1
|
Hou Z, Zhang L, Guo X, Li H, Li X, Dong J, Zhang G, Liu D. A method for quantification of human transporter proteins by liquid chromatography–tandem mass spectrometry. SEPARATION SCIENCE PLUS 2023. [DOI: 10.1002/sscp.202200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Affiliation(s)
- Zhe Hou
- Institute of Medical Innovation and Research Peking University Third Hospital Beijing P. R. China
- School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing P. R. China
| | - Lei Zhang
- Institute of Medical Innovation and Research Peking University Third Hospital Beijing P. R. China
- Department of Cardiology and Institute of Vascular Medicine Peking University Third Hospital Beijing P. R. China
- Medical Metabolomics Center Center for Reproductive Medicine Peking University Third Hospital Beijing P. R. China
| | - Xuan Guo
- Institute of Medical Innovation and Research Peking University Third Hospital Beijing P. R. China
- School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing P. R. China
| | - Hui Li
- Institute of Medical Innovation and Research Peking University Third Hospital Beijing P. R. China
- School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing P. R. China
| | - Xiaodong Li
- Shimadzu China Innovation Center Beijing P. R. China
| | - Jing Dong
- Shimadzu China Innovation Center Beijing P. R. China
| | - Guangqin Zhang
- School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing P. R. China
| | - Dongyang Liu
- Institute of Medical Innovation and Research Peking University Third Hospital Beijing P. R. China
| |
Collapse
|
2
|
Buyssens L, Valenzuela A, Prims S, Ayuso M, Thymann T, Van Ginneken C, Van Cruchten S. Ontogeny of CYP3A and UGT activity in preterm piglets: a translational model for drug metabolism in preterm newborns. Front Pharmacol 2023; 14:1177541. [PMID: 37124224 PMCID: PMC10133700 DOI: 10.3389/fphar.2023.1177541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Despite considerable progress in understanding drug metabolism in the human pediatric population, data remains scarce in preterm neonates. Improving our knowledge of the ADME properties in this vulnerable age group is of utmost importance to avoid suboptimal dosing, which may lead to adverse drug reactions. The juvenile (mini)pig is a representative model for hepatic drug metabolism in human neonates and infants, especially phase I reactions. However, the effect of prematurity on the onset of hepatic phase I and phase II enzyme activity has yet to be investigated in this animal model. Therefore, the aim of this study was to assess the ontogeny of CYP3A and UGT enzyme activity in the liver of preterm (gestational day 105-107) and term-born (gestational day 115-117) domestic piglets. In addition, the ontogeny pattern between the preterm and term group was compared to examine whether postconceptional or postnatal age affects the onset of enzyme activity. The following age groups were included: preterm postnatal day (PND) 0 (n = 10), PND 5 (n = 10), PND 11 (n = 8), PND 26 (n = 10) and term PND 0 (n = 10), PND 5 (n = 10), PND 11 (n = 8), PND 19 (n = 18) and PND 26 (n = 10). Liver microsomes were extracted, and the metabolism of CYP3A and UGT-specific substrates assessed enzyme activity. Preterm CYP3A activity was only detectable at PND 26, whereas term CYP3A activity showed a gradual postnatal increase from PND 11 onwards. UGT activity gradually increased between PND 0 and PND 26 in preterm and term-born piglets, albeit, being systematically lower in the preterm group. Thus, postconceptional age is suggested as the main driver affecting porcine CYP3A and UGT enzyme ontogeny. These data are a valuable step forward in the characterization of the preterm piglet as a translational model for hepatic drug metabolism in the preterm human neonate.
