101
|
Phillips MC, Dheer R, Santaolalla R, Davies JM, Burgueño J, Lang JK, Toborek M, Abreu MT. Intestinal exposure to PCB 153 induces inflammation via the ATM/NEMO pathway. Toxicol Appl Pharmacol 2017; 339:24-33. [PMID: 29197519 DOI: 10.1016/j.taap.2017.11.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/08/2017] [Accepted: 11/28/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Polychlorinated biphenyls (PCBs) are persistent organic pollutants that adversely affect human health. PCBs bio-accumulate in organisms important for human consumption. PCBs accumulation in the body leads to activation of the transcription factor NF-κB, a major driver of inflammation. Despite dietary exposure being one of the main routes of exposure to PCBs, the gut has been widely ignored when studying the effects of PCBs. OBJECTIVES We investigated the effects of PCB 153 on the intestine and addressed whether PCB 153 affected intestinal permeability or inflammation and the mechanism by which this occurred. METHODS Mice were orally exposed to PCB 153 and gut permeability was assessed. Intestinal epithelial cells (IECs) were collected and evaluated for evidence of genotoxicity and inflammation. A human IEC line (SW480) was used to examine the direct effects of PCB 153 on epithelial function. NF-кB activation was measured using a reporter assay, DNA damage was assessed, and cytokine expression was ascertained with real-time PCR. RESULTS Mice orally exposed to PCB 153 had an increase in intestinal permeability and inflammatory cytokine expression in their IECs; inhibition of NF-кB ameliorated both these effects. This inflammation was associated with genotoxic damage and NF-кB activation. Exposure of SW480 cells to PCB 153 led to similar effects as seen in vivo. We found that activation of the ATM/NEMO pathway by genotoxic stress was upstream of NF-kB activation. CONCLUSIONS These results demonstrate that oral exposure to PCB 153 is genotoxic to IECs and induces downstream inflammation and barrier dysfunction in the intestinal epithelium.
Collapse
Affiliation(s)
- Matthew C Phillips
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rishu Dheer
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rebeca Santaolalla
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Julie M Davies
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Burgueño
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jessica K Lang
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria T Abreu
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA; Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
102
|
Dachineni R, Kumar DR, Callegari E, Kesharwani SS, Sankaranarayanan R, Seefeldt T, Tummala H, Bhat GJ. Salicylic acid metabolites and derivatives inhibit CDK activity: Novel insights into aspirin's chemopreventive effects against colorectal cancer. Int J Oncol 2017; 51:1661-1673. [PMID: 29075787 PMCID: PMC5673027 DOI: 10.3892/ijo.2017.4167] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022] Open
Abstract
Aspirin's potential as a drug continues to be evaluated for the prevention of colorectal cancer (CRC). Although multiple targets for aspirin and its metabolite, salicylic acid, have been identified, no unifying mechanism has been proposed to clearly explain its chemopreventive effects. Our goal here was to investigate the ability of salicylic acid metabolites, known to be generated through cytochrome P450 (CYP450) enzymes, and its derivatives as cyclin dependent kinase (CDK) inhibitors to gain new insights into aspirin's chemopreventive actions. Using in vitro kinase assays, for the first time, we demonstrate that salicylic acid metabolites, 2,3-dihydroxy-benzoic acid (2,3-DHBA) and 2,5-dihydroxybenzoic acid (2,5-DHBA), as well as derivatives 2,4-dihydroxybenzoic acid (2,4-DHBA), 2,6-dihydroxybenzoic acid (2,6-DHBA), inhibited CDK1 enzyme activity. 2,3-DHBA and 2,6-DHBA did not inhibit CDK2 and 4; however, both inhibited CDK-6 activity. Interestingly, another derivative, 2,4,6-trihydroxybenzoic acid (2,4,6-THBA) was highly effective in inhibiting CDK1, 2, 4 and 6 activity. Molecular docking studies showed that these compounds potentially interact with CDK1. Immunoblotting experiments showed that aspirin acetylated CDK1, and pre-incubation with salicylic acid and its derivatives prevented aspirin-mediated CDK1 acetylation, which supported the data obtained from molecular docking studies. We suggest that intracellularly generated salicylic acid metabolites through CYP450 enzymes within the colonic epithelial cells, or the salicylic acid metabolites generated by gut microflora may significantly contribute to the preferential chemopreventive effect of aspirin against CRC through inhibition of CDKs. This novel hypothesis and mechanism of action in aspirin's chemopreventive effects opens a new area for future research. In addition, structural modification to salicylic acid derivatives may prove useful in the development of novel CDK inhibitors in cancer prevention and treatment.
Collapse
Affiliation(s)
- Rakesh Dachineni
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - D Ramesh Kumar
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - Eduardo Callegari
- SD-BRIN Proteomic Facility, University of South Dakota School of Medicine, Vermillion, SD 57069, USA
| | - Siddharth S Kesharwani
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - Ranjini Sankaranarayanan
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - Teresa Seefeldt
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - Hemachand Tummala
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| | - G Jayarama Bhat
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD 57007, USA
| |
Collapse
|
103
|
Xu J, Lin Y, Boulas P, Peterson ML. Low colonic absorption drugs: risks and opportunities in the development of oral extended release products. Expert Opin Drug Deliv 2017; 15:197-211. [PMID: 28988504 DOI: 10.1080/17425247.2018.1389889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Currently numerous drugs have been observed with lower colonic absorption than small intestine absorption, which can significantly impact in vivo performance of their oral extended release (ER) products. AREAS COVERED We reviewed over 300 publications, patents, book chapters, and commercial reports of drug products from regulatory agencies for low colonic absorption (LCA) drugs and critical findings are discussed. The focuses of this article are (1) current findings on the causes of low colonic absorption to support early assessment of LCA candidates, and (2) current knowledge on successful ER strategies and technical platforms used for LCA drugs in commercial drug products to facilitate oral ER product development. EXPERT OPINION Colonic drug absorption is one of the critical considerations in successful development of oral ER products. The root causes of low colonic absorption in many LCA drugs are still unclear. It is recommended to evaluate colonic drug absorption of drug candidate at early stage of oral ER product development. After evaluation, the selection of a formulation platform to develop an oral ER product needs to be carefully considered for LCA drugs. Based on the current commercial oral ER formulation platforms for LCA drugs, compounds are first divided into five types (I-V) and different ER formulation approaches with higher success rate are recommended for each type.
Collapse
Affiliation(s)
- Jin Xu
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | - Yiqing Lin
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | - Pierre Boulas
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | | |
Collapse
|
104
|
Hatley OJD, Jones CR, Galetin A, Rostami-Hodjegan A. Optimization of intestinal microsomal preparation in the rat: A systematic approach to assess the influence of various methodologies on metabolic activity and scaling factors. Biopharm Drug Dispos 2017; 38:187-208. [PMID: 28207929 PMCID: PMC5413848 DOI: 10.1002/bdd.2070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/31/2017] [Accepted: 02/11/2017] [Indexed: 01/27/2023]
Abstract
The metabolic capacity of the intestine and its importance as the initial barrier to systemic exposure can lead to underestimation of first‐pass, and thus overestimation of oral bioavailability. However, the in vitro tools informing estimates of in vivo intestinal metabolism are limited by the complexity of the in vitro matrix preparation and uncertainty with the scaling factors for in vitro to in vivo extrapolation. A number of methods currently exist in the literature for the preparation of intestinal microsomes; however, the impact of key steps in the preparation procedure has not been critically assessed. In the current study, changes in enterocyte isolation, the impact of buffer constituents heparin and glycerol, as well as sonication as a direct method of homogenization were assessed systematically. Furthermore, fresh vs. frozen tissue samples and the impact of microsome freeze thawing was assessed. The rat intestinal microsomes were characterized for CYP content as well as metabolic activity using testosterone and 4‐nitropheonol as probes for CYP and UGT activity, respectively. Comparisons in metabolic activity and scaled unbound intestinal intrinsic clearance (CLintu,gut) were made to commercially available microsomes using 25 drugs with a diverse range of metabolic pathways and intestinal metabolic stabilities. An optimal, robust and reproducible microsomal preparation method for investigation of intestinal metabolism is proposed. The importance of characterization of the in vitro matrix and the potential impact of intestinal scaling factors on the in vitro–in vivo extrapolation of FG needs to be investigated further. © 2017 The Authors Biopharmaceutics & Drug Disposition Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Oliver J D Hatley
- Certara, Blades Enterprise Centre, Sheffield, S2 4SU, UK.,Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, M13 9PT, UK
| | | | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, M13 9PT, UK
| | - Amin Rostami-Hodjegan
- Certara, Blades Enterprise Centre, Sheffield, S2 4SU, UK.,Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
105
|
Haraya K, Kato M, Chiba K, Sugiyama Y. Prediction of inter-individual variability on the pharmacokinetics of CYP2C8 substrates in human. Drug Metab Pharmacokinet 2017; 32:277-285. [PMID: 29174535 DOI: 10.1016/j.dmpk.2017.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/06/2017] [Accepted: 09/06/2017] [Indexed: 01/10/2023]
Abstract
Inter-individual variability in pharmacokinetics can lead to unexpected side effects and treatment failure, and is therefore an important factor in drug development. CYP2C8 is a major drug-metabolizing enzyme known to be involved in the metabolism of over 100 drugs. In this study, we predicted the inter-individual variability in AUC/Dose of CYP2C8 substrates in healthy volunteers using the Monte Carlo simulation. Inter-individual variability in the hepatic intrinsic clearance of CYP2C8 substrates (CLint,h,2C8) was estimated from the inter-individual variability in pharmacokinetics of pioglitazone, which is a major CYP2C8 substrate. The coefficient of variation (CV) of CLint,h,2C8 was estimated to be 40%. Using this value, the CVs of AUC/Dose of other major CYP2C8 substrates, rosiglitazone and amodiaquine, were predicted to validate the estimated CV of CLint,h,2C8. As a result, the reported CVs of both substrates were within the 2.5-97.5 percentile range of the predicted CVs. Furthermore, the CVs of AUC/Dose of the CYP2C8 substrates loperamide and chloroquine, which are affected by renal clearance, were also successfully predicted. Combining this value with previously reported CVs of other CYPs, we were able to successfully predict the inter-individual variability in pharmacokinetics of various drugs in clinical.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd., Singapore.
