1
|
Najjar A, Hamadeh A, Krause S, Schepky A, Edginton A. Global sensitivity analysis of Open Systems Pharmacology Suite physiologically based pharmacokinetic models. CPT Pharmacometrics Syst Pharmacol 2024. [PMID: 39498820 DOI: 10.1002/psp4.13256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Sensitivity analyses are important components of physiologically based pharmacokinetic (PBPK) model development and are required by regulatory agencies for PBPK submissions. They assess the impact of parametric uncertainty and variability on model estimates, aid model optimization by identifying parameters requiring calibration, and enable the testing of assumptions within PBPK models. One-at-a-time (OAT) sensitivity analyses quantify the impact on a model output in response to changes in a single parameter while holding others fixed. Global sensitivity analysis (GSA) methods provide more comprehensive assessments by accounting for changes in all uncertain or variable parameters, though at a higher computational cost. This tutorial article presents a software package for conducting both OAT and GSA of PBPK models built in the Open Systems Pharmacology (OSP) Suite. The tool is accessible through either an R script or a graphical user interface, and the outputs consist of sensitivity metrics of pharmacokinetic (PK) parameters, such as Cmax and AUC, evaluated with respect to model input parameters. Results are formatted according to regulatory standards. The OAT analysis methods comprise two-way local sensitivity analyses and probabilistic uncertainty analyses, whereas the GSA methods include the Morris, Sobol, and EFAST methods. These analyses can be conducted on single PBPK models or pairs of models for the evaluation of the sensitivity of PK parameter ratios in drug-drug interaction studies. The practical application of the package is demonstrated through three illustrative case studies.
Collapse
Affiliation(s)
| | - Abdullah Hamadeh
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
- Systems In Silico Ltd., Waterloo, Ontario, Canada
| | | | | | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| |
Collapse
|
2
|
Robins D, Lehmann A, Krollik K, Vertzoni M. Analyzing parametric influences driving age-associated changes in absorption using a PBPK-GSA approach. Eur J Pharm Sci 2024; 202:106881. [PMID: 39179162 DOI: 10.1016/j.ejps.2024.106881] [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: 05/28/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
The advanced age population may be susceptible to an increased risk of adverse effects due to increased drug exposure after oral dosing. Factors such as high-interindividual variability and lack of data has led to poor characterization of absorption's role in pharmacokinetic changes in this population. Physiologically based pharmacokinetic (PBPK) models are increasingly being used during the drug development process, as their unique qualities are advantageous in atypical scenarios such as drug-drug interactions or special populations such as older people. Along with relying on various sources of data, auxiliary tools including parameter estimation and sensitivity analysis techniques are employed to support model development and other applications. However, sensitivity analyses have mostly been limited to localized techniques in the majority of reported PBPK models using them. This is disadvantageous, since local sensitivity analyses are unsuitable for risk analysis, which require assessment of parametric interactions and proper coverage of the input space to better estimate and subsequently mitigate the effects of the phenomenon of interest. For this reason, this study seeks to integrate a global sensitivity analysis screening method with PBPK models based in PK-Sim® to characterize the consequences of potential changes in absorption that are often associated with advanced age. The Elementary Effects (Morris) method and visualization of the results are implemented in R and three model drugs representing Biopharmaceutical Classification System classes I-III that are expected to exhibit some sensitivity to three age-associated hypotheses were successfully tested.
Collapse
Affiliation(s)
- Donnia Robins
- Global Drug Product Development, Global CMC Development, Merck KGaA, Frankfurter Straße 250, Darmstadt, Germany; Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Andreas Lehmann
- Global Drug Product Development, Global CMC Development, Merck KGaA, Frankfurter Straße 250, Darmstadt, Germany.
| | - Katharina Krollik
- Global Drug Product Development, Global CMC Development, Merck KGaA, Frankfurter Straße 250, Darmstadt, Germany
| | - Maria Vertzoni
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
3
|
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
|
4
|
Tsiros P, Minadakis V, Li D, Sarimveis H. Parameter grouping and co-estimation in physiologically based kinetic models using genetic algorithms. Toxicol Sci 2024; 200:31-46. [PMID: 38637946 PMCID: PMC11199918 DOI: 10.1093/toxsci/kfae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Physiologically based kinetic (PBK) models are widely used in pharmacology and toxicology for predicting the internal disposition of substances upon exposure, voluntarily or not. Due to their complexity, a large number of model parameters need to be estimated, either through in silico tools, in vitro experiments, or by fitting the model to in vivo data. In the latter case, fitting complex structural models on in vivo data can result in overparameterization and produce unrealistic parameter estimates. To address these issues, we propose a novel parameter grouping approach, which reduces the parametric space by co-estimating groups of parameters across compartments. Grouping of parameters is performed using genetic algorithms and is fully automated, based on a novel goodness-of-fit metric. To illustrate the practical application of the proposed methodology, two case studies were conducted. The first case study demonstrates the development of a new PBK model, while the second focuses on model refinement. In the first case study, a PBK model was developed to elucidate the biodistribution of titanium dioxide (TiO2) nanoparticles in rats following intravenous injection. A variety of parameter estimation schemes were employed. Comparative analysis based on goodness-of-fit metrics demonstrated that the proposed methodology yields models that outperform standard estimation approaches, while utilizing a reduced number of parameters. In the second case study, an existing PBK model for perfluorooctanoic acid (PFOA) in rats was extended to incorporate additional tissues, providing a more comprehensive portrayal of PFOA biodistribution. Both models were validated through independent in vivo studies to ensure their reliability.
Collapse
Affiliation(s)
- Periklis Tsiros
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Vasileios Minadakis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Dingsheng Li
- School of Public Health, University of Nevada, Reno, Nevada 89557-0274, USA
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| |
Collapse
|
5
|
Toshimoto K. Beyond the basics: A deep dive into parameter estimation for advanced PBPK and QSP models. Drug Metab Pharmacokinet 2024; 56:101011. [PMID: 38833901 DOI: 10.1016/j.dmpk.2024.101011] [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: 11/06/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 06/06/2024]
Abstract
Physiologically-based pharmacokinetic (PBPK) models and quantitative systems pharmacology (QSP) models have contributed to drug development strategies. The parameters of these models are commonly estimated by capturing observed values using the nonlinear least-squares method. Software packages for PBPK and QSP modeling provide a range of parameter estimation algorithms. To choose the most appropriate method, modelers need to understand the basic concept of each approach. This review provides a general introduction to the key points of parameter estimation with a focus on the PBPK and QSP models, and the respective parameter estimation algorithms. The latter part assesses the performance of five parameter estimation algorithms - the quasi-Newton method, Nelder-Mead method, genetic algorithm, particle swarm optimization, and Cluster Gauss-Newton method - using three examples of PBPK and QSP modeling. The assessment revealed that some parameter estimation results were significantly influenced by the initial values. Moreover, the choice of algorithms demonstrating good estimation results heavily depends on factors such as model structure and the parameters to be estimated. To obtain credible parameter estimation results, it is advisable to conduct multiple rounds of parameter estimation under different conditions, employing various estimation algorithms.
Collapse
Affiliation(s)
- Kota Toshimoto
- Systems Pharmacology, Non-Clinical Biomedical Science, Applied Research & Operations, Astellas Pharma Inc., Ibaraki, Japan.
| |
Collapse
|
6
|
Mann J, Meshkin H, Zirkle J, Han X, Thrasher B, Chaturbedi A, Arabidarrehdor G, Li Z. Mechanism-based organization of neural networks to emulate systems biology and pharmacology models. Sci Rep 2024; 14:12082. [PMID: 38802422 PMCID: PMC11130269 DOI: 10.1038/s41598-024-59378-9] [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: 11/22/2023] [Accepted: 04/10/2024] [Indexed: 05/29/2024] Open
Abstract
Deep learning neural networks are often described as black boxes, as it is difficult to trace model outputs back to model inputs due to a lack of clarity over the internal mechanisms. This is even true for those neural networks designed to emulate mechanistic models, which simply learn a mapping between the inputs and outputs of mechanistic models, ignoring the underlying processes. Using a mechanistic model studying the pharmacological interaction between opioids and naloxone as a proof-of-concept example, we demonstrated that by reorganizing the neural networks' layers to mimic the structure of the mechanistic model, it is possible to achieve better training rates and prediction accuracy relative to the previously proposed black-box neural networks, while maintaining the interpretability of the mechanistic simulations. Our framework can be used to emulate mechanistic models in a large parameter space and offers an example on the utility of increasing the interpretability of deep learning networks.
Collapse
Affiliation(s)
- John Mann
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Hamed Meshkin
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Joel Zirkle
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Xiaomei Han
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Bradlee Thrasher
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Anik Chaturbedi
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Ghazal Arabidarrehdor
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Zhihua Li
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, WO Bldg 64 Rm 2084, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
7
|
Lumen A, Zhang X, Dutta S, Upreti VV. Predicting Clinical Pharmacokinetics/Pharmacodynamics and Impact of Organ Impairment on siRNA-Based Therapeutics Using a Mechanistic Physiologically-Based Pharmacokinetic-Pharmacodynamic Model. Clin Pharmacol Ther 2024; 115:1054-1064. [PMID: 38282246 DOI: 10.1002/cpt.3160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Approved and emerging siRNA therapeutics are primarily designed for targeted delivery to liver where the therapeutic gene silencing effects occurs. Impairment of hepatic/renal function and its impact on siRNA pharmacokinetics/pharmacodynamics (PKs/PDs) are yet to be mechanistically evaluated to describe the unanticipated clinical observations for this novel modality. We developed pathophysiologically relevant models for organ impairment within a physiologically-based PK-PD (PBPK-PD) modeling framework focusing on modality-specific mechanistic factors to evaluate impact on siRNA PKs and PDs. PBPK-PD models for two US Food and Drug Administration (FDA) approved siRNAs inclisiran and vutrisiran were developed as case studies leveraging available tissue-specific data and translated to humans. Key determinants of the clinical PK and PD of N-acetylgalactosamine conjugated siRNAs (GalNAc-siRNAs) with varying sequences were also identified to inform effective clinical translation strategies for emerging GalNAc-siRNA candidates. A 30-70% reduction in hepatic asialoglycoprotein receptors concentrations still allowed for sufficient amount of free cytoplasmic siRNA for RISC-loading to produce PD effects comparable in extent and duration to normal liver function. This included severe hepatic impairment for which no clinical data are available. Inclusion of other modality agnostic physiological changes relevant to organ impairment did not alter the findings. Changes in renal physiologies, including changes in GFR across various degrees of impairment, well predicted minimal changes in PD for inclisiran and vutrisiran. This work provides a quantitative mechanistic framework and insights on modality-specific factors that drive clinical translation and patient/disease-related factors that impact specific dosing considerations and clinical outcomes to help accelerate the optimal development of siRNA therapeutics.