Collapse
Affiliation(s)
- Laura Buyssens
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Allan Valenzuela
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sara Prims
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Miriam Ayuso
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Chris Van Ginneken
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
- *Correspondence: Steven Van Cruchten,
| |
Collapse
|
3
|
El-Khateeb E, Darwich AS, Achour B, Athwal V, Rostami-Hodjegan A. Review article: time to revisit Child-Pugh score as the basis for predicting drug clearance in hepatic impairment. Aliment Pharmacol Ther 2021; 54:388-401. [PMID: 34218453 DOI: 10.1111/apt.16489] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/14/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prescription information for many drugs entering the market lacks dosage guidance for hepatic impairment. Dedicated studies for assessing the fate of drugs in hepatic impairment commonly stratify patients using Child-Pugh score. Child-Pugh is a prognostic clinical score with limitations in reflecting the liver's metabolic capacity. AIMS To demonstrate the need for better drug dosing approaches in hepatic impairment, summarise the current status, identify knowledge gaps related to drug kinetic parameters in hepatic impairment, propose solutions for predicting the liver disease impact on drug exposure and discuss barriers to dosing guidance in those patients. METHODS Relevant reports on dosage adjustment in hepatic impairment were analysed concerning the prediction of the impairment impact on drug kinetics using physiologically-based pharmacokinetic (PBPK) modelling. RESULTS PBPK models are suggested as a potential framework to understand drug clearance changes in hepatic impairment. Quantifying changes in abundance and activity of drug-metabolising enzymes and transporters, understanding the impact of shunting, and accounting for interindividual variations in drug absorption could help in extending the success of these models in hepatically-impaired populations. These variables might not correlate with Child-Pugh score as a whole. Therefore, new metabolic activity markers, imaging techniques and other scoring systems are proposed to either support or substitute Child-Pugh score. CONCLUSIONS Many physiological changes in hepatic impairment determining the fate of drugs do not necessarily correlate with Child-Pugh score. Quantifying these changes in individual patients is essential in future hepatic impairment studies. Further studies assessing Child-Pugh alternatives are recommended to allow better prediction of drug exposure.
Collapse
Affiliation(s)
- Eman El-Khateeb
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Adam S Darwich
- Logistics and Informatics in Health Care, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Varinder Athwal
- Wellcome Centre for Cell-Matrix Research, Division of Diabetes, Endocrinology and Gastroenterology, University of Manchester, Manchester, UK.,Research and Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Certara UK Ltd. (Simcyp Division), Sheffield, UK
| |
Collapse
|
4
|
Buyssens L, De Clerck L, Schelstraete W, Dhaenens M, Deforce D, Ayuso M, Van Ginneken C, Van Cruchten S. Hepatic Cytochrome P450 Abundance and Activity in the Developing and Adult Göttingen Minipig: Pivotal Data for PBPK Modeling. Front Pharmacol 2021; 12:665644. [PMID: 33935788 PMCID: PMC8082684 DOI: 10.3389/fphar.2021.665644] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/26/2021] [Indexed: 12/28/2022] Open
Abstract
The Göttingen Minipig is gaining ground as nonrodent species in safety testing of drugs for pediatric indications. Due to developmental changes in pharmacokinetics and pharmacodynamics, physiologically based pharmacokinetic (PBPK) models are built to better predict drug exposure in children and to aid species selection for nonclinical safety studies. These PBPK models require high quality physiological and ADME data such as protein abundance of drug metabolizing enzymes. These data are available for man and rat, but scarce for the Göttingen Minipig. The aim of this study was to assess hepatic cytochrome P450 (CYP) protein abundance in the developing Göttingen Minipig by using mass spectrometry. In addition, sex-related differences in CYP protein abundance and correlation of CYP enzyme activity with CYP protein abundance were assessed. The following age groups were included: gestational day (GD) 84–86 (n = 8), GD 108 (n = 6), postnatal day (PND) 1 (n = 8), PND 3 (n = 8), PND 7 (n = 8), PND 28 (n = 8) and adult (n = 8). Liver microsomes were extracted and protein abundance was compared to that in adult animals. Next, the CYP protein abundance was correlated to CYP enzyme activity in the same biological samples. In general, CYP protein abundance gradually increased during development. However, we observed a stable protein expression over time for CYP4A24 and CYP20A1 and for CYP51A1, a high protein expression during the fetal stages was followed by a decrease during the first month of life and an increase toward adulthood. Sex-related differences were observed for CYP4V2_2a and CYP20A1 at PND 1 with highest expression in females for both isoforms. In the adult samples, sex-related differences were detected for CYP1A1, CYP1A2, CYP2A19, CYP2E1_2, CYP3A22, CYP4V2_2a and CYP4V2_2b with higher values in female compared to male Göttingen Minipigs. The correlation analysis between CYP protein abundance and CYP enzyme activity showed that CYP3A22 protein abundance correlated clearly with the metabolism of midazolam at PND 7. These data are remarkably comparable to human data and provide a valuable step forward in the construction of a neonatal and juvenile Göttingen Minipig PBPK model.