| | | | - Koji Chiba
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Japan; Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Japan
| |
Collapse
|
106
|
Berry DJ, Steed JW. Pharmaceutical cocrystals, salts and multicomponent systems; intermolecular interactions and property based design. Adv Drug Deliv Rev 2017; 117:3-24. [PMID: 28344021 DOI: 10.1016/j.addr.2017.03.003] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 01/01/2023]
Abstract
As small molecule drugs become harder to develop and less cost effective for patient use, efficient strategies for their property improvement become increasingly important to global health initiatives. Improvements in the physical properties of Active Pharmaceutical Ingredients (APIs), without changes in the covalent chemistry, have long been possible through the application of binary component solids. This was first achieved through the use of pharmaceutical salts, within the last 10-15years with cocrystals and more recently coamorphous systems have also been consciously applied to this problem. In order to rationally discover the best multicomponent phase for drug development, intermolecular interactions need to be considered at all stages of the process. This review highlights the current thinking in this area and the state of the art in: pharmaceutical multicomponent phase design, the intermolecular interactions in these phases, the implications of these interactions on the material properties and the pharmacokinetics in a patient.
Collapse
Affiliation(s)
- David J Berry
- Durham University, Division of Pharmacy, Queen's Campus, Stockton on Tees, TS17 6BH, UK.
| | - Jonathan W Steed
- Department of Chemistry, Durham University, University Science Laboratories, South Road, Durham, DH1 3LE, UK
| |
Collapse
|
107
|
He G, Troberg J, Lv X, Xia YL, Zhu LL, Ning J, Ge GB, Finel M, Yang L. Identification and characterization of human UDP-glucuronosyltransferases responsible for xanthotoxol glucuronidation. Xenobiotica 2017; 48:109-116. [DOI: 10.1080/00498254.2017.1283719] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Guiyuan He
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
- University of Chinese Academy of Sciences, Beijing, China,
| | - Johanna Troberg
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland, and
| | - Xia Lv
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
- University of Chinese Academy of Sciences, Beijing, China,
| | - Yang-Liu Xia
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
- University of Chinese Academy of Sciences, Beijing, China,
| | - Liang-Liang Zhu
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
| | - Jing Ning
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
| | - Guang-Bo Ge
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland, and
| | - Ling Yang
- Laboratory of Pharmaceutical Resource Discovery, Biotechnology Department, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China,
- Centre for System Pharmacokinetics, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
108
|
Dahlinger D, Aslan S, Pietsch M, Frechen S, Fuhr U. Assessment of inhibitory effects on major human cytochrome P450 enzymes by spasmolytics used in the treatment of overactive bladder syndrome. Ther Adv Urol 2017; 9:163-177. [PMID: 28747995 PMCID: PMC5510770 DOI: 10.1177/1756287217708951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/19/2017] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The objective of this study was to examine the inhibitory potential of darifenacin, fesoterodine, oxybutynin, propiverine, solifenacin, tolterodine and trospium chloride on the seven major human cytochrome P450 enzymes (CYP) by using a standardized and validated seven-in-one cytochrome P450 cocktail inhibition assay. METHODS An in vitro cocktail of seven highly selective probe substrates was incubated with human liver microsomes and varying concentrations of the seven test compounds. The major metabolites of the probe substrates were simultaneously analysed using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Enzyme kinetics were estimated by determining IC50 and Ki values via nonlinear regression. Obtained Ki values were used for predictions of potential clinical impact of the inhibition using a static mechanistic prediction model. RESULTS In this study, 49 IC50 experiments were conducted. In six cases, IC50 values lower than the calculated threshold for drug-drug interactions (DDIs) in the gut wall were observed. In these cases, no increase in inhibition was determined after a 30 min preincubation. Considering a typical dosing regimen and applying the obtained Ki values of 0.72 µM (darifenacin, 15 mg daily) and 7.2 µM [propiverine, 30 mg daily, immediate release (IR)] for the inhibition of CYP2D6 yielded a predicted 1.9-fold and 1.4-fold increase in the area under the curve (AUC) of debrisoquine (CYP2D6 substrate), respectively. Due to the inhibition of the particular intestinal CYP3A4, the obtained Ki values of 14 µM of propiverine (30 mg daily, IR) resulted in a predicted doubling of the AUC for midazolam (CYP3A4 substrate). CONCLUSIONS In vitro/in vivo extrapolation based on pharmacokinetic data and the conducted screening experiments yielded similar effects of darifenacin on CYP2D6 and propiverine on CYP3A4 as obtained in separately conducted in vivo DDI studies. As a novel finding, propiverine was identified to potentially inhibit CYP2D6 at clinically occurring concentrations.
Collapse
Affiliation(s)
- Dominik Dahlinger
- Department I of Pharmacology, University Hospital Cologne, Köln, Germany
| | - Sevinc Aslan
- Department I of Pharmacology, University Hospital Cologne, Köln, Germany
| | - Markus Pietsch
- Department II of Pharmacology, University Hospital Cologne, Köln, Germany
| | - Sebastian Frechen
- Department I of Pharmacology, University Hospital Cologne, Köln, Germany
| | - Uwe Fuhr
- Department I of Pharmacology, Center for Pharmacology, Clinical Pharmacology Unit, University Hospital Cologne (AöR), Gleueler Straße 24, 50931 Köln, Germany
| |
Collapse
|
109
|
Kawakami M, Takenoshita-Nakaya S, Takeba Y, Nishimura Y, Oda M, Watanabe M, Ohta Y, Kobayashi S, Ohtsubo T, Kobayashi S, Uchida N, Matsumoto N. Evaluation of CYP2D6 Protein Expression and Activity in the Small Intestine to Determine Its Metabolic Capability in the Japanese Population. Biol Pharm Bull 2017. [PMID: 28626197 DOI: 10.1248/bpb.b16-00370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CYP2D6 plays an important role in the metabolism of many drugs such as opioids and antidepressants. Polymorphisms of the CYP2D6 gene are widely observed in the Japanese population, and can affect the first-pass metabolism of orally administered drugs. Several CYP enzymes have been identified in the small intestine of Caucasians, but intestinal CYP enzymes have not been reported in the Japanese population, except for CYP3A4 and CYP2C19. In this study, we evaluated the CYP2D6 metabolic capacity by measurement of CYP2D6 mRNA and protein levels and activity in the small intestine of Japanese individuals. Normal jejunal tissues were obtained from 31 patients who had undergone pancreaticoduodenectomy, and the CYP2D6*10 variant was identified in these tissues. CYP2D6 mRNA and CYP2D6 protein levels were analyzed using real-time RT-PCR and Western blotting, respectively. Bufuralol 1'-hydroxylation, a marker of CYP2D6 activity, was analyzed using HPLC. Frequencies of the CYP2D6*1/*1, *1/*10, and *10/*10 genotypes in the jejunal tissue were 29.0% (n=9), 35.5% (n=11), and 35.5% (n=11), respectively. CYP2D6 protein and activity levels did not differ significantly between the genotypes. A positive correlation was found between CYP2D6 protein and activity levels. Furthermore, CYP2D6 protein levels and activity in the small intestine were significantly lower than those in the liver. These findings suggest that the metabolic capacity of CYP2D6 in the small intestine of the Japanese population has a relatively small effect on drug metabolism.
Collapse
Affiliation(s)
- Momoko Kawakami
- Department of Clinical Pharmacology, Showa University School of Medicine.,Department of Pharmacology, St. Marianna University School of Medicine
| | | | - Yuko Takeba
- Department of Pharmacology, St. Marianna University School of Medicine
| | - Yuki Nishimura
- Department of Pharmacology, Showa University School of Medicine
| | - Masayuki Oda
- Department of Pharmacogenomics, St. Marianna University Graduate School of Medicine
| | - Minoru Watanabe
- Institute of Animal Experimentation, St. Marianna University Graduate School of Medicine
| | - Yuki Ohta
- Department of Pharmacology, St. Marianna University School of Medicine
| | - Shinjiro Kobayashi
- Division of Gastroenterological and General Surgery, St. Marianna University School of Medicine
| | - Takehito Ohtsubo
- Division of Gastroenterological and General Surgery, St. Marianna University School of Medicine
| | - Shinichi Kobayashi
- Showa University Clinical Research Institute for Clinical Pharmacology and Therapeutics
| | - Naoki Uchida
- Department of Clinical Pharmacology, Showa University School of Medicine.,Showa University Clinical Research Institute for Clinical Pharmacology and Therapeutics
| | - Naoki Matsumoto
- Department of Pharmacology, St. Marianna University School of Medicine
| |
Collapse
|
110
|
Three-dimensional in vitro gut model on a villi-shaped collagen scaffold. BIOCHIP JOURNAL 2017. [DOI: 10.1007/s13206-017-1307-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
111
|
Samant TS, Lukacova V, Schmidt S. Development and Qualification of Physiologically Based Pharmacokinetic Models for Drugs With Atypical Distribution Behavior: A Desipramine Case Study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:315-321. [PMID: 28398693 PMCID: PMC5697013 DOI: 10.1002/psp4.12180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/04/2022]
Abstract
Desipramine is a secondary tricyclic amine, which is primarily metabolized by cytochrome 2D6. It shows a high volume of distribution (Vss) (10–50 L/kg) due to its high lipophilicity, unspecific phospholipid binding, and lysosomal trapping. The objective of this study was to develop and qualify a physiologically based pharmacokinetic (PBPK) model for desipramine, which accounts for the high Vss of the drug following intravenous and oral administration of doses up to 100 mg. The model also accounts for the extended time to reach maximum concentration after oral dosing due to enterocyte trapping. Once developed and qualified in adults, we characterized the dynamic changes in metabolism and pharmacokinetics of desipramine after birth by scaling the system‐specific parameters of the model from adults to pediatrics. The developed modeling strategy provides a prototypical workflow that can also be applied to other drugs with similar properties and a high volume of distribution.
