1
|
Schoenmakers S, Li L, Kluivers ACM, Broekhuizen M, Harhangi MS, Danser AHJ, Reiss I, Allegaert K, van den Berg SAA, van Zelst BD, van Schaik RHN, DeKoninck PLJ, Ronde E, Sassen SDT, Simons SHP, Koch BCP. Pharmacokinetics of betamethasone in pre-eclampsia: An in vivo and ex vivo study. Br J Clin Pharmacol 2025. [PMID: 40083164 DOI: 10.1002/bcp.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
AIMS To enhance understanding of betamethasone and its metabolites' pharmacokinetics in pregnancy, specifically early-onset pre-eclampsia, through a population pharmacokinetic model. Additionally, to investigate the placental metabolism and transfer of betamethasone and its main metabolites. METHODS A prospective, single-centre pharmacokinetic study was conducted in pregnant women (n = 28) with imminent preterm birth treated with intramuscular betamethasone. Betamethasone serum concentrations were determined from serial venous blood samples (n = 194). Placental transfer and metabolism were studied using ex vivo human placental perfusion (healthy term; n = 3) and placental explant experiments (healthy term, n = 4; early-onset pre-eclampsia, n = 4). Additionally, placental mRNA expression of CYP3A4, CYP3A7, 11β-hydroxysteroid dehydrogenase (HSD) 1 and 11β-HSD2 were quantified in healthy and early-onset pre-eclampsia placentas. RESULTS The population pharmacokinetic model was best described by a 2-compartment nonlinear mixed effects model. Betamethasone clearance in early-onset pre-eclamptic women was 60% lower of that observed in women without pre-eclampsia (9.35 vs. 15.78 L/h), resulting in a 40% median increase in maternal betamethasone exposure (1567 vs. 1114 ng h/mL). Ex vivo experiments showed placental transfer of betamethasone to the foetal circulation (foetal-to-maternal ratio 0.76 ± 0.05 [in a perfused placental cotyledon]). The placenta only converted betamethasone into 11-ketobetamethasone, with similar ratios in early-onset pre-eclampsia and healthy placental explants (3.0 ± 2.2 vs. 1.4 ± 0.4 per mg tissue, P = .27). The expression of 11β-HSD1 mRNA was lower in early-onset pre-eclampsia placentas (P = .015), while placental CYP3A7 and 11β-HSD2 mRNA expression were similar. CONCLUSION Women with early-onset pre-eclampsia have elevated betamethasone exposure. Betamethasone transfers freely into the foetal circulation, with placental metabolism resulting only in 11-ketobetamethasone. Decreased placental 11β-HSD1 expression may play a role in increased betamethasone exposure in early-onset pre-eclampsia.
Collapse
Affiliation(s)
- Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Division of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Letao Li
- Department of Hospital Pharmacy, Erasmus University Medical Center, The Netherlands
| | - Anna C M Kluivers
- Department of Obstetrics and Gynaecology, Division of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC University Hospital, Rotterdam, The Netherlands
| | - Michelle Broekhuizen
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC University Hospital, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Madhavi S Harhangi
- Department of Obstetrics and Gynaecology, Division of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC University Hospital, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC University Hospital, Rotterdam, The Netherlands
| | - Irwin Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus University Medical Center, The Netherlands
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, Leuven, Belgium
| | - Sjoerd A A van den Berg
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center, the Netherlands
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bertrand D van Zelst
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Philip L J DeKoninck
- Department of Obstetrics and Gynaecology, Division of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Emma Ronde
- Department of Obstetrics and Gynaecology, Division of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sebastiaan D T Sassen
- Department of Hospital Pharmacy, Erasmus University Medical Center, The Netherlands
- Center for Antimicrobial Treatment Optimization Rotterdam (CATOR), the Netherlands
- Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
| | - Sinno H P Simons
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center, The Netherlands
- Center for Antimicrobial Treatment Optimization Rotterdam (CATOR), the Netherlands
- Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
| |
Collapse
|
2
|
Terrier J, Abouir K, Gaspar F, Daali Y, Samer CF. The Impact of Dexamethasone and Prednisone on Apixaban and Rivaroxaban Exposure in COVID-19 Patients: A Physiologically Based Pharmacokinetic Modeling Study. Clin Pharmacol Ther 2025; 117:554-560. [PMID: 39535447 PMCID: PMC11739736 DOI: 10.1002/cpt.3491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Dexamethasone (DEX) is currently the treatment of choice for patients with oxygen-dependent COVID-19. It has been observed, primarily in vitro, that dexamethasone induces the expression of CYP3A and the ABCB1 gene, which encodes P-glycoprotein (P-gp). This has raised concerns about potential interactions between DEX and substrates of CYP3A and P-gp, such as direct oral anticoagulants (DOAC). Currently, there is limited robust evidence to support a clinically significant interaction between DEX and DOAC. Using physiologically based pharmacokinetic modeling (PBPK), we investigated the impact of DEX administered in the context of SARS-CoV-2 infection on the pharmacokinetics of apixaban (APX) and rivaroxaban (RVX). After validating the induction effect of the DEX compound on two CYP3A4 substrates using the limited available studies, we optimized the compound in a COVID-19 patient population, where significantly higher DEX plasma concentrations were observed compared to healthy volunteers. Our PBPK-based PK simulations showed a 20% decrease in the AUC of APX and RVX in a worst-case scenario and when DEX was administered at 6 mg PO for 10 days. This finding confirms the limited clinical data currently available and supports the use of APX and RVX with DEX in COVID-19 patients at low-risk for thrombo-embolism. In addition, our results suggest that prednisone (PRED), when used at an equipotent dose, could serve as a viable alternative to DEX, given its less pronounced induction effect on APX and RVX. Further research is needed to validate these findings and to explore the clinical implications of using PRED in place of DEX in such scenarios.
Collapse
Affiliation(s)
- Jean Terrier
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Kenza Abouir
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Frederic Gaspar
- Center for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of GenevaGenevaSwitzerland
| | - Caroline Flora Samer
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
3
|
Van Der Heijden JEM, Van Hove H, Van Elst NM, Van Den Broek P, Van Drongelen J, Scheepers HCJ, De Wildt SN, Greupink R. Optimization of the betamethasone and dexamethasone dosing regimen during pregnancy: a combined placenta perfusion and pregnancy physiologically based pharmacokinetic modeling approach. Am J Obstet Gynecol 2025; 232:228.e1-228.e9. [PMID: 38763343 DOI: 10.1016/j.ajog.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Antenatal betamethasone and dexamethasone are prescribed to women who are at high risk of premature birth to prevent neonatal respiratory distress syndrome (RDS). The current treatment regimens, effective to prevent neonatal RDS, may be suboptimal. Recently, concerns have been raised regarding possible adverse long-term neurological outcomes due to high fetal drug exposures. Data from nonhuman primates and sheep suggest maintaining a fetal plasma concentration above 1 ng/mL for 48 hours to retain efficacy, while avoiding undesirable high fetal plasma levels. OBJECTIVE We aimed to re-evaluate the current betamethasone and dexamethasone dosing strategies to assess estimated fetal exposure and provide new dosing proposals that meet the efficacy target but avoid excessive peak exposures. STUDY DESIGN A pregnancy physiologically based pharmacokinetic (PBPK) model was used to predict fetal drug exposures. To allow prediction of the extent of betamethasone and dexamethasone exposure in the fetus, placenta perfusion experiments were conducted to determine placental transfer. Placental transfer rates were integrated in the PBPK model to predict fetal exposure and model performance was verified using published maternal and fetal pharmacokinetic data. The verified pregnancy PBPK models were then used to simulate alternative dosing regimens to establish a model-informed dose. RESULTS Ex vivo data showed that both drugs extensively cross the placenta. For betamethasone 15.7±1.7% and for dexamethasone 14.4±1.5%, the initial maternal perfusate concentration reached the fetal circulations at the end of the 3-hour perfusion period. Pregnancy PBPK models that include these ex vivo-derived placental transfer rates accurately predicted maternal and fetal exposures resulting from current dosing regimens. The dose simulations suggest that for betamethasone intramuscular, a dose reduction from 2 dosages 11.4 mg, 24 hours apart, to 4 dosages 1.425 mg, 12 hours apart would avoid excessive peak exposures and still meet the fetal response threshold. For dexamethasone, the dose may be reduced from 4 times 6 mg every 12 hours to 8 times 1.5 mg every 6 hours. CONCLUSION A combined placenta perfusion and pregnancy PBPK modeling approach adequately predicted both maternal and fetal drug exposures of 2 antenatal corticosteroids (ACSs). Strikingly, our PBPK simulations suggest that drug doses might be reduced drastically to still meet earlier proposed efficacy targets and minimize peak exposures. We propose the provided model-informed dosing regimens are used to support further discussion on an updated ACS scheme and design of clinical trials to confirm the effectiveness and safety of lower doses.