Collapse
Affiliation(s)
- Annie Lumen
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| | - Xinwen Zhang
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| | - Sandeep Dutta
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling, and Simulation, Amgen Inc., South San Francisco, California, USA
| |
Collapse
|
8
|
Kesharwani SS, Louit G, Ibrahim F. The Use of Global Sensitivity Analysis to Assess the Oral Absorption of Weakly Basic Compounds: A Case Example of Dipyridamole. Pharm Res 2024; 41:877-890. [PMID: 38538971 DOI: 10.1007/s11095-024-03688-0] [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: 01/23/2024] [Accepted: 03/04/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE To utilize the global system analysis (GSA) in oral absorption modeling to gain a deeper understanding of system behavior, improve model accuracy, and make informed decisions during drug development. METHODS GSA was utilized to give insight into which drug substance (DS), drug product (DP), and/or physiological parameter would have an impact on peak plasma concentration (Cmax) and area under the curve (AUC) of dipyridamole as a model weakly basic compound. GSA guided the design of in vitro experiments and oral absorption risk assessment using FormulatedProducts v2202.1.0. The solubility and precipitation profiles of dipyridamole in different bile salt concentrations were measured. The results were then used to build a mechanistic oral absorption model. RESULTS GSA warranted further investigation into the precipitation kinetics and its link to the levels of bile salt concentrations. Mechanistic modeling studies demonstrated that a precipitation-integrated modeling approach appropriately predicted the mean plasma profiles, Cmax, and AUC from the clinical studies. CONCLUSIONS This work shows the value of GSA utilization in early development to guide in vitro experimentation and build more confidence in identifying the critical parameters for the mathematical models.
Collapse
Affiliation(s)
- Siddharth S Kesharwani
- US Early Development Biopharmacy, Synthetics Platform, Sanofi, 350 Water St, Cambridge, MA, 02141, USA
| | - Guillaume Louit
- Siemens K.K, DI SW Division, 1-6-1 Miyahara, Osaka, 532-0003, Japan
| | - Fady Ibrahim
- US Early Development Biopharmacy, Synthetics Platform, Sanofi, 350 Water St, Cambridge, MA, 02141, USA.
| |
Collapse
|
9
|
Husøy T, Caspersen IH, Thépaut E, Knutsen H, Haug LS, Andreassen M, Gkrillas A, Lindeman B, Thomsen C, Herzke D, Dirven H, Wojewodzic MW. Comparison of aggregated exposure to perfluorooctanoic acid (PFOA) from diet and personal care products with concentrations in blood using a PBPK model - Results from the Norwegian biomonitoring study in EuroMix. ENVIRONMENTAL RESEARCH 2023; 239:117341. [PMID: 37839534 DOI: 10.1016/j.envres.2023.117341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) constitute a large group of compounds that are water, stain, and oil repellent. Numerous sources contribute to the blood levels of PFAS in the European population. The main contributor for perfluorooctanoic acid (PFOA) is food, house dust, consumer products and personal care products (PCPs). OBJECTIVES The purpose of the present work is to calculate the dietary and dermal external exposure to PFOA, estimate the aggregated internal exposure from diet and PCPs using a PBPK model, and compare estimates with measured concentrations. METHODS Detailed information on diet and PCP use from the EuroMix study is combined with concentration data of PFOA in food and PCPs in a probabilistic exposure assessment. A physiologically based pharmacokinetic model (PBPK) was further refined by incorporating a dermal exposure pathway, and changes in the kidney and faecal excretion. RESULTS The aggregated internal exposure using the PBPK model shows that the major contributor to the internal exposure is diet for both males and females. The estimated internal exposure of PFOA for the EuroMix population was in the same range but lower than the measured blood concentrations using the lower bound (LB) external exposure estimates, showing that the LB estimates are underestimations. For seven females the internal exposure of PFOA were higher from PCPs than from diet. CONCLUSION PCPs and diet contributed in the same range to the internal PFOA exposure for several women participating in EuroMix. This calls for additional studies on exposure to PFOA and possibly other PFAS from PCPs, especially for women. Overall, PBPK modelling was shown as valuable tool in understanding the sources of PFOA exposure and in guiding risk assessments and regulatory decisions.
Collapse
Affiliation(s)
- T Husøy
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway.
| | - I H Caspersen
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - E Thépaut
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
| | - H Knutsen
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway
| | - L S Haug
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway
| | - M Andreassen
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
| | - A Gkrillas
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
| | - B Lindeman
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway
| | - C Thomsen
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway
| | - D Herzke
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
| | - H Dirven
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
| | - M W Wojewodzic
- The Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway; The Norwegian Institute of Public Health, Centre for Sustainable Diets, Oslo, Norway; Cancer Registry of Norway, Section for Molecular Epidemiology and Infections, Oslo, Norway
| |
Collapse
|
10
|
Moreau M, Jamalpoor A, Hall JC, Fisher J, Hartvelt S, Hendriks G, Nong A. Animal-free assessment of developmental toxicity: Combining PBPK modeling with the ReproTracker assay. Toxicology 2023; 500:153684. [PMID: 38029956 PMCID: PMC10842933 DOI: 10.1016/j.tox.2023.153684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
in vitro screening platforms to assess teratogenic potential of compounds are emerging rapidly. ReproTracker is a human induced pluripotent stem cells (hiPSCs)-based biomarker assay that is shown to identify the teratogenicity potential of new pharmaceuticals and chemicals reliably. In its current state, the assay is limited to identifying the potential teratogenic effects and does not immediately quantify a clinical dose relevant to the exposure of chemicals or drugs observable in mothers or fetuses. The goal of this study was to evaluate whether the ReproTracker assay can be extrapolated in vivo and quantitatively predict developmental toxicity exposure levels of two known human teratogens, thalidomide, and carbamazepine. Here, we utilized Physiologically Based Pharmacokinetic (PBPK) modeling to describe the pharmacokinetic behavior of these compounds and conducted an in vitro to in vivo extrapolation (IVIVE) approach to predict human equivalent effect doses (HEDs) that correspond with in vitro concentrations potentially associated with adverse outcomes in ReproTracker. The HEDs derived from the ReproTracker concentration predicted to cause developmental toxicity were close to the reported teratogenic human clinical doses and the HED derived from the rat or rabbit developmental toxicity study. The ReproTracker derived-HED revealed to be sensitive and protective of humans. Overall, this pilot study demonstrated the importance of integrating PBPK model in extrapolating and assessing developmental toxicity in vitro. The combination of these tools demonstrated that they could improve the safety assessment of drugs and chemicals without animal testing.
Collapse
Affiliation(s)
- Marjory Moreau
- ScitoVation, LLC, Research Triangle Park, NC 27713, USA.
| | - Amer Jamalpoor
- Toxys, Leiden Bioscience Park, Oegstgeest, the Netherlands
| | | | | | | | - Giel Hendriks
- Toxys, Leiden Bioscience Park, Oegstgeest, the Netherlands
| | - Andy Nong
- ScitoVation, LLC, Research Triangle Park, NC 27713, USA
| |
Collapse
|
11
|
Chen M, Du R, Zhang T, Li C, Bao W, Xin F, Hou S, Yang Q, Chen L, Wang Q, Zhu A. The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment. TOXICS 2023; 11:874. [PMID: 37888724 PMCID: PMC10611306 DOI: 10.3390/toxics11100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Toxicokinetics plays a crucial role in the health risk assessments of xenobiotics. Classical compartmental models are limited in their ability to determine chemical concentrations in specific organs or tissues, particularly target organs or tissues, and their limited interspecific and exposure route extrapolation hinders satisfactory health risk assessment. In contrast, physiologically based toxicokinetic (PBTK) models quantitatively describe the absorption, distribution, metabolism, and excretion of chemicals across various exposure routes and doses in organisms, establishing correlations with toxic effects. Consequently, PBTK models serve as potent tools for extrapolation and provide a theoretical foundation for health risk assessment and management. This review outlines the construction and application of PBTK models in health risk assessment while analyzing their limitations and future perspectives.
Collapse
Affiliation(s)
- Mengting Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Ruihu Du
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Tao Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Chutao Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Wenqiang Bao
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Fan Xin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qiaomei Yang
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Li Chen
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, China
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| |
Collapse
|
12
|
Demeester C, Robins D, Edwina AE, Tournoy J, Augustijns P, Ince I, Lehmann A, Vertzoni M, Schlender JF. Physiologically based pharmacokinetic (PBPK) modelling of oral drug absorption in older adults - an AGePOP review. Eur J Pharm Sci 2023; 188:106496. [PMID: 37329924 DOI: 10.1016/j.ejps.2023.106496] [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: 03/28/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
The older population consisting of persons aged 65 years or older is the fastest-growing population group and also the major consumer of pharmaceutical products. Due to the heterogenous ageing process, this age group shows high interindividual variability in the dose-exposure-response relationship and, thus, a prediction of drug safety and efficacy is challenging. Although physiologically based pharmacokinetic (PBPK) modelling is a well-established tool to inform and confirm drug dosing strategies during drug development for special population groups, age-related changes in absorption are poorly accounted for in current PBPK models. The purpose of this review is to summarise the current state-of-knowledge in terms of physiological changes with increasing age that can influence the oral absorption of dosage forms. The capacity of common PBPK platforms to incorporate these changes and describe the older population is also discussed, as well as the implications of extrinsic factors such as drug-drug interactions associated with polypharmacy on the model development process. The future potential of this field will rely on addressing the gaps identified in this article, which can subsequently supplement in-vitro and in-vivo data for more robust decision-making on the adequacy of the formulation for use in older adults and inform pharmacotherapy.
