1
|
Deferm N, Dinh J, Pansari A, Jamei M, Abduljalil K. Postpartum changes in maternal physiology and milk composition: a comprehensive database for developing lactation physiologically-based pharmacokinetic models. Front Pharmacol 2025; 16:1517069. [PMID: 39963246 PMCID: PMC11830814 DOI: 10.3389/fphar.2025.1517069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Pharmacotherapy during lactation often lacks reliable drug safety data, resulting in delayed treatment or early cessation of breastfeeding. In silico tools, such as physiologically-based pharmacokinetic (PBPK) models, can help to bridge this knowledge gap. To increase the accuracy of these models, it is essential to account for the physiological changes that occur throughout the postpartum period. Methods This study aimed to collect and analyze data on the longitudinal changes in various physiological parameters that can affect drug distribution into breast milk during lactation. Following meta-analysis of the collated data, mathematical functions were fitted to the available data for each parameter. The best-performing functions were selected through numerical and visual diagnostics. Results and Discussion The literature search identified 230 studies, yielding a dataset of 36,689 data points from 20,801 postpartum women, covering data from immediately after childbirth to 12 months postpartum. Sufficient data were obtained to describe postpartum changes in maternal plasma volume, breast volume, cardiac output, glomerular filtration rate, haematocrit, human serum albumin, alpha-1-acid glycoprotein, milk pH, milk volume, milk fat, milk protein, milk water content, and daily infant milk intake. Although data beyond 7 months postpartum were limited for some parameters, mathematical functions were generated for all parameters. These functions can be integrated into lactation PBPK models to increase their predictive power and better inform medication efficacy and safety for breastfeeding women.
Collapse
Affiliation(s)
- Neel Deferm
- Predictive Technologies Division, Certara UK Limited, Sheffield, United Kingdom
- Pharmides BV, Pelt, Belgium
| | - Jean Dinh
- Predictive Technologies Division, Certara UK Limited, Sheffield, United Kingdom
- School of Pharmacy, Department of Pharmacy, University of Washington, Seattle, WA, United States
| | - Amita Pansari
- Predictive Technologies Division, Certara UK Limited, Sheffield, United Kingdom
| | - Masoud Jamei
- Predictive Technologies Division, Certara UK Limited, Sheffield, United Kingdom
| | - Khaled Abduljalil
- Predictive Technologies Division, Certara UK Limited, Sheffield, United Kingdom
| |
Collapse
|
2
|
Cloostermans L, Allegaert K, Smits A, Van Neste M. The Deuterium Oxide Dilution Method to Quantify Human Milk Intake Volume of Infants: A Systematic Review-A Contribution from the ConcePTION Project. Nutrients 2024; 16:4205. [PMID: 39683598 DOI: 10.3390/nu16234205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Global health organizations recommend breastfeeding, but maternal pharmacotherapy can disrupt this due to safety concerns. Physiologically based pharmacokinetic (PBPK) models predict medication transfer through breastfeeding, relying on validated milk intake volume data. However, the literature is mainly focused on different measurement methods, or such intake data have been collected without systematic review. This systematic review therefore aims to gather data on human milk intake volume derived using the (dose-to-the-mother) deuterium oxide dilution method, allowing for comparison with the literature. Additionally, it aims to explore the effects of maternal conditions on milk intake volume. Methods: PubMed, Embase, Web of science, Cochrane library, Scopus and CINAHL were searched for studies on the dilution method and breastfeeding in healthy infants. Risk of bias was assessed using the Newcastle-Ottawa scale (NOS) and the Risk of Bias 2 (RoB2) tool. Data on mean human milk intake volume were extracted and synthesized (mL/day and mL/kg/day) throughout infancy. Results: Sixty studies (34 countries) reported on the milk intake volume of 5502 infants. This intake was best described by logarithmic regression y(mL/kg/day) = 149.4002 - 0.2268 × x - 0.1365 × log(x) (x = postnatal age, days). Maternal conditions showed no significant influence on human milk intake, except for maternal smoking (reduction). Conclusions: This function corresponds with previous research, particularly for infants aged between 1.5 and 12 months. The limited availability of early infancy data underscores the need for additional data for future PBPK modeling to enhance informed healthcare decisions and improve outcomes for mothers and infants.
Collapse
Affiliation(s)
| | - Karel Allegaert
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Anne Smits
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Martje Van Neste
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium
| |
Collapse
|
3
|
Suryavanshi SV, Wang S, Hajducek DM, Hamadeh A, Yeung CHT, Maglalang PD, Ito S, Autmizguine J, Gonzalez D, Edginton AN. Coupling Pre- and Postnatal Infant Exposures with Physiologically Based Pharmacokinetic Modeling to Predict Cumulative Maternal Levetiracetam Exposure During Breastfeeding. Clin Pharmacokinet 2024; 63:1735-1748. [PMID: 39586935 PMCID: PMC11726907 DOI: 10.1007/s40262-024-01447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND AND OBJECTIVE Although breastfeeding ensures optimal infant development and maternal health, mothers taking medications may abandon breastfeeding because of uncertainties regarding toxicity to infants. Current methods in predicting infant risk to maternal medication exposure do not account for breastfeeding-related variability or in utero exposure via the umbilical cord (UC). Previously, our workflow integrated variability in infant anatomy and physiology, breast milk intake volume, and drug concentrations in breast milk using physiologically based pharmacokinetic (PBPK) modeling. The upper area under the curve ratio (UAR) was then calculated to assess infant risk from maternal drug. Herein, we enhanced this workflow by coupling pre- and postnatal exposures to predict the overall levetiracetam exposure in breastfeeding infants. METHODS A published pediatric PBPK model of levetiracetam was used to simulate an infant population (n = 100). Daily infant doses were simulated using a weight-normalized milk intake model to calculate volumes ingested across age groups, alongside literature-derived or simulated milk concentrations across maternal doses to predict infant concentrations. Published UC concentrations were used to develop a cord-coupled neonatal model (CCM), which was integrated with the PBPK and milk intake models and evaluated by comparing observed and simulated infant blood concentrations using a 90% prediction interval (PI). RESULTS UC concentration data from 14 mothers were used to develop the CCM. A total of 16 paired (known milk concentrations) and two unpaired (unknown milk concentrations) individual infant concentrations were identified for evaluating the model along with population values of 64 infants from two age groups (2-4 and 7-31 days). The CCM improved the predictions overall compared with the original workflow, largely due to improvements for the youngest age group evaluated. Overall, 83% (10 of 12) of the individual infant plasma concentrations were successfully captured within the 90% PI for the paired, quantifiable (i.e. above the limit of quantification) evaluation datasets. After administration of a maternal dose of levetiracetam 2000 mg, the calculated UAR ranged from 0.13 to 0.27 for the 95th percentile infants. CONCLUSIONS To our knowledge, this is the first report to combine prenatal levetiracetam exposures from the UC and postnatal exposures from breastfeeding to predict overall infant drug exposure. The results indicate that infant exposure in infants aged 0-7 days may approach therapeutic levels of levetiracetam in the highest-risk infants (i.e. 95th percentile), with a low likelihood of adverse effects based on published clinical studies. This integrated modeling approach provides a more holistic analysis of neonatal exposures. It can be applied in future studies to derive the UAR of drugs administered during breastfeeding to identify infants at risk of potential toxicity.
