1
|
Huang H, Zhao W, Qin N, Duan X. Recent Progress on Physiologically Based Pharmacokinetic (PBPK) Model: A Review Based on Bibliometrics. TOXICS 2024; 12:433. [PMID: 38922113 PMCID: PMC11209072 DOI: 10.3390/toxics12060433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Physiologically based pharmacokinetic/toxicokinetic (PBPK/PBTK) models are designed to elucidate the mechanism of chemical compound action in organisms based on the physiological, biochemical, anatomical, and thermodynamic properties of organisms. After nearly a century of research and practice, good results have been achieved in the fields of medicine, environmental science, and ecology. However, there is currently a lack of a more systematic review of progress in the main research directions of PBPK models, especially a more comprehensive understanding of the application in aquatic environmental research. In this review, a total of 3974 articles related to PBPK models from 1996 to 24 March 2024 were collected. Then, the main research areas of the PBPK model were categorized based on the keyword co-occurrence maps and cluster maps obtained by CiteSpace. The results showed that research related to medicine is the main application area of PBPK. Four major research directions included in the medical field were "drug assessment", "cross-species prediction", "drug-drug interactions", and "pediatrics and pregnancy drug development", in which "drug assessment" accounted for 55% of the total publication volume. In addition, bibliometric analyses indicated a rapid growth trend in the application in the field of environmental research, especially in predicting the residual levels in organisms and revealing the relationship between internal and external exposure. Despite facing the limitation of insufficient species-specific parameters, the PBPK model is still an effective tool for improving the understanding of chemical-biological effectiveness and will provide a theoretical basis for accurately assessing potential risks to ecosystems and human health. The combination with the quantitative structure-activity relationship model, Bayesian method, and machine learning technology are potential solutions to the previous research gaps.
Collapse
Affiliation(s)
| | | | - Ning Qin
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| | - Xiaoli Duan
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| |
Collapse
|
2
|
Silva MH. Investigating open access new approach methods (NAM) to assess biological points of departure: A case study with 4 neurotoxic pesticides. Curr Res Toxicol 2024; 6:100156. [PMID: 38404712 PMCID: PMC10891343 DOI: 10.1016/j.crtox.2024.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/28/2023] [Accepted: 02/09/2024] [Indexed: 02/27/2024] Open
Abstract
Open access new approach methods (NAM) in the US EPA ToxCast program and NTP Integrated Chemical Environment (ICE) were used to investigate activities of four neurotoxic pesticides: endosulfan, fipronil, propyzamide and carbaryl. Concordance of in vivo regulatory points of departure (POD) adjusted for interspecies extrapolation (AdjPOD) to modelled human Administered Equivalent Dose (AEDHuman) was assessed using 3-compartment or Adult/Fetal PBTK in vitro to in vivo extrapolation. Model inputs were from Tier 1 (High throughput transcriptomics: HTTr, high throughput phenotypic profiling: HTPP) and Tier 2 (single target: ToxCast) assays. HTTr identified gene expression signatures associated with potential neurotoxicity for endosulfan, propyzamide and carbaryl in non-neuronal MCF-7 and HepaRG cells. The HTPP assay in U-2 OS cells detected potent effects on DNA endpoints for endosulfan and carbaryl, and mitochondria with fipronil (propyzamide was inactive). The most potent ToxCast assays were concordant with specific components of each chemical mode of action (MOA). Predictive adult IVIVE models produced fold differences (FD) < 10 between the AEDHuman and the measured in vivo AdjPOD. The 3-compartment model was concordant (i.e., smallest FD) for endosulfan, fipronil and carbaryl, and PBTK was concordant for propyzamide. The most potent AEDHuman predictions for each chemical showed HTTr, HTPP and ToxCast were mainly concordant with in vivo AdjPODs but assays were less concordant with MOAs. This was likely due to the cell types used for testing and/or lack of metabolic capabilities and pathways available in vivo. The Fetal PBTK model had larger FDs than adult models and was less predictive overall.
Collapse
|
3
|
Fomina YY, Byrne JJ, Spong CY. Evaluating strength of recommendations for commonly administered medications in lactating women. J Matern Fetal Neonatal Med 2023; 36:2163626. [PMID: 36597824 DOI: 10.1080/14767058.2022.2163626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To characterize the data on medications for lactating people in the LactMed database and evaluate the strength of the data for the most commonly administered medications in lactating women. METHODS A retrospective analysis of all medications in the LactMed database in 12/2020 was performed. Each medication was classified into one of three categories: absent data, minimal-moderate data, strong data pertaining to safety in lactation. No data was defined as no available research studies associated with the medication. Minimal-moderate data was defined as absent research studies in one or more of the four LactMed categories: maternal drug levels, infant drug levels, effects on infants, effects on lactation, or if data was limited to a case report or observational study. Strong data was classified as availability of research studies in all four LactMed categories with data derived from pharmacokinetic/pharmacodynamic, cohort, case control, or randomized control studies. Additionally, the most commonly used medications in lactating women as defined by prior literature were analyzed for strength of data. RESULTS 1408 medications were evaluated: 714 (51%) had no associated data, 664 (47%) had minimal-moderate data, and 30 (2%) had strong data. Maternal drug level category had the highest proportion of rigorous supportive data while the effect on lactation category had the least supportive data. Of the most common mediations used in lactating women, sex hormones (contraception) and the nervous system medication classes had the most robust supportive data while respiratory, blood forming organs, and galactogogues had the weakest supportive data. CONCLUSION There is significant variability and dearth in the quality of data guiding recommendations for use of medications in lactation providing numerous opportunities for research.
Collapse
Affiliation(s)
- Yevgenia Y Fomina
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Parkland Health, Dallas, TX, USA
| | - John J Byrne
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Parkland Health, Dallas, TX, USA
| | - Catherine Y Spong
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Parkland Health, Dallas, TX, USA
| |
Collapse
|
4
|
Chen M, Du R, Zhang T, Li C, Bao W, Xin F, Hou S, Yang Q, Chen L, Wang Q, Zhu A. The Application of a Physiologically Based Toxicokinetic Model in Health Risk Assessment. TOXICS 2023; 11:874. [PMID: 37888724 PMCID: PMC10611306 DOI: 10.3390/toxics11100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Toxicokinetics plays a crucial role in the health risk assessments of xenobiotics. Classical compartmental models are limited in their ability to determine chemical concentrations in specific organs or tissues, particularly target organs or tissues, and their limited interspecific and exposure route extrapolation hinders satisfactory health risk assessment. In contrast, physiologically based toxicokinetic (PBTK) models quantitatively describe the absorption, distribution, metabolism, and excretion of chemicals across various exposure routes and doses in organisms, establishing correlations with toxic effects. Consequently, PBTK models serve as potent tools for extrapolation and provide a theoretical foundation for health risk assessment and management. This review outlines the construction and application of PBTK models in health risk assessment while analyzing their limitations and future perspectives.
Collapse
Affiliation(s)
- Mengting Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Ruihu Du
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Tao Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Chutao Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Wenqiang Bao
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Fan Xin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qiaomei Yang
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Li Chen
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, China
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| |
Collapse
|
5
|
Cardoso E, Guidi M, Nauwelaerts N, Nordeng H, Teil M, Allegaert K, Smits A, Gandia P, Edginton A, Ito S, Annaert P, Panchaud A. Safety of medicines during breastfeeding - from case report to modeling : A contribution from the ConcePTION project. Expert Opin Drug Metab Toxicol 2023. [PMID: 37269321 DOI: 10.1080/17425255.2023.2221847] [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: 12/06/2022] [Accepted: 06/01/2023] [Indexed: 06/05/2023]
Abstract
INTRODUCTION Despite many research efforts, current data on the safety of medicines during breastfeeding are either fragmented or lacking, resulting in restrictive labeling of most medicines. In the absence of pharmacoepidemiologic safety studies, risk estimation for breastfed infants is mainly derived from pharmacokinetic (PK) information on the medicine. This manuscript provides a description and a comparison of the different methodological approaches that can yield reliable information on medicine transfer into human milk and the resulting infant exposure. AREA COVERED Currently, most information on medicine transfer in human milk relies on case reports or traditional PK studies, which generate data that can hardly be generalized to the population. Some methodological approaches, such as population PK (popPK) and physiologically-based PK (PBPK) modeling, can be used to provide a more complete characterization of infant medicine exposure through human milk and simulate the most extreme situations, while decreasing the burden of sampling in breastfeeding women. EXPERT OPINION PBPK and popPK modeling are promising approaches to fill the gap of knowledge in medicine safety in breastfeeding, as illustrated with our escitalopram example.
