1
|
Magel V, Blum J, Dolde X, Leisner H, Grillberger K, Khalidi H, Gardner I, Ecker GF, Pallocca G, Dreser N, Leist M. Inhibition of Neural Crest Cell Migration by Strobilurin Fungicides and Other Mitochondrial Toxicants. Cells 2024; 13:2057. [PMID: 39768149 PMCID: PMC11674305 DOI: 10.3390/cells13242057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Cell-based test methods with a phenotypic readout are frequently used for toxicity screening. However, guidance on how to validate the hits and how to integrate this information with other data for purposes of risk assessment is missing. We present here such a procedure and exemplify it with a case study on neural crest cell (NCC)-based developmental toxicity of picoxystrobin. A library of potential environmental toxicants was screened in the UKN2 assay, which simultaneously measures migration and cytotoxicity in NCC. Several strobilurin fungicides, known as inhibitors of the mitochondrial respiratory chain complex III, emerged as specific hits. From these, picoxystrobin was chosen to exemplify a roadmap leading from cell-based testing towards toxicological predictions. Following a stringent confirmatory testing, an adverse outcome pathway was developed to provide a testable toxicity hypothesis. Mechanistic studies showed that the oxygen consumption rate was inhibited at sub-µM picoxystrobin concentrations after a 24 h pre-exposure. Migration was inhibited in the 100 nM range, under assay conditions forcing cells to rely on mitochondria. Biokinetic modeling was used to predict intracellular concentrations. Assuming an oral intake of picoxystrobin, consistent with the acceptable daily intake level, physiologically based kinetic modeling suggested that brain concentrations of 0.1-1 µM may be reached. Using this broad array of hazard and toxicokinetics data, we calculated a margin of exposure ≥ 80 between the lowest in vitro point of departure and the highest predicted tissue concentration. Thus, our study exemplifies a hit follow-up strategy and contributes to paving the way to next-generation risk assessment.
Collapse
Affiliation(s)
- Viktoria Magel
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Jonathan Blum
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Xenia Dolde
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Heidrun Leisner
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Karin Grillberger
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Hiba Khalidi
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Iain Gardner
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Gerhard F. Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Giorgia Pallocca
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Nadine Dreser
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
2
|
Dibbets AC, Koldeweij C, Osinga EP, Scheepers HCJ, de Wildt SN. Barriers and Facilitators for Bringing Model-Informed Precision Dosing to the Patient's Bedside: A Systematic Review. Clin Pharmacol Ther 2024. [PMID: 39659053 DOI: 10.1002/cpt.3510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Model-informed precision dosing (MIPD) utilizes mathematical models to predict optimal medication doses for a specific patient or patient population. However, the factors influencing the implementation of MIPD have not been fully elucidated, hindering its widespread use in clinical practice. A systematic review was conducted in PubMed from inception to December 2022, aiming to identify barriers and facilitators for the implementation of MIPD into patient care. Articles with a focus on implementation of MIPD were eligible for this review. After screening titles and abstracts, full articles investigating the clinical implementation of MIPD were included for data extraction. Of 790 records identified, 15 publications were included. A total of 72 barriers and facilitators across seven categories were extracted through a hybrid thematic analysis. Barriers comprised limited data for model validation, unclear regulatory pathways for model endorsement and additional drug level measurements required for certain types of MIPD. Facilitators encompassed the development of user-friendly MIPD tools continuously updated based on user feedback and data. Collaborative efforts among diverse stakeholders for model validation and implementation, along with education of end-users, may promote the utilization of MIPD in patient care. Despite ongoing challenges, this systematic review revealed various strategies to facilitate the clinical implementation of MIPD.
Collapse
Affiliation(s)
- Anna Caroline Dibbets
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW, Institute for Oncology and Reproduction, Maastricht, The Netherlands
| | - Charlotte Koldeweij
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esra P Osinga
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hubertina C J Scheepers
- Department of Obstetrics and Gynaecology, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW, Institute for Oncology and Reproduction, Maastricht, The Netherlands
| | - Saskia N de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Intensive Care, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Jiang C, Zheng X, Li P, Qian J, Li Q. A retrospective pharmacovigilance study of post-marketing safety concerns with cefuroxime. Ther Adv Drug Saf 2024; 15:20420986241258049. [PMID: 38881538 PMCID: PMC11177735 DOI: 10.1177/20420986241258049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/09/2024] [Indexed: 06/18/2024] Open
Abstract
Background Cefuroxime has played a crucial role in the prevention and treatment of bacterial infections. However, the differences in adverse events across formulations and routes remain unclear. Objectives This study aimed to investigate the post-marketing safety of cefuroxime, particularly concerning formulations and routes. Design A retrospective pharmacovigilance study of cefuroxime was conducted using the data from Food and Drug Administration Adverse Event Reporting System database. Methods The clinical characteristics and concomitant drugs reported with cefuroxime were investigated. Adverse event signals of cefuroxime were identified based on four disproportionality algorithms. The signal differences of cefuroxime across formulations and routes were further examined. Results A total of 1810 adverse event reports associated with cefuroxime were identified, and 181 cefuroxime-associated signals were detected. Compared with tablets, injections were more likely to cause preferred terms 'blood pressure decreased' and 'anaphylactic shock'. In addition, system organ class 'eye disorders' significantly increased when cefuroxime was administered intraocularly, underscoring the importance of exercising caution regarding ocular toxicity. Conclusion The adverse events associated with cefuroxime were significantly different across formulations and routes, which deserve special attention in clinical use.