Collapse
Affiliation(s)
| | - Shirley Wang
- School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| | | | - Abdullah Hamadeh
- School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| | - Cindy H T Yeung
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Patricia D Maglalang
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Autmizguine
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
- Department of Clinical Pharmacology Unit, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
| | - Daniel Gonzalez
- Division of Clinical Pharmacology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | | |
Collapse
|
4
|
Humerickhouse C, Pressly M, Lin Z, Guinn D, Samuels S, Pfuma Fletcher E, Schmidt S. Informing the risk assessment related to lactation and drug exposure: A physiologically based pharmacokinetic lactation model for pregabalin. CPT Pharmacometrics Syst Pharmacol 2024; 13:1953-1966. [PMID: 39460526 DOI: 10.1002/psp4.13266] [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: 05/16/2024] [Revised: 08/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Breastfeeding is important in childhood development, and medications are often necessary for lactating individuals, yet information on the potential risk of infant drug exposure through human milk is limited. Establishing a lactation modeling framework can advance our understanding of this topic and potentiate clinical decision making. We expanded the modeling framework previously developed for sotalol using pregabalin as a second prototypical probe compound with similar absorption, distribution, metabolism, and elimination (ADME) properties. Adult oral models were developed in PK-Sim® and used to build a lactation model in MoBi® to simulate drug transfer into human milk. The adult model was applied to breastfeeding pediatrics (ages 1 to 23 months) and subsequently integrated with the lactation model to simulate infant drug exposure according to age, size, and breastfeeding frequency. Physiologically based pharmacokinetic (PBPK) model simulations captured the data used for verification both in adults and pediatrics. Lactation simulations captured observed milk and plasma data corresponding to doses of 150 mg administered twice daily to lactating individuals, and estimated a relative infant dose (RID) of approximately 7% of the maternal dose. The infant drug exposure simulations showed peak plasma concentrations of 0.44 μg/mL occurring within the first 2 weeks of life, followed by gradual decline with age after week four. The modeling framework performs well for this second prototypical drug and warrants expansion to other drugs for further validation. PBPK modeling and simulation approaches together with clinical lactation data could ultimately help inform infant drug exposure risk assessments to guide clinical decision making.
Collapse
Affiliation(s)
- Cameron Humerickhouse
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Michelle Pressly
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhoumeng Lin
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - Daphne Guinn
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sherbet Samuels
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elimika Pfuma Fletcher
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| |
Collapse
|
5
|
Ojara FW, Kawuma AN, Nakalema S, Kyohairwe I, Nakijoba R, Lamorde M, Pertinez H, Khoo S, Waitt C. Population pharmacokinetic modeling of paired plasma-breast milk lamivudine data for estimation of infant exposure in breastfeeding mother-infant pairs. CPT Pharmacometrics Syst Pharmacol 2024; 13:1978-1989. [PMID: 39508333 PMCID: PMC11578128 DOI: 10.1002/psp4.13274] [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: 05/29/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Around 1.2 million women living with HIV give birth annually, majority of whom will breastfeed their infants while receiving antiretroviral therapy (ART). Lamivudine, a component of first-line ART regimens crosses from maternal plasma to breast milk, with measurable concentrations in some breastfed infants. Wide variability in plasma-to-breast milk transfer has been reported within- or across studies, probably due to differences in sampling framework. This work sought to characterize the milk-to-plasma transfer of lamivudine, quantify inter-patient variability and associated factors, and predict exposure of a breastfed infant. We explored data from an observational pharmacokinetic study that included 35 Ugandan mothers and their infants. Mothers received lamivudine doses of 150 mg twice daily or 300 mg once daily as part of their antiretroviral regimen. Pharmacokinetic sampling was undertaken across two visits approximately 8 weeks apart, providing 248 maternal plasma, 256 breast milk-, and 151 infant blood concentrations, measured across a 24-h sampling interval. A one-compartmental model best described the plasma disposition of lamivudine, with first-order absorption, interindividual variability on clearance and volume of distribution, and a proportional residual error model. A lag in time of plasma-to-breast milk drug accumulation was described using an effect compartment model with a milk-to-plasma ratio of 1.77. An estimated daily infant dose of 179.3 μg/kg (range: 125.8, 282.3) closely predicted the observed infant steady-state concentrations and translated into 3.34% (2.13, 7.20) and 3.35% (1.10, 7.15) of the standard daily maternal dose in visits 1 and 2, respectively. The established modeling framework can be extended to other drugs.
