Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (256)

Search Parameters:
Keywords = physiologically based pharmacokinetic (PBPK) modelling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4756 KiB  
Article
Quasi-3D Mechanistic Model for Predicting Eye Drop Distribution in the Human Tear Film
by Harsha T. Garimella, Carly Norris, Carrie German, Andrzej Przekwas, Ross Walenga, Andrew Babiskin and Ming-Liang Tan
Bioengineering 2025, 12(8), 825; https://doi.org/10.3390/bioengineering12080825 - 30 Jul 2025
Viewed by 216
Abstract
Topical drug administration is a common method of delivering medications to the eye to treat various ocular conditions, including glaucoma, dry eye, and inflammation. Drug efficacy following topical administration, including the drug’s distribution within the eye, absorption and elimination rates, and physiological responses [...] Read more.
Topical drug administration is a common method of delivering medications to the eye to treat various ocular conditions, including glaucoma, dry eye, and inflammation. Drug efficacy following topical administration, including the drug’s distribution within the eye, absorption and elimination rates, and physiological responses can be predicted using physiologically based pharmacokinetic (PBPK) modeling. High-resolution computational models of the eye are desirable to improve simulations of drug delivery; however, these approaches can have long run times. In this study, a fast-running computational quasi-3D (Q3D) model of the human tear film was developed to account for absorption, blinking, drainage, and evaporation. Visualization of blinking mechanics and flow distributions throughout the tear film were enabled using this Q3D approach. Average drug absorption throughout the tear film subregions was quantified using a high-resolution compartment model based on a system of ordinary differential equations (ODEs). Simulations were validated by comparing them with experimental data from topical administration of 0.1% dexamethasone suspension in the tear film (R2 = 0.76, RMSE = 8.7, AARD = 28.8%). Overall, the Q3D tear film model accounts for critical mechanistic factors (e.g., blinking and drainage) not previously included in fast-running models. Further, this work demonstrated methods toward improved computational efficiency, where central processing unit (CPU) time was decreased while maintaining accuracy. Building upon this work, this Q3D approach applied to the tear film will allow for more seamless integration into full-body models, which will be an extremely valuable tool in the development of treatments for ocular conditions. Full article
Show Figures

Figure 1

18 pages, 4976 KiB  
Article
Mechanistic Insights into Cytokine Antagonist-Drug Interactions: A Physiologically Based Pharmacokinetic Modelling Approach with Tocilizumab as a Case Study
by Xian Pan, Cong Liu, Felix Stader, Abdallah Derbalah, Masoud Jamei and Iain Gardner
Pharmaceutics 2025, 17(7), 896; https://doi.org/10.3390/pharmaceutics17070896 - 10 Jul 2025
Viewed by 557
Abstract
Background: Understanding interactions between cytokine antagonists and drugs is essential for effective medication management in inflammatory conditions. Recent regulatory authority guidelines emphasise a systematic, risk-based approach to evaluating these interactions, underscoring the need for mechanistic insight. Proinflammatory cytokines, such as interleukin-6 (IL-6), modulate [...] Read more.
Background: Understanding interactions between cytokine antagonists and drugs is essential for effective medication management in inflammatory conditions. Recent regulatory authority guidelines emphasise a systematic, risk-based approach to evaluating these interactions, underscoring the need for mechanistic insight. Proinflammatory cytokines, such as interleukin-6 (IL-6), modulate cytochrome P450 (CYP) enzymes, reducing the metabolism of CYP substrates. Cytokine antagonists (such as IL-6 receptor antagonists) can counteract this effect, restoring CYP activity and increasing drug clearance. However, quantitative prediction of cytokine-mediated drug interactions remains challenging, as existing models often lack the mechanistic detail needed to capture the dynamic relationship between cytokine signalling, receptor engagement, and downstream modulation of drug metabolism. Methods: A physiologically based pharmacokinetic (PBPK) framework incorporating cytokine–receptor binding, subsequent downregulation of CYP expression, and blockade of the cytokine signalling by a therapeutic protein antagonist was developed to simulate and investigate cytokine antagonist-drug interactions. Tocilizumab, a humanised IL-6 receptor antagonist used to treat several inflammatory conditions associated with elevated IL-6 levels, was selected as a model drug to demonstrate the utility of the framework. Results: The developed PBPK model accurately predicted the pharmacokinetics profiles of tocilizumab and captured clinically observed dynamic changes in simvastatin exposure before and after tocilizumab treatment in rheumatoid arthritis (RA) patients. Simulated IL-6 dynamics aligned with observed clinical profiles, showing transient elevation following receptor blockade and associated restoration of CYP3A4 activity. Prospective simulations with commonly co-administered CYP substrates (celecoxib, chloroquine, cyclosporine, ibuprofen, prednisone, simvastatin, and theophylline) in RA patients revealed dose regimen- and drug-dependent differences in interaction magnitude. Conclusions: This study demonstrated the utility of PBPK models in providing a mechanistic understanding of cytokine antagonist-drug interactions, supporting enhanced therapeutic decision-making and optimising patient care in inflammatory conditions. Full article
Show Figures

