1
|
Kang DW, Kim JH, Choi GW, Cho SJ, Cho HY. PBPK model-based gender-specific risk assessment of N-nitrosodimethylamine (NDMA) using human biomonitoring data. Arch Toxicol 2024; 98:3269-3288. [PMID: 39096368 DOI: 10.1007/s00204-024-03828-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Despite several screening levels for NDMA reported in water, soil, air, and drugs, the human risk assessment using biomonitoring concentrations has not been performed. In this study, gender-specific exposure guidance values were determined in humans, then biomonitoring measurements in healthy Korean subjects (32 men and 40 women) were compared to the exposure guidance values to evaluate the current exposure level to NDMA. For the human risk assessment of NDMA, the gender-specific physiologically based pharmacokinetic (PBPK) model was developed in humans using proper physiological parameters, partition coefficients, and biochemical parameters. Using the PBPK model, a Monte Carlo simulation was performed to describe the magnitudes of inter-individual variability and uncertainty on the single model predictions. The PBPK modeling and Monte Carlo simulation allowed the estimation of the relationship between external dose and blood concentration for the risk assessment. The procedure for the human risk assessment was summarized as follows: (1) estimating a steady-state blood concentration (Cavg) corresponding to the daily no observed adverse effect level (NOAEL) administration in rats; (2) applying uncertainty factors (UFs) for deriving the human Cavg; (3) determining the exposure guidance values as screening criteria; (4) interpreting the human biomonitoring measurements by forward and reverse dosimetry approaches. Using the biomonitoring concentrations, current daily exposures to NDMA were estimated to be 3.95 μg/day/kg for men and 10.60 μg/day/kg for women, respectively. The result of the study could be used as a basis for implementing further risk management and regulatory decision-making for NDMA.
Collapse
Affiliation(s)
- Dong Wook Kang
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Ju Hee Kim
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Go-Wun Choi
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seok-Jin Cho
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hea-Young Cho
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
2
|
Zhu J, Zhou S, Wang L, Zhao Y, Wang J, Zhao T, Li T, Shao F. Characterization of Pediatric Rectal Absorption, Drug Disposition, and Sedation Level for Midazolam Gel Using Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling. Mol Pharm 2024; 21:2187-2197. [PMID: 38551309 DOI: 10.1021/acs.molpharmaceut.3c00778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
This study aims to explore and characterize the role of pediatric sedation via rectal route. A pediatric physiologically based pharmacokinetic-pharmacodynamic (PBPK/PD) model of midazolam gel was built and validated to support dose selection for pediatric clinical trials. Before developing the rectal PBPK model, an intravenous PBPK model was developed to determine drug disposition, specifically by describing the ontogeny model of the metabolic enzyme. Pediatric rectal absorption was developed based on the rectal PBPK model of adults. The improved Weibull function with permeability, surface area, and fluid volume parameters was used to extrapolate pediatric rectal absorption. A logistic regression model was used to characterize the relationship between the free concentrations of midazolam and the probability of sedation. All models successfully described the PK profiles with absolute average fold error (AAFE) < 2, especially our intravenous PBPK model that extended the predicted age to preterm. The simulation results of the PD model showed that when the free concentrations of midazolam ranged from 3.9 to 18.4 ng/mL, the probability of "Sedation" was greater than that of "Not-sedation" states. Combined with the rectal PBPK model, the recommended sedation doses were in the ranges of 0.44-2.08 mg/kg for children aged 2-3 years, 0.35-1.65 mg/kg for children aged 4-7 years, 0.24-1.27 mg/kg for children aged 8-12 years, and 0.20-1.10 mg/kg for adolescents aged 13-18 years. Overall, this model mechanistically quantified drug disposition and effect of midazolam gel in the pediatric population, accurately predicted the observed clinical data, and simulated the drug exposure for sedation that will inform dose selection for following pediatric clinical trials.
Collapse
Affiliation(s)
- Jinying Zhu
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| | - Sufeng Zhou
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Lu Wang
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yuqing Zhao
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jie Wang
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tangping Zhao
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| | - Tongtong Li
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| | - Feng Shao
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
3
|
Kang DW, Kim JH, Choi GW, Cho SJ, Cho HY. Physiologically-based pharmacokinetic model for evaluating gender-specific exposures of N-nitrosodimethylamine (NDMA). Arch Toxicol 2024; 98:821-835. [PMID: 38127128 DOI: 10.1007/s00204-023-03652-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
N-nitrosodimethylamine (NDMA) is classified as a human carcinogen and could be produced by both natural and industrial processes. Although its toxicity and histopathology have been well-studied in animal species, there is insufficient data on the blood and tissue exposures that can be correlated with the toxicity of NDMA. The purpose of this study was to evaluate gender-specific pharmacokinetics/toxicokinetics (PKs/TKs), tissue distribution, and excretion after the oral administration of three different doses of NDMA in rats using a physiologically-based pharmacokinetic (PBPK) model. The major target tissues for developing the PBPK model and evaluating dose metrics of NDMA included blood, gastrointestinal (GI) tract, liver, kidney, lung, heart, and brain. The predictive performance of the model was validated using sensitivity analysis, (average) fold error, and visual inspection of observations versus predictions. Then, a Monte Carlo simulation was performed to describe the magnitudes of inter-individual variability and uncertainty of the single model predictions. The developed PBPK model was applied for the exposure simulation of daily oral NDMA to estimate blood concentration ranges affecting health effects following acute-duration (≤ 14 days), intermediate-duration (15-364 days), and chronic-duration (≥ 365 days) intakes. The results of the study could be used as a scientific basis for interpreting the correlation between in vivo exposures and toxicological effects of NDMA.