Collapse
Affiliation(s)
- Cleo Demeester
- Systems Pharmacology & Medicine, Pharmaceuticals, Bayer AG, Leverkusen 51373, Germany; Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Gasthuisberg O&N II, Leuven, Belgium
| | - Donnia Robins
- Global CMC Development, Merck KGaA, Frankfurter Straße 250, Darmstadt, Germany; Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Angela Elma Edwina
- Gerontology and Geriatrics Unit, Department of Public Health and Primary care, KU Leuven - University of Leuven, Leuven, Belgium
| | - Jos Tournoy
- Gerontology and Geriatrics Unit, Department of Public Health and Primary care, KU Leuven - University of Leuven, Leuven, Belgium; Department of Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Gasthuisberg O&N II, Leuven, Belgium
| | - Ibrahim Ince
- Systems Pharmacology & Medicine, Pharmaceuticals, Bayer AG, Leverkusen 51373, Germany
| | - Andreas Lehmann
- Global CMC Development, Merck KGaA, Frankfurter Straße 250, Darmstadt, Germany
| | - Maria Vertzoni
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | | |
Collapse
|
13
|
Viel A, Nouichi A, Le Van Suu M, Rolland JG, Sanders P, Laurentie M, Manceau J, Henri J. PBPK Model To Predict Marbofloxacin Distribution in Edible Tissues and Intestinal Exposure in Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4358-4370. [PMID: 36877630 DOI: 10.1021/acs.jafc.2c06561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Marbofloxacin (MAR) is a fluoroquinolone antibiotic used in food-producing animals in European Union, especially in pigs. In this study, MAR concentrations in plasma, comestible tissues, and intestinal segments were determined in pigs injected with MAR. Based on these data and the literature, a flow-limited PBPK model was developed to predict the tissue distribution of MAR and estimate the withdrawal period after label-use in Europe. A submodel describing the different segments of the intestinal lumen was also developed to assess the intestinal exposure of MAR for the commensal bacteria. During model calibration, only four parameters were estimated. Then, Monte Carlo simulations were performed to generate a virtual population of pigs. The simulation results were compared with the observations from an independent data set during the validation step. A global sensitivity analysis was also carried out to identify the most influential parameters. Overall, the PBPK model was able to adequately predict the MAR kinetics in plasma and edible tissues, as well as in small intestines. However, the simulated concentrations in the large intestine were mostly underestimated, highlighting the need for improvements in the field of PBPK modeling to assess the intestinal exposure of antimicrobials in food animals.
Collapse
Affiliation(s)
- Alexis Viel
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Anis Nouichi
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Mélanie Le Van Suu
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Jean-Guy Rolland
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Pascal Sanders
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Michel Laurentie
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Jacqueline Manceau
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| | - Jérôme Henri
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 10B rue Claude Bourgelat, Fougères 35306, France
| |
Collapse
|
14
|
Hügl B, Horlitz M, Fischer K, Kreutz R. Clinical significance of the rivaroxaban-dronedarone interaction: insights from physiologically based pharmacokinetic modelling. EUROPEAN HEART JOURNAL OPEN 2023; 3:oead004. [PMID: 36820238 PMCID: PMC9938521 DOI: 10.1093/ehjopen/oead004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
Patients with atrial fibrillation may require rhythm control therapy in addition to anticoagulation therapy for the prevention of stroke. Since 2012, the European Society of Cardiology and European Heart Rhythm Association guidelines have recommended non-vitamin K antagonist oral anticoagulants, including rivaroxaban, for the prevention of stroke in patients with atrial fibrillation. During the same period, these guidelines have also recommended dronedarone or amiodarone as second-line rhythm control agents in certain patients with atrial fibrillation and no contraindications. Amiodarone and dronedarone both strongly inhibit P-glycoprotein, while dronedarone is a moderate and amiodarone a weak inhibitor of cytochrome P450 3A4 (CYP3A4). Based on these data and evidence from physiologically based pharmacokinetic modelling, amiodarone and dronedarone are expected to have similar effects on rivaroxaban exposure resulting from P-glycoprotein and CYP3A4 inhibition. However, the rivaroxaban label recommends against the concomitant use of dronedarone, but not amiodarone, citing a lack of evidence on the concomitant use of rivaroxaban and dronedarone as the reason for the different recommendations. In this report, we discuss evidence from clinical studies and physiologically based pharmacokinetic modelling on the potential for increased rivaroxaban exposure resulting from drug-drug interaction between rivaroxaban and dronedarone or amiodarone. The current evidence supports the same clinical status and concomitant use of either amiodarone or dronedarone with rivaroxaban, which could be considered in future recommendations.
Collapse
Affiliation(s)
- Burkhard Hügl
- Clinic for Cardiology and Rhythmology, Marienhaus Klinikum St Elisabeth Neuwied, Neuwied, Germany
| | - Marc Horlitz
- Klinik für Kardiologie, Elektrophysiologie und Rhythmologie, Krankenhaus Porz am Rhein, Universität Witten/Herdecke, Köln, Germany
| | - Kerstin Fischer
- Bayer AG, Research & Development, Pharmaceuticals Therapeutic Opportunity Expansion, Berlin, Germany
| | - Reinhold Kreutz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Clinical Pharmacology and Toxicology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
15
|
McNally K, Sams C, Loizou G. Development, testing, parameterisation, and calibration of a human PBK model for the plasticiser, di (2-ethylhexyl) adipate (DEHA) using in silico, in vitro and human biomonitoring data. Front Pharmacol 2023; 14:1165770. [PMID: 37033641 PMCID: PMC10076754 DOI: 10.3389/fphar.2023.1165770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: A physiologically based biokinetic model for di (2-ethylhexyl) adipate (DEHA) based on a refined model for di-(2-propylheptyl) phthalate (DPHP) was developed to interpret the metabolism and biokinetics of DEHA following a single oral dosage of 50 mg to two male and two female volunteers. Methods: The model was parameterized using in vitro and in silico methods such as, measured intrinsic hepatic clearance scaled from in vitro to in vivo and algorithmically predicted parameters such as plasma unbound fraction and tissue:blood partition coefficients (PCs). Calibration of the DEHA model was achieved using concentrations of specific downstream metabolites of DEHA excreted in urine. The total fractions of ingested DEHA eliminated as specific metabolites were estimated and were sufficient for interpreting the human biomonitoring data. Results: The specific metabolites of DEHA, mono-2-ethyl-5-hydroxyhexyl adipate (5OH-MEHA), mono-2-ethyl-5-oxohexyl adipate (5oxo-MEHA), mono-5-carboxy-2-ethylpentyl adipate (5cx-MEPA) only accounted for ∼0.45% of the ingested DEHA. Importantly, the measurements of adipic acid, a non-specific metabolite of DEHA, proved to be important in model calibration. Discussion: The very prominent trends in the urinary excretion of the metabolites, 5cx-MEPA and 5OH-MEHA allowed the important absorption mechanisms of DEHA to be modelled. The model should be useful for the study of exposure to DEHA of the general human population.
Collapse
|
16
|
McNally K, Sams C, Hogg A, Loizou G. Development, testing, parameterisation, and calibration of a human PBPK model for the plasticiser, di-(2-ethylhexyl) terephthalate (DEHTP) using in silico, in vitro and human biomonitoring data. Front Pharmacol 2023; 14:1140852. [PMID: 36891271 PMCID: PMC9986446 DOI: 10.3389/fphar.2023.1140852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
A physiologically based pharmacokinetic model for di-(2-ethylhexyl) terephthalate (DEHTP) based on a refined model for di-(2-propylheptyl) phthalate (DPHP) was developed to interpret the metabolism and biokinetics of DEHTP following a single oral dose of 50 mg to three male volunteers. In vitro and in silico methods were used to generate parameters for the model. For example, measured intrinsic hepatic clearance scaled from in vitro to in vivo and plasma unbound fraction and tissue:blood partition coefficients (PCs) were predicted algorithmically. Whereas the development and calibration of the DPHP model was based upon two data streams, blood concentrations of parent chemical and first metabolite and the urinary excretion of metabolites, the model for DEHTP was calibrated against a single data stream, the urinary excretion of metabolites. Despite the model form and structure being identical significant quantitative differences in lymphatic uptake between the models were observed. In contrast to DPHP the fraction of ingested DEHTP entering lymphatic circulation was much greater and of a similar magnitude to that entering the liver with evidence for the dual uptake mechanisms discernible in the urinary excretion data. Further, the absolute amounts absorbed by the study participants, were much higher for DEHTP relative to DPHP. The in silico algorithm for predicting protein binding performed poorly with an error of more than two orders of magnitude. The extent of plasma protein binding has important implications for the persistence of parent chemical in venous blood-inferences on the behaviour of this class of highly lipophilic chemicals, based on calculations of chemical properties, should be made with extreme caution. Attempting read across for this class of highly lipophilic chemicals should be undertaken with caution since basic adjustments to PCs and metabolism parameters would be insufficient, even when the structure of the model itself is appropriate. Therefore, validation of a model parameterized entirely with in vitro and in silico derived parameters would need to be calibrated against several human biomonitoring data streams to constitute a data rich source chemical to afford confidence for future evaluations of other similar chemicals using the read-across approach.
Collapse
|
17
|
Sawyer CW, Tuey SM, West RE, Nolin TD, Joy MS. Physiologically Based Pharmacokinetic Modeling of Vitamin D 3 and Metabolites in Vitamin D-Insufficient Patients. Drug Metab Dispos 2022; 50:1161-1169. [PMID: 35779863 PMCID: PMC9450961 DOI: 10.1124/dmd.121.000609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
A physiologically based pharmacokinetic (PBPK) model of vitamin D3 and metabolites [25(OH)D3, 1,25(OH)2D3, and 24,25(OH)2D3] is presented. In this study, patients with 25(OH)D3 plasma concentrations below 30 ng/ml were studied after a single dose of 5000 I.U. (125 µg) cholecalciferol, provided with 5000 I.U. daily cholecalciferol supplementation until vitamin D replete [25(OH)D3 plasma concentrations above 30 ng/ml], and had serial plasma samples were collected at each phase for 14 days. Total concentrations of vitamin D3 and metabolites were measured by ultra-high performance liquid chromatography tandem mass spectrometry. A nine-compartment PBPK model was built using MATLAB to represent the triphasic study nature (insufficient, replenishing, and sufficient). The stimulatory and inhibitory effect of 1,25(OH)2D3 were incorporated by fold-changes in the primary metabolic enzymes CYP27B1 and CYP24A1, respectively. Incorporation of dynamic adipose partition coefficients for vitamin D3 and 25(OH)D3 and variable enzymatic reactions aided in model fitting. Measures of model predictions agreed well with data from metabolites, with 97%, 88%, and 98% of the data for 25(OH)D3, 24,25(OH)2D3, and 1,25(OH)2D3, respectively, within twofold of unity (fold error values between 0.5 and 2.0). Bootstrapping was performed and optimized parameters were reported with 95% confidence intervals. This PBPK model could be a useful tool for understanding the connections between vitamin D and its metabolites under a variety of clinical situations. SIGNIFICANCE STATEMENT: This study developed a physiologically based pharmacokinetic (PBPK) model of vitamin D3 and metabolites for patients moving from an insufficient to a repleted state over a period of 16 weeks.