Figure 1

15 pages, 1404 KiB  
Article
Physiologically Based Pharmacokinetic Modeling for Predicting Drug Levels After Bariatric Surgery: Vardenafil Exposure Before vs. After Gastric Sleeve/Bypass
by Daniel Porat, Oleg Dukhno, Sandra Cvijić and Arik Dahan
Biomolecules 2025, 15(7), 975; https://doi.org/10.3390/biom15070975 - 7 Jul 2025
Viewed by 393
Abstract
Bariatric surgery involves major changes in the anatomy and physiology of the gastrointestinal tract, which may alter oral drug bioavailability and efficacy. Phosphodiesterase-5 inhibitor (PDE5i) drugs are the first-line treatment of erectile dysfunction, a condition associated with a higher BMI. In this paper, [...] Read more.
Bariatric surgery involves major changes in the anatomy and physiology of the gastrointestinal tract, which may alter oral drug bioavailability and efficacy. Phosphodiesterase-5 inhibitor (PDE5i) drugs are the first-line treatment of erectile dysfunction, a condition associated with a higher BMI. In this paper, we examine the PDE5i vardenafil for possible post-bariatric changes in solubility/dissolution and absorption. Vardenafil solubility was determined in vitro, as well as ex vivo using aspirated gastric contents from patients prior to vs. following bariatric procedures. Dissolution was tested in vitro under unoperated stomach vs. post-gastric sleeve/bypass conditions. Lastly, the gathered solubility/dissolution data were used to produce an in silico physiologically based pharmacokinetic (PBPK) model (GastroPlus®), where gastric volume, pH, and transit time, as well as proximal GI bypass (when relevant) were all adjusted for, evaluating vardenafil dissolution, gastrointestinal compartmental absorption, and pharmacokinetics before vs. after different bariatric procedures. pH-dependent solubility was demonstrated for vardenafil with low (pH 7) vs. high solubility (pH 1–5), which was confirmed ex vivo. The impaired dissolution of all vardenafil doses under post-gastric bypass conditions was demonstrated, contrary to complete (100%) dissolution under pre-surgery and post-sleeve gastrectomy conditions. Compared to unoperated individuals, PBPK simulations revealed altered pharmacokinetics post-gastric bypass (but not after sleeve gastrectomy), with 30% lower peak plasma concentration (Cmax) and 40% longer time to Cmax (Tmax). Complete absorption after gastric bypass is predicted for vardenafil, which is attributable to significant absorption from the large intestine. The biopharmaceutics and PBPK analysis indicate that vardenafil may be similarly effective after sleeve gastrectomy as before the procedure. However, results after gastric bypass question the effectiveness of this PDE5i. Specifically, vardenafil’s onset of action might be delayed and unpredictable, negatively affecting the practicality of the intended use. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