Collapse
Affiliation(s)
- Evelina Cardoso
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nina Nauwelaerts
- Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Hedvig Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, PharmaTox Strategic Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Karel Allegaert
- Child and Youth Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy,erasmus MC, Rotterdam, GA, The Netherlands
| | - Anne Smits
- Child and Youth Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Peggy Gandia
- Laboratory of Pharmacokinetics and Toxicology, Purpan Hospital, University Hospital of Toulouse
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, ON, Canada
| | - Pieter Annaert
- Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Alice Panchaud
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Quinney SK, Bies RR, Grannis SJ, Bartlett CW, Mendonca E, Rogerson CM, Backes CH, Shah DK, Tillman EM, Costantine MM, Aruldhas BW, Allam R, Grant A, Abbasi MY, Kandasamy M, Zang Y, Wang L, Shendre A, Li L. The MPRINT Hub Data, Model, Knowledge and Research Coordination Center: Bridging the gap in maternal-pediatric therapeutics research through data integration and pharmacometrics. Pharmacotherapy 2023; 43:391-402. [PMID: 36625779 PMCID: PMC10192201 DOI: 10.1002/phar.2765] [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: 08/15/2022] [Revised: 11/13/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023]
Abstract
Maternal and pediatric populations have historically been considered "therapeutic orphans" due to their limited inclusion in clinical trials. Physiologic changes during pregnancy and lactation and growth and maturation of children alter pharmacokinetics (PK) and pharmacodynamics (PD) of drugs. Precision therapy in these populations requires knowledge of these effects. Efforts to enhance maternal and pediatric participation in clinical studies have increased over the past few decades. However, studies supporting precision therapeutics in these populations are often small and, in isolation, may have limited impact. Integration of data from various studies, for example through physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling or bioinformatics approaches, can augment the value of data from these studies, and help identify gaps in understanding. To catalyze research in maternal and pediatric precision therapeutics, the Obstetric and Pediatric Pharmacology and Therapeutics Branch of the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) established the Maternal and Pediatric Precision in Therapeutics (MPRINT) Hub. Herein, we provide an overview of the status of maternal-pediatric therapeutics research and introduce the Indiana University-Ohio State University MPRINT Hub Data, Model, Knowledge and Research Coordination Center (DMKRCC), which aims to facilitate research in maternal and pediatric precision therapeutics through the integration and assessment of existing knowledge, supporting pharmacometrics and clinical trials design, development of new real-world evidence resources, educational initiatives, and building collaborations among public and private partners, including other NICHD-funded networks. By fostering use of existing data and resources, the DMKRCC will identify critical gaps in knowledge and support efforts to overcome these gaps to enhance maternal-pediatric precision therapeutics.
Collapse
Affiliation(s)
- Sara K Quinney
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert R Bies
- Department of Pharmaceutical Sciences, University at Buffalo School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Institute for Computational and Data Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, USA
| | - Shaun J Grannis
- Department of Family Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Christopher W Bartlett
- The Steve & Cindy Rasmussen Institute for Genomic Medicine, Battelle Center for Computational Biology, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Eneida Mendonca
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department Biostatistics and Health Data Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Colin M Rogerson
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Carl H Backes
- Division of Neonatology, Nationwide Children’s Hospital; Departments of Pediatrics and Obstetrics and Gynecology, The Ohio State University College of Medicine; Center for Perinatal Research and The Ohio Perinatal Research Network, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, USA; The Heart Center at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, University at Buffalo School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Emma M Tillman
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Maged M Costantine
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - Blessed W Aruldhas
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Department of Pharmacology and Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Reva Allam
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Amelia Grant
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Mohammed Yaseen Abbasi
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Murugesh Kandasamy
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Yong Zang
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department Biostatistics and Health Data Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lei Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Aditi Shendre
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Dubbelboer IR, Le Roux-Pullen L, Gehring R. Systematic review of physiologically based kinetic lactation models for transfer of xenobiotic compounds to milk. Toxicol Appl Pharmacol 2023; 467:116495. [PMID: 36996912 DOI: 10.1016/j.taap.2023.116495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Lactational elimination has been described mathematically for nearly 50 years. Over 40 published articles, containing >50 physiologically based kinetic (PBK) lactation models were included in the systematic review. These PBK models described the lactational elimination of xenobiotic compounds in humans, rats, mice, and dairy cows and goats. A total of 78 compounds have been modelled, ranging from industrial chemicals, pesticides, to pain medication, antibiotics, and caffeine. Few models included several species or compounds, and models were thus generally not translational or generic. Three dairy cow models mechanistically described the intramammary disposition of pharmaceuticals after intramammary administration, including volume changes caused by milking, while empirically describing the remaining pharmacokinetics. The remaining models were semi- or whole body PBK models, describing long-term exposure of environmental pollutants, or short-term exposure of pharmaceuticals. The absolute majority described the disposition to the mammary gland or milk with perfusion limited compartments, but permeability limited models were available as well. With long-term exposure, models often included changes in milk volume and/or consumption by the offspring, and changes in body weight of offspring. Periodic emptying of the mammary gland, as with feeding or milking, was sparsely applied. Rodent models used similar physiological parameters, while values of physiological parameters applied in human models could range widely. When milk composition was included in the models, it most often included the fat content. The review gives an extensive overview of the applied functions and modelling strategies of PBK lactation models.
Collapse
|
8
|
Kapraun DF, Sfeir M, Pearce RG, Davidson-Fritz SE, Lumen A, Dallmann A, Judson RS, Wambaugh JF. Evaluation of a rapid, generic human gestational dose model. Reprod Toxicol 2022; 113:172-188. [PMID: 36122840 PMCID: PMC9761697 DOI: 10.1016/j.reprotox.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/30/2022] [Accepted: 09/14/2022] [Indexed: 10/14/2022]
Abstract
Chemical risk assessment considers potentially susceptible populations including pregnant women and developing fetuses. Humans encounter thousands of chemicals in their environments, few of which have been fully characterized. Toxicokinetic (TK) information is needed to relate chemical exposure to potentially bioactive tissue concentrations. Observational data describing human gestational exposures are unavailable for most chemicals, but physiologically based TK (PBTK) models estimate such exposures. Development of chemical-specific PBTK models requires considerable time and resources. As an alternative, generic PBTK approaches describe a standardized physiology and characterize chemicals with a set of standard physical and TK descriptors - primarily plasma protein binding and hepatic clearance. Here we report and evaluate a generic PBTK model of a human mother and developing fetus. We used a published set of formulas describing the major anatomical and physiological changes that occur during pregnancy to augment the High-Throughput Toxicokinetics (httk) software package. We simulated the ratio of concentrations in maternal and fetal plasma and compared to literature in vivo measurements. We evaluated the model with literature in vivo time-course measurements of maternal plasma concentrations in pregnant and non-pregnant women. Finally, we prioritized chemicals measured in maternal serum based on predicted fetal brain concentrations. This new model can be used for TK simulations of 859 chemicals with existing human-specific in vitro TK data as well as any new chemicals for which such data become available. This gestational model may allow for in vitro to in vivo extrapolation of point of departure doses relevant to reproductive and developmental toxicity.
Collapse
Affiliation(s)
- Dustin F Kapraun
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Mark Sfeir
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA
| | - Robert G Pearce
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA
| | - Sarah E Davidson-Fritz
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Annie Lumen
- National Center for Toxicological Research, US Food and Drug Administration, USA
| | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - Richard S Judson
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - John F Wambaugh
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
9
|
Nauwelaerts N, Deferm N, Smits A, Bernardini C, Lammens B, Gandia P, Panchaud A, Nordeng H, Bacci ML, Forni M, Ventrella D, Van Calsteren K, DeLise A, Huys I, Bouisset-Leonard M, Allegaert K, Annaert P. A comprehensive review on non-clinical methods to study transfer of medication into breast milk - A contribution from the ConcePTION project. Biomed Pharmacother 2021; 136:111038. [PMID: 33526310 DOI: 10.1016/j.biopha.2020.111038] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022] Open
Abstract
Breastfeeding plays a major role in the health and wellbeing of mother and infant. However, information on the safety of maternal medication during breastfeeding is lacking for most medications. This leads to discontinuation of either breastfeeding or maternal therapy, although many medications are likely to be safe. Since human lactation studies are costly and challenging, validated non-clinical methods would offer an attractive alternative. This review gives an extensive overview of the non-clinical methods (in vitro, in vivo and in silico) to study the transfer of maternal medication into the human breast milk, and subsequent neonatal systemic exposure. Several in vitro models are available, but model characterization, including quantitative medication transport data across the in vitro blood-milk barrier, remains rather limited. Furthermore, animal in vivo models have been used successfully in the past. However, these models don't always mimic human physiology due to species-specific differences. Several efforts have been made to predict medication transfer into the milk based on physicochemical characteristics. However, the role of transporter proteins and several physiological factors (e.g., variable milk lipid content) are not accounted for by these methods. Physiologically-based pharmacokinetic (PBPK) modelling offers a mechanism-oriented strategy with bio-relevance. Recently, lactation PBPK models have been reported for some medications, showing at least the feasibility and value of PBPK modelling to predict transfer of medication into the human milk. However, reliable data as input for PBPK models is often missing. The iterative development of in vitro, animal in vivo and PBPK modelling methods seems to be a promising approach. Human in vitro models will deliver essential data on the transepithelial transport of medication, whereas the combination of animal in vitro and in vivo methods will deliver information to establish accurate in vitro/in vivo extrapolation (IVIVE) algorithms and mechanistic insights. Such a non-clinical platform will be developed and thoroughly evaluated by the Innovative Medicines Initiative ConcePTION.
Collapse
Affiliation(s)
- Nina Nauwelaerts
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| | - Neel Deferm
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| | - Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, UZ Leuven, Neonatology, Herestraat 49, 3000, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Belgium.
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | | | - Peggy Gandia
- Laboratoire de Pharmacocinétique et Toxicologie, Centre Hospitalier Universitaire de Toulouse, France.
| | - Alice Panchaud
- Service of Pharmacy Service, Lausanne University Hospital and University of Lausanne, Switzerland; Institute of Primary Health Care (BIHAM), University of Bern, Switzerland
| | - Hedvig Nordeng
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, University of Oslo, PB. 1068 Blindern, 0316, Oslo, Norway.