Collapse
Affiliation(s)
- T S Samant
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Lake Nona (Orlando), Florida, USA
| | - V Lukacova
- Simulations Plus, Inc., Lancaster, California, USA
| | - S Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Lake Nona (Orlando), Florida, USA
| |
Collapse
|
112
|
Constitutive expression and activity of cytochrome P450 in conventional pigs. Res Vet Sci 2017; 111:75-80. [DOI: 10.1016/j.rvsc.2016.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022]
|
113
|
Müller MF, Zhou Y, Adams DJ, Arends MJ. Effects of long-term ethanol consumption and Aldh1b1 depletion on intestinal tumourigenesis in mice. J Pathol 2017; 241:649-660. [PMID: 28026023 DOI: 10.1002/path.4869] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
Ethanol and its metabolite acetaldehyde have been classified as carcinogens for the upper aerodigestive tract, liver, breast, and colorectum. Whereas mechanisms related to oxidative stress and Cyp2e1 induction seem to prevail in the liver, and acetaldehyde has been proposed to play a crucial role in the upper aerodigestive tract, pathological mechanisms in the colorectum have not yet been clarified. Moreover, all evidence for a pro-carcinogenic role of ethanol in colorectal cancer is derived from correlations observed in epidemiological studies or from rodent studies with additional carcinogen application or tumour suppressor gene inactivation. In the current study, wild-type mice and mice with depletion of aldehyde dehydrogenase 1b1 (Aldh1b1), an enzyme which has been proposed to play an important role in acetaldehyde detoxification in the intestines, received ethanol in drinking water for 1 year. Long-term ethanol consumption led to intestinal tumour development in wild-type and Aldh1b1-depleted mice, but no intestinal tumours were observed in water-treated controls. Moreover, a significant increase in DNA damage was detected in the large intestinal epithelium of ethanol-treated mice of both genotypes compared with the respective water-treated groups, along with increased proliferation of the small and large intestinal epithelium. Aldh1b1 depletion led to increased plasma acetaldehyde levels in ethanol-treated mice, to a significant aggravation of ethanol-induced intestinal hyperproliferation, and to more advanced features of intestinal tumours, but it did not affect intestinal tumour incidence. These data indicate that ethanol consumption can initiate intestinal tumourigenesis without any additional carcinogen treatment or tumour suppressor gene inactivation, and we provide evidence for a role of Aldh1b1 in protection of the intestines from ethanol-induced damage, as well as for both carcinogenic and tumour-promoting functions of acetaldehyde, including increased progression of ethanol-induced tumours. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mike F Müller
- University of Edinburgh, Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Ying Zhou
- University of Edinburgh, Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Mark J Arends
- University of Edinburgh, Division of Pathology, Centre for Comparative Pathology, Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, UK
| |
Collapse
|
114
|
Johansson S, Rosenbaum DP, Ahlqvist M, Rollison H, Knutsson M, Stefansson B, Elebring M. Effects of Tenapanor on Cytochrome P450-Mediated Drug-Drug Interactions. Clin Pharmacol Drug Dev 2017; 6:466-475. [PMID: 28301096 PMCID: PMC5599994 DOI: 10.1002/cpdd.346] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023]
Abstract
Tenapanor (RDX5791, AZD1722) is an inhibitor of sodium/hydrogen exchanger isoform 3 in development for the treatment of constipation‐predominant irritable bowel syndrome and the treatment of hyperphosphatemia in patients with chronic kidney disease on dialysis. We aimed to investigate whether tenapanor inhibits or induces cytochrome P450s (CYPs). In vitro experiments assessing the potential of tenapanor to affect various CYPs indicated that it could inhibit CYP3A4/5 (IC50 0.4‐0.7 μM). An open‐label, phase 1 clinical study (NCT02140268) evaluated the pharmacokinetics of the CYP3A4 substrate midazolam when administered with and without tenapanor. Healthy volunteers received a single oral dose of midazolam 7.5 mg on day 1 followed by tenapanor 15 mg twice daily on days 2 to 15, with an additional single 7.5‐mg midazolam dose coadministered on day 15. Midazolam exposure was similar whether it was administered alone or with tenapanor (geometric least‐squares mean ratio [90%CI] for [midazolam + tenapanor]/midazolam: area under the concentration‐time curve, 107% [101% to 113%]; Cmax 104% [89.6% to 122%]). Findings were similar for metabolites of midazolam. These results indicate that tenapanor 15 mg twice daily does not have a clinically relevant impact on CYP3A4 in humans and suggest that tenapanor can be coadministered with CYP3A4‐metabolized drugs without affecting their exposure.
Collapse
|
115
|
Gupta P, Gramatke A, Einspanier R, Schütte C, von Kleist M, Sharbati J. In silico cytotoxicity assessment on cultured rat intestinal cells deduced from cellular impedance measurements. Toxicol In Vitro 2017; 41:179-188. [PMID: 28263893 DOI: 10.1016/j.tiv.2017.02.021] [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: 12/05/2016] [Revised: 02/16/2017] [Accepted: 02/27/2017] [Indexed: 11/16/2022]
Abstract
Early and reliable identification of chemical toxicity is of utmost importance. At the same time, reduction of animal testing is paramount. Therefore, methods that improve the interpretability and usability of in vitro assays are essential. xCELLigence's real-time cell analyzer (RTCA) provides a novel, fast and cost effective in vitro method to probe compound toxicity. We developed a simple mathematical framework for the qualitative and quantitative assessment of toxicity for RTCA measurements. Compound toxicity, in terms of its 50% inhibitory concentration IC50 on cell growth, and parameters related to cell turnover were estimated on cultured IEC-6 cells exposed to 10 chemicals at varying concentrations. Our method estimated IC50 values of 113.05, 7.16, 28.69 and 725.15 μM for the apparently toxic compounds 2-acetylamino-fluorene, aflatoxin B1, benzo-[a]-pyrene and chloramphenicol in the tested cell line, in agreement with literature knowledge. IC50 values of all apparent in vivo non-toxic compounds were estimated to be non-toxic by our method. Corresponding estimates from RTCA's in-built model gave false positive (toxicity) predictions in 5/10 cases. Taken together, our proposed method reduces false positive predictions and reliably identifies chemical toxicity based on impedance measurements. The source code for the developed method including instructions is available at https://git.zib.de/bzfgupta/toxfit/tree/master.
Collapse
Affiliation(s)
- P Gupta
- Department of Mathematics and Informatics, Freie Universität Berlin, Arnimallee 6, Berlin 14195, Germany; Department of Mathematics for Life and Materials Sciences, Zuse Institute Berlin, Takustrasse 7, Berlin 14195, Germany.
| | - A Gramatke
- Department of Veterinary Biochemistry, Freie Universität Berlin, Oertzenweg 19b, Building 12, Berlin 14163, Germany.
| | - R Einspanier
- Department of Veterinary Biochemistry, Freie Universität Berlin, Oertzenweg 19b, Building 12, Berlin 14163, Germany.
| | - C Schütte
- Department of Mathematics and Informatics, Freie Universität Berlin, Arnimallee 6, Berlin 14195, Germany; Department of Mathematics for Life and Materials Sciences, Zuse Institute Berlin, Takustrasse 7, Berlin 14195, Germany.
| | - M von Kleist
- Department of Mathematics and Informatics, Freie Universität Berlin, Arnimallee 6, Berlin 14195, Germany.
| | - J Sharbati
- Department of Veterinary Biochemistry, Freie Universität Berlin, Oertzenweg 19b, Building 12, Berlin 14163, Germany.
| |
Collapse
|
116
|
Kim A, Yu BY, Dueker SR, Shin KH, Kim HS, Ahn H, Cho JY, Yu KS, Jang IJ, Lee H. An Accelerator Mass Spectrometry-Enabled Microtracer Study to Evaluate the First-Pass Effect on the Absorption of YH4808. Clin Pharmacol Ther 2017; 102:537-546. [PMID: 28214288 DOI: 10.1002/cpt.672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/23/2017] [Accepted: 02/14/2017] [Indexed: 12/25/2022]
Abstract
14 C-labeled YH4808, a novel potassium-competitive acid blocker, was intravenously administered as a microtracer at 80 μg (11.8 kBq or 320 nCi) concomitantly with the nonradiolabeled oral drug at 200 mg to determine the absolute bioavailability and to assess the effect of pharmacogenomics on the oral absorption of YH4808. The absolute bioavailability was low and highly variable (mean, 10.1%; range, 2.3-19.3%), and M3 and M8, active metabolites of YH4808, were formed 22.6- and 38.5-fold higher after oral administration than intravenous administration, respectively. The product of the fraction of an oral YH4808 dose entering the gut wall and the fraction of YH4808 passing on to the portal circulation was larger in subjects carrying the variants of the CHST3, SLC15A1, and SULT1B1 genes. A combined LC+AMS is a useful tool to construct a rich and highly informative pharmacokinetic knowledge core in early clinical drug development at a reasonable cost.