15 pages, 1142 KiB  
Article
Prediction of Monoclonal Antibody Pharmacokinetics in Pediatric Populations Using PBPK Modeling and Simulation
by Chiara Zunino, Virginie Gualano, Haiying Zhou, Viera Lukacova and Maxime Le Merdy
Pharmaceutics 2025, 17(7), 884; https://doi.org/10.3390/pharmaceutics17070884 - 5 Jul 2025
Viewed by 571
Abstract
Background: Accurately determining pediatric dosing is essential prior to initiating clinical trials or administering medications in routine clinical settings. In children, ethical considerations demand careful evaluation of both safety and effectiveness. Typically, dosing recommendations for therapeutic proteins, such as monoclonal antibodies (mAbs), [...] Read more.
Background: Accurately determining pediatric dosing is essential prior to initiating clinical trials or administering medications in routine clinical settings. In children, ethical considerations demand careful evaluation of both safety and effectiveness. Typically, dosing recommendations for therapeutic proteins, such as monoclonal antibodies (mAbs), are derived from adult dosages using body weight as a scaling factor. However, this method overlooks key physiological and biochemical distinctions between pediatric and adult patients. Therefore, this could lead to the underexposure of mAbs, limiting their efficacy in this population. Additional methods are necessary to predict pediatric doses mechanistically. For small molecules, physiologically based pharmacokinetic (PBPK) models have been extensively used to predict pediatric doses based on physiological age-related changes and enzymes/transporters ontogeny. This study aims to evaluate the ability of PBPK models to predict mAbs’ pediatric exposure. Methods: Three mAbs were used for model development and validation: bevacizumab, infliximab, and atezolizumab. The PBPK models were built using GastroPlus© Biologics module. For each mAb, the PBPK model was developed based on observed data in healthy and/or patient adults. Then, the physiological parameters were scaled to describe the pediatric physiology to predict exposure to the pediatric populations. Predicted plasma concentration–time courses were overlaid with reported observed data to assess the ability of the PBPK model to predict pediatric exposure. Results: Results showed that PBPK models accurately predicted pediatric pharmacokinetics for mAbs. Conclusions: This research marks a significant step in validating mechanistic extrapolation methods for biologics exposure prediction in children using PBPK models. Full article
Show Figures

Figure 1

25 pages, 2704 KiB  
Article
A Parent–Metabolite Middle-Out PBPK Model for Genistein and Its Glucuronide Metabolite in Rats: Integrating Liver and Enteric Metabolism with Hepatobiliary and Enteroluminal Transport to Assess Glucuronide Recycling
by Bhargavi Srija Ramisetty, Rashim Singh, Ming Hu and Michael Zhuo Wang
Pharmaceutics 2025, 17(7), 814; https://doi.org/10.3390/pharmaceutics17070814 - 23 Jun 2025
Viewed by 474
Abstract
Background: Glucuronide recycling in the gut and liver profoundly affects the systemic and/or local exposure of drugs and their glucuronide metabolites, impacting both clinical efficacy and toxicity. This recycling also alters drug exposure in the colon, making it critical to establish local [...] Read more.
Background: Glucuronide recycling in the gut and liver profoundly affects the systemic and/or local exposure of drugs and their glucuronide metabolites, impacting both clinical efficacy and toxicity. This recycling also alters drug exposure in the colon, making it critical to establish local concentration for drugs targeting colon (e.g., drugs for colon cancer and inflammatory bowel disease). Methods: In this study, a parent–metabolite middle-out physiologically based pharmacokinetic (PBPK) model was built for genistein and its glucuronide metabolite to estimate the systemic and local exposure of the glucuronide and its corresponding aglycone in rats by incorporating UDP-glucuronosyltransferase (UGT)-mediated metabolism and transporter-dependent glucuronide disposition in the liver and intestine, as well as gut microbial-mediated deglucuronidation that enables the recycling of the parent compound. Results: This parent–metabolite middle-out rat PBPK model utilized in vitro-to-in vivo extrapolated (IVIVE) metabolic and transporter clearance values based on in vitro kinetic parameters from surrogate species, the rat tissue abundance of relevant proteins, and saturable Michaelis–Menten mechanisms. Inter-system extrapolation factors (ISEFs) were used to account for transporter protein abundance differences between in vitro systems and tissues and between rats and surrogate species. Model performance was evaluated at multiple dose levels for genistein and its glucuronide. Model sensitivity analyses demonstrated the impact of key parameters on the plasma concentrations and local exposure of genistein and its glucuronide. Our model was applied to simulate the quantitative impact of glucuronide recycling on the pharmacokinetic profiles in both plasma and colonocytes. Conclusions: Our study underlines the importance of glucuronide recycling in determining local drug concentrations in the intestine and provides a preliminary modeling tool to assess the influence of transporter-mediated drug–drug interactions on glucuronide recycling and local drug exposure, which are often misrepresented by systemic plasma concentrations. Full article
(This article belongs to the Special Issue Development of Physiologically Based Pharmacokinetic (PBPK) Modeling)
Show Figures

