The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment
Abstract
:1. Introduction
2. The Construction of the PBTK Model
2.1. Model Characterization
2.1.1. Determination of the Model Structure
2.1.2. Building Mathematical Equations
- (1)
- Perfusion-limited model: in this model, xenobiotic concentrations in organs, tissues, and blood reach instant equilibrium without concentration differences. Organs, tissues, and blood are considered as a single compartment with a homogeneous distribution of xenobiotics. The only limiting factor for xenobiotic distribution is blood flow velocity [12,13]. This model is suitable for small lipid-soluble molecules that can readily cross membrane barriers and when organs or tissues are small-sized with high blood flow [7,14,15];
- (2)
- Permeability-limited model: in this model, xenobiotics penetrate organs or tissues through membrane barriers, resulting in a concentration gradient between organs or tissues and blood. Depending on the number of membrane barriers, the permeability-limited model can have two or three sub-compartments. This model is suitable for molecules with polarity or large molecular weight [14,15];
- (3)
- Dispersion model: in this model, xenobiotics are distributed in organs or tissues with a gradient, and the degree of dispersion is evaluated using the dispersion coefficient (DN). A higher DN indicates a greater dispersion of xenobiotics in organs or tissues. When DN approaches infinity, the dispersion model is similar to the perfusion-limited model [16,17,18,19,20,21]. This model is suitable for xenobiotics with high hepatic clearance [8].
2.2. Definition of Model Parameters
2.3. Model Simulation
2.3.1. Algorithm
2.3.2. Software
2.4. Model Evaluation
2.4.1. Verification of Measured Data
2.4.2. Uncertainty Analysis
2.4.3. Variation Analysis
2.4.4. Sensitivity Analysis
2.4.5. Model Optimization
3. Applications of Physiologically Based Toxicokinetics (PBTK) in Health Risk Assessment
3.1. Exposure Assessment
3.2. Extrapolation
3.2.1. IVIVE
3.2.2. Dose Extrapolation
3.2.3. Exposure Route Extrapolation
3.2.4. Interspecific Extrapolation
3.3. PBTK of the Special Population
3.4. Metabolic Characteristics and Mechanisms
3.5. Mixture Risk Assessment
4. Prospects
- (1)
- The PBTK model is based on the understanding of the in vivo ADME processes of chemicals. However, it is challenging to construct a corresponding PBTK model for certain drugs, such as Class 3 and 4 drugs in the Biopharmaceutics Classification System (BCS), traditional Chinese medicine ingredients, heavy metals, etc., due to limited knowledge about their ADME processes.
- (2)
- The parameters required by the PBTK model are typically obtained through in vivo, in vitro, and in silico assays. However, uncertainties may exist regarding parameters measured using in vitro and in silico assays that require validation with further in vivo experimental data. Moreover, special population groups, such as different races, children, pregnant women, obese individuals, and those who are ill, exhibit distinct physiological and ADME characteristics. Therefore, constructing PBTK models for these special groups necessitates extensive experimental research support.
- (3)
- Validating exposure characteristics among different subjects under various exposure conditions is crucial for establishing a reliable PBTK model. Unfortunately, in toxicological studies, the lack of relevant experimental data poses challenges for verifying the accuracy of PBTK models.
- (4)
- Constructing PBTK models can be relatively complex as it requires researchers to possess fundamental knowledge of toxicokinetics, toxicology, physiology, mathematics, and modeling. This complexity limits accessibility to these models.
- (5)
- For mixtures composed of different types of chemicals, it becomes difficult to construct mixed PBTK models due to variations observed during their respective ADME processes within an organism and also because interaction processes between them can become intricate.
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Dixit, R.; Riviere, J.; Krishnan, K.; Andersen, M.E. Toxicokinetics and physiologically based toxicokinetics in toxicology and risk assessment. J. Toxicol. Environ. Health B Crit. Rev. 2003, 6, 1–40. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Li, Z. Status and prospects in drug toxicokinetics. Chin. J. New Drugs 2017, 26, 742–748. [Google Scholar]
- Lipscomb, J.C.; Haddad, S.; Poet, T.; Krishnan, K. Physiologically-based pharmacokinetic (PBPK) models in toxicity testing and risk assessment. Adv. Exp. Med. Biol. 2012, 745, 76–95. [Google Scholar] [PubMed]