Collapse
Affiliation(s)
- Joyce E M Van Der Heijden
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Hedwig Van Hove
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niki M Van Elst
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Petra Van Den Broek
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joris Van Drongelen
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hubertina C J Scheepers
- Department of Obstetrics and Gynecology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Saskia N De Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Rick Greupink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Khadka KB, Koirala N, Ivanova O, Bastola R, Singh D, Magar KR, Banstola B, Adhikari RP, Giedraitis V, Paudel D, Froeschl G. Newborn morbidities and care procedures at the special newborn care units of Gandaki Province, Nepal: a retrospective study. BMC Pregnancy Childbirth 2024; 24:883. [PMID: 39736626 DOI: 10.1186/s12884-024-07120-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Despite recent improvements in the overall health status of Nepal's population, newborn morbidities and mortalities have remained a challenge. This study explores the situation and care strategies for newborn health problems in the Gandaki Province of Nepal. METHODS This is a retrospective hospital records analysis. A structured questionnaire was employed to collect data on socio-demographic, clinical, and outcome variables in 1,355 newborns admitted to the Special Newborn Care Unit (SNCU) between May 1, 2021, and April 30, 2022, in five hospitals within the Gandaki Province. RESULTS Among all newborns, 60% were male, and 40% belonged to Janajati indigenous families. The mean age of mothers at the time of delivery was 24.4 years; the average birth weight of babies was 2.8 kg; and the gestational week was 38 weeks. Around 96% of births occurred in healthcare facilities. The average inpatient hospital stay was 4.7 days. The reasons for SNCU admission were newborn sepsis (51%), neonatal hyperbilirubinemia (23%), respiratory distress syndrome (18%), and low birth weight (11%). Approximately 7% of the newborns were found to have died due to various causes, including sepsis, asphyxia, and indirect medical reasons. Female newborns had a 0.45-times (CI: 0.23-0.84) lower risk of mortality compared to male newborns. Underweight newborns had 8.8 times (CI: 4.5-17.2) higher risk of death than newborns with a normal birth weight, even after adjusting for other factors like sex, delivery site, mode of delivery, mother's age, respiratory distress syndrome, neonatal hyperbilirubinemia, neonatal sepsis, and age at admission to SNCU. The most common treatments included injectable antibiotics (73%), intravenous fluids (53%), oxygen delivery (39%), and phototherapy (36%), while 3% received "Kangaroo Mother Care (KMC)". CONCLUSIONS The study showed that newborns suffered from multiple health complications such as sepsis, hyperbilirubinemia, or asphyxia, and many newborns received essential medical services from hospitals. Birth weight, sex of the newborn, and respiratory distress syndrome were significantly associated with neonatal mortality. Hospitals should focus on reinforcing KMC, neonatal resuscitation, and early infection control measures.
Collapse
MESH Headings
- Humans
- Nepal/epidemiology
- Infant, Newborn
- Retrospective Studies
- Female
- Male
- Adult
- Young Adult
- Infant Mortality
- Intensive Care Units, Neonatal/statistics & numerical data
- Infant, Newborn, Diseases/epidemiology
- Infant, Newborn, Diseases/therapy
- Respiratory Distress Syndrome, Newborn/therapy
- Respiratory Distress Syndrome, Newborn/epidemiology
- Infant, Low Birth Weight
- Neonatal Sepsis/epidemiology
- Hyperbilirubinemia, Neonatal/therapy
- Hyperbilirubinemia, Neonatal/epidemiology
- Pregnancy
- Infant
- Length of Stay/statistics & numerical data
- Morbidity
Collapse
Affiliation(s)
- Khim Bahadur Khadka
- Health Directorate, Ministry of Social Development and Health, Pokhara, Gandaki Province, Nepal.
- Center for International Health, LMU, Munich, Germany.
| | - Nabina Koirala
- Health Directorate, Ministry of Social Development and Health, Pokhara, Gandaki Province, Nepal
| | - Olena Ivanova
- Center for International Health, LMU, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU, Munich, Germany
| | - Ramchandra Bastola
- Pokhara Academy of Health Sciences, Western Regional Hospital, Pokhara, Gandaki Province, Nepal
| | - Dela Singh
- Pokhara Academy of Health Sciences, Western Regional Hospital, Pokhara, Gandaki Province, Nepal
| | - Kamala Rana Magar
- Pokhara Academy of Health Sciences, Western Regional Hospital, Pokhara, Gandaki Province, Nepal
| | - Bidhya Banstola
- Pokhara Academy of Health Sciences, Western Regional Hospital, Pokhara, Gandaki Province, Nepal
| | - Ramesh Prasad Adhikari
- Health Training Center, Ministry of Social Development and Health, Pokhara, Gandaki Province, Nepal
| | | | | | - Guenter Froeschl
- Center for International Health, LMU, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU, Munich, Germany
| |
Collapse
|
5
|
Zhang J, Li SP, Li QQ, Zhang YT, Dong GH, Canchola A, Zeng X, Chou WC. Development of a Physiologically Based Pharmacokinetic (PBPK) Model for F-53B in Pregnant Mice and Its Extrapolation to Humans. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18928-18939. [PMID: 39394996 PMCID: PMC11500426 DOI: 10.1021/acs.est.4c05405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024]
Abstract
Chlorinated polyfluorinated ether sulfonic acid (F-53B), a commonly utilized alternative for perfluorooctane sulfonate, was detected in pregnant women and cord blood recently. However, the lack of detailed toxicokinetic information poses a significant challenge in assessing the human risk assessment for F-53B exposure. Our study aimed to develop a physiologically based pharmacokinetic (PBPK) model for pregnant mice, based on toxicokinetic experiments, and extrapolating it to humans. Pregnant mice were administered 80 μg/kg F-53B orally and intravenously on gestational day 13. F-53B concentrations in biological samples were analyzed via ultraperformance liquid chromatography-mass spectrometry. Results showed the highest F-53B accumulation in the brain, followed by the placenta, amniotic fluid, and liver in fetal mice. These toxicokinetic data were applied to F-53B PBPK model development and evaluation, and Monte Carlo simulations were used to characterize the variability and uncertainty in the human population. Most of the predictive values were within a 2-fold range of experimental data (>72%) and had a coefficient of determination (R2) greater than 0.68. The developed mouse model was then extrapolated to the human and evaluated with human biomonitoring data. Our study provides an important step toward improving the understanding of toxicokinetics of F-53B and enhancing the quantitative risk assessments in sensitive populations, particularly in pregnant women and fetuses.
Collapse
Affiliation(s)
- Jing Zhang
- Joint
International Research Laboratory of Environment and Health, Ministry
of Education, Guangdong Provincial Engineering Technology Research
Center of Environmental Pollution and Health Risk Assessment, Department
of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen-Pan Li
- Joint
International Research Laboratory of Environment and Health, Ministry
of Education, Guangdong Provincial Engineering Technology Research
Center of Environmental Pollution and Health Risk Assessment, Department
of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qing-Qing Li
- Acacia
Lab for Implementation Science, Institute for Global Health, Dermatology Hospital of Southern Medical University, Guangzhou 510515, China
| | - Yun-Ting Zhang
- Joint
International Research Laboratory of Environment and Health, Ministry
of Education, Guangdong Provincial Engineering Technology Research
Center of Environmental Pollution and Health Risk Assessment, Department
of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Guang-Hui Dong
- Joint
International Research Laboratory of Environment and Health, Ministry
of Education, Guangdong Provincial Engineering Technology Research
Center of Environmental Pollution and Health Risk Assessment, Department
of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Alexa Canchola
- Department
of Environmental Sciences, University of
California, Riverside, California 92521, United States
- Environmental
Toxicology Graduate Program, University
of California, Riverside, California 92521, United States
| | - Xiaowen Zeng
- Joint
International Research Laboratory of Environment and Health, Ministry
of Education, Guangdong Provincial Engineering Technology Research
Center of Environmental Pollution and Health Risk Assessment, Department
of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei-Chun Chou
- Department
of Environmental Sciences, University of
California, Riverside, California 92521, United States
- Environmental
Toxicology Graduate Program, University
of California, Riverside, California 92521, United States
| |
Collapse
|
6
|
Gosavi A, Amin Z, Carter SWD, Choolani MA, Fee EL, Milad MA, Jobe AH, Kemp MW. Antenatal corticosteroids in Singapore: a clinical and scientific assessment. Singapore Med J 2024; 65:479-487. [PMID: 36254928 PMCID: PMC11479002 DOI: 10.4103/singaporemedj.smj-2022-014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
ABSTRACT Preterm birth (PTB; delivery prior to 37 weeks' gestation) is the leading cause of early childhood death in Singapore today. Approximately 9% of Singaporean babies are born preterm; the PTB rate is likely to increase given the increased use of assisted reproduction technologies, changes in the incidence of gestational diabetes/high body mass index and the ageing maternal population. Antenatal administration of dexamethasone phosphate is a key component of the obstetric management of Singaporean women who are at risk of imminent preterm labour. Dexamethasone improves preterm outcomes by crossing the placenta to functionally mature the fetal lung. The dexamethasone regimen used in Singapore today affords a very high maternofetal drug exposure over a brief period of time. Drawing on clinical and experimental data, we reviewed the pharmacokinetic profile and pharmacodynamic effects of dexamethasone treatment regimen in Singapore, with a view to creating a development pipeline for optimising this critically important antenatal therapy.