Collapse
Affiliation(s)
- Cheng Jiang
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoxiao Zheng
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ping Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiancheng Qian
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou, Zhejiang 310012, China
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Berezowska M, Sharma P, Pilla Reddy V, Coppola P. Physiologically Based Pharmacokinetic modelling of drugs in pregnancy: A mini-review on availability and limitations. Fundam Clin Pharmacol 2024; 38:402-409. [PMID: 37968879 DOI: 10.1111/fcp.12967] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/14/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modelling in pregnancy is a relatively new approach that is increasingly being used to assess drug systemic exposure in pregnant women to potentially inform dosing adjustments. Physiological changes throughout pregnancy are incorporated into mathematical models to simulate drug disposition in the maternal and fetal compartments as well as the transfer of drugs across the placenta. This mini-review gathers currently available pregnancy PBPK models for drugs commonly used during pregnancy. In addition, information about the main PBPK modelling platforms used, metabolism pathways, drug transporters, data availability and drug labels were collected. The aim of this mini-review is to provide a concise overview, demonstrate trends in the field, highlight understudied areas and identify current gaps of PBPK modelling in pregnancy. Possible future applications of this PBPK approach are discussed from a clinical, regulatory and industry perspective.
Collapse
Affiliation(s)
- Monika Berezowska
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Pradeep Sharma
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Venkatesh Pilla Reddy
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Paola Coppola
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
5
|
Werdan Romão MA, Pinto L, Cavalli RC, Duarte G, de Moraes NV, Abduljalil K, Moreira FDL. Mechanistic Framework to Predict Maternal-Placental-Fetal Pharmacokinetics of Nifedipine Employing Physiologically Based Pharmacokinetic Modeling Approach. J Clin Pharmacol 2024; 64:568-577. [PMID: 38305718 DOI: 10.1002/jcph.2404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024]
Abstract
Nifedipine is used for treating mild to severe hypertension and preventing preterm labor in pregnant women. Nevertheless, concerns about nifedipine fetal exposure and safety are always raised. The aim of this study was to develop and validate a maternal-placental-fetal nifedipine physiologically based pharmacokinetic (PBPK) model and apply the model to predict maternal, placental, and fetal exposure to nifedipine at different pregnancy stages. A nifedipine PBPK model was verified with nonpregnant data and extended to the pregnant population after the inclusion of the fetoplacental multicompartment model that accounts for the placental tissue and different fetal organs within the Simcyp Simulator version 22. Model parametrization involved scaling nifedipine transplacental clearance based on Caco-2 permeability, and fetal hepatic clearance was obtained from in vitro to in vivo extrapolation encompassing cytochrome P450 3A7 and 3A4 activities. Predicted concentration profiles were compared with in vivo observations and the transplacental transfer results were evaluated using 2-fold criteria. The PBPK model predicted a mean cord-to-maternal plasma ratio of 0.98 (range, 0.86-1.06) at term, which agrees with experimental observations of 0.78 (range, 0.59-0.93). Predicted nifedipine exposure was 1.4-, 2.0-, and 3.0-fold lower at 15, 27, and 39 weeks of gestation when compared with nonpregnant exposure, respectively. This innovative PBPK model can be applied to support maternal and fetal safety assessment for nifedipine at various stages of pregnancy.
Collapse
Affiliation(s)
- Marya Antônya Werdan Romão
- Laboratório de Farmacometria (LabFarma), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leonardo Pinto
- Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Ricardo Carvalho Cavalli
- Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Geraldo Duarte
- Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Natália Valadares de Moraes
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | | | - Fernanda de Lima Moreira
- Laboratório de Farmacometria (LabFarma), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
6
|
Yan Y, Wang Q, Wu W, Yi H, Xie F. Evaluation of Various Approaches to Estimate Transplacental Clearance of Vancomycin for Predicting Fetal Concentrations using a Maternal-Fetal Physiologically Based Pharmacokinetic Model. Pharm Res 2024; 41:899-910. [PMID: 38684563 DOI: 10.1007/s11095-024-03705-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Evaluating drug transplacental clearance is vital for forecasting fetal drug exposure. Ex vivo human placenta perfusion experiments are the most suitable approach for this assessment. Various in silico methods are also proposed. This study aims to compare these prediction methods for drug transplacental clearance, focusing on the large molecular weight drug vancomycin (1449.3 g/mol), using maternal-fetal physiologically based pharmacokinetic (m-f PBPK) modeling. METHODS Ex vivo human placenta perfusion experiments, in silico approaches using intestinal permeability as a substitute (quantitative structure property relationship (QSPR) model and Caco-2 permeability in vitro-in vivo correlation model) and midazolam calibration model with Caco-2 scaling were assessed for determining the transplacental clearance (CLPD) of vancomycin. The m-f PBPK model was developed stepwise using Simcyp, incorporating the determined CLPD values as a crucial input parameter for transplacental kinetics. RESULTS The developed PBPK model of vancomycin for non-pregnant adults demonstrated excellent predictive performance. By incorporating the CLPD parameterization derived from ex vivo human placenta perfusion experiments, the extrapolated m-f PBPK model consistently predicted maternal and fetal concentrations of vancomycin across diverse doses and distinct gestational ages. However, when the CLPD parameter was derived from alternative prediction methods, none of the extrapolated maternal-fetal PBPK models produced fetal predictions in line with the observed data. CONCLUSION Our study showcased that combination of ex vivo human placenta perfusion experiments and m-f PBPK model has the capability to predict fetal exposure for the large molecular weight drug vancomycin, whereas other in silico approaches failed to achieve the same level of accuracy.