Graphical abstract

20 pages, 7892 KiB  
Article
Tissue Distribution and Pharmacokinetic Characteristics of Aztreonam Based on Multi-Species PBPK Model
by Xiao Ye, Xiaolong Sun, Jianing Zhang, Min Yu, Nie Wen, Xingchao Geng and Ying Liu
Pharmaceutics 2025, 17(6), 748; https://doi.org/10.3390/pharmaceutics17060748 - 6 Jun 2025
Viewed by 698
Abstract
Background/Objectives: As a monocyclic β-lactam antibiotic, aztreonam has regained attention recently because combining it with β-lactamase inhibitors helps fight drug-resistant bacteria. This study aimed to systematically characterize the plasma and tissue concentration-time profiles of aztreonam in rats, mice, dogs, monkeys, and humans [...] Read more.
Background/Objectives: As a monocyclic β-lactam antibiotic, aztreonam has regained attention recently because combining it with β-lactamase inhibitors helps fight drug-resistant bacteria. This study aimed to systematically characterize the plasma and tissue concentration-time profiles of aztreonam in rats, mice, dogs, monkeys, and humans by developing a multi-species, physiologically based pharmacokinetic (PBPK) model. Methods: A rat PBPK model was optimized and validated using plasma concentration-time curves determined by liquid chromatography–tandem mass spectrometry (LC-MS/MS) following intravenous administration, with reliability confirmed through another dose experiment. The rat model characteristics, modeling experience, ADMET Predictor (11.0) software prediction results, and allometric scaling were used to extrapolate to mouse, human, dog, and monkey models. The tissue-to-plasma partition coefficients (Kp values) were predicted using GastroPlus (9.0) software, and the sensitivity analyses of key parameters were evaluated. Finally, the cross-species validation was performed using the average fold error (AFE) and absolute relative error (ARE). Results: The cross-species validation showed that the model predictions were highly consistent with the experimental data (AFE < 2, ARE < 30%), but the deviation of the volume of distribution (Vss) in dogs and monkeys suggested the need to supplement the species-specific parameters to optimize the prediction accuracy. The Kp values revealed a high distribution of aztreonam in the kidneys (Kp = 2.0–3.0), which was consistent with its clearance mechanism dominated by renal excretion. Conclusions: The PBPK model developed in this study can be used to predict aztreonam pharmacokinetics across species, elucidating its renal-targeted distribution and providing key theoretical support for the clinical dose optimization of aztreonam, the assessment of target tissue exposure in drug-resistant bacterial infections, and the development of combination therapy strategies. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Figure 1

37 pages, 1088 KiB  
Review
A Review on New Frontiers in Drug-Drug Interaction Predictions and Safety Evaluations with In Vitro Cellular Models
by Lara Marques and Nuno Vale
Pharmaceutics 2025, 17(6), 747; https://doi.org/10.3390/pharmaceutics17060747 - 6 Jun 2025
Viewed by 1179
Abstract
The characterization of a drug’s ADME (absorption, distribution, metabolism, and excretion) profile is crucial for accurately determining its safety and efficacy. The rising prevalence of polypharmacy has significantly increased the risk of drug-drug interactions (DDIs). These interactions can lead to altered drug exposure, [...] Read more.
The characterization of a drug’s ADME (absorption, distribution, metabolism, and excretion) profile is crucial for accurately determining its safety and efficacy. The rising prevalence of polypharmacy has significantly increased the risk of drug-drug interactions (DDIs). These interactions can lead to altered drug exposure, potentially compromising efficacy or increasing the risk of adverse drug reactions (ADRs), thereby posing significant clinical and regulatory concerns. Traditional methods for assessing potential DDIs rely heavily on in vitro models, including enzymatic assays and transporter studies. While indispensable, these approaches have inherent limitations in scalability, cost, and ability to predict complex interactions. Recent advancements in analytical technologies, particularly the development of more sophisticated cellular models and computational modeling, have paved the way for more accurate and efficient DDI assessments. Emerging methodologies, such as organoids, physiologically based pharmacokinetic (PBPK) modeling, and artificial intelligence (AI), demonstrate significant potential in this field. A powerful and increasingly adopted approach is the integration of in vitro data with in silico modeling, which can lead to better in vitro-in vivo extrapolation (IVIVE). This review provides a comprehensive overview of both conventional and novel strategies for DDI predictions, highlighting their strengths and limitations. Equipping researchers with a structured framework for selecting optimal methodologies improves safety and efficacy evaluation and regulatory decision-making and deepens the understanding of DDIs. Full article
Show Figures

