - Xiao-Nan Li
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA
| | - Cynthia Hawkins
- Department of Pathology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Veena Rajaram
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA. .,Molecular and Cellular Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.,Brain Tumor Center, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for Cancer Epigenetics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.,The University of Texas MD Anderson Cancer Center-University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
100
|
Li S, Zhao J, Huang R, Santillo MF, Houck KA, Xia M. Use of high-throughput enzyme-based assay with xenobiotic metabolic capability to evaluate the inhibition of acetylcholinesterase activity by organophosphorous pesticides. Toxicol In Vitro 2019; 56:93-100. [PMID: 30625376 DOI: 10.1016/j.tiv.2019.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
Abstract
The inhibition of acetylcholinesterase (AChE) has pharmaceutical applications as well as potential neurotoxic effects. The in vivo metabolites of some chemicals including organophosphorus pesticides can become more potent AChE inhibitors compared to their parental compounds. To account for the effects of biotransformation, we have developed and characterized a high-throughput screening method for identifying AChE inhibitors that become active or more potent following xenobiotic metabolism. In this study, an enzyme-based assay was developed in 1536-well plates using recombinant human AChE combined with human or rat liver microsomes. The AChE activity was measured by two methods with different readouts: colorimetric and fluorescent. The assay exhibited exceptional performance characteristics including large assay signal window, low well-to-well variability and high reproducibility. The performance of the assays with microsomes was characterized by testing a group of known AChE inhibitors including parent compounds and their metabolites. Large potency differences between the parent compounds and the metabolites were observed in the assay with microsome addition. Both assay readouts were required for maximal sensitivity. These results demonstrate that this platform is a promising method to profile large numbers of chemicals that require metabolic activation for inhibiting AChE activity.
Collapse
Affiliation(s)
- Shuaizhang Li
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jinghua Zhao
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Ruili Huang
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Michael F Santillo
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Keith A Houck
- National Center for Computational Toxicology, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|