Collapse
Affiliation(s)
- Yunan Yan
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China
| | - Qiushi Wang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China
| | - Wei Wu
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Hanxi Yi
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China.
| |
Collapse
|
7
|
Abduljalil K, Gardner I, Jamei M. An Application of a Physiologically Based Pharmacokinetic Approach to Predict Ceftazidime Pharmacokinetics in a Pregnant Population. Pharmaceutics 2024; 16:474. [PMID: 38675135 PMCID: PMC11054561 DOI: 10.3390/pharmaceutics16040474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Physiological changes during pregnancy can alter maternal and fetal drug exposure. The objective of this work was to predict maternal and umbilical ceftazidime pharmacokinetics during pregnancy. Ceftazidime transplacental permeability was predicted from its physicochemical properties and incorporated into the model. Predicted concentrations and parameters from the PBPK model were compared to the observed data. PBPK predicted ceftazidime concentrations in non-pregnant and pregnant subjects of different gestational weeks were within 2-fold of the observations, and the observed concentrations fell within the 5th-95th prediction interval from the PBPK simulations. The calculated transplacental clearance (0.00137 L/h/mL of placenta volume) predicted an average umbilical cord-to-maternal plasma ratio of 0.7 after the first dose, increasing to about 1.0 at a steady state, which also agrees well with clinical observations. The developed maternal PBPK model adequately predicted the observed exposure and kinetics of ceftazidime in the pregnant population. Using a verified population-based PBPK model provides valuable insights into the disposition of drug concentrations in special individuals that are otherwise difficult to study and, in addition, offers the possibility of supplementing sparse samples obtained in vulnerable populations with additional knowledge, informing the dosing adjustment and study design, and improving the efficacy and safety of drugs in target populations.
Collapse
Affiliation(s)
- Khaled Abduljalil
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | | | | |
Collapse
|
8
|
van Hoogdalem MW, Tanaka R, Abduljalil K, Johnson TN, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T. Forecasting Fetal Buprenorphine Exposure through Maternal-Fetal Physiologically Based Pharmacokinetic Modeling. Pharmaceutics 2024; 16:375. [PMID: 38543269 PMCID: PMC10975492 DOI: 10.3390/pharmaceutics16030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 04/01/2024] Open
Abstract
Buprenorphine readily crosses the placenta, and with greater prenatal exposure, neonatal opioid withdrawal syndrome (NOWS) likely grows more severe. Current dosing strategies can be further improved by tailoring doses to expected NOWS severity. To allow the conceptualization of fetal buprenorphine exposure, a maternal-fetal physiologically based pharmacokinetic (PBPK) model for sublingual buprenorphine was developed using Simcyp (v21.0). Buprenorphine transplacental passage was predicted from its physicochemical properties. The maternal-fetal PBPK model integrated reduced transmucosal absorption driven by lower salivary pH and induced metabolism observed during pregnancy. Maternal pharmacokinetics was adequately predicted in the second trimester, third trimester, and postpartum period, with the simulated area under the curve from 0 to 12 h, apparent clearance, and peak concentration falling within the 1.25-fold prediction error range. Following post hoc adjustment of the likely degree of individual maternal sublingual absorption, umbilical cord blood concentrations at delivery (n = 21) were adequately predicted, with a geometric mean ratio between predicted and observed fetal concentrations of 1.15 and with 95.2% falling within the 2-fold prediction error range. The maternal-fetal PBPK model developed in this study can be used to forecast fetal buprenorphine exposure and would be valuable to investigate its correlation to NOWS severity.
Collapse
Affiliation(s)
- Matthijs W. van Hoogdalem
- Division of Translational and Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.W.v.H.); (R.T.); (A.A.V.)
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ryota Tanaka
- Division of Translational and Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.W.v.H.); (R.T.); (A.A.V.)
| | - Khaled Abduljalil
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, UK; (K.A.); (T.N.J.)
| | - Trevor N. Johnson
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, UK; (K.A.); (T.N.J.)
| | - Scott L. Wexelblatt
- Division of Neonatology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (S.L.W.); (H.T.A.)
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Henry T. Akinbi
- Division of Neonatology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (S.L.W.); (H.T.A.)
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Alexander A. Vinks
- Division of Translational and Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.W.v.H.); (R.T.); (A.A.V.)
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Tomoyuki Mizuno
- Division of Translational and Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.W.v.H.); (R.T.); (A.A.V.)