Figure 1

16 pages, 596 KiB  
Review
Targeted but Troubling: CYP450 Inhibition by Kinase and PARP Inhibitors and Its Clinical Implications
by Martin Kondža, Josipa Bukić, Ivan Ćavar and Biljana Tubić
Drugs Drug Candidates 2025, 4(2), 24; https://doi.org/10.3390/ddc4020024 - 26 May 2025
Viewed by 1165
Abstract
Cytochrome P450 (CYP450) enzymes are pivotal in the metabolism of numerous anticancer agents, with CYP3A4 being the predominant isoform involved. Inhibition of CYP450 enzymes is a major mechanism underlying clinically significant drug-drug interactions (DDIs), particularly in oncology, where polypharmacy is frequent. This review [...] Read more.
Cytochrome P450 (CYP450) enzymes are pivotal in the metabolism of numerous anticancer agents, with CYP3A4 being the predominant isoform involved. Inhibition of CYP450 enzymes is a major mechanism underlying clinically significant drug-drug interactions (DDIs), particularly in oncology, where polypharmacy is frequent. This review aims to provide a comprehensive and critical overview of CYP450 enzyme inhibition, focusing specifically on the impact of kinase inhibitors (KIs) and poly adenosine diphosphate-ribose polymerase (PARP) inhibitors. A systematic review of the current literature was conducted, focusing on the molecular mechanisms of CYP450 inhibition, including reversible, time-dependent, mechanism-based, and pseudo-irreversible inhibition. Specific attention was given to the inhibitory profiles of clinically relevant KIs and PARP inhibitors, with analysis of pharmacokinetic consequences and regulatory considerations. Many KIs, such as abemaciclib and ibrutinib, demonstrate time-dependent or quasi-irreversible inhibition of CYP3A4, while PARP inhibitors like olaparib and rucaparib exhibit moderate reversible and time-dependent CYP3A4 inhibition. These inhibitory activities can significantly alter the pharmacokinetics of co-administered drugs, leading to increased risk of toxicity or therapeutic failure. Regulatory guidelines now recommend early identification of time-dependent and mechanism-based inhibition using physiologically based pharmacokinetic) (PBPK) modeling. CYP450 inhibition by KIs and PARP inhibitors represents a critical but often underappreciated challenge in oncology pharmacotherapy. Understanding the mechanistic basis of these interactions is essential for optimizing treatment regimens, improving patient safety, and supporting personalized oncology care. Greater clinical vigilance and the integration of predictive modeling tools are necessary to mitigate the risks associated with CYP-mediated DDIs. Full article
(This article belongs to the Section Marketed Drugs)
Show Figures

Figure 1

18 pages, 513 KiB  
Review
Physiologically Based Pharmacokinetic Modeling of Antibiotics in Children: Perspectives on Model-Informed Precision Dosing
by Ryota Tanaka, Kei Irie and Tomoyuki Mizuno
Antibiotics 2025, 14(6), 541; https://doi.org/10.3390/antibiotics14060541 - 24 May 2025
Viewed by 1365
Abstract
The appropriate use of antibiotics is crucial and involves selecting an optimal dosing regimen based on pharmacokinetic (PK) and pharmacodynamic (PD) indicators. Physiologically based pharmacokinetic (PBPK) modeling is a powerful tool that integrates drugs’ physicochemical properties with anatomical and physiological data to predict [...] Read more.
The appropriate use of antibiotics is crucial and involves selecting an optimal dosing regimen based on pharmacokinetic (PK) and pharmacodynamic (PD) indicators. Physiologically based pharmacokinetic (PBPK) modeling is a powerful tool that integrates drugs’ physicochemical properties with anatomical and physiological data to predict PK behavior. In pediatric populations, PBPK modeling accounts for developmental changes in organ function, making it particularly useful for optimizing antibiotic dosing across different age groups, from neonates to adolescents. In recent decades, PBPK modeling has been widely applied to predict antibiotic disposition in pediatric patients for various clinical and research purposes. Model-informed precision dosing (MIPD) is an evolving approach that enhances traditional therapeutic drug monitoring by integrating multiple information sources into a mathematical framework. By incorporating PBPK modeling, MIPD could offer a more optimized antibiotic dosing that accounts for PK/PD parameters at the site of infection, improving therapeutic outcomes while minimizing toxicity. This review summarizes currently published pediatric PBPK modeling studies on antibiotics, covering various objectives such as evaluating drug–drug interactions, PK/PD analyses in targeted tissues, predicting PK in specific populations (e.g., maternal/fetal, renal impairment, obesity), and PK predictions for preterm neonates. Based on these reports, the review discusses the implications of PBPK modeling for MIPD in pediatric antibiotic therapy. Full article
Show Figures

