1
|
Niemeijer M, Więcek W, Fu S, Huppelschoten S, Bouwman P, Baze A, Parmentier C, Richert L, Paules RS, Bois FY, van de Water B. Mapping Interindividual Variability of Toxicodynamics Using High-Throughput Transcriptomics and Primary Human Hepatocytes from Fifty Donors. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:37005. [PMID: 38498338 PMCID: PMC10947137 DOI: 10.1289/ehp11891] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Understanding the variability across the human population with respect to toxicodynamic responses after exposure to chemicals, such as environmental toxicants or drugs, is essential to define safety factors for risk assessment to protect the entire population. Activation of cellular stress response pathways are early adverse outcome pathway (AOP) key events of chemical-induced toxicity and would elucidate the estimation of population variability of toxicodynamic responses. OBJECTIVES We aimed to map the variability in cellular stress response activation in a large panel of primary human hepatocyte (PHH) donors to aid in the quantification of toxicodynamic interindividual variability to derive safety uncertainty factors. METHODS High-throughput transcriptomics of over 8,000 samples in total was performed covering a panel of 50 individual PHH donors upon 8 to 24 h exposure to broad concentration ranges of four different toxicological relevant stimuli: tunicamycin for the unfolded protein response (UPR), diethyl maleate for the oxidative stress response (OSR), cisplatin for the DNA damage response (DDR), and tumor necrosis factor alpha (TNF α ) for NF- κ B signaling. Using a population mixed-effect framework, the distribution of benchmark concentrations (BMCs) and maximum fold change were modeled to evaluate the influence of PHH donor panel size on the correct estimation of interindividual variability for the various stimuli. RESULTS Transcriptome mapping allowed the investigation of the interindividual variability in concentration-dependent stress response activation, where the average of BMCs had a maximum difference of 864-, 13-, 13-, and 259-fold between different PHHs for UPR, OSR, DDR, and NF- κ B signaling-related genes, respectively. Population modeling revealed that small PHH panel sizes systematically underestimated the variance and gave low probabilities in estimating the correct human population variance. Estimated toxicodynamic variability factors of stress response activation in PHHs based on this dataset ranged between 1.6 and 6.3. DISCUSSION Overall, by combining high-throughput transcriptomics and population modeling, improved understanding of interindividual variability in chemical-induced activation of toxicity relevant stress pathways across the human population using a large panel of plated cryopreserved PHHs was established, thereby contributing toward increasing the confidence of in vitro-based prediction of adverse responses, in particular hepatotoxicity. https://doi.org/10.1289/EHP11891.
Collapse
Affiliation(s)
- Marije Niemeijer
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | | | - Shuai Fu
- Simcyp Division, CERTARA, Sheffield, UK
| | - Suzanna Huppelschoten
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | - Peter Bouwman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | | | | | | | - Richard S. Paules
- Division of the National Toxicology Program, NIEHS, NIH, Research Triangle Park, North Carolina, USA
| | | | - Bob van de Water
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Elmokadem A, Zhang Y, Knab T, Jordie E, Gillespie WR. Bayesian PBPK modeling using R/Stan/Torsten and Julia/SciML/Turing.Jl. CPT Pharmacometrics Syst Pharmacol 2023; 12:300-310. [PMID: 36661183 PMCID: PMC10014045 DOI: 10.1002/psp4.12926] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Physiologically-based pharmacokinetic (PBPK) models are mechanistic models that are built based on an investigator's prior knowledge of the in vivo system of interest. Bayesian inference incorporates an investigator's prior knowledge of parameters while using the data to update this knowledge. As such, Bayesian tools are well-suited to infer PBPK model parameters using the strong prior knowledge available while quantifying the uncertainty on these parameters. This tutorial demonstrates a full population Bayesian PBPK analysis framework using R/Stan/Torsten and Julia/SciML/Turing.jl.
Collapse
Affiliation(s)
| | - Yi Zhang
- Sage Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Timothy Knab
- Metrum Research Group, Tariffville, Connecticut, USA
| | - Eric Jordie
- Metrum Research Group, Tariffville, Connecticut, USA
| | | |
Collapse
|
5
|
Middleton AM, Reynolds J, Cable S, Baltazar MT, Li H, Bevan S, Carmichael PL, Dent MP, Hatherell S, Houghton J, Kukic P, Liddell M, Malcomber S, Nicol B, Park B, Patel H, Scott S, Sparham C, Walker P, White A. Are Non-animal Systemic Safety Assessments Protective? A Toolbox and Workflow. Toxicol Sci 2022; 189:124-147. [PMID: 35822611 PMCID: PMC9412174 DOI: 10.1093/toxsci/kfac068] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An important question in toxicological risk assessment is whether non-animal new approach methodologies (NAMs) can be used to make safety decisions that are protective of human health, without being overly conservative. In this work, we propose a core NAM toolbox and workflow for conducting systemic safety assessments for adult consumers. We also present an approach for evaluating how protective and useful the toolbox and workflow are by benchmarking against historical safety decisions. The toolbox includes physiologically based kinetic (PBK) models to estimate systemic Cmax levels in humans, and 3 bioactivity platforms, comprising high-throughput transcriptomics, a cell stress panel, and in vitro pharmacological profiling, from which points of departure are estimated. A Bayesian model was developed to quantify the uncertainty in the Cmax estimates depending on how the PBK models were parameterized. The feasibility of the evaluation approach was tested using 24 exposure scenarios from 10 chemicals, some of which would be considered high risk from a consumer goods perspective (eg, drugs that are systemically bioactive) and some low risk (eg, existing food or cosmetic ingredients). Using novel protectiveness and utility metrics, it was shown that up to 69% (9/13) of the low risk scenarios could be identified as such using the toolbox, whilst being protective against all (5/5) the high-risk ones. The results demonstrated how robust safety decisions could be made without using animal data. This work will enable a full evaluation to assess how protective and useful the toolbox and workflow are across a broader range of chemical-exposure scenarios.