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
9
|
Isoherranen N. Physiologically Based Pharmacokinetic (PBPK) Modeling of small molecules: How Much Progress Have We Made?. Drug Metab Dispos 2024; 53:DMD-MR-2022-000960. [PMID: 38326033 DOI: 10.1124/dmd.123.000960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/10/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models of small molecules have become mainstream in drug development and academic research. The use of PBPK models is continuously expanding with the majority of work now focusing on predictions of drug-drug interactions, drug-disease interactions, and changes in drug disposition across lifespan. Recently, publications that use PBPK modeling to predict drug disposition during pregnancy and in organ impairment have increased reflecting the advances in incorporating diverse physiological changes into the models. Due to the expanding computational power and diversity of modeling platforms available, the complexity of PBPK models has also increased. Academic efforts have provided clear advances in better capturing human physiology in PBPK models and incorporating more complex mathematical concepts into PBPK models. Examples of such advances include the segregated gut model with a series gut compartments allowing modeling of physiological blood flow distribution within an organ and zonation of metabolic enzymes, and series compartment liver models allowing simulations of hepatic clearance for high extraction drugs. Despite these advances in academic research, the progress in assessing model quality and defining model acceptance criteria based on the intended use of the models has not kept pace. This review suggests that awareness of the need for predefined criteria for model acceptance has increased but many manuscripts still lack description of scientific justification and/or rationale for chosen acceptance criteria. As artificial intelligence and machine learning approaches become more broadly accepted, these tools offer promise for development of comprehensive assessment for existing observed data and analysis of model performance. Significance Statement PBPK modeling has become a mainstream application in academic literature and is broadly used for predictions, analysis and evaluation of pharmacokinetic data. Many significant advances have been made in developing advanced PBPK models that better capture human physiology but oftentimes sufficient justification for the chosen model acceptance criterion and model structure is still missing. This review provides a summary of the current landscape of PBPK applications used and highlights the needs for advancing PBPK modeling science and training in academia.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, University of Washington, United States
| |
Collapse
|
10
|
Liu XI, Green DJ, van den Anker J, Ahmadzia HK, Burckart GJ, Dallmann A. Development of a Generic Fetal Physiologically Based Pharmacokinetic Model and Prediction of Human Maternal and Fetal Organ Concentrations of Cefuroxime. Clin Pharmacokinet 2024; 63:69-78. [PMID: 37962827 DOI: 10.1007/s40262-023-01323-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Physiologically based pharmacokinetic (PBPK) models for pregnant women have recently been successfully used to predict maternal and umbilical cord pharmacokinetics (PK). Because there is very limited opportunity for conducting clinical and PK investigations for fetal drug exposure, PBPK models may provide further insights. The objectives of this study were to extend a whole-body pregnancy PBPK model by multiple compartments representing fetal organs, and to predict the PK of cefuroxime in the maternal and fetal plasma, the amniotic fluid, and several fetal organs. METHODS To this end, a previously developed pregnancy PBPK model for cefuroxime was updated using the open-source software Open Systems Pharmacology (PK-Sim®/MoBi®). Multiple compartments were implemented to represent fetal organs including brain, heart, liver, lungs, kidneys, the gastrointestinal tract (GI), muscles, and fat tissue, as well as another compartment lumping organs and tissues not explicitly represented. RESULTS This novel PBPK model successfully predicted cefuroxime concentrations in maternal blood, umbilical cord, amniotic fluid, and several fetal organs including heart, liver, and lungs. Further model validation with additional clinical PK data is needed to build confidence in the model. CONCLUSIONS Being developed with an open-source software, the presented generic model can be freely re-used and tailored to address specific questions at hand, e.g., to assist the design of clinical studies in the context of drug research or to predict fetal organ concentrations of chemicals in the context of fetal health risk assessment.
Collapse
Affiliation(s)
- Xiaomei I Liu
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.
| | - Dionna J Green
- Office of Pediatric Therapeutics, US Food and Drug Administration, Silver Spring, MD, USA
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
| | - Homa K Ahmadzia
- Division of Maternal-Fetal Medicine, Department of OB/Gyn, George Washington University, Washington, DC, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, MD, USA
| | - André Dallmann
- Bayer HealthCare SAS, Loos, France
- On Behalf of: Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| |
Collapse
|
11
|
Benzi JRDL, Melli PPDS, Duarte G, Unadkat JD, Lanchote VL. The Impact of Inflammation on the In Vivo Activity of the Renal Transporters OAT1/3 in Pregnant Women Diagnosed with Acute Pyelonephritis. Pharmaceutics 2023; 15:2427. [PMID: 37896187 PMCID: PMC10610490 DOI: 10.3390/pharmaceutics15102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Inflammation can regulate hepatic drug metabolism enzymes and transporters. The impact of inflammation on renal drug transporters remains to be elucidated. We aimed to quantify the effect of inflammation (caused by acute pyelonephritis) on the in vivo activity of renal OAT1/3, using the probe drug furosemide. Pregnant women (second or third trimester) received a single oral dose of furosemide 40 mg during acute pyelonephritis (Phase 1; n = 7) and after its resolution (Phase 2; n = 7; by treatment with intravenous cefuroxime 750 mg TID for 3-7 days), separated by 10 to 14 days. The IL-6, IFN-γ, TNF-α, MCP-1, and C-reactive protein plasma concentrations were higher in Phase I vs. Phase II. The pregnant women had a lower geometric mean [CV%] furosemide CLsecretion (3.9 [43.4] vs. 6.7 [43.8] L/h) and formation clearance to the glucuronide (1.1 [85.9] vs. 2.3 [64.1] L/h) in Phase 1 vs. Phase 2. Inflammation reduced the in vivo activity of renal OAT1/3 (mediating furosemide CLsecretion) and UGT1A9/1A1 (mediating the formation of furosemide glucuronide) by approximately 40% and 54%, respectively, presumably by elevating the plasma cytokine concentrations. The dosing regimens of narrow therapeutic window OAT drug substrates may need to be adjusted during inflammatory conditions.
Collapse
Affiliation(s)
- Jhohann Richard de Lima Benzi
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, São Paulo, Brazil;
| | - Patrícia Pereira dos Santos Melli
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (P.P.d.S.M.)
| | - Geraldo Duarte
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (P.P.d.S.M.)