Figure 1

14 pages, 1451 KiB  
Article
In Silico Evaluation of the Biopharmaceutical and Pharmacokinetic Behavior of Metronidazole from Coated Colonic Release Matrix Tablets
by Roberto Arévalo-Pérez, Cristina Maderuelo and José M. Lanao
Pharmaceutics 2025, 17(5), 647; https://doi.org/10.3390/pharmaceutics17050647 - 14 May 2025
Viewed by 586
Abstract
Background: Physiologically based biopharmaceutics modeling (PBBM) models can help to predict drug release and in vivo absorption behaviors. Colon drug delivery systems have gained interest over the past few years due to the advantages they provide in treating certain diseases in a local [...] Read more.
Background: Physiologically based biopharmaceutics modeling (PBBM) models can help to predict drug release and in vivo absorption behaviors. Colon drug delivery systems have gained interest over the past few years due to the advantages they provide in treating certain diseases in a local way. The objectives of this work were to simulate the biopharmaceutical and pharmacokinetic behavior of metronidazole hydrophilic matrices coated with different enteric polymers and to highlight the factors with a significant impact on the simulated pharmacokinetic parameters. Methods: Physicochemical properties of metronidazole were introduced into Simcyp® simulator platform, and the Advanced Dissolution Absorption Model (ADAM) was employed to simulate the in vivo intestinal absorption and colonic concentrations of metronidazole using a PBBM model. A Kruskal–Wallis test was carried out in order to determine which one of the factors studied has a statistically significant impact on the pharmacokinetic parameters (AUC, Cmax, and Tmax) simulated. Results: Enteric-coated matrix tablets are capable of avoiding metronidazole absorption in the small intestine and releasing it in the colonic region. The release and absorption rates of metronidazole depend largely on the percentage of weight gain of the coating and also on the coating agent. Coated tablets with a time-dependent coating show less variability. Conclusions: PBBM models can help predict the release from drug delivery systems and the pharmacokinetics in vivo of metronidazole from data obtained in vitro, although complementary in vivo studies should be needed. Full article
Show Figures

Figure 1

28 pages, 7155 KiB  
Review
Accelerating Biologics PBPK Modelling with Automated Model Building: A Tutorial
by Abdallah Derbalah, Tariq Abdulla, Mailys De Sousa Mendes, Qier Wu, Felix Stader, Masoud Jamei, Iain Gardner and Armin Sepp
Pharmaceutics 2025, 17(5), 604; https://doi.org/10.3390/pharmaceutics17050604 - 2 May 2025
Viewed by 1708
Abstract
Physiologically based pharmacokinetic (PBPK) modelling for biologics, such as monoclonal antibodies and therapeutic proteins, involves capturing complex processes, including target-mediated drug disposition (TMDD), FcRn-mediated recycling, and tissue-specific distribution. The Simcyp Designer Biologics PBPK Platform Model offers an intuitive and efficient platform for constructing [...] Read more.
Physiologically based pharmacokinetic (PBPK) modelling for biologics, such as monoclonal antibodies and therapeutic proteins, involves capturing complex processes, including target-mediated drug disposition (TMDD), FcRn-mediated recycling, and tissue-specific distribution. The Simcyp Designer Biologics PBPK Platform Model offers an intuitive and efficient platform for constructing mechanistic PBPK models with pre-defined templates and automated model assembly, reducing manual input and improving reproducibility. This tutorial provides a step-by-step guide to using the platform, highlighting features such as cross-species scaling, population variability simulations, and flexibility for model customization. Practical case studies demonstrate the platform’s capability to streamline workflows, enabling rapid, mechanistic model development to address key questions in biologics drug development. By automating critical processes, this tool enhances decision-making in translational research, optimizing the modelling and simulation of large molecules across discovery and clinical stages. Full article
Show Figures