Collapse
Affiliation(s)
- Francis Williams Ojara
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
- Department of Pharmacology and TherapeuticsGulu UniversityGuluUganda
| | - Aida N. Kawuma
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
| | - Shadia Nakalema
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
| | - Isabella Kyohairwe
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
| | - Ritah Nakijoba
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
| | - Mohammed Lamorde
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
| | - Henry Pertinez
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Saye Khoo
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Catriona Waitt
- Infectious Diseases InstituteMakerere University College of Health SciencesKampalaUganda
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
6
|
Monfort A, Cardoso E, Eap CB, Ansermot N, Crettol S, Fischer Fumeaux CJ, Graz MB, Harari MM, Weisskopf E, Gandia P, Allegaert K, Annaert P, Nordeng H, Hascoët JM, Claris O, Epiney M, Ferreira E, Leclair G, Csajka C, Panchaud A, Guidi M. A population pharmacokinetic model for sertraline in women during the perinatal period-A contribution from the ConcePTION project. Br J Clin Pharmacol 2024; 90:2849-2860. [PMID: 39030897 DOI: 10.1111/bcp.16177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/17/2024] [Accepted: 06/17/2024] [Indexed: 07/22/2024] Open
Abstract
AIMS Sertraline is frequently prescribed for mental health conditions in both pregnant and breastfeeding women. According to the limited available data, only small amounts of sertraline are transferred into human milk, yet with a large amount of unexplained interindividual variability. This study aimed to develop a population pharmacokinetic (popPK) model to describe the pharmacokinetics of sertraline during the perinatal period and explain interindividual variability. METHODS Pregnant women treated with sertraline were enrolled in the multicenter prospective cohort SSRI-Breast Milk study. A popPK model for sertraline maternal plasma and breast milk concentrations was developed and allowed estimating the milk-to-plasma ratio (MPR). An additional fetal compartment allowed cord blood concentrations to be described. Several covariates were tested for significance. Ultimately, model-based simulations allowed infant drug exposure through placenta and breast milk under various conditions to be predicted. RESULTS Thirty-eight women treated with sertraline were included in the study and provided 89 maternal plasma, 29 cord blood and 107 breast milk samples. Sertraline clearance was reduced by 42% in CYP2C19 poor metabolizers compared to other phenotypes. Doubling milk fat content increased the MPR by 95%. Simulations suggested a median daily infant dosage of 6.9 μg kg-1 after a 50 mg maternal daily dose, representing 0.95% of the weight-adjusted maternal dose. Median cord blood concentrations could range from 3.29 to 33.23 ng mL-1 after maternal daily doses between 25 and 150 mg. CONCLUSIONS Infant exposure to sertraline, influenced by CYP2C19 phenotype and breast milk fat content, remains low, providing reassurance regarding the use of sertraline during pregnancy and breastfeeding.
Collapse
Affiliation(s)
- Anaëlle Monfort
- CHU Sainte-Justine, Montréal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Evelina Cardoso
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chin B Eap
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva and University of Lausanne, Lausanne and Geneva, Switzerland
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Nicolas Ansermot
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Severine Crettol
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Céline J Fischer Fumeaux
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Myriam Bickle Graz
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Mathilde Morisod Harari
- Division of Child and Adolescent Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Etienne Weisskopf
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Peggy Gandia
- Laboratory of Pharmacokinetics and Toxicology, Purpan Hospital, University Hospital of Toulouse, Toulouse, France
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands
| | - Pieter Annaert
- Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Hedvig Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Jean-Michel Hascoët
- Department of Neonatology, Maternité Régionale, Université de Lorraine, Nancy, France
| | - Olivier Claris
- Department of Neonatology, Hospices Civils de Lyon, Lyon, France
- Claude Bernard University, Lyon, France
| | - Manuella Epiney
- Department of Women, Child and Adolescent, Geneva University Hospital, Geneva, Switzerland
| | - Ema Ferreira
- CHU Sainte-Justine, Montréal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Grégoire Leclair
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Chantal Csajka
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva and University of Lausanne, Lausanne and Geneva, Switzerland
| | - Alice Panchaud
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Monia Guidi
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva and University of Lausanne, Lausanne and Geneva, Switzerland
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
Cole S, Malamatari M, Butler A, Arshad M, Kerwash E. Investigation of a fully mechanistic physiologically based pharmacokinetics model of absorption to support predictions of milk concentrations in breastfeeding women and the exposure of infants: A case study for albendazole. CPT Pharmacometrics Syst Pharmacol 2024; 13:1990-2001. [PMID: 39558864 DOI: 10.1002/psp4.13260] [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: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024] Open
Abstract
Due to limited non-clinical and clinical data, European guidance recommends to discontinue breastfeeding when taking albendazole. The aim of this study was to consider the use of PBPK modeling to support the expected exposure in breastfed infants. A fully mechanistic PBPK approach was used to provide quantitative predictions of albendazole and its main active metabolite, albendazole sulfoxide, concentrations in plasma and breast milk of lactating women. The model predicted the exposure in adults and the large food effect, however, it does not predict all the clinical data for the exposure in children. Milk/plasma ratio predictions were also largely over-predicted for this lipophilic compound, but not for the less lipophilic metabolite. Predictions using the observed ratio and a worse-case exposure based on Cmax predictions, suggest doses to children through milk will be low. However, more clinical data are required before full exposure predictions can be made to breastfed infants.
Collapse
Affiliation(s)
- Susan Cole
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | | | - Andrew Butler
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | | | - Essam Kerwash
- Medicines and Healthcare Products Regulatory Agency, London, UK
| |
Collapse
|
8
|
Ratier A, Casas M, Grazuleviciene R, Slama R, Småstuen Haug L, Thomsen C, Vafeiadi M, Wright J, Zeman FA, Vrijheid M, Brochot C. Estimating the dynamic early life exposure to PFOA and PFOS of the HELIX children: Emerging profiles via prenatal exposure, breastfeeding, and diet. ENVIRONMENT INTERNATIONAL 2024; 186:108621. [PMID: 38593693 DOI: 10.1016/j.envint.2024.108621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/11/2024]
Abstract
In utero and children's exposure to per- and polyfluoroalkyl substances (PFAS) is a major concern in health risk assessment as early life exposures are suspected to induce adverse health effects. Our work aims to estimate children's exposure (from birth to 12 years old) to PFOA and PFOS, using a Physiologically-Based Pharmacokinetic (PBPK) modelling approach. A model for PFAS was updated to simulate the internal PFAS exposures during the in utero life and childhood, and including individual characteristics and exposure scenarios (e.g., duration of breastfeeding, weight at birth, etc.). Our approach was applied to the HELIX cohort, involving 1,239 mother-child pairs with measured PFOA and PFOS plasma concentrations at two sampling times: maternal and child plasma concentrations (6 to 12 y.o). Our model predicted an increase in plasma concentrations during fetal development and childhood until 2 y.o when the maximum concentrations were reached. Higher plasma concentrations of PFOA than PFOS were predicted until 2 y.o, and then PFOS concentrations gradually became higher than PFOA concentrations. From 2 to 8 y.o, mean concentrations decreased from 3.1 to 1.88 µg/L or ng/mL (PFOA) and from 4.77 to 3.56 µg/L (PFOS). The concentration-time profiles vary with the age and were mostly influenced by in utero exposure (on the first 4 months after birth), breastfeeding (from 5 months to 2 (PFOA) or 5 (PFOS) y.o of the children), and food intake (after 3 (PFOA) or 6 (PFOS) y.o of the children). Similar measured biomarker levels can correspond to large differences in the simulated internal exposures, highlighting the importance to investigate the children's exposure over the early life to improve exposure classification. Our approach demonstrates the possibility to simulate individual internal exposures using PBPK models when measured biomarkers are scarce, helping risk assessors in gaining insight into internal exposure during critical windows, such as early life.