Collapse
Affiliation(s)
- Dong Wook Kang
- College of Pharmacy, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Ju Hee Kim
- College of Pharmacy, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Go-Wun Choi
- College of Pharmacy, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Seok-Jin Cho
- College of Pharmacy, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Hea-Young Cho
- College of Pharmacy, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
4
|
Mukherjee D, Collins M, Dylla DE, Kaur J, Semizarov D, Martinez A, Conway B, Khan T, Mostafa NM. Assessment of Drug-Drug Interaction Risk Between Intravenous Fentanyl and the Glecaprevir/Pibrentasvir Combination Regimen in Hepatitis C Patients Using Physiologically Based Pharmacokinetic Modeling and Simulations. Infect Dis Ther 2023; 12:2057-2070. [PMID: 37470926 PMCID: PMC10505123 DOI: 10.1007/s40121-023-00830-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/23/2023] [Indexed: 07/21/2023] Open
Abstract
INTRODUCTION An unsafe injection practice is one of the major contributors to new hepatitis C virus (HCV) infections; thus, people who inject drugs are a key population to prioritize to achieve HCV elimination. The introduction of highly effective and well-tolerated pangenotypic direct-acting antivirals, including glecaprevir/pibrentasvir (GLE/PIB), has revolutionized the HCV treatment landscape. Glecaprevir is a weak cytochrome P450 3A4 (CYP3A4) inhibitor, so there is the potential for drug-drug interactions (DDIs) with some opioids metabolized by CYP3A4, such as fentanyl. This study estimated the impact of GLE/PIB on the pharmacokinetics of intravenous fentanyl by building a physiologically based pharmacokinetic (PBPK) model. METHODS A PBPK model was developed for intravenous fentanyl by incorporating published information on fentanyl metabolism, distribution, and elimination in healthy individuals. Three clinical DDI studies were used to verify DDIs within the fentanyl PBPK model. This model was integrated with a previously developed GLE/PIB PBPK model. After model validation, DDI simulations were conducted by coadministering GLE 300 mg + PIB 120 mg with a single dose of intravenous fentanyl (0.5 µg/kg). RESULTS The predicted maximum plasma concentration ratio between GLE/PIB + fentanyl and fentanyl alone was 1.00, and the predicted area under the curve ratio was 1.04, suggesting an increase of only 4% in fentanyl exposure. CONCLUSION The administration of a therapeutic dose of GLE/PIB has very little effect on the pharmacokinetics of intravenous fentanyl. This negligible increase would not be expected to increase the risk of fentanyl overdose beyond the inherent risks related to the amount and purity of the fentanyl received during recreational use.
Collapse
Affiliation(s)
| | | | | | | | | | - Anthony Martinez
- Jacobs School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Brian Conway
- Vancouver Infectious Diseases Centre, Vancouver, Canada
- Simon Fraser University, Burnaby, Canada
| | - Tipu Khan
- Ventura County Medical Center, Ventura, CA, USA
- USC Keck School of Medicine, Los Angeles, CA, USA
| | | |
Collapse
|
5
|
Fairman K, Choi MK, Gonnabathula P, Lumen A, Worth A, Paini A, Li M. An Overview of Physiologically-Based Pharmacokinetic Models for Forensic Science. TOXICS 2023; 11:126. [PMID: 36851001 PMCID: PMC9964742 DOI: 10.3390/toxics11020126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
A physiologically-based pharmacokinetic (PBPK) model represents the structural components of the body with physiologically relevant compartments connected via blood flow rates described by mathematical equations to determine drug disposition. PBPK models are used in the pharmaceutical sector for drug development, precision medicine, and the chemical industry to predict safe levels of exposure during the registration of chemical substances. However, one area of application where PBPK models have been scarcely used is forensic science. In this review, we give an overview of PBPK models successfully developed for several illicit drugs and environmental chemicals that could be applied for forensic interpretation, highlighting the gaps, uncertainties, and limitations.
Collapse
Affiliation(s)
- Kiara Fairman
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Me-Kyoung Choi
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Pavani Gonnabathula
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | | | - Miao Li
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
6
|
Mehta D, Li M, Nakamura N, Chidambaram M, He X, Bryant MS, Patton R, Davis K, Fisher J. In vivo pharmacokinetic analyses of placental transfer of three drugs of different physicochemical properties in pregnant rats. Reprod Toxicol 2022; 111:194-203. [PMID: 35714934 DOI: 10.1016/j.reprotox.2022.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022]
Abstract
Although the use of medication during pregnancy is common, information on exposure to the developing fetus and potential teratogenic effects is often lacking. This study used a rat model to examine the placental transfer of three small-molecule drugs with molecular weights ranging from approximately 300 to 800 Da with different physicochemical properties. Time-mated Sprague Dawley (Hsd:SD) rats aged 11-13 weeks were administered either glyburide, rifaximin, or fentanyl at gestational day 15. Maternal blood, placentae, and fetuses were collected at 5 min, 30 min, 1 h, 4 h, 8 h, 24 h, 48 h, and 96 h post-dose. To characterize the rate and extent of placental drug transfer, we calculated several pharmacokinetic parameters such as maximum concentration (Cmax), time to maximum concentration (Tmax), area under the concentration-time curve (AUC), half-life (t1/2), clearance (CL), and volume of distribution (Vd) for plasma, placenta, and fetus tissues. The results indicated showed that fetal exposure was lowest for glyburide, accounting for only 2.2 % of maternal plasma exposure as measured by their corresponding AUC ratio, followed by rifaximin (37.9 %) and fentanyl (172.4 %). The fetus/placenta AUC ratios were found to be 10.7 % for glyburide, 11.8 % for rifaximin, and 39.1 % for fentanyl. These findings suggest that although the placenta acts as a protective shield for the fetus, the extent of protection varies for different drugs and depends on factors such as molecular weight, lipid solubility, transporter-mediated efflux, and binding to maternal and fetal plasma proteins.