Figure 1

15 pages, 2870 KiB  
Article
5-Fluorouracil Toxicity: Revisiting the Relevance of Pharmacokinetic Parameters
by Hans Mielke, Engi Abd Elhady Algharably and Ursula Gundert-Remy
Pharmaceuticals 2025, 18(5), 653; https://doi.org/10.3390/ph18050653 - 29 Apr 2025
Viewed by 1064
Abstract
Background/Objectives: 5-fluorouracil (5-FU) is used in the treatment of solid cancer types. Because of its narrow therapeutic window, drug monitoring is recommended. We were confronted to answer a question on the relevance of concentration as opposed to the area under the plasma [...] Read more.
Background/Objectives: 5-fluorouracil (5-FU) is used in the treatment of solid cancer types. Because of its narrow therapeutic window, drug monitoring is recommended. We were confronted to answer a question on the relevance of concentration as opposed to the area under the plasma concentration–time curve (AUC) to predict toxicity when we had to assess the case of a patient who died after an erroneously high infusion rate. Methods: We used physiologically-based pharmacokinetic modeling (PBPK) to simulate the concentration–time profile of 5-FU data on doses, dosing schedules and life-threatening toxicity for both the patient in question as well as data from the literature. Furthermore, steady-state 5-FU concentrations were calculated from an additional set of data found in the literature on AUCs and non-life-threatening toxicity. Results: The model predictions matched well with experimental data, confirming the suitability of the model. Life-threatening toxicity was related to a concentration above 6 mg/L, whereas non-life-threatening toxicity was low at concentrations less than 3 mg/L but steeply increased between 3 and 4 mg/L. Data analysis supported by a decision algorithm suggests that the 5-FU steady-state plasma concentration is a better toxicity predictor than the AUC. Conclusions: We recommend monitoring the concentration one hour after infusion starts when about 50% of the steady state is reached in patients for whom higher doses are clinically considered relevant. Monitoring the concentration one hour after starting the infusion has the advantage that dose correction could be made early before toxicity can be observed. Full article
(This article belongs to the Special Issue Therapeutic Drug Monitoring and Adverse Drug Reactions: 2nd Edition)
Show Figures

Graphical abstract

15 pages, 2715 KiB  
Article
Physiologically Based Pharmacokinetic Modeling to Predict Lamotrigine Exposure in Special Populations to Facilitate Therapeutic Drug Monitoring and Guide Dosing Regimens
by Ji-Cheng Li, Chen-Fang Miao, Yun Lei and Ai-Lin Liu
Pharmaceuticals 2025, 18(5), 637; https://doi.org/10.3390/ph18050637 - 27 Apr 2025
Viewed by 889
Abstract
Background: Lamotrigine plays a crucial role in the treatment of epilepsy and bipolar disorder in adults and children. However, its pharmacokinetic (PK) behavior in first or long-term treatment in pediatric patients and the changes in drug exposure in patients with renal impairment [...] Read more.
Background: Lamotrigine plays a crucial role in the treatment of epilepsy and bipolar disorder in adults and children. However, its pharmacokinetic (PK) behavior in first or long-term treatment in pediatric patients and the changes in drug exposure in patients with renal impairment are not well characterized. The purpose of the research was to build a robust physiologically based pharmacokinetic (PBPK) model of lamotrigine for the prediction of drug exposure in diverse populations to facilitate therapeutic drug monitoring (TDM) and guide dosing regimens. Methods: The physicochemical parameter values of lamotrigine were integrated to establish and validate the model in an adult population in PK-sim. This adult PBPK model can be extrapolated to children and patients with renal impairment to predict PK changes. Results: Most of the observed data were within the 5th and 95th percentile intervals of the variability around the predicted plasma concentrations. The model predicted pharmacokinetic thresholds and exposure values for clinically safe and effective doses recommended by the FDA for initial and long-term treatment of epilepsy in adults and children aged 2–12 years. Notably, patients with severe renal impairment and end-stage renal disease experienced an average increase in the area under the curve of 1.51 folds and 1.62 folds, respectively. This scenario necessitates further lamotrigine dose adjustments. Conclusions: The developed lamotrigine PBPK model offers a strategy for assisting clinicians in TDM and dose adjustment for special populations, thereby offering a reference (PK parameters, as well as peak and valley concentrations to reach a steady state) for a safer administration regimen in clinical treatment. Full article
Show Figures