Collapse
Affiliation(s)
- Colton W Sawyer
- Department of Mathematics, Southern New Hampshire University, Manchester, New Hampshire (C.W.S.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (S.M.T., M.S.J.); and University of Pittsburgh, School of Pharmacy, Department of Pharmacy and Therapeutics, Pittsburgh Pennsylvania (R.E.W., T.D.N.)
| | - Stacey M Tuey
- Department of Mathematics, Southern New Hampshire University, Manchester, New Hampshire (C.W.S.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (S.M.T., M.S.J.); and University of Pittsburgh, School of Pharmacy, Department of Pharmacy and Therapeutics, Pittsburgh Pennsylvania (R.E.W., T.D.N.)
| | - Raymond E West
- Department of Mathematics, Southern New Hampshire University, Manchester, New Hampshire (C.W.S.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (S.M.T., M.S.J.); and University of Pittsburgh, School of Pharmacy, Department of Pharmacy and Therapeutics, Pittsburgh Pennsylvania (R.E.W., T.D.N.)
| | - Thomas D Nolin
- Department of Mathematics, Southern New Hampshire University, Manchester, New Hampshire (C.W.S.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (S.M.T., M.S.J.); and University of Pittsburgh, School of Pharmacy, Department of Pharmacy and Therapeutics, Pittsburgh Pennsylvania (R.E.W., T.D.N.)
| | - Melanie S Joy
- Department of Mathematics, Southern New Hampshire University, Manchester, New Hampshire (C.W.S.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (S.M.T., M.S.J.); and University of Pittsburgh, School of Pharmacy, Department of Pharmacy and Therapeutics, Pittsburgh Pennsylvania (R.E.W., T.D.N.)
| |
Collapse
|
18
|
A web-based interactive physiologically based pharmacokinetic (iPBPK) model for meloxicam in broiler chickens and laying hens. Food Chem Toxicol 2022; 168:113332. [DOI: 10.1016/j.fct.2022.113332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
|
19
|
Chou WC, Tell LA, Baynes RE, Davis JL, Maunsell FP, Riviere JE, Lin Z. An Interactive Generic Physiologically Based Pharmacokinetic (igPBPK) Modeling Platform to Predict Drug Withdrawal Intervals in Cattle and Swine: A Case Study on Flunixin, Florfenicol and Penicillin G. Toxicol Sci 2022; 188:180-197. [PMID: 35642931 PMCID: PMC9333411 DOI: 10.1093/toxsci/kfac056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Violative chemical residues in edible tissues from food-producing animals are of global public health concern. Great efforts have been made to develop physiologically based pharmacokinetic (PBPK) models for estimating withdrawal intervals (WDIs) for extralabel prescribed drugs in food animals. Existing models are insufficient to address the food safety concern as these models are either limited to 1 specific drug or difficult to be used by non-modelers. This study aimed to develop a user-friendly generic PBPK platform that can predict tissue residues and estimate WDIs for multiple drugs including flunixin, florfenicol, and penicillin G in cattle and swine. Mechanism-based in silico methods were used to predict tissue/plasma partition coefficients and the models were calibrated and evaluated with pharmacokinetic data from Food Animal Residue Avoidance Databank (FARAD). Results showed that model predictions were, in general, within a 2-fold factor of experimental data for all 3 drugs in both species. Following extralabel administration and respective U.S. FDA-approved tolerances, predicted WDIs for both cattle and swine were close to or slightly longer than FDA-approved label withdrawal times (eg, predicted 8, 28, and 7 days vs labeled 4, 28, and 4 days for flunixin, florfenicol, and penicillin G in cattle, respectively). The final model was converted to a web-based interactive generic PBPK platform. This PBPK platform serves as a user-friendly quantitative tool for real-time predictions of WDIs for flunixin, florfenicol, and penicillin G following FDA-approved label or extralabel use in both cattle and swine, and provides a basis for extrapolating to other drugs and species.
Collapse
Affiliation(s)
- Wei-Chun Chou
- 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
| | - Lisa A Tell
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, 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, USA
| | - Jennifer L Davis
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24060, USA
| | - Fiona P Maunsell
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Jim E Riviere
- Center for Chemical Toxicology Research and Pharmacokinetics, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA.,1Data Consortium,Kansas State University, Olathe, KS, 66061, USA
| | - Zhoumeng Lin
- 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
|
20
|
Le A, Wearing HJ, Li D. Streamlining physiologically‐based pharmacokinetic model design for intravenous delivery of nanoparticle drugs. CPT Pharmacometrics Syst Pharmacol 2022; 11:409-424. [PMID: 35045205 PMCID: PMC9007599 DOI: 10.1002/psp4.12762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/19/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Physiologically‐based pharmacokinetic (PBPK) modeling for nanoparticles elucidates the nanoparticle drug’s disposition in the body and serves a vital role in drug development and clinical studies. This paper offers a systematic and tutorial‐like approach to developing a model structure and writing distribution ordinary differential equations based on asking binary questions involving the physicochemical nature of the drug in question. Further, by synthesizing existing knowledge, we summarize pertinent aspects in PBPK modeling and create a guide for building model structure and distribution equations, optimizing nanoparticle and non‐nanoparticle specific parameters, and performing sensitivity analysis and model validation. The purpose of this paper is to facilitate a streamlined model development process for students and practitioners in the field.
Collapse
Affiliation(s)
- Anh‐Dung Le
- Nanoscience & Microsystems Engineering University of New Mexico Albuquerque New Mexico USA
| | - Helen J. Wearing
- Department of Biology Department of Mathematics & Statistics University of New Mexico Albuquerque New Mexico USA
| | - Dingsheng Li
- School of Community Health Sciences University of Nevada Reno Nevada USA
| |
Collapse
|
21
|
Loizou G, McNally K, Paini A, Hogg A. Derivation of a Human In Vivo Benchmark Dose for Bisphenol A from ToxCast In Vitro Concentration Response Data Using a Computational Workflow for Probabilistic Quantitative In Vitro to In Vivo Extrapolation. Front Pharmacol 2022; 12:754408. [PMID: 35222005 PMCID: PMC8874249 DOI: 10.3389/fphar.2021.754408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022] Open
Abstract
A computational workflow which integrates physiologically based kinetic (PBK) modelling; global sensitivity analysis (GSA), Approximate Bayesian Computation (ABC), Markov Chain Monte Carlo (MCMC) simulation and the Virtual Cell Based Assay (VCBA) for the estimation of the active, free in vitro concentration of chemical in the reaction medium was developed to facilitate quantitative in vitro to in vivo extrapolation (QIVIVE). The workflow was designed to estimate parameter and model uncertainty within a computationally efficient framework. The workflow was tested using a human PBK model for bisphenol A (BPA) and high throughput screening (HTS) in vitro concentration-response data, for estrogen and pregnane X receptor activation determined in human liver and kidney cell lines, from the ToxCast/Tox21 database. In vivo benchmark dose 10% lower confidence limits (BMDL10) for oral uptake of BPA (ng/kg BW/day) were calculated from the in vivo dose-responses and compared to the human equivalent dose (HED) BMDL10 for relative kidney weight change in the mouse derived by European Food Safety Authority (EFSA). Three from four in vivo BMDL10 values calculated in this study were similar to the EFSA values whereas the fourth was much smaller. The derivation of an uncertainty factor (UF) to accommodate the uncertainties associated with measurements using human cell lines in vitro, extrapolated to in vivo, could be useful for the derivation of Health Based Guidance Values (HBGV).
Collapse
Affiliation(s)
- George Loizou
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| | - Kevin McNally
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| | - Alicia Paini
- European Commission Joint Research Centre, Ispra, Italy
| | - Alex Hogg
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| |
Collapse
|
22
|
Mast MP, Modh H, Champanhac C, Wang JW, Storm G, Krämer J, Mailänder V, Pastorin G, Wacker MG. Nanomedicine at the crossroads - A quick guide for IVIVC. Adv Drug Deliv Rev 2021; 179:113829. [PMID: 34174332 DOI: 10.1016/j.addr.2021.113829] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
For many years, nanomedicine is pushing the boundaries of drug delivery. When applying these novel therapeutics, safety considerations are not only a key concern when entering clinical trials but also an important decision point in product development. Standing at the crossroads, nanomedicine may be able to escape the niche markets and achieve wider acceptance by the pharmaceutical industry. While there is a new generation of drug delivery systems, the extracellular vesicles, standing on the starting line, unresolved issues and new challenges emerge from their translation from bench to bedside. Some key features of injectable nanomedicines contribute to the predictability of the pharmacological and toxicological effects. So far, only a few of the physicochemical attributes of nanomedicines can be justified by a direct mathematical relationship between the in vitro and the in vivo responses. To further develop extracellular vesicles as drug carriers, we have to learn from more than 40 years of clinical experience in liposomal delivery and pass on this knowledge to the next generation. Our quick guide discusses relationships between physicochemical characteristics and the in vivo response, commonly referred to as in vitro-in vivo correlation. Further, we highlight the key role of computational methods, lay open current knowledge gaps, and question the established design strategies. Has the recent progress improved the predictability of targeted delivery or do we need another change in perspective?
Collapse
|
23
|
A Bayesian population physiologically based pharmacokinetic absorption modeling approach to support generic drug development: application to bupropion hydrochloride oral dosage forms. J Pharmacokinet Pharmacodyn 2021; 48:893-908. [PMID: 34553275 DOI: 10.1007/s10928-021-09778-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/22/2021] [Indexed: 12/13/2022]
Abstract
We propose a Bayesian population modeling and virtual bioequivalence assessment approach to establishing dissolution specifications for oral dosage forms. A generalizable semi-physiologically based pharmacokinetic absorption model with six gut segments and liver, connected to a two-compartment model of systemic disposition for bupropion hydrochloride oral dosage forms was developed. Prior information on model parameters for gut physiology, bupropion physicochemical properties, and drug product properties were obtained from the literature. The release of bupropion hydrochloride from immediate-, sustained- and extended-release oral dosage forms was described by a Weibull function. In vitro dissolution data were used to assign priors to the in vivo release properties of the three bupropion formulations. We applied global sensitivity analysis to identify the influential parameters for plasma bupropion concentrations and calibrated them. To quantify inter- and intra-individual variability, plasma concentration profiles in healthy volunteers that received the three dosage forms, each at two doses, were used. The calibrated model was in good agreement with both in vitro dissolution and in vivo exposure data. Markov Chain Monte Carlo samples from the joint posterior parameter distribution were used to simulate virtual crossover clinical trials for each formulation with distinct drug dissolution profiles. For each trial, an allowable range of dissolution parameters ("safe space") in which bioequivalence can be anticipated was established. These findings can be used to assure consistent product performance throughout the drug product life-cycle and to support manufacturing changes. Our framework provides a comprehensive approach to support decision-making in drug product development.