Collapse
Affiliation(s)
- Darshan Mehta
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Miao Li
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Noriko Nakamura
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Mani Chidambaram
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Xiaobo He
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Matthew S Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Ralph Patton
- Toxicologic Pathology Associates, Jefferson, AR 72079, USA
| | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR 72079, USA
| | - Jeffrey Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
7
|
Zhu J, Zhao Y, Wang L, Zhou C, Zhou S, Chen T, Chen J, Zhang Z, Zhu Y, Ding S, Shao F. Physiologically based pharmacokinetic/pharmacodynamic modeling to evaluate the absorption of midazolam rectal gel. Eur J Pharm Sci 2021; 167:106006. [PMID: 34520836 DOI: 10.1016/j.ejps.2021.106006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We aimed to 1) develop physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) models of a novel midazolam rectal gel in healthy adults, 2) assess the contribution of different physiologically relevant factors in rectal absorption, and 3) to provide supports for future clinical studies of midazolam rectal gel. METHODS We developed the rectal PBPK model after built the intravenous and the oral PBPK model. Then, the physiological progress of rectal route was described in terms of the drug release, the rectal absorption and the particle first-pass elimination. Next, the validated PBPK model was combined with the sigmoid Emax PD model. This PBPK/PD model was used to identify the dose range and the critical parameters to ensure safety sedation. RESULTS Based on the simulations, the recommended maximum dose for adults' sedation was 15 mg. And the retention time of midazolam rectal gel should be longer than 3 h to reach over 80% pharmacokinetics and pharmacodynamics effects. CONCLUSION We successfully developed a PBPK/PD model for the midazolam rectal gel, which accurately described the PK/PD behavior in healthy adults and indicated the transit time of rectum was the most sensitive parameter for absorption. This PBPK/PD model would be expected to support the future clinical studies and pediatric application.
Collapse
Affiliation(s)
- Jinying Zhu
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| | - Yuqing Zhao
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Lu Wang
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Chen Zhou
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Sufeng Zhou
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Tao Chen
- Shanghai PharmoGo Co., Ltd, 3F, Block B, Weitai Building, No. 58, Lane 91, Shanghai, 200127, China
| | - Juan Chen
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Zeru Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhu
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China
| | - Sijia Ding
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Feng Shao
- Phase I Clinical Trial Unit, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
8
|
Riad MH, Baynes RE, Tell LA, Davis JL, Maunsell FP, Riviere JE, Lin Z. Development and Application of an interactive Physiologically Based Pharmacokinetic (iPBPK) Model to Predict Oxytetracycline Tissue Distribution and Withdrawal Intervals in Market-Age Sheep and Goats. Toxicol Sci 2021; 183:253-268. [PMID: 34329480 DOI: 10.1093/toxsci/kfab095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oxytetracycline (OTC) is a widely used antibiotic in food-producing animals. Extralabel use of OTC is common and may lead to violative residues in edible tissues. It is important to have a quantitative tool to predict scientifically-based withdrawal intervals (WDIs) after extralabel use in food animals to ensure human food safety. This study focuses on developing a physiologically based pharmacokinetic (PBPK) model for OTC in sheep and goats. The model included seven compartments: plasma, lung, liver, kidneys, muscle, fat, and rest of the body. The model was calibrated with serum and tissue (liver, muscle, kidney, and fat) concentration data following a single intramuscular (IM, 20 mg/kg) and/or intravenous (IV, 10 mg/kg) administration of a long-acting formulation in sheep and goats. The model was evaluated with independent datasets from Food Animal Residue Avoidance Databank (FARAD). Results showed that the model adequately simulated the calibration datasets with an overall estimated coefficient of determination (R2) of 0.95 and 0.92, respectively, for sheep and goat models and had acceptable accuracy for the validation datasets. Monte Carlo sampling technique was applied to predict the time needed for drug concentrations in edible tissues to fall below tolerances for the 99th percentiles of the population. The model was converted to a web-based interactive PBPK (iPBPK) interface to facilitate model applications. This iPBPK model provides a useful tool to estimate WDIs for OTC after extralabel use in small ruminants to ensure food safety and serves as a basis for extrapolation to other tetracycline drugs and other food animals.
Collapse
Affiliation(s)
- Mahbubul H Riad
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506.,Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL 32608, USA
| | - Ronald E Baynes
- Center for Chemical Toxicology Research and Pharmacokinetics, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606
| | - Lisa A Tell
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616
| | - Jennifer L Davis
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24060
| | - Fiona P Maunsell
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608
| | - Jim E Riviere
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506.,Center for Chemical Toxicology Research and Pharmacokinetics, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506.,Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL 32608, USA
| |
Collapse
|
9
|
Chou WC, Lin Z. Development of a Gestational and Lactational Physiologically Based Pharmacokinetic (PBPK) Model for Perfluorooctane Sulfonate (PFOS) in Rats and Humans and Its Implications in the Derivation of Health-Based Toxicity Values. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:37004. [PMID: 33730865 PMCID: PMC7969127 DOI: 10.1289/ehp7671] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 05/03/2023]
Abstract
BACKGROUND There is a great concern on potential adverse effects of exposure to perfluorooctane sulfonate (PFOS) in sensitive subpopulations, such as pregnant women, fetuses, and neonates, due to its reported transplacental and lactational transfer and reproductive and developmental toxicities in animals and humans. OBJECTIVES This study aimed to develop a gestational and lactational physiologically based pharmacokinetic (PBPK) model in rats and humans for PFOS to aid risk assessment in sensitive human subpopulations. METHODS Based upon existing PBPK models for PFOS, the present model addressed a data gap of including a physiologically based description of basolateral and apical membrane transporter-mediated renal reabsorption and excretion in kidneys during gestation and lactation. The model was calibrated with published rat toxicokinetic and human biomonitoring data and was independently evaluated with separate data. Monte Carlo simulation was used to address the interindividual variability. RESULTS Model simulations were generally within 2-fold of observed PFOS concentrations in maternal/fetal/neonatal plasma and liver in rats and humans. Estimated fifth percentile human equivalent doses (HEDs) based on selected critical toxicity studies in rats following U.S. Environmental Protection Agency (EPA) guidelines ranged from 0.08 to 0.91 μ g / kg per day . These values are lower than the HEDs estimated in U.S. EPA guidance (0.51 - 1.6 μ g / kg per day ) using an empirical toxicokinetic model in adults. CONCLUSIONS The results support the importance of renal reabsorption/excretion during pregnancy and lactation in PFOS dosimetry and suggest that the derivation of health-based toxicity values based on developmental toxicity studies should consider gestational/lactational dosimetry estimated from a life stage-appropriate PBPK model. This study provides a quantitative tool to aid risk reevaluation of PFOS, especially in sensitive human subpopulations, and it provides a basis for extrapolating to other per- and polyfluoroalkyl substances (PFAS). All model codes and detailed tutorials are provided in the Supplemental Materials to allow readers to reproduce our results and to use this model. https://doi.org/10.1289/EHP7671.