Collapse
Affiliation(s)
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Cable
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Matthew Philip Dent
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Jade Houghton
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Mark Liddell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Beate Nicol
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hiral Patel
- Charles River Laboratories, Cambridgeshire, CB10 1XL, UK
| | - Sharon Scott
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Chris Sparham
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Cheshire SK10 4TG, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
6
|
Wedagedera JR, Afuape A, Chirumamilla SK, Momiji H, Leary R, Dunlavey M, Matthews R, Abduljalil K, Jamei M, Bois FY. Population PBPK modeling using parametric and nonparametric methods of the Simcyp Simulator, and Bayesian samplers. CPT Pharmacometrics Syst Pharmacol 2022; 11:755-765. [PMID: 35385609 PMCID: PMC9197540 DOI: 10.1002/psp4.12787] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Physiologically‐based pharmacokinetic (PBPK) models usually include a large number of parameters whose values are obtained using in vitro to in vivo extrapolation. However, such extrapolations can be uncertain and may benefit from inclusion of evidence from clinical observations via parametric inference. When clinical interindividual variability is high, or the data sparse, it is essential to use a population pharmacokinetics inferential framework to estimate unknown or uncertain parameters. Several approaches are available for that purpose, but their relative advantages for PBPK modeling are unclear. We compare the results obtained using a minimal PBPK model of a canonical theophylline dataset with quasi‐random parametric expectation maximization (QRPEM), nonparametric adaptive grid estimation (NPAG), Bayesian Metropolis‐Hastings (MH), and Hamiltonian Markov Chain Monte Carlo sampling. QRPEM and NPAG gave consistent population and individual parameter estimates, mostly agreeing with Bayesian estimates. MH simulations ran faster than the others methods, which together had similar performance.
Collapse
Affiliation(s)
| | | | | | | | - Robert Leary
- CERTARA UK Limited, Simcyp Division, Sheffield, UK
| | | | | | | | - Masoud Jamei
- CERTARA UK Limited, Simcyp Division, Sheffield, UK
| | | |
Collapse
|
7
|
Talkington AM, McSweeney MD, Wessler T, Rath MK, Li Z, Zhang T, Yuan H, Frank JE, Forest MG, Cao Y, Lai SK. A PBPK model recapitulates early kinetics of anti-PEG antibody-mediated clearance of PEG-liposomes. J Control Release 2022; 343:518-527. [PMID: 35066099 PMCID: PMC9080587 DOI: 10.1016/j.jconrel.2022.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/23/2022]
Abstract
PEGylation is routinely used to extend the systemic circulation of various protein therapeutics and nanomedicines. Nonetheless, mounting evidence is emerging that individuals exposed to select PEGylated therapeutics can develop antibodies specific to PEG, i.e., anti-PEG antibodies (APA). In turn, APA increase both the risk of hypersensitivity to the drug as well as potential loss of efficacy due to accelerated blood clearance of the drug. Despite the broad implications of APA, the timescales and systemic specificity by which APA can alter the pharmacokinetics and biodistribution of PEGylated drugs remain not well understood. Here, we developed a physiologically based pharmacokinetic (PBPK) model designed to resolve APA's impact on both early- and late-phase pharmacokinetics and biodistribution of intravenously administered PEGylated drugs. Our model accurately recapitulates PK and biodistribution data obtained from PET/CT imaging of radiolabeled PEG-liposomes and PEG-uricase in mice with and without APA, as well as serum levels of PEG-uricase in humans. Our work provides another illustration of the power of high-resolution PBPK models for understanding the pharmacokinetic impacts of anti-drug antibodies and the dynamics with which antibodies can mediate clearance of foreign species.
Collapse
Affiliation(s)
- Anne M Talkington
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Morgan D McSweeney
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy Wessler
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA; Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Marielle K Rath
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Zibo Li
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Tao Zhang
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Yuan
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, UNC Chapel Hill, USA
| | | | - M Gregory Forest
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA; Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel K Lai
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Talkington AM, Wessler T, Lai SK, Cao Y, Forest MG. Experimental Data and PBPK Modeling Quantify Antibody Interference in PEGylated Drug Carrier Delivery. Bull Math Biol 2021; 83:123. [PMID: 34751832 PMCID: PMC8576315 DOI: 10.1007/s11538-021-00950-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling is a popular drug development tool that integrates physiology, drug physicochemical properties, preclinical data, and clinical information to predict drug systemic disposition. Since PBPK models seek to capture complex physiology, parameter uncertainty and variability is a prevailing challenge: there are often more compartments (e.g., organs, each with drug flux and retention mechanisms, and associated model parameters) than can be simultaneously measured. To improve the fidelity of PBPK modeling, one approach is to search and optimize within the high-dimensional model parameter space, based on experimental time-series measurements of drug distributions. Here, we employ Latin Hypercube Sampling (LHS) on a PBPK model of PEG-liposomes (PL) that tracks biodistribution in an 8-compartment mouse circulatory system, in the presence (APA+) or absence (naïve) of anti-PEG antibodies (APA). Near-continuous experimental measurements of PL concentration during the first hour post-injection from the liver, spleen, kidney, muscle, lung, and blood plasma, based on PET/CT imaging in live mice, are used as truth sets with LHS to infer optimal parameter ranges for the full PBPK model. The data and model quantify that PL retention in the liver is the primary differentiator of biodistribution patterns in naïve versus APA+ mice, and spleen the secondary differentiator. Retention of PEGylated nanomedicines is substantially amplified in APA+ mice, likely due to PL-bound APA engaging specific receptors in the liver and spleen that bind antibody Fc domains. Our work illustrates how applying LHS to PBPK models can further mechanistic understanding of the biodistribution and antibody-mediated clearance of specific drugs.
Collapse
Affiliation(s)
- Anne M Talkington
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA.
| | - Timothy Wessler
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel K Lai
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, NC, USA
| | - M Gregory Forest
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA.