Collapse
Affiliation(s)
- Arundhati Gosavi
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zubair Amin
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sean William David Carter
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Mahesh Arjandas Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Erin Lesley Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Mark Amir Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, USA
| | - Alan Hall Jobe
- Perinatal Research, Department of Pediatrics, Cincinnati Children’s Hospital Medical Centre, University of Cincinnati, Cincinnati, Ohio, USA
| | - Matthew Warren Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
- School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
7
|
Ait-Chikh C, Page G, Thoreau V. Physiologically-based pharmacokinetic models to predict drug exposure during pregnancy. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:236-242. [PMID: 37739215 DOI: 10.1016/j.pharma.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/24/2023]
Abstract
As pregnant women are constantly exposed to drugs during pregnancy, either to treat long-term conditions or acute illnesses, drug safety is a major concern for the fetus and the mother. Clinical trials are rarely made in this population due to strict regulation and ethical reasons. However, drug pharmacokinetic (PK) parameters vary during pregnancy with an increase in distribution volume, renal clearance and more. In addition, the fetal distribution should be evaluated with the importance of placental diffusion, both active and passive. Therefore, there is a recent interest in the use of physiologically-based pharmacokinetic (PBPK) modeling to characterize these changes and complete the sparse data available on drug PK during pregnancy. Indeed, PBPK models integrate drug physicochemical and physiological parameters corresponding to each compartment of the body to estimate drug concentrations. This review establishes an overview on the current use of PBPK models in drug dosage determination for the pregnant woman, fetal exposure and drug interactions in the fetal compartment.
Collapse
Affiliation(s)
- Celia Ait-Chikh
- Faculté de médecine et pharmacie, université de Poitiers, UFR médecine et pharmacie, bâtiment D1, 6, rue de la Milétrie, TSA 51115, 86073 Poitiers cedex 9, France.
| | - Guylène Page
- Faculté de médecine et pharmacie, université de Poitiers, UFR médecine et pharmacie, bâtiment D1, 6, rue de la Milétrie, TSA 51115, 86073 Poitiers cedex 9, France; Neurovascular Unit and Cognitive Disorders (NEUVACOD), pôle Biologie santé, université de Poitiers, Poitiers, France
| | - Vincent Thoreau
- Faculté de médecine et pharmacie, université de Poitiers, UFR médecine et pharmacie, bâtiment D1, 6, rue de la Milétrie, TSA 51115, 86073 Poitiers cedex 9, France; Neurovascular Unit and Cognitive Disorders (NEUVACOD), pôle Biologie santé, université de Poitiers, Poitiers, France
| |
Collapse
|
8
|
Du R, Zhao X, Song L, Wang H, Liu D, Wang Q. A physiologically based toxicokinetic model of P-glycoprotein transporter-mediated placenta perfusion of dexamethasone in the pregnant rat. Food Chem Toxicol 2024; 183:114213. [PMID: 38052401 DOI: 10.1016/j.fct.2023.114213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/09/2023] [Accepted: 11/19/2023] [Indexed: 12/07/2023]
Abstract
The present dosage of Dexamethasone (DEX) administered to pregnant women may pose a risk of toxicity to their unborn offspring. We aimed to develop a maternal-fetal physiologically based toxicokinetic (PBTK) model for DEX in pregnant rats, with a specific focus on the role of the P-glycoprotein (P-gp) transporter in placenta perfusion, and finally facilitate the optimization of clinical DEX dosage. We conducted animal experiments to determine DEX concentrations in various rat tissues, and constructed the PBTK model using MATLAB software. Sensitivity analysis was performed to assess input parameters and the model stability, with fold error (FE) values serving as evaluation indices. Our results indicate the successful construction of the PBTK model, with the fitting key parameters such as the absorption rate constant (Ka), intrinsic hepatic clearance (CLh,int) and intrinsic P-gp clearance (CLint,P-gp). The median concentration of DEX in maternal plasma, fetal plasma, fetal lung, and fetal brain were determined, which allowed us to fit the tissue-to-plasma partition coefficients for the fetal lung (Kp,lung,f) and fetal brain (Kp,brain,f). After making adjustments, all calculated FE values were found to be less than 2, demonstrating the acceptability and accuracy of our model's predictions. Our model integrated external literature data and internal animal experimentation to comprehensively evaluate the maternal-fetal PK characteristics of DEX. These findings provide valuable support for the optimization of clinical DEX dosing.
Collapse
Affiliation(s)
- Ruihu Du
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, China
| | - Xiaoqi Zhao
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Ling Song
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China; Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, 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; Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China.
| |
Collapse
|
9
|
Krzyzanski W, Milad MA, Jobe AH, Jusko WJ. Minimal physiologically-based hybrid model of pharmacokinetics in pregnant women: Application to antenatal corticosteroids. CPT Pharmacometrics Syst Pharmacol 2023; 12:668-680. [PMID: 36917704 PMCID: PMC10196440 DOI: 10.1002/psp4.12899] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 03/16/2023] Open
Abstract
Minimal physiologically-based pharmacokinetic (mPBPK) models are an alternative to full physiologically-based pharmacokinetic (PBPK) models as they offer reduced complexity while maintaining the physiological interpretation of key model components. Full PBPK models have been developed for pregnancy, but a mPBPK model eases the ability to perform a "top-down" meta-analysis melding all available pharmacokinetic (PK) data in the mother and fetus. Our hybrid mPBPK model consists of mPBPK models for the mother and fetus with connection by the placenta. This model was applied to describe the rich PK data of antenatal corticosteroid betamethasone (BET) jointly with the limited data for dexamethasone (DEX) in the mother and fetus. Physiologic model parameters were obtained from the literature while drug-dependent parameters were estimated by the simultaneous fitting of all available data for DEX and BET. Maternal clearances of DEX and BET confirmed the literature values, and the expected fetal-to-maternal plasma ratios ranged from 0.3 to 0.4 for both drugs. Simulations of maternal plasma concentrations for the dosing regimens of BET and DEX recommended by the World Health Organization based on our findings revealed up to 60% lower exposures than found in nonpregnant women and offers a means of devising alternative dosing regimens. Our hybrid mPBPK model and meta-analysis approach could facilitate assessment of other classes of drugs indicated for the treatment of pregnant women.
Collapse
Affiliation(s)
- Wojciech Krzyzanski
- School of Pharmacy and Pharmaceutical Sciences, State University of New YorkUniversity of BuffaloBuffaloNew YorkUSA
| | - Mark A. Milad
- Milad Pharmaceutical Consulting LLCPlymouthMichiganUSA
| | - Alan H. Jobe
- Division of Pulmonary BiologyCincinnati Children's Hospital Medical Center, University of CincinnatiCincinnatiOhioUSA
| | - William J. Jusko
- School of Pharmacy and Pharmaceutical Sciences, State University of New YorkUniversity of BuffaloBuffaloNew YorkUSA
| |
Collapse
|
10
|
Takahashi T, Jobe AH, Fee EL, Newnham JP, Schmidt AF, Usuda H, Kemp MW. The complex challenge of antenatal steroid therapy nonresponsiveness. Am J Obstet Gynecol 2022; 227:696-704. [PMID: 35932879 DOI: 10.1016/j.ajog.2022.07.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/01/2022]
Abstract
Antenatal steroid therapy is standard care for women at imminent risk of preterm delivery. When deliveries occur within 7 days of treatment, antenatal steroid therapy reduces the risk of neonatal death and improves preterm outcomes by exerting diverse developmental effects on the fetal organs, in particular the preterm lung and cardiovascular system. There is, however, sizable variability in antenatal steroid treatment efficacy, and an important percentage of fetuses exposed to antenatal steroid therapy do not respond sufficiently to derive benefit. Respiratory distress syndrome, for example, is a central metric of clinical trials to assess antenatal steroid outcomes. In the present analysis, we addressed the concept of antenatal steroid nonresponsiveness, and defined a failed or suboptimal response to antenatal steroids as death or a diagnosis of respiratory distress syndrome following treatment. For deliveries at 24 to 35 weeks' gestation, the number needed to treat to prevent 1 case of respiratory distress syndrome was 19 (95% confidence interval, 14-28). Reflecting gestation-dependent risk, for deliveries at >34 weeks' gestation the number needed to treat was 55 (95% confidence interval, 30-304), whereas for elective surgical deliveries at term this number was 106 (95% confidence interval, 61-421). We reviewed data from clinical and animal studies investigating antenatal steroid therapy to highlight the significant incidence of antenatal steroid therapy nonresponsiveness (ie, residual mortality or respiratory distress syndrome after treatment), and the potential mechanisms underpinning this outcome variability. The origins of this variability may be related to both the manner in which the therapy is applied (ie, the treatment regimen itself) and factors specific to the individual (ie, genetic variation, stress, infection). The primary aims of this review were: (1) to emphasize to the obstetrical and neonatal communities the extent of antenatal steroid response variability and its potential impact; (2) to propose approaches by which antenatal steroid therapy may be better applied to improve overall benefit; and (3) to stimulate further research toward the empirical optimization of this important antenatal therapy.