Collapse
Affiliation(s)
- Laura Buyssens
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Laura De Clerck
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Wim Schelstraete
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Maarten Dhaenens
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Miriam Ayuso
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Chris Van Ginneken
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Comparative Perinatal Development, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
5
|
Sharma S, Suresh Ahire D, Prasad B. Utility of Quantitative Proteomics for Enhancing the Predictive Ability of Physiologically Based Pharmacokinetic Models Across Disease States. J Clin Pharmacol 2020; 60 Suppl 1:S17-S35. [DOI: 10.1002/jcph.1709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Sheena Sharma
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| | - Deepak Suresh Ahire
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| |
Collapse
|
6
|
Physiologically-based pharmacokinetic models for children: Starting to reach maturation? Pharmacol Ther 2020; 211:107541. [DOI: 10.1016/j.pharmthera.2020.107541] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022]
|
7
|
Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Proteomics of Drug-Metabolizing Enzymes and Transporters. Molecules 2020; 25:molecules25112718. [PMID: 32545386 PMCID: PMC7321193 DOI: 10.3390/molecules25112718] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics is a powerful tool for identifying and quantifying proteins in biological samples, outperforming conventional antibody-based methods in many aspects. LC-MS/MS-based proteomics studies have revealed the protein abundances of many drug-metabolizing enzymes and transporters (DMETs) in tissues relevant to drug metabolism and disposition. Previous studies have consistently demonstrated marked interindividual variability in DMET protein expression, suggesting that varied DMET function is an important contributing factor for interindividual variability in pharmacokinetics (PK) and pharmacodynamics (PD) of medications. Moreover, differential DMET expression profiles were observed across different species and in vitro models. Therefore, caution must be exercised when extrapolating animal and in vitro DMET proteomics findings to humans. In recent years, DMET proteomics has been increasingly utilized for the development of physiologically based pharmacokinetic models, and DMET proteins have also been proposed as biomarkers for prediction of the PK and PD of the corresponding substrate drugs. In sum, despite the existence of many challenges in the analytical technology and data analysis methods of LC-MS/MS-based proteomics, DMET proteomics holds great potential to advance our understanding of PK behavior at the individual level and to optimize treatment regimens via the DMET protein biomarker-guided precision pharmacotherapy.
Collapse
|
8
|
Elmorsi Y, Al Feteisi H, Al-Majdoub ZM, Barber J, Rostami-Hodjegan A, Achour B. Proteomic characterisation of drug metabolising enzymes and drug transporters in pig liver. Xenobiotica 2020; 50:1208-1219. [DOI: 10.1080/00498254.2020.1763513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yasmine Elmorsi
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Hajar Al Feteisi
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Zubida M. Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
- Certara UK Ltd., Simcyp Division, Sheffield, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Quantitative mass spectrometry-based proteomics in the era of model-informed drug development: Applications in translational pharmacology and recommendations for best practice. Pharmacol Ther 2019; 203:107397. [DOI: 10.1016/j.pharmthera.2019.107397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/29/2019] [Indexed: 02/08/2023]
|
10
|
Harwood MD, Zhang M, Pathak SM, Neuhoff S. The Regional-Specific Relative and Absolute Expression of Gut Transporters in Adult Caucasians: A Meta-Analysis. Drug Metab Dispos 2019; 47:854-864. [DOI: 10.1124/dmd.119.086959] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/30/2019] [Indexed: 01/04/2023] Open
|
11
|
Jankovskaja S, Kamiie J, Rezeli M, Gustavsson L, Sugihara Y, Miliotis T, Ruzgas T, Marko-Varga G. Optimization of sample preparation for transporter protein quantification in tissues by LC-MS/MS. J Pharm Biomed Anal 2019; 164:9-15. [PMID: 30339949 DOI: 10.1016/j.jpba.2018.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Reproducible quantification of drug transporter protein expression in tissues is important for predicting transporter mediated drug disposition. Many mass-spectrometry based transporter protein quantification methods result in high variability of the estimated transporter quantities. Therefore, we aimed to evaluate and optimize mass spectrometry-based quantification method for drug transporter proteins in tissues. MATERIALS AND METHODS Plasma membrane (PM) proteins from mouse tissues were isolated by applying three extraction protocols: commercial plasma membrane extraction kit, tissue homogenization by Potter-Elvehjem homogenizer in combination with sucrose-cushion ultracentrifugation, and PM enrichment with Tween 40. Moreover, five different protein digestion protocols were applied on the same PM fraction. PM isolation and digestion protocols were evaluated by measuring the amount of transporter proteins by liquid chromatography-tandem mass spectrometry in selected reaction monitoring mode. RESULTS Mouse liver homogenization by Potter-Elvehjem homogenizer in combination with sucrose-cushion ultracentrifugation and PM enrichment with Tween 40 resulted in two times higher transporter protein quantity (Breast cancer resistance protein (Bcrp) 18.0 fmol/μg protein) in comparison with the PM samples isolated by extraction kit (Bcrp 9.8 fmol/μg protein). The evaluation of protein digestion protocols revealed that the most optimal protocol for PM protein digestion is with Lys-C and trypsin, in combination with trypsin enhancer and heat denaturation. Overall, quantities of Bcrp and Na+/K + ATPase proteins evaluated in mouse liver and kidney cortex by using our optimized PM isolation method, as well as, established digestion protocol were two to three times higher than previously reported and coefficient of variation (CV) for technical replicates was below 10%. CONCLUSION We have established an improved transporter protein quantification methodology by optimizing PM isolation and protein digestion procedures. The optimized procedure resulted in a higher transporter protein yield and improved precision.