Collapse
Affiliation(s)
- Aude Ratier
- INERIS, Unit of Experimental Toxicology and Modelling, Verneuil-en-Halatte, France; PériTox Laboratory, UMR-I 01 INERIS, Université de Picardie Jules Verne, Amiens, France.
| | - Maribel Casas
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | | | - Remy Slama
- Team of Environmental Epidemiology, IAB, Institute for Advanced Biosciences, Inserm, CNRS, CHU-Grenoble-Alpes, University Grenoble-Alpes, CNRS, Grenoble, France
| | - Line Småstuen Haug
- Norwegian Institute of Public Health, Department of Food Safety, Oslo, Norway
| | - Cathrine Thomsen
- Norwegian Institute of Public Health, Department of Food Safety, Oslo, Norway
| | - Marina Vafeiadi
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Florence A Zeman
- INERIS, Unit of Experimental Toxicology and Modelling, Verneuil-en-Halatte, France
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | - Céline Brochot
- INERIS, Unit of Experimental Toxicology and Modelling, Verneuil-en-Halatte, France; Certara UK Ltd, Simcyp Division, Sheffield, UK
| |
Collapse
|
9
|
Yeung CHT, Autmizguine J, Dalvi P, Denoncourt A, Ito S, Katz P, Rahman M, Theoret Y, Edginton AN. Maternal Ezetimibe Concentrations Measured in Breast Milk and Its Use in Breastfeeding Infant Exposure Predictions. Clin Pharmacokinet 2024; 63:317-332. [PMID: 38278872 DOI: 10.1007/s40262-023-01345-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Lactating mothers taking ezetimibe, an antihyperlipidemic agent, may be hesitant to breastfeed despite the known benefit of breastfeeding to both mother and infant. Currently, no data exist on the presence or concentration of ezetimibe and its main active metabolite, ezetimibe-glucuronide (EZE-glucuronide), in human breast milk. METHODS Voluntary breast milk samples containing ezetimibe and EZE-glucuronide were attained from lactating mothers taking ezetimibe as part of their treatment. An assay was developed and validated to measure ezetimibe and EZE-glucuronide concentrations in breast milk. A workflow that utilized a developed and evaluated pediatric physiologically based pharmacokinetic (PBPK) model, the measured concentrations in milk, and weight-normalized breast milk intake volumes was applied to predict infant exposures and determine the upper area under the curve ratio (UAR). RESULTS Fifteen breast milk samples from two maternal-infant pairs were collected. The developed liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay showed an analytical range of 0.039-5.0 ng/mL and 0.39-50.0 ng/mL for ezetimibe and EZE-glucuronide, respectively. The measured concentrations in the breast milk samples were 0.17-1.02 ng/mL and 0.42-2.65 ng/mL of ezetimibe and EZE-glucuronide, respectively. The evaluated pediatric PBPK model demonstrated minimal exposure overlap in adult therapeutic dose and breastfed infant simulated area under the concentration-time curve from time zero to 24 h (AUC24). Calculated UAR across infant age groups ranged from 0.0015 to 0.0026. CONCLUSIONS PBPK model-predicted ezetimibe and EZE-glucuronide exposures and UAR suggest that breastfeeding infants would receive non-therapeutic exposures. Future work should involve a 'mother-infant pair study' to ascertain breastfed infant plasma ezetimibe and EZE-glucuronide concentrations to confirm the findings of this work.
Collapse
Affiliation(s)
- Cindy H T Yeung
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Autmizguine
- Department of Clinical Pharmacology Unit, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Universite de Montreal, Montreal, QC, Canada
| | - Pooja Dalvi
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
| | - Audrey Denoncourt
- Department of Clinical Pharmacology Unit, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Pamela Katz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Mehzabin Rahman
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
| | - Yves Theoret
- Department of Clinical Pharmacology Unit, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, 10 Victoria St S A, Kitchener, ON, N2G 1C5, Canada.
| |
Collapse
|
10
|
Langevin B, Gopalakrishnan M, Kuttamperoor J, Van Den Anker J, Murphy J, Arcaro KF, Daines D, Sylvetsky AC. The MILK study: Investigating intergenerational transmission of low-calorie sweeteners in breast milk. Contemp Clin Trials Commun 2023; 36:101212. [PMID: 37881407 PMCID: PMC10594547 DOI: 10.1016/j.conctc.2023.101212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/27/2023] [Accepted: 10/01/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction Forty-four percent of lactating women in the United States consume beverages containing low calorie sweeteners (LCS), and the presence of LCS in the food supply has continued to increase in recent years. While LCS are approved by the United States Food and Drug Administration (FDA) and are believed to be safe for human consumption, intergenerational LCS transmission and the health impacts of early life LCS exposure are severely understudied. Methods and analysis In a tightly controlled, single site, prospective interventional study, mothers' plasma and breast milk, and infants' plasma will be collected from 40 mother-infant dyads over the course of 72 h, with rich sampling following maternal ingestion of a LCS sweetened beverage containing sucralose and acesulfame potassium (ace-K). Concentration-time data will be used to build maternal and infant pharmacokinetic models for future simulations and analysis. Conclusion This study aims to measure LCS concentrations in breast milk, maternal plasma, and infant plasma, to gain insight into infant exposure and inform recommendations for LCS consumption during breastfeeding.