- Sarigiannis, D.A.; Karakitsios, S.; Dominguez-Romero, E.; Papadaki, K.; Brochot, C.; Kumar, V.; Schuhmacher, M.; Sy, M.; Mielke, H.; Greiner, M.; et al. Physiology-based toxicokinetic modelling in the frame of the European Human Biomonitoring Initiative. Environ. Res. 2019, 172, 216–230. [Google Scholar] [CrossRef]
- Hu, M.; Zhang, Y.; Zhan, M.; He, G.; Qu, W.; Zhou, Y. Physiologically-based toxicokinetic modeling of human dermal exposure to diethyl phthalate: Application to health risk assessment. Chemosphere 2022, 307, 135931. [Google Scholar] [CrossRef]
- Yao, M.; Jiang, X.; Lu, Y.; Qiao, Y. PBPK Model and Its Application in TCM Studies. Mod. Tradi. Chin. Med. Mater. Medica-World Sci.Tech. 2007, 3, 55–59. [Google Scholar]
- Zhang, S.; Wang, Z.; Chen, J. Application of physiologically based toxicokinetics models in risk assessment of chemicals. Chin. Sci. Bull. 2017, 62, 4139–4150. [Google Scholar] [CrossRef]
- Jin, Y.; Ma, Y. Progress in methodology of establishing physiologically based pharmacokinetic models. Acta Pharm. Sinica 2014, 49, 16–22. [Google Scholar]
- Niu, Z.; Zang, X. Research advances of the physioloaically based pharmacokinetic(PBPK) models and their applications in health risk assessment. J. Health Saf. Environ. 2015, 15, 30–34. [Google Scholar]
- Peters, S.A. Evaluation of a generic physiologically based pharmacokinetic model for lineshape analysis. Clin. Pharmacokinet 2008, 47, 261–275. [Google Scholar] [CrossRef]
- Krishnan, K.; Johanson, G. Physiologically-based pharmacokinetic and toxicokinetic models in cancer risk assessment. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2005, 23, 31–53. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Zhao, L.; Wu, P.; Wang, J. Characteristics and Research Progress of Physiologically Based Pharmacokinetic Model. Chin. J. ETMF 2012, 18, 247–250. [Google Scholar]
- Nestorov, I. Whole body pharmacokinetic models. Clin. Pharmacokinet 2003, 42, 883–908. [Google Scholar] [CrossRef] [PubMed]
- Jones, H.M.; Gardner, I.B.; Watson, K.J. Modelling and PBPK simulation in drug discovery. AAPS J. 2009, 11, 155–166. [Google Scholar] [CrossRef] [PubMed]
- Krishnan, K.; Loizou, G.D.; Spendiff, M.; Lipscomb, J.C.; Andersen, M.E. Quantitative Modeling in Toxicology, 3rd ed.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2010; pp. 24–26. [Google Scholar]
- Barboriak, D.P.; MacFall, J.R.; Viglianti, B.L.; Dewhirst Dvm, M.W. Comparison of three physiologically-based pharmacokinetic models for the prediction of contrast agent distribution measured by dynamic MR imaging. J. Magn. Reson. Imaging 2008, 27, 1388–1398. [Google Scholar] [CrossRef] [PubMed]
- Oliver, R.E.; Jones, A.F.; Rowland, M. A whole-body physiologically based pharmacokinetic model incorporating dispersion concepts: Short and long time characteristics. J. Pharmacokinet. Pharmacodyn. 2001, 28, 27–55. [Google Scholar] [CrossRef]
- Pang, K.S.; Durk, M.R. Physiologically-based pharmacokinetic modeling for absorption, transport, metabolism and excretion. J. Pharmacokinet. Pharmacodyn. 2010, 37, 591–615. [Google Scholar] [CrossRef] [PubMed]
- Roberts, M.S.; Rowland, M. A dispersion model of hepatic elimination: 1. Formulation of the model and bolus considerations. J. Pharmacokinet. Biopharm. 1986, 14, 227–260. [Google Scholar] [CrossRef]
- Roberts, M.S.; Rowland, M. A dispersion model of hepatic elimination: 2. Steady-state considerations--influence of hepatic blood flow, binding within blood, and hepatocellular enzyme activity. J. Pharmacokinet. Biopharm. 1986, 14, 261–288. [Google Scholar] [CrossRef]
- Roberts, M.S.; Rowland, M. A dispersion model of hepatic elimination: 3. Application to metabolite formation and elimination kinetics. J. Pharmacokinet. Biopharm. 1986, 14, 289–308. [Google Scholar] [CrossRef]
- Brown, R.P.; Delp, M.D.; Lindstedt, S.L.; Rhomberg, L.R.; Beliles, R.P. Physiological parameter values for physiologically based pharmacokinetic models. Toxicol. Ind. Health 1997, 13, 407–484. [Google Scholar] [CrossRef] [PubMed]
- Davies, B.; Morris, T. Physiological parameters in laboratory animals and humans. Pharm. Res. 1993, 10, 1093–1095. [Google Scholar] [CrossRef] [PubMed]
- Johns, D.O.; Owens, E.O.; Thompson, C.M.; Sonawane, B.; Hattis, D.; Krishnan, K. Physiological Parameters and Databases for PBPK Modeling. In Quantitative Modeling in Toxicology, 3rd ed.; Krishnan, K., Loizou, G.D., Spendiff, M., Lipscomb, J.C., Andersen, M.E., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2010; pp. 107–134. [Google Scholar]
- Thompson, C.M.; Johns, D.O.; Sonawane, B.; Barton, H.A.; Hattis, D.; Tardif, R.; Krishnan, K. Database for physiologically based pharmacokinetic (PBPK) modeling: Physiological data for healthy and health-impaired elderly. J. Toxicol. Environ. Health B. Crit. Rev. 2009, 12, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Caster, W.O.; Poncelet, J.; Simon, A.B.; Armstrong, W.D. Tissue weights of the rat. I. Normal values determined by dissection and chemical methods. Proc. Soc. Exp. Biol. Med. 1956, 91, 122–126. [Google Scholar] [CrossRef] [PubMed]