Collapse
Affiliation(s)
- Skaidre Jankovskaja
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden; Department of Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms -Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Junichi Kamiie
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden; Laboratory of Veterinary Pathology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Melinda Rezeli
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Lena Gustavsson
- Department of Drug Metabolism, H. Lundbeck A/S, Valby, Denmark
| | - Yutaka Sugihara
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden; Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Tasso Miliotis
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden; Translational Science, Cardiovascular Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Tautgirdas Ruzgas
- Department of Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms -Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - György Marko-Varga
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden; Centre of Excellence in Biological and Medical Mass Spectrometry "CEBMMS", Biomedical Centre D13, Lund University, Lund, Sweden.
| |
Collapse
|
12
|
Drozdzik M, Busch D, Lapczuk J, Müller J, Ostrowski M, Kurzawski M, Oswald S. Protein Abundance of Clinically Relevant Drug Transporters in the Human Liver and Intestine: A Comparative Analysis in Paired Tissue Specimens. Clin Pharmacol Ther 2019; 105:1204-1212. [PMID: 30447067 DOI: 10.1002/cpt.1301] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/20/2018] [Indexed: 11/11/2022]
Abstract
Bioavailability of orally administered drugs is partly determined by function of drug transporters in the liver and intestine. Therefore, we explored adenosine triphosphate-binding cassette (ABC) and solute carriers family transporters expression (quantitative polymerase chain reaction) and protein abundance (liquid chromatography tandem mass spectrometry (LC-MS/MS)) in human liver and duodenum, jejunum, ileum, and colon in paired tissue specimens from nine organ donors. The transporter proteins were detected in the liver (permeability-glycoprotein (P-gp), multidrug resistance protein (MRP)2, MRP3, breast cancer resistance protein (BCRP), organic anion-transporting polypeptide (OATP)1B1, OATP1B3, OATP2B1, organic cation transporter (OCT)1, OCT3, organic anion transporter 2, Na+-taurocholate cotransporting polypeptide, monocarboxylate transporter (MCT)1, and multidrug and toxin extrusion 1) and the intestine (P-gp, multidrug-resistance protein (MRP)2, MRP3, MRP4, BCRP, OATP2B1, OCT1, apical sodium-bile acid transporter (only ileum), MCT1, and peptide transporter (PEPT1)). Significantly higher hepatic gene expression and protein abundance of ABCC2/MRP2, SLC22A1/OCT1, and SLCO2B1/OATP2B1 were found, as compared to all intestinal segments. No correlations between hepatic and small intestinal protein levels were observed. These observations provide a description of drug transporters distribution without the impact of interindividual variability bias and may help in construction of superior physiologically based pharmacokinetic and humanized animal models.
Collapse
Affiliation(s)
- Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Diana Busch
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| | - Joanna Lapczuk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Janett Müller
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| | - Marek Ostrowski
- Department of General and Transplantation Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Stefan Oswald
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| |
Collapse
|
13
|
Wittenburg LA, Ramirez D, Conger H, Gustafson DL. Simultaneous absolute quantitation of ATP-binding cassette transporters in normal dog tissues by signature peptide analysis using a LC/MS/MS method. Res Vet Sci 2018; 122:93-101. [PMID: 30500618 DOI: 10.1016/j.rvsc.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 12/21/2022]
Abstract
Membrane transport proteins are fundamental components of blood-tissue barriers and affect the absorption, distribution and elimination, and interactions of many of the drugs commonly used in veterinary medicine. A quantitative, simultaneous measurement of these proteins across dog tissues is not currently available, nor is it possible with current immune-based assays such as western blot. In the present study, we aimed to develop a sensitive and specific liquid chromatography tandem-mass spectrometry (LC/MS/MS) based quantitation method that can simultaneously quantitate 14 ATP-binding cassette transporters. We applied this method to a panel of normal canine tissues and compared the LC/MS/MS results with relative messenger RNA (mRNA) abundance using quantitative real-time polymerase chain reaction (qRT-PCR). Our LC/MS/MS method is sensitive, with lower limits of quantitation ranging from 5 to 10 fmol/μg of protein. We were able to detect and/or quantitate each of the 14 transporters in at least one normal dog tissue. Relative protein and mRNA abundance within tissues did not demonstrate a significant correlation in all cases. The results presented here will provide for more accurate predictions of drug movement in dogs through incorporation into physiologically based pharmacokinetic (PBPK) models; the method described here has wide applicability to the quantitation of virtually any proteins of interest in biologic samples where validated canine antibodies do not exist.