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | - Domenico Ventrella
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | | | - Anthony DeLise
- Novartis Pharmaceuticals Corporation, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, NJ, 07936, USA.
| | - Isabelle Huys
- KU Leuven, Department of Clinical Pharmacology and Pharmacotherapy, ON II Herestraat 49 - bus, 521 3000, Leuven, Belgium.
| | - Michele Bouisset-Leonard
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Werk Klybeck Postfach, Basel, CH-4002, Switzerland.
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Belgium; KU Leuven, Department of Clinical Pharmacology and Pharmacotherapy, ON II Herestraat 49 - bus, 521 3000, Leuven, Belgium; Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands.
| | - Pieter Annaert
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| |
Collapse
|
10
|
Coppola P, Kerwash E, Cole S. Physiologically Based Pharmacokinetics Model in Pregnancy: A Regulatory Perspective on Model Evaluation. Front Pediatr 2021; 9:687978. [PMID: 34249817 PMCID: PMC8262675 DOI: 10.3389/fped.2021.687978] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Physiologically based pharmacokinetics (PBPK) modelling is widely used in medicine development and regulatory submissions. The lack of clinical pharmacokinetic data in pregnancy is widely acknowledged; therefore, one area of current interest is in the use of PBPK modelling to describe the potential impact of anatomical and physiological changes during pregnancy on the medicine's pharmacokinetics. PBPK modelling could possibly represent a predictive tool to support the medicine benefit-risk decision and inform dose adjustment in this population and also to investigate medicine levels in the foetus to support the risk assessment to the foetus. In the context of regulatory application, there are, however, a number of considerations around model evaluation, and this should be tailored to the model purpose, in order to inform the confidence in the model for the intended application. A number of gestational age-related physiological changes are expected to alter the pharmacokinetics of medicines during pregnancy, and there are uncertainties on some parameters; therefore, well-qualified models are needed to improve assurance in the model prediction before this approach can be used to inform with confidence high-impact decisions as part of regulatory submissions.
Collapse
Affiliation(s)
- Paola Coppola
- Medicines and Healthcare Products Regulatory Agency, London, United Kingdom
| | - Essam Kerwash
- Medicines and Healthcare Products Regulatory Agency, London, United Kingdom
| | - Susan Cole
- Medicines and Healthcare Products Regulatory Agency, London, United Kingdom
| |
Collapse
|
11
|
Eke AC, Olagunju A, Best BM, Mirochnick M, Momper JD, Abrams E, Penazzato M, Cressey TR, Colbers A. Innovative Approaches for Pharmacology Studies in Pregnant and Lactating Women: A Viewpoint and Lessons from HIV. Clin Pharmacokinet 2020; 59:1185-1194. [PMID: 32757103 PMCID: PMC7550310 DOI: 10.1007/s40262-020-00915-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Medication use during pregnancy in the absence of pharmacokinetic and safety data is common, particularly for antiretrovirals, as pregnant women are not usually included in clinical trials leading to drug licensure. To date, data are typically generated through opportunistic pregnancy studies performed in the postmarketing setting, leading to a substantial time-lag between initial regulatory approval of a drug and availability of essential pregnancy-specific pharmacokinetic and safety data. During this period, health care providers lack key information on human placental transfer, fetal exposure, optimal maternal dosing in pregnancy, and maternal and fetal drug toxicity, including teratogenicity risk. We discuss new approaches that could facilitate the acquisition of these critical data earlier in the drug development process, aiding clinicians and patients in making informed decisions on drug selection and dosing during pregnancy. An integrated approach utilizing multiple novel methodologies (in vitro, ex vivo, in silico and in vivo) is needed to accelerate the availability of pharmacology data in pregnancy and lactation.
Collapse
Affiliation(s)
- Ahizechukwu C Eke
- Division of Maternal-Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, 600N Wolfe Street, Phipps 215, Baltimore, MD, 21287, USA
| | - Adeniyi Olagunju
- Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Brookie M Best
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, USA
- Pediatrics Department, University of California San Diego School of Medicine-Rady Children's Hospital San Diego, San Diego, CA, USA
| | | | - Jeremiah D Momper
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, USA
| | - Elaine Abrams
- Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Martina Penazzato
- HIV, Hepatitis and STI Department, World Health Organization, Geneva, Switzerland
| | - Tim R Cressey
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- PHPT/IRD 174, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Immunology and Infectious Diseases, Harvard T.H Chan School of Public Health, Boston, MA, USA
| | - Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Abduljalil K, Jamei M, Johnson TN. Fetal Physiologically Based Pharmacokinetic Models: Systems Information on Fetal Blood Components and Binding Proteins. Clin Pharmacokinet 2019; 59:629-642. [PMID: 31696406 DOI: 10.1007/s40262-019-00836-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fetal blood and plasma volume and binding components are critical parameters in a fetal physiologically based pharmacokinetic model. To date, a comprehensive review of their changes during fetal development has not been reported. OBJECTIVE The objective of this work was to collate and analyze physiological information on fetal blood and plasma volume and binding component data during development and to provide a mathematical description of these parameters that can be integrated within a fetal physiologically based pharmacokinetic model. METHODS A comprehensive literature search was conducted on fetal blood and plasma volume and binding component parameters and their changes during growth from apparently healthy fetuses from uncomplicated pregnancies. Collated data were assessed, integrated, and analyzed to establish continuous mathematical functions describing their growth trends with fetal age and weight. RESULTS Data were available from 14 studies for blood, ten studies for hematocrit, 12 studies for albumin, and four studies for alpha-1-acid glycoprotein, while plasma and red blood cell volumes were described based on blood and hematocrit data. Fetal physiologically based pharmacokinetic parameters, including blood, plasma and red blood cell volumes, hematocrit, serum albumin, and acid glycoprotein were quantified as a function of fetal age and weight. Variability around the mean parameters at different fetal ages was also investigated. The growth of each of these parameters was different (with respect to direction and monotonicity). CONCLUSIONS Despite the limitations identified in the availability of some values, the collected data presented in this article provide a useful resource for fetal physiologically based pharmacokinetic modeling. Potential applications include predicting xenobiotic exposure and risk assessment in the fetus following maternally administered drugs or unintended exposure to environmental toxicants.
Collapse
Affiliation(s)
- Khaled Abduljalil
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | - Masoud Jamei
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Trevor N Johnson
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| |
Collapse
|
13
|
Codaccioni M, Bois F, Brochot C. Placental transfer of xenobiotics in pregnancy physiologically-based pharmacokinetic models: Structure and data. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.comtox.2019.100111] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Kapraun DF, Wambaugh JF, Setzer RW, Judson RS. Empirical models for anatomical and physiological changes in a human mother and fetus during pregnancy and gestation. PLoS One 2019; 14:e0215906. [PMID: 31048866 PMCID: PMC6497258 DOI: 10.1371/journal.pone.0215906] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/10/2019] [Indexed: 12/28/2022] Open
Abstract
Many parameters treated as constants in traditional physiologically based pharmacokinetic models must be formulated as time-varying quantities when modeling pregnancy and gestation due to the dramatic physiological and anatomical changes that occur during this period. While several collections of empirical models for such parameters have been published, each has shortcomings. We sought to create a repository of empirical models for tissue volumes, blood flow rates, and other quantities that undergo substantial changes in a human mother and her fetus during the time between conception and birth, and to address deficiencies with similar, previously published repositories. We used maximum likelihood estimation to calibrate various models for the time-varying quantities of interest, and then used the Akaike information criterion to select an optimal model for each quantity. For quantities of interest for which time-course data were not available, we constructed composite models using percentages and/or models describing related quantities. In this way, we developed a comprehensive collection of formulae describing parameters essential for constructing a PBPK model of a human mother and her fetus throughout the approximately 40 weeks of pregnancy and gestation. We included models describing blood flow rates through various fetal blood routes that have no counterparts in adults. Our repository of mathematical models for anatomical and physiological quantities of interest provides a basis for PBPK models of human pregnancy and gestation, and as such, it can ultimately be used to support decision-making with respect to optimal pharmacological dosing and risk assessment for pregnant women and their developing fetuses. The views expressed in this article are those of the authors and do not necessarily represent the views or policies of the U.S. Environmental Protection Agency.
Collapse
Affiliation(s)
- Dustin F. Kapraun
- National Center for Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| | - John F. Wambaugh
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - R. Woodrow Setzer
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Richard S. Judson
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
15
|
Biesdorf C, Martins FS, Sy SKB, Diniz A. Physiologically-based pharmacokinetics of ziprasidone in pregnant women. Br J Clin Pharmacol 2019; 85:914-923. [PMID: 30669177 DOI: 10.1111/bcp.13872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/29/2018] [Accepted: 01/06/2019] [Indexed: 01/19/2023] Open
Abstract
AIMS Pregnancy is associated with physiological changes that alter the pharmacokinetics (PK) of drugs. The aim of this study was to predict the PK of ziprasidone in pregnant women. METHODS A full physiologically-based pharmacokinetic (PBPK) model of ziprasidone was developed and validated for the non-pregnant population (healthy adults, paediatrics, geriatrics), and this was extended to the pregnant state to assess the change in PK profile of ziprasidone throughout pregnancy. RESULTS The PBPK model successfully predicted the ziprasidone disposition in healthy adult volunteers, wherein the predicted and observed AUC, Cmax and tmax were within the fold-difference of 0.94-1.09, 0.89-1.40 and 0.80-1.08, respectively. The paediatric and geriatric population, also showed predicted AUC, Cmax and tmax within a two-fold range of the observed values. The simulated exposure in pregnant women using a p-PBPK model showed no significant difference when compared to non-pregnant women. CONCLUSIONS The PBPK model predicted the impact of physiological changes during pregnancy on PK and exposure of ziprasidone, suggesting that dose adjustment is not necessary in this special population.