Collapse
Affiliation(s)
- A Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Clinical Trial Center, Ajou University Medical Center, Suwon, Korea
| | - B-Y Yu
- Korea Institute of Science and Technology, Seoul, Korea
| | | | - K-H Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - H S Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - H Ahn
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - J-Y Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - K-S Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - I-J Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - H Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
117
|
Billat PA, Roger E, Faure S, Lagarce F. Models for drug absorption from the small intestine: where are we and where are we going? Drug Discov Today 2017; 22:761-775. [PMID: 28115264 DOI: 10.1016/j.drudis.2017.01.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/12/2016] [Accepted: 01/13/2017] [Indexed: 02/07/2023]
Abstract
The small intestine is a complex organ with movements, flora, mucus and flows. Despite this, the most widely used absorption models consider the organ a cylindrical monoepithelial tube. This review presents the recent evolution of models to take into consideration the complex nature of gut physiology. The most commonly encountered issues are ethical (in vivo models) and differences in drug transport as a result of a modified expression of drug transporters or metabolic enzymes compared with human (in vitro and in vivo models). Finally, this review discusses the way forward to reach an ideal equilibrium between reproducibility, predictability and efficiency for predicting permeability. The features of an ideal model are listed as a guideline for future development.
Collapse
Affiliation(s)
- Pierre-André Billat
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, France
| | - Emilie Roger
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, France
| | - Sébastien Faure
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, France
| | - Frédéric Lagarce
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, France; Pharmacy Department, Angers University Hospital, Angers, France.
| |
Collapse
|
118
|
Abstract
The lining of the gastrointestinal tract needs to be easily accessible to nutrients and, at the same time, defend against pathogens and chemical challenges. This lining is the largest and most vulnerable surface that faces the outside world. To manage the dual problems of effective nutrient conversion and defence, the gut lining has a sophisticated system for detection of individual chemical entities, pathogenic organisms and their products, and physico-chemical properties of its contents. Detection is through specific receptors that signal to the gut endocrine system, the nervous system, the immune system and local tissue defence systems. These effectors, in turn, modify digestive functions and contribute to tissue defence. Receptors for nutrients include taste receptors for sweet, bitter and savoury, free fatty acid receptors, peptide and phytochemical receptors, that are primarily located on enteroendocrine cells. Hormones released by enteroendocrine cells act locally, through the circulation and via the nervous system, to optimise digestion and mucosal health. Pathogen detection is both through antigen presentation to T-cells and through pattern-recognition receptors (PRRs). Activation of PRRs triggers local tissue defence, for example, by causing release of antimicrobials from Paneth cells. Toxic chemicals, including plant toxins, are sensed and then avoided, expelled or metabolised. It continues to be a major challenge to develop a comprehensive understanding of the integrated responses of the gastrointestinal tract to its luminal contents.
Collapse
|
119
|
Pang KS, Yang QJ, Noh K. Unequivocal evidence supporting the segregated flow intestinal model that discriminates intestine versus liver first-pass removal with PBPK modeling. Biopharm Drug Dispos 2016; 38:231-250. [PMID: 27977852 DOI: 10.1002/bdd.2056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 11/08/2022]
Abstract
Merits of the segregated flow model (SFM), highlighting the intestine as inert serosa and active enterocyte regions, with a smaller fractional (fQ < 0.3) intestinal flow (QI ) perfusing the enterocyte region, are described. Less drug in the circulation reaches the enterocytes due to the lower flow (fQ QI ) in comparison with drug administered into the gut lumen, fostering the idea of route-dependent intestinal removal. The SFM has been found superior to the traditional model (TM), which views the serosa and enterocytes totally as a well-mixed tissue perfused by 100% of the intestinal flow, QI . The SFM model is able to explain the lower extents of intestinal metabolism of enalapril, morphine and midazolam with i.v. vs. p.o. dosing. For morphine, the urine/bile ratio of the metabolite, morphine glucuronide MGurineMGbile for p.o. was 2.6× that of i.v. This was due to the higher proportion of intestinally formed morphine glucuronide, appearing more in urine than in bile due to its low permeability and greater extent of intestinal formation with p.o. administration. By contrast, the TM predicted the same MGurineMGbile for p.o. vs. i.v. The TM predicted that the contributions of the intestine:liver to first-pass removal were 46%:54% for both p.o. and i.v. The SFM predicted same 46%:54% (intestine:liver) for p.o., but 9%:91% for i.v. By contrast, the kinetics of codeine, the precursor of morphine, was described equally well by the SFM- and TM-PBPK models, a trend suggesting that intestinal metabolism of codeine is negligible. Fits to these PBPK models further provide insightful information towards metabolite formation: available fractions and the fractions of hepatic and total clearances that form the metabolite in question. The SFM-PBPK model is useful to identify not only the presence of intestinal metabolism but the contributions of the intestine and liver for metabolite formation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Qi Joy Yang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Keumhan Noh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
120
|
Kuepfer L, Niederalt C, Wendl T, Schlender JF, Willmann S, Lippert J, Block M, Eissing T, Teutonico D. Applied Concepts in PBPK Modeling: How to Build a PBPK/PD Model. CPT Pharmacometrics Syst Pharmacol 2016; 5:516-531. [PMID: 27653238 PMCID: PMC5080648 DOI: 10.1002/psp4.12134] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this tutorial is to introduce the fundamental concepts of physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling with a special focus on their practical implementation in a typical PBPK model building workflow. To illustrate basic steps in PBPK model building, a PBPK model for ciprofloxacin will be constructed and coupled to a pharmacodynamic model to simulate the antibacterial activity of ciprofloxacin treatment.
Collapse
Affiliation(s)
- L Kuepfer
- Bayer Technology Services, Leverkusen, Germany
| | - C Niederalt
- Bayer Technology Services, Leverkusen, Germany
| | - T Wendl
- Bayer Technology Services, Leverkusen, Germany
| | | | | | - J Lippert
- Bayer HealthCare, Wuppertal, Germany
| | - M Block
- Bayer Technology Services, Leverkusen, Germany
| | - T Eissing
- Bayer Technology Services, Leverkusen, Germany
| | - D Teutonico
- Bayer Technology Services, Leverkusen, Germany.
| |
Collapse
|
121
|
Xie F, Ding X, Zhang QY. An update on the role of intestinal cytochrome P450 enzymes in drug disposition. Acta Pharm Sin B 2016; 6:374-383. [PMID: 27709006 PMCID: PMC5045550 DOI: 10.1016/j.apsb.2016.07.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/05/2022] Open
Abstract
Oral administration is the most commonly used route for drug treatment. Intestinal cytochrome P450 (CYP)-mediated metabolism can eliminate a large proportion of some orally administered drugs before they reach systemic circulation, while leaving the passage of other drugs unimpeded. A better understanding of the ability of intestinal P450 enzymes to metabolize various clinical drugs in both humans and preclinical animal species, including the identification of the CYP enzymes expressed, their regulation, and the relative importance of intestinal metabolism compared to hepatic metabolism, is important for improving bioavailability of current drugs and new drugs in development. Here, we briefly review the expression of drug-metabolizing P450 enzymes in the small intestine of humans and several preclinical animal species, and provide an update of the various factors or events that regulate intestinal P450 expression, including a cross talk between the liver and the intestine. We further compare various clinical and preclinical approaches for assessing the impact of intestinal drug metabolism on bioavailability, and discuss the utility of the intestinal epithelium–specific NADPH-cytochrome P450 reductase-null (IECN) mouse as a useful model for studying in vivo roles of intestinal P450 in the disposition of orally administered drugs.
Collapse
|
122
|
Manjunatha UH, Chao AT, Leong FJ, Diagana TT. Cryptosporidiosis Drug Discovery: Opportunities and Challenges. ACS Infect Dis 2016; 2:530-7. [PMID: 27626293 DOI: 10.1021/acsinfecdis.6b00094] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The apicomplexan parasite Cryptosporidium is the second most important diarrheal pathogen causing life-threatening diarrhea in children, which is also associated with long-term growth faltering and cognitive deficiency. Cryptosporidiosis is a parasitic disease of public health concern caused by Cryptosporidium parvum and Cryptosporidium hominis. Currently, nitazoxanide is the only approved treatment for cryptosporidium infections. Unfortunately, it has limited efficacy in the most vulnerable patients, thus there is an urgent need for a safe and efficacious cryptosporidiosis drug. In this work, we present our current perspectives on the target product profile for novel cryptosporidiosis therapies and the perceived challenges and possible mitigation plans at different stages in the cryptosporidiosis drug discovery process.