Collapse
Affiliation(s)
- Luke A Wittenburg
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80525, United States.
| | - Dominique Ramirez
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80525, United States
| | - Holly Conger
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80525, United States
| | - Daniel L Gustafson
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80525, United States
| |
Collapse
|
14
|
Perez Jimenez TE, Mealey KL, Schnider D, Grubb TL, Greene SA, Court MH. Identification of canine cytochrome P-450s (CYPs) metabolizing the tramadol (+)-M1 and (+)-M2 metabolites to the tramadol (+)-M5 metabolite in dog liver microsomes. J Vet Pharmacol Ther 2018; 41:815-824. [PMID: 30113702 DOI: 10.1111/jvp.12706] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
We previously showed that (+)-tramadol is metabolized in dog liver to (+)-M1 exclusively by CYP2D15 and to (+)-M2 by multiple CYPs, but primarily CYP2B11. However, (+)-M1 and (+)-M2 are further metabolized in dogs to (+)-M5, which is the major metabolite found in dog plasma and urine. In this study, we identified canine CYPs involved in metabolizing (+)-M1 and (+)-M2 using recombinant enzymes, untreated dog liver microsomes (DLMs), inhibitor-treated DLMs, and DLMs from CYP inducer-treated dogs. A canine P-glycoprotein expressing cell line was also used to evaluate whether (+)-tramadol, (+)-M1, (+)-M2, or (+)-M5 are substrates of canine P-glycoprotein, thereby limiting their distribution into the central nervous system. (+)-M5 was largely formed from (+)-M1 by recombinant CYP2C21 with minor contributions from CYP2C41 and CYP2B11. (+)-M5 formation in DLMs from (+)-M1 was potently inhibited by sulfaphenazole (CYP2C inhibitor) and chloramphenicol (CYP2B11 inhibitor) and was greatly increased in DLMs from phenobarbital-treated dogs. (+)-M5 was formed from (+)-M2 predominantly by CYP2D15. (+)-M5 formation from (+)-M1 in DLMs was potently inhibited by quinidine (CYP2D inhibitor) but had only a minor impact from all CYP inducers tested. Intrinsic clearance estimates showed over 50 times higher values for (+)-M5 formation from (+)-M2 compared with (+)-M1 in DLMs. This was largely attributed to the higher enzyme affinity (lower Km) for (+)-M2 compared with (+)-M1 as substrate. (+)-tramadol, (+)-M1, (+)-M2, or (+)-M5 were not p-glycoprotein substrates. This study provides a clearer picture of the role of individual CYPs in the complex metabolism of tramadol in dogs.
Collapse
Affiliation(s)
- Tania E Perez Jimenez
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| | - Katrina L Mealey
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| | - Darren Schnider
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| | - Tamara L Grubb
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| | - Stephen A Greene
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| | - Michael H Court
- Program in Individualized Medicine (PrIMe), Department of Veterinary Clinical Sciences, Pharmacogenomics Laboratory, Washington State University College of Veterinary Medicine, Pullman, Washington
| |
Collapse
|
15
|
Han LW, Gao C, Mao Q. An update on expression and function of P-gp/ABCB1 and BCRP/ABCG2 in the placenta and fetus. Expert Opin Drug Metab Toxicol 2018; 14:817-829. [PMID: 30010462 DOI: 10.1080/17425255.2018.1499726] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION P-glycoprotein (P-gp)/ABCB1 and breast cancer resistance protein (BCRP)/ABCG2 are highly expressed in the placenta and fetus throughout gestation and can modulate exposure and toxicity of drugs and xenobiotics to the vulnerable fetus during the sensitive times of growth and development. We aim to provide an update on current knowledge on placental and fetal expressions of the two transporters in different species, and to provide insight on interpreting transporter expression and fetal exposure relative to the concept of fraction of drug transported. Areas covered: Comprehensive literature review through PubMed (primarily from July 2010 to February 2018) on P-gp and BCRP expression and function in the placenta and fetus of primarily human, mouse, rat, and guinea pig. Expert opinion: While there are many commonalities in the expression and function of P-gp and BCRP in the placenta and fetal tissues across species, there are distinct differences in expression levels and temporal changes. Further studies are needed to quantify protein abundance of these transporters and functionally assess their activities at various gestational stages. Combining the knowledge of interspecies differences and the concept of fraction of drug transported, we may better predict the magnitude of impact these transporters have on fetal drug exposure.