Collapse
Affiliation(s)
- Brooke Langevin
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, USA
| | - Mathangi Gopalakrishnan
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, USA
| | - Janae Kuttamperoor
- Department of Exercise and Nutrition Sciences, The George Washington University, Washington D.C., USA
| | - John Van Den Anker
- Division of Clinical Pharmacology, Children's National Medical Center, Washington D.C., USA
| | - Jeanne Murphy
- School of Nursing, The George Washington University, Washington D.C., USA
| | - Kathleen F Arcaro
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, USA
| | - Dina Daines
- Department of Obstetrics and Gynecology, The George Washington University, Washington D.C., USA
| | - Allison C Sylvetsky
- Department of Exercise and Nutrition Sciences, The George Washington University, Washington D.C., USA
| |
Collapse
|
11
|
Van Neste M, Bogaerts A, Nauwelaerts N, Macente J, Smits A, Annaert P, Allegaert K. Challenges Related to Acquisition of Physiological Data for Physiologically Based Pharmacokinetic (PBPK) Models in Postpartum, Lactating Women and Breastfed Infants-A Contribution from the ConcePTION Project. Pharmaceutics 2023; 15:2618. [PMID: 38004596 PMCID: PMC10674226 DOI: 10.3390/pharmaceutics15112618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/21/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modelling is a bottom-up approach to predict pharmacokinetics in specific populations based on population-specific and medicine-specific data. Using an illustrative approach, this review aims to highlight the challenges of incorporating physiological data to develop postpartum, lactating women and breastfed infant PBPK models. For instance, most women retain pregnancy weight during the postpartum period, especially after excessive gestational weight gain, while breastfeeding might be associated with lower postpartum weight retention and long-term weight control. Based on a structured search, an equation for human milk intake reported the maximum intake of 153 mL/kg/day in exclusively breastfed infants at 20 days, which correlates with a high risk for medicine reactions at 2-4 weeks in breastfed infants. Furthermore, the changing composition of human milk and its enzymatic activities could affect pharmacokinetics in breastfed infants. Growth in breastfed infants is slower and gastric emptying faster than in formula-fed infants, while a slower maturation of specific metabolizing enzymes in breastfed infants has been described. The currently available PBPK models for these populations lack structured systematic acquisition of population-specific data. Future directions include systematic searches to fully identify physiological data. Following data integration as mathematical equations, this holds the promise to improve postpartum, lactation and infant PBPK models.
Collapse
Affiliation(s)
- Martje Van Neste
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium;
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium; (A.B.); (A.S.)
| | - Annick Bogaerts
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium; (A.B.); (A.S.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Faculty of Health, University of Plymouth, Devon PL4 8AA, UK
| | - Nina Nauwelaerts
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (N.N.); (J.M.); (P.A.)
| | - Julia Macente
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (N.N.); (J.M.); (P.A.)
| | - Anne Smits
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium; (A.B.); (A.S.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (N.N.); (J.M.); (P.A.)
- BioNotus GCV, 2845 Niel, Belgium
| | - Karel Allegaert
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium;
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium; (A.B.); (A.S.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
12
|
Yeung CHT, Bertrand KA, Best BM, Capparelli E, Chambers CD, Hajducek DM, Hamadeh A, Ito S, Momper JD, Edginton AN. Cannabidiol Exposure Through Maternal Marijuana Use: Predictions in Breastfed Infants. Clin Pharmacokinet 2023; 62:1611-1619. [PMID: 37735347 PMCID: PMC10659070 DOI: 10.1007/s40262-023-01307-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND AND OBJECTIVE Knowledge about exposure to cannabidiol (CBD) in breastfed infants can provide an improved understanding of potential risk. The aim was to predict CBD exposure in breastfed infants from mothers taking CBD and CBD-containing products. METHODS Cannabidiol concentrations in milk previously attained from data collected through an existing human milk research biorepository were used to simulate infant doses and identify subgroups. A developed pediatric physiologically based pharmacokinetic model produced virtual breastfed infants administered the simulated CBD doses. Predicted breastfed infant exposures and upper area under the curve ratios were compared to the lowest therapeutic dose for approved indications in children. RESULTS The existing human milk research biorepository contained 200 samples from 181 unique breastfeeding mothers for whom self-reported administration data and CBD concentrations had previously been measured. Samples that were above the lower limit of quantification with only one maternal administration type revealed that administration type, i.e., joint/blunt or edible versus oil or pipe, resulted in significantly different subgroups in terms of milk concentrations. Resulting simulated infant doses (ng/kg) were described by lognormal distributions with geometric means and geometric standard deviations: 0.61 ± 2.41 all concentrations, 0.10 ± 0.37 joint/blunt or edible, and 2.23 ± 8.15 oil or pipe. Doses administered to breastfed infants had exposures magnitudes lower than exposures in children aged 4-11 years administered the lowest therapeutic dose for approved indications, and low upper area under the curve ratios. CONCLUSIONS Based on real-world use, breastfeeding infants are predicted to receive very small exposures of CBD through milk. Studies examining adverse reactions will provide further insight into potential risk.
Collapse
Affiliation(s)
- Cindy H T Yeung
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
| | - Kerri A Bertrand
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Brookie M Best
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Edmund Capparelli
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Christina D Chambers
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Division of Epidemiology, Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, San Diego, CA, USA
| | - Dagmar M Hajducek
- School of Pharmacy, University of Waterloo, 10 Victoria St S A, Kitchener, ON, N2G 1C5, Canada
| | - Abdullah Hamadeh
- School of Pharmacy, University of Waterloo, 10 Victoria St S A, Kitchener, ON, N2G 1C5, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, 10 Victoria St S A, Kitchener, ON, N2G 1C5, Canada.
| |
Collapse
|
13
|
Ponsonby-Thomas E, Salim M, Klein LD, Clulow AJ, Seibt S, Boyd BJ. Evaluating human milk as a drug delivery vehicle for clofazimine to premature infants. J Control Release 2023; 362:257-267. [PMID: 37619865 DOI: 10.1016/j.jconrel.2023.08.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Human milk is proposed as a drug delivery vehicle suitable for use in neonatal patients. Clofazimine, traditionally used for the treatment of leprosy and tuberculosis, is emerging as a treatment for cryptosporidiosis in infants, however its poor aqueous solubility has led to its commercial formulation as a waxy lipid formulation in a capsule, a format that is not suitable for infants. In this study, the evaluation of pasteurised human milk for the delivery of clofazimine was investigated using an in vitro lipolysis model to simulate gastric and intestinal digestion. The total lipid composition of the human milk was characterised alongside the liberated fatty acid species following digestion for comparison to alternative lipid-based delivery systems. Small-angle X-ray scattering was used to measure the presence of crystalline clofazimine during digestion and hence the extent of drug solubilisation. High-performance liquid chromatography was used to quantify the mass of clofazimine solubilised per gram of human milk fat (drug-to-fat ratio) in digested and undigested human milk. The digestion process was essential for the solubilisation of clofazimine, with digested human milk solubilising a sufficient dose of clofazimine for treatment of a premature infant. Human milk solubilised the clofazimine to a greater extent than bovine milk and infant formula during digestion, most likely as a result of differing lipid composition and increased long-chain fatty acid concentrations. These findings show that human milk enhances the solubility of clofazimine as a model drug and may be a suitable drug delivery vehicle for infant populations requiring therapeutic treatment.