Collapse
Affiliation(s)
- Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Alan H Jobe
- Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - John P Newnham
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | | | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Matthew W Kemp
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Australia; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
11
|
Prediction of Maternal and Fetal Doravirine Exposure by Integrating Physiologically Based Pharmacokinetic Modeling and Human Placenta Perfusion Experiments. Clin Pharmacokinet 2022; 61:1129-1141. [PMID: 35579825 PMCID: PMC9349081 DOI: 10.1007/s40262-022-01127-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
Background and Objective Doravirine is currently not recommended for pregnant women living with human immunodeficiency virus because efficacy and safety data are lacking. This study aimed to predict maternal and fetal doravirine exposure by integrating human placenta perfusion experiments with pregnancy physiologically based pharmacokinetic (PBPK) modeling. Methods Ex vivo placenta perfusions were performed in a closed–closed configuration, in both maternal-to-fetal and fetal-to-maternal directions (n = 8). To derive intrinsic placental transfer parameters from perfusion data, we developed a mechanistic placenta model. Next, we developed a maternal and fetal full-body pregnancy PBPK model for doravirine in Simcyp, which was parameterized with the derived intrinsic placental transfer parameters to predict in vivo maternal and fetal doravirine exposure at 26, 32, and 40 weeks of pregnancy. The predicted total geometric mean (GM) trough plasma concentration (Ctrough) values were compared with the target (0.23 mg/L) derived from in vivo exposure–response analysis. Results A decrease of 55% in maternal doravirine area under the plasma concentration–time curve (AUC)0–24h was predicted in pregnant women at 40 weeks of pregnancy compared with nonpregnant women. At 26, 32, and 40 weeks of pregnancy, predicted maternal total doravirine GM Ctrough values were below the predefined efficacy target of 0.23 mg/L. Perfusion experiments showed that doravirine extensively crossed the placenta, and PBPK modeling predicted considerable fetal doravirine exposure. Conclusion Substantially reduced maternal doravirine exposure was predicted during pregnancy, possibly resulting in impaired efficacy. Therapeutic drug and viral load monitoring are advised for pregnant women treated with doravirine. Considerable fetal doravirine exposure was predicted, highlighting the need for clinical fetal safety data. Supplementary Information The online version contains supplementary material available at 10.1007/s40262-022-01127-0.
Collapse
|
12
|
Adiwidjaja J, Adattini JA, Boddy AV, McLachlan AJ. Physiologically-Based Pharmacokinetic Modeling Approaches for Patients with SARS-CoV-2 Infection: A Case Study with Imatinib. J Clin Pharmacol 2022; 62:1285-1296. [PMID: 35460539 PMCID: PMC9088354 DOI: 10.1002/jcph.2065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/16/2022] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection, which causes coronavirus disease 2019 (COVID‐19), manifests as mild respiratory symptoms to severe respiratory failure and is associated with inflammation and other physiological changes. Of note, substantial increases in plasma concentrations of α1‐acid‐glycoprotein and interleukin‐6 have been observed among patients admitted to the hospital with advanced SARS‐CoV‐2 infection. A physiologically based pharmacokinetic (PBPK) approach is a useful tool to evaluate and predict disease‐related changes on drug pharmacokinetics. A PBPK model of imatinib has previously been developed and verified in healthy people and patients with cancer. In this study, the PBPK model of imatinib was successfully extrapolated to patients with SARS‐CoV‐2 infection by accounting for disease‐related changes in plasma α1‐acid‐glycoprotein concentrations and the potential drug interaction between imatinib and dexamethasone. The model demonstrated a good predictive performance in describing total and unbound imatinib concentrations in patients with SARS‐CoV‐2 infection. PBPK simulations highlight that an equivalent dose of imatinib may lead to substantially higher total drug concentrations in patients with SARS‐CoV‐2 infection compared to that in patients with cancer, while the unbound concentrations remain comparable between the 2 patient populations. This supports the notion that unbound trough concentration is a better exposure metric for dose adjustment of imatinib in patients with SARS‐CoV‐2 infection, compared to the corresponding total drug concentration. Potential strategies for refinement and generalization of the PBPK modeling approach in the patient population with SARS‐CoV‐2 are also provided in this article, which could be used to guide study design and inform dose adjustment in the future.
Collapse
Affiliation(s)
- Jeffry Adiwidjaja
- Sydney Pharmacy SchoolFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Division of Pharmacotherapy and Experimental TherapeuticsUNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Josephine A. Adattini
- Sydney Pharmacy SchoolFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Alan V. Boddy
- UniSA Cancer Research Institute and UniSA Clinical & Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Andrew J. McLachlan
- Sydney Pharmacy SchoolFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
13
|
Mehta J, Rolta R, Mehta BB, Kaushik N, Choi EH, Kaushik NK. Role of Dexamethasone and Methylprednisolone Corticosteroids in Coronavirus Disease 2019 Hospitalized Patients: A Review. Front Microbiol 2022; 13:813358. [PMID: 35242118 PMCID: PMC8886296 DOI: 10.3389/fmicb.2022.813358] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
The WHO announced coronavirus disease 2019 (COVID-19) as a pandemic disease globally on March 11, 2020, after it emerged in China. The emergence of COVID-19 has lasted over a year, and despite promising vaccine reports that have been produced, we still have a long way to go until such remedies are accessible to everyone. The immunomodulatory strategy has been kept at the top priority for the research agenda for COVID-19. Corticosteroids have been used to modulate the immune response in a wide range of diseases for the last 70 years. These drugs have been shown to avoid and reduce inflammation in tissues and the bloodstream through non-genomic and genomic effects. Now, the use of corticosteroids increased the chance of survival and relief by combating the viral strong inflammatory impacts and has moved to the forefront in the management of patients seeking supplemental oxygen. The goal of this review is to illuminate dexamethasone and methylprednisolone, i.e., in terms of their chemical and physical properties, role in COVID-19 patients suffering from pneumonia, the proposed mode of action in COVID-19, pharmacokinetics, pharmacodynamics, clinical outcomes in immunocompromised populations with COVID-19, interaction with other drugs, and contradiction to explore the trends and perspectives for future research. Literature was searched from scientific databases such as Science Direct, Wiley, Springer, PubMed, and books for the preparation of this review. The RECOVERY trial, a massive, multidisciplinary, randomized, and open-label trial, is mainly accountable for recommendations over the usage of corticosteroids in COVID-19 patients. The corticosteroids such as dexamethasone and methylprednisolone in the form of medication have anti-inflammatory, analgesic, and anti-allergic characteristics, including the ability to inhibit the immune system. These drugs are also recommended for treating symptoms of multiple ailments such as rheumatic and autoimmune diseases, leukemia, multiple myeloma, and Hodgkin’s and non-Hodgkin’s lymphoma along with other drugs. Toxicology studies proved them safe usually at low dosage via oral or other routes.
Collapse
Affiliation(s)
- Jyoti Mehta
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Rajan Rolta
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | | | - Neha Kaushik
- Department of Biotechnology, The University of Suwon, Hwaseong, South Korea
| | - Eun Ha Choi
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, South Korea
| | - Nagendra Kumar Kaushik
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, South Korea
| |
Collapse
|
14
|
Montanha MC, Cottura N, Booth M, Hodge D, Bunglawala F, Kinvig H, Grañana-Castillo S, Lloyd A, Khoo S, Siccardi M. PBPK Modelling of Dexamethasone in Patients With COVID-19 and Liver Disease. Front Pharmacol 2022; 13:814134. [PMID: 35153785 PMCID: PMC8832977 DOI: 10.3389/fphar.2022.814134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
The aim of the study was to apply Physiologically-Based Pharmacokinetic (PBPK) modelling to predict the effect of liver disease (LD) on the pharmacokinetics (PK) of dexamethasone (DEX) in the treatment of COVID-19. A whole-body PBPK model was created to simulate 100 adult individuals aged 18–60 years. Physiological changes (e.g., plasma protein concentration, liver size, CP450 expression, hepatic blood flow) and portal vein shunt were incorporated into the LD model. The changes were implemented by using the Child-Pugh (CP) classification system. DEX was qualified using clinical data in healthy adults for both oral (PO) and intravenous (IV) administrations and similarly propranolol (PRO) and midazolam (MDZ) were qualified with PO and IV clinical data in healthy and LD adults. The qualified model was subsequently used to simulate a 6 mg PO and 20 mg IV dose of DEX in patients with varying degrees of LD, with and without shunting. The PBPK model was successfully qualified across DEX, MDZ and PRO. In contrast to healthy adults, the simulated systemic clearance of DEX decreased (35%–60%) and the plasma concentrations increased (170%–400%) in patients with LD. Moreover, at higher doses of DEX, the AUC ratio between healthy/LD individuals remained comparable to lower doses. The exposure of DEX in different stages of LD was predicted through PBPK modelling, providing a rational framework to predict PK in complex clinical scenarios related to COVID-19. Model simulations suggest dose adjustments of DEX in LD patients are not necessary considering the low dose administered in the COVID-19 protocol.