Collapse
Affiliation(s)
- Lyrialle W Han
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| | - Chunying Gao
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| | - Qingcheng Mao
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| |
Collapse
|
16
|
Taddio MF, Mu L, Keller C, Schibli R, Krämer SD. Physiologically Based Pharmacokinetic Modelling with Dynamic PET Data to Study the In Vivo Effects of Transporter Inhibition on Hepatobiliary Clearance in Mice. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:5849047. [PMID: 29967572 PMCID: PMC6008768 DOI: 10.1155/2018/5849047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/20/2018] [Indexed: 01/16/2023]
Abstract
Physiologically based pharmacokinetic modelling (PBPK) is a powerful tool to predict in vivo pharmacokinetics based on physiological parameters and data from in vivo studies and in vitro assays. In vivo PBPK modelling in laboratory animals by noninvasive imaging could help to improve the in vivo-in vivo translation towards human pharmacokinetics modelling. We evaluated the feasibility of PBPK modelling with PET data from mice. We used data from two of our PET tracers under development, [11C]AM7 and [11C]MT107. PET images suggested hepatobiliary excretion which was reduced after cyclosporine administration. We fitted the time-activity curves of blood, liver, gallbladder/intestine, kidney, and peripheral tissue to a compartment model and compared the resulting pharmacokinetic parameters under control conditions ([11C]AM7 n = 2; [11C]MT107, n = 4) and after administration of cyclosporine ([11C]MT107, n = 4). The modelling revealed a significant reduction in [11C]MT107 hepatobiliary clearance from 35.2 ± 10.9 to 17.1 ± 5.6 μl/min after cyclosporine administration. The excretion profile of [11C]MT107 was shifted from predominantly hepatobiliary (CLH/CLR = 3.8 ± 3.0) to equal hepatobiliary and renal clearance (CLH/CLR = 0.9 ± 0.2). Our results show the potential of PBPK modelling for characterizing the in vivo effects of transporter inhibition on whole-body and organ-specific pharmacokinetics.
Collapse
Affiliation(s)
- Marco F. Taddio
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland
| | - Claudia Keller
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roger Schibli
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Stefanie D. Krämer
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Emami Riedmaier A, Burt H, Abduljalil K, Neuhoff S. More Power to OATP1B1: An Evaluation of Sample Size in Pharmacogenetic Studies Using a Rosuvastatin PBPK Model for Intestinal, Hepatic, and Renal Transporter-Mediated Clearances. J Clin Pharmacol 2017; 56 Suppl 7:S132-42. [PMID: 27385171 PMCID: PMC5096019 DOI: 10.1002/jcph.669] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/26/2015] [Indexed: 11/07/2022]
Abstract
Rosuvastatin is a substrate of choice in clinical studies of organic anion-transporting polypeptide (OATP)1B1- and OATP1B3-associated drug interactions; thus, understanding the effect of OATP1B1 polymorphisms on the pharmacokinetics of rosuvastatin is crucial. Here, physiologically based pharmacokinetic (PBPK) modeling was coupled with a power calculation algorithm to evaluate the influence of sample size on the ability to detect an effect (80% power) of OATP1B1 phenotype on pharmacokinetics of rosuvastatin. Intestinal, hepatic, and renal transporters were mechanistically incorporated into a rosuvastatin PBPK model using permeability-limited models for intestine, liver, and kidney, respectively, nested within a full PBPK model. Simulated plasma rosuvastatin concentrations in healthy volunteers were in agreement with previously reported clinical data. Power calculations were used to determine the influence of sample size on study power while accounting for OATP1B1 haplotype frequency and abundance in addition to its correlation with OATP1B3 abundance. It was determined that 10 poor-transporter and 45 intermediate-transporter individuals are required to achieve 80% power to discriminate the AUC0-48h of rosuvastatin from that of the extensive-transporter phenotype. This number was reduced to 7 poor-transporter and 40 intermediate-transporter individuals when the reported correlation between OATP1B1 and 1B3 abundance was taken into account. The current study represents the first example in which PBPK modeling in conjunction with power analysis has been used to investigate sample size in clinical studies of OATP1B1 polymorphisms. This approach highlights the influence of interindividual variability and correlation of transporter abundance on study power and should allow more informed decision making in pharmacogenomic study design.