Collapse
Affiliation(s)
- Wei-Chun Chou
- Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
10
|
George B, Lumen A, Nguyen C, Wesley B, Wang J, Beitz J, Crentsil V. Application of physiologically based pharmacokinetic modeling for sertraline dosing recommendations in pregnancy. NPJ Syst Biol Appl 2020; 6:36. [PMID: 33159093 PMCID: PMC7648747 DOI: 10.1038/s41540-020-00157-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/02/2020] [Indexed: 01/26/2023] Open
Abstract
Pregnancy is a period of significant change that impacts physiological and metabolic status leading to alterations in the disposition of drugs. Uncertainty in drug dosing in pregnancy can lead to suboptimal therapy, which can contribute to disease exacerbation. A few studies show there are increased dosing requirements for antidepressants in late pregnancy; however, the quantitative data to guide dose adjustments are sparse. We aimed to develop a physiologically based pharmacokinetic (PBPK) model that allows gestational-age dependent prediction of sertraline dosing in pregnancy. A minimal physiological model with defined gut, liver, plasma, and lumped placental-fetal compartments was constructed using the ordinary differential equation solver package, ‘mrgsolve’, in R. We extracted data from the literature to parameterize the model, including sertraline physicochemical properties, in vitro metabolism studies, disposition in nonpregnant women, and physiological changes during pregnancy. The model predicted the pharmacokinetic parameters from a clinical study with eight subjects for the second trimester and six subjects for the third trimester. Based on the model, gestational-dependent changes in physiology and metabolism account for increased clearance of sertraline (up to 143% at 40 weeks gestational age), potentially leading to under-dosing of pregnant women when nonpregnancy doses are used. The PBPK model was converted to a prototype web-based interactive dosing tool to demonstrate how the output of a PBPK model may translate into optimal sertraline dosing in pregnancy. Quantitative prediction of drug exposure using PBPK modeling in pregnancy will support clinically appropriate dosing and increase the therapeutic benefit for pregnant women.
Collapse
Affiliation(s)
- Blessy George
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA.,Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Annie Lumen
- National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Christine Nguyen
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Barbara Wesley
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Jian Wang
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Julie Beitz
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Victor Crentsil
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA.
| |
Collapse
|
11
|
Physiologically-Based Pharmacokinetic (PBPK) Modeling Providing Insights into Fentanyl Pharmacokinetics in Adults and Pediatric Patients. Pharmaceutics 2020; 12:pharmaceutics12100908. [PMID: 32977559 PMCID: PMC7598194 DOI: 10.3390/pharmaceutics12100908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 11/17/2022] Open
Abstract
Fentanyl is widely used for analgesia, sedation, and anesthesia both in adult and pediatric populations. Yet, only few pharmacokinetic studies of fentanyl in pediatrics exist as conducting clinical trials in this population is especially challenging. Physiologically-based pharmacokinetic (PBPK) modeling is a mechanistic approach to explore drug pharmacokinetics and allows extrapolation from adult to pediatric populations based on age-related physiological differences. The aim of this study was to develop a PBPK model of fentanyl and norfentanyl for both adult and pediatric populations. The adult PBPK model was established in PK-Sim® using data from 16 clinical studies and was scaled to several pediatric subpopulations. ~93% of the predicted AUClast values in adults and ~88% in pediatrics were within 2-fold of the corresponding value observed. The adult PBPK model predicted a fraction of fentanyl dose metabolized to norfentanyl of ~33% and a fraction excreted in urine of ~7%. In addition, the pediatric PBPK model was used to simulate differences in peak plasma concentrations after bolus injections and short infusions. The novel PBPK models could be helpful to further investigate fentanyl pharmacokinetics in both adult and pediatric populations.
Collapse
|
12
|
Tan YM, Chan M, Chukwudebe A, Domoradzki J, Fisher J, Hack CE, Hinderliter P, Hirasawa K, Leonard J, Lumen A, Paini A, Qian H, Ruiz P, Wambaugh J, Zhang F, Embry M. PBPK model reporting template for chemical risk assessment applications. Regul Toxicol Pharmacol 2020; 115:104691. [PMID: 32502513 PMCID: PMC8188465 DOI: 10.1016/j.yrtph.2020.104691] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/04/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling analysis does not stand on its own for regulatory purposes but is a robust tool to support drug/chemical safety assessment. While the development of PBPK models have grown steadily since their emergence, only a handful of models have been accepted to support regulatory purposes due to obstacles such as the lack of a standardized template for reporting PBPK analysis. Here, we expand the existing guidances designed for pharmaceutical applications by recommending additional elements that are relevant to environmental chemicals. This harmonized reporting template can be adopted and customized by public health agencies receiving PBPK model submission, and it can also serve as general guidance for submitting PBPK-related studies for publication in journals or other modeling sharing purposes. The current effort represents one of several ongoing collaborations among the PBPK modeling and risk assessment communities to promote, when appropriate, incorporating PBPK modeling to characterize the influence of pharmacokinetics on safety decisions made by regulatory agencies.