Collapse
Affiliation(s)
- Ellie Ponsonby-Thomas
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Malinda Salim
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Laura D Klein
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Strategy and Growth, Australian Red Cross Lifeblood, 17 O'Riordan St, Alexandria, NSW 2015, Australia
| | - Andrew J Clulow
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Australian Synchrotron, ANSTO, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Susi Seibt
- Australian Synchrotron, ANSTO, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Ben J Boyd
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
14
|
Cardoso E, Guidi M, Nauwelaerts N, Nordeng H, Teil M, Allegaert K, Smits A, Gandia P, Edginton A, Ito S, Annaert P, Panchaud A. Safety of medicines during breastfeeding - from case report to modeling : A contribution from the ConcePTION project. Expert Opin Drug Metab Toxicol 2023. [PMID: 37269321 DOI: 10.1080/17425255.2023.2221847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/01/2023] [Indexed: 06/05/2023]
Abstract
INTRODUCTION Despite many research efforts, current data on the safety of medicines during breastfeeding are either fragmented or lacking, resulting in restrictive labeling of most medicines. In the absence of pharmacoepidemiologic safety studies, risk estimation for breastfed infants is mainly derived from pharmacokinetic (PK) information on the medicine. This manuscript provides a description and a comparison of the different methodological approaches that can yield reliable information on medicine transfer into human milk and the resulting infant exposure. AREA COVERED Currently, most information on medicine transfer in human milk relies on case reports or traditional PK studies, which generate data that can hardly be generalized to the population. Some methodological approaches, such as population PK (popPK) and physiologically-based PK (PBPK) modeling, can be used to provide a more complete characterization of infant medicine exposure through human milk and simulate the most extreme situations, while decreasing the burden of sampling in breastfeeding women. EXPERT OPINION PBPK and popPK modeling are promising approaches to fill the gap of knowledge in medicine safety in breastfeeding, as illustrated with our escitalopram example.
Collapse
Affiliation(s)
- Evelina Cardoso
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nina Nauwelaerts
- Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Hedvig Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, PharmaTox Strategic Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Karel Allegaert
- Child and Youth Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy,erasmus MC, Rotterdam, GA, The Netherlands
| | - Anne Smits
- Child and Youth Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Peggy Gandia
- Laboratory of Pharmacokinetics and Toxicology, Purpan Hospital, University Hospital of Toulouse
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, ON, Canada
| | - Pieter Annaert
- Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Alice Panchaud
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| |
Collapse
|
15
|
Pressly MA, Schmidt S, Guinn D, Liu Z, Ceresa C, Samuels S, Madabushi R, Florian J, Fletcher EP. Informing a Comprehensive Risk Assessment of Infant Drug Exposure From Human Milk: Application of a Physiologically Based Pharmacokinetic Lactation Model for Sotalol. J Clin Pharmacol 2023; 63 Suppl 1:S106-S116. [PMID: 37317500 DOI: 10.1002/jcph.2242] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/26/2023] [Indexed: 06/16/2023]
Abstract
Characterization of infant drug exposure through human milk is important and underexplored. Because infant plasma concentrations are not frequently collected in clinical lactation studies, modeling and simulation approaches can integrate physiology, available milk concentrations, and pediatric data to inform exposure in breastfeeding infants. A physiologically based pharmacokinetic model was built for sotalol, a renally eliminated drug, to simulate infant drug exposure from human milk. Intravenous and oral adult models were built, optimized, and scaled to an oral pediatric model for a breastfeeding-relevant age group (<2 years). Model simulations captured the data that were put aside for verification. The resulting pediatric model was applied to predict the impacts of sex, infant body size, breastfeeding frequency, age, and maternal dose (240 and 433 mg) on drug exposure during breastfeeding. Simulations suggest a minimal effect of sex or frequency on total sotalol exposure. Infants in the 90th percentile in height and weight have predicted exposures ≈20% higher than infants of the same age in the 10th percentile due to increased milk intake. The simulated infant exposures increase throughout the first 2 weeks of life and are maintained at the highest concentrations in weeks 2-4, with a consistent decrease observed as infants age. Simulations suggest that breastfeeding infants will have plasma concentrations in the lower range observed in infants administered sotalol. With further validation on additional drugs, physiologically based pharmacokinetic modeling approaches could use lactation data to a greater extent and provide comprehensive information to support decisions regarding medication use during breastfeeding.
Collapse
Affiliation(s)
- Michelle A Pressly
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida, USA
| | - Daphne Guinn
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhichao Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Carrie Ceresa
- Division of Pediatrics and Maternal Health, Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Office of New Drugs, Center for Drug Evaluation and Research, Silver Spring, Maryland, USA
| | - Sherbet Samuels
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rajanikanth Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffry Florian
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elimika Pfuma Fletcher
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
16
|
Nauwelaerts N, Macente J, Deferm N, Bonan RH, Huang MC, Van Neste M, Bibi D, Badee J, Martins FS, Smits A, Allegaert K, Bouillon T, Annaert P. Generic Workflow to Predict Medicine Concentrations in Human Milk Using Physiologically-Based Pharmacokinetic (PBPK) Modelling-A Contribution from the ConcePTION Project. Pharmaceutics 2023; 15:pharmaceutics15051469. [PMID: 37242712 DOI: 10.3390/pharmaceutics15051469] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Women commonly take medication during lactation. Currently, there is little information about the exposure-related safety of maternal medicines for breastfed infants. The aim was to explore the performance of a generic physiologically-based pharmacokinetic (PBPK) model to predict concentrations in human milk for ten physiochemically diverse medicines. First, PBPK models were developed for "non-lactating" adult individuals in PK-Sim/MoBi v9.1 (Open Systems Pharmacology). The PBPK models predicted the area-under-the-curve (AUC) and maximum concentrations (Cmax) in plasma within a two-fold error. Next, the PBPK models were extended to include lactation physiology. Plasma and human milk concentrations were simulated for a three-months postpartum population, and the corresponding AUC-based milk-to-plasma (M/P) ratios and relative infant doses were calculated. The lactation PBPK models resulted in reasonable predictions for eight medicines, while an overprediction of human milk concentrations and M/P ratios (>2-fold) was observed for two medicines. From a safety perspective, none of the models resulted in underpredictions of observed human milk concentrations. The present effort resulted in a generic workflow to predict medicine concentrations in human milk. This generic PBPK model represents an important step towards an evidence-based safety assessment of maternal medication during lactation, applicable in an early drug development stage.