Collapse
Affiliation(s)
- Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - Alexander T. Chao
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - F. Joel Leong
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| |
Collapse
|
123
|
Miyauchi E, Tachikawa M, Declèves X, Uchida Y, Bouillot JL, Poitou C, Oppert JM, Mouly S, Bergmann JF, Terasaki T, Scherrmann JM, Lloret-Linares C. Quantitative Atlas of Cytochrome P450, UDP-Glucuronosyltransferase, and Transporter Proteins in Jejunum of Morbidly Obese Subjects. Mol Pharm 2016; 13:2631-40. [DOI: 10.1021/acs.molpharmaceut.6b00085] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Eisuke Miyauchi
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masanori Tachikawa
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Xavier Declèves
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Pharmacokinetics and Pharmacochemistry Unit, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris F-75014, France
| | - Yasuo Uchida
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Jean-Luc Bouillot
- Department of Surgery, Université
Versailles Saint Quentin, Hôpital Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne 92100, France
| | - Christine Poitou
- Institut cardiométabolisme et nutrition
(ICAN), Université Pierre et Marie Curie, Service de Nutrition,
Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris F-75013, France
| | - Jean-Michel Oppert
- Institut cardiométabolisme et nutrition
(ICAN), Université Pierre et Marie Curie, Service de Nutrition,
Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris F-75013, France
| | - Stéphane Mouly
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| | - Jean-François Bergmann
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| | - Tetsuya Terasaki
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
| | - Célia Lloret-Linares
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| |
Collapse
|
124
|
Andreas CJ, Tomaszewska I, Muenster U, van der Mey D, Mueck W, Dressman JB. Can dosage form-dependent food effects be predicted using biorelevant dissolution tests? Case example extended release nifedipine. Eur J Pharm Biopharm 2016; 105:193-202. [PMID: 27322002 DOI: 10.1016/j.ejpb.2016.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/06/2016] [Accepted: 06/13/2016] [Indexed: 12/31/2022]
Abstract
AIMS Food intake is known to have various effects on gastrointestinal luminal conditions in terms of transit times, hydrodynamic forces and/or luminal fluid composition and can therefore affect the dissolution behavior of solid oral dosage forms. The aim of this study was to investigate and detect the dosage form-dependent food effect that has been observed for two extended-release formulations of nifedipine using in vitro dissolution tests. METHODS Two monolithic extended release formulations, the osmotic pump Adalat® XL 60mg and matrix-type Adalat® Eins 30mg formulation, were investigated with biorelevant dissolution methods using the USP apparatus III and IV under both simulated prandial states, and their corresponding quality control dissolution method. In vitro data were compared to published and unpublished in vivo data using deconvolution-based in vitro - in vivo correlation (IVIVC) approaches. RESULTS Quality control dissolution methods tended to overestimate the dissolution rate due to the excessive solubilizing capabilities of the sodium dodecyl sulfate (SDS)-containing dissolution media. Using Level II biorelevant media the dosage form dependent food effect for nifedipine was described well when studied with the USP apparatus III, whereas the USP apparatus IV failed to detect the positive food effect for the matrix-type dosage form. CONCLUSIONS It was demonstrated that biorelevant methods can serve as a useful tool during formulation development as they were able to qualitatively reflect the in vivo data.
Collapse
Affiliation(s)
- Cord J Andreas
- Institute of Pharmaceutical Technology, Goethe University Frankfurt am Main, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
| | - Irena Tomaszewska
- Pfizer Ltd., Discovery Park House, Sandwich, Kent CT13 9NJ, England, United Kingdom
| | - Uwe Muenster
- Bayer Pharma AG, Research Center Wuppertal-Aprath, Wuppertal, Germany
| | | | - Wolfgang Mueck
- Bayer Pharma AG, Research Center Wuppertal-Aprath, Wuppertal, Germany
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University Frankfurt am Main, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
125
|
Peters SA, Jones CR, Ungell AL, Hatley OJD. Predicting Drug Extraction in the Human Gut Wall: Assessing Contributions from Drug Metabolizing Enzymes and Transporter Proteins using Preclinical Models. Clin Pharmacokinet 2016; 55:673-96. [PMID: 26895020 PMCID: PMC4875961 DOI: 10.1007/s40262-015-0351-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Frankfurter Str. 250, F130/005, 64293, Darmstadt, Germany.
| | | | - Anna-Lena Ungell
- Investigative ADME, Non-Clinical Development, UCB New Medicines, BioPharma SPRL, Braine l'Alleud, Belgium
| | - Oliver J D Hatley
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
126
|
Kumagai T, Aratsu Y, Sugawara R, Sasaki T, Miyairi S, Nagata K. Indirubin, a component of Ban-Lan-Gen, activates CYP3A4 gene transcription through the human pregnane X receptor. Drug Metab Pharmacokinet 2016; 31:139-45. [DOI: 10.1016/j.dmpk.2016.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 12/20/2022]
|
127
|
Vanhove T, Annaert P, Kuypers DRJ. Clinical determinants of calcineurin inhibitor disposition: a mechanistic review. Drug Metab Rev 2016; 48:88-112. [DOI: 10.3109/03602532.2016.1151037] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
128
|
Foufelle F, Fromenty B. Role of endoplasmic reticulum stress in drug-induced toxicity. Pharmacol Res Perspect 2016; 4:e00211. [PMID: 26977301 PMCID: PMC4777263 DOI: 10.1002/prp2.211] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Drug‐induced toxicity is a key issue for public health because some side effects can be severe and life‐threatening. These adverse effects can also be a major concern for the pharmaceutical companies since significant toxicity can lead to the interruption of clinical trials, or the withdrawal of the incriminated drugs from the market. Recent studies suggested that endoplasmic reticulum (ER) stress could be an important event involved in drug liability, in addition to other key mechanisms such as mitochondrial dysfunction and oxidative stress. Indeed, drug‐induced ER stress could lead to several deleterious effects within cells and tissues including accumulation of lipids, cell death, cytolysis, and inflammation. After recalling important information regarding drug‐induced adverse reactions and ER stress in diverse pathophysiological situations, this review summarizes the main data pertaining to drug‐induced ER stress and its potential involvement in different adverse effects. Drugs presented in this review are for instance acetaminophen (APAP), arsenic trioxide and other anticancer drugs, diclofenac, and different antiretroviral compounds. We also included data on tunicamycin (an antibiotic not used in human medicine because of its toxicity) and thapsigargin (a toxic compound of the Mediterranean plant Thapsia garganica) since both molecules are commonly used as prototypical toxins to induce ER stress in cellular and animal models.
Collapse
|
129
|
Yi JM, Kim YA, Lee YJ, Bang OS, Kim NS. Effect of an ethanol extract of Descurainia sophia seeds on Phase I and II drug metabolizing enzymes and P-glycoprotein activity in vitro. Altern Ther Health Med 2015; 15:441. [PMID: 26683337 PMCID: PMC4683934 DOI: 10.1186/s12906-015-0965-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/09/2015] [Indexed: 12/29/2022]
Abstract
Background Descurainia sophia seeds have a variety of pharmacological functions and been widely used in traditional folk medicine. However, their effects on human drug metabolizing enzyme (DME) activities have not been elucidated. The present study investigated the inhibitory effects of an ethanol extract of D. sophia seeds (EEDS) on human Phase I/II (DMEs) and P-glycoprotein (p-gp) in vitro. Methods The enzyme activities of human Phase I (cytochrome P450s, CYPs), Phase II (uridine diphosphate glucuronosyltransferases, UGTs) DMEs, and the drug transporter P-gp were determined in the presence of various concentrations of EEDS using commercially available luminogenic assay systems. The mode of enzyme inhibition and the inhibitory constant (Ki) value of EEDS were graphically determined with Lineweaver-Burk double reciprocal plots and secondary plots, respectively. Results The enzyme activity assays showed that EEDS moderately inhibited the CYP1A2, CYP2C9, and CYP2C19 isoforms with half maximal inhibitory concentrations (IC50) of 47.3, 25.8, and 38.7 μg/mL, respectively. Graphical analyses with Lineweaver-Burk double reciprocal plots and secondary plots indicated that EEDS competitively inhibited CYP2C9 with a Ki value of 19.8 μg/mL; however, it inhibited CYP2C9 and CYP2C19 in a mixed mode with Ki values of 5.2, and 11.9 μg/mL, respectively. Other Phase I (CYP2C8, CYP2D6, and CYP3A4) and Phase II (UGT1A1 and UGT2B7) enzymes as well as P-gp were weakly or negligibly affected by EEDS with concentrations up to 500 μg/mL. Conclusions EEDS is a selective inhibitor of CYP1A2, CYP2C9, and CYP2C19 with moderate enzymatic inhibition. Clinically, full consideration should be given to a potential toxic adverse effect from a herb-drug interaction when drugs that are particularly susceptible to CYP1A2, CYP2C9, or CYP2C19-mediated metabolism are taken together with EEDS. Characterization of metabolic profiles of specific herbal drugs could help consumers and medical specialists to use them safely as a complementary and alternative medicine.
Collapse
|
130
|
Gene copy number variation analysis reveals dosage-insensitive expression of CYP2E1. THE PHARMACOGENOMICS JOURNAL 2015; 16:551-558. [PMID: 26503817 DOI: 10.1038/tpj.2015.69] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 01/21/2023]
Abstract
Gene copy number variants (CNVs) of CYP2E1 have been described but not functionally characterized. Here we investigated effects of CNVs on hepatic and lymphoblastoid CYP2E1 expression. Using available single-nuleotide polymorphism microarray data and quantitative PCR, CYP2E1 gene duplication and deletion carriers were identified. CYP2E1 mRNA, protein and enzyme activity (chlorzoxazone-6-hydroxylation) phenotypes of CYP2E1 were not associated with gene copy number. Analysis of gene expression in lymphoblastoid cell lines in relation to CNV confirmed this finding in an extrahepatic tissue and for other ethnicities. Further analyses identified a linked haplotype cluster with possible influence on gene expression. In summary, our data suggest a homeostatic, gene dosage-insensitive regulation of CYP2E1 expression by unknown gene dosage compensation mechanisms. This is in striking contrast to well-known structural variations of CYP2A6 and CYP2D6 that have a strong impact on expression and activity. These findings are important in the context of pharmacogenetic prediction.
Collapse
|
131
|
Roerig JL, Steffen K. Psychopharmacology and Bariatric Surgery. EUROPEAN EATING DISORDERS REVIEW 2015; 23:463-9. [PMID: 26338011 DOI: 10.1002/erv.2396] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/30/2015] [Indexed: 12/20/2022]
Abstract
Currently, it has been demonstrated that psychotropic drugs, particularly antidepressants, are frequently prescribed for patients who seek bariatric surgery. Many bariatric surgery patients have a history of a mood disorder. Unlike medications for diabetes, hypertension or hyperlipidemia, which are generally reduced and at times discontinued, postsurgery antidepressants use is only slightly reduced. The Roux-en-Y procedure is most frequently associated with alteration in drug exposure. Medication disintegration, dissolution, absorption, metabolism and excretion have been found to be altered in postbariatric patients, although data are sparse at this time. This paper will review the current evidence regarding the effect of bariatric surgery on drug treatment including mechanism of interference as well as the extent of changes identified to date. Data will be presented as controlled trials followed by case series and reports.