Collapse
|
24
|
McNally K, Sams C, Hogg A, Lumen A, Loizou G. Development, Testing, Parameterisation and Calibration of a Human PBPK Model for the Plasticiser, Di-(2-propylheptyl) Phthalate (DPHP) Using in Silico, in vitro and Human Biomonitoring Data. Front Pharmacol 2021; 12:692442. [PMID: 34539393 PMCID: PMC8443793 DOI: 10.3389/fphar.2021.692442] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
A physiologically based pharmacokinetic model for Di-(2-propylheptyl) phthalate (DPHP) was developed to interpret the biokinetics in humans after single oral doses. The model was parameterized with in vitro and in silico derived parameters and uncertainty and sensitivity analysis was used during the model development process to assess structure, biological plausibility and behaviour prior to simulation and analysis of human biological monitoring data. To provide possible explanations for some of the counter-intuitive behaviour of the biological monitoring data the model included a simple lymphatic uptake process for DPHP and enterohepatic recirculation (EHR) for DPHP and the mono ester metabolite mono-(2-propylheptyl) phthalate (MPHP). The model was used to simultaneously simulate the concentration-time profiles of blood DPHP, MPHP and the urinary excretion of two metabolites, mono-(2-propyl-6-hydroxyheptyl) phthalate (OH-MPHP) and mono-(2-propyl-6-carboxyhexyl) phthalate (cx-MPHP). The availability of blood and urine measurements permitted a more robust qualitative and quantitative investigation of the importance of EHR and lymphatic uptake. Satisfactory prediction of blood DPHP and urinary metabolites was obtained whereas blood MPHP was less satisfactory. However, the delayed peak of DPHP concentration relative to MPHP in blood and second order metabolites in urine could be explained as a result of three processes: 1) DPHP entering the systemic circulation from the lymph, 2) rapid and very high protein binding and 3) the efficiency of the liver in removing DPHP absorbed via the hepatic route. The use of sensitivity analysis is considered important in the evaluation of uncertainty around in vitro and in silico derived parameters. By quantifying their impact on model output sufficient confidence in the use of a model should be afforded. This approach could expand the use of PBPK models since parameterization with in silico techniques allows for rapid model development. This in turn could assist in reducing the use of animals in toxicological evaluations by enhancing the utility of “read across” techniques.
Collapse
Affiliation(s)
| | - Craig Sams
- Health and Safety Executive, Buxton, United Kingdom
| | - Alex Hogg
- Health and Safety Executive, Buxton, United Kingdom
| | - Annie Lumen
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | | |
Collapse
|
25
|
Dalaijamts C, Cichocki JA, Luo YS, Rusyn I, Chiu WA. Quantitative Characterization of Population-Wide Tissue- and Metabolite-Specific Variability in Perchloroethylene Toxicokinetics in Male Mice. Toxicol Sci 2021; 182:168-182. [PMID: 33988684 DOI: 10.1093/toxsci/kfab057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Quantification of interindividual variability is a continuing challenge in risk assessment, particularly for compounds with complex metabolism and multi-organ toxicity. Toxicokinetic variability for perchloroethylene (perc) was previously characterized across 3 mouse strains and in 1 mouse strain with various degrees of liver steatosis. To further characterize the role of genetic variability in toxicokinetics of perc, we applied Bayesian population physiologically based pharmacokinetic (PBPK) modeling to the data on perc and metabolites in blood/plasma and tissues of male mice from 45 inbred strains from the Collaborative Cross (CC) mouse population. After identifying the most influential PBPK parameters based on global sensitivity analysis, we fit the model with a hierarchical Bayesian population analysis using Markov chain Monte Carlo simulation. We found that the data from 3 commonly used strains were not representative of the full range of variability in perc and metabolite blood/plasma and tissue concentrations across the CC population. Using interstrain variability as a surrogate for human interindividual variability, we calculated dose-dependent, chemical-, and tissue-specific toxicokinetic variability factors (TKVFs) as candidate science-based replacements for the default uncertainty factor for human toxicokinetic variability of 100.5. We found that toxicokinetic variability factors for glutathione conjugation metabolites of perc showed the greatest variability, often exceeding the default, whereas those for oxidative metabolites and perc itself were generally less than the default. Overall, we demonstrate how a combination of a population-based mouse model such as the CC with Bayesian population PBPK modeling can reduce uncertainty in human toxicokinetic variability and increase accuracy and precision in quantitative risk assessment.
Collapse
Affiliation(s)
- Chimeddulam Dalaijamts
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843-4458, USA.,Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4458, USA
| | - Joseph A Cichocki
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843-4458, USA.,Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4458, USA
| | - Yu-Syuan Luo
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843-4458, USA.,Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4458, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843-4458, USA.,Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4458, USA
| | - Weihsueh A Chiu
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843-4458, USA.,Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4458, USA
| |
Collapse
|
26
|
Predicted limited redistribution of T cells to secondary lymphoid tissue correlates with increased risk of haematological malignancies in asplenic patients. Sci Rep 2021; 11:16394. [PMID: 34385480 PMCID: PMC8360980 DOI: 10.1038/s41598-021-95225-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022] Open
Abstract
The spleen, a secondary lymphoid tissue (SLT), has an important role in generation of adaptive immune responses. Although splenectomy remains a common procedure, recent studies reported poor prognosis and increased risk of haematological malignancies in asplenic patients. The high baseline trafficking of T lymphocytes to splenic tissue suggests splenectomy may lead to loss of blood-borne malignant immunosurveillance that is not compensated for by the remaining SLT. To date, no quantitative analysis of the impact of splenectomy on the human T cell trafficking dynamics and tissue localisation has been reported. We developed a quantitative computational model that describes organ distribution and trafficking of human lymphocytes to explore the likely impact of splenectomy on immune cell distributions. In silico splenectomy resulted in an average reduction of T cell numbers in SLT by 35% (95%CI 0.12–0.97) and a comparatively lower, 9% (95%CI 0.17–1.43), mean decrease of T cell concentration in SLT. These results suggest that the surveillance capacity of the remaining SLT insufficiently compensates for the absence of the spleen. This may, in part, explain haematological malignancy risk in asplenic patients and raises the question of whether splenectomy has a clinically meaningful impact on patient responses to immunotherapy.
Collapse
|
27
|
Melillo N, Darwich AS. A latent variable approach to account for correlated inputs in global sensitivity analysis. J Pharmacokinet Pharmacodyn 2021; 48:671-686. [PMID: 34032996 PMCID: PMC8405496 DOI: 10.1007/s10928-021-09764-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
In drug development decision-making is often supported through model-based methods, such as physiologically-based pharmacokinetics (PBPK). Global sensitivity analysis (GSA) is gaining use for quality assessment of model-informed inference. However, the inclusion and interpretation of correlated factors in GSA has proven an issue. Here we developed and evaluated a latent variable approach for dealing with correlated factors in GSA. An approach was developed that describes the correlation between two model inputs through the causal relationship of three independent factors: the latent variable and the unique variances of the two correlated parameters. The latent variable approach was applied to a set of algebraic models and a case from PBPK. Then, this method was compared to Sobol’s GSA assuming no correlations, Sobol’s GSA with groups and the Kucherenko approach. For the latent variable approach, GSA was performed with Sobol’s method. By using the latent variable approach, it is possible to devise a unique and easy interpretation of the sensitivity indices while maintaining the correlation between the factors. Compared methods either consider the parameters independent, group the dependent variables into one unique factor or present difficulties in the interpretation of the sensitivity indices. In situations where GSA is called upon to support model-informed decision-making, the latent variable approach offers a practical method, in terms of ease of implementation and interpretability, for applying GSA to models with correlated inputs that does not violate the independence assumption. Prerequisites and limitations of the approach are discussed.
Collapse
Affiliation(s)
- Nicola Melillo
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy & Optometry, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Adam S Darwich
- Division of Health Informatics and Logistics, Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
28
|
Loizou G, McNally K, Dorne JLCM, Hogg A. Derivation of a Human In Vivo Benchmark Dose for Perfluorooctanoic Acid From ToxCast In Vitro Concentration-Response Data Using a Computational Workflow for Probabilistic Quantitative In Vitro to In Vivo Extrapolation. Front Pharmacol 2021; 12:630457. [PMID: 34045957 PMCID: PMC8144460 DOI: 10.3389/fphar.2021.630457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/01/2021] [Indexed: 01/11/2023] Open
Abstract
A computational workflow which integrates physiologically based kinetic (PBK) modeling, global sensitivity analysis (GSA), approximate Bayesian computation (ABC), and Markov Chain Monte Carlo (MCMC) simulation was developed to facilitate quantitative in vitro to in vivo extrapolation (QIVIVE). The workflow accounts for parameter and model uncertainty within a computationally efficient framework. The workflow was tested using a human PBK model for perfluorooctanoic acid (PFOA) and high throughput screening (HTS) in vitro concentration–response data, determined in a human liver cell line, from the ToxCast/Tox21 database. In vivo benchmark doses (BMDs) for PFOA intake (ng/kg BW/day) and drinking water exposure concentrations (µg/L) were calculated from the in vivo dose responses and compared to intake values derived by the European Food Safety Authority (EFSA). The intake benchmark dose lower confidence limit (BMDL5) of 0.82 was similar to 0.86 ng/kg BW/day for altered serum cholesterol levels derived by EFSA, whereas the intake BMDL5 of 6.88 was six-fold higher than the value of 1.14 ng/kg BW/day for altered antibody titer also derived by the EFSA. Application of a chemical-specific adjustment factor (CSAF) of 1.4, allowing for inter-individual variability in kinetics, based on biological half-life, gave an intake BMDL5 of 0.59 for serum cholesterol and 4.91 (ng/kg BW/day), for decreased antibody titer, which were 0.69 and 4.31 the EFSA-derived values, respectively. The corresponding BMDL5 for drinking water concentrations, for estrogen receptor binding activation associated with breast cancer, pregnane X receptor binding associated with altered serum cholesterol levels, thyroid hormone receptor α binding leading to thyroid disease, and decreased antibody titer (pro-inflammation from cytokines) were 0.883, 0.139, 0.086, and 0.295 ng/ml, respectively, with application of no uncertainty factors. These concentrations are 5.7-, 36-, 58.5-, and 16.9-fold lower than the median measured drinking water level for the general US population which is approximately, 5 ng/ml.