Collapse
Affiliation(s)
- Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Health Effects Division, 109 TW Alexander Dr, Research Triangle Park, NC, 27709, USA.
| | - Melissa Chan
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19714, USA.
| | - Amechi Chukwudebe
- BASF Corporation, 26 Davis Drive, Research Triangle Park, NC, 27709, USA.
| | - Jeanne Domoradzki
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19714, USA
| | - Jeffrey Fisher
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - C Eric Hack
- ScitoVation, 100 Capitola Drive, Durham, NC, 27713, USA.
| | - Paul Hinderliter
- Syngenta Crop Protection, LLC, 410 Swing Rd, Greensboro, NC, 27409, USA.
| | - Kota Hirasawa
- Sumitomo Chemical Co, Ltd, 1-98, Kasugadenaka 3-chome, Konohana-ku, Osaka, 554-8558, Japan.
| | - Jeremy Leonard
- Oak Ridge Institute for Science and Education, 100 ORAU Way, Oak Ridge, TN, 37830, USA.
| | - Annie Lumen
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - Alicia Paini
- European Commission Joint Research Centre, Via E. Fermi 2749, Ispra I, 21027, Italy.
| | - Hua Qian
- ExxonMobil Biomedical Sciences, Inc, 1545 US Hwy 22 East, Annandale, NJ, 08801, USA.
| | - Patricia Ruiz
- CDC-ATSDR, 4770 Buford Hwy, Mailstop S102-1, Chamblee, GA, 3034, USA.
| | - John Wambaugh
- US Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr, Research Triangle Park, NC, 27711, USA.
| | - Fagen Zhang
- The Dow Chemical Company, 1803 Building, Midland, MI, 48674, USA.
| | - Michelle Embry
- Health and Environmental Sciences Institute, 740 15th Street, NW, Suite 600, Washington, DC, 20005, USA.
| |
Collapse
|
13
|
Ji B, Liu S, Xue Y, He X, Man VH, Xie XQ, Wang J. Prediction of Drug-Drug Interactions Between Opioids and Overdosed Benzodiazepines Using Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulation. Drugs R D 2020; 19:297-305. [PMID: 31482303 PMCID: PMC6738369 DOI: 10.1007/s40268-019-00282-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Researchers have long been interested in the potential drug–drug interactions (DDIs) between opioids and benzodiazepines. However, much remains unknown concerning the interactions between these two drug classes. The objective of this work is to study the mechanism underlying the DDIs between opioids and benzodiazepines from the perspective of their pharmacokinetic (PK) interactions. A PK interaction occurs when two drugs are metabolized by the same cytochrome P450 enzymes and is one of the most common reasons for DDIs. Methods We quantitatively predicted the DDIs between three opioids (fentanyl, oxycodone and buprenorphine) and four benzodiazepines (alprazolam, diazepam, midazolam and triazolam) using a physiologically based pharmacokinetic (PBPK) modeling approach. A set of PBPK models was first constructed for these common opioids and benzodiazepines using SimCYP software, and the DDIs between them were then explored at various dosages. Results Our simulation results suggested there were no PK interactions between normal doses of opioids and benzodiazepines; but weak interactions can be expected with the combination of opioids and overdosed benzodiazepines. Particular attention should be given to the combination of fentanyl and overdosed alprazolam since a PK interaction can be observed between them. Conclusion Our results appear to indicate that pharmacodynamics may play a more important role than PKs in causing DDIs between opioids and benzodiazepines. This study also demonstrated that molecular modeling can be a very useful tool to mitigate the problem of “missing metabolic reaction parameters” in PK modeling and simulation. Electronic supplementary material The online version of this article (10.1007/s40268-019-00282-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beihong Ji
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Shuhan Liu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Ying Xue
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Xibing He
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Viet Hoang Man
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 3501 Terrace, St Pittsburgh, PA 15261 USA
| |
Collapse
|
14
|
Computational framework for predictive PBPK-PD-Tox simulations of opioids and antidotes. J Pharmacokinet Pharmacodyn 2019; 46:513-529. [PMID: 31396799 DOI: 10.1007/s10928-019-09648-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/29/2019] [Indexed: 10/26/2022]
Abstract
The primary goal of this work was to develop a computational tool to enable personalized prediction of pharmacological disposition and associated responses for opioids and antidotes. Here we present a computational framework for physiologically-based pharmacokinetic (PBPK) modeling of an opioid (morphine) and an antidote (naloxone). At present, the model is solely personalized according to an individual's mass. These PK models are integrated with a minimal pharmacodynamic model of respiratory depression induction (associated with opioid administration) and reversal (associated with antidote administration). The model was developed and validated on human data for IV administration of morphine and naloxone. The model can be further extended to consider different routes of administration, as well as to study different combinations of opioid receptor agonists and antagonists. This work provides the framework for a tool that could be used in model-based management of pain, pharmacological treatment of opioid addiction, appropriate use of antidotes for opioid overdose and evaluation of abuse deterrent formulations.
Collapse
|
15
|
Tan YM, Worley RR, Leonard JA, Fisher JW. Challenges Associated With Applying Physiologically Based Pharmacokinetic Modeling for Public Health Decision-Making. Toxicol Sci 2019; 162:341-348. [PMID: 29385573 DOI: 10.1093/toxsci/kfy010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The development and application of physiologically based pharmacokinetic (PBPK) models in chemical toxicology have grown steadily since their emergence in the 1980s. However, critical evaluation of PBPK models to support public health decision-making across federal agencies has thus far occurred for only a few environmental chemicals. In order to encourage decision-makers to embrace the critical role of PBPK modeling in risk assessment, several important challenges require immediate attention from the modeling community. The objective of this contemporary review is to highlight 3 of these challenges, including: (1) difficulties in recruiting peer reviewers with appropriate modeling expertise and experience; (2) lack of confidence in PBPK models for which no tissue/plasma concentration data exist for model evaluation; and (3) lack of transferability across modeling platforms. Several recommendations for addressing these 3 issues are provided to initiate dialog among members of the PBPK modeling community, as these issues must be overcome for the field of PBPK modeling to advance and for PBPK models to be more routinely applied in support of public health decision-making.