| | - Jashvant D. Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Vera Lucia Lanchote
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, São Paulo, Brazil;
| |
Collapse
|
12
|
Nihad M, Sen U, Chaudhury D, Das UN, Shenoy P S, Bose B. Arachidonic acid modulates the cellular energetics of human pluripotent stem cells and protects the embryoid bodies from embryotoxicity effects in vitro. Reprod Toxicol 2023; 120:108438. [PMID: 37454977 DOI: 10.1016/j.reprotox.2023.108438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Arachidonic acid (AA), an ω-6 polyunsaturated fatty acid involved in signalling pathways that drive cell fate decisions, has an enhancing role in the immunomodulatory effect on mesenchymal stem cells and the vasculogenesis of embryonic stem cells. 3D embryoid bodies (EBs) from pluripotent stem cells (PSCs) have been used as in vitro models for embryotoxicity for various compounds/drugs. Valproic acid (VA), a common anti-epileptic drug, is known to be embryotoxic and cause malformations in embryos. As early embryogenesis depends on AA, we investigated the embryo protective effects of AA against the embryotoxic drug VA in this study. The effects of AA on the proliferation and cell cycle parameters of PSCs were studied. In particular, the potential of AA to abrogate VA-induced embryotoxicity in vitro was evaluated using ROS detection and antioxidant assays. In response to AA, we observed modulation in cell proliferation of induced pluripotent stem cells (iPSCs) and pluripotent NTERA-2 embryonal carcinoma (EC) cells. The present study substantiates the cytoprotective effects of AA against VA. These results imply that AA plays a critical role in the proliferation and differentiation of iPSCs and EC cells and protects the EBs from cytotoxic damage, thereby ensuring normal embryogenesis. Thus, the bioactive lipid AA may be explored for supplementation to benefit pregnant women treated with long-term anti-epileptic drugs to prevent in-utero fetal growth malformations.
Collapse
Affiliation(s)
- Muhammad Nihad
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, Mangalore 575018, Karnataka, India
| | - Utsav Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, 15th Floor, New York, NY 10029, USA
| | - Debajit Chaudhury
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, Mangalore 575018, Karnataka, India
| | - Undurti N Das
- BioScience Research Centre, Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India; UND Life Sciences, 2221 NW 5th St., Battle Ground, WA 98604, USA
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, Mangalore 575018, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, Mangalore 575018, Karnataka, India.
| |
Collapse
|
13
|
Krzyzanski W, Milad MA, Jobe AH, Jusko WJ. Minimal physiologically-based hybrid model of pharmacokinetics in pregnant women: Application to antenatal corticosteroids. CPT Pharmacometrics Syst Pharmacol 2023; 12:668-680. [PMID: 36917704 PMCID: PMC10196440 DOI: 10.1002/psp4.12899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 03/16/2023] Open
Abstract
Minimal physiologically-based pharmacokinetic (mPBPK) models are an alternative to full physiologically-based pharmacokinetic (PBPK) models as they offer reduced complexity while maintaining the physiological interpretation of key model components. Full PBPK models have been developed for pregnancy, but a mPBPK model eases the ability to perform a "top-down" meta-analysis melding all available pharmacokinetic (PK) data in the mother and fetus. Our hybrid mPBPK model consists of mPBPK models for the mother and fetus with connection by the placenta. This model was applied to describe the rich PK data of antenatal corticosteroid betamethasone (BET) jointly with the limited data for dexamethasone (DEX) in the mother and fetus. Physiologic model parameters were obtained from the literature while drug-dependent parameters were estimated by the simultaneous fitting of all available data for DEX and BET. Maternal clearances of DEX and BET confirmed the literature values, and the expected fetal-to-maternal plasma ratios ranged from 0.3 to 0.4 for both drugs. Simulations of maternal plasma concentrations for the dosing regimens of BET and DEX recommended by the World Health Organization based on our findings revealed up to 60% lower exposures than found in nonpregnant women and offers a means of devising alternative dosing regimens. Our hybrid mPBPK model and meta-analysis approach could facilitate assessment of other classes of drugs indicated for the treatment of pregnant women.
Collapse
Affiliation(s)
- Wojciech Krzyzanski
- School of Pharmacy and Pharmaceutical Sciences, State University of New YorkUniversity of BuffaloBuffaloNew YorkUSA
| | - Mark A. Milad
- Milad Pharmaceutical Consulting LLCPlymouthMichiganUSA
| | - Alan H. Jobe
- Division of Pulmonary BiologyCincinnati Children's Hospital Medical Center, University of CincinnatiCincinnatiOhioUSA
| | - William J. Jusko
- School of Pharmacy and Pharmaceutical Sciences, State University of New YorkUniversity of BuffaloBuffaloNew YorkUSA
| |
Collapse
|
14
|
De Sutter PJ, De Cock P, Johnson TN, Musther H, Gasthuys E, Vermeulen A. Predictive Performance of Physiologically Based Pharmacokinetic Modelling of Beta-Lactam Antibiotic Concentrations in Adipose, Bone, and Muscle Tissues. Drug Metab Dispos 2023; 51:499-508. [PMID: 36639242 DOI: 10.1124/dmd.122.001129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models consist of compartments representing different tissues. As most models are only verified based on plasma concentrations, it is unclear how reliable associated tissue profiles are. This study aimed to assess the accuracy of PBPK-predicted beta-lactam antibiotic concentrations in different tissues and assess the impact of using effect site concentrations for evaluation of target attainment. Adipose, bone, and muscle concentrations of five beta-lactams (piperacillin, cefazolin, cefuroxime, ceftazidime, and meropenem) in healthy adults were collected from literature and compared with PBPK predictions. Model performance was evaluated with average fold errors (AFEs) and absolute AFEs (AAFEs) between predicted and observed concentrations. In total, 26 studies were included, 14 of which reported total tissue concentrations and 12 unbound interstitial fluid (uISF) concentrations. Concurrent plasma concentrations, used as baseline verification of the models, were fairly accurate (AFE: 1.14, AAFE: 1.50). Predicted total tissue concentrations were less accurate (AFE: 0.68, AAFE: 1.89). A slight trend for underprediction was observed but none of the studies had AFE or AAFE values outside threefold. Similarly, predictions of microdialysis-derived uISF concentrations were less accurate than plasma concentration predictions (AFE: 1.52, AAFE: 2.32). uISF concentrations tended to be overpredicted and two studies had AFEs and AAFEs outside threefold. Pharmacodynamic simulations in our case showed only a limited impact of using uISF concentrations instead of unbound plasma concentrations on target attainment rates. The results of this study illustrate the limitations of current PBPK models to predict tissue concentrations and the associated need for more accurate models. SIGNIFICANCE STATEMENT: Clinical inaccessibility of local effect site concentrations precipitates a need for predictive methods for the estimation of tissue concentrations. This is the first study in which the accuracy of PBPK-predicted tissue concentrations of beta-lactam antibiotics in humans were assessed. Predicted tissue concentrations were found to be less accurate than concurrent predicted plasma concentrations. When using PBPK models to predict tissue concentrations, this potential relative loss of accuracy should be acknowledged when clinical tissue concentrations are unavailable to verify predictions.