Collapse
|
15
|
Zheng L, Yang H, Dallmann A, Jiang X, Wang L, Hu W. Physiologically Based Pharmacokinetic Modeling in Pregnant Women Suggests Minor Decrease in Maternal Exposure to Olanzapine. Front Pharmacol 2022; 12:793346. [PMID: 35126130 PMCID: PMC8807508 DOI: 10.3389/fphar.2021.793346] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023] Open
Abstract
Pregnancy is accompanied by significant physiological changes that might affect the in vivo drug disposition. Olanzapine is prescribed to pregnant women with schizophrenia, while its pharmacokinetics during pregnancy remains unclear. This study aimed to develop a physiologically based pharmacokinetic (PBPK) model of olanzapine in the pregnant population. With the contributions of each clearance pathway determined beforehand, a full PBPK model was developed and validated in the non-pregnant population. This model was then extrapolated to predict steady-state pharmacokinetics in the three trimesters of pregnancy by introducing gestation-related alterations. The model adequately simulated the reported time-concentration curves. The geometric mean fold error of Cmax and AUC was 1.14 and 1.09, respectively. The model predicted that under 10 mg daily dose, the systematic exposure of olanzapine had minor changes (less than 28%) throughout pregnancy. We proposed that the reduction in cytochrome P4501A2 activity is counteracted by the induction of other enzymes, especially glucuronyltransferase1A4. In conclusion, the PBPK model simulations suggest that, at least at the tested stages of pregnancy, dose adjustment of olanzapine can hardly be recommended for pregnant women if effective treatment was achieved before the onset of pregnancy and if fetal toxicity can be ruled out.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hongyi Yang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals Bayer AG, Leverkusen, Germany
| | - Xuehua Jiang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Ling Wang, ; Wei Hu,
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ling Wang, ; Wei Hu,
| |
Collapse
|
16
|
Rodrigues GDFP, Benzi JRDL, Matos LHDC, de Freitas SF, Marques MP, Cavalli RDC, Moisés ECD, Duarte G, Lanchote VL, Marcolin AC. Enhanced elimination of betamethasone in dichorionic twin pregnancies. Br J Clin Pharmacol 2021; 88:1897-1903. [PMID: 34665470 DOI: 10.1111/bcp.15111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/27/2022] Open
Abstract
AIM No study has evaluated the betamethasone pharmacokinetics in twin pregnancies according to chorionicity. This study aimed to describe and compare the betamethasone pharmacokinetic parameters in singleton and dichorionic (DC) and monochorionic twin pregnancies in the third trimester of pregnancy. METHODS Twenty-six pregnant women received 2 intramuscular doses of 6 mg of betamethasone sodium phosphate plus 6 mg betamethasone acetate due to preterm labour. Serial blood samples were collected for 24 hours after the first intramuscular dose of betamethasone esters. Betamethasone plasma concentrations were quantified using a validated liquid chromatography-tandem mass spectrometry analytical method, and the pharmacokinetic parameters were obtained employing a noncompartmental model. Preliminary data on the betamethasone placental transfer are also presented. RESULTS The geometric mean (95% confidence interval) of AUC0-∞ 645.1 (504.3-825.2) vs. 409.8 (311.2-539.6) ng.h/mL and CL/F 17.70 (13.84-22.65) vs. 27.87 (21.17-36.69) were significantly different, respectively, in singleton pregnancies when compared to DC twins. CONCLUSION Data from this study suggest that the presence of 2 foetoplacental units may increase the betamethasone metabolism by hepatic CYP3A4 and/or placental 11β-HSD2 enzymes. Pharmacokinetic-pharmacodynamic clinical studies are needed to investigate whether these betamethasone pharmacokinetic changes have clinical repercussions for the newborns and require dose adjustment in DC twin pregnancies.
Collapse
Affiliation(s)
| | - Jhohann Richard de Lima Benzi
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luísa Helena de Castro Matos
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Stella Felippe de Freitas
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria Paula Marques
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elaine Christine Dantas Moisés
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Geraldo Duarte
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Alessandra Cristina Marcolin
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
17
|
Silva LL, Silvola RM, Haas DM, Quinney SK. Physiologically based pharmacokinetic modelling in pregnancy: Model reproducibility and external validation. Br J Clin Pharmacol 2021; 88:1441-1451. [PMID: 34337764 DOI: 10.1111/bcp.15018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 02/02/2023] Open
Abstract
AIMS Physiologically based pharmacokinetic (PBPK) models have been previously developed for betamethasone and buprenorphine for pregnant women. The goal of this work was to replicate and reassess these models using data from recently completed studies. METHODS Betamethasone and buprenorphine PBPK models were developed in Simcyp V19 based on prior publications using V17 and V15. Ability to replicate models was verified by comparing predictions in V19 to those previously published. Once replication was verified, models were reassessed by comparing predictions to observed data from additional studies in pregnant women. Model performance was based upon visual inspection of concentration vs. time profiles, and comparison of pharmacokinetic parameters. Models were deemed reproducible if parameter estimates were within 10% of previously reported values. External validations were considered acceptable if the predicted area under the concentration-time curve (AUC) and peak plasma concentration fell within 2-fold of the observed. RESULTS The betamethasone model was successfully replicated using Simcyp V19, with ratios of reported (V17) to reproduced (V19) peak plasma concentration of 0.98-1.04 and AUC of 0.95-1.07. The model-predicted AUC ratios ranged from 0.98-1.79 compared to external data. The previously published buprenorphine PBPK model was not reproducible, as we predicted intravenous clearance of 70% that reported previously (both in Simcyp V15). CONCLUSION While high interstudy variability was observed in the newly available clinical data, the PBPK model sufficiently predicted changes in betamethasone exposure across gestation. Model reproducibility and reassessment with external data are important for the advancement of the discipline. PBPK modelling publications should contain sufficient detail and clarity to enable reproducibility.
Collapse
Affiliation(s)
- Larissa L Silva
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rebecca M Silvola
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David M Haas
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara K Quinney
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
18
|
Anoshchenko O, Milad MA, Unadkat JD. Estimating fetal exposure to the P-gp substrates, corticosteroids, by PBPK modeling to inform prevention of neonatal respiratory distress syndrome. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1057-1070. [PMID: 34273255 PMCID: PMC8452292 DOI: 10.1002/psp4.12674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 11/08/2022]
Abstract
We have previously developed a maternal-fetal physiologically-based pharmacokinetic (m-f PBPK) model to dynamically predict (and verify) fetal-maternal exposure to drugs that passively diffuse across the placenta. Here, we extended the application of this model to dynamically predict fetal exposure to drugs which are effluxed by placental P-glycoprotein, namely the antenatal corticosteroids (ACS; dexamethasone [DEX], and betamethasone [BET]). To do so, we estimated both the placental P-gp mediated efflux clearance (CL) and the passive diffusion CL of the ACS. The efficacy and toxicity of the currently used maternal ACS dosing regimens to prevent neonatal respiratory distress syndrome could be improved by altering their dosing regimens. Therefore, to illustrate the utility of our m-f PBPK model, we used it to design alternative dosing regimens of DEX and BET that could potentially improve their efficacy and reduce their toxicity. The redesigned dosing regimens are convenient to administer, maintain maternal-fetal exposure (area under the concentration-time curve [AUC]) or maximum plasma concentration (Cmax ) or both (DEX and BET) or minimize maternal exposure while maintaining fetal drug plasma concentrations above the minimum therapeutic threshold of 1 ng/ml for 48 h (BET only; based on efficacy data in sheep). To our knowledge, this is the first study to dynamically predict fetal plasma concentrations of placental P-gp effluxed drugs. Our approach and our m-f PBPK model could be used in the future to predict maternal-fetal exposure to any drug and to design alternative dosing regimens of the drug.