Collapse
Affiliation(s)
- Yu-Mei Tan
- National Exposure Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27709
| | - Rachel R Worley
- Agency for Toxic Substances and Disease Registry, Atlanta, Georgia 30341
| | - Jeremy A Leonard
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830
| | - Jeffrey W Fisher
- National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, Arizona 72079
| |
Collapse
|
16
|
Elwell-Cuddy T, Li M, KuKanich B, Lin Z. The construction and application of a population physiologically based pharmacokinetic model for methadone in Beagles and Greyhounds. J Vet Pharmacol Ther 2018; 41:670-683. [DOI: 10.1111/jvp.12676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Trevor Elwell-Cuddy
- Institute of Computational Comparative Medicine (ICCM); Department of Anatomy and Physiology; College of Veterinary Medicine; Kansas State University; Manhattan Kansas
| | - Miao Li
- Institute of Computational Comparative Medicine (ICCM); Department of Anatomy and Physiology; College of Veterinary Medicine; Kansas State University; Manhattan Kansas
| | - Butch KuKanich
- Institute of Computational Comparative Medicine (ICCM); Department of Anatomy and Physiology; College of Veterinary Medicine; Kansas State University; Manhattan Kansas
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine (ICCM); Department of Anatomy and Physiology; College of Veterinary Medicine; Kansas State University; Manhattan Kansas
| |
Collapse
|
17
|
Li M, Gehring R, Riviere JE, Lin Z. Probabilistic Physiologically Based Pharmacokinetic Model for Penicillin G in Milk From Dairy Cows Following Intramammary or Intramuscular Administrations. Toxicol Sci 2018; 164:85-100. [DOI: 10.1093/toxsci/kfy067] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Miao Li
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506
| | - Ronette Gehring
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506
| | - Jim E Riviere
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine (ICCM), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506
| |
Collapse
|
18
|
Worley RR, Yang X, Fisher J. Physiologically based pharmacokinetic modeling of human exposure to perfluorooctanoic acid suggests historical non drinking-water exposures are important for predicting current serum concentrations. Toxicol Appl Pharmacol 2017; 330:9-21. [PMID: 28684146 PMCID: PMC5664934 DOI: 10.1016/j.taap.2017.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 01/09/2023]
Abstract
Manufacturing of perfluorooctanoic acid (PFOA), a synthetic chemical with a long half-life in humans, peaked between 1970 and 2002, and has since diminished. In the United States, PFOA is detected in the blood of >99% of people tested, but serum concentrations have decreased since 1999. Much is known about exposure to PFOA in drinking water; however, the impact of non-drinking water PFOA exposure on serum PFOA concentrations is not well characterized. The objective of this research is to apply physiologically based pharmacokinetic (PBPK) modeling and Monte Carlo analysis to evaluate the impact of historic non-drinking water PFOA exposure on serum PFOA concentrations. In vitro to in vivo extrapolation was utilized to inform descriptions of PFOA transport in the kidney. Monte Carlo simulations were incorporated to evaluate factors that account for the large inter-individual variability of serum PFOA concentrations measured in individuals from North Alabama in 2010 and 2016, and the Mid-Ohio River Valley between 2005 and 2008. Predicted serum PFOA concentrations were within two-fold of experimental data. With incorporation of Monte Carlo simulations, the model successfully tracked the large variability of serum PFOA concentrations measured in populations from the Mid-Ohio River Valley. Simulation of exposure in a population of 45 adults from North Alabama successfully predicted 98% of individual serum PFOA concentrations measured in 2010 and 2016, respectively, when non-drinking water ingestion of PFOA exposure was included. Variation in serum PFOA concentrations may be due to inter-individual variability in the disposition of PFOA and potentially elevated historical non-drinking water exposures.
Collapse
Affiliation(s)
- Rachel Rogers Worley
- Division of Community Health Investigations, Agency for Toxic Substances and Disease Registry, Atlanta, GA, USA; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA.
| | - Xiaoxia Yang
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | - Jeffrey Fisher
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA; National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
19
|
Li M, Gehring R, Riviere JE, Lin Z. Development and application of a population physiologically based pharmacokinetic model for penicillin G in swine and cattle for food safety assessment. Food Chem Toxicol 2017. [DOI: 10.1016/j.fct.2017.06.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
20
|
Clipp RB, Bray A, Metoyer R, Thames MC, Webb JB. Pharmacokinetic and pharmacodynamic modeling in BioGears. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:1467-1470. [PMID: 28268603 DOI: 10.1109/embc.2016.7590986] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Pharmacokinetics/pharmacodynamics models were designed and integrated into the BioGears® physiology engine to address the need for real time drug effects for varying patients and injury profiles. Ten drugs were validated using experimental and subject matter expert data. The plasma concentration curves had a good fit with experimental data and 48 of 50 physiologic parameters displayed a less than 10% error compared to the validation data.
Collapse
|
21
|
Yang X, Duan J, Fisher J. Application of Physiologically Based Absorption Modeling to Characterize the Pharmacokinetic Profiles of Oral Extended Release Methylphenidate Products in Adults. PLoS One 2016; 11:e0164641. [PMID: 27723791 PMCID: PMC5056674 DOI: 10.1371/journal.pone.0164641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/28/2016] [Indexed: 11/30/2022] Open
Abstract
A previously presented physiologically-based pharmacokinetic model for immediate release (IR) methylphenidate (MPH) was extended to characterize the pharmacokinetic behaviors of oral extended release (ER) MPH formulations in adults for the first time. Information on the anatomy and physiology of the gastrointestinal (GI) tract, together with the biopharmaceutical properties of MPH, was integrated into the original model, with model parameters representing hepatic metabolism and intestinal non-specific loss recalibrated against in vitro and in vivo kinetic data sets with IR MPH. A Weibull function was implemented to describe the dissolution of different ER formulations. A variety of mathematical functions can be utilized to account for the engineered release/dissolution technologies to achieve better model performance. The physiological absorption model tracked well the plasma concentration profiles in adults receiving a multilayer-release MPH formulation or Metadate CD, while some degree of discrepancy was observed between predicted and observed plasma concentration profiles for Ritalin LA and Medikinet Retard. A local sensitivity analysis demonstrated that model parameters associated with the GI tract significantly influenced model predicted plasma MPH concentrations, albeit to varying degrees, suggesting the importance of better understanding the GI tract physiology, along with the intestinal non-specific loss of MPH. The model provides a quantitative tool to predict the biphasic plasma time course data for ER MPH, helping elucidate factors responsible for the diverse plasma MPH concentration profiles following oral dosing of different ER formulations.