- Domenech, R.J.; Hoffman, J.I.; Noble, M.I.; Saunders, K.B.; Henson, J.R.; Subijanto, S. Total and regional coronary blood flow measured by radioactive microspheres in conscious and anesthetized dogs. Circ. Res. 1969, 25, 581–596. [Google Scholar] [CrossRef] [PubMed]
- Mauderly, J.L. Measurement of Respiration and Respiratory Responses During Inhalation Exposures. J. Am. Coll. Toxicol. 1990, 9, 397–405. [Google Scholar] [CrossRef]
- Schoeffner, D.J.; Warren, D.A.; Muralidara, S.; Bruckner, J.V.; Simmons, J.E. Organ weights and fat volume in rats as a function of strain and age. J. Toxicol. Environ. Health A 1999, 56, 449–462. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.S.; Gross, J.F. Estimation of tissue-to-plasma partition coefficients used in physiological pharmacokinetic models. J. Pharmacokinet. Biopharm. 1979, 7, 117–125. [Google Scholar] [CrossRef]
- Dedrick, R.L.; Zaharko, D.S.; Lutz, R.J. Transport and binding of methotrexate in vivo. J. Pharm. Sci. 1973, 62, 882–890. [Google Scholar] [CrossRef]
- Gabrielsson, J.L.; Paalzow, L.K.; Nordström, L. A physiologically based pharmacokinetic model for theophylline disposition in the pregnant and nonpregnant rat. J. Pharmacokinet. Biopharm. 1984, 12, 149–165. [Google Scholar] [CrossRef]
- Gallo, J.M.; Lam, F.C.; Perrier, D.G. Area method for the estimation of partition coefficients for physiological pharmacokinetic models. J. Pharmacokinet. Biopharm. 1987, 15, 271–280. [Google Scholar] [CrossRef] [PubMed]
- Lam, G.; Chen, M.L.; Chiou, W.L. Determination of tissue to blood partition coefficients in physiologically-based pharmacokinetic studies. J. Pharm. Sci. 1982, 71, 454–456. [Google Scholar] [CrossRef] [PubMed]
- Bertelsen, S.L.; Hoffman, A.D.; Gallinat, C.A.; Elonen, C.M.; Nichols, J.W. Evaluation of log KOW and tissue lipid content as predictors of chemical partitioning to fish tissues. Environ. Toxicol. Chem. 1998, 17, 1447–1455. [Google Scholar] [CrossRef]
- Endo, S.; Bauerfeind, J.; Goss, K.U. Partitioning of neutral organic compounds to structural proteins. Environ. Sci. Technol. 2012, 46, 12697–12703. [Google Scholar] [CrossRef] [PubMed]
- Endo, S.; Escher, B.I.; Goss, K.U. Capacities of membrane lipids to accumulate neutral organic chemicals. Environ. Sci. Technol. 2011, 45, 5912–5921. [Google Scholar] [CrossRef] [PubMed]
- Endo, S.; Goss, K.U. Serum albumin binding of structurally diverse neutral organic compounds: Data and models. Chem. Res. Toxicol. 2011, 24, 2293–2301. [Google Scholar] [CrossRef] [PubMed]
- Fiserova-Bergerova, V.; Diaz, M.L. Determination and prediction of tissue-gas partition coefficients. Int. Arch. Occup. Environ. Health 1986, 58, 75–87. [Google Scholar] [CrossRef] [PubMed]
- Gargas, M.L.; Burgess, R.J.; Voisard, D.E.; Cason, G.H.; Andersen, M.E. Partition coefficients of low-molecular-weight volatile chemicals in various liquids and tissues. Toxicol. Appl. Pharmacol. 1989, 98, 87–99. [Google Scholar] [CrossRef]
- Gargas, M.L.; Seybold, P.G.; Andersen, M.E. Modeling the tissue solubilities and metabolic rate constant (Vmax) of halogenated methanes, ethanes, and ethylenes. Toxicol. Lett. 1988, 43, 235–256. [Google Scholar] [CrossRef]
- Geisler, A.; Endo, S.; Goss, K.U. Partitioning of organic chemicals to storage lipids: Elucidating the dependence on fatty acid composition and temperature. Environ. Sci. Technol. 2012, 46, 9519–9524. [Google Scholar] [CrossRef]
- Johanson, G.; Dynésius, B. Liquid/air partition coefficients of six commonly used glycol ethers. Br. J. Ind. Med. 1988, 45, 561–564. [Google Scholar] [CrossRef] [PubMed]
- Sato, A.; Nakajima, T. Partition coefficients of some aromatic hydrocarbons and ketones in water, blood and oil. Br. J. Ind. Med. 1979, 36, 231–234. [Google Scholar] [CrossRef] [PubMed]
- Igari, Y.; Sugiyama, Y.; Sawada, Y.; Iga, T.; Hanano, M. Prediction of diazepam disposition in the rat and man by a physiologically based pharmacokinetic model. J. Pharmacokinet. Biopharm. 1983, 11, 577–593. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.H.; Sugiyama, Y.; Awazu, S.; Hanano, M. In vitro and in vivo evaluation of the tissue-to-blood partition coefficient for physiological pharmacokinetic models. J. Pharmacokinet. Biopharm. 1982, 10, 637–647. [Google Scholar] [CrossRef] [PubMed]
- Sultatos, L.G.; Kim, B.; Woods, L. Evaluation of estimations in vitro of tissue/blood distribution coefficients for organothiophosphate insecticides. Toxicol. Appl. Pharmacol. 1990, 103, 52–55. [Google Scholar] [CrossRef] [PubMed]