Collapse
Affiliation(s)
- Carla Biesdorf
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| | | | - Sherwin K B Sy
- Department of Statistics, State University of Maringá, Maringá, Brazil
| | - Andrea Diniz
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| |
Collapse
|
16
|
Chebekoue SF, Krishnan K. A framework for application of quantitative property-property relationships (QPPRs) in physiologically based pharmacokinetic (PBPK) models for high-throughput prediction of internal dose of inhaled organic chemicals. CHEMOSPHERE 2019; 215:634-646. [PMID: 30347358 DOI: 10.1016/j.chemosphere.2018.10.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/03/2018] [Accepted: 10/06/2018] [Indexed: 06/08/2023]
Abstract
New generation of toxicological tests and assessment strategies require validated toxicokinetic data or models that are lacking for most chemicals. This study aimed at developing a quantitative property-property relationship (QPPR)-based human physiologically based pharmacokinetic (PBPK) modeling framework for high-throughput predictions of inhalation toxicokinetics of organic chemicals. A PBPK model was parameterized with QPPR-derived values for hepatic clearance (CLh) and partition coefficients (P) [blood:air (Pba) and tissue:air (Pta) and tissue:blood (Ptb)]. The model was initially applied to an evaluation dataset of 40 organic chemicals in the applicability domain, and then to an expanded dataset of 249 organic chemicals from diverse chemical classes. 'Batch' analyses were performed for rapid assessments of hundreds of chemicals. The simulations of inhalation toxicokinetics following an 8-h exposure to 1 ppm of each chemical were successful. The mean ratios of their predicted-to-experimental values were within a factor of 1.36-2.36 for Ptb and 1.18 for CLh, for 80% of the chemicals in the evaluation dataset. The predicted 24-h area under the venous blood concentration-time curve (AUC24) values were within the predicted envelopes obtained while using experimental values of Pba and considering either no or maximal hepatic extraction. The reliability analysis (based on combined sensitivity and uncertainty analyses) indicated that AUC24 predictions for 55% of the expanded dataset were moderately to highly reliable, with 46% exhibiting highly reliable values. Overall, the modeling framework suggests that molecular structure and chemical properties can together be effectively used to obtain first-cut estimates of the toxicokinetics of data-poor organic chemicals for screening and prioritization purposes.
Collapse
Affiliation(s)
- Sandrine F Chebekoue
- École de Santé Publique de l'Université de Montréal (ESPUM), Montréal, Québec, Canada.
| | - Kannan Krishnan
- École de Santé Publique de l'Université de Montréal (ESPUM), Montréal, Québec, Canada; Institut de recherche Robert-Sauvé en santé et en sécurité du travail (IRSST), Montréal, Québec, Canada.
| |
Collapse
|
17
|
Neal-Kluever A, Fisher J, Grylack L, Kakiuchi-Kiyota S, Halpern W. Physiology of the Neonatal Gastrointestinal System Relevant to the Disposition of Orally Administered Medications. Drug Metab Dispos 2018; 47:296-313. [DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
|
18
|
Sharma RP, Schuhmacher M, Kumar V. The development of a pregnancy PBPK Model for Bisphenol A and its evaluation with the available biomonitoring data. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 624:55-68. [PMID: 29247905 DOI: 10.1016/j.scitotenv.2017.12.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/03/2017] [Accepted: 12/03/2017] [Indexed: 05/20/2023]
Abstract
Recent studies suggest universal fetal exposure to Bisphenol A (BPA) and its association with the adverse birth outcomes. Estimation of the fetal plasma BPA concentration from the maternal plasma BPA would be highly useful to predict its associated risk to this specific population. The objective of current work is to develop a pregnancy-physiologically based pharmacokinetic (P-PBPK) model to predict the toxicokinetic profile of BPA in the fetus during gestational growth, and to evaluate the developed model using biomonitoring data obtained from different pregnancy cohort studies. To achieve this objective, first, the adult PBPK model was developed and validated with the human BPA toxicokinetic data. This validated human PBPK model was extended to develop a P-PBPK model, which included the physiological changes during pregnancy and the fetus sub-model. The developed model would be able to predict the BPA pharmacokinetics (PKs) in both mother and fetus. Transplacental BPA kinetics parameters for this study were taken from the previous pregnant mice study. Both oral and dermal exposure routes were included into the model to simulate total BPA internal exposure. The impact of conjugation and deconjugation of the BPA and its metabolites on fetal PKs was investigated. The developed P-PBPK model was evaluated against the observed BPA concentrations in cord blood, fetus liver and amniotic fluid considering maternal blood concentration as an exposure source. A range of maternal exposure dose for the oral and dermal routes was estimated, so that simulation concentration matched the observed highest and lowest mother plasma concentration in different cohorts' studies. The developed model could be used to address the concerns regarding possible adverse health effects in the fetus being exposed to BPA and might be useful in identifying critical windows of exposure during pregnancy.
Collapse
Affiliation(s)
- Raju Prasad Sharma
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Marta Schuhmacher
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Vikas Kumar
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain.
| |
Collapse
|
19
|
Olagunju A, Rajoli RKR, Atoyebi SA, Khoo S, Owen A, Siccardi M. Physiologically-based pharmacokinetic modelling of infant exposure to efavirenz through breastfeeding. AAS Open Res 2018. [DOI: 10.12688/aasopenres.12860.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background: Very little is known about the level of infant exposure to many drugs commonly used during breastfeeding. The aim of this study was to develop a physiologically-based pharmacokinetic (PBPK) model for predicting infant exposure to maternal efavirenz through breastmilk. Methods: A breastfeeding PBPK model combining whole-body maternal and infant sub-models was constructed from drug-specific and system parameters affecting drug disposition using mathematical descriptions. The model was validated against published data on the pharmacokinetics of efavirenz in nursing mother-infant pairs. Further simulations were conducted to assess exposure in the context of the 400 mg reduced dose of efavirenz as well as best- and worse-case scenarios. Results: The model adequately described efavirenz pharmacokinetics, with over 80% of observed data points (203 matched breast milk and plasma pairs) within the predictive interval. All parameters were within 2-fold difference of clinical data. Median (range) predicted versus observed breast milk AUC0-24, Cmax and Cmin at the standard 600 mg dose were 75.0 (18.5-324) versus 68.5 (26.3-257) µg.hr/mL, 4.56 (1.17-16.0) versus 5.39 (1.43-18.4) µg/mL, and 2.11 (0.38-12.3) versus 1.68 (0.316-9.57) µg/mL, respectively. Predicted plasma AUC0-24, Cmax and Cmin at 400 mg reduced dose were similar to clinical data from non-breastfeeding adults. Model-predicted infant plasma concentrations were similar to clinical data, 0.15 (0.026–0.78) μg/mL at the 400 mg maternal dose in pooled analysis, approximately 25% lower than simulated exposure at 600 mg. The maximum exposure index was observed in the youngest infants, 5.9% (2.2-20) at 400 mg and 8.7% (3.2-29) at 600 mg. Thirteen and 36% of 10 days-1 month old infants were predicted to have exposure index above the 10% recommended threshold at 400 mg and 600 mg maternal dose, respectively. Conclusions: This application of PBPK modelling opens up opportunities for expanding our understanding of infant exposure to maternal drugs through breastfeeding.
Collapse
|
20
|
Abstract
One impediment to breastfeeding is the lack of information on the use of many drugs during lactation, especially newer ones. The principles of drug passage into breastmilk are well established, but have often not been optimally applied prospectively. Commonly used preclinical rodent models for determining drug excretion into milk are very unreliable because of marked differences in milk composition and transporters compared to those of humans. Measurement of drug concentrations in humans remains the gold standard, but computer modeling is promising. New FDA labeling requirements present an opportunity to apply modeling to preclinical drug development in place of conventional animal testing for drug excretion into breastmilk, which should improve the use of medications in nursing mothers.
Collapse
|
21
|
Illamola SM, Bucci‐Rechtweg C, Costantine MM, Tsilou E, Sherwin CM, Zajicek A. Inclusion of pregnant and breastfeeding women in research - efforts and initiatives. Br J Clin Pharmacol 2018; 84:215-222. [PMID: 28925019 PMCID: PMC5777434 DOI: 10.1111/bcp.13438] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/01/2017] [Accepted: 09/09/2017] [Indexed: 01/06/2023] Open
Abstract
Pregnant and breastfeeding women have been rendered therapeutic orphans as they have been historically excluded from clinical trials. Labelling for most approved drugs does not provide information about safety and efficacy during pregnancy. This lack of data is mainly due to ethico-legal challenges that have remained entrenched in the post-diethylstilbestrol and thalidomide era, and that have led to pregnancy being viewed in the clinical trial setting primarily through a pharmacovigilance lens. Policy considerations that encourage and/or require the inclusion of pregnant or lactating women in clinical trials may address the current lack of available information. However, there are additional pragmatic strategies, such the employment of pharmacometric tools and the introduction of innovative clinical trial designs, which could improve knowledge about the safety and efficacy of medication use during pregnancy and lactation. This paper provides a broad overview of the pharmacoepidemiology of drugs used during pregnancy and lactation, and offers recommendations for regulators and researchers in academia and industry to increase the available pharmacokinetic and -dynamic understanding of medication use in pregnancy.