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
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
|
10
|
Lin HC, Chen WY. Bayesian population physiologically-based pharmacokinetic model for robustness evaluation of withdrawal time in tilapia aquaculture administrated to florfenicol. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 210:111867. [PMID: 33387907 DOI: 10.1016/j.ecoenv.2020.111867] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 06/12/2023]
Abstract
The antimicrobial residues of aquacultural production is a growing public concern, leading to reexamine the method for establishing robust withdrawal time and ensuring food safety. Our study aims to develop the optimizing population physiologically-based pharmacokinetic (PBPK) model for assessing florfenicol residues in the tilapia tissues, and for evaluating the robustness of the withdrawal time (WT). Fitting with published pharmacokinetic profiles that experimented under temperatures of 22 and 28 °C, a PBPK model was constructed by applying with the Bayesian Markov chain Monte Carol (MCMC) algorithm to estimate WTs under different physiological, environmental and dosing scenarios. Results show that the MCMC algorithm improves the estimates of uncertainty and variability of PBPK-related parameters, and optimizes the simulation of the PBPK model. It is noteworthy that posterior sets generated from temperature-associated datasets to be respectively used for simulating residues under corresponding temperature conditions. Simulating the residues under regulated regimen and overdosing scenarios for Taiwan, the estimated WTs were 12-16 days at 22 °C and 9-12 days at 28 °C, while for the USA, the estimated WTs were 14-18 and 11-14 days, respectively. Comparison with the regulated WT of 15 days, results indicate that the current WT has well robustness and resilience in the environment of higher temperatures. The optimal Bayesian population PBPK model provides effective analysis for determining WTs under scenario-specific conditions. It is a new insight into the increasing body of literature on developing the Bayesian-PBPK model and has practical implications for improving the regulation of food safety.
Collapse
Affiliation(s)
- Hsing-Chieh Lin
- Department of Ecology and Environmental Resources, National University of Tainan, Tainan, Taiwan
| | - Wei-Yu Chen
- Department of Ecology and Environmental Resources, National University of Tainan, Tainan, Taiwan.
| |
Collapse
|
11
|
Loisios-Konstantinidis I, Dressman J. Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling to Support Waivers of In Vivo Clinical Studies: Current Status, Challenges, and Opportunities. Mol Pharm 2020; 18:1-17. [PMID: 33320002 DOI: 10.1021/acs.molpharmaceut.0c00903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling has been extensively applied to quantitatively translate in vitro data, predict the in vivo performance, and ultimately support waivers of in vivo clinical studies. In the area of biopharmaceutics and within the context of model-informed drug discovery and development (MID3), there is a rapidly growing interest in applying verified and validated mechanistic PBPK models to waive in vivo clinical studies. However, the regulatory acceptance of PBPK analyses for biopharmaceutics and oral drug absorption applications, which is also referred to variously as "PBPK absorption modeling" [Zhang et al. CPT: Pharmacometrics Syst. Pharmacol. 2017, 6, 492], "physiologically based absorption modeling", or "physiologically based biopharmaceutics modeling" (PBBM), remains rather low [Kesisoglou et al. J. Pharm. Sci. 2016, 105, 2723] [Heimbach et al. AAPS J. 2019, 21, 29]. Despite considerable progress in the understanding of gastrointestinal (GI) physiology, in vitro biopharmaceutic and in silico tools, PBPK models for oral absorption often suffer from an incomplete understanding of the physiology, overparameterization, and insufficient model validation and/or platform verification, all of which can represent limitations to their translatability and predictive performance. The complex interactions of drug substances and (bioenabling) formulations with the highly dynamic and heterogeneous environment of the GI tract in different age, ethnic, and genetic groups as well as disease states have not been yet fully elucidated, and they deserve further research. Along with advancements in the understanding of GI physiology and refinement of current or development of fully mechanistic in silico tools, we strongly believe that harmonization, interdisciplinary interaction, and enhancement of the translational link between in vitro, in silico, and in vivo will determine the future of PBBM. This Perspective provides an overview of the current status of PBBM, reflects on challenges and knowledge gaps, and discusses future opportunities around PBPK/PD models for oral absorption of small and large molecules to waive in vivo clinical studies.
Collapse
Affiliation(s)
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main 60438, Germany.,Fraunhofer Institute of Translational Pharmacology and Medicine (ITMP), Carl-von-Noorden Platz 9, Frankfurt am Main 60438, Germany
| |
Collapse
|
12
|
Dalaijamts C, Cichocki JA, Luo YS, Rusyn I, Chiu WA. PBPK modeling of impact of nonalcoholic fatty liver disease on toxicokinetics of perchloroethylene in mice. Toxicol Appl Pharmacol 2020; 400:115069. [PMID: 32445755 DOI: 10.1016/j.taap.2020.115069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD), a major cause of chronic liver disease in the Western countries with increasing prevalence worldwide, may substantially affect chemical toxicokinetics and thereby modulate chemical toxicity. OBJECTIVES This study aims to use physiologically-based pharmacokinetic (PBPK) modeling to characterize the impact of NAFLD on toxicokinetics of perchloroethylene (perc). METHODS Quantitative measures of physiological and biochemical changes associated with the presence of NAFLD induced by high-fat or methionine/choline-deficient diets in C57B1/6 J mice are incorporated into a previously developed PBPK model for perc and its oxidative and conjugative metabolites. Impacts on liver fat and volume, as well as blood:air and liver:air partition coefficients, are incorporated into the model. Hierarchical Bayesian population analysis using Markov chain Monte Carlo simulation is conducted to characterize uncertainty, as well as disease-induced variability in toxicokinetics. RESULTS NAFLD has a major effect on toxicokinetics of perc, with greater oxidative and lower conjugative metabolism as compared to healthy mice. The NAFLD-updated PBPK model accurately predicts in vivo metabolism of perc through oxidative and conjugative pathways in all tissues across disease states and strains, but underestimated parent compound concentrations in blood and liver of NAFLD mice. CONCLUSIONS We demonstrate the application of PBPK modeling to predict the effects of pre-existing disease conditions as a variability factor in perc metabolism. These results suggest that non-genetic factors such as diet and pre-existing disease can be as influential as genetic factors in altering toxicokinetics of perc, and thus are likely contribute substantially to population variation in its adverse effects.