- Assmus, F.; Houston, J.B.; Galetin, A. Incorporation of lysosomal sequestration in the mechanistic model for prediction of tissue distribution of basic drugs. Eur. J. Pharm. Sci. 2017, 109, 419–430. [Google Scholar] [CrossRef] [PubMed]
- Gombar, V.K.; Hall, S.D. Quantitative structure-activity relationship models of clinical pharmacokinetics: Clearance and volume of distribution. J. Chem. Inf. Model 2013, 53, 948–957. [Google Scholar] [CrossRef] [PubMed]
- Jansson, R.; Bredberg, U.; Ashton, M. Prediction of drug tissue to plasma concentration ratios using a measured volume of distribution in combination with lipophilicity. J. Pharm. Sci. 2008, 97, 2324–2339. [Google Scholar] [CrossRef]
- Poulin, P. Drug Distribution to Human Tissues: Prediction and Examination of the Basic Assumption in In Vivo Pharmacokinetics-Pharmacodynamics (PK/PD) Research. J. Pharm. Sci. 2015, 104, 2110–2118. [Google Scholar] [CrossRef]
- Poulin, P.; Haddad, S. Advancing prediction of tissue distribution and volume of distribution of highly lipophilic compounds from a simplified tissue-composition-based model as a mechanistic animal alternative method. J. Pharm. Sci. 2012, 101, 2250–2261. [Google Scholar] [CrossRef]
- Poulin, P.; Krishnan, K. An algorithm for predicting tissue: Blood partition coefficients of organic chemicals from n-octanol: Water partition coefficient data. J. Toxicol. Environ. Health 1995, 46, 117–129. [Google Scholar] [CrossRef] [PubMed]
- Poulin, P.; Krishnan, K. Molecular Structure-Based Prediction of the Partition Coefficients of Organic Chemicals for Physiological Pharmacokinetic Models. Toxicol. Mech. Methods 1996, 6, 117–137. [Google Scholar] [CrossRef]
- Poulin, P.; Theil, F.P. Prediction of pharmacokinetics prior to in vivo studies. 1. Mechanism-based prediction of volume of distribution. J. Pharm. Sci. 2002, 91, 129–156. [Google Scholar] [CrossRef] [PubMed]
- Poulin, P.; Theil, F.P. A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically-based pharmacokinetic models in drug discovery. J. Pharm. Sci. 2000, 89, 16–35. [Google Scholar] [CrossRef]
- Rodgers, T.; Leahy, D.; Rowland, M. Physiologically based pharmacokinetic modeling 1: Predicting the tissue distribution of moderate-to-strong bases. J. Pharm. Sci. 2005, 94, 1259–1276. [Google Scholar] [CrossRef] [PubMed]
- Rodgers, T.; Rowland, M. Mechanistic approaches to volume of distribution predictions: Understanding the processes. Pharm. Res. 2007, 24, 918–933. [Google Scholar] [CrossRef] [PubMed]
- DeJongh, J.; Blaauboer, B.J. Simulation of toluene kinetics in the rat by a physiologically based pharmacokinetic model with application of biotransformation parameters derived independently in vitro and in vivo. Fundam. Appl. Toxicol. 1996, 32, 260–268. [Google Scholar] [CrossRef]
- Bauman, J.N.; Kelly, J.M.; Tripathy, S.; Zhao, S.X.; Lam, W.W.; Kalgutkar, A.S.; Obach, R.S. Can in vitro metabolism-dependent covalent binding data distinguish hepatotoxic from nonhepatotoxic drugs? An analysis using human hepatocytes and liver S-9 fraction. Chem. Res. Toxicol. 2009, 22, 332–340. [Google Scholar] [CrossRef]
- Iwatsubo, T.; Suzuki, H.; Sugiyama, Y. Prediction of species differences (rats, dogs, humans) in the in vivo metabolic clearance of YM796 by the liver from in vitro data. J. Pharmacol. Exp. Ther. 1997, 283, 462–469. [Google Scholar]
- Kedderis, G.L.; Held, S.D. Prediction of furan pharmacokinetics from hepatocyte studies: Comparison of bioactivation and hepatic dosimetry in rats, mice, and humans. Toxicol. Appl. Pharmacol. 1996, 140, 124–130. [Google Scholar] [CrossRef]
- Lipscomb, J.C.; Fisher, J.W.; Confer, P.D.; Byczkowski, J.Z. In vitro to in vivo extrapolation for trichloroethylene metabolism in humans. Toxicol. Appl. Pharmacol. 1998, 152, 376–387. [Google Scholar] [CrossRef] [PubMed]
- Mortensen, B.; Nilsen, O.G. Allometric species comparison of toluene and n-hexane metabolism: Prediction of hepatic clearance in man from experiments with rodent liver S9 in a head space vial equilibration system. Pharmacol. Toxicol. 1998, 82, 183–188. [Google Scholar] [CrossRef] [PubMed]
- Obach, R.S.; Kalgutkar, A.S.; Soglia, J.R.; Zhao, S.X. Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from nonhepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose. Chem. Res. Toxicol. 2008, 21, 1814–1822. [Google Scholar] [CrossRef] [PubMed]
- Doddareddy, M.R.; Cho, Y.S.; Koh, H.Y.; Kim, D.H.; Pae, A.N. In silico renal clearance model using classical Volsurf approach. J. Chem. Inf. Model 2006, 46, 1312–1320. [Google Scholar] [CrossRef] [PubMed]
- Manga, N.n.; Duffy, J.C.; Rowe, P.H.; Cronin, M.T.D. A Hierarchical QSAR Model for Urinary Excretion of Drugs in Humans as a Predictive Tool for Biotransformation. Mol. Inform. 2003, 22, 263–273. [Google Scholar] [CrossRef]