Collapse
Affiliation(s)
- Pieter-Jan De Sutter
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Pieter De Cock
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Trevor N Johnson
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Helen Musther
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Elke Gasthuys
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| |
Collapse
|
15
|
Li S, Xie F. Foetal and neonatal exposure prediction and dosing evaluation for ampicillin using a physiologically-based pharmacokinetic modelling approach. Br J Clin Pharmacol 2023; 89:1402-1412. [PMID: 36357171 DOI: 10.1111/bcp.15589] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Ampicillin is frequently used in neonates for the treatment of sepsis and as an intrapartum prophylaxis option for Group B Streptococcus. Pharmacokinetic data to guide ampicillin dosing in neonates and during the intrapartum period are limited. The objective of this study was to build a physiologically-based pharmacokinetic (PBPK) model to characterize the disposition of ampicillin in neonates and foetuses and to inform corresponding optimal dosing regimens. METHODS An adult ampicillin PBPK model was first developed using the Simcyp® simulator. The adult model was then scaled to neonates by accounting for maturational changes in physiological parameters and age-dependent drug disposition or extended to a pregnancy model for mothers and foetuses. Models were verified using collected mean or individual-level concentration data from the literature. RESULTS The developed adult PBPK model included elimination via glomerular filtration, OAT3-mediated tubular secretion and biliary excretion as well as hepatic metabolism, and 89.8% of the observed mean concentrations in adults were within a 2-fold range of model mean predictions. Most of the observed individual-level observations in neonates (78.4%) and foetuses (about 65% in two studies) were within the 90% prediction intervals. The recommended 50 mg/kg every 8 h (q8h) ampicillin regimen achieved the 75% fraction time of total drug concentration above minimum inhibitory concentration (T > MIC) target for an MIC ≤8 mg/L in >90% virtual neonates, and 1 g ampicillin for pregnant women provided adequate foetal exposure (>0.25 mg/L) for 4 h prior to delivery. CONCLUSIONS A PBPK model was developed to characterize ampicillin's disposition in neonates, pregnant women, and foetuses, and the model supported optimal dosing evaluation in these vulnerable populations.
Collapse
Affiliation(s)
- Sanwang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
16
|
Hudson RE, Metz TD, Ward RM, McKnite AM, Enioutina EY, Sherwin CM, Watt KM, Job KM. Drug exposure during pregnancy: Current understanding and approaches to measure maternal-fetal drug exposure. Front Pharmacol 2023; 14:1111601. [PMID: 37033628 PMCID: PMC10076747 DOI: 10.3389/fphar.2023.1111601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Prescription drug use is prevalent during pregnancy, yet there is limited knowledge about maternal-fetal safety and efficacy of this drug use because pregnant individuals have historically been excluded from clinical trials. Underrepresentation has resulted in a lack of data available to estimate or predict fetal drug exposure. Approaches to study fetal drug pharmacology are limited and must be evaluated for feasibility and accuracy. Anatomic and physiological changes throughout pregnancy fluctuate based on gestational age and can affect drug pharmacokinetics (PK) for both mother and fetus. Drug concentrations have been studied throughout different stages of gestation and at or following delivery in tissue and fluid biospecimens. Sampling amniotic fluid, umbilical cord blood, placental tissue, meconium, umbilical cord tissue, and neonatal hair present surrogate options to quantify and characterize fetal drug exposure. These sampling methods can be applied to all therapeutics including small molecule drugs, large molecule drugs, conjugated nanoparticles, and chemical exposures. Alternative approaches to determine PK have been explored, including physiologically based PK modeling, in vitro methods, and traditional animal models. These alternative approaches along with convenience sampling of tissue or fluid biospecimens can address challenges in studying maternal-fetal pharmacology. In this narrative review, we 1) present an overview of the current understanding of maternal-fetal drug exposure; 2) discuss biospecimen-guided sampling design and methods for measuring fetal drug concentrations throughout gestation; and 3) propose methods for advancing pharmacology research in the maternal-fetal population.