Collapse
Affiliation(s)
- George Loizou
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| | - Kevin McNally
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| | - Jean-Lou C M Dorne
- Scientific Committee and Emerging Risks Unit, European Food Safety Authority, Parma, Italy
| | - Alex Hogg
- Health and Safety Executive, Harpur Hill, Buxton, United Kingdom
| |
Collapse
|
29
|
Willmann S, Coboeken K, Kapsa S, Thelen K, Mundhenke M, Fischer K, Hügl B, Mück W. Applications of Physiologically Based Pharmacokinetic Modeling of Rivaroxaban-Renal and Hepatic Impairment and Drug-Drug Interaction Potential. J Clin Pharmacol 2021; 61:656-665. [PMID: 33205449 PMCID: PMC8048900 DOI: 10.1002/jcph.1784] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
The non–vitamin K antagonist oral anticoagulant rivaroxaban is used in several thromboembolic disorders. Rivaroxaban is eliminated via both metabolic degradation and renal elimination as unchanged drug. Therefore, renal and hepatic impairment may reduce rivaroxaban clearance, and medications inhibiting these clearance pathways could lead to drug‐drug interactions. This physiologically based pharmacokinetic (PBPK) study investigated the pharmacokinetic behavior of rivaroxaban in clinical situations where drug clearance is impaired. A PBPK model was developed using mass balance and bioavailability data from adults and qualified using clinically observed data. Renal and hepatic impairment were simulated by adjusting disease‐specific parameters, and concomitant drug use was simulated by varying enzyme activity in virtual populations (n = 1000) and compared with pharmacokinetic predictions in virtual healthy populations and clinical observations. Rivaroxaban doses of 10 mg or 20 mg were used. Mild to moderate renal impairment had a minor effect on area under the concentration‐time curve and maximum plasma concentration of rivaroxaban, whereas severe renal impairment caused a more pronounced increase in these parameters vs normal renal function. Area under the concentration‐time curve and maximum plasma concentration increased with severity of hepatic impairment. These effects were smaller in the simulations compared with clinical observations. AUC and Cmax increased with the strength of cytochrome P450 3A4 and P‐glycoprotein inhibitors in simulations and clinical observations. This PBPK model can be useful for estimating the effects of impaired drug clearance on rivaroxaban pharmacokinetics. Identifying other factors that affect the pharmacokinetics of rivaroxaban could facilitate the development of models that approximate real‐world pharmacokinetics more accurately.
Collapse
Affiliation(s)
| | | | - Stefanie Kapsa
- Clinical Pharmacokinetics Cardiovascular, Bayer AG, Wuppertal, Germany
| | - Kirstin Thelen
- Clinical Pharmacokinetics Cardiovascular, Bayer AG, Wuppertal, Germany
| | - Markus Mundhenke
- Medical Affairs Cardiovascular, Bayer Vital GmbH, Leverkusen, Germany
| | | | - Burkhard Hügl
- Clinic for Cardiology and Rhythmology, Marienhaus Klinikum St Elisabeth Neuwied, Neuwied, Germany
| | - Wolfgang Mück
- Clinical Pharmacokinetics Cardiovascular, Bayer AG, Wuppertal, Germany
| |
Collapse
|
30
|
Hardiansyah D, Kletting P, Begum NJ, Eiber M, Beer AJ, Pawiro SA, Glatting G. Important pharmacokinetic parameters for individualization of 177 Lu-PSMA therapy: A global sensitivity analysis for a physiologically-based pharmacokinetic model. Med Phys 2020; 48:556-568. [PMID: 33244792 DOI: 10.1002/mp.14622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/26/2020] [Accepted: 11/13/2020] [Indexed: 11/06/2022] Open
Abstract
PURPOSE The knowledge of the contribution of anatomical and physiological parameters to interindividual pharmacokinetic differences could potentially be used to improve individualized treatment planning for radionuclide therapy. The aim of this study was therefore to identify the physiologically based pharmacokinetic (PBPK) model parameters that determine the interindividual variability of absorbed doses (ADs) to kidneys and tumor lesions in therapy with 177 Lu-labeled PSMA-targeting radioligands. METHODS A global sensitivity analysis (GSA) with the extended Fourier Amplitude Sensitivity Test (eFAST) algorithm was performed. The whole-body PBPK model for PSMA-targeting radioligand therapy from our previous studies was used in this study. The model parameters of interest (input of the GSA) were the organ receptor densities [R0 ], the organ blood flows f, and the organ release rates λ. These parameters were systematically sampled NE times according to their distribution in the patient population. The corresponding pharmacokinetics were simulated and the ADs (model output) to kidneys and tumor lesions were collected. The main effect S i and total effect S Ti were calculated using the eFAST algorithm based on the variability of the model output: The main effect S i of input parameter i represents the reduction in variance of the output if the "true" value of parameter i would be known. The total effect S Ti of an input parameter i represents the proportion of variance remaining if the "true" values of all other input parameters except for i are known. The numbers of samples NE were increased up to 8193 to check the stability (i.e., convergence) of the calculated main effects S i and total effects S Ti . RESULTS From the simulations, the relative interindividual variability of ADs in the kidneys (coefficient of variation CV = 31%) was lower than that of ADs in the tumors (CV up to 59%). Based on the GSA, the most important parameters that determine the ADs to the kidneys were kidneys flow ( S i = 0.36, S Ti = 0.43) and kidneys receptor density ( S i = 0.25, S Ti = 0.30). Tumor receptor density was identified as the most important parameter determining the ADs to tumors ( S i and S Ti up to 0.72). CONCLUSIONS The results suggest that an accurate measurement of receptor density and flow before therapy could be a promising approach for developing an individualized treatment with 177 Lu-labeled PSMA-targeting radioligands.
Collapse
Affiliation(s)
- Deni Hardiansyah
- Medical Physics and Biophysics, Physics Department, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16424, Indonesia
| | - Peter Kletting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany
| | - Nusrat J Begum
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, München, 81675, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany
| | - Supriyanto A Pawiro
- Medical Physics and Biophysics, Physics Department, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16424, Indonesia
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, 89081, Germany
| |
Collapse
|
31
|
Toutain PL, Pelligand L, Lees P, Bousquet-Mélou A, Ferran AA, Turnidge JD. The pharmacokinetic/pharmacodynamic paradigm for antimicrobial drugs in veterinary medicine: Recent advances and critical appraisal. J Vet Pharmacol Ther 2020; 44:172-200. [PMID: 33089523 DOI: 10.1111/jvp.12917] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/16/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Pharmacokinetic/pharmacodynamic (PK/PD) modelling is the initial step in the semi-mechanistic approach for optimizing dosage regimens for systemically acting antimicrobial drugs (AMDs). Numerical values of PK/PD indices are used to predict dose and dosing interval on a rational basis followed by confirmation in clinical trials. The value of PK/PD indices lies in their universal applicability amongst animal species. Two PK/PD indices are routinely used in veterinary medicine, the ratio of the area under the curve of the free drug plasma concentration to the minimum inhibitory concentration (MIC) (fAUC/MIC) and the time that free plasma concentration exceeds the MIC over the dosing interval (fT > MIC). The basic concepts of PK/PD modelling of AMDs were established some 20 years ago. Earlier studies have been reviewed previously and are not reconsidered in this review. This review describes and provides a critical appraisal of more recent, advanced PK/PD approaches, with particular reference to their application in veterinary medicine. Also discussed are some hypotheses and new areas for future developments.First, a brief overview of PK/PD principles is presented as the basis for then reviewing more advanced mechanistic considerations on the precise nature of selected indices. Then, several new approaches to selecting PK/PD indices and establishing their numerical values are reviewed, including (a) the modelling of time-kill curves and (b) the use of population PK investigations. PK/PD indices can be used for dose determination, and they are required to establish clinical breakpoints for antimicrobial susceptibility testing. A particular consideration is given to the precise nature of MIC, because it is pivotal in establishing PK/PD indices, explaining that it is not a "pharmacodynamic parameter" in the usual sense of this term.
Collapse
Affiliation(s)
- Pierre-Louis Toutain
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France.,Royal Veterinary College, University of London, London, UK
| | | | - Peter Lees
- Royal Veterinary College, University of London, London, UK
| | | | - Aude A Ferran
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France
| | - John D Turnidge
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
32
|
Kenyon EM, Eklund C, Pegram RA, Lipscomb JC. Comparison of in vivo derived and scaled in vitro metabolic rate constants for several volatile organic compounds (VOCs). Toxicol In Vitro 2020; 69:105002. [PMID: 32946980 DOI: 10.1016/j.tiv.2020.105002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 10/23/2022]
Abstract
Metabolic rate parameters estimation using in vitro data is necessary due to numbers of chemicals for which data are needed, trend towards minimizing laboratory animal use, and limited opportunity to collect data in human subjects. We evaluated how well metabolic rate parameters derived from in vitro data predict overall in vivo metabolism for a set of environmental chemicals for which well validated and established methods exist. We compared values of VmaxC derived from in vivo vapor uptake studies with estimates of VmaxC scaled up from in vitro hepatic microsomal metabolism studies for VOCs for which data were available in male F344 rats. For 6 of 7 VOCs, differences between the in vivo and scaled up in vitro VmaxC estimates were less than 2.6-fold. For bromodichloromethane (BDCM), the in vivo derived VmaxC was approximately 4.4-fold higher than the in vitro derived and scaled up VmaxC. The more rapid rate of BDCM metabolism estimated based in vivo studies suggests other factors such as extrahepatic metabolism, binding or other non-specific losses making a significant contribution to overall clearance. Systematic and reliable utilization of scaled up in vitro biotransformation rate parameters in PBPK models will require development of methods to predict cases in which extrahepatic metabolism and binding as well as other factors are likely to be significant contributors.
Collapse
Affiliation(s)
- Elaina M Kenyon
- Center for Computational Toxicology and Exposure, U.S. EPA, Office of Research and Development, Research Triangle Park, NC, United States.
| | - Christopher Eklund
- Center for Computational Toxicology and Exposure, U.S. EPA, Office of Research and Development, Research Triangle Park, NC, United States
| | - Rex A Pegram
- Center for Computational Toxicology and Exposure, U.S. EPA, Office of Research and Development, Research Triangle Park, NC, United States
| | | |
Collapse
|
33
|
Melillo N, Grandoni S, Cesari N, Brogin G, Puccini P, Magni P. Inter-compound and Intra-compound Global Sensitivity Analysis of a Physiological Model for Pulmonary Absorption of Inhaled Compounds. AAPS J 2020; 22:116. [PMID: 32862303 PMCID: PMC7456635 DOI: 10.1208/s12248-020-00499-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/06/2020] [Indexed: 12/25/2022] Open
Abstract
In recent years, global sensitivity analysis (GSA) has gained interest in physiologically based pharmacokinetics (PBPK) modelling and simulation from pharmaceutical industry, regulatory authorities, and academia. With the case study of an in-house PBPK model for inhaled compounds in rats, the aim of this work is to show how GSA can contribute in PBPK model development and daily use. We identified two types of GSA that differ in the aims and, thus, in the parameter variability: inter-compound and intra-compound GSA. The inter-compound GSA aims to understand which are the parameters that mostly influence the variability of the metrics of interest in the whole space of the drugs' properties, and thus, it is useful during the model development. On the other hand, the intra-compound GSA aims to highlight how much the uncertainty associated with the parameters of a given drug impacts the uncertainty in the model prediction and so, it is useful during routine PBPK use. In this work, inter-compound GSA highlighted that dissolution- and formulation-related parameters were mostly important for the prediction of the fraction absorbed, while the permeability is the most important parameter for lung AUC and MRT. Intra-compound GSA highlighted that, for all the considered compounds, the permeability was one of the most important parameters for lung AUC, MRT and plasma MRT, while the extraction ratio and the dose for the plasma AUC. GSA is a crucial instrument for the quality assessment of model-based inference; for this reason, we suggest its use during both PBPK model development and use.