Collapse
Affiliation(s)
- James L Roerig
- Department of Psychiatry and Behavioral Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA.,Neuropsychiatric Research Institute, Fargo, ND, USA
| | - Kristine Steffen
- Neuropsychiatric Research Institute, Fargo, ND, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, North Dakota State University, Grand Forks, ND, USA
| |
Collapse
|
132
|
Nguyen MA, Staubach P, Wolffram S, Langguth P. The Influence of Single-Dose and Short-Term Administration of Quercetin on the Pharmacokinetics of Midazolam in Humans. J Pharm Sci 2015; 104:3199-207. [DOI: 10.1002/jps.24500] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023]
|
133
|
Hatton GB, Yadav V, Basit AW, Merchant HA. Animal Farm: Considerations in Animal Gastrointestinal Physiology and Relevance to Drug Delivery in Humans. J Pharm Sci 2015; 104:2747-76. [DOI: 10.1002/jps.24365] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 12/30/2022]
|
134
|
Barbieri S, Buttini F, Rossi A, Bettini R, Colombo P, Ponchel G, Sonvico F, Colombo G. Ex vivo permeation of tamoxifen and its 4-OH metabolite through rat intestine from lecithin/chitosan nanoparticles. Int J Pharm 2015; 491:99-104. [DOI: 10.1016/j.ijpharm.2015.06.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/12/2015] [Accepted: 06/13/2015] [Indexed: 12/17/2022]
|
135
|
Foo WYB, Tay HY, Chan ECY, Lau AJ. Meclizine, a pregnane X receptor agonist, is a direct inhibitor and mechanism-based inactivator of human cytochrome P450 3A. Biochem Pharmacol 2015; 97:320-30. [PMID: 26239802 DOI: 10.1016/j.bcp.2015.07.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/29/2015] [Indexed: 12/15/2022]
Abstract
Meclizine is an agonist of human pregnane X receptor (PXR). It increases CYP3A4 mRNA expression, but decreases CYP3A-catalyzed testosterone 6β-hydroxylation in primary cultures of human hepatocytes, as assessed at 24h after the last dose of meclizine. Therefore, the hypothesis to be tested is that meclizine inactivates human CYP3A enzymes. Our findings indicated that meclizine directly inhibited testosterone 6β-hydroxylation catalyzed by human liver microsomes, recombinant CYP3A4, and recombinant CYP3A5. The inhibition of human liver microsomal testosterone 6β-hydroxylation by meclizine occurred by a mixed mode and with an apparent Ki of 31±6μM. Preincubation of meclizine with human liver microsomes and NADPH resulted in a time- and concentration-dependent decrease in testosterone 6β-hydroxylation. The extent of inactivation required the presence of NADPH, was unaffected by nucleophilic trapping agents or reactive oxygen species scavengers, attenuated by a CYP3A substrate, and not reversed by dialysis. Meclizine selectively inactivated CYP3A4, but not CYP3A5. In contrast to meclizine, which has a di-substituted piperazine ring, norchlorcyclizine, which is a N-debenzylated meclizine metabolite with a mono-substituted piperazine ring, did not inactivate but directly inhibited hepatic microsomal CYP3A activity. In conclusion, meclizine inhibited human CYP3A enzymes by both direct inhibition and mechanism-based inactivation. In contrast, norchlorcyclizine is a direct inhibitor but not a mechanism-based inactivator. Furthermore, a PXR agonist may also be an inhibitor of a PXR-regulated enzyme, thereby giving rise to opposing effects on the functional activity of the enzyme and indicating the importance of measuring the catalytic activity of nuclear receptor-regulated enzymes.
Collapse
Affiliation(s)
- Winnie Yin Bing Foo
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | - Hwee Ying Tay
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | - Aik Jiang Lau
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
136
|
Ogaki S, Taguchi K, Maeda H, Watanabe H, Ishima Y, Otagiri M, Maruyama T. Kupffer cell inactivation by carbon monoxide bound to red blood cells preserves hepatic cytochrome P450 via anti-oxidant and anti-inflammatory effects exerted through the HMGB1/TLR-4 pathway during resuscitation from hemorrhagic shock. Biochem Pharmacol 2015; 97:310-9. [PMID: 26232728 DOI: 10.1016/j.bcp.2015.07.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/27/2015] [Indexed: 12/18/2022]
Abstract
Red blood cell (RBC) transfusions for controlling hemorrhaging induce systemic ischemia reperfusion, resulting in a decrease in hepatic cytochrome P450 (CYP) levels. Carbon monoxide (CO), when bound to red blood cells (CO-RBC) has the potential to protect the hepatic CYP protein to produce a resuscitative effect in a hemorrhagic shock rat model. The aim of this study was to investigate the mechanism by which CO-RBC resuscitation from a massive hemorrhage protects against a decrease in hepatic CYP. In the early phase (∼1h) after a hemorrhage and RBC resuscitation, hepatic CYP protein levels were significantly decreased with increasing hepatic free heme levels, but were maintained by a pre-treatment of gadolinium chloride (GdCl3), a Kupffer cell inhibitor, and Trolox, an anti-oxidant agent, as well as CO-RBC resuscitation. Under these conditions, the production of reactive oxygen species (ROS) derived from activated Kupffer cells was increased, but this increase was suppressed by CO-RBC resuscitation. At a late phase (6∼24h), CYP mRNA levels decreased after hemorrhage and RBC resuscitation, but not in the case of CO-RBC resuscitation. The increases in plasma IL-6 and TNF-α levels were decreased by CO-RBC resuscitation via the suppression of the toll-like receptor-4 (TLR-4) and the expression of the high mobility group box-1 (HMGB-1). Hepatic CYP protection after a hemorrhage and CO-RBC resuscitation can be attributed to the inactivation of Kupffer cells, resulting in the suppression of ROS production in the early phase and the suppression of inflammatory cytokine production via the TLR-4/HMGB-1signal pathway in the late phase.
Collapse
Affiliation(s)
- Shigeru Ogaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan
| | - Kazuaki Taguchi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, 860-0082 Kumamoto, Japan
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan,; Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan
| | - Yu Ishima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan,; Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan
| | - Masaki Otagiri
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan,; Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, 860-0082 Kumamoto, Japan,; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, 860-0082 Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan,; Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973 Kumamoto, Japan,.
| |
Collapse
|
137
|
Kwon SS, Kim JH, Jeong HU, Ahn KS, Oh SR, Lee HS. Role of cytochrome P450 and UDP-glucuronosyltransferases in metabolic pathway of homoegonol in human liver microsomes. Drug Metab Pharmacokinet 2015; 30:305-13. [PMID: 26163112 DOI: 10.1016/j.dmpk.2015.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022]
Abstract
Homoegonol is being evaluated for the development of a new antiasthmatic drug. Based on a pharmacokinetic study of homoegonol in rats, homoegonol is almost completely eliminated via metabolism, but no study on its metabolism has been reported in animals and humans. Incubation of homoegonol in human liver microsomes in the presence of the reduced form of nicotinamide adenine dinucleotide phosphate and UDP-glucuronic acid resulted in the formation of five metabolites: 4-O-demethylhomoegonol (M1), hydroxyhomoegonol (M2 and M3), 4-O-demethylhomoegonol glucuronide (M4), and homoegonol glucuronide (M5). We characterized the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes responsible for homoegonol metabolism using human liver microsomes, and cDNA-expressed CYP and UGT enzymes. CYP1A2 played a more prominent role than CYP3A4 and CYP2D6 in the 4-O-demethylation of homoegonol to M1. CYP3A4 was responsible for the hydroxylation of homoegonol to M2. The hydroxylation of homoegonol to M3 was insufficient to characterize CYP enzymes. Glucuronidation of homoegonol to M5 was mediated by UGT1A1, UGT1A3, UGT1A4, and UGT2B7 enzymes, whereas M4 was formed from 4-O-demethylhomoegonol by UGT1A1, UGT1A8, UGT1A10, and UGT2B15 enzymes.
Collapse
Affiliation(s)
- Soon Sang Kwon
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Republic of Korea
| | - Ju Hyun Kim
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Republic of Korea
| | - Hyeon-Uk Jeong
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk 363-883, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk 363-883, Republic of Korea
| | - Hye Suk Lee
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Republic of Korea.
| |
Collapse
|
138
|
Dorokhov YL, Shindyapina AV, Sheshukova EV, Komarova TV. Metabolic methanol: molecular pathways and physiological roles. Physiol Rev 2015; 95:603-44. [PMID: 25834233 DOI: 10.1152/physrev.00034.2014] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Methanol has been historically considered an exogenous product that leads only to pathological changes in the human body when consumed. However, in normal, healthy individuals, methanol and its short-lived oxidized product, formaldehyde, are naturally occurring compounds whose functions and origins have received limited attention. There are several sources of human physiological methanol. Fruits, vegetables, and alcoholic beverages are likely the main sources of exogenous methanol in the healthy human body. Metabolic methanol may occur as a result of fermentation by gut bacteria and metabolic processes involving S-adenosyl methionine. Regardless of its source, low levels of methanol in the body are maintained by physiological and metabolic clearance mechanisms. Although human blood contains small amounts of methanol and formaldehyde, the content of these molecules increases sharply after receiving even methanol-free ethanol, indicating an endogenous source of the metabolic methanol present at low levels in the blood regulated by a cluster of genes. Recent studies of the pathogenesis of neurological disorders indicate metabolic formaldehyde as a putative causative agent. The detection of increased formaldehyde content in the blood of both neurological patients and the elderly indicates the important role of genetic and biochemical mechanisms of maintaining low levels of methanol and formaldehyde.