Collapse
Affiliation(s)
- Rachel E. Hudson
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Torri D. Metz
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Robert M. Ward
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Autumn M. McKnite
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Utah, Salt Lake City, UT, United States
| | - Elena Y. Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Catherine M. Sherwin
- Department of Pediatrics, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Kevin M. Watt
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Kathleen M. Job
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, The University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
17
|
Macente J, Nauwelaerts N, Russo FM, Deprest J, Allegaert K, Lammens B, Hernandes Bonan R, Turner JM, Kumar S, Diniz A, Martins FS, Annaert P. PBPK-based dose finding for sildenafil in pregnant women for antenatal treatment of congenital diaphragmatic hernia. Front Pharmacol 2023; 14:1068153. [PMID: 36998614 PMCID: PMC10043195 DOI: 10.3389/fphar.2023.1068153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
Sildenafil is a potent vasodilator and phosphodiesterase type five inhibitor, commercially known as Revatio® and approved for the treatment of pulmonary arterial hypertension. Maternal administration of sildenafil during pregnancy is being evaluated for antenatal treatment of several conditions, including the prevention of pulmonary hypertension in fetuses with congenital diaphragmatic hernia. However, determination of a safe and effective maternal dose to achieve adequate fetal exposure to sildenafil remains challenging, as pregnancy almost always is an exclusion criterion in clinical studies. Physiologically-based pharmacokinetic (PBPK) modelling offers an attractive approach for dose finding in this specific population. The aim of this study is to exploit physiologically-based pharmacokinetic modelling to predict the required maternal dose to achieve therapeutic fetal exposure for the treatment congenital diaphragmatic hernia. A full-PBPK model was developed for sildenafil and N-desmethyl-sildenafil using the Simcyp simulator V21 platform, and verified in adult reference individuals, as well as in pregnant women, taking into account maternal and fetal physiology, along with factors known to determine hepatic disposition of sildenafil. Clinical pharmacokinetic data in mother and fetus were previously obtained in the RIDSTRESS study and were used for model verification purposes. Subsequent simulations were performed relying either on measured values for fetal fraction unbound (fu = 0.108) or on values predicted by the simulator (fu = 0.044). Adequate doses were predicted according to the efficacy target of 15 ng/mL (or 38 ng/mL) and safety target of 166 ng/mL (or 409 ng/mL), assuming measured (or predicted) fu values, respectively. Considering simulated median profiles for average steady state sildenafil concentrations, dosing regimens of 130 mg/day or 150 mg/day (administered as t.i.d.), were within the therapeutic window, assuming either measured or predicted fu values, respectively. For safety reasons, dosing should be initiated at 130 mg/day, under therapeutic drug monitoring. Additional experimental measurements should be performed to confirm accurate fetal (and maternal) values for fu. Additional characterization of pharmacodynamics in this specific population is required and may lead to further optimization of the dosing regimen.
Collapse
Affiliation(s)
- Julia Macente
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Nina Nauwelaerts
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | | | - Jan Deprest
- Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Karel Allegaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Jessica M. Turner
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Sailesh Kumar
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Andrea Diniz
- Pharmacokinetics and Biopharmaceutical Laboratory (PKBio), Department of Pharmacy, State University of Maringa, Maringa, Brazil
| | - Frederico S. Martins
- Pharmacokinetics and Biopharmaceutical Laboratory (PKBio), Department of Pharmacy, State University of Maringa, Maringa, Brazil
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- BioNotus GCV, Niel, Belgium
- *Correspondence: Pieter Annaert,
| |
Collapse
|
18
|
Wei Z, Jeong HC, Kim MG, Shin KH. Prediction of the Drug–Drug Interaction Potential between Tegoprazan and Amoxicillin/Clarithromycin Using the Physiologically Based Pharmacokinetic and Pharmacodynamic Model. Pharmaceuticals (Basel) 2023; 16:ph16030360. [PMID: 36986460 PMCID: PMC10054914 DOI: 10.3390/ph16030360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Tegoprazan is a novel potassium-competitive acid blocker. This study investigated the effect of drug–drug interaction on the pharmacokinetics and pharmacodynamics of tegoprazan co-administered with amoxicillin and clarithromycin, the first-line therapy for the eradication of Helicobacter pylori, using physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) modeling. The previously reported tegoprazan PBPK/PD model was modified and applied. The clarithromycin PBPK model was developed based on the model provided by the SimCYP® compound library. The amoxicillin model was constructed using the middle-out approach. All of the observed concentration–time profiles were covered well by the predicted profiles with the 5th and 95th percentiles. The mean ratios of predicted to observed PK parameters, including the area under the curve (AUC), maximum plasma drug concentration (Cmax), and clearance, were within the 30% intervals for the developed models. Two-fold ratios of predicted fold-changes of Cmax and AUC from time 0 to 24 h to observed data were satisfied. The predicted PD endpoints, including median intragastric pH and percentage holding rate at pH above 4 or 6 on day 1 and day 7, were close to the corresponding observed data. This investigation allows evaluation of the effects of CYP3A4 perpetrators on tegoprazan PK and PD changes, thus providing clinicians with the rationale for co-administration dosing adjustment.
Collapse
Affiliation(s)
- Zhuodu Wei
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Hyeon-Cheol Jeong
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Min-Gul Kim
- Department of Pharmacology, Medical School, Jeonbuk National University, Jeonju 54907, Republic of Korea
| | - Kwang-Hee Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
- Correspondence: ; Tel.: +82-53-950-8582
| |
Collapse
|
19
|
Sorzano C, Moreno MPDLC, Vilas J. An Analytical Solution for Saturable Absorption in Pharmacokinetics Models. Pharm Res 2023; 40:481-485. [PMID: 36543972 PMCID: PMC9944386 DOI: 10.1007/s11095-022-03455-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The first-order absorption is a common model used in Pharmacokinetics. The absorption of some drugs follows carrier mediated transport. It has been proposed that the amount of drug available may saturate the transport mechanism resulting in an absorption slower than the one predicted by the first-order model. Saturable absorption has been modeled at the differential equation level by substituting the constant rate absorption by a Hill kinetics absorption. However, its exact solution is so far unknown. The goal of this is to know the exact solution of different Hill kinetic absorption models. METHODS We start defining different absorption models and increasing then their complexity. The simplest case is the first-order absorption model and the most complex will be a generalized Hill kinetic absorption model. The differential equation of each model is integrated. RESULTS The complexity of the models their solutions may be not expressed in a close-form, or in term of elementary functions. We obtain and discuss the exact solutions of the different Hill kinetics absorption models. To do that, the solutions are studied according to the possible values of the free parameters of the models. We show the differences between models through simulations. CONCLUSIONS The knowledge of closed-form solutions allows to illustrate the differences between the different absorption models and minimizes the errors of numerical integration.