Collapse
Affiliation(s)
- Ariane Emami Riedmaier
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Howard Burt
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Khaled Abduljalil
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Sibylle Neuhoff
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| |
Collapse
|
18
|
Rodrigues AD, Taskar KS, Kusuhara H, Sugiyama Y. Endogenous Probes for Drug Transporters: Balancing Vision With Reality. Clin Pharmacol Ther 2017; 103:434-448. [DOI: 10.1002/cpt.749] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/04/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022]
Affiliation(s)
- AD Rodrigues
- Pharmacokinetics; Dynamics & Metabolism, Medicine Design, Pfizer Inc.; Groton Connecticut USA
| | - KS Taskar
- Mechanistic Safety and Disposition; IVIVT, GlaxoSmithKline; Ware Hertfordshire UK
| | - H Kusuhara
- Laboratory of Molecular Pharmacokinetics; Graduate School of Pharmaceutical Sciences, University of Tokyo; Tokyo Japan
| | - Y Sugiyama
- RIKEN Innovation Center; Research Cluster for Innovation; RIKEN Kanagawa Japan
| |
Collapse
|
19
|
Hermann KF, Neuhaus CS, Micallef V, Wagner B, Hatibovic M, Aschmann HE, Paech F, Alvarez-Sanchez R, Krämer SD, Belli S. Kinetics of lipid bilayer permeation of a series of ionisable drugs and their correlation with human transporter-independent intestinal permeability. Eur J Pharm Sci 2017; 104:150-161. [PMID: 28366650 DOI: 10.1016/j.ejps.2017.03.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/21/2017] [Accepted: 03/29/2017] [Indexed: 12/28/2022]
Abstract
For low molecular weight drugs, lipid bilayer permeation is considered the major route for in vivo cell barrier passage. We recently introduced a fluorescence assay with liposomes to determine permeation kinetics of ionisable compounds across the lipid bilayer by monitoring drug-induced pH changes inside the liposomes. Here, we determined the permeability coefficients (PFLipP, FLipP for "Fluorescence Liposomal Permeability") across 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayers of 35 ionisable drugs at pH6.0 and compared them to available in vivo human jejunal permeability (Peff) data. PFLipP values were furthermore compared with published Caco-2 cell permeability coefficients (PCaco-2), permeability coefficients determined with the parallel artificial membrane permeability assay (PAMPA) and with log D (pH6.0). The log PFLipP, corrected for predicted para-cellular diffusion, and log PCaco-2 correlated best with log Peff, with similar adjusted R2 (0.75 and 0.74, n=12). Our results suggest that transporter-independent intestinal drug absorption is predictable from liposomal permeability.
Collapse
Affiliation(s)
- Katharina F Hermann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Claudia S Neuhaus
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Virgine Micallef
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Björn Wagner
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Maja Hatibovic
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Hélène E Aschmann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Franziska Paech
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Rubén Alvarez-Sanchez
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland.
| | - Sara Belli
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland.
| |
Collapse
|
20
|
Lin Z, Gehring R, Mochel JP, Lavé T, Riviere JE. Mathematical modeling and simulation in animal health – Part
II
: principles, methods, applications, and value of physiologically based pharmacokinetic modeling in veterinary medicine and food safety assessment. J Vet Pharmacol Ther 2016; 39:421-38. [DOI: 10.1111/jvp.12311] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/21/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Z. Lin
- Institute of Computational Comparative Medicine (ICCM) Department of Anatomy and Physiology College of Veterinary Medicine Kansas State University Manhattan KS USA
| | - R. Gehring
- Institute of Computational Comparative Medicine (ICCM) Department of Anatomy and Physiology College of Veterinary Medicine Kansas State University Manhattan KS USA
| | - J. P. Mochel
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Switzerland
| | - T. Lavé
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Switzerland
| | - J. E. Riviere
- Institute of Computational Comparative Medicine (ICCM) Department of Anatomy and Physiology College of Veterinary Medicine Kansas State University Manhattan KS USA
| |
Collapse
|
21
|
Karamanos Y, Pottiez G. Proteomics and the blood-brain barrier: how recent findings help drug development. Expert Rev Proteomics 2016; 13:251-8. [PMID: 26778576 DOI: 10.1586/14789450.2016.1143780] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The drug discovery and development processes are divided into different stages separated by milestones to indicate that significant progress has been made and that certain criteria for target validation, hits, leads and candidate drugs have been met. Proteomics is a promising approach for the identification of protein targets and biochemical pathways involved in disease process and thus plays an important role in several stages of the drug development. The blood-brain barrier is considered as a major bottleneck when trying to target new compounds to treat neurodegenerative diseases. Based on the survey of recent findings and with a projection on expected improvements, this report attempt to address how proteomics participates in drug development.