Collapse
Affiliation(s)
- Xiaoxia Yang
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| | - John Duan
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jeffrey Fisher
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas, United States of America
| |
Collapse
|
22
|
Rezaei Kolahchi A, Khadem Mohtaram N, Pezeshgi Modarres H, Mohammadi MH, Geraili A, Jafari P, Akbari M, Sanati-Nezhad A. Microfluidic-Based Multi-Organ Platforms for Drug Discovery. MICROMACHINES 2016; 7:E162. [PMID: 30404334 PMCID: PMC6189912 DOI: 10.3390/mi7090162] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022]
Abstract
Development of predictive multi-organ models before implementing costly clinical trials is central for screening the toxicity, efficacy, and side effects of new therapeutic agents. Despite significant efforts that have been recently made to develop biomimetic in vitro tissue models, the clinical application of such platforms is still far from reality. Recent advances in physiologically-based pharmacokinetic and pharmacodynamic (PBPK-PD) modeling, micro- and nanotechnology, and in silico modeling have enabled single- and multi-organ platforms for investigation of new chemical agents and tissue-tissue interactions. This review provides an overview of the principles of designing microfluidic-based organ-on-chip models for drug testing and highlights current state-of-the-art in developing predictive multi-organ models for studying the cross-talk of interconnected organs. We further discuss the challenges associated with establishing a predictive body-on-chip (BOC) model such as the scaling, cell types, the common medium, and principles of the study design for characterizing the interaction of drugs with multiple targets.
Collapse
Affiliation(s)
- Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | - Nima Khadem Mohtaram
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | - Mohammad Hossein Mohammadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Armin Geraili
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Parya Jafari
- Department of Electrical Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada.
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
- Center for Bioengineering Research and Education, Biomedical Engineering Program, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
23
|
Yang X, Doerge DR, Teeguarden JG, Fisher JW. Development of a physiologically based pharmacokinetic model for assessment of human exposure to bisphenol A. Toxicol Appl Pharmacol 2015; 289:442-56. [PMID: 26522835 DOI: 10.1016/j.taap.2015.10.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 01/24/2023]
Abstract
A previously developed physiologically based pharmacokinetic (PBPK) model for bisphenol A (BPA) in adult rhesus monkeys was modified to characterize the pharmacokinetics of BPA and its phase II conjugates in adult humans following oral ingestion. Coupled with in vitro studies on BPA metabolism in the liver and the small intestine, the PBPK model was parameterized using oral pharmacokinetic data with deuterated-BPA (d6-BPA) delivered in cookies to adult humans after overnight fasting. The availability of the serum concentration time course of unconjugated d6-BPA offered direct empirical evidence for the calibration of BPA model parameters. The recalibrated PBPK adult human model for BPA was then evaluated against published human pharmacokinetic studies with BPA. A hypothesis of decreased oral uptake was needed to account for the reduced peak levels observed in adult humans, where d6-BPA was delivered in soup and food was provided prior to BPA ingestion, suggesting the potential impact of dosing vehicles and/or fasting on BPA disposition. With the incorporation of Monte Carlo analysis, the recalibrated adult human model was used to address the inter-individual variability in the internal dose metrics of BPA for the U.S. general population. Model-predicted peak BPA serum levels were in the range of pM, with 95% of human variability falling within an order of magnitude. This recalibrated PBPK model for BPA in adult humans provides a scientific basis for assessing human exposure to BPA that can serve to minimize uncertainties incurred during extrapolations across doses and species.
Collapse
Affiliation(s)
- Xiaoxia Yang
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States.
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Justin G Teeguarden
- Health Effects and Exposure Science, Pacific Northwest National Laboratory, Richland, WA 99352, United States; Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Jeffrey W Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| |
Collapse
|
24
|
Musther H, Gill KL, Chetty M, Rostami-Hodjegan A, Rowland M, Jamei M. Are Physiologically Based Pharmacokinetic Models Reporting the Right C(max)? Central Venous Versus Peripheral Sampling Site. AAPS JOURNAL 2015; 17:1268-79. [PMID: 26100012 PMCID: PMC4540731 DOI: 10.1208/s12248-015-9796-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/03/2015] [Indexed: 11/30/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) models can over-predict maximum plasma concentrations (Cmax) following intravenous administration. A proposed explanation is that invariably PBPK models report the concentration in the central venous compartment, rather than the site where the samples are drawn. The purpose of this study was to identify and validate potential corrective models based on anatomy and physiology governing the blood supply at the site of sampling and incorporate them into a PBPK platform. Four models were developed and scrutinised for their corrective potential. All assumed the peripheral sampling site concentration could be described by contributions from surrounding tissues and utilised tissue-specific concentration-time profiles reported from the full-PBPK model within the Simcyp Simulator. Predicted concentrations for the peripheral site were compared to the observed Cmax. The models results were compared to clinical data for 15 studies over seven compounds (alprazolam, imipramine, metoprolol, midazolam, omeprazole, rosiglitazone and theophylline). The final model utilised tissue concentrations from adipose, skin, muscle and a contribution from artery. Predicted Cmax values considering the central venous compartment can over-predict the observed values up to 10-fold whereas the new sampling site predictions were within 2-fold of observed values. The model was particularly relevant for studies where traditional PBPK models over-predict early time point concentrations. A successful corrective model for Cmax prediction has been developed, subject to further validation. These models can be enrolled as built-up modules into PBPK platforms and potentially account for factors that may affect the initial mixing of the blood at the site of sampling.