Collapse
Affiliation(s)
- Olena Anoshchenko
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Washington, Seattle, USA
| | - Mark A Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Washington, Seattle, USA
| |
Collapse
|
19
|
Krzyzanski W, Milad MA, Jobe AH, Peppard T, Bies RR, Jusko WJ. Population pharmacokinetic modeling of intramuscular and oral dexamethasone and betamethasone in Indian women. J Pharmacokinet Pharmacodyn 2021; 48:261-272. [PMID: 33389521 PMCID: PMC7778726 DOI: 10.1007/s10928-020-09730-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/20/2020] [Indexed: 11/01/2022]
Abstract
Population analysis of pharmacokinetic data for five differing dosage forms and routes for dexamethasone and betamethasone in 48 healthy nonpregnant Indian women was performed that accounted for a partial and complex cross-over design. Single doses of 6 mg dexamethasone phosphate (DEX-P), betamethasone phosphate (BET-P), or 1:1 mixture of betamethasone phosphate and acetate (BET-PA) were administered orally (PO) or intramuscularly (IM). Plasma concentrations collected for two periods over 96 h were described with a two-compartment model with differing PO and IM first-order absorption inputs. Clearances and volumes were divided by the IM bioavailability [Formula: see text]. The homogeneous ages, body weights, and ethnicity of the women obviated covariate analysis. Parameter estimates were obtained by the Laplace estimation method implemented in NONMEM 7.4. Typical values for dexamethasone were clearance ([Formula: see text] of 9.29 L/h, steady-state volume ([Formula: see text] of 56.4 L, IM absorption constant [Formula: see text] of 0.460 1/h and oral absorption constant ([Formula: see text] of 0.936 1/h. Betamethasone parameters were CL/FIM of 5.95 L/h, [Formula: see text] of 72.4 L, [Formula: see text] of 0.971 1/h, and [Formula: see text] of 1.21 1/h. The PO to IM F values were close to 1.0 for both drugs. The terminal half-lives averaged about 7.5 h for DEX, 17 h for BET, and 78 h for BET from BET-PA with the latter reflecting very slow release of BET from the acetate ester. Overall, BET exhibited slower clearance, larger volume of distribution, faster absorption, and longer persistence than DEX. These data may be useful in considering exposures when substituting one form of corticosteroid for another.
Collapse
Affiliation(s)
- Wojciech Krzyzanski
- School of Pharmacy and Pharmaceutical Sciences, State University of New York, University of Buffalo, Buffalo, NY, USA
| | - Mark A Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, MI, USA
| | - Alan H Jobe
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - Robert R Bies
- School of Pharmacy and Pharmaceutical Sciences, State University of New York, University of Buffalo, Buffalo, NY, USA
| | - William J Jusko
- School of Pharmacy and Pharmaceutical Sciences, State University of New York, University of Buffalo, Buffalo, NY, USA.
| |
Collapse
|
20
|
Song D, Jusko WJ. Across‐species meta‐analysis of dexamethasone pharmacokinetics utilizing allometric and scaling modeling approaches. Biopharm Drug Dispos 2021; 42:191-203. [DOI: 10.1002/bdd.2266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Dawei Song
- Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences State University of New York at Buffalo Buffalo New York USA
| | - William J Jusko
- Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences State University of New York at Buffalo Buffalo New York USA
| |
Collapse
|
21
|
Vazquez B, Tomson T, Dobrinsky C, Schuck E, O'Brien TJ. Perampanel and pregnancy. Epilepsia 2021; 62:698-708. [PMID: 33666943 PMCID: PMC7986165 DOI: 10.1111/epi.16821] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/15/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022]
Abstract
Objective The objective was to summarize pregnancy and fetal/postnatal outcomes following maternal perampanel exposure using preclinical and clinical data, and to use physiologically based pharmacokinetic (PBPK) modeling to improve understanding of perampanel pharmacokinetics (PK) during pregnancy. Methods Preclinical developmental studies with perampanel were conducted in pregnant rats and rabbits. Clinical data were collated from the Eisai global perampanel safety database, comprising reports of perampanel exposure during pregnancy from routine clinical settings, interventional studies, and non‐interventional post‐marketing studies, searched for events coded to Medical Dictionary for Regulatory Activities (MedDRA) high‐level group terms of Pregnancy, Labor, Delivery, and Postpartum Conditions and/or the Standardized MedDRA Query terms of Congenital, Familiar, and Genetic Disorders. A PBPK model was used to predict clinical perampanel PK throughout pregnancy. Results Preclinical studies indicated that perampanel may be linked with post‐implantation loss and/or some specific physical development delays but not fertility and early embryonic development. As of August 31, 2018, 96 pregnancies in 90 women receiving perampanel had been reported. No concomitant medications were reported in 26 (28.9%) women taking perampanel. Overall, 43 pregnancies reached full term (all normal live births), 28 did not reach term (induced abortion, n = 18; spontaneous miscarriage, n = 6; incomplete spontaneous miscarriage, n = 2; premature delivery, n = 1; stillbirth [Fallot’s tetralogy], n = 1), 18 were lost to follow‐up, and seven were ongoing at data cut‐off. Adverse events were reported in five full‐term neonates (low Apgar score, n = 2; fatal neonatal aspiration, n = 1; cystic fibrosis and congenital deafness, n = 1; poor sucking reflex and shallow breathing, n = 1). PK simulations predicted perampanel exposure decreases throughout pregnancy and is up to four‐ and three‐fold lower towards the end of pregnancy compared with non‐pregnant women for total and unbound perampanel, respectively. Significance These data provide preliminary information on perampanel use during pregnancy and should be interpreted with caution. Further outcome data are required to estimate the prevalence of adverse pregnancy outcomes with perampanel exposure.
Collapse
Affiliation(s)
- Blanca Vazquez
- NYU Langone Comprehensive Epilepsy Center, New York, New York, USA
| | - Torbjörn Tomson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
22
|
Jobe AH, Kemp M, Schmidt A, Takahashi T, Newnham J, Milad M. Antenatal corticosteroids: a reappraisal of the drug formulation and dose. Pediatr Res 2021; 89:318-325. [PMID: 33177675 PMCID: PMC7892336 DOI: 10.1038/s41390-020-01249-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022]
Abstract
We review the history of antenatal corticosteroid therapy (ACS) and present recent experimental data to demonstrate that this, one of the pillars of perinatal care, has been inadequately evaluated to minimize fetal exposure to these powerful medications. There have been concerns since 1972 that fetal exposures to ACS convey risk. However, this developmental modulator, with its multiple widespread biologic effects, has not been evaluated for drug choice, dose, or duration of treatment, despite over 30 randomized trials. The treatment used in the United States is two intramuscular doses of a mixture of 6 mg betamethasone phosphate (Beta P) and 6 mg betamethasone acetate (Beta Ac). To optimize outcomes with ACS, the goal should be to minimize fetal drug exposure. We have determined that the minimum exposure needed for fetal lung maturation in sheep, monkeys, and humans (based on published cord blood corticosteroid concentrations) is about 1 ng/ml for a 48-h continuous exposure, far lower than the concentration reached by the current dosing. Because the slowly released Beta Ac results in prolonged fetal exposure, a drug containing Beta Ac is not ideal for ACS use. IMPACT: Using sheep and monkey models, we have defined the minimum corticosteroid exposure for a fetal lung maturation. These results should generate new clinical trials of antenatal corticosteroids (ACS) at much lower fetal exposures to ACS, possibly given orally, with fewer risks for the fetus.
Collapse
Affiliation(s)
- Alan H. Jobe
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.24827.3b0000 0001 2179 9593Perinatal Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA
| | - Matthew Kemp
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.412757.20000 0004 0641 778XCentre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan ,grid.1025.60000 0004 0436 6763School of Veterinary and Life Sciences, Murdoch University, Perth, WA Australia
| | - Augusto Schmidt
- grid.26790.3a0000 0004 1936 8606Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL USA
| | - Tsukasa Takahashi
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.412757.20000 0004 0641 778XCentre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - John Newnham
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia
| | - Mark Milad
- Milad Pharmaceutical Consulting, Plymouth, MI USA
| |
Collapse
|
23
|
Chaphekar N, Dodeja P, Shaik IH, Caritis S, Venkataramanan R. Maternal-Fetal Pharmacology of Drugs: A Review of Current Status of the Application of Physiologically Based Pharmacokinetic Models. Front Pediatr 2021; 9:733823. [PMID: 34805038 PMCID: PMC8596611 DOI: 10.3389/fped.2021.733823] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/16/2021] [Indexed: 12/31/2022] Open
Abstract
Pregnancy and the postpartum period are associated with several physiological changes that can alter the pharmacokinetics (PK) and pharmacodynamics (PD) of drugs. For certain drugs, dosing changes may be required during pregnancy and postpartum to achieve drug exposures comparable to what is observed in non-pregnant subjects. There is very limited data on fetal exposure of drugs during pregnancy, and neonatal exposure through transfer of drugs via human milk during breastfeeding. Very few systematic clinical pharmacology studies have been conducted in pregnant and postpartum women due to ethical issues, concern for the fetus safety as well as potential legal ramifications. Over the past several years, there has been an increase in the application of modeling and simulation approaches such as population PK (PopPK) and physiologically based PK (PBPK) modeling to provide guidance on drug dosing in those special patient populations. Population PK models rely on measured PK data, whereas physiologically based PK models incorporate physiological, preclinical, and clinical data into the model to predict drug exposure during pregnancy. These modeling strategies offer a promising approach to identify the drugs with PK changes during pregnancy to guide dose optimization in pregnancy, when there is lack of clinical data. PBPK modeling is also utilized to predict the fetal exposure of drugs and drug transfer via human milk following maternal exposure. This review focuses on the current status of the application of PBPK modeling to predict maternal and fetal exposure of drugs and thereby guide drug therapy during pregnancy.