- Paine, S.W.; Barton, P.; Bird, J.; Denton, R.; Menochet, K.; Smith, A.; Tomkinson, N.P.; Chohan, K.K. A rapid computational filter for predicting the rate of human renal clearance. J. Mol. Graph. Model. 2010, 29, 529–537. [Google Scholar] [CrossRef] [PubMed]
- Price, K.; Krishnan, K. An integrated QSAR-PBPK modelling approach for predicting the inhalation toxicokinetics of mixtures of volatile organic chemicals in the rat. SAR QSAR Environ. Res. 2011, 22, 107–128. [Google Scholar] [CrossRef] [PubMed]
- Nichols, J.W.; Fitzsimmons, P.N.; Burkhard, L.P. In vitro-in vivo extrapolation of quantitative hepatic biotransformation data for fish. II. Modeled effects on chemical bioaccumulation. Environ. Toxicol. Chem. 2007, 26, 1304–1319. [Google Scholar] [CrossRef]
- Nichols, J.W.; Schultz, I.R.; Fitzsimmons, P.N. In vitro-in vivo extrapolation of quantitative hepatic biotransformation data for fish. I. A review of methods, and strategies for incorporating intrinsic clearance estimates into chemical kinetic models. Aquat. Toxicol. 2006, 78, 74–90. [Google Scholar] [CrossRef]
- Schneckener, S.; Preuss, T.G.; Kuepfer, L.; Witt, J. A workflow to build PBTK models for novel species. Arch. Toxicol. 2020, 94, 3847–3860. [Google Scholar] [CrossRef]
- Haddad, S.; Pelekis, M.; Krishnan, K. A methodology for solving physiologically based pharmacokinetic models without the use of simulation softwares. Toxicol. Lett. 1996, 85, 113–126. [Google Scholar] [CrossRef]
- Johanson, G.; Näslund, P.H. Spreadsheet programming—A new approach in physiologically based modeling of solvent toxicokinetics. Toxicol. Lett. 1988, 41, 115–127. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.; Jaberi-Douraki, M.; He, C.; Jin, S.; Yang, R.S.H.; Fisher, J.W.; Riviere, J.E. Performance Assessment and Translation of Physiologically Based Pharmacokinetic Models From acslX to Berkeley Madonna, MATLAB, and R Language: Oxytetracycline and Gold Nanoparticles As Case Examples. Toxicol. Sci. 2017, 158, 23–35. [Google Scholar] [CrossRef] [PubMed]
- Peters, S.A. Physiologically-based pharmacokinetic (PBPK) modeling and simulations: Principles, methods, and applications in the pharmaceutical industry. In Quantitative Modeling in Toxicology, 3rd ed.; Krishnan, K., Loizou, G.D., Spendiff, M., Lipscomb, J.C., Andersen, M.E., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2012; pp. 158–159. [Google Scholar]
- Wang, D.; Yang, F.; Sun, L.; Bai, J.; LI, Y. Physiologically based pharmacokinetic modeling based on different softwares. Chin. J. Vet. Med. 2015, 51, 58–60. [Google Scholar]
- Mahdi, W.A.; Hussain, A.; Altamimi, M.A.; Alshehri, S.; Bukhari, S.I.; Ahsan, M.N. Experimental Solubility, Thermodynamic/Computational Validations, and GastroPlus-Based In Silico Prediction for Subcutaneous Delivery of Rifampicin. AAPS Pharm. 2021, 22, 116. [Google Scholar] [CrossRef] [PubMed]
- Hanke, N.; Gómez-Mantilla, J.D.; Ishiguro, N.; Stopfer, P.; Nock, V. Physiologically Based Pharmacokinetic Modeling of Rosuvastatin to Predict Transporter-Mediated Drug-Drug Interactions. Pharm. Res. 2021, 38, 1645–1661. [Google Scholar] [CrossRef] [PubMed]
- WHO. Characterization and Application of Physiologically Based Pharmacokinetic Models in Risk Assessment; WHO: Geneva, Switzerland, 2010; pp. 21–37. [Google Scholar]
- Zhao, H.; Ge, P.; Chang, X.; Ren, X.; Chen, L.; Gao, J.; Hu, J.; Li, X. Physiologically based toxicokinetic model for nickel. J. Hyg. Res. 2017, 46, 797–801. [Google Scholar]
- Vrieze, S.I. Model selection and psychological theory: A discussion of the differences between the Akaike information criterion (AIC) and the Bayesian information criterion (BIC). Psychol. Methods 2012, 17, 228–243. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.; Li, M.; Gehring, R.; Riviere, J.E. Development and application of a multiroute physiologically based pharmacokinetic model for oxytetracycline in dogs and humans. J. Pharm. Sci. 2015, 104, 233–243. [Google Scholar] [CrossRef]
- Portet, S. A primer on model selection using the Akaike Information Criterion. Infect. Dis. Model. 2020, 5, 111–128. [Google Scholar] [CrossRef]
- Cox, L.A., Jr. Reassessing benzene risks using internal doses and Monte-Carlo uncertainty analysis. Environ. Health Perspect. 1996, 104, 1413–1429. [Google Scholar] [PubMed]
- Farrar, D.; Allen, B.; Crump, K.; Shipp, A. Evaluation of uncertainty in input parameters to pharmacokinetic models and the resulting uncertainty in output. Toxicol. Lett. 1989, 49, 371–385. [Google Scholar] [CrossRef] [PubMed]
- Iman, R.L.; Helton, J.C. An Investigation of Uncertainty and Sensitivity Analysis Techniques for Computer Models. Risk Anals. 1988, 8, 71–90. [Google Scholar] [CrossRef]