Collapse
Affiliation(s)
- Helen Musther
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK,
| | | | | | | | | | | |
Collapse
|
25
|
Blanco ME, Encinas E, González O, Rico E, Vozmediano V, Suárez E, Alonso RM. Quantitative determination of fentanyl in newborn pig plasma and cerebrospinal fluid samples by HPLC-MS/MS. Drug Test Anal 2015; 7:804-11. [PMID: 25755165 DOI: 10.1002/dta.1778] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 11/07/2022]
Abstract
In this study, a selective and sensitive high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method requiring low sample volume (≤100 μL) was developed and validated for the quantitative determination of the opioid drug fentanyl in plasma and cerebrospinal fluid (CSF). A protein precipitation extraction with acetonitrile was used for plasma samples whereas CSF samples were injected directly on the HPLC column. Fentanyl and (13) C6 -fentanyl (Internal Standard) were analyzed in an electrospray ionization source in positive mode, with multiple reaction monitoring (MRM) of the transitions m/z 337.0/188.0 and m/z 337.0/105.0 for quantification and confirmation of fentanyl, and m/z 343.0/188.0 for (13) C6 -fentanyl. The respective lowest limits of quantification for plasma and CSF were 0.2 and 0.25 ng/mL. Intra- and inter-assay precision and accuracy did not exceed 15%, in accordance with bioanalytical validation guidelines. The described analytical method was proven to be robust and was successfully applied to the determination of fentanyl in plasma and CSF samples from a pharmacokinetic and pharmacodynamic study in newborn piglets receiving intravenous fentanyl (5 µg/kg bolus immediately followed by a 90-min infusion of 3 µg/kg/h).
Collapse
Affiliation(s)
- M E Blanco
- Analytical Chemistry Department, Science and Technology Faculty, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - E Encinas
- Pharmacology Department, Faculty of Medicine, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - O González
- Analytical Chemistry Department, Science and Technology Faculty, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain.,Analytical Bioscience Division, LACDR, Leiden University, Leiden, the Netherlands
| | - E Rico
- Analytical Chemistry Department, Science and Technology Faculty, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - V Vozmediano
- Drug Modeling & Consulting, Dynakin, SL, Bilbao, Basque Country, Spain
| | - E Suárez
- Pharmacology Department, Faculty of Medicine, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - R M Alonso
- Analytical Chemistry Department, Science and Technology Faculty, University of the Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| |
Collapse
|
26
|
Xia B, Yang Z, Zhou H, Lukacova V, Zhu W, Milewski M, Kesisoglou F. Development of a Novel Oral Cavity Compartmental Absorption and Transit Model for Sublingual Administration: Illustration with Zolpidem. AAPS JOURNAL 2015; 17:631-42. [PMID: 25716146 DOI: 10.1208/s12248-015-9727-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/29/2015] [Indexed: 11/30/2022]
Abstract
Intraoral (IO) delivery is an alternative administration route to deliver a drug substance via the mouth that provides several advantages over conventional oral dosage forms. The purpose of this work was to develop and evaluate a novel, physiologically based oral cavity model for projection and mechanistic analysis of the clinical pharmacokinetics of intraoral formulations. The GastroPlus™ Oral Cavity Compartmental Absorption and Transit (OCCAT™) model was used to simulate the plasma concentration versus time profiles and the fraction and rate of intraoral drug transit/absorption for Intermezzo® sublingual tablets (zolpidem tartrate). The model was evaluated by the goodness-of-fit between simulated and observed concentrations and the deviation of key PK parameters (e.g., C max, T max, and AUC). In addition, a sensitivity analysis was conducted to demonstrate the interplay and impact of key modeling parameters on the fraction absorbed via oral mucosa (F a_IO). The OCCAT™ model captured the observed pharmacokinetics for Intermezzo® sublingual tablets (R (2) > 0.9). The predicted deviations (%) for C max, AUC0-inf, AUC0-20 min, and T max were 5.7, 28.0, 11.8, and 28.6%, respectively, indicating good prediction accuracy. The model also estimated ~18% of total drug was absorbed via the IO route. Furthermore, the sensitivity analysis indicated that the F a_IO was not only associated with drug diffusivity and unbound fraction in epithelium tissue (f ut) but also depended on the physicochemical properties of compounds for IO delivery (e.g., solubility and logD pH = 7.4). The novel physiologically based IO absorption OCCAT™ model showed satisfactory performance and will be helpful to guide development of future intraoral formulations.
Collapse
Affiliation(s)
- Binfeng Xia
- Biopharmaceutics, Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc., West Point, Pennsylvania, 19486, USA,
| | | | | | | | | | | | | |
Collapse
|
27
|
Yang X, Zhou YF, Yu Y, Zhao DH, Shi W, Fang BH, Liu YH. A physiologically based pharmacokinetic model for quinoxaline-2-carboxylic acid in rats, extrapolation to pigs. J Vet Pharmacol Ther 2014; 38:55-64. [DOI: 10.1111/jvp.12143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/25/2014] [Indexed: 11/26/2022]
Affiliation(s)
- X. Yang
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - Y.-F. Zhou
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - Y. Yu
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - D.-H. Zhao
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - W. Shi
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - B.-H. Fang
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| | - Y.-H. Liu
- Laboratory of Veterinary Pharmacology; College of Veterinary Medicine; South China Agricultural University; Guangzhou China
| |
Collapse
|
28
|
Takaku T, Nagahori H, Sogame Y. Metabolism and physiologically based pharmacokinetic modeling of flumioxazin in pregnant animals. Toxicol Appl Pharmacol 2014; 277:242-9. [DOI: 10.1016/j.taap.2014.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/19/2014] [Accepted: 03/23/2014] [Indexed: 01/13/2023]
|