1
|
Saib S, Hodin S, He Z, Delézay O, Delavenne X. Is the human model RPTEC/TERT1 a relevant model for assessing renal drug efflux? Fundam Clin Pharmacol 2020; 35:732-743. [PMID: 33185296 DOI: 10.1111/fcp.12631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/14/2023]
Abstract
Active tubular secretion plays a major role in renal excretion of drugs thanks to the presence of many membrane transporters such as ABC transporters. These proteins facilitate drug transfer into the urine and could be a source of pharmacokinetic variabilities. Up to now, several human in vitro models of proximal tubule have been proposed but few of them have been characterized for predicting drugs renal efflux. The aim of this study was to determine whether the human model RPTEC/TERT1 meets all the criteria expected of a good model to assess renal drug transport. First, in vitro barrier properties were investigated. Then, the expression of several ABC transporters was assessed by immunofluorescence and relative quantification by liquid chromatography-high-resolution mass spectrometry (LC-HRMS) in comparison to the MDCK model. Finally, bidirectional transport studies were performed to evaluate the functionality of transporters and the abilities of model to discriminate several drugs. The RPTEC/TERT1 model formed a tight structure (192 Ω.cm2 ) that was confirmed by paracellular permeability assays. Proteomic analysis and immunofluorescence staining showed the expression of several ABC transporters. Then, only the functionality of P-gp was confirmed by the active efflux of apixaban in this study. In addition, the RPTEC/TERT1 model presents the key criteria of a renal barrier and expresses several ABC transporters. Nevertheless, the BCRP and MRP's functionality was not confirmed and further investigations are required to valid this model as in vitro model for assessing renal drug efflux.
Collapse
Affiliation(s)
- Sonia Saib
- Dysfonction Vasculaire et Hémostase, INSERM U1059, Université Jean Monnet, Saint-Etienne, France
| | - Sophie Hodin
- Dysfonction Vasculaire et Hémostase, INSERM U1059, Université Jean Monnet, Saint-Etienne, France
| | - Zhiguo He
- Laboratoire de Biologie, d'Ingénierie et d'Imagerie de la Greffe de Cornée, BiiGC, Saint-Etienne, France
| | - Olivier Delézay
- Dysfonction Vasculaire et Hémostase, INSERM U1059, Université Jean Monnet, Saint-Etienne, France
| | - Xavier Delavenne
- Dysfonction Vasculaire et Hémostase, INSERM U1059, Université Jean Monnet, Saint-Etienne, France.,Laboratoire de Pharmacologie Toxicologie Gaz du sang, CHU de Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
2
|
Exclusive prenatal exposure to a 16 PAH mixture does not impact anxiety-related behaviours and regional brain metabolism in adult male rats: A role for the period of exposure in the modulation of PAH neurotoxicity. Toxicol Lett 2013; 221:40-6. [DOI: 10.1016/j.toxlet.2013.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 11/20/2022]
|
3
|
SUD DHRUV, JOSEPH IANMP, KIRSCHNER DENISE. PREDICTING EFFICACY OF PROTON PUMP INHIBITORS IN REGULATING GASTRIC ACID SECRETION. J BIOL SYST 2011. [DOI: 10.1142/s0218339004000999] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Developing drugs to treat gastric acid related illnesses such as ulcers and acid reflux disease is the leading focus of pharmaceutical companies. In fact, expenditure for treating these disorders is highest among all illnesses in the US. Over the last few decades, a class of drugs known as a proton pump inhibitors (PPIs) appeared on the market and are highly effective at abating gastric illnesses by raising stomach pH (reducing gastric acid levels). While much is known about the action of PPIs , there are still open questions regarding their efficacy, dosing and long-term effects. Here we extend a previous gastric acid secretion model developed by our group to incorporate a pharmacodynamic/pharmacokinetic model to study proton pump inhibitor (PPI) action. Model-relevant parameters for specific drugs such as omeprazole (OPZ) , lansoprazole (LPZ) and pantoprazole (PPZ) were used from published data, and we conducted simulations to study various aspects of PPI treatment. Clinical data suggests that duration of acid suppression is dependent on proton pump turnover rates and this is supported by our model. We found the order of efficacy of the different PPIs to be OPZ>PPZ>LPZ for clinically recommended dose values, and OPZ>PPZ=LPZ for equal doses. Our results indicate that a breakfast dose for once-daily dosing regimens and a breakfast-lunch dose for twice-daily dosing regimens is recommended. Simulation of other gastric disorders using our model provides atypical applications for the study of drug treatment on homeostatic systems and identification of potential side-effects.