Collapse
Affiliation(s)
- Sílvia M. Illamola
- Division of Clinical Pharmacology, Department of PediatricsUniversity of Utah School of MedicineSalt Lake CityUTUSA
| | - Christina Bucci‐Rechtweg
- Pediatric & Maternal Health Policy, Global Drug Regulatory AffairsNovartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Maged M. Costantine
- Department of Obstetrics and Gynecology, Division of Maternal‐Fetal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Ekaterini Tsilou
- Obstetric and Pediatric Pharmacology and Therapeutics Branch at the Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMDUSA
| | - Catherine M. Sherwin
- Division of Clinical Pharmacology, Department of PediatricsUniversity of Utah School of MedicineSalt Lake CityUTUSA
- Department of PharmacotherapyUniversity of Utah College of PharmacySalt Lake CityUTUSA
| | - Anne Zajicek
- Obstetric and Pediatric Pharmacology and Therapeutics Branch at the Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMDUSA
| |
Collapse
|
22
|
Ke AB, Greupink R, Abduljalil K. Drug Dosing in Pregnant Women: Challenges and Opportunities in Using Physiologically Based Pharmacokinetic Modeling and Simulations. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:103-110. [PMID: 29349870 PMCID: PMC5824116 DOI: 10.1002/psp4.12274] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/22/2017] [Accepted: 12/28/2017] [Indexed: 01/04/2023]
Abstract
The unmet medical need of providing evidence‐based pharmacotherapy for pregnant women is recognized by the regulatory bodies. Physiologically based pharmacokinetic (PBPK) modeling offers an attractive platform to quantify anticipated changes in the pharmacokinetics (PKs) of drugs during pregnancy. Recent publications applying a pregnancy PBPK module to the prediction of maternal and fetal exposure of drugs are summarized. Future opportunities to use PBPK models to predict breast milk exposure and assess human fetotoxicity risks are presented.
Collapse
Affiliation(s)
- Alice Ban Ke
- Simcyp Limited (a Certara company), Sheffield, UK
| | - Rick Greupink
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | | |
Collapse
|
23
|
Quinney SK, Gullapelli R, Haas DM. Translational Systems Pharmacology Studies in Pregnant Women. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 7:69-81. [PMID: 29239132 PMCID: PMC5824114 DOI: 10.1002/psp4.12269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/26/2022]
Abstract
Pregnancy involves rapid physiological adaptation and complex interplay between mother and fetus. New analytic technologies provide large amounts of genomic, proteomic, and metabolomics data. The integration of these data through bioinformatics, statistical, and systems pharmacology techniques can improve our understanding of the mechanisms of normal maternal physiologic changes and fetal development. New insights into the mechanisms of pregnancy‐related disorders, such as preterm birth (PTB), may lead to the development of new therapeutic interventions and novel biomarkers.
Collapse
Affiliation(s)
- Sara K Quinney
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rakesh Gullapelli
- School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - David M Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Poet TS, Timchalk C, Bartels MJ, Smith JN, McDougal R, Juberg DR, Price PS. Use of a probabilistic PBPK/PD model to calculate Data Derived Extrapolation Factors for chlorpyrifos. Regul Toxicol Pharmacol 2017; 86:59-73. [PMID: 28238854 DOI: 10.1016/j.yrtph.2017.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/24/2017] [Accepted: 02/17/2017] [Indexed: 11/16/2022]
Abstract
A physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) model combined with Monte Carlo analysis of inter-individual variation was used to assess the effects of the insecticide, chlorpyrifos and its active metabolite, chlorpyrifos oxon in humans. The PBPK/PD model has previously been validated and used to describe physiological changes in typical individuals as they grow from birth to adulthood. This model was updated to include physiological and metabolic changes that occur with pregnancy. The model was then used to assess the impact of inter-individual variability in physiology and biochemistry on predictions of internal dose metrics and quantitatively assess the impact of major sources of parameter uncertainty and biological diversity on the pharmacodynamics of red blood cell acetylcholinesterase inhibition. These metrics were determined in potentially sensitive populations of infants, adult women, pregnant women, and a combined population of adult men and women. The parameters primarily responsible for inter-individual variation in RBC acetylcholinesterase inhibition were related to metabolic clearance of CPF and CPF-oxon. Data Derived Extrapolation Factors that address intra-species physiology and biochemistry to replace uncertainty factors with quantitative differences in metrics were developed in these same populations. The DDEFs were less than 4 for all populations. These data and modeling approach will be useful in ongoing and future human health risk assessments for CPF and could be used for other chemicals with potential human exposure.
Collapse
Affiliation(s)
| | | | | | - Jordan N Smith
- Battelle, Pacific Northwest Division, Richland, WA, 99354, USA
| | - Robin McDougal
- Dug Safety and Metabolism, AstraZeneca, Gatehouse Park, Waltham, Boston, 02451, USA
| | | | | |
Collapse
|
25
|
State of the art on public risk assessment of combined human exposure to multiple chemical contaminants. Trends Food Sci Technol 2016. [DOI: 10.1016/j.tifs.2016.06.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
26
|
Emond C, DeVito M, Warner M, Eskenazi B, Mocarelli P, Birnbaum LS. An assessment of dioxin exposure across gestation and lactation using a PBPK model and new data from Seveso. ENVIRONMENT INTERNATIONAL 2016; 92-93:23-32. [PMID: 27045706 PMCID: PMC4902767 DOI: 10.1016/j.envint.2016.03.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/27/2016] [Accepted: 03/14/2016] [Indexed: 05/09/2023]
Abstract
On July 10, 1976, an explosion at a chemical plant in Seveso, Italy, released up to 30kg of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-the most potent dioxin congener. Twenty years later, the Seveso Women's Health Study (SWHS) initiated a follow-up assessment of a cohort of female Seveso residents. Researchers collected serial blood, measured for TCDD levels, and recorded information about the women's medical history after the explosion. The study's aims were to: 1) modify the human PBPK model for TCDD (Emond et al. 2004; Emond et al. 2005; NCEA-USEPA, 2010) to include repetitive gestation and lactation; 2) simulate TCDD blood concentrations during different life stages including pregnancy and lactation, under different exposure scenarios; and 3) use this PBPK model to compare the influence of gestation and lactation on elimination of TCDD. After optimization of the model, it was assessed using data from the SWHS cohort. The 23 women in Subcohort A, were 4-39years old and in Subcohort B, the 18 women were 3-17years old when the explosion occurred. The model accurately predicted the blood concentrations during the 20years post-exposure, including periods of pregnancy and lactation. The model was also used to analyze the contribution of gestation and lactation to the mother's elimination of TCDD. The results suggest that gestation and lactation do not significantly impact TCDD blood elimination. Future efforts will focus on using additional data to evaluate the PBPK model and improving the mathematical descriptions of lactation and multiple gestations.
Collapse
Affiliation(s)
- C Emond
- BioSimulation Consulting Inc., Newark, DE, USA; Department of Environmental and Occupational Health, University of Montreal, Quebec, Canada.
| | - M DeVito
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, NC, USA
| | - M Warner
- School of Public Health, University of California, Berkeley, CA, USA
| | - B Eskenazi
- School of Public Health, University of California, Berkeley, CA, USA
| | - P Mocarelli
- Department of Laboratory Medicine, University of Milano-Bicocca, School of Medicine, Hospital of Desio, Desio, Milano, Italy
| | - L S Birnbaum
- National Cancer Institute, Research Triangle Park, NC, USA
| |
Collapse
|
27
|
Anderson PO, Sauberan JB. Modeling drug passage into human milk. Clin Pharmacol Ther 2016; 100:42-52. [PMID: 27060684 DOI: 10.1002/cpt.377] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/16/2016] [Accepted: 04/01/2016] [Indexed: 01/16/2023]
Abstract
Breastfeeding has positive health consequences for both the breastfed infant and the nursing mother.(1,2) Although information on drug use during lactation is available through sites such as LactMed,(3) available information is often incomplete. Unlike pregnancy, in which large numbers of pregnant women need to be studied to assure safety, measurement of drug concentrations in breastmilk in a relatively few subjects can provide valuable information to assess drug safety. This article reviews methods of measuring and predicting drug passage into breastmilk.
Collapse
Affiliation(s)
- P O Anderson
- Health Sciences Clinical Professor, University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, California, USA
| | - J B Sauberan
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, California, USA
| |
Collapse
|
28
|
Lin Z, Fisher JW, Wang R, Ross MK, Filipov NM. Estimation of placental and lactational transfer and tissue distribution of atrazine and its main metabolites in rodent dams, fetuses, and neonates with physiologically based pharmacokinetic modeling. Toxicol Appl Pharmacol 2013; 273:140-58. [DOI: 10.1016/j.taap.2013.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/07/2013] [Accepted: 08/10/2013] [Indexed: 11/27/2022]
|
29
|
Xia B, Heimbach T, Gollen R, Nanavati C, He H. A simplified PBPK modeling approach for prediction of pharmacokinetics of four primarily renally excreted and CYP3A metabolized compounds during pregnancy. AAPS JOURNAL 2013; 15:1012-24. [PMID: 23835676 DOI: 10.1208/s12248-013-9505-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 06/12/2013] [Indexed: 02/02/2023]
Abstract
During pregnancy, a drug's pharmacokinetics may be altered and hence anticipation of potential systemic exposure changes is highly desirable. Physiologically based pharmacokinetics (PBPK) models have recently been used to influence clinical trial design or to facilitate regulatory interactions. Ideally, whole-body PBPK models can be used to predict a drug's systemic exposure in pregnant women based on major physiological changes which can impact drug clearance (i.e., in the kidney and liver) and distribution (i.e., adipose and fetoplacental unit). We described a simple and readily implementable multitissue/organ whole-body PBPK model with key pregnancy-related physiological parameters to characterize the PK of reference drugs (metformin, digoxin, midazolam, and emtricitabine) in pregnant women compared with the PK in nonpregnant or postpartum (PP) women. Physiological data related to changes in maternal body weight, tissue volume, cardiac output, renal function, blood flows, and cytochrome P450 activity were collected from the literature and incorporated into the structural PBPK model that describes HV or PP women PK data. Subsequently, the changes in exposure (area under the curve (AUC) and maximum concentration (C max)) in pregnant women were simulated. Model-simulated PK profiles were overall in agreement with observed data. The prediction fold error for C max and AUC ratio (pregnant vs. nonpregnant) was less than 1.3-fold, indicating that the pregnant PBPK model is useful. The utilization of this simplified model in drug development may aid in designing clinical studies to identify potential exposure changes in pregnant women a priori for compounds which are mainly eliminated renally or metabolized by CYP3A4.
