Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicology ; 332: 52-66, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-24863738

ABSTRACT

In vitro-derived information has been increasingly used to support and improve human health risk assessment for exposure to chemicals. Physiologically based pharmacokinetic (PBPK) modeling is a key component in the movement toward in vitro-based risk assessment, providing a tool to integrate diverse experimental data and mechanistic information to relate in vitro effective concentrations to equivalent human exposures. One of the challenges, however, in the use of PBPK models for this purpose has been the need for extensive chemical-specific parameters. With the remarkable advances in in vitro methodologies in recent years, in vitro-derived parameters can now be easily incorporated into PBPK models. In this study we demonstrate an in vitro data based parameterization approach to develop a physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) model, using carbaryl as a case study. In vitro experiments were performed to provide the chemical-specific pharmacokinetic (PK) and pharmacodynamic (PD) parameters for carbaryl in the PBPK model for this compound. Metabolic clearance and cholinesterase (ChE) interaction parameters for carbaryl were measured in rat and human tissues. These in vitro PK and PD data were extrapolated to parameters in the whole body PBPK model using biologically appropriate scaling. The PBPK model was then used to predict the kinetics and ChE inhibition dynamics of carbaryl in vivo. This case study with carbaryl provides a reasonably successful example of utilizing the in vitro to in vivo extrapolation (IVIVE) approach for PBPK model development. This approach can be applied to other carbamates with an anticholinesterase mode of action as well as to environmental chemicals in general with further refinement of the current shortcomings in the approach. It will contribute to minimizing the need for in vivo human data for PBPK model parameterization and evaluation in human risk assessments.


Subject(s)
Carbaryl/pharmacokinetics , Carbaryl/toxicity , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/toxicity , Cholinesterases/metabolism , Models, Biological , Toxicity Tests/methods , Adult , Animal Testing Alternatives , Animals , Biotransformation , Brain/drug effects , Brain/enzymology , Carbaryl/blood , Cells, Cultured , Cholinesterase Inhibitors/blood , Computer Simulation , Erythrocytes/drug effects , Erythrocytes/enzymology , Female , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Linear Models , Male , Metabolic Clearance Rate , Middle Aged , Rats, Sprague-Dawley , Risk Assessment , Risk Factors , Species Specificity , Young Adult
2.
Drug Metab Dispos ; 41(11): 1949-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990525

ABSTRACT

Prediction of clinical efficacy, toxicity, and drug-drug interactions may be improved by accounting for the intracellular unbound drug concentration (C(unbound)) in vitro and in vivo. Furthermore, subcellular drug distribution may aid in predicting efficacy, toxicity, and risk assessment. The present study was designed to quantify the intracellular C(unbound) and subcellular localization of drugs in rat sandwich-cultured hepatocytes (SCH) compared with rat isolated perfused liver (IPL) tissue. Probe drugs with distinct mechanisms of hepatocellular uptake and accumulation were selected for investigation. Following drug treatment, SCH and IPL tissues were homogenized and fractionated by differential centrifugation to enrich for subcellular compartments. Binding in crude lysate and cytosol was determined by equilibrium dialysis; the C(unbound) and intracellular-to-extracellular C(unbound) ratio (K(pu,u)) were used to describe accumulation of unbound drug. Total accumulation (K(pobserved)) in whole tissue was well predicted by the SCH model (within 2- to 3-fold) for the selected drugs. Ritonavir (K(pu,u) ∼1) was evenly distributed among cellular compartments, but highly bound, which explained the observed accumulation within liver tissue. Rosuvastatin was recovered primarily in the cytosolic fraction, but did not exhibit extensive binding, resulting in a K(pu,u) >1 in liver tissue and SCH, consistent with efficient hepatic uptake. Despite extensive binding and sequestration of furamidine within liver tissue, a significant portion of cellular accumulation was attributed to unbound drug (K(pu,u) >16), as expected for a charged, hepatically derived metabolite. Data demonstrate the utility of SCH to predict quantitatively total tissue accumulation and elucidate mechanisms of hepatocellular drug accumulation such as active uptake versus binding/sequestration.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , Pharmaceutical Preparations/metabolism , Tissue Distribution/physiology , Animals , Cells, Cultured , Drug Interactions , Male , Rats , Rats, Wistar
3.
Drug Metab Dispos ; 40(1): 6-17, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21953913