Collapse
Affiliation(s)
- DHRUV SUD
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - IAN M. P. JOSEPH
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - DENISE KIRSCHNER
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Suhre WM, Ekins S, Chang C, Swaan PW, Wright SH. Molecular determinants of substrate/inhibitor binding to the human and rabbit renal organic cation transporters hOCT2 and rbOCT2. Mol Pharmacol 2005; 67:1067-77. [PMID: 15630081 DOI: 10.1124/mol.104.004713] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Organic cation transporters are important for the elimination of many drugs and toxins from the body. In the present study, substrate-transporter interactions were investigated in Chinese hamster ovary cells stably transfected with either the human or rabbit orthologs of the principal organic cation transporter in the kidney, OCT2. IC(50) values, ranging from 0.04 muM to >3 mM, for inhibition of [(14)C]tetraethylammonium transport were determined for more than 30 structurally diverse compounds. Although the two OCT orthologs displayed similar IC(50) values for some of these compounds, the majority varied by as much as 20-fold. Marked differences in substrate affinity were also noted when comparing hOCT2 to the closely related homolog hOCT1. These data suggest the molecular determinants of substrate binding differ markedly among both homologous and orthologous OCT transporters. The software package Cerius(2) (Accelrys, San Diego, CA) was used to generate a descriptor-based, two-dimensional, quantitative structure-activity relationship (QSAR) to produce a model relating the affinity of hOCT2 to particular physicochemical features of substrate/inhibitor molecules (r(2) = 0.81). Comparative molecular field analysis (Tripos, St. Louis, MO) was used to generate three-dimensional QSARs describing the structural basis of substrate binding to hOCT2 and rbOCT2 (q(2) = 0.60 and 0.53, respectively, and each with r(2) = 0.97). The quality of the models was assessed by their ability to successfully predict the inhibition of a set of test compounds. The current models enabled prediction of OCT2 affinity and may prove useful in the prediction of unwanted drug interactions at the level of the renal secretory process.
Collapse
Affiliation(s)
- Wendy M Suhre
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | |
Collapse
|
5
|
Zhou S, Kestell P, Paxton JW. Predicting pharmacokinetics and drug interactions in patients from in vitro and in vivo models: the experience with 5,6-dimethylxanthenone-4-acetic acid (DMXAA), an anti-cancer drug eliminated mainly by conjugation. Drug Metab Rev 2002; 34:751-90. [PMID: 12487149 DOI: 10.1081/dmr-120015693] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The novel anti-tumor agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) was developed in the Auckland Cancer Society Research Center. Its pharmacokinetic properties have been investigated using both in vitro and in vivo models, and the resulting data extrapolated to patients. The metabolism of DMXAA has been extensively studied mainly using hepatic microsomes, which indicated that UGT1A9 and UGT2B7-catalyzed glucuronidation on its acetic acid side chain and to a lesser extent CYP1A2-catalyzed hydroxylation of the 6-methyl group are the major metabolic pathways, resulting in DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid. The predominant metabolite in human urine (up to 60% of total dose) was identified as DMXAA-G, which was chemically reactive, undergoing hydrolysis, intramolecular rearrangement, and covalent binding to plasma proteins. In vivo formation of DMXAA-protein adducts were also observed in cancer patients receiving DMXAA treatment. The comparison of the in vitro human hepatic microsomal metabolism and inhibition of DMXA by UGT and/or CYP substrates with animal species indicated species differences. Renal microsomes from all animal species examined had glucuronidation activity for DMXAA, but lower than the liver. In vitro-in vivo extrapolations based on human microsomal data indicated a 7-fold underestimation of plasma clearance in patients. In contrast, allometric scaling using in vivo data from the mouse, rat, and rabbit predicted a plasma clearance of 3.5 mL/min/kg, similar to that observed in patients (3.7 mL/min/kg). Based on in vitro metabolic inhibition studies, it appears possible to predict the effects on the plasma kinetic profile of DMXAA of drugs such as diclofenac, which are mainly metabolized by UGT2B7. However, it did not appear possible to predict the effect of thalidomide on the pharmacokinetics of DMXAA in patients based on in vitro inhibition and animal studies. These data indicate that preclincial pharmacokinetic studies using both in vitro and in vivo models play an important but different role in predicting pharmacokinetics and drug interactions in patients.