Collapse
Affiliation(s)
- C.O.S. Sorzano
- National Center of Biotechnology, CSIC., Madrid, Spain
- Kinestat Pharma, Madrid, Spain
| | | | - J.L. Vilas
- National Center of Biotechnology, CSIC., Madrid, Spain
| |
Collapse
|
20
|
Mao Q, Chen X. An update on placental drug transport and its relevance to fetal drug exposure. MEDICAL REVIEW (2021) 2022; 2:501-511. [PMID: 37724167 PMCID: PMC10388746 DOI: 10.1515/mr-2022-0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/27/2022] [Indexed: 09/20/2023]
Abstract
Pregnant women are often complicated with diseases that require treatment with medication. Most drugs administered to pregnant women are off-label without the necessary dose, efficacy, and safety information. Knowledge concerning drug transfer across the placental barrier is essential for understanding fetal drug exposure and hence drug safety and efficacy to the fetus. Transporters expressed in the placenta, including adenosine triphosphate (ATP)-binding cassette efflux transporters and solute carrier uptake transporters, play important roles in determining drug transfer across the placental barrier, leading to fetal exposure to the drugs. In this review, we provide an update on placental drug transport, including in vitro cell/tissue, ex vivo human placenta perfusion, and in vivo animal studies that can be used to determine the expression and function of drug transporters in the placenta as well as placental drug transfer and fetal drug exposure. We also describe how the knowledge of placental drug transfer through passive diffusion or active transport can be combined with physiologically based pharmacokinetic modeling and simulation to predict systemic fetal drug exposure. Finally, we highlight knowledge gaps in studying placental drug transport and predicting fetal drug exposure and discuss future research directions to fill these gaps.
Collapse
Affiliation(s)
- Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Xin Chen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
Coppola P, Kerwash E, Cole S. The Use of Pregnancy Physiologically Based Pharmacokinetic Modeling for Renally Cleared Drugs. J Clin Pharmacol 2022; 62 Suppl 1:S129-S139. [PMID: 36106785 DOI: 10.1002/jcph.2110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/09/2022] [Indexed: 11/06/2022]
Abstract
Physiologically based pharmacokinetic modeling (PBPK) could be used to predict changes in exposure during pregnancy and possibly inform medicine use in pregnancy in situations where there are currently no available clinical data. The Medicines and Healthcare Product Regulatory Agency has been evaluating the available models for a number of medicines cleared by the kidney. Models were evaluated for ceftazidime, cefuroxime, metformin, oseltamivir, and amoxicillin. Because the passive renal process contributes significantly to the renal elimination of these drugs and changes of the process during pregnancy have been implemented in existing pregnancy physiology models, simulations using these models can reasonably describe the pharmacokinetics of ceftazidime changes during pregnancy and appears to generally capture the changes in the other medicines; however, there are insufficient data on drugs solely passively cleared to fully qualify the models. In addition, in many cases, active transport processes are involved in a drug's renal clearance. Knowledge of changes in renal transport functions during pregnancy is emerging, and incorporation of such changes in current physiologically based pharmacokinetic modeling software is a work in progress. Filling this gap is expected to further enhance predictive performance of the models and increase the confidence in predicting pharmacokinetic changes in pregnant women for other renally cleared drugs.
Collapse
Affiliation(s)
- Paola Coppola
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Essam Kerwash
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Susan Cole
- Medicines and Healthcare Products Regulatory Agency, London, UK
| |
Collapse
|
22
|
Application of a Physiologically Based Pharmacokinetic Model to Predict Cefazolin and Cefuroxime Disposition in Obese Pregnant Women Undergoing Caesarean Section. Pharmaceutics 2022; 14:pharmaceutics14061162. [PMID: 35745736 PMCID: PMC9229966 DOI: 10.3390/pharmaceutics14061162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
Intravenous (IV) cefuroxime and cefazolin are used prophylactically in caesarean sections (CS). Currently, there are concerns regarding sub-optimal dosing in obese pregnant women compared to lean pregnant women prior to CS. The current study used a physiologically based pharmacokinetic (PBPK) approach to predict cefazolin and cefuroxime pharmacokinetics in obese pregnant women at the time of CS as well as the duration that these drug concentrations remain above a target concentration (2, 4 or 8 µg/mL or µg/g) in plasma or adipose tissue. Cefazolin and cefuroxime PBPK models were first built using clinical data in lean and in obese non–pregnant populations. Models were then used to predict cefazolin and cefuroxime pharmacokinetics data in lean and obese pregnant populations. Both cefazolin and cefuroxime models sufficiently described their total and free levels in the plasma and in the adipose interstitial fluid (ISF) in non–pregnant and pregnant populations. The obese pregnant cefazolin model predicted adipose exposure adequately at different reference time points and indicated that an IV dose of 2000 mg can maintain unbound plasma and adipose ISF concentration above 8 µg/mL for 3.5 h post dose. Predictions indicated that an IV 1500 mg cefuroxime dose can achieve unbound plasma and unbound ISF cefuroxime concentration of ≥8 µg/mL up to 2 h post dose in obese pregnant women. Re-dosing should be considered if CS was not completed within 2 h post cefuroxime administration for both lean or obese pregnant if cefuroxime concentrations of ≥8 µg/mL is required. A clinical study to measure cefuroxime adipose concentration in pregnant and obese pregnant women is warranted.
Collapse
|