Collapse
Affiliation(s)
- Yannis Karamanos
- a Laboratoire de la Barrière Hématoencéphalique (LBHE) , Univesrité d'Artois EA2465 , Lens , France
| | - Gwënaël Pottiez
- a Laboratoire de la Barrière Hématoencéphalique (LBHE) , Univesrité d'Artois EA2465 , Lens , France
| |
Collapse
|
22
|
Quantitative aspects of drug permeation across in vitro and in vivo barriers. Eur J Pharm Sci 2015; 87:30-46. [PMID: 26493585 DOI: 10.1016/j.ejps.2015.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/16/2015] [Accepted: 10/17/2015] [Indexed: 01/09/2023]
Abstract
The kinetics of permeation across epithelial and endothelial cell sheets and across cell membranes is determinant for the pharmacokinetics of a drug. In vitro transport experiments with cultured cells or artificial barriers have tremendously improved the predictability of the in vivo behaviour of tested compounds. This article focuses on the parameters and calculation methods that are used to describe permeation quantitatively, with a focus on in vitro experiments and the prediction of intestinal absorption and blood-brain barrier passage. It shows under which in vitro experimental conditions standard calculations are adequate and under which conditions equations should be adapted to the experimental details. The impact of volume differences between donor and receiver compartments, pH gradients, addition of albumin, accumulation in the barrier and unidirectional transport by an efflux transporter on the results is shown in simulations. The article should make researchers aware of experimental factors that affect the outcome of a permeation experiment and how to account for this during data analysis. Finally, strategies to predict the in vivo behaviour of a compound based on the in vitro data are discussed. The goal of the article is to support researchers in choosing experimental conditions and calculation methods that deliver appropriate and reproducible results in permeation studies in vitro.
Collapse
|
23
|
Bioanalytical approaches for characterizing catabolism of antibody–drug conjugates. Bioanalysis 2015; 7:1583-604. [DOI: 10.4155/bio.15.87] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The in vivo stability and catabolism of antibody–drug conjugates (ADCs) directly impact their PK, efficacy and safety, and metabolites of the cytotoxic or small molecule drug component of an ADC can further complicate these factors. This perspective highlights the importance of understanding ADC catabolism and the associated bioanalytical challenges. We evaluated different bioanalytical approaches to qualitatively and quantitatively characterize ADC catabolites. Here we review and discuss the rationale and experimental strategies used to design bioanalytical assays for characterization of ADC catabolism and supporting ADME studies during ADC clinical development. This review covers both large and small molecule approaches, and uses examples from Kadcyla® (T-DM1) and a THIOMAB™ antibody–drug conjugate to illustrate the process.
Collapse
|
24
|
Al Feteisi H, Achour B, Rostami-Hodjegan A, Barber J. Translational value of liquid chromatography coupled with tandem mass spectrometry-based quantitative proteomics forin vitro–in vivoextrapolation of drug metabolism and transport and considerations in selecting appropriate techniques. Expert Opin Drug Metab Toxicol 2015; 11:1357-69. [DOI: 10.1517/17425255.2015.1055245] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
25
|
Cutler P, Voshol H. Proteomics in pharmaceutical research and development. Proteomics Clin Appl 2015; 9:643-50. [PMID: 25763573 DOI: 10.1002/prca.201400181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/10/2015] [Accepted: 03/09/2015] [Indexed: 01/07/2023]
Abstract
In the 20 years since its inception, the evolution of proteomics in pharmaceutical industry has mirrored the developments within academia and indeed other industries. From initial enthusiasm and subsequent disappointment in global protein expression profiling, pharma research saw the biggest impact when relating to more focused approaches, such as those exploring the interaction between proteins and drugs. Nowadays, proteomics technologies have been integrated in many areas of pharmaceutical R&D, ranging from the analysis of therapeutic proteins to the monitoring of clinical trials. Here, we review the development of proteomics in the drug discovery process, placing it in a historical context as well as reviewing the current status in light of the contributions to this special issue, which reflect some of the diverse demands of the drug and biomarker pipelines.
Collapse
Affiliation(s)
- Paul Cutler
- Translational Technologies and Bioinformatics, Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Hans Voshol
- Novartis Institutes for BioMedical Research, Analytical Sciences and Imaging, Basel, Switzerland
| |
Collapse
|