ABSTRACT

Dose selection during antiparasitic drug development in animal models and humans traditionally has relied on correlations between plasma concentrations obtained at or below maximally tolerated doses that are efficacious. The objective of this study was to improve the understanding of the relationship between dose and plasma/tissue exposure of the model antiparasitic agent, pafuramidine, using a semiphysiologically based pharmacokinetic (semi-PBPK) modeling approach. Preclinical and clinical data generated during the development of pafuramidine, a prodrug of the active metabolite, furamidine, were used. A whole-body semi-PBPK model for rats was developed based on a whole-liver PBPK model using rat isolated perfused liver data. A whole-body semi-PBPK model for humans was developed on the basis of the whole-body rat model. Scaling factors were calculated using metabolic and transport clearance data generated from rat and human sandwich-cultured hepatocytes. Both whole-body models described pafuramidine and furamidine disposition in plasma and predicted furamidine tissue (liver and kidney) exposure and excretion profiles (biliary and renal). The whole-body models predicted that the intestine contributes significantly (30-40%) to presystemic furamidine formation in both rats and humans. The predicted terminal elimination half-life of furamidine in plasma was 3- to 4-fold longer than that of pafuramidine in rats (170 versus 47 h) and humans (64 versus 19 h). The dose-plasma/tissue exposure relationship for the prodrug/active metabolite pair was determined using the whole-body models. The human model proposed a dose regimen of pafuramidine (40 mg once daily) based on a predefined efficacy-safety index. A similar approach could be used to guide dose-ranging studies in humans for next-in-class compounds.


Subject(s)
Antiparasitic Agents/pharmacology , Antiparasitic Agents/pharmacokinetics , Models, Biological , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Humans , Male , Middle Aged , Predictive Value of Tests , Rats , Rats, Sprague-Dawley , Rats, Wistar , Tissue Distribution/drug effects , Tissue Distribution/physiology
4.
J Pharmacol Exp Ther ; 337(2): 503-12, 2011 May.
Article in English | MEDLINE | ID: mdl-21320872

ABSTRACT

Selection of in vitro models that accurately characterize metabolite systemic and hepatobiliary exposure remains a challenge in drug development. In the present study, mechanisms underlying differences in systemic exposure of two active metabolites, furamidine and 2,5-bis (5-amidino)-2-pyridyl furan (CPD-0801), were examined using two hepatic models from rats: isolated perfused livers (IPLs) and sandwich-cultured hepatocytes (SCH). Pafuramidine, a prodrug of furamidine, and 2,5-bis [5-(N-methoxyamidino)-2-pyridyl] furan (CPD-0868), a prodrug of CPD-0801, were selected for investigation because CPD-0801 exhibits greater systemic exposure than furamidine, despite remarkable structural similarity between these two active metabolites. In both IPLs and SCH, the extent of conversion of CPD-0868 to CPD-0801 was consistently higher than that of pafuramidine to furamidine over time (at most 2.5-fold); area under the curve (AUC) of CPD-0801 in IPL perfusate and SCH medium was at least 7-fold higher than that of furamidine. Pharmacokinetic modeling revealed that the rate constant for basolateral (liver to blood) net efflux (k(A_net efflux)) of total formed CPD-0801 (bound + unbound) was 6-fold higher than that of furamidine. Hepatic accumulation of both active metabolites was extensive (>95% of total formed); the hepatic unbound fraction (f(u,L)) of CPD-0801 was 5-fold higher than that of furamidine (1.6 versus 0.3%). Incorporation of f(u,L) into the pharmacokinetic model resulted in comparable k(A_net efflux,u) between furamidine and CPD-0801. In conclusion, intrahepatic binding markedly influenced the disposition of these active metabolites. A higher f(u,L) explained, in part, the enhanced perfusate AUC of CPD-0801 compared with furamidine in IPLs. SCH predicted the disposition of prodrug/metabolite in IPLs.


Subject(s)
Amidines/pharmacology , Amidines/pharmacokinetics , Benzamidines/pharmacology , Benzamidines/pharmacokinetics , Liver/metabolism , Pharmaceutical Preparations/metabolism , Animals , Bile/metabolism , Biotransformation , Cell Survival/physiology , Cells, Cultured , Chromatography, High Pressure Liquid , Data Interpretation, Statistical , Hepatocytes/metabolism , Male , Mass Spectrometry , Models, Biological , Pharmacokinetics , Prodrugs , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...