Collapse
Affiliation(s)
- Nicola Melillo
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering, Università degli Studi di Pavia, Via Ferrata 5, I-27100, Pavia, Italy
| | - Silvia Grandoni
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering, Università degli Studi di Pavia, Via Ferrata 5, I-27100, Pavia, Italy
| | - Nicola Cesari
- Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Giandomenico Brogin
- Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Paola Puccini
- Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Paolo Magni
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering, Università degli Studi di Pavia, Via Ferrata 5, I-27100, Pavia, Italy.
| |
Collapse
|
34
|
Liu D, Li L, Rostami-Hodjegan A, Bois FY, Jamei M. Considerations and Caveats when Applying Global Sensitivity Analysis Methods to Physiologically Based Pharmacokinetic Models. AAPS JOURNAL 2020; 22:93. [PMID: 32681207 PMCID: PMC7367914 DOI: 10.1208/s12248-020-00480-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023]
Abstract
Three global sensitivity analysis (GSA) methods (Morris, Sobol and extended Sobol) are applied to a minimal physiologically based PK (mPBPK) model using three model drugs given orally, namely quinidine, alprazolam, and midazolam. We investigated how correlations among input parameters affect the determination of the key parameters influencing pharmacokinetic (PK) properties of general interest, i.e., the maximal plasma concentration (Cmax) time at which Cmax is reached (Tmax), and area under plasma concentration (AUC). The influential parameters determined by the Morris and Sobol methods (suitable for independent model parameters) were compared to those determined by the extended Sobol method (which considers model parameter correlations). For the three drugs investigated, the Morris method was as informative as the Sobol method. The extended Sobol method identified different sets of influential parameters to Morris and Sobol. These methods overestimated the influence of volume of distribution at steady state (Vss) on AUC24h for quinidine and alprazolam. They also underestimated the effect of volume of liver (Vliver) for all three drugs, the impact of enzyme intrinsic clearance of CYP2C9 and CYP2E1 for quinidine, and that of UGT1A4 abundance for midazolam. Our investigation showed that the interpretation of GSA results is not straightforward. Dismissing existing model parameter correlations, GSA methods such as Morris and Sobol can lead to biased determination of the key parameters for the selected outputs of interest. Decisions regarding parameters’ influence (or otherwise) should be made in light of available knowledge including the model assumptions, GSA method limitations, and inter-correlations between model parameters, particularly in complex models. Graphical abstract ![]()
Collapse
Affiliation(s)
- Dan Liu
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | - Linzhong Li
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Amin Rostami-Hodjegan
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Frederic Y Bois
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Masoud Jamei
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| |
Collapse
|
35
|
Hsieh NH, Reisfeld B, Chiu WA. pksensi: An R package to apply global sensitivity analysis in physiologically based kinetic modeling. SOFTWAREX 2020; 12:100609. [PMID: 33426260 PMCID: PMC7790364 DOI: 10.1016/j.softx.2020.100609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Sensitivity analysis (SA) is an essential tool for modelers to understand the influence of model parameters on model outputs. It is also increasingly used in developing and assessing physiologically based kinetic (PBK) models. For instance, several studies have applied global SA to reduce the computational burden in the Bayesian Markov chain Monte Carlo-based calibration process PBK models. Although several SA algorithms and software packages are available, no comprehensive software package exists that allows users to seamlessly solve differential equations in a PBK model, conduct and visualize SA results, and discriminate between the non-influential model parameters that can be fixed and those that need calibration. Therefore, we developed an R package, named pksensi, to make global SA more accessible in PBK modeling. This package can investigate both uncertainty and sensitivity in PBK models, including those with multivariate model outputs. It also includes functions to check the convergence of the global SA results. Overall, pksensi improves the user experience of performing global SA and can create robust and reproducible results for decision making in PBK model calibration.
Collapse
Affiliation(s)
- Nan-Hung Hsieh
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Brad Reisfeld
- Chemical and Biological Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Weihsueh A. Chiu
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
36
|
Utembe W, Clewell H, Sanabria N, Doganis P, Gulumian M. Current Approaches and Techniques in Physiologically Based Pharmacokinetic (PBPK) Modelling of Nanomaterials. NANOMATERIALS 2020; 10:nano10071267. [PMID: 32610468 PMCID: PMC7407857 DOI: 10.3390/nano10071267] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/03/2020] [Accepted: 06/13/2020] [Indexed: 02/08/2023]
Abstract
There have been efforts to develop physiologically based pharmacokinetic (PBPK) models for nanomaterials (NMs). Since NMs have quite different kinetic behaviors, the applicability of the approaches and techniques that are utilized in current PBPK models for NMs is warranted. Most PBPK models simulate a size-independent endocytosis from tissues or blood. In the lungs, dosimetry and the air-liquid interface (ALI) models have sometimes been used to estimate NM deposition and translocation into the circulatory system. In the gastrointestinal (GI) tract, kinetics data are needed for mechanistic understanding of NM behavior as well as their absorption through GI mucus and their subsequent hepatobiliary excretion into feces. Following absorption, permeability (Pt) and partition coefficients (PCs) are needed to simulate partitioning from the circulatory system into various organs. Furthermore, mechanistic modelling of organ- and species-specific NM corona formation is in its infancy. More recently, some PBPK models have included the mononuclear phagocyte system (MPS). Most notably, dissolution, a key elimination process for NMs, is only empirically added in some PBPK models. Nevertheless, despite the many challenges still present, there have been great advances in the development and application of PBPK models for hazard assessment and risk assessment of NMs.
Collapse
Affiliation(s)
- Wells Utembe
- National Institute for Occupational Health, P.O. Box 4788, Johannesburg 2000, South Africa; (W.U.); (N.S.)
| | - Harvey Clewell
- Ramboll US Corporation, Research Triangle Park, NC 27709, USA;
| | - Natasha Sanabria
- National Institute for Occupational Health, P.O. Box 4788, Johannesburg 2000, South Africa; (W.U.); (N.S.)
| | - Philip Doganis
- School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15780 Athens, Greece;
| | - Mary Gulumian
- National Institute for Occupational Health, P.O. Box 4788, Johannesburg 2000, South Africa; (W.U.); (N.S.)
- Hematology and Molecular Medicine, University of the Witwatersrand, Johannesburg 2000, South Africa
- Correspondence: ; Tel.: +27-11-712-6428
| |
Collapse
|
37
|
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: 6.0] [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
|
38
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
39
|
Sayre RR, Wambaugh JF, Grulke CM. Database of pharmacokinetic time-series data and parameters for 144 environmental chemicals. Sci Data 2020; 7:122. [PMID: 32313097 PMCID: PMC7170868 DOI: 10.1038/s41597-020-0455-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/12/2020] [Indexed: 12/20/2022] Open
Abstract
Time courses of compound concentrations in plasma are used in chemical safety analysis to evaluate the relationship between external administered doses and internal tissue exposures. This type of experimental data is rarely available for the thousands of non-pharmaceutical chemicals to which people may potentially be unknowingly exposed but is necessary to properly assess the risk of such exposures. In vitro assays and in silico models are often used to craft an understanding of a chemical's pharmacokinetics; however, the certainty of the quantitative application of these estimates for chemical safety evaluations cannot be determined without in vivo data for external validation. To address this need, we present a public database of chemical time-series concentration data from 567 studies in humans or test animals for 144 environmentally-relevant chemicals and their metabolites (187 analytes total). All major administration routes are incorporated, with concentrations measured in blood/plasma, tissues, and excreta. We also include calculated pharmacokinetic parameters for some studies, and a bibliography of additional source documents to support future extraction of time-series. In addition to pharmacokinetic model calibration and validation, these data may be used for analyses of differential chemical distribution across chemicals, species, doses, or routes, and for meta-analyses on pharmacokinetic studies.
Collapse
Affiliation(s)
- Risa R Sayre
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA.
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - John F Wambaugh
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA
| | - Christopher M Grulke
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
40
|
Díaz de León-Ortega R, D'Arcy DM, Lamprou DA, Fotaki N. In vitro - in vivo relations for the parenteral liposomal formulation of Amphotericin B: A clinically relevant approach with PBPK modeling. Eur J Pharm Biopharm 2020; 159:177-187. [PMID: 32147578 DOI: 10.1016/j.ejpb.2020.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/22/2020] [Accepted: 03/03/2020] [Indexed: 12/26/2022]
Abstract
In vitro release testing is a useful tool for the quality control of controlled release parenteral formulations, but in vitro release test conditions that reflect or are able to predict the in vivo performance are advantageous. Therefore, it is important to investigate the factors that could affect drug release from formulations and relate them to in vivo performance. In this study the effect of media composition including albumin presence, type of buffer and hydrodynamics on drug release were evaluated on a liposomal Amphotericin B formulation (Ambisome®). A physiologically based pharmacokinetic (PBPK) model was developed using plasma concentration profiles from healthy subjects, in order to investigate the impact of each variable from the in vitro release tests on the prediction of the in vivo performance. It was found that albumin presence was the most important factor for the release of Amphotericin B from Ambisome®; both hydrodynamics setups, coupled with the PBPK model, had comparable predictive ability for simulating in vivo plasma concentration profiles. The PBPK model was extrapolated to a hypothetical hypoalbuminaemic population and the Amphotericin B plasma concentration and its activity against fungal cells were simulated. Selected in vitro release tests for these controlled release parenteral formulations were able to predict the in vivo AmB exposure, and this PBPK driven approach to release test development could benefit development of such formulations.
Collapse
Affiliation(s)
| | - D M D'Arcy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - D A Lamprou
- School of Pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - N Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom.
| |
Collapse
|
41
|
Lautz LS, Nebbia C, Hoeks S, Oldenkamp R, Hendriks AJ, Ragas AMJ, Dorne JLCM. An open source physiologically based kinetic model for the chicken (Gallus gallus domesticus): Calibration and validation for the prediction residues in tissues and eggs. ENVIRONMENT INTERNATIONAL 2020; 136:105488. [PMID: 31991240 DOI: 10.1016/j.envint.2020.105488] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 06/10/2023]
Abstract
Xenobiotics from anthropogenic and natural origin enter animal feed and human food as regulated compounds, environmental contaminants or as part of components of the diet. After dietary exposure, a chemical is absorbed and distributed systematically to a range of organs and tissues, metabolised, and excreted. Physiologically based kinetic (PBK) models have been developed to estimate internal concentrations from external doses. In this study, a generic multi-compartment PBK model was developed for chicken. The PBK model was implemented for seven compounds (with log Kow range -1.37-6.2) to quantitatively link external dose and internal dose for risk assessment of chemicals. Global sensitivity analysis was performed for a hydrophilic and a lipophilic compound to identify the most sensitive parameters in the PBK model. Model predictions were compared to measured data according to dataset-specific exposure scenarios. Globally, 71% of the model predictions were within a 3-fold change of the measured data for chicken and only 7% of the PBK predictions were outside a 10-fold change. While most model input parameters still rely on in vivo experiments, in vitro data were also used as model input to predict internal concentration of the coccidiostat monensin. Future developments of generic PBK models in chicken and other species of relevance to animal health risk assessment are discussed.