Collapse
Affiliation(s)
- Binfeng Xia
- Novartis Institutes for Biomedical Research, DMPK-Translational Sciences, One Health Plaza 436/3253, East Hanover, New Jersey, 07470, USA
| | | | | | | | | |
Collapse
|
30
|
van Hasselt JGC, Andrew MA, Hebert MF, Tarning J, Vicini P, Mattison DR. The status of pharmacometrics in pregnancy: highlights from the 3(rd) American conference on pharmacometrics. Br J Clin Pharmacol 2013; 74:932-9. [PMID: 22452385 PMCID: PMC3522806 DOI: 10.1111/j.1365-2125.2012.04280.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Physiological changes during pregnancy may alter drug pharmacokinetics. Therefore, mechanistic understanding of these changes and, ultimately, clinical studies in pregnant women are necessary to determine if and how dosing regimens should be adjusted. Because of the typically limited number of patients who can be recruited in this patient group, efficient design and analysis of these studies is of special relevance. This paper is a summary of a conference session organized at the American Conference of Pharmacometrics in April 2011, around the topic of applying pharmacometric methodology to this important problem. The discussion included both design and analysis of clinical studies during pregnancy and in silico predictions. An overview of different pharmacometric methods relevant to this subject was given. The impact of pharmacometrics was illustrated using a range of case examples of studies around pregnancy.
Collapse
Affiliation(s)
- J G Coen van Hasselt
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | | | | | | | | | | |
Collapse
|
31
|
Gaohua L, Abduljalil K, Jamei M, Johnson TN, Rostami-Hodjegan A. A pregnancy physiologically based pharmacokinetic (p-PBPK) model for disposition of drugs metabolized by CYP1A2, CYP2D6 and CYP3A4. Br J Clin Pharmacol 2013; 74:873-85. [PMID: 22725721 DOI: 10.1111/j.1365-2125.2012.04363.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AIMS Pregnant women are usually not part of the traditional drug development programme. Pregnancy is associated with major biological and physiological changes that alter the pharmacokinetics (PK) of drugs. Prediction of the changes to drug exposure in this group of patients may help to prevent under- or overtreatment. We have used a pregnancy physiologically based pharmacokinetic (p-PBPK) model to assess the likely impact of pregnancy on three model compounds, namely caffeine, metoprolol and midazolam, based on the knowledge of their disposition in nonpregnant women and information from in vitro studies. METHODS A perfusion-limited form of a 13-compartment full-PBPK model (Simcyp® Simulator) was used for the nonpregnant women, and this was extended to the pregnant state by applying known changes to all model components (including the gestational related activity of specific cytochrome P450 enzymes) and through the addition of an extra compartment to represent the fetoplacental unit. The uterus and the mammary glands were grouped into the muscle compartment. The model was implemented in Matlab Simulink and validated using clinical observations. RESULTS The p-PBPK model predicted the PK changes of three model compounds (namely caffeine, metoprolol and midazolam) for CYP1A2, CYP2D6 and CYP3A4 during pregnancy within twofold of observed values. The changes during the third trimester were predicted to be a 100% increase, a 30% decrease and a 35% decrease in the exposure of caffeine, metoprolol and midazolam, respectively, compared with the nonpregnant women. CONCLUSIONS In the absence of clinical data, the in silico prediction of PK behaviour during pregnancy can provide a valuable aid to dose adjustment in pregnant women. The performance of the model for drugs metabolized by a single enzyme to different degrees (high and low extraction) and for drugs that are eliminated by several different routes warrants further study.
Collapse
|
32
|
A Semi-Mechanistic Metabolism Model of CYP3A Substrates in Pregnancy: Predicting Changes in Midazolam and Nifedipine Pharmacokinetics. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2012; 1:e2. [PMID: 23835882 PMCID: PMC3603475 DOI: 10.1038/psp.2012.5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Physiological changes in pregnancy, including changes in body composition and metabolic enzyme activity, can alter drug pharmacokinetics. A semi-mechanistic metabolism model was developed to describe the pharmacokinetics of two cytochrome P450 3A (CYP3A) substrates, midazolam and nifedipine, in obstetrics patients. The model parameters were optimized to fit the data of oral midazolam pharmacokinetics in pregnant women, by increasing CYP3A-induced hepatic metabolism 1.6-fold in the model with no change in gut wall metabolism. Fetal metabolism had a negligible effect on maternal plasma drug concentrations. Validation of the model was performed by applying changes in volume of distribution and metabolism, consistent with those observed for midazolam, to the pharmacokinetics parameters of immediate-release nifedipine in healthy volunteers. The predicted steady-state areas under the concentration–time curve (AUCs) for nifedipine were within 15% of the data observed in pregnant women undergoing treatment for preterm labor. This model predicts the pharmacokinetics of two CYP3A substrates in pregnancy, and may be applicable to other CYP3A substrates as well.
Collapse
|
33
|
Saghir SA, Khan SA, McCoy AT. Ontogeny of mammalian metabolizing enzymes in humans and animals used in toxicological studies. Crit Rev Toxicol 2012; 42:323-57. [PMID: 22512665 DOI: 10.3109/10408444.2012.674100] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
It is well recognized that expression of enzymes varies during development and growth. However, an in-depth review of this acquired knowledge is needed to translate the understanding of enzyme expression and activity into the prediction of change in effects (e.g. kinetics and toxicity) of xenobiotics with age. Age-related changes in metabolic capacity are critical for understanding and predicting the potential differences resulting from exposure. Such information may be especially useful in the evaluation of the risk of exposure to very low (µg/kg/day or ng/kg/day) levels of environmental chemicals. This review is to better understand the ontogeny of metabolizing enzymes in converting chemicals to either less-toxic metabolite(s) or more toxic products (e.g. reactive intermediate[s]) during stages before birth and during early development (neonate/infant/child). In this review, we evaluated the ontogeny of major "phase I" and "phase II" metabolizing enzymes in humans and commonly used experimental animals (e.g. mouse, rat, and others) in order to fill the information gap.
Collapse
Affiliation(s)
- Shakil Ahmed Saghir
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, Michigan, USA.
| | | | | |
Collapse
|
34
|
Abduljalil K, Furness P, Johnson TN, Rostami-Hodjegan A, Soltani H. Anatomical, Physiological and Metabolic Changes with Gestational Age during Normal Pregnancy. Clin Pharmacokinet 2012; 51:365-96. [DOI: 10.2165/11597440-000000000-00000] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
35
|
Dorlo TPC, Balasegaram M, Lima MA, de Vries PJ, Beijnen JH, Huitema ADR. Translational pharmacokinetic modelling and simulation for the assessment of duration of contraceptive use after treatment with miltefosine. J Antimicrob Chemother 2012; 67:1996-2004. [DOI: 10.1093/jac/dks164] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
36
|
Gentry PR, Clewell HJ, Clewell R, Campbell J, Van Landingham C, Shipp AM. Challenges in the application of quantitative approaches in risk assessment: a case study with di-(2-ethylhexyl)phthalate. Crit Rev Toxicol 2011; 41 Suppl 2:1-72. [DOI: 10.3109/10408444.2011.599366] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
37
|
Yoon M, Schroeter JD, Nong A, Taylor MD, Dorman DC, Andersen ME, Clewell HJ. Physiologically based pharmacokinetic modeling of fetal and neonatal manganese exposure in humans: describing manganese homeostasis during development. Toxicol Sci 2011; 122:297-316. [PMID: 21622944 DOI: 10.1093/toxsci/kfr141] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Concerns for potential vulnerability to manganese (Mn) neurotoxicity during fetal and neonatal development have been raised due to increased needs for Mn for normal growth, different sources of exposure to Mn, and pharmacokinetic differences between the young and adults. A physiologically based pharmacokinetic (PBPK) model for Mn during human gestation and lactation was developed to predict Mn in fetal and neonatal brain using a parallelogram approach based upon extrapolation across life stages in rats and cross-species extrapolation to humans. Based on the rodent modeling, key physiological processes controlling Mn kinetics during gestation and lactation were incorporated, including alterations in Mn uptake, excretion, tissue-specific distributions, and placental and lactational transfer of Mn. Parameters for Mn kinetics were estimated based on human Mn data for milk, placenta, and fetal/neonatal tissues, along with allometric scaling from the human adult model. The model was evaluated by comparison with published Mn levels in cord blood, milk, and infant blood. Maternal Mn homeostasis during pregnancy and lactation, placenta and milk Mn, and fetal/neonatal tissue Mn were simulated for normal dietary intake and with inhalation exposure to environmental Mn. Model predictions indicate similar or lower internal exposures to Mn in the brains of fetus/neonate compared with the adult at or above typical environmental air Mn concentrations. This PBPK approach can assess expected Mn tissue concentration during early life and compares contributions of different Mn sources, such as breast or cow milk, formula, food, drinking water, and inhalation, with tissue concentration.