- Khazaee, M.; Ng, C.A. Evaluating parameter availability for physiologically based pharmacokinetic (PBPK) modeling of perfluorooctanoic acid (PFOA) in zebrafish. Environ. Sci. Process Impacts 2018, 20, 105–119. [Google Scholar] [CrossRef] [PubMed]
- Krewski, D.; Wang, Y.; Bartlett, S.; Krishnan, K. Uncertainty, variability, and sensitivity analysis in physiological pharmacokinetic models. J. Biopharm. Stat. 1995, 5, 245–271. [Google Scholar] [CrossRef] [PubMed]
- Radomyski, A.; Giubilato, E.; Ciffroy, P.; Critto, A.; Brochot, C.; Marcomini, A. Modelling ecological and human exposure to POPs in Venice lagoon—Part II: Quantitative uncertainty and sensitivity analysis in coupled exposure models. Sci. Total Environ. 2016, 569–570, 1635–1649. [Google Scholar] [CrossRef] [PubMed]
- Gueorguieva, I.I.; Nestorov, I.A.; Rowland, M. Fuzzy simulation of pharmacokinetic models: Case study of whole body physiologically based model of diazepam. J. Pharmacokinet. Pharmacodyn. 2004, 31, 185–213. [Google Scholar] [CrossRef]
- Nestorov, I. Modelling and simulation of variability and uncertainty in toxicokinetics and pharmacokinetics. Toxicol. Lett. 2001, 120, 411–420. [Google Scholar] [CrossRef]
- Seng, K.Y.; Nestorov, I.; Vicini, P. Simulating pharmacokinetic and pharmacodynamic fuzzy-parameterized models: A comparison of numerical methods. J. Pharmacokinet. Pharmacodyn. 2007, 34, 595–621. [Google Scholar] [CrossRef]
- Nong, A.; Krishnan, K. Estimation of interindividual pharmacokinetic variability factor for inhaled volatile organic chemicals using a probability-bounds approach. Regul. Toxicol. Pharmacol. 2007, 48, 93–101. [Google Scholar] [CrossRef]
- Bois, F.Y.; Jamei, M.; Clewell, H.J. PBPK modelling of inter-individual variability in the pharmacokinetics of environmental chemicals. Toxicology 2010, 278, 256–267. [Google Scholar] [CrossRef] [PubMed]
- Krauss, M.; Burghaus, R.; Lippert, J.; Niemi, M.; Neuvonen, P.; Schuppert, A.; Willmann, S.; Kuepfer, L.; Görlitz, L. Using Bayesian-PBPK modeling for assessment of inter-individual variability and subgroup stratification. Silico. Pharmacol. 2013, 1, 6. [Google Scholar] [CrossRef] [PubMed]
- Krauss, M.; Tappe, K.; Schuppert, A.; Kuepfer, L.; Goerlitz, L. Bayesian Population Physiologically-Based Pharmacokinetic (PBPK) Approach for a Physiologically Realistic Characterization of Interindividual Variability in Clinically Relevant Populations. PLoS ONE 2015, 10, e0139423. [Google Scholar] [CrossRef] [PubMed]
- Weijs, L.; Yang, R.S.; Das, K.; Covaci, A.; Blust, R. Application of Bayesian population physiologically based pharmacokinetic (PBPK) modeling and Markov chain Monte Carlo simulations to pesticide kinetics studies in protected marine mammals: DDT, DDE, and DDD in harbor porpoises. Environ. Sci. Technol. 2013, 47, 4365–4374. [Google Scholar] [CrossRef] [PubMed]
- Huet, R.; Johanson, G. 1,1-Difluoroethane Detection Time in Blood after Inhalation Abuse Estimated by Monte Carlo PBPK Modeling. Pharmaceutics 2020, 12, 997. [Google Scholar] [CrossRef] [PubMed]
- Saltelli, A.; Ratto, M.; Tarantola, S.; Campolongo, F. Update 1 of: Sensitivity analysis for chemical models. Chem. Rev. 2012, 112, Pr1–Pr21. [Google Scholar] [CrossRef]
- Brochot, C.; Smith, T.J.; Bois, F.Y. Development of a physiologically based toxicokinetic model for butadiene and four major metabolites in humans: Global sensitivity analysis for experimental design issues. Chem. Biol. Interact. 2007, 167, 168–183. [Google Scholar] [CrossRef] [PubMed]
- Saltelli, A.; Tarantola, S.; Chan, K.P.S. A Quantitative Model-Independent Method for Global Sensitivity Analysis of Model Output. Technometrics 1999, 41, 39–56. [Google Scholar] [CrossRef]
- Jeong, H.C.; Chae, Y.J.; Shin, K.H. Predicting the systemic exposure and lung concentration of nafamostat using physiologically-based pharmacokinetic modeling. Transl. Clin. Pharmacol. 2022, 30, 201–211. [Google Scholar] [CrossRef]
- Yu, L.; Guo, J.; Guo, J.; Yin j Cui, L.; Yang, Y.; Zhao, J.; Peng, S. Prediction of drug toxicity by reverse dosimetry based on PBPK modeling. In Proceedings of the 3rd International Conference on Toxicity Testing Alternative and Translational Toxicology, Nanjing, China, 9–12 July 2017. [Google Scholar]
- Louisse, J.; Beekmann, K.; Rietjens, I.M. Use of Physiologically Based Kinetic Modeling-Based Reverse Dosimetry to Predict in Vivo Toxicity from in Vitro Data. Chem. Res. Toxicol. 2017, 30, 114–125. [Google Scholar] [CrossRef]