Collapse
Affiliation(s)
- Shufeng Zhou
- Division of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
6
|
|
7
|
Gelman A, Bois F, Jiang J. Physiological Pharmacokinetic Analysis Using Population Modeling and Informative Prior Distributions. J Am Stat Assoc 1996. [DOI: 10.1080/01621459.1996.10476708] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
8
|
Charnick SB, Kawai R, Nedelman JR, Lemaire M, Niederberger W, Sato H. Perspectives in pharmacokinetics. Physiologically based pharmacokinetic modeling as a tool for drug development. JOURNAL OF PHARMACOKINETICS AND BIOPHARMACEUTICS 1995; 23:217-29. [PMID: 8719238 DOI: 10.1007/bf02354273] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Since the pioneering work of Haggard and Teorell in the first half of the 20th century, and of Bischoff and Dedrick in the late 1960s, physiologically based pharmacokinetic (PBPK) modeling has gone through cycles of general acceptance, and of healthy skepticism. Recently, however, the trend in the pharmaceuticals industry has been away from PBPK models. This is understandable when one considers the time and effort necessary to develop, test, and implement a typical PBPK model, and the fact that in the present-day environment for drug development, efficacy and safety must be demonstrated and drugs brought to market more rapidly. Although there are many modeling tools available to the pharmacokineticist today, many of which are preferable to PBPK modeling in most circumstances, there are several situations in which PBPK modeling provides distinct benefits that outweigh the drawbacks of increased time and effort for implementation. In this Commentary, we draw on our experience with this modeling technique in an industry setting to provide guidelines on when PBPK modeling techniques could be applied in an industrial setting to satisfy the needs of regulatory customers. We hope these guidelines will assist researchers in deciding when to apply PBPK modeling techniques. It is our contention that PBPK modeling should be viewed as one of many modeling tools for drug development.
Collapse
Affiliation(s)
- S B Charnick
- Department of Clinical Pharmacology, Sandoz Pharmaceuticals Corporation, East Hanover, New Jersey 07936, USA
| | | | | | | | | | | |
Collapse
|
9
|
Hildebrand M. Inter-species extrapolation of pharmacokinetic data of three prostacyclin-mimetics. PROSTAGLANDINS 1994; 48:297-312. [PMID: 7531864 DOI: 10.1016/0090-6980(94)90030-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cica-, eptalo- and iloprost are chemically and metabolically stabilized derivatives of prostacyclin which maintain the pharmacodynamic profile of the endogenous precursor. While iloprost is still subject to beta-oxidative degradation of the upper side chain, cicaprost is highly metabolically stable. Eptaloprost was synthesized to realize the pro-drug concept in PGI2-mimetics and was designed to be activated to cicaprost by single beta-oxidation. All three prostacyclin-mimetics were studied in various animal species (mouse, rat, rabbit, monkey, dog and pig) and in man to determine their pharmacokinetic profiles. Based upon this data, it was of interest whether an inter-species extrapolation of pharmacokinetic parameters can be performed to show the predictive value of animal experimentation. Allometric inter-species extrapolation is performed by modelling pharmacokinetic data (Y) as exponential functions (x) of species characteristics (e.g. body weight, W) as: Y = .aWx. For total clearance and volumes of distribution at steady state, a clear-cut correlation with x-values of 0.6-0.8 and 1.0-1.1 could be shown for all three compounds. For cicaprost, which was excreted unchanged in several species, renal and non-renal clearance was also mathematically scalable. Due to the use of different compartment models to describe plasma disposition, different sets of half-life data were obtained and could not be extrapolated reasonably. However, mean residence time showed a dependency on body weight with 0.25 as power function. In case of cicaprost, only the dog, which extensively metabolizes the compound, could not be enrolled in inter-species extrapolation. Excretion half-lives or residence times did not show a significant correlation to body weight or maximum life time potential. The present inter-species extrapolation showed a dependency from species body weight for model-independent pharmacokinetic data, e.g. clearance, volume of distribution at steady state and correspondingly mean residence time. The disposition profile of these compounds can therefore be predicted. Preliminary information on bio-degradation is an additional prerequisite for extrapolation. These data demonstrate that basic physiologically determined processes, which show some evolutionary allometric dependency, also influence the disposition of prostacyclin-mimetics. An extrapolation of data from animal to man could easily be realized giving additional justification for animal studies in pharmacology, toxicology and pharmacokinetics.