Collapse
Affiliation(s)
- Chimeddulam Dalaijamts
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Joseph A Cichocki
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Yu-Syuan Luo
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Weihsueh A Chiu
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
13
|
Maharao N, Antontsev V, Wright M, Varshney J. Entering the era of computationally driven drug development. Drug Metab Rev 2020; 52:283-298. [PMID: 32083960 DOI: 10.1080/03602532.2020.1726944] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Historically, failure rates in drug development are high; increased sophistication and investment throughout the process has shifted the reasons for attrition, but the overall success rates have remained stubbornly and consistently low. Only 8% of new entities entering clinical testing gain regulatory approval, indicating that significant obstacles still exist for efficient therapeutic development. The continued high failure rate can be partially attributed to the inability to link drug exposure with the magnitude of observed safety and efficacy-related pharmacodynamic (PD) responses; frequently, this is a result of nonclinical models exhibiting poor prediction of human outcomes across a wide range of disease conditions, resulting in faulty evaluation of drug toxicology and efficacy. However, the increasing quality and standardization of experimental methods in preclinical stages of testing has created valuable data sets within companies that can be leveraged to further improve the efficiency and accuracy of preclinical prediction for both pharmacokinetics (PK) and PD. Models of Quantitative structure-activity relationships (QSAR), physiologically based pharmacokinetics (PBPK), and PK/PD relationships have also improved efficiency. Founded on a core understanding of biochemistry and physiological interactions of xenobiotics, these in silico methods have the potential to increase the probability of compound success in clinical trials. Integration of traditional computational methods with machine-learning approaches and existing internal pharma databases stands to make a fundamental impact on the speed and accuracy of predictions during the process of drug development and approval.
Collapse
|
14
|
Vizirianakis IS, Miliotou AN, Mystridis GA, Andriotis EG, Andreadis II, Papadopoulou LC, Fatouros DG. Tackling pharmacological response heterogeneity by PBPK modeling to advance precision medicine productivity of nanotechnology and genomics therapeutics. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1605828] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George A. Mystridis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios G. Andriotis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis I. Andreadis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Wagner JB, Abdel-Rahman S, Gaedigk R, Gaedigk A, Raghuveer G, Staggs VS, Kauffman R, Van Haandel L, Leeder JS. Impact of Genetic Variation on Pravastatin Systemic Exposure in Pediatric Hypercholesterolemia. Clin Pharmacol Ther 2019; 105:1501-1512. [PMID: 30549267 DOI: 10.1002/cpt.1330] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/20/2018] [Indexed: 11/07/2022]
Abstract
This study investigated the impact of SLCO1B1 genotype on pravastatin systemic exposure in children and adolescents with hypercholesterolemia. Participants (8-20 years) with at least one allelic variant of SLCO1B1 c.521T>C (521TC, n = 15; 521CC, n = 2) and wild-type controls (521TT, n = 15) completed a single oral dose pharmacokinetic study. Interindividual variability of pravastatin acid (PVA) exposure within SLCO1B1 genotype groups exceeded the approximately twofold difference in mean PVA exposure observed between SLCO1B1 genotype groups (P > 0.05, q > 0.10). The 3'α-iso-pravastatin acid and lactone isomer formation in the acidic environment of the stomach prior to absorption also was variable and affected PVA exposure in all genotype groups. The SLCO1B1 c.521 gene variant contributing to variability in systemic exposure to PVA in our pediatric cohort was comparable to previous studies in adults. However, other demographic and physicochemical factors seem to also contribute to interindividual variability in the dose-exposure relationship.
Collapse
Affiliation(s)
- Jonathan B Wagner
- Ward Family Heart Center, Children's Mercy, Kansas City, Missouri, USA
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Susan Abdel-Rahman
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Roger Gaedigk
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Geetha Raghuveer
- Ward Family Heart Center, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Vincent S Staggs
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
- Health Services & Outcomes Research, Children's Mercy, Kansas City, Missouri, USA
| | - Ralph Kauffman
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Leon Van Haandel
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Medical Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| |
Collapse
|
16
|
Evaluation of the whole body physiologically based pharmacokinetic (WB-PBPK) modeling of drugs. J Theor Biol 2018; 451:1-9. [DOI: 10.1016/j.jtbi.2018.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 11/17/2022]
|
17
|
Christ B, Dahmen U, Herrmann KH, König M, Reichenbach JR, Ricken T, Schleicher J, Ole Schwen L, Vlaic S, Waschinsky N. Computational Modeling in Liver Surgery. Front Physiol 2017; 8:906. [PMID: 29249974 PMCID: PMC5715340 DOI: 10.3389/fphys.2017.00906] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
The need for extended liver resection is increasing due to the growing incidence of liver tumors in aging societies. Individualized surgical planning is the key for identifying the optimal resection strategy and to minimize the risk of postoperative liver failure and tumor recurrence. Current computational tools provide virtual planning of liver resection by taking into account the spatial relationship between the tumor and the hepatic vascular trees, as well as the size of the future liver remnant. However, size and function of the liver are not necessarily equivalent. Hence, determining the future liver volume might misestimate the future liver function, especially in cases of hepatic comorbidities such as hepatic steatosis. A systems medicine approach could be applied, including biological, medical, and surgical aspects, by integrating all available anatomical and functional information of the individual patient. Such an approach holds promise for better prediction of postoperative liver function and hence improved risk assessment. This review provides an overview of mathematical models related to the liver and its function and explores their potential relevance for computational liver surgery. We first summarize key facts of hepatic anatomy, physiology, and pathology relevant for hepatic surgery, followed by a description of the computational tools currently used in liver surgical planning. Then we present selected state-of-the-art computational liver models potentially useful to support liver surgery. Finally, we discuss the main challenges that will need to be addressed when developing advanced computational planning tools in the context of liver surgery.