Collapse
Affiliation(s)
- Miyoung Yoon
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Pruthi S, Haakenson C, Brost BC, Bryant K, Reid JM, Singh R, Netzel B, Boughey JC, Degnim AC. Pharmacokinetics of methylene blue dye for lymphatic mapping in breast cancer—implications for use in pregnancy. Am J Surg 2011; 201:70-5. [DOI: 10.1016/j.amjsurg.2009.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 03/31/2009] [Accepted: 03/31/2009] [Indexed: 10/18/2022]
|
39
|
Spurgeon DJ, Jones OAH, Dorne JLCM, Svendsen C, Swain S, Stürzenbaum SR. Systems toxicology approaches for understanding the joint effects of environmental chemical mixtures. THE SCIENCE OF THE TOTAL ENVIRONMENT 2010; 408:3725-3734. [PMID: 20231031 DOI: 10.1016/j.scitotenv.2010.02.038] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 02/15/2010] [Accepted: 02/17/2010] [Indexed: 05/28/2023]
Abstract
Environmental mixtures of chemicals constitute a prevalent issue in ecotoxicology and the development of new methods to reduce the uncertainties associated with their ecological risk assessment is a critical research need. Historically, a number of models have been explored to predict the potential combined effects of chemicals on species. These models, especially concentration addition and the independent action, have been applied to a number of mixtures. While often providing a good prediction of joint effect, there are cases where these models can have limitations: notably in cases where there are interactions for which they fail to adequately predict joint effects. To support the better mechanistic understanding of interactions in mixture toxicology a framework to support experimental studies to investigate the basis of observed interactions is proposed. The conceptual framework is derived from the extension of a three stage scheme which has previously been applied to understand chemical bioavailability. The framework considers that interactions in mixtures result from processes related to 1) the speciation, binding and transport of chemicals in the exposure medium (external exposure); 2) the adsorption, distribution, metabolism and excretion of chemicals within the organisms (toxicokinetics); 3) associations governing the binding and toxicity of the chemical(s) at the target site (toxicodynamics). The current state of the art in (eco)toxicology in relation to investigation of the mechanisms of interactions between chemicals is discussed with particular emphasis towards the multi-disciplinary tools and techniques within environmental chemistry; toxicology; biochemistry and systems biology that can be used to address such effects.
Collapse
Affiliation(s)
- David J Spurgeon
- Centre for Ecology and Hydrology, Maclean Building, Benson Lane, Crowmarsh Gifford, Wallingford, Oxon, OX10 8BB, UK.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Summarize recent studies exploring the relationship between paternal and maternal environmental exposures to chemicals before, at the time of and after conception to adverse developmental outcomes including preterm birth, death, structural and functional abnormalities and growth restriction. RECENT FINDINGS Recent studies have demonstrated that human pregnancy and development are vulnerable to environmental exposures of the father and mother to chemical, biological and physical agents. Exposures associated with adverse developmental outcomes include air and water pollution, chemicals in foods, occupational exposures, agricultural chemicals, metals, persistent and volatile organics. Developmental endpoints which are linked with these exposures include growth restriction, functional abnormalities, structural abnormalities, preterm delivery and death. Despite this general understanding we still have incomplete knowledge concerning most exposures and the biological interactions responsible for impaired development and preterm delivery. SUMMARY Whereas single genes and individual chemical exposures are responsible for some instances of adverse pregnancy outcome or developmental disease, gene-environment interactions are responsible for the majority. These gene-environment interactions may occur in the father, mother, placenta or fetus, suggesting that critical attention be given to maternal and paternal exposures and gene expression as they relate to the mode of action of the putative developmental toxicant both prior to and during pregnancy.
Collapse
|
41
|
Poet TS, Kirman CR, Bader M, van Thriel C, Gargas ML, Hinderliter PM. Quantitative risk analysis for N-methyl pyrrolidone using physiologically based pharmacokinetic and benchmark dose modeling. Toxicol Sci 2009; 113:468-82. [PMID: 19875680 DOI: 10.1093/toxsci/kfp264] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Establishing an occupational exposure limit (OEL) for N-methyl pyrrolidone (NMP) is important due to its widespread use as a solvent. Based on studies in rodents, the most sensitive toxic end point is a decrease in fetal/pup body weights observed after oral, dermal, and inhalation exposures of dams to NMP. Evidence indicates that the parent compound is the causative agent. To reduce the uncertainty in rat to human extrapolations, physiologically based pharmacokinetic (PBPK) models were developed to describe the pharmacokinetics of NMP in both species. Since in utero exposures are of concern, the models considered major physiological changes occurring in the dam or mother over the course of gestation. The rat PBPK model was used to determine the relationship between NMP concentrations in maternal blood and decrements in fetal/pup body weights following exposures to NMP vapor. Body weight decrements seen after vapor exposures occurred at lower NMP blood levels than those observed after oral and dermal exposures. Benchmark dose modeling was used to better define a point of departure (POD) for fetal/pup body weight changes based on dose-response information from two inhalation studies in rats. The POD and human PBPK model were then used to estimate the human equivalent concentrations (HECs) that could be used to derive an OEL value for NMP. The geometric mean of the PODs derived from the rat studies was estimated to be 350 mg h/l (expressed in terms of internal dose), a value which corresponds to an HEC of 480 ppm (occupational exposure of 8 h/day, 5 days/week). The HEC is much higher than recently developed internationally recognized OELs for NMP of 10-20 ppm, suggesting that these OELs adequately protect workers exposed to NMP vapor.
Collapse
Affiliation(s)
- Torka S Poet
- Battelle Pacific Northwest Division, Center for Biological Monitoring and Modeling, Richland, Washington 99352, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Andrew MA, Hebert MF, Vicini P. Physiologically based pharmacokinetic model of midazolam disposition during pregnancy. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2009; 2008:5454-7. [PMID: 19163951 DOI: 10.1109/iembs.2008.4650448] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Disposition of drugs in pregnant women is poorly understood in spite of widespread prescription of drugs to women during gestation. We have developed a whole body physiologically based pharmacokinetic (PBPK) model to explore the effects of pregnancy on pharmacokinetics. The model accounts for maternofetal changes over the course of gestation; physiological and drug-specific parameters are taken from literature. Here we preliminarily demonstrate the model's utility to predict midazolam pharmacokinetics following intravenous bolus dosing in women undergoing Caesarian section. Simulations of maternal venous plasma concentrations compare favorably with data extracted from historical studies.
Collapse
Affiliation(s)
- Marilee A Andrew
- Applied Physics Laboratory and the Department of Bioengineering at the University of Washington, Seattle, WA 98105-6698, USA.
| | | | | |
Collapse
|
43
|
Lowe ER, Poet TS, Rick DL, Marty MS, Mattsson JL, Timchalk C, Bartels MJ. The effect of plasma lipids on the pharmacokinetics of chlorpyrifos and the impact on interpretation of blood biomonitoring data. Toxicol Sci 2009; 108:258-72. [PMID: 19223661 DOI: 10.1093/toxsci/kfp034] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Lipophilic molecules, like chlorpyrifos (CPF), present a special problem for interpretation of biomonitoring data because both the environmental dose of CPF and the physiological (pregnancy, diet, etc.) or pathological levels of blood lipids will affect the concentrations of CPF measured in blood. The objective of this study was to investigate the distribution of CPF between plasma and tissues when lipid levels are altered in late pregnancy. CPF was sequestered more in the low-density lipid fraction of the blood during the late stages of gestation in the rat and returned to nonpregnant patterns in the dam after birth. Plasma partitioning of CPF increased with increases in plasma lipid levels and the increased partitioning of CPF into plasma lipids resulted in less CPF in other tissue compartments. Gavage dosing with corn oil also increased plasma lipids that led to a moderate increase of CPF partitioning into the plasma. To mechanistically investigate the potential pharmacokinetic effects of blood lipid changes, an existing CPF physiologically based pharmacokinetic/pharmacodynamic model for rats and humans was modified to account for altered lipid-tissue partition coefficients and for major physiological and biochemical changes of pregnancy. The model indicated that plasma CPF levels are expected to be proportional to the well-known changes in plasma lipids during gestation. There is a rapidly growing literature on the relationship of lipid profiles with different disease conditions and on birth outcomes. Increased blood concentrations of lipophilic chemicals like CPF may point to altered lipid status, as well as possibly higher levels of exposure. Thus, proper interpretation of blood biomonitoring data of lipophilic chemicals requires a careful consideration of blood lipids.