Collapse
Affiliation(s)
- Yuri L Dorokhov
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia; and N. I. Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russia
| | - Anastasia V Shindyapina
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia; and N. I. Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russia
| | - Ekaterina V Sheshukova
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia; and N. I. Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russia
| | - Tatiana V Komarova
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia; and N. I. Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
139
|
Pinheiro P, Marinho A, Antunes A, Marques M, Pereira S, Miranda J. Sex differences in hepatic and intestinal contributions to nevirapine biotransformation in rats. Chem Biol Interact 2015; 233:115-21. [DOI: 10.1016/j.cbi.2015.03.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/10/2015] [Accepted: 03/22/2015] [Indexed: 01/01/2023]
|
140
|
Van Landeghem L, Santoro MA, Mah AT, Krebs AE, Dehmer JJ, McNaughton KK, Helmrath MA, Magness ST, Lund PK. IGF1 stimulates crypt expansion via differential activation of 2 intestinal stem cell populations. FASEB J 2015; 29:2828-42. [PMID: 25837582 DOI: 10.1096/fj.14-264010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/06/2015] [Indexed: 01/24/2023]
Abstract
Insulin-like growth factor 1 (IGF1) has potent trophic effects on normal or injured intestinal epithelium, but specific effects on intestinal stem cells (ISCs) are undefined. We used Sox9-enhanced green fluorescent protein (EGFP) reporter mice that permit analyses of both actively cycling ISCs (Sox9-EGFP(Low)) and reserve/facultative ISCs (Sox9-EGFP(High)) to study IGF1 action on ISCs in normal intestine or during crypt regeneration after high-dose radiation-induced injury. We hypothesized that IGF1 differentially regulates proliferation and gene expression in actively cycling and reserve/facultative ISCs. IGF1 was delivered for 5 days using subcutaneously implanted mini-pumps in uninjured mice or after 14 Gy abdominal radiation. ISC numbers, proliferation, and transcriptome were assessed. IGF1 increased epithelial growth in nonirradiated mice and enhanced crypt regeneration after radiation. In uninjured and regenerating intestines, IGF1 increased total numbers of Sox9-EGFP(Low) ISCs and percentage of these cells in M-phase. IGF1 increased percentages of Sox9-EGFP(High) ISCs in S-phase but did not expand this population. Microarray revealed that IGF1 activated distinct gene expression signatures in the 2 Sox9-EGFP ISC populations. In vitro IGF1 enhanced enteroid formation by Sox9-EGFP(High) facultative ISCs but not Sox9-EGFP(Low) actively cycling ISCs. Our data provide new evidence that IGF1 activates 2 ISC populations via distinct regulatory pathways to promote growth of normal intestinal epithelium and crypt regeneration after irradiation.
Collapse
Affiliation(s)
- Laurianne Van Landeghem
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - M Agostina Santoro
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Amanda T Mah
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Adrienne E Krebs
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Jeffrey J Dehmer
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Kirk K McNaughton
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Michael A Helmrath
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Scott T Magness
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - P Kay Lund
- *Department of Cell Biology and Physiology, Department of Nutrition, Department of Surgery, and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA; and University of North Carolina/North Carolina State Biomedical Engineering, Chapel Hill, North Carolina, USA
| |
Collapse
|
141
|
Herbrink M, Nuijen B, Schellens JHM, Beijnen JH. Variability in bioavailability of small molecular tyrosine kinase inhibitors. Cancer Treat Rev 2015; 41:412-22. [PMID: 25818541 DOI: 10.1016/j.ctrv.2015.03.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 01/23/2023]
Abstract
Small molecular tyrosine kinase inhibitors (smTKIs) are in the centre of the very quickly expanding area of personalized chemotherapy and oral applicability thereof. The number of drugs in this class is rapidly growing, with twenty current approvals by both the European Medicines Agency (EMA) and the Food and Drug Administration (FDA). The drugs are, however, generally characterized by a poor oral, and thus variable, bioavailability. This results in significant variation in plasma levels and exposure. The cause is a complex interplay of factors, including poor aqueous solubility, issued permeability, membrane transport and enzymatic metabolism. Additionally, food and drug-drug interactions can play a significant role. The issues related with an impaired bioavailability generally receive little attention. To the best of our knowledge, this article is the first to provide an overview of the factors that determine the bioavailability of the smTKIs.
Collapse
Affiliation(s)
- Maikel Herbrink
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Louwesweg 6, 1006 BK Amsterdam, The Netherlands.
| | - Bastiaan Nuijen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Louwesweg 6, 1006 BK Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Louwesweg 6, 1006 BK Amsterdam, The Netherlands; Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Louwesweg 6, 1006 BK Amsterdam, The Netherlands; Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
142
|
Ye N, Tang M, Ye H, Wang C, Wang C, Yang Q, Wan L, Chen L. 13C stable isotope labeling followed by ultra-high performance liquid chromatography/quadrupole time-of-flight tandem mass spectrometry (UHPLC/Q-TOF MS) was applied to identify the metabolites of honokiol in rat small intestines. ANALYTICAL METHODS 2015. [DOI: 10.1039/c4ay02403f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Honokiol, as a pharmacological active small-molecule, has received significant attention for its strong pharmacological effects without remarkable toxicity.
Collapse
Affiliation(s)
- Na Ye
- School of Pharmacy
- Chengdu University of TCM
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
| | - Minghai Tang
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu 610041
| | - Haoyu Ye
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu 610041
| | - Chunyan Wang
- School of Pharmacy
- Chengdu University of TCM
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
| | - Chunyu Wang
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu 610041
| | - Qiunan Yang
- School of Pharmacy
- Chengdu University of TCM
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
| | - Li Wan
- School of Pharmacy
- Chengdu University of TCM
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
| | - Lijuan Chen
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu 610041
| |
Collapse
|
143
|
Zelasko S, Arnold WR, Das A. Endocannabinoid metabolism by cytochrome P450 monooxygenases. Prostaglandins Other Lipid Mediat 2014; 116-117:112-23. [PMID: 25461979 DOI: 10.1016/j.prostaglandins.2014.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 01/01/2023]
Abstract
The endogenous cannabinoid system was first uncovered following studies of the recreational drug Cannabis sativa. It is now recognized as a vital network of signaling pathways that regulate several physiological processes. Following the initial discovery of the cannabinoid receptors 1 (CB1) and 2 (CB2), activated by Cannabis-derived analogs, many endogenous fatty acids termed "endocannabinoids" are now known to be partial agonists of the CB receptors. At present, the most thoroughly studied endocannabinoid signaling molecules are anandamide (AEA) and 2-arachidonylglycerol (2-AG), which are both derived from arachidonic acid. Both AEA and 2-AG are also substrates for the eicosanoid-synthesizing pathways, namely, certain cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes. In the past, research in the endocannabinoid field focused on the interaction of AEA and 2-AG with the COX and LOX enzymes, but accumulating evidence also points to the involvement of CYPs in modulating endocannabinoid signaling. The focus of this review is to explore the current understanding of CYP-mediated metabolism of endocannabinoids.
Collapse
Affiliation(s)
- Susan Zelasko
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States
| | - William R Arnold
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States
| | - Aditi Das
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States; Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61802, United States.
| |
Collapse
|
144
|
Shahabi P, Siest G, Meyer UA, Visvikis-Siest S. Human cytochrome P450 epoxygenases: Variability in expression and role in inflammation-related disorders. Pharmacol Ther 2014; 144:134-61. [DOI: 10.1016/j.pharmthera.2014.05.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/15/2014] [Indexed: 12/19/2022]
|
145
|
Tydén E, Tjälve H, Larsson P. Gene and protein expression and cellular localisation of cytochrome P450 enzymes of the 1A, 2A, 2C, 2D and 2E subfamilies in equine intestine and liver. Acta Vet Scand 2014; 56:69. [PMID: 25288196 PMCID: PMC4192735 DOI: 10.1186/s13028-014-0069-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/03/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Among the cytochrome P450 enzymes (CYP), families 1-3 constitute almost half of total CYPs in mammals and play a central role in metabolism of a wide range of pharmaceuticals. This study investigated gene and protein expression and cellular localisation of CYP1A, CYP2A, CYP2C, CYP2D and CYP2E in equine intestine and liver. Real-time polymerase chain reaction (RT-PCR) was used to analyse gene expression, western blot to examine protein expression and immunohistochemical analyses to investigate cellular localisation. RESULTS CYP1A and CYP2C were the CYPs with the highest gene expression in the intestine and also showed considerable gene expression in the liver. CYP2E and CYP2A showed the highest gene expression in the liver. CYP2E showed moderate intestinal gene expression, whereas that of CYP2A was very low or undetectable. For CYP2D, rather low gene expression levels were found in both intestine and the liver. In the intestine, CYP gene expression levels, except for CYP2E, exhibited patterns resembling those of the proteins, indicating that intestinal protein expression of these CYPs is regulated at the transcriptional level. For CYP2E, the results showed that the intestinal gene expression did not correlate to any visible protein expression, indicating that intestinal protein expression of this CYP is regulated at the post-transcriptional level. Immunostaining of intestine tissue samples showed preferential CYP staining in enterocytes at the tips of intestinal villi in the small intestine. In the liver, all CYPs showed preferential localisation in the centrilobular hepatocytes. CONCLUSIONS Overall, different gene expression profiles were displayed by the CYPs examined in equine intestine and liver. The CYPs present in the intestine may act in concert with those in the liver to affect the oral bioavailability and therapeutic efficiency of substrate drugs. In addition, they may play a role in first-pass metabolism of feed constituents and of herbal supplements used in equine practice.