Collapse
Affiliation(s)
- Bruno Christ
- Molecular Hepatology Lab, Clinics of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Matthias König
- Department of Biology, Institute for Theoretical Biology, Humboldt University of Berlin, Berlin, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Tim Ricken
- Mechanics, Structural Analysis, and Dynamics, TU Dortmund University, Dortmund, Germany
| | - Jana Schleicher
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany.,Department of Bioinformatics, Friedrich Schiller University Jena, Jena, Germany
| | | | - Sebastian Vlaic
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Navina Waschinsky
- Mechanics, Structural Analysis, and Dynamics, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
18
|
Li M, Gehring R, Riviere JE, Lin Z. Development and application of a population physiologically based pharmacokinetic model for penicillin G in swine and cattle for food safety assessment. Food Chem Toxicol 2017. [DOI: 10.1016/j.fct.2017.06.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
19
|
Delannée V, Langouët S, Théret N, Siegel A. A modeling approach to evaluate the balance between bioactivation and detoxification of MeIQx in human hepatocytes. PeerJ 2017; 5:e3703. [PMID: 28879062 PMCID: PMC5582613 DOI: 10.7717/peerj.3703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/27/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Heterocyclic aromatic amines (HAA) are environmental and food contaminants that are potentially carcinogenic for humans. 2-Amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) is one of the most abundant HAA formed in cooked meat. MeIQx is metabolized by cytochrome P450 1A2 in the human liver into detoxificated and bioactivated products. Once bioactivated, MeIQx metabolites can lead to DNA adduct formation responsible for further genome instability. METHODS Using a computational approach, we developed a numerical model for MeIQx metabolism in the liver that predicts the MeIQx biotransformation into detoxification or bioactivation pathways according to the concentration of MeIQx. RESULTS Our results demonstrate that (1) the detoxification pathway predominates, (2) the ratio between detoxification and bioactivation pathways is not linear and shows a maximum at 10 µM of MeIQx in hepatocyte cell models, and (3) CYP1A2 is a key enzyme in the system that regulates the balance between bioactivation and detoxification. Our analysis suggests that such a ratio could be considered as an indicator of MeIQx genotoxicity at a low concentration of MeIQx. CONCLUSIONS Our model permits the investigation of the balance between bioactivation (i.e., DNA adduct formation pathway through the prediction of potential genotoxic compounds) and detoxification of MeIQx in order to predict the behaviour of this environmental contaminant in the human liver. It highlights the importance of complex regulations of enzyme competitions that should be taken into account in any further multi-organ models.
Collapse
Affiliation(s)
- Victorien Delannée
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France.,UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Sophie Langouët
- UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Nathalie Théret
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France.,UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Anne Siegel
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France
| |
Collapse
|
20
|
Ballnus B, Hug S, Hatz K, Görlitz L, Hasenauer J, Theis FJ. Comprehensive benchmarking of Markov chain Monte Carlo methods for dynamical systems. BMC SYSTEMS BIOLOGY 2017; 11:63. [PMID: 28646868 PMCID: PMC5482939 DOI: 10.1186/s12918-017-0433-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/10/2017] [Indexed: 11/12/2022]
Abstract
BACKGROUND In quantitative biology, mathematical models are used to describe and analyze biological processes. The parameters of these models are usually unknown and need to be estimated from experimental data using statistical methods. In particular, Markov chain Monte Carlo (MCMC) methods have become increasingly popular as they allow for a rigorous analysis of parameter and prediction uncertainties without the need for assuming parameter identifiability or removing non-identifiable parameters. A broad spectrum of MCMC algorithms have been proposed, including single- and multi-chain approaches. However, selecting and tuning sampling algorithms suited for a given problem remains challenging and a comprehensive comparison of different methods is so far not available. RESULTS We present the results of a thorough benchmarking of state-of-the-art single- and multi-chain sampling methods, including Adaptive Metropolis, Delayed Rejection Adaptive Metropolis, Metropolis adjusted Langevin algorithm, Parallel Tempering and Parallel Hierarchical Sampling. Different initialization and adaptation schemes are considered. To ensure a comprehensive and fair comparison, we consider problems with a range of features such as bifurcations, periodical orbits, multistability of steady-state solutions and chaotic regimes. These problem properties give rise to various posterior distributions including uni- and multi-modal distributions and non-normally distributed mode tails. For an objective comparison, we developed a pipeline for the semi-automatic comparison of sampling results. CONCLUSION The comparison of MCMC algorithms, initialization and adaptation schemes revealed that overall multi-chain algorithms perform better than single-chain algorithms. In some cases this performance can be further increased by using a preceding multi-start local optimization scheme. These results can inform the selection of sampling methods and the benchmark collection can serve for the evaluation of new algorithms. Furthermore, our results confirm the need to address exploration quality of MCMC chains before applying the commonly used quality measure of effective sample size to prevent false analysis conclusions.