Collapse
Affiliation(s)
- Nupur Chaphekar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Prerna Dodeja
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Imam H Shaik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Steve Caritis
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee Women's Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Obstetrics, Gynecology and Reproductive Sciences, Magee Women's Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Ninan K, Morfaw F, Murphy KE, Beyene J, McDonald SD. Neonatal and Maternal Outcomes of Lower Versus Standard Doses of Antenatal Corticosteroids for Women at Risk of Preterm Delivery: A Systematic Review of Randomized Controlled Trials. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2021; 43:74-81. [PMID: 32660867 DOI: 10.1016/j.jogc.2020.02.127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Our objective was to systematically review randomized and quasi-randomized trials on the neonatal and maternal effects of lower doses of antenatal corticosteroids (<24 mg of betamethasone or dexamethasone) compared with standard double doses of antenatal corticosteroids (24 mg of betamethasone or dexamethasone) administered to women at risk of preterm delivery. DATA SOURCES Medline, Embase, CINAHL, Web of Science, Cochrane CENTRAL, ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform, and the Australia New Zealand Clinical Trials Registry were searched from inception to December 8, 2019. STUDY SELECTION A total of 2401 titles, abstracts, and protocols were independently screened by two reviewers, and subsequently 113 full-text articles were reviewed. DATA EXTRACTION Our primary outcomes were perinatal death and severe respiratory distress syndrome. DATA SYNTHESIS We identified one large in-progress trial comparing 11.4 mg versus 22.8 mg betamethasone and one published randomized controlled trial that compared a lower dose of dexamethasone (16 mg) to a standard dose of betamethasone (24 mg). The only relevant data from the published trial suggests minor changes in fetal heart rate variability between baseline and 24- to 48-hour follow-up between the two groups. Data for other outcomes had to be excluded due to the administration of weekly courses of antenatal corticosteroids. CONCLUSIONS Randomized trial data comparing lower doses of antenatal corticosteroids to standard double doses are scarce. Given concerns regarding current antenatal corticosteroids dosing patterns, there is an urgent need for randomized controlled trials examining lower versus standard double doses of antenatal corticosteroids.
Collapse
Affiliation(s)
- Kiran Ninan
- Department of Obstetrics and Gynaecology, McMaster University, Hamilton, ON
| | - Frederick Morfaw
- Department of Obstetrics and Gynaecology, McMaster University, Hamilton, ON
| | - Kellie E Murphy
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON
| | - Joseph Beyene
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON
| | - Sarah D McDonald
- Department of Obstetrics and Gynaecology, McMaster University, Hamilton, ON; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON; Department of Radiology, McMaster University, Hamilton, ON.
| |
Collapse
|
25
|
Zheng L, Tang S, Tang R, Xu M, Jiang X, Wang L. Dose Adjustment of Quetiapine and Aripiprazole for Pregnant Women Using Physiologically Based Pharmacokinetic Modeling and Simulation. Clin Pharmacokinet 2020; 60:623-635. [DOI: 10.1007/s40262-020-00962-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 12/12/2022]
|
26
|
George B, Lumen A, Nguyen C, Wesley B, Wang J, Beitz J, Crentsil V. Application of physiologically based pharmacokinetic modeling for sertraline dosing recommendations in pregnancy. NPJ Syst Biol Appl 2020; 6:36. [PMID: 33159093 PMCID: PMC7648747 DOI: 10.1038/s41540-020-00157-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/02/2020] [Indexed: 01/26/2023] Open
Abstract
Pregnancy is a period of significant change that impacts physiological and metabolic status leading to alterations in the disposition of drugs. Uncertainty in drug dosing in pregnancy can lead to suboptimal therapy, which can contribute to disease exacerbation. A few studies show there are increased dosing requirements for antidepressants in late pregnancy; however, the quantitative data to guide dose adjustments are sparse. We aimed to develop a physiologically based pharmacokinetic (PBPK) model that allows gestational-age dependent prediction of sertraline dosing in pregnancy. A minimal physiological model with defined gut, liver, plasma, and lumped placental-fetal compartments was constructed using the ordinary differential equation solver package, ‘mrgsolve’, in R. We extracted data from the literature to parameterize the model, including sertraline physicochemical properties, in vitro metabolism studies, disposition in nonpregnant women, and physiological changes during pregnancy. The model predicted the pharmacokinetic parameters from a clinical study with eight subjects for the second trimester and six subjects for the third trimester. Based on the model, gestational-dependent changes in physiology and metabolism account for increased clearance of sertraline (up to 143% at 40 weeks gestational age), potentially leading to under-dosing of pregnant women when nonpregnancy doses are used. The PBPK model was converted to a prototype web-based interactive dosing tool to demonstrate how the output of a PBPK model may translate into optimal sertraline dosing in pregnancy. Quantitative prediction of drug exposure using PBPK modeling in pregnancy will support clinically appropriate dosing and increase the therapeutic benefit for pregnant women.
Collapse
Affiliation(s)
- Blessy George
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA.,Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Annie Lumen
- National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Christine Nguyen
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Barbara Wesley
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Jian Wang
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Julie Beitz
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA
| | - Victor Crentsil
- Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, USA.
| |
Collapse
|
27
|
Song D, Sun L, DuBois DC, Almon RR, Meng S, Jusko WJ. Physiologically Based Pharmacokinetics of Dexamethasone in Rats. Drug Metab Dispos 2020; 48:811-818. [PMID: 32601175 PMCID: PMC7448200 DOI: 10.1124/dmd.120.091017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Blood and multitissue concentration-time profiles for dexamethasone (DEX), a synthetic corticosteroid, were measured in male rats after subcutaneous bolus and infusion dosing. A physiologically based pharmacokinetics (PBPK) model was applied for 12 measured tissues. Tissue partition coefficients (K p ) and metabolic clearance were assessed from infusion studies. Blood cell to plasma partitioning (0.664) and plasma free fraction (0.175) for DEX were found to be moderate. DEX was extensively partitioned into liver (K p = 6.76), whereas the calculated K p values of most tissues ranged between 0.1 and 1.5. Despite the moderate lipophilicity of DEX (log P = 1.8), adipose exhibited very limited distribution (K p = 0.17). Presumably due to P-glycoprotein-mediated efflux, DEX concentrations were very low in brain compared with its expected high permeability. Infusion studies yielded K p values from male and female rats at steady state that were similar. In silico K p values calculated for different tissues by using GastroPlus software were similar to in vivo values except for adipose and liver. Glucocorticoid receptors are found in diverse tissues, and these PBPK modeling results may help provide exposure profiles driving pharmacodynamic effects of DEX. SIGNIFICANCE STATEMENT: Our physiologically based pharmacokinetics model describes the experimentally determined tissue and plasma dexamethasone (DEX) pharmacokinetics (PK) profiles in rats reasonably well. This model can serve for further investigation of DEX tissue distribution in rats as the PK driving force for PD effects in different tissues. No major sex differences were found for DEX tissue distribution. Knowledge gained in this study may be translatable to higher-order species including humans.