Collapse
Affiliation(s)
- Nina Nauwelaerts
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Julia Macente
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Neel Deferm
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Simcyp Division, Certara UK Ltd., Sheffield S1 2BJ, UK
| | | | - Miao-Chan Huang
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Martje Van Neste
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - David Bibi
- Global Research and Development, Teva Pharmaceutical Industries Ltd., Netanya 42504, Israel
| | - Justine Badee
- Novartis Institutes for BioMedical Research, Novartis, CH-4056 Basel, Switzerland
| | - Frederico S Martins
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Karel Allegaert
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- L-C&Y, KU Leuven Child & Youth Institute, 3000 Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | | | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- BioNotus GCV, 2845 Niel, Belgium
| |
Collapse
|
17
|
Statelova M, Holm R, Fotaki N, Reppas C, Vertzoni M. Usefulness of the Beagle Model in the Evaluation of Paracetamol and Ibuprofen Exposure after Oral Administration to Pediatric Populations: An Exploratory Study. Mol Pharm 2023. [PMID: 37125690 DOI: 10.1021/acs.molpharmaceut.2c00926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The present study aimed to explore the usefulness of beagle dogs in combination with physiologically based pharmacokinetic (PBPK) modeling in the evaluation of drug exposure after oral administration to pediatric populations at an early stage of pharmaceutical product development. An exploratory, single-dose, crossover bioavailability study in six beagles was performed. A paracetamol suspension and an ibuprofen suspension were coadministered in the fasted-state conditions, under reference-meal fed-state conditions, and under infant-formula fed-state conditions. PBPK models developed with GastroPlus v9.7 were used to inform the extrapolation of beagle data to human infants and children. Beagle-based simulation outcomes were compared with published human-adult-based simulations. For paracetamol, fasted-state conditions and reference-meal fed-state conditions in beagles appeared to provide adequate information for the applied scaling approach. Fasted-state and/or reference-meal fed-state conditions in beagles appeared suitable to simulate the performance of ibuprofen suspension in pediatric populations. Contrary to human-adult-based translations, extrapolations based on beagle data collected under infant-formula fed-state conditions appeared less useful for informing simulations of plasma levels in pediatric populations. Beagle data collected under fasted and/or reference-meal fed-state conditions appeared to be useful in the investigation of pediatric product performance of the two investigated highly permeable and highly soluble drugs in the upper small intestine. The suitability of the beagle as a preclinical model to understand pediatric drug product performance under different dosing conditions deserves further evaluation with a broader spectrum of drugs and drug products and comparisons with pediatric in vivo data.
Collapse
Affiliation(s)
- Marina Statelova
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens 157 84, Greece
| | - René Holm
- Drug Product Development, Janssen Research and Development, Johnson & Johnson, Beerse B-2340, Belgium
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense 5230, Denmark
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens 157 84, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens 157 84, Greece
| |
Collapse
|
18
|
Guinn D, Pressly MA, Liu Z, Ceresa C, Samuels S, Wang YM, Madabushi R, Schmidt S, Fletcher EP. Exploring the Knowledge Gaps in Infant Drug Exposure From Human Milk: A Clinical Pharmacology Perspective. J Clin Pharmacol 2023; 63:273-276. [PMID: 36355399 DOI: 10.1002/jcph.2177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Daphne Guinn
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michelle A Pressly
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhichao Liu
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas, USA
| | - Carrie Ceresa
- Division of Pediatrics and Maternal Health, Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Office of New Drugs, Center for Drug Evaluation and Research, Silver Spring, Maryland, USA
| | - Sherbet Samuels
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yow-Ming Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rajanikanth Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida, USA
| | - Elimika Pfuma Fletcher
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
19
|
Yeung CHT, Houle SKD, Anderson PO, Best BM, Dubinsky S, Edginton AN. Addressing maternal medication use during breastfeeding using clinical resources and a novel physiologically based pharmacokinetic model-derived metric: A qualitative study. Front Pediatr 2023; 11:1147566. [PMID: 37077336 PMCID: PMC10106747 DOI: 10.3389/fped.2023.1147566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/17/2023] [Indexed: 04/21/2023] Open
Abstract
Introduction Breastfeeding has major benefits to the maternal-infant dyad and yet healthcare providers have expressed uncertainty about advocating breastfeeding when mothers are taking medications. The tendency for some providers to be more cautious in their advising approach is likely a consequence of limited, unfamiliar, and unreliable existing information on medication use during lactation. A novel risk metric termed the Upper Area Under the Curve Ratio (UAR) was developed to overcome existing resource shortcomings. However, the perception and use of the UAR in practice by providers is not known. The aim of this study was to understand existing resource use and potential UAR use in practice, their advantages and disadvantages, and areas of improvement for the UAR. Methods Healthcare providers mainly practicing in California with experience advising on medication use during lactation were recruited. One-on-one semi-structured interviews that included questions on current practices when advising medication use during breastfeeding, and approaches to a given a scenario with and without information about the UAR were conducted. The Framework Method was applied for data analysis to construct themes and codes. Results Twenty-eight providers representing multiple professions and disciplines were interviewed. Six main themes emerged: (1) Current Practice Approaches, (2) Advantages of Existing Resources, (3) Disadvantages of Existing Resources, (4) Advantages of the UAR, (5) Disadvantages of the UAR, and (6) Strategies to Improve the UAR. Overall, 108 codes were identified that illustrated theme topics ranging from a general lack of metric use to the realities of advising. A workflow describing current practice approaches connected all other themes. Almost all disadvantages of existing resources could be overcome by advantages of other resources and the UAR. Several improvements to the UAR were identified to address its shortcomings. Conclusion Through interviews with providers who use resources to advise on medication use during breastfeeding, an improved understanding of current practice approaches and accessed resources was ascertained. Ultimately, it was found that the UAR would confer multiple benefits over existing resources, and improvements of the UAR were identified. Future work should focus on implementing the suggested recommendations to ensure optimal uptake of the UAR to improve advising practices.