Collapse
Affiliation(s)
- Eva Tydén
- Department of Biomedical Sciences and Veterinary Public Health, Division of Pathology, Pharmacology and Toxicology, Swedish University of Agricultural Sciences, S-750 07 Uppsala, Sweden
| | - Hans Tjälve
- Department of Biomedical Sciences and Veterinary Public Health, Division of Pathology, Pharmacology and Toxicology, Swedish University of Agricultural Sciences, S-750 07 Uppsala, Sweden
| | - Pia Larsson
- Department of Biomedical Sciences and Veterinary Public Health, Division of Pathology, Pharmacology and Toxicology, Swedish University of Agricultural Sciences, S-750 07 Uppsala, Sweden
| |
Collapse
|
146
|
Yang X, Morris SM, Gearhart JM, Ruark CD, Paule MG, Slikker W, Mattison DR, Vitiello B, Twaddle NC, Doerge DR, Young JF, Fisher JW. Development of a physiologically based model to describe the pharmacokinetics of methylphenidate in juvenile and adult humans and nonhuman primates. PLoS One 2014; 9:e106101. [PMID: 25184666 PMCID: PMC4153582 DOI: 10.1371/journal.pone.0106101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
The widespread usage of methylphenidate (MPH) in the pediatric population has received considerable attention due to its potential effect on child development. For the first time a physiologically based pharmacokinetic (PBPK) model has been developed in juvenile and adult humans and nonhuman primates to quantitatively evaluate species- and age-dependent enantiomer specific pharmacokinetics of MPH and its primary metabolite ritalinic acid. The PBPK model was first calibrated in adult humans using in vitro enzyme kinetic data of MPH enantiomers, together with plasma and urine pharmacokinetic data with MPH in adult humans. Metabolism of MPH in the small intestine was assumed to account for the low oral bioavailability of MPH. Due to lack of information, model development for children and juvenile and adult nonhuman primates primarily relied on intra- and interspecies extrapolation using allometric scaling. The juvenile monkeys appear to metabolize MPH more rapidly than adult monkeys and humans, both adults and children. Model prediction performance is comparable between juvenile monkeys and children, with average root mean squared error values of 4.1 and 2.1, providing scientific basis for interspecies extrapolation of toxicity findings. Model estimated human equivalent doses in children that achieve similar internal dose metrics to those associated with pubertal delays in juvenile monkeys were found to be close to the therapeutic doses of MPH used in pediatric patients. This computational analysis suggests that continued pharmacovigilance assessment is prudent for the safe use of MPH.
Collapse
Affiliation(s)
- Xiaoxia Yang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| | - Suzanne M. Morris
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffery M. Gearhart
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Christopher D. Ruark
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Merle G. Paule
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - William Slikker
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Donald R. Mattison
- Risk Sciences International, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Benedetto Vitiello
- National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Nathan C. Twaddle
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Daniel R. Doerge
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - John F. Young
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffrey W. Fisher
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| |
Collapse
|
147
|
Wetmore BA, Allen B, Clewell HJ, Parker T, Wambaugh JF, Almond LM, Sochaski MA, Thomas RS. Incorporating population variability and susceptible subpopulations into dosimetry for high-throughput toxicity testing. Toxicol Sci 2014; 142:210-24. [PMID: 25145659 DOI: 10.1093/toxsci/kfu169] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Momentum is growing worldwide to use in vitro high-throughput screening (HTS) to evaluate human health effects of chemicals. However, the integration of dosimetry into HTS assays and incorporation of population variability will be essential before its application in a risk assessment context. Previously, we employed in vitro hepatic metabolic clearance and plasma protein binding data with in vitro in vivo extrapolation (IVIVE) modeling to estimate oral equivalent doses, or daily oral chemical doses required to achieve steady-state blood concentrations (Css) equivalent to media concentrations having a defined effect in an in vitro HTS assay. In this study, hepatic clearance rates of selected ToxCast chemicals were measured in vitro for 13 cytochrome P450 and five uridine 5'-diphospho-glucuronysyltransferase isozymes using recombinantly expressed enzymes. The isozyme-specific clearance rates were then incorporated into an IVIVE model that captures known differences in isozyme expression across several life stages and ethnic populations. Comparison of the median Css for a healthy population against the median or the upper 95th percentile for more sensitive populations revealed differences of 1.3- to 4.3-fold or 3.1- to 13.1-fold, respectively. Such values may be used to derive chemical-specific human toxicokinetic adjustment factors. The IVIVE model was also used to estimate subpopulation-specific oral equivalent doses that were directly compared with subpopulation-specific exposure estimates. This study successfully combines isozyme and physiologic differences to quantitate subpopulation pharmacokinetic variability. Incorporation of these values with dosimetry and in vitro bioactivities provides a viable approach that could be employed within a high-throughput risk assessment framework.
Collapse
Affiliation(s)
- Barbara A Wetmore
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| | - Brittany Allen
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| | - Harvey J Clewell
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| | - Timothy Parker
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| | - John F Wambaugh
- United States Environmental Protection Agency, Office of Research and Development, National Center for Computational Toxicology, Research Triangle Park, North Carolina 27711
| | - Lisa M Almond
- Simcyp Limited (a Certara company), Blades Enterprise Centre, John Street, Sheffield S2 4SU, UK
| | - Mark A Sochaski
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| | - Russell S Thomas
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709-2137
| |
Collapse
|
148
|
Cruz HG, Hay JL, Hoever P, Alessi F, te Beek ET, van Gerven JMA, Dingemanse J. Pharmacokinetic and pharmacodynamic interactions between almorexant, a dual orexin receptor antagonist, and desipramine. Eur Neuropsychopharmacol 2014; 24:1257-68. [PMID: 24880753 DOI: 10.1016/j.euroneuro.2014.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 04/03/2014] [Accepted: 05/01/2014] [Indexed: 01/18/2023]
Abstract
Almorexant is a dual orexin receptor antagonist (DORA) with sleep-enabling effects in humans. Insomnia is often associated with mental health problems, including depression. Hence, potential interactions with antidepressants deserve attention. Desipramine was selected as a model drug because it is mainly metabolized by CYP2D6, which is inhibited by almorexant in vitro. A single-center, randomized, placebo-controlled, two-way crossover study in 20 healthy male subjects was conducted to evaluate the pharmacokinetic and pharmacodynamic interactions between almorexant and desipramine. Almorexant 200mg or matching placebo (double-blind) was administered orally once daily in the morning for 10 days, and a single oral dose of 50mg desipramine (open-label) was administered on Day 5. Almorexant increased the exposure to desipramine 3.7-fold, suggesting that almorexant is a moderate inhibitor of desipramine metabolism through inhibition of CYP2D6. Conversely, desipramine showed no relevant effects on the pharmacokinetics of almorexant. Pharmacodynamic evaluations indicated that almorexant alone reduced visuomotor coordination, postural stability, and alertness, and slightly increased calmness. Desipramine induced a reduction in subjective alertness and an increase in pupil/iris ratio. Despite the increase in exposure to desipramine, almorexant and desipramine in combination showed the same pharmacodynamic profile as almorexant alone, except for prolonging reduced alertness and preventing the miotic effect of almorexant. Co-administration also prolonged the mydriatic effect of desipramine. Overall, repeated administration of almorexant alone or with single-dose desipramine was well tolerated. The lack of a relevant interaction with antidepressants, if confirmed for other DORAs, would be a key feature for a safer class of hypnotics.
Collapse
Affiliation(s)
- Hans G Cruz
- Actelion Pharmaceuticals Ltd, Clinical Pharmacology, Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Justin L Hay
- Centre for Human Drug Research, Leiden, The Netherlands
| | - Petra Hoever
- Actelion Pharmaceuticals Ltd, Clinical Pharmacology, Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Federica Alessi
- Actelion Pharmaceuticals Ltd, Biostatistics, Allschwil, Switzerland
| | | | | | - Jasper Dingemanse
- Actelion Pharmaceuticals Ltd, Clinical Pharmacology, Gewerbestrasse 16, CH-4123 Allschwil, Switzerland.
| |
Collapse
|
149
|
Sjögren E, Abrahamsson B, Augustijns P, Becker D, Bolger MB, Brewster M, Brouwers J, Flanagan T, Harwood M, Heinen C, Holm R, Juretschke HP, Kubbinga M, Lindahl A, Lukacova V, Münster U, Neuhoff S, Nguyen MA, Peer AV, Reppas C, Hodjegan AR, Tannergren C, Weitschies W, Wilson C, Zane P, Lennernäs H, Langguth P. In vivo methods for drug absorption – Comparative physiologies, model selection, correlations with in vitro methods (IVIVC), and applications for formulation/API/excipient characterization including food effects. Eur J Pharm Sci 2014; 57:99-151. [PMID: 24637348 DOI: 10.1016/j.ejps.2014.02.010] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 01/11/2023]
|
150
|
Perspective and potential of oral lipid-based delivery to optimize pharmacological therapies against cardiovascular diseases. J Control Release 2014; 193:174-87. [PMID: 24852093 DOI: 10.1016/j.jconrel.2014.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/04/2014] [Accepted: 05/07/2014] [Indexed: 02/01/2023]
Abstract
Cardiovascular diseases (CVDs) remain the major cause of morbidity and mortality globally. Despite the large number of cardiovascular drugs available for pharmacological therapies, factors limiting the efficient oral use are identified, including low water solubility, pre-systemic metabolism, food intake effects and short half-life. Numerous in vivo proof-of-concepts studies are presented to highlight the viability of lipid-based delivery to optimize the oral delivery of cardiovascular drugs. In particular, the key performance enhancement roles of oral lipid-based drug delivery systems (LBDDSs) are identified, which include i) improving the oral bioavailability, ii) sustaining/controlling drug release, iii) improving drug stability, iv) reducing food intake effect, v) targeting to injured sites, and vi) potential for combination therapy. Mechanisms involved in achieving these features, range of applicability, and limits of available systems are detailed. Future research and development efforts to address these issues are discussed, which is of significant value in directing future research work in fostering translation of lipid-based formulations into clinical applications to reduce the prevalence of CVDs.
Collapse
|