Collapse
Affiliation(s)
- Dawei Song
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| | - Le Sun
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| | - Debra C DuBois
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| | - Richard R Almon
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| | - Shengnan Meng
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| | - William J Jusko
- Departments of Pharmaceutical Sciences (D.S., L.S., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York; and Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China (S.M.)
| |
Collapse
|
28
|
Abduljalil K, Badhan RKS. Drug dosing during pregnancy-opportunities for physiologically based pharmacokinetic models. J Pharmacokinet Pharmacodyn 2020; 47:319-340. [PMID: 32592111 DOI: 10.1007/s10928-020-09698-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/20/2020] [Indexed: 12/15/2022]
Abstract
Drugs can have harmful effects on the embryo or the fetus at any point during pregnancy. Not all the damaging effects of intrauterine exposure to drugs are obvious at birth, some may only manifest later in life. Thus, drugs should be prescribed in pregnancy only if the expected benefit to the mother is thought to be greater than the risk to the fetus. Dosing of drugs during pregnancy is often empirically determined and based upon evidence from studies of non-pregnant subjects, which may lead to suboptimal dosing, particularly during the third trimester. This review collates examples of drugs with known recommendations for dose adjustment during pregnancy, in addition to providing an example of the potential use of PBPK models in dose adjustment recommendation during pregnancy within the context of drug-drug interactions. For many drugs, such as antidepressants and antiretroviral drugs, dose adjustment has been recommended based on pharmacokinetic studies demonstrating a reduction in drug concentrations. However, there is relatively limited (and sometimes inconsistent) information regarding the clinical impact of these pharmacokinetic changes during pregnancy and the effect of subsequent dose adjustments. Examples of using pregnancy PBPK models to predict feto-maternal drug exposures and their applications to facilitate and guide dose assessment throughout gestation are discussed.
Collapse
Affiliation(s)
- Khaled Abduljalil
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | | |
Collapse
|
29
|
Ayyar VS, Jusko WJ. Transitioning from Basic toward Systems Pharmacodynamic Models: Lessons from Corticosteroids. Pharmacol Rev 2020; 72:414-438. [PMID: 32123034 PMCID: PMC7058984 DOI: 10.1124/pr.119.018101] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Technology in bioanalysis, -omics, and computation have evolved over the past half century to allow for comprehensive assessments of the molecular to whole body pharmacology of diverse corticosteroids. Such studies have advanced pharmacokinetic and pharmacodynamic (PK/PD) concepts and models that often generalize across various classes of drugs. These models encompass the "pillars" of pharmacology, namely PK and target drug exposure, the mass-law interactions of drugs with receptors/targets, and the consequent turnover and homeostatic control of genes, biomarkers, physiologic responses, and disease symptoms. Pharmacokinetic methodology utilizes noncompartmental, compartmental, reversible, physiologic [full physiologically based pharmacokinetic (PBPK) and minimal PBPK], and target-mediated drug disposition models using a growing array of pharmacometric considerations and software. Basic PK/PD models have emerged (simple direct, biophase, slow receptor binding, indirect response, irreversible, turnover with inactivation, and transduction models) that place emphasis on parsimony, are mechanistic in nature, and serve as highly useful "top-down" methods of quantitating the actions of diverse drugs. These are often components of more complex quantitative systems pharmacology (QSP) models that explain the array of responses to various drugs, including corticosteroids. Progressively deeper mechanistic appreciation of PBPK, drug-target interactions, and systems physiology from the molecular (genomic, proteomic, metabolomic) to cellular to whole body levels provides the foundation for enhanced PK/PD to comprehensive QSP models. Our research based on cell, animal, clinical, and theoretical studies with corticosteroids have provided ideas and quantitative methods that have broadly advanced the fields of PK/PD and QSP modeling and illustrates the transition toward a global, systems understanding of actions of diverse drugs. SIGNIFICANCE STATEMENT: Over the past half century, pharmacokinetics (PK) and pharmacokinetics/pharmacodynamics (PK/PD) have evolved to provide an array of mechanism-based models that help quantitate the disposition and actions of most drugs. We describe how many basic PK and PK/PD model components were identified and often applied to the diverse properties of corticosteroids (CS). The CS have complications in disposition and a wide array of simple receptor-to complex gene-mediated actions in multiple organs. Continued assessments of such complexities have offered opportunities to develop models ranging from simple PK to enhanced PK/PD to quantitative systems pharmacology (QSP) that help explain therapeutic and adverse CS effects. Concurrent development of state-of-the-art PK, PK/PD, and QSP models are described alongside experimental studies that revealed diverse CS actions.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| |
Collapse
|
30
|
Jobe AH, Milad MA, Peppard T, Jusko WJ. Pharmacokinetics and Pharmacodynamics of Intramuscular and Oral Betamethasone and Dexamethasone in Reproductive Age Women in India. Clin Transl Sci 2020; 13:391-399. [PMID: 31808984 PMCID: PMC7070803 DOI: 10.1111/cts.12724] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/24/2019] [Indexed: 01/23/2023] Open
Abstract
High-dose betamethasone and dexamethasone are standard of care treatments for women at risk of preterm delivery to improve neonatal respiratory and mortality outcomes. The dose in current use has never been evaluated to minimize exposures while assuring efficacy. We report the pharmacokinetics and pharmacodynamics (PDs) of oral and intramuscular treatments with single 6 mg doses of dexamethasone phosphate, betamethasone phosphate, or a 1:1 mixture of betamethasone phosphate and betamethasone acetate in reproductive age South Asian women. Intramuscular or oral betamethasone has a terminal half-life of 11 hours, about twice as long as the 5.5 hours for oral and intramuscular dexamethasone. The 1:1 mixture of betamethasone phosphate and betamethasone acetate shows an immediate release of betamethasone followed by a slow release where plasma betamethasone can be measured out to 14 days after the single dose administration, likely from a depo formed at the injection site by the acetate. PD responses were: increased glucose, suppressed cortisol, increased neutrophils, and suppressed basophils, CD3CD4 and CD3CD8 lymphocytes. PD responses were comparable for betamethasone and dexamethasone, but with longer times to return to baseline for betamethasone. The 1:1 mixture of betamethasone phosphate and betamethasone acetate caused much longer adrenal suppression because of the slow release. These results will guide the development of better treatment strategies to minimize fetal and maternal drug exposures for women at risk of preterm delivery.
Collapse
Affiliation(s)
- Alan H. Jobe
- Division of Pulmonary BiologyCincinnati Children's Hospital Medical CenterUniversity of CincinnatiCincinnatiOhioUSA
| | - Mark A. Milad
- Milad Pharmaceutical Consulting LLCPlymouthMichiganUSA
| | | | - William J. Jusko
- State University of New YorkSchool of Pharmacy and Pharmaceutical SciencesUniversity of BuffaloBuffaloNew YorkUSA
| |
Collapse
|
31
|
Schmidt AF, Kemp MW, Milad M, Miller LA, Bridges JP, Clarke MW, Kannan PS, Jobe AH. Oral dosing for antenatal corticosteroids in the Rhesus macaque. PLoS One 2019; 14:e0222817. [PMID: 31536601 PMCID: PMC6752828 DOI: 10.1371/journal.pone.0222817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/06/2019] [Indexed: 02/04/2023] Open
Abstract
Antenatal corticosteroids (ACS) are standard of care for women at risk of preterm delivery, although choice of drug, dose or route have not been systematically evaluated. Further, ACS are infrequently used in low resource environments where most of the mortality from prematurity occurs. We report proof of principle experiments to test betamethasone-phosphate (Beta-P) or dexamethasone-phosphate (Dex-P) given orally in comparison to the clinical treatment with the intramuscular combination drug beta-phosphate plus beta-acetate in a Rhesus Macaque model. First, we performed pharmacokinetic studies in non-pregnant monkeys to compare blood levels of the steroids using oral dosing with Beta-P, Dex-P and an effective maternal intramuscular dose of the beta-acetate component of the clinical treatment. We then evaluated maternal and fetal blood steroid levels with limited fetal sampling under ultrasound guidance in pregnant macaques. We found that oral Beta is more slowly cleared from plasma than oral Dex. The blood levels of both drugs were lower in maternal plasma of pregnant than in non-pregnant macaques. Using the pharmacokinetic data, we treated groups of 6–8 pregnant monkeys with oral Beta-P, oral Dex-P, or the maternal intramuscular clinical treatment and saline controls and measured pressure-volume curves to assess corticosteroid effects on lung maturation at 5d. Oral Beta-P improved the pressure-volume curves similarly to the clinical treatment. Oral Dex-P gave more variable and nonsignificant responses. We then compared gene expression in the fetal lung, liver and hippocampus between oral Beta-P and the clinical treatment by RNA-sequencing. The transcriptomes were largely similar with small gene expression differences in the lung and liver, and no differences in the hippocampus between the groups. As proof of principle, ACS therapy can be effective using inexpensive and widely available oral drugs. Clinical dosing strategies must carefully consider the pharmacokinetics of oral Beta-P or Dex-P to minimize fetal exposure while achieving the desired treatment responses.
Collapse
Affiliation(s)
- Augusto F. Schmidt
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | | | - Mark Milad
- Milad Pharmaceutical consulting LLC, Plymouth, Michigan, United States of America
| | - Lisa A. Miller
- California National Primate Research Center, University of California, Davis, Davis, California, United States of America
| | - James P. Bridges
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Michael W. Clarke
- Metabolomics Australia, Centre for Microscopy, Characterization and Analysis, The University of Western Australia, Perth, WA, Australia
| | - Paranthaman S. Kannan
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Alan H. Jobe
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
- University of Western Australia, Perth, Australia
- * E-mail:
| |
Collapse
|