Collapse
Affiliation(s)
- Benjamin Ballnus
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Ingolstädter Landstraße 1, Neuherberg, 85764 Germany
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, Boltzmannstraße 15, Garching, 85748 Germany
| | - Sabine Hug
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Ingolstädter Landstraße 1, Neuherberg, 85764 Germany
| | - Kathrin Hatz
- Bayer AG, Engineering & Technologies, Applied Mathematics, Kaiser-Wilhelm-Allee, Leverkusen, 51368 Germany
| | - Linus Görlitz
- Bayer AG, Engineering & Technologies, Applied Mathematics, Kaiser-Wilhelm-Allee, Leverkusen, 51368 Germany
| | - Jan Hasenauer
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Ingolstädter Landstraße 1, Neuherberg, 85764 Germany
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, Boltzmannstraße 15, Garching, 85748 Germany
| | - Fabian J. Theis
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Ingolstädter Landstraße 1, Neuherberg, 85764 Germany
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, Boltzmannstraße 15, Garching, 85748 Germany
| |
Collapse
|
21
|
Translational learning from clinical studies predicts drug pharmacokinetics across patient populations. NPJ Syst Biol Appl 2017. [PMID: 28649438 PMCID: PMC5460240 DOI: 10.1038/s41540-017-0012-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Early indication of late-stage failure of novel candidate drugs could be facilitated by continuous integration, assessment, and transfer of knowledge acquired along pharmaceutical development programs. We here present a translational systems pharmacology workflow that combines drug cocktail probing in a specifically designed clinical study, physiologically based pharmacokinetic modeling, and Bayesian statistics to identify and transfer (patho-)physiological and drug-specific knowledge across distinct patient populations. Our work builds on two clinical investigations, one with 103 healthy volunteers and one with 79 diseased patients from which we systematically derived physiological information from pharmacokinetic data for a reference probe drug (midazolam) at the single-patient level. Taking into account the acquired knowledge describing (patho-)physiological alterations in the patient cohort allowed the successful prediction of the population pharmacokinetics of a second, candidate probe drug (torsemide) in the patient population. In addition, we identified significant relations of the acquired physiological processes to patient metadata from liver biopsies. The presented prototypical systems pharmacology approach is a proof of concept for model-based translation across different stages of pharmaceutical development programs. Applied consistently, it has the potential to systematically improve predictivity of pharmacokinetic simulations by incorporating the results of clinical trials and translating them to subsequent studies. Physiologically based modeling together with Bayesian statistics allows the prediction of drug pharmacokinetics in specific patient populations. An interdisciplinary group of clinicians and computational scientists led by Dr. Lars Kuepfer from Bayer developed a generic workflow consisting of several consecutive learning steps where knowledge about both individual physiology as well as drug physicochemistry can be efficiently derived from plasma concentration profiles. The acquired information is then be used for the prediction of the pharmacokinetic behavior of a new drug candidate in a diseased population. This allows to simulate the variability in drug exposure virtually before starting clinical investigation in real patients in order to evaluate drug safety or efficacy through the simulation of virtual populations. Further development of this workflow could improve the safety of clinical development programs to assess the risk-benefit ratio of novel drug candidates in silico.
Collapse
|
22
|
Kuepfer L, Niederalt C, Wendl T, Schlender JF, Willmann S, Lippert J, Block M, Eissing T, Teutonico D. Applied Concepts in PBPK Modeling: How to Build a PBPK/PD Model. CPT Pharmacometrics Syst Pharmacol 2016; 5:516-531. [PMID: 27653238 PMCID: PMC5080648 DOI: 10.1002/psp4.12134] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this tutorial is to introduce the fundamental concepts of physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling with a special focus on their practical implementation in a typical PBPK model building workflow. To illustrate basic steps in PBPK model building, a PBPK model for ciprofloxacin will be constructed and coupled to a pharmacodynamic model to simulate the antibacterial activity of ciprofloxacin treatment.
Collapse
Affiliation(s)
- L Kuepfer
- Bayer Technology Services, Leverkusen, Germany
| | - C Niederalt
- Bayer Technology Services, Leverkusen, Germany
| | - T Wendl
- Bayer Technology Services, Leverkusen, Germany
| | | | | | - J Lippert
- Bayer HealthCare, Wuppertal, Germany
| | - M Block
- Bayer Technology Services, Leverkusen, Germany
| | - T Eissing
- Bayer Technology Services, Leverkusen, Germany
| | - D Teutonico
- Bayer Technology Services, Leverkusen, Germany.
| |
Collapse
|
23
|
Abstract
The development of new drug therapies requires substantial and ever increasing investments from the pharmaceutical company. Ten years ago, the average time from early target identification and optimization until initial market authorization of a new drug compound took more than 10 years and involved costs in the order of one billion US dollars. Recent studies indicate even a significant growth of costs in the meanwhile, mainly driven by the increasing complexity of diseases addressed by pharmaceutical research.Modeling and simulation are proven approaches to handle highly complex systems; hence, systems medicine is expected to control the spiral of complexity of diseases and increasing costs. Today, the main focus of systems medicine applications in industry is on mechanistic modeling. Biological mechanisms are represented by explicit equations enabling insight into the cooperation of all relevant mechanisms. Mechanistic modeling is widely accepted in pharmacokinetics, but prediction from cell behavior to patients is rarely possible due to lacks in our understanding of the controlling mechanisms. Data-driven modeling aims to compensate these lacks by the use of advanced statistical and machine learning methods. Future progress in pharmaceutical research and development will require integrated hybrid modeling technologies allowing realization of the benefits of both mechanistic and data-driven modeling. In this chapter, we sketch typical industrial application areas for both modeling techniques and derive the requirements for future technology development.
Collapse
Affiliation(s)
- Lars Kuepfer
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
- Institute of Applied Microbiology, RWTH Aachen University, Aachen, Germany
| | - Andreas Schuppert
- Lehrstuhl für datenbasierte Modellierung in CES, Joint Research Center for Computational Biomedicine, AICES, RWTH Aachen University, Augustinerbach 2, Aachen, 52062, Germany.
| |
Collapse
|
24
|
Krauss M, Schuppert A. Assessing interindividual variability by Bayesian-PBPK modeling. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.ddmod.2017.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Tsamandouras N, Rostami-Hodjegan A, Aarons L. Combining the 'bottom up' and 'top down' approaches in pharmacokinetic modelling: fitting PBPK models to observed clinical data. Br J Clin Pharmacol 2015; 79:48-55. [PMID: 24033787 DOI: 10.1111/bcp.12234] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/22/2013] [Indexed: 01/07/2023] Open
Abstract
Pharmacokinetic models range from being entirely exploratory and empirical, to semi-mechanistic and ultimately complex physiologically based pharmacokinetic (PBPK) models. This choice is conditional on the modelling purpose as well as the amount and quality of the available data. The main advantage of PBPK models is that they can be used to extrapolate outside the studied population and experimental conditions. The trade-off for this advantage is a complex system of differential equations with a considerable number of model parameters. When these parameters cannot be informed from in vitro or in silico experiments they are usually optimized with respect to observed clinical data. Parameter estimation in complex models is a challenging task associated with many methodological issues which are discussed here with specific recommendations. Concepts such as structural and practical identifiability are described with regards to PBPK modelling and the value of experimental design and sensitivity analyses is sketched out. Parameter estimation approaches are discussed, while we also highlight the importance of not neglecting the covariance structure between model parameters and the uncertainty and population variability that is associated with them. Finally the possibility of using model order reduction techniques and minimal semi-mechanistic models that retain the physiological-mechanistic nature only in the parts of the model which are relevant to the desired modelling purpose is emphasized. Careful attention to all the above issues allows us to integrate successfully information from in vitro or in silico experiments together with information deriving from observed clinical data and develop mechanistically sound models with clinical relevance.