Collapse
Affiliation(s)
- Ezra R Lowe
- Toxicology & Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan 48674, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Makris SL, Thompson CM, Euling SY, Selevan SG, Sonawane B. A lifestage-specific approach to hazard and dose-response characterization for children's health risk assessment. ACTA ACUST UNITED AC 2009; 83:530-46. [PMID: 19085945 DOI: 10.1002/bdrb.20176] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In 2006, the U.S. EPA published a report entitled A Framework for Assessing Health Risks of Environmental Exposures to Children (hereafter referred to as the "Framework") describing a lifestage approach to risk assessment that includes the evaluation of existing data from a temporal perspective (i.e., the timing of both the exposure and the outcome). This article summarizes the lifestage-specific issues discussed in the Framework related to the qualitative and the quantitative hazard and dose-response characterization. Lifestage-specific hazard characterization includes an evaluation of relevant human and experimental animal studies, focusing on the identification of critical windows of development (i.e., exposure intervals of maximum susceptibility) for observed outcomes, evaluation of differential exposure at individual lifestages, the relevance and impact of lifestage-specific toxicokinetic and toxicodynamic data, mode of action information, variability and latency of effects from early lifestage exposure, and describing uncertainties. The interpretation of the hazard data to determine the strength of association between early life exposures and the timing and type of outcomes depends upon the overall weight of evidence. Lifestage-specific dose-response characterization relies on the identification of susceptible lifestages in order to quantify health risk, information on the point of departure, key default assumptions, and descriptions of uncertainty, sensitivity, and variability. Discussion of the strength and limitations of the hazard and dose-response data provides a basis for confidence in risk determinations. Applying a lifestage approach to hazard and dose-response characterization is likely to improve children's health risk assessment by identifying data gaps and providing a better understanding of sources of uncertainty.
Collapse
Affiliation(s)
- Susan L Makris
- National Center for Environmental Assessment (NCEA), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), 1200 Pennsylvania Avenue, NW, Washington, DC 20460, USA.
| | | | | | | | | |
Collapse
|
45
|
Lee SK, Hamer D, Bedwell CL, Lohitnavy M, Yang RSH. Effect of PCBs on the lactational transfer of methyl mercury in mice: PBPK modeling. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2009; 27:75-83. [PMID: 20046988 PMCID: PMC2707941 DOI: 10.1016/j.etap.2008.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
MeHg and PCB exposure to lactating mice were analyzed and a physiologically-based pharmacokinetic (PBPK) model was developed to describe the lactational transfer of MeHg in mice. The influence of albumin on the lactational transfer of MeHg was incorporated into the PBPK model. Experimental results with lactating mice and their pups showed that co-exposure with PCB congeners increased the lactational transfer of MeHg to the pups, which was associated with the rise of albumin levels in maternal blood. Observed results were matched with PBPK model simulations conducted under the assumptions that (1) MeHg bound to plasma albumin is transferred to maternal milk, and (2) PCB congeners may increase the lactational transfer of MeHg by escalating albumin levels in maternal blood. Further refinement of PBPK model quantitatively described the pharmacokinetic changes of MeHg by co-exposure with PCBs in pup's tissues.
Collapse
Affiliation(s)
- Sun Ku Lee
- Quantitative and Computational Toxicology Group, Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collin, CO 80523
| | - Dwayne Hamer
- Veterinary Diagnostic Laboratory, Colorado State University, Fort Collins, CO 80523
| | - Cathy L. Bedwell
- Veterinary Diagnostic Laboratory, Colorado State University, Fort Collins, CO 80523
| | | | - Raymond S. H. Yang
- Quantitative and Computational Toxicology Group, Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collin, CO 80523
| |
Collapse
|
46
|
García-Algar O, Vall Combelles O, Puig Sola C, Mur Sierra A, Scaravelli G, Pacifici R, Monleón Getino T, Pichini S. [Prenatal exposure to drugs of abuse using meconium analysis in a low socioeconomic population in Barcelona]. An Pediatr (Barc) 2008; 70:151-8. [PMID: 19217571 DOI: 10.1016/j.anpedi.2008.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 08/12/2008] [Accepted: 08/25/2008] [Indexed: 10/20/2022] Open
Abstract
INTRODUCTION For the first time in Europe, the <<Meconium Project>> aimed to estimate the prevalence of drug use by pregnant women and the subsequent foetal exposure to illicit drugs. PATIENTS AND METHOD Between October 2002 and February 2004, 1209 mother-infant dyads from the Hospital del Mar, Barcelona, Spain met eligibility criteria and agreed to participate in the study. Data on socio-economic and demographic characteristics and on drug habits during pregnancy were collected using a structured questionnaire. Neonatal meconium was collected within 24h after birth and analyzed by standardized chromatographic techniques for the presence of opiates, cocaine, cannabinoids and amphetamines. RESULTS Meconium analysis showed an overall 10.9% positivity for drugs of abuse, with a specific prevalence of heroin, cocaine and cannabis with foetal exposure of 4.7, 2.6 and 5.3%, respectively. Structured interviews also revealed that 0.3, 1.2 and 1.5% of mothers used heroin, cocaine and cannabis, respectively, while only one mother declared ecstasy consumption, confirmed by meconium analysis. Parental ethnicity and working class was not associated with drug use. Drug consuming mothers were shown to have a higher number of previous abortions when compared to non-consumer mothers, which was probably due to a lack of family planning. Significantly lower birth weight and length was found in newborns from mothers exposed to cocaine alone or in combination with other drugs. CONCLUSIONS This study, although developed in a low socio-economic-status cohort, may serve as an eye opener for any hidden non-negligible drug consumption during pregnancy. In this sense, meconium analysis can be important to identify neonates with a high suspicion of exposure to drugs of abuse in utero, and provides the basis for appropriate treatment and adequate medical and social follow-up.
Collapse
Affiliation(s)
- O García-Algar
- Servicio de Pediatría, Hospital del Mar, Barcelona, España.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Leavens TL, Parkinson CU, James RA, House D, Elswick B, Dorman DC. Respiration in Sprague-Dawley Rats During Pregnancy. Inhal Toxicol 2008; 18:305-12. [DOI: 10.1080/08958370500444361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
48
|
Thompson CM, Sonawane B, Barton HA, DeWoskin RS, Lipscomb JC, Schlosser P, Chiu WA, Krishnan K. Approaches for applications of physiologically based pharmacokinetic models in risk assessment. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2008; 11:519-47. [PMID: 18584453 DOI: 10.1080/10937400701724337] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) models are particularly useful for simulating exposures to environmental toxicants for which, unlike pharmaceuticals, there is often little or no human data available to estimate the internal dose of a putative toxic moiety in a target tissue or an appropriate surrogate. This article reviews the current state of knowledge and approaches for application of PBPK models in the process of deriving reference dose, reference concentration, and cancer risk estimates. Examples drawn from previous U.S. Environmental Protection Agency (EPA) risk assessments and human health risk assessments in peer-reviewed literature illustrate the ways and means of using PBPK models to quantify the pharmacokinetic component of the interspecies and intraspecies uncertainty factors as well as to conduct route to route, high dose to low dose and duration extrapolations. The choice of the appropriate dose metric is key to the use of the PBPK models for the various applications in risk assessment. Issues related to whether uncertainty factors are most appropriately applied before or after derivation of human equivalent dose (or concentration) continue to be explored. Scientific progress in the understanding of life stage and genetic differences in dosimetry and their impacts on variability in susceptibility, as well as ongoing development of analytical methods to characterize uncertainty in PBPK models, will make their use in risk assessment increasingly likely. As such, it is anticipated that when PBPK models are used to express adverse tissue responses in terms of the internal target tissue dose of the toxic moiety rather than the external concentration, the scientific basis of, and confidence in, risk assessments will be enhanced.
Collapse
Affiliation(s)
- Chad M Thompson
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington, DC, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
McMullin TS, Lowe ER, Bartels MJ, Marty MS. Dynamic changes in lipids and proteins of maternal, fetal, and pup blood and milk during perinatal development in CD and Wistar rats. Toxicol Sci 2008; 105:260-74. [PMID: 18593729 DOI: 10.1093/toxsci/kfn124] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
An understanding of the physiological factors that regulate perinatal dosimetry is essential to improve the ability of physiologically based (PB) pharmacokinetic (PK) models to predict chemical risks to children. However, the impact of changing maternal/offspring physiology on PK during gestation and lactation remains poorly understood. This research determined lipid and protein changes in blood, milk and amniotic fluid of CD and Wistar dams, fetuses and neonates to improve the precision of perinatal PBPK modeling. Samples were collected from time-mated CD dams, fetuses, and pups on gestation day (GD) 18 and 20 (sperm positive = GD 0) or lactation day 0 (day of birth), 1, 3, 5, 10, 15, and 20 (n > or = 5 per time point). Fewer time points were sampled in Wistar rats, which showed similar patterns to CDs. Relative to nonpregnant dams, maternal serum protein levels (albumin, total protein and globulin) each decreased by approximately 20% during late gestation, whereas maternal serum lipids (triglycerides, low density lipoproteins, and phospholipids) increased up to fourfold. These physiological changes can impact maternal PK of both protein-bound and lipophilic chemicals. During lactation, triglycerides in milk were greater than 100-fold higher than maternal serum, favoring the disposition of lipophilic chemicals into milk and potentially increasing neonatal rodent exposure during critical stages of postnatal development. Serum protein levels in pups were two- to threefold lower than adults at birth, which may increase the bioavailability of protein-bound compounds. These data will aid in the interpretation of perinatal toxicity studies and improve the accuracy of predictive perinatal PBPK models.
Collapse
|
50
|
Lipscomb JC, Poet TS. In vitro measurements of metabolism for application in pharmacokinetic modeling. Pharmacol Ther 2008; 118:82-103. [DOI: 10.1016/j.pharmthera.2008.01.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Accepted: 01/24/2008] [Indexed: 11/25/2022]
|