- Cooper, A.B.; Aggarwal, M.; Bartels, M.J.; Morriss, A.; Terry, C.; Lord, G.A.; Gant, T.W. PBTK model for assessment of operator exposure to haloxyfop using human biomonitoring and toxicokinetic data. Regul. Toxicol. Pharmacol. 2019, 102, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Poulin, P.; Burczynski, F.J.; Haddad, S. The Role of Extracellular Binding Proteins in the Cellular Uptake of Drugs: Impact on Quantitative In Vitro-to-In Vivo Extrapolations of Toxicity and Efficacy in Physiologically Based Pharmacokinetic-Pharmacodynamic Research. J. Pharm. Sci. 2016, 105, 497–508. [Google Scholar] [CrossRef] [PubMed]
- Jaroch, K.; Jaroch, A.; Bojko, B. Cell cultures in drug discovery and development: The need of reliable in vitro-in vivo extrapolation for pharmacodynamics and pharmacokinetics assessment. J. Pharm. Biomed. Anal. 2018, 147, 297–312. [Google Scholar] [CrossRef] [PubMed]
- Fabian, E.; Gomes, C.; Birk, B.; Williford, T.; Hernandez, T.R.; Haase, C.; Zbranek, R.; van Ravenzwaay, B.; Landsiedel, R. In vitro-to-in vivo extrapolation (IVIVE) by PBTK modeling for animal-free risk assessment approaches of potential endocrine-disrupting compounds. Arch. Toxicol. 2019, 93, 401–416. [Google Scholar] [CrossRef] [PubMed]
- Dejongh, J.; Forsby, A.; Houston, J.B.; Beckman, M.; Combes, R.; Blaauboer, B.J. An Integrated Approach to the Prediction of Systemic Toxicity using Computer-based Biokinetic Models and Biological In vitro Test Methods: Overview of a Prevalidation Study Based on the ECITTS Project. Toxicol. Vitr. 1999, 13, 549–554. [Google Scholar] [CrossRef] [PubMed]
- Brinkmann, M.; Eichbaum, K.; Buchinger, S.; Reifferscheid, G.; Bui, T.; Schäffer, A.; Hollert, H.; Preuss, T.G. Understanding receptor-mediated effects in rainbow trout: In vitro-in vivo extrapolation using physiologically based toxicokinetic models. Environ. Sci. Technol. 2014, 48, 3303–3309. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Liu, M. Solution of lnternal Doses for lnhaled Vinyl Chloride by Physiologically Based Toxicokinetic (PBTK) Model. Ind. Health Occup. Dis. 2009, 35, 280–284. [Google Scholar]
- Gajewska, M.; Worth, A.; Urani, C.; Briesen, H.; Schramm, K.W. Application of physiologically-based toxicokinetic modelling in oral-to-dermal extrapolation of threshold doses of cosmetic ingredients. Toxicol. Lett. 2014, 227, 189–202. [Google Scholar] [CrossRef]
- Sweeney, L.M.; Kirman, C.R.; Gargas, M.L.; Carson, M.L.; Tardiff, R.G. Development of a physiologically-based toxicokinetic model of acrylamide and glycidamide in rats and humans. Food Chem. Toxicol. 2010, 48, 668–685. [Google Scholar] [CrossRef]
- Abduljalil, K.; Furness, P.; Johnson, T.N.; Rostami-Hodjegan, A.; Soltani, H. Anatomical, physiological and metabolic changes with gestational age during normal pregnancy: A database for parameters required in physiologically based pharmacokinetic modelling. Clin. Pharmacokinet 2012, 51, 365–396. [Google Scholar] [CrossRef]
- Xia, B.; Heimbach, T.; Gollen, R.; Nanavati, C.; He, H. A simplified PBPK modeling approach for prediction of pharmacokinetics of four primarily renally excreted and CYP3A metabolized compounds during pregnancy. AAPS J. 2013, 15, 1012–1024. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.; Shi, A.X. Current application of physiologically based pharmacokinetic model and its applications in pregnant women’s drug research. Chin. J. Clin. Pharmacol. 2017, 33, 2209–2211. [Google Scholar]
- Dallmann, A.; Pfister, M.; van den Anker, J.; Eissing, T. Physiologically Based Pharmacokinetic Modeling in Pregnancy: A Systematic Review of Published Models. Clin. Pharmacol. Ther. 2018, 104, 1110–1124. [Google Scholar] [CrossRef] [PubMed]
- Corley, R.A.; Mast, T.J.; Carney, E.W.; Rogers, J.M.; Daston, G.P. Evaluation of physiologically based models of pregnancy and lactation for their application in children’s health risk assessments. Crit. Rev. Toxicol 2003, 33, 137–211. [Google Scholar] [CrossRef] [PubMed]
- Gaohua, L.; Abduljalil, K.; Jamei, M.; Johnson, T.N.; Rostami-Hodjegan, A. A pregnancy physiologically based pharmacokinetic (p-PBPK) model for disposition of drugs metabolized by CYP1A2, CYP2D6 and CYP3A4. Br. J. Clin. Pharmacol. 2012, 74, 873–885. [Google Scholar] [CrossRef] [PubMed]
- Edginton, A.N.; Schmitt, W.; Willmann, S. Development and evaluation of a generic physiologically based pharmacokinetic model for children. Clin. Pharmacokinet 2006, 45, 1013–1034. [Google Scholar] [CrossRef] [PubMed]
- Kovar, L.; Schräpel, C.; Selzer, D.; Kohl, Y.; Bals, R.; Schwab, M.; Lehr, T. Physiologically-Based Pharmacokinetic (PBPK) Modeling of Buprenorphine in Adults, Children and Preterm Neonates. Pharmaceutics 2020, 12, 578. [Google Scholar] [CrossRef]
- Khalil, F.; Läer, S. Physiologically based pharmacokinetic modeling: Methodology, applications, and limitations with a focus on its role in pediatric drug development. J. Biomed. Biotechnol. 2011, 2011, 907461. [Google Scholar] [CrossRef]