Collapse
Affiliation(s)
- M Hildebrand
- Institute of Pharmacokinetics, Schering AG, Berlin, Germany
| |
Collapse
|
10
|
Filser JG, Schwegler U, Csanády GA, Greim H, Kreuzer PE, Kessler W. Species-specific pharmacokinetics of styrene in rat and mouse. Arch Toxicol 1993; 67:517-30. [PMID: 8285850 DOI: 10.1007/bf01969264] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The pharmacokinetics of styrene were investigated in male Sprague-Dawley rats and male B6C3F1 mice using the closed chamber technique. Animals were exposed to styrene vapors of initial concentrations ranging from 550 to 5000 ppm, or received intraperitoneal (i.p.) doses of styrene from 20 to 340 mg/kg or oral (p.o.) doses of styrene in olive oil from 100 to 350 mg/kg. Concentration-time courses of styrene in the chamber atmosphere were monitored and analyzed by a pharmacokinetic two-compartment model. In both species, the rate of metabolism of inhaled styrene was concentration dependent. At steady state it increased linearly with exposure concentration up to about 300 ppm; more than 95% of inhaled styrene was metabolized and only small amounts were exhaled unchanged. At these low concentrations transport to the metabolizing enzymes and not their metabolic capacity was the rate limiting step for metabolism. Pharmacokinetic behaviour of styrene was strongly influenced by physiological parameters such as blood flow and especially the alveolar ventilation rate. At exposure concentrations of styrene above 300 ppm the rate of metabolism at steady state was progressively limited by biochemical parameters of the metabolizing enzymes. Saturation of metabolism (Vmax) was reached at atmospheric concentrations of about 700 ppm in rats and 800 ppm in mice, Vmax being 224 mumol/(h.kg) and 625 mumol/(h.kg), respectively. The atmospheric concentrations at Vmax/2 were 190 ppm in rats and 270 ppm in mice. Styrene accumulates preferentially in the fatty tissue as can be deduced from its partition coefficients in olive oil:air and water:air which have been determined in vitro at 37 degrees C to be 5600 and 15. In rats and mice exposed to styrene vapors below 300 ppm, there was little accumulation since the uptake was rate limiting. The bioaccumulation factor body:air at steady state (K'st*) was rather low in comparison to the thermodynamic partition coefficient body:air (Keq) which was determined to be 420. K'st* increased from 2.7 at 10 ppm to 13 at 310 ppm in the rat and from 5.9 at 20 ppm to 13 at 310 ppm in the mouse. Above 300 ppm, K'st* increased considerably with increasing concentration since metabolism became saturated in both species. At levels above 2000 ppm K'st* reached its maximum of 420 being equivalent to Keq. Pretreatment with diethyldithiocarbamate, administered intraperitoneally (200 mg/kg in rats, 400 mg/kg in mice) 15 min prior to exposure of styrene vapours, resulted in effective inhibition of styrene metabolism, indicating that most of the styrene is metabolized by cytochrome P450-dependent monooxygenases.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- J G Filser
- GSF-Forschungszentrum für Umwelt und Gesundheit, Institut für Toxicologie, Neuherberg, Germany
| | | | | | | | | | | |
Collapse
|