Collapse
Affiliation(s)
- Nikolaos Tsamandouras
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
26
|
Zurlinden TJ, Heard K, Reisfeld B. A novel approach for estimating ingested dose associated with paracetamol overdose. Br J Clin Pharmacol 2015; 81:634-45. [PMID: 26441245 DOI: 10.1111/bcp.12796] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/04/2015] [Accepted: 10/01/2015] [Indexed: 11/28/2022] Open
Abstract
AIM In cases of paracetamol (acetaminophen, APAP) overdose, an accurate estimate of tissue-specific paracetamol pharmacokinetics (PK) and ingested dose can offer health care providers important information for the individualized treatment and follow-up of affected patients. Here a novel methodology is presented to make such estimates using a standard serum paracetamol measurement and a computational framework. METHODS The core component of the computational framework was a physiologically-based pharmacokinetic (PBPK) model developed and evaluated using an extensive set of human PK data. Bayesian inference was used for parameter and dose estimation, allowing the incorporation of inter-study variability, and facilitating the calculation of uncertainty in model outputs. RESULTS Simulations of paracetamol time course concentrations in the blood were in close agreement with experimental data under a wide range of dosing conditions. Also, predictions of administered dose showed good agreement with a large collection of clinical and emergency setting PK data over a broad dose range. In addition to dose estimation, the platform was applied for the determination of optimal blood sampling times for dose reconstruction and quantitation of the potential role of paracetamol conjugate measurement on dose estimation. CONCLUSIONS Current therapies for paracetamol overdose rely on a generic methodology involving the use of a clinical nomogram. By using the computational framework developed in this study, serum sample data, and the individual patient's anthropometric and physiological information, personalized serum and liver pharmacokinetic profiles and dose estimate could be generated to help inform an individualized overdose treatment and follow-up plan.
Collapse
Affiliation(s)
- Todd J Zurlinden
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523-1370
| | - Kennon Heard
- Department of Emergency Medicine, University of Colorado School of Medicine, 12401 E. 17th Avenue Campus Box B-215, Aurora, CO, 80045.,Rocky Mountain Poison and Drug Center, Denver, CO, 80204
| | - Brad Reisfeld
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523-1370.,School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523-1376, USA
| |
Collapse
|
27
|
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. [PMID: 26431198 PMCID: PMC4592188 DOI: 10.1371/journal.pone.0139423] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 09/14/2015] [Indexed: 01/26/2023] Open
Abstract
Interindividual variability in anatomical and physiological properties results in significant differences in drug pharmacokinetics. The consideration of such pharmacokinetic variability supports optimal drug efficacy and safety for each single individual, e.g. by identification of individual-specific dosings. One clear objective in clinical drug development is therefore a thorough characterization of the physiological sources of interindividual variability. In this work, we present a Bayesian population physiologically-based pharmacokinetic (PBPK) approach for the mechanistically and physiologically realistic identification of interindividual variability. The consideration of a generic and highly detailed mechanistic PBPK model structure enables the integration of large amounts of prior physiological knowledge, which is then updated with new experimental data in a Bayesian framework. A covariate model integrates known relationships of physiological parameters to age, gender and body height. We further provide a framework for estimation of the a posteriori parameter dependency structure at the population level. The approach is demonstrated considering a cohort of healthy individuals and theophylline as an application example. The variability and co-variability of physiological parameters are specified within the population; respectively. Significant correlations are identified between population parameters and are applied for individual- and population-specific visual predictive checks of the pharmacokinetic behavior, which leads to improved results compared to present population approaches. In the future, the integration of a generic PBPK model into an hierarchical approach allows for extrapolations to other populations or drugs, while the Bayesian paradigm allows for an iterative application of the approach and thereby a continuous updating of physiological knowledge with new data. This will facilitate decision making e.g. from preclinical to clinical development or extrapolation of PK behavior from healthy to clinically significant populations.