- Strougo, A.; Yassen, A.; Krauwinkel, W.; Danhof, M.; Freijer, J. A semiphysiological population model for prediction of the pharmacokinetics of drugs under liver and renal disease conditions. Drug Metab. Dispos. 2011, 39, 1278–1287. [Google Scholar] [CrossRef]
- Edginton, A.N.; Willmann, S. Physiology-based simulations of a pathological condition: Prediction of pharmacokinetics in patients with liver cirrhosis. Clin. Pharmacokinet 2008, 47, 743–752. [Google Scholar] [CrossRef]
- Schuppan, D.; Afdhal, N.H. Liver cirrhosis. Lancet 2008, 371, 838–851. [Google Scholar] [CrossRef] [PubMed]
- Nolin, T.D.; Naud, J.; Leblond, F.A.; Pichette, V. Emerging evidence of the impact of kidney disease on drug metabolism and transport. Clin. Pharmacol. Ther. 2008, 83, 898–903. [Google Scholar] [CrossRef] [PubMed]
- Li, G.F.; Wang, K.; Chen, R.; Zhao, H.R.; Yang, J.; Zheng, Q.S. Simulation of the pharmacokinetics of bisoprolol in healthy adults and patients with impaired renal function using whole-body physiologically based pharmacokinetic modeling. Acta Pharmacol. Sin. 2012, 33, 1359–1371. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Liu, X.; Yang, J. A semi-physiologically based pharmacokinetic pharmacodynamic model for glycyrrhizin-induced pseudoaldosteronism and prediction of the dose limit causing hypokalemia in a virtual elderly population. PLoS ONE 2014, 9, e114049. [Google Scholar] [CrossRef] [PubMed]
- Haddad, S.; Tardif, R.; Charest-Tardif, G.; Krishnan, K. Physiological modeling of the toxicokinetic interactions in a quaternary mixture of aromatic hydrocarbons. Toxicol. Appl. Pharmacol. 1999, 161, 249–257. [Google Scholar] [CrossRef] [PubMed]
- Plowchalk, D.R.; Andersen, M.E.; Bogdanffy, M.S. Physiologically based modeling of vinyl acetate uptake, metabolism, and intracellular pH changes in the rat nasal cavity. Toxicol. Appl. Pharmacol. 1997, 142, 386–400. [Google Scholar] [CrossRef] [PubMed]
- McNally, K.; Cotton, R.; Loizou, G.D. A Workflow for Global Sensitivity Analysis of PBPK Models. Front. Pharmacol. 2011, 2, 31. [Google Scholar] [CrossRef] [PubMed]
- Lin, W.; Chen, Y.; Unadkat, J.D.; Zhang, X.; Wu, D.; Heimbach, T. Applications, Challenges, and Outlook for PBPK Modeling and Simulation: A Regulatory, Industrial and Academic Perspective. Pharm. Res. 2022, 39, 1701–1731. [Google Scholar] [CrossRef]
- OECD. Guidance Document on the Characterization, Validation and Reporting of Physiologically Based Kinetic (PBK) Models for Regulatory Purposes; OECD Series on Testing and Assessment, No. 331, Environment, Health and Safety, Environment Directorate; OECD: Paris, France, 2021. [Google Scholar]
Toxicokinetic Process | Equation | |
---|---|---|
Absorption | ||
Respiratory tract | (2) | |
Percutaneous | (3) | |
Oral | (4) | |
Intravenous | (5) | |
Distribution | ||
Protein binding | (6) | |
Irrigation rate-limiting structure | (7) | |
Membrane rate-limiting tissue | (8) | |
Metabolism | ||
First-order kinetic | (9) | |
Second-order kinetic | (10) | |
Saturation process | (11) | |
Excretion | ||
Kidney | (12) | |
Lung | (13) |
Parameter | Data Source | |
---|---|---|
Physiological parameter | Body weight Cardiac output Blood flow to organ or tissue Volume of an organ or tissue Alveolar ventilation | Literature In vivo experiment. |
Physicochemical parameter | Blood-air distribution coefficient Tissue-blood distribution coefficient | Literature In vivo experiment In vitro experiment In silico prediction |
Biochemical parameter | Maximum velocity Michaelis constant First-order rate constant Second-order rate constant | Literature In vivo experiment In vitro experiment In silico prediction |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chen, M.; Du, R.; Zhang, T.; Li, C.; Bao, W.; Xin, F.; Hou, S.; Yang, Q.; Chen, L.; Wang, Q.; et al. The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment. Toxics 2023, 11, 874. https://doi.org/10.3390/toxics11100874
Chen M, Du R, Zhang T, Li C, Bao W, Xin F, Hou S, Yang Q, Chen L, Wang Q, et al. The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment. Toxics. 2023; 11(10):874. https://doi.org/10.3390/toxics11100874
Chicago/Turabian StyleChen, Mengting, Ruihu Du, Tao Zhang, Chutao Li, Wenqiang Bao, Fan Xin, Shaozhang Hou, Qiaomei Yang, Li Chen, Qi Wang, and et al. 2023. "The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment" Toxics 11, no. 10: 874. https://doi.org/10.3390/toxics11100874
APA StyleChen, M., Du, R., Zhang, T., Li, C., Bao, W., Xin, F., Hou, S., Yang, Q., Chen, L., Wang, Q., & Zhu, A. (2023). The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment. Toxics, 11(10), 874. https://doi.org/10.3390/toxics11100874