Collapse
Affiliation(s)
- L S Lautz
- Department of Environmental Science, Radboud University Nijmegen, Houtlaan 4, 6525 XZ Nijmegen, the Netherlands.
| | - C Nebbia
- Department of Veterinary Sciences, University of Torino, Largo P. Braccini 2, 10095 Grugliasco, Italy
| | - S Hoeks
- Department of Environmental Science, Radboud University Nijmegen, Houtlaan 4, 6525 XZ Nijmegen, the Netherlands
| | - R Oldenkamp
- Department of Environmental Science, Radboud University Nijmegen, Houtlaan 4, 6525 XZ Nijmegen, the Netherlands
| | - A J Hendriks
- Department of Environmental Science, Radboud University Nijmegen, Houtlaan 4, 6525 XZ Nijmegen, the Netherlands
| | - A M J Ragas
- Department of Environmental Science, Radboud University Nijmegen, Houtlaan 4, 6525 XZ Nijmegen, the Netherlands; Department of Science, Faculty of Management, Science &Technology, Open University, 6419 AT Heerlen, the Netherlands
| | - J L C M Dorne
- European Food Safety Authority, Via Carlo Magno 1A, 43126 Parma, Italy
| |
Collapse
|
42
|
Lautz L, Dorne J, Oldenkamp R, Hendriks A, Ragas A. Generic physiologically based kinetic modelling for farm animals: Part I. Data collection of physiological parameters in swine, cattle and sheep. Toxicol Lett 2020; 319:95-101. [DOI: 10.1016/j.toxlet.2019.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/09/2019] [Accepted: 10/22/2019] [Indexed: 11/30/2022]
|
43
|
McNally K, Sams C, Loizou G. Development, Testing, Parameterization, and Calibration of a Human Physiologically Based Pharmacokinetic Model for the Plasticizer, Hexamoll ® Diisononyl-Cyclohexane-1, 2-Dicarboxylate Using In Silico, In Vitro, and Human Biomonitoring Data. Front Pharmacol 2019; 10:1394. [PMID: 31849656 PMCID: PMC6897292 DOI: 10.3389/fphar.2019.01394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/31/2019] [Indexed: 11/13/2022] Open
Abstract
A physiologically based pharmacokinetic model for Hexamoll® diisononyl-cyclohexane-1, 2-dicarboxylate was developed to interpret the biokinetics in humans after single oral doses. The model was parameterized with in vitro and in silico derived parameters and uncertainty and sensitivity analysis was used during the model development process to assess structure, biological plausibility and behavior prior to simulation and analysis of human biological monitoring data. The model provided good simulations of the urinary excretion (Curine) of two metabolites; cyclohexane-1,2-dicarboxylic acid mono hydroxyisononyl ester (OH-MINCH) and cyclohexane-1, 2-dicarboxylic acid mono carboxyisononyl ester (cx-MINCH) from the biotransformation of mono-isononyl-cyclohexane-1, 2-dicarboxylate (MINCH), the monoester metabolite of di-isononyl-cyclohexane-1,2-dicarboxylate. However, good simulations could be obtained, with and without, a lymphatic compartment. Selection of an appropriate model structure was informed by sensitivity analysis which could identify and quantify the contribution to variability in Curine by parameters, such as, the fraction of oral dose that directly entered the lymphatic compartment and therefore by-passed the liver and the fraction of MINCH bio-transformed to cx-MINCH and OH-MINCH. By constraining these parameters within biologically plausible limits the presence of a lymphatic compartment was deemed an important component of model structure. Furthermore, the use of sensitivity analysis is important in the evaluation of uncertainty around in silico derived parameters. By quantifying their impact on model output sufficient confidence in the use of a model should be afforded. This type of approach could expand the use of physiologically based pharmacokinetic models since parameterization with in silico techniques allows for rapid model development. This in turn could assist in reducing the use of animals in toxicological evaluations by enhancing the utility of “read across” techniques.
Collapse
Affiliation(s)
- Kevin McNally
- Exposure and Health Consequences, Health and Safety Executive, Buxton, United Kingdom
| | - Craig Sams
- Exposure and Health Consequences, Health and Safety Executive, Buxton, United Kingdom
| | - George Loizou
- Exposure and Health Consequences, Health and Safety Executive, Buxton, United Kingdom
| |
Collapse
|
44
|
Peters SA, Dolgos H. Requirements to Establishing Confidence in Physiologically Based Pharmacokinetic (PBPK) Models and Overcoming Some of the Challenges to Meeting Them. Clin Pharmacokinet 2019; 58:1355-1371. [PMID: 31236775 PMCID: PMC6856026 DOI: 10.1007/s40262-019-00790-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
When scientifically well-founded, the mechanistic basis of physiologically based pharmacokinetic (PBPK) models can help reduce the uncertainty and increase confidence in extrapolations outside the studied scenarios or studied populations. However, it is not always possible to establish mechanistically credible PBPK models. Requirements to establishing confidence in PBPK models, and challenges to meeting these requirements, are presented in this article. Parameter non-identifiability is the most challenging among the barriers to establishing confidence in PBPK models. Using case examples of small molecule drugs, this article examines the use of hypothesis testing to overcome parameter non-identifiability issues, with the objective of enhancing confidence in the mechanistic basis of PBPK models and thereby improving the quality of predictions that are meant for internal decisions and regulatory submissions. When the mechanistic basis of a PBPK model cannot be established, we propose the use of simpler models or evidence-based approaches.
Collapse
Affiliation(s)
| | - Hugues Dolgos
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| |
Collapse
|
45
|
Lautz L, Oldenkamp R, Dorne J, Ragas A. Physiologically based kinetic models for farm animals: Critical review of published models and future perspectives for their use in chemical risk assessment. Toxicol In Vitro 2019; 60:61-70. [DOI: 10.1016/j.tiv.2019.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/28/2019] [Accepted: 05/05/2019] [Indexed: 10/26/2022]
|
46
|
Joshi K, Tiwari B, Cullen PJ, Frias JM. Predicting quality attributes of strawberry packed under modified atmosphere throughout the cold chain. Food Packag Shelf Life 2019. [DOI: 10.1016/j.fpsl.2019.100354] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
47
|
Mavroudis PD, Ayyar VS, Jusko WJ. ATLAS mPBPK: A MATLAB-Based Tool for Modeling and Simulation of Minimal Physiologically-Based Pharmacokinetic Models. CPT Pharmacometrics Syst Pharmacol 2019; 8:557-566. [PMID: 31154668 PMCID: PMC6709424 DOI: 10.1002/psp4.12441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/06/2019] [Indexed: 01/24/2023] Open
Abstract
Minimal physiologically-based pharmacokinetic (mPBPK) models are frequently used to model plasma pharmacokinetic (PK) data and utilize and yield physiologically relevant parameters. Compared with classical compartment and whole-body physiologically-based pharmacokinetic modeling approaches, mPBPK models maintain a structure of intermediate physiological complexity that can be adequately informed by plasma PK data. In this tutorial, we present a MATLAB-based tool for the modeling and simulation of mPBPK models (ATLAS mPBPK) of small and large molecules. This tool enables the users to perform the following: (i) PK data visualization, (ii) simulation, (iii) parameter optimization, and (iv) local sensitivity analysis of mPBPK models in a simple and efficient manner. In addition to the theoretical background and implementation of the different tool functionalities, this tutorial includes simulation and sensitivity analysis showcases of small and large molecules with and without target-mediated drug disposition.
Collapse
Affiliation(s)
| | - Vivaswath S. Ayyar
- School of Pharmacy and Pharmaceutical SciencesUniversity at BuffaloBuffaloNew YorkUSA
| | - William J. Jusko
- School of Pharmacy and Pharmaceutical SciencesUniversity at BuffaloBuffaloNew YorkUSA
| |
Collapse
|
48
|
The importance of mathematical modelling in chemical risk assessment and the associated quantification of uncertainty. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.comtox.2018.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
49
|
Dong Z, Naidu R. Response to comment on: Dong et al. (2017) "issues raised by the reference doses for perfluorooctonate sulfonate and perfluorooctanoic acid". ENVIRONMENT INTERNATIONAL 2019; 126:802-803. [PMID: 30718018 DOI: 10.1016/j.envint.2019.01.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 05/20/2023]
Affiliation(s)
- Zhaomin Dong
- School of Space and Environment, Beihang University, China.
| | - Ravi Naidu
- Global Centre for Environmental Remediation, The Faculty of Science and Information Technology, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia.
| |
Collapse
|
50
|
Integration of Food Animal Residue Avoidance Databank (FARAD) empirical methods for drug withdrawal interval determination with a mechanistic population-based interactive physiologically based pharmacokinetic (iPBPK) modeling platform: example for flunixin meglumine administration. Arch Toxicol 2019; 93:1865-1880. [PMID: 31025081 DOI: 10.1007/s00204-019-02464-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/18/2019] [Indexed: 12/31/2022]
Abstract
Violative chemical residues in animal-derived food products affect food safety globally and have impact on the trade of international agricultural products. The Food Animal Residue Avoidance Databank program has been developing scientific tools to provide appropriate withdrawal interval (WDI) estimations after extralabel drug use in food animals for the past three decades. One of the tools is physiologically based pharmacokinetic (PBPK) modeling, which is a mechanistic-based approach that can be used to predict tissue residues and WDIs. However, PBPK models are complicated and difficult to use by non-modelers. Therefore, a user-friendly PBPK modeling framework is needed to move this field forward. Flunixin was one of the top five violative drug residues identified in the United States from 2010 to 2016. The objective of this study was to establish a web-based user-friendly framework for the development of new PBPK models for drugs administered to food animals. Specifically, a new PBPK model for both cattle and swine after administration of flunixin meglumine was developed. Population analysis using Monte Carlo simulations was incorporated into the model to predict WDIs following extralabel administration of flunixin meglumine. The population PBPK model was converted to a web-based interactive PBPK (iPBPK) framework to facilitate its application. This iPBPK framework serves as a proof-of-concept for further improvements in the future and it can be applied to develop new models for other drugs in other food animal species, thereby facilitating the application of PBPK modeling in WDI estimation and food safety assessment.
Collapse
|