Figure 1

11 pages, 9036 KiB  
Article
Physiologically Based Pharmacokinetic Modeling of Biologic Case Studies in Monkeys and Humans Reveals the Necessity of an Additional Clearance Term
by Felix Stader, Pradeep Sharma, Weize Huang, Mary P. Choules, Marie-Emilie Willemin, Xinwen Zhang, Estelle Yau, Abdallah Derbalah, Adriana Zyla, Cong Liu and Armin Sepp
Pharmaceutics 2025, 17(5), 560; https://doi.org/10.3390/pharmaceutics17050560 - 24 Apr 2025
Viewed by 1331
Abstract
Background/Objectives: Physiologically based pharmacokinetic (PBPK) modeling is an important tool in biologic drug development. However, a standardized modeling strategy is currently missing. A cross-industry collaboration developed PBPK models for seven case studies, including monoclonal antibodies, antibody–drug conjugates, and bispecific T-cell engagers, to [...] Read more.
Background/Objectives: Physiologically based pharmacokinetic (PBPK) modeling is an important tool in biologic drug development. However, a standardized modeling strategy is currently missing. A cross-industry collaboration developed PBPK models for seven case studies, including monoclonal antibodies, antibody–drug conjugates, and bispecific T-cell engagers, to identify key parameters and establish a workflow to simulate biologic drugs in monkeys and in humans. Methods: PBPK models were developed in the monkey with limited data, including the molecular weight, the binding affinity to FcRn, and the additional systemic clearance of IgG, which is 20% of the total clearance. The binding affinity was only available for human FcRn and corrected for the known species-dependent differences in IgG binding. The strategy of monkey simulations was evaluated with an additional 14 studies published in the literature. Three different scenarios were simulated in humans afterwards: without, with allometrically scaled, and with optimized additional systemic clearance. Results: The plasma peak concentration and the area under the curve were predicted within 50% of the observed data for all studied case examples in the monkey, which demonstrates that sparse input parameters are sufficient for successful predictions in the monkey. Simulations in humans demonstrated the need for additional systemic clearance, because drug exposure was highly overpredicted without an additional systemic clearance term. Allometric scaling improved the predictions, but optimization led to the best fit, which is currently a limitation in the translation from animals to humans. Conclusions: This work highlights the importance of understanding the general mechanisms of drug uptake in different tissue types and cells in both target-dependent and -independent processes. Full article
Show Figures

Figure 1

20 pages, 2054 KiB  
Article
Matrix Approach Assessment of Cabotegravir Drug–Drug Interactions with OAT1/OAT3 Substrates and UGT1A1/UGT1A9 Inhibitors Using Physiologically-Based Pharmacokinetic Modeling
by Helen Tracey, Simon T. Bate, Susan Ford, Parul Patel, Jackie Bloomer, Aarti Patel and Kunal S. Taskar
Pharmaceutics 2025, 17(4), 531; https://doi.org/10.3390/pharmaceutics17040531 - 18 Apr 2025
Viewed by 924
Abstract
Background/Objective: Cabotegravir (CAB), available as an oral tablet and as a long-acting (LA) nanosuspension for intramuscular injection, is approved as a combination therapy for the treatment, and as a monotherapy for the prevention, of HIV-1 infection. People living with HIV may receive multiple [...] Read more.
Background/Objective: Cabotegravir (CAB), available as an oral tablet and as a long-acting (LA) nanosuspension for intramuscular injection, is approved as a combination therapy for the treatment, and as a monotherapy for the prevention, of HIV-1 infection. People living with HIV may receive multiple concomitant medications, with the associated risk of drug–drug interactions (DDIs). CAB is an inhibitor of OAT1/OAT3 renal transporters and a substrate of the UDP-glucuronosyltransferase enzymes UGT1A1 and 1A9, in vitro. While the effect of induction of UGT1A1/UGT1A9 on CAB exposure had been investigated in the clinic, the effect of the risk of DDIs with CAB via inhibition of these enzymes, or as an inhibitor of OAT1/OAT3 transporters, had not been evaluated. Methods: A physiologically-based pharmacokinetic (PBPK) model was developed and verified for orally dosed CAB to investigate the DDI risks associated with CAB, using a matrix approach to extensively qualify the PBPK platform and the substrates and/or inhibitors of either OAT1/OAT3 or UGT1A1/UGT1A9. The effect of uncertainties in in vitro inhibition values for OAT1/OAT3 was assessed via sensitivity analysis. Results: A mean increase of less than 25% in systemic exposure for OAT1/OAT3 substrates was predicted, with the potential for an increase of up to 80% based on the sensitivity analysis. On co-dosing with UGT1A1/UGT1A9 inhibitors, the predicted mean increase in CAB exposure was within 11%. Conclusions: PBPK modelling indicated that clinically relevant DDIs are not anticipated with OAT1/3 substrates or UGT1A1/1A9 inhibitors and CAB. With maximal exposure of the LA formulation of CAB being lower than the oral, the results of these simulations can be extrapolated to LA injectable dosing. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Figure 1

Back to TopTop