With respect to drug disposition, children (especially neonates and infants) are different than the adults. Therefore it is not appropriate to select dosing regimen for this special population based on empirical scaling (e.g., based on body weight) of the adult dose. This issue becomes more significant as it is not always possible to establish safety and efficacy of drugs in the children at clinic due to logistical, ethical, safetyand medico- legal concerns. For instance, out of 399 prescribed drugs to neonates/infants between 1997-2010, only 28 drugs were studied for the safety and/or efficacy. Thus, it is imperative that novel alternative approaches are developed to predict safe and efficacious dosing regimens for children. One such approach is to integrate age- dependent physiological parameters with drug specific parameters (e.g., in vitro enzyme/transport kinetic data) to develop a pediatric physiologically based pharmacokinetic (pPBPK) model. Once validated, such fully mechanistic pPBPK model can be generalized for any drug. However, the biggest hurdle in developing such models for children are the lack of absolute ontogeny data on the proteins that are related to drug disposition, i.e,, drug metabolizing enzymes (DMEs) and transporters. It is known that developmental pattern exists in the expression of major hepatic DMEs, but the available data are either qualitative/semi-quantitative or completely missing for most of the DMEs/transporters. Therefore, as a first step towards rectifying this gap in knowledge we propose to quantify the hepatic expression of DMEs and transporters in our unique pediatric livers (n=220) and compare this expression with that in adults. We will use selective and robust multiple reaction monitoring (MRM) proteomic approach to quantify these proteins. Once the age-dependent protein abundance data are available, these data can be integrated with in vitro kinetics and other developmental (physiological) information to construct a pPBPK models. Such rationally designed models can be validated using available clinical data on the model compounds, and then generalized to drugs that are eliminated by these mechanisms in the liver. Because mechanistic pPBPK models can delineate fractional role of individual metabolic/transport pathways in drug disposition, such mechanistic tools are also capable of accurately predicting drug-drug interactions (DDIs) and pharmacogenetic variability mediated by these pathways. Hence, this proposal addresses the mechanisms of hepatic drug disposition in neonates to adolescents. The pPBPK model generated in this study will be of enormous value with respect to child health as these will be important to assess the risk associated with the first use of drug (or other xenobiotics) in children (including neonates/infants, where the ontogeny matters most).

Public Health Relevance

Completion of this project will provide a drug metabolizing enzyme/transporter ontogeny based refined pediatric physiologically based pharmacokinetic (pPBPK) model for accurate prediction of in vivo drug disposition in pediatric population (neonates to adolescence). The refined pPBPK model will be a significant advancement in the field of pediatric clinical pharmacology as it provides a tool to estimate safe and effective dose of hepatically cleared drugs in children in the absence of pediatric safety and efficacy data.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD081299-02
Application #
9035414
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Ren, Zhaoxia
Project Start
2015-04-01
Project End
2020-02-29
Budget Start
2016-03-01
Budget End
2017-02-28
Support Year
2
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Washington
Department
Pharmacology
Type
Schools of Pharmacy
DUNS #
605799469
City
Seattle
State
WA
Country
United States
Zip Code
98195
Xu, Meijuan; Saxena, Neha; Vrana, Marc et al. (2018) Targeted LC-MS/MS Proteomics-Based Strategy To Characterize in Vitro Models Used in Drug Metabolism and Transport Studies. Anal Chem 90:11873-11882
Zhang, Haeyoung; Basit, Abdul; Busch, Diana et al. (2018) Quantitative characterization of UDP-glucuronosyltransferase 2B17 in human liver and intestine and its role in testosterone first-pass metabolism. Biochem Pharmacol 156:32-42
Prasad, Bhagwat; Bhatt, Deepak Kumar; Johnson, Katherine et al. (2018) Abundance of Phase 1 and 2 Drug-Metabolizing Enzymes in Alcoholic and Hepatitis C Cirrhotic Livers: A Quantitative Targeted Proteomics Study. Drug Metab Dispos 46:943-952
Basit, Abdul; Amory, John K; Prasad, Bhagwat (2018) Effect of Dose and 5?-Reductase Inhibition on the Circulating Testosterone Metabolite Profile of Men Administered Oral Testosterone. Clin Transl Sci 11:513-522
Bhatt, Deepak Kumar; Prasad, Bhagwat (2018) Critical Issues and Optimized Practices in Quantification of Protein Abundance Level to Determine Interindividual Variability in DMET Proteins by LC-MS/MS Proteomics. Clin Pharmacol Ther 103:619-630
Bhatt, Deepak Kumar; Basit, Abdul; Zhang, Haeyoung et al. (2018) Hepatic Abundance and Activity of Androgen- and Drug-Metabolizing Enzyme UGT2B17 Are Associated with Genotype, Age, and Sex. Drug Metab Dispos 46:888-896
Prasad, Bhagwat; Vrana, Marc; Mehrotra, Aanchal et al. (2017) The Promises of Quantitative Proteomics in Precision Medicine. J Pharm Sci 106:738-744
Bhatt, Deepak Kumar; Gaedigk, Andrea; Pearce, Robin E et al. (2017) Age-dependent Protein Abundance of Cytosolic Alcohol and Aldehyde Dehydrogenases in Human Liver. Drug Metab Dispos 45:1044-1048
Xu, Meijuan; Bhatt, Deepak Kumar; Yeung, Catherine K et al. (2017) Genetic and Nongenetic Factors Associated with Protein Abundance of Flavin-Containing Monooxygenase 3 in Human Liver. J Pharmacol Exp Ther 363:265-274
Nyquist, Michael D; Prasad, Bhagwat; Mostaghel, Elahe A (2017) Harnessing Solute Carrier Transporters for Precision Oncology. Molecules 22:

Showing the most recent 10 out of 13 publications