Collapse
Affiliation(s)
- Markus Krauss
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany; Aachen Institute for Advanced Study in Computational Engineering Sciences, RWTH Aachen, Aachen, Germany
| | - Kai Tappe
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Andreas Schuppert
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany; Aachen Institute for Advanced Study in Computational Engineering Sciences, RWTH Aachen, Aachen, Germany
| | - Lars Kuepfer
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Linus Goerlitz
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| |
Collapse
|
28
|
Drasdo D, Bode J, Dahmen U, Dirsch O, Dooley S, Gebhardt R, Ghallab A, Godoy P, Häussinger D, Hammad S, Hoehme S, Holzhütter HG, Klingmüller U, Kuepfer L, Timmer J, Zerial M, Hengstler JG. The virtual liver: state of the art and future perspectives. Arch Toxicol 2015; 88:2071-5. [PMID: 25331938 DOI: 10.1007/s00204-014-1384-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Dirk Drasdo
- Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau - Rocquencourt, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stader F, Wuerthwein G, Groll AH, Vehreschild JJ, Cornely OA, Hempel G. Physiology-Based Pharmacokinetics of Caspofungin for Adults and Paediatrics. Pharm Res 2014; 32:2029-37. [DOI: 10.1007/s11095-014-1595-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022]
|
30
|
Tsamandouras N, Dickinson G, Guo Y, Hall S, Rostami-Hodjegan A, Galetin A, Aarons L. Development and Application of a Mechanistic Pharmacokinetic Model for Simvastatin and its Active Metabolite Simvastatin Acid Using an Integrated Population PBPK Approach. Pharm Res 2014; 32:1864-83. [PMID: 25446771 DOI: 10.1007/s11095-014-1581-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Abstract
PURPOSE To develop a population physiologically-based pharmacokinetic (PBPK) model for simvastatin (SV) and its active metabolite, simvastatin acid (SVA), that allows extrapolation and prediction of their concentration profiles in liver (efficacy) and muscle (toxicity). METHODS SV/SVA plasma concentrations (34 healthy volunteers) were simultaneously analysed with NONMEM 7.2. The implemented mechanistic model has a complex compartmental structure allowing inter-conversion between SV and SVA in different tissues. Prior information for model parameters was extracted from different sources to construct appropriate prior distributions that support parameter estimation. The model was employed to provide predictions regarding the effects of a range of clinically important conditions on the SV and SVA disposition. RESULTS The developed model offered a very good description of the available plasma SV/SVA data. It was also able to describe previously observed effects of an OATP1B1 polymorphism (c.521 T > C) and a range of drug-drug interactions (CYP inhibition) on SV/SVA plasma concentrations. The predicted SV/SVA liver and muscle tissue concentrations were in agreement with the clinically observed efficacy and toxicity outcomes of the investigated conditions. CONCLUSIONS A mechanistically sound SV/SVA population model with clinical applications (e.g., assessment of drug-drug interaction and myopathy risk) was developed, illustrating the advantages of an integrated population PBPK approach.
Collapse
Affiliation(s)
- Nikolaos Tsamandouras
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Stopford Building, Room 3.32, Oxford Road, Manchester, M13 9PT, UK,
| | | | | | | | | | | | | |
Collapse
|
31
|
Selen A, Dickinson PA, Müllertz A, Crison JR, Mistry HB, Cruañes MT, Martinez MN, Lennernäs H, Wigal TL, Swinney DC, Polli JE, Serajuddin AT, Cook JA, Dressman JB. The Biopharmaceutics Risk Assessment Roadmap for Optimizing Clinical Drug Product Performance. J Pharm Sci 2014; 103:3377-3397. [DOI: 10.1002/jps.24162] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 02/06/2023]
|
32
|
Clinical translation in the virtual liver network. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e127. [PMID: 25076067 PMCID: PMC4120019 DOI: 10.1038/psp.2014.25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/22/2014] [Indexed: 02/04/2023]
Abstract
The liver is the central detoxifying organ, continuously removing xenobiotics from the vascular system. Given its role in drug metabolism, a functional understanding of liver physiology is crucial to optimizing drug efficacy and patient safety. The Virtual Liver Network (VLN), a German national flagship research program, focuses on producing validated computer models of human liver physiology. These models are used to analyze patient-derived data and thereby gain mechanistic insights in the processes underlying drug pharmacokinetics (PK).
Collapse
|
33
|
Lahoz-Beneytez J, Schnizler K, Eissing T. A pharma perspective on the systems medicine and pharmacology of inflammation. Math Biosci 2014; 260:2-5. [PMID: 25057776 DOI: 10.1016/j.mbs.2014.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/10/2014] [Indexed: 10/25/2022]
Abstract
Biological systems are complex and comprehend multiple scales of organisation. Hence, holistic approaches are necessary to capture the behaviour of these entities from the molecular and cellular to the whole organism level. This also applies to the understanding and treatment of different diseases. Traditional systems biology has been successful in describing different biological phenomena at the cellular level, but it still lacks of a holistic description of the multi-scale interactions within the body. The importance of the physiological context is of particular interest in inflammation. Regulatory agencies have urged the scientific community to increase the translational power of bio-medical research and it has been recognised that modelling and simulation could be a path to follow. Interestingly, in pharma R&D, modelling and simulation has been employed since a long time ago. Systems pharmacology, and particularly physiologically based pharmacokinetic/pharmacodynamic models, serve as a suitable framework to integrate the available and emerging knowledge at different levels of the drug development process. Systems medicine and pharmacology of inflammation will potentially benefit from this framework in order to better understand inflammatory diseases and to help to transfer the vast knowledge on the molecular and cellular level into a more physiological context. Ultimately, this may lead to reliable predictions of clinical outcomes such as disease progression or treatment efficacy, contributing thereby to a better care of patients.
Collapse
Affiliation(s)
- Julio Lahoz-Beneytez
- Bayer Technology Services GmbH, Computational Systems Biology, Leverkusen 51368, Germany.
| | - Katrin Schnizler
- Bayer Technology Services GmbH, Computational Systems Biology, Leverkusen 51368, Germany.
| | - Thomas Eissing
- Bayer Technology Services GmbH, Computational Systems Biology, Leverkusen 51368, Germany.
| |
Collapse
|
34
|
Schaeftlein A, Minichmayr IK, Kloft C. Population pharmacokinetics meets microdialysis: Benefits, pitfalls and necessities of new analysis approaches for human microdialysis data. Eur J Pharm Sci 2014; 57:68-73. [DOI: 10.1016/j.ejps.2013.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
|
35
|
Bois FY. Computational pharmacokinetics at a crossroads. In Silico Pharmacol 2013; 1:5. [PMID: 25505650 PMCID: PMC4230653 DOI: 10.1186/2193-9616-1-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 02/20/2013] [Indexed: 01/07/2023] Open
Abstract
This first special issue of In Silico Pharmacology focuses on computational pharmacokinetics since they are an important part of integrated applications in computational pharmacology. The important topics of model structure, model parameterization, improved organ description, and modeling of drug-drug interactions are covered. They are actually at the crossroads between several emerging disciplines which will shape the future of therapeutic treatments and public health.
Collapse
Affiliation(s)
- Frédéric Y Bois
- DRC/VIVA/METO, INERIS and Chair of Mathematical Modeling for Systems Toxicology, Technological University of Compiegne, Compiegne, France
| |
Collapse
|