Collapse
Affiliation(s)
- Cindy Hoi Ting Yeung
- School of Pharmacy, Faculty of Science, University of Waterloo, Kitchener, ON, Canada
| | - Sherilyn K. D. Houle
- School of Pharmacy, Faculty of Science, University of Waterloo, Kitchener, ON, Canada
| | - Philip O. Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Brookie M. Best
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Samuel Dubinsky
- School of Pharmacy, Faculty of Science, University of Waterloo, Kitchener, ON, Canada
| | - Andrea N. Edginton
- School of Pharmacy, Faculty of Science, University of Waterloo, Kitchener, ON, Canada
- Correspondence: Andrea N. Edginton
| |
Collapse
|
20
|
Job KM, Dallmann A, Parry S, Saade G, Haas DM, Hughes B, Berens P, Chen JY, Fu C, Humphrey K, Hornik C, Balevic S, Zimmerman K, Watt K. Development of a Generic Physiologically-Based Pharmacokinetic Model for Lactation and Prediction of Maternal and Infant Exposure to Ondansetron via Breast Milk. Clin Pharmacol Ther 2022; 111:1111-1120. [PMID: 35076931 PMCID: PMC10267851 DOI: 10.1002/cpt.2530] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 11/11/2022]
Abstract
Ondansetron is commonly used in breastfeeding mothers to treat nausea and vomiting. There is limited information in humans regarding safety of ondansetron exposure to nursing infants and no adequate study looking at ondansetron pharmacokinetics during lactation. We developed a generic physiologically-based pharmacokinetic lactation model for small molecule drugs and applied this model to predict ondansetron transfer into breast milk and characterize infant exposure. Drug-specific model inputs were parameterized using data from the literature. Population-specific inputs were derived from a previously conducted systematic literature review of anatomic and physiologic changes in postpartum women. Model predictions were evaluated using ondansetron plasma and breast milk concentration data collected prospectively from 78 women in the Commonly Used Drugs During Lactation and infant Exposure (CUDDLE) study. The final model predicted breast milk and plasma exposures following a single 4 mg dose of intravenous ondansetron in 1,000 simulated women who were 2 days postpartum. Model predictions showed good agreement with observed data. Breast milk median prediction error (MPE) was 18.4% and median absolute prediction error (MAPE) was 53.0%. Plasma MPE was 32.5% and MAPE was 43.2%. The model-predicted daily and relative infant doses were 0.005 mg/kg/day and 3.0%, respectively. This model adequately predicted ondansetron passage into breast milk. The calculated low relative infant dose indicates that mothers receiving ondansetron can safely breastfeed. The model building blocks and population database are open-source and can be adapted to other drugs.
Collapse
Affiliation(s)
- Kathleen M. Job
- Division of Clinical Pharmacology, Department of Pediatrics, The University of Utah, Salt Lake City, Utah, USA
| | - André Dallmann
- Pharmacometrics/Modeling & Simulation, Research & Development, Bayer AG, Leverkusen, Germany
| | - Samuel Parry
- Division of Maternal-Fetal Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George Saade
- University of Texas Medical Branch–Galveston, Galveston, Texas, USA
| | - David M. Haas
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brenna Hughes
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Pamela Berens
- McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Jia-Yu Chen
- The Emmes Company, LLC, Rockville, Maryland, USA
| | - Christina Fu
- The Emmes Company, LLC, Rockville, Maryland, USA
| | | | - Christoph Hornik
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Stephen Balevic
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Kanecia Zimmerman
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Kevin Watt
- Division of Clinical Pharmacology, Department of Pediatrics, The University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
21
|
Pierezan MD, Dalla Nora FM, Verruck S. Correlation between As, Cd, Hg, Pb and Sn concentration in human milk and breastfeeding mothers' food consumption: a systematic review and infants' health risk assessment. Crit Rev Food Sci Nutr 2022; 63:8261-8274. [PMID: 35352976 DOI: 10.1080/10408398.2022.2056869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mothers' food and water consumption appear to be determining factors for infants' potentially toxic elements exposure through human milk. Therefore, this systematic review aimed to assess correlations between As, Cd, Hg, Pb and/or Sn concentration in human milk and breastfeeding mothers' food consumption, with later infants' health risk assessment. Estimated Daily Intakes of such elements by infants were also calculated and compared with reference values (RfD or BMDL01). Among 5.663 identified studies, 23 papers remained for analysis. Potentially toxic elements concentration in human milk presented positive correlation with seafood (As, Hg), fresh vegetables (Hg, Cd), cereals (Hg, Cd), cheese, rice, potatoes, private and well-water supply (Pb), wild meat (Pb, Cd) and milk, dairy products, dried fruits and oilseeds (Cd) mothers' consumption. Red meat, caffeinated drinks, and dairy products consume presented negative correlations (Pb). No correlations were found for Sn. Infants from three studies presented high Hg exposition through human milk (> 0. 1 μg/kg PC-1 day-1), as well as observed for Pb in one study (> 0. 5 μg/kg PC-1 day-1). Potentially toxic elements can damage infants' health when they are present in mothers' diet due to the infants' high vulnerability. Therefore, these results raise important issues for public health.Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2022.2056869 .
Collapse
Affiliation(s)
- Milena Dutra Pierezan
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Silvani Verruck
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
22
|
Smith LS, Barkmeier-Kraemer JM. Conceptual Framework Behind the Development of a Level of Confidence Tool: The Pediatric Videofluoroscopic Swallow Study Value Scale. AMERICAN JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2022; 31:689-704. [PMID: 35201847 DOI: 10.1044/2021_ajslp-20-00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
PURPOSE The videofluoroscopic swallow study (VFSS) is the most commonly used instrumental procedure for evaluating swallowing in pediatric populations suspected of having dysphagia. Assessment and interpretation of a VFSS in pediatric populations is frequently challenged by testing-specific factors that can raise concerns regarding the representativeness of swallow events observed during testing compared to daily feeding/swallowing physiology. When VFSS findings do not represent typical swallowing patterns, treatment recommendations can result in suboptimal outcomes. To address this current challenge to pediatric VFSS interpretation and associated treatment recommendations, the pediatric VFSS Value Scale (pVFSS Value Scale) was developed within a tertiary regional pediatric medical center. This clinical focus article summarizes the initial scale development phases and resulting conceptual framework for rating clinical testing factors that influence a clinician's level of confidence regarding pediatric VFSS findings. Future goals for scientific evaluation and clinical utilization of this new rating scale are also reported. CONCLUSIONS The pVFSS Value Scale was developed to assist clinicians with interpretation of pediatric VFSS assessment outcomes and to efficiently communicate factors influencing impressions and treatment recommendations with team members and caregivers. This clinical concept article summarizes potential uses of this tool to inform treatment planning as well as future clinical research to evaluate its psychometrics and clinical utility.
Collapse
Affiliation(s)
- Leann Schow Smith
- Department of Rehabilitation, Primary Children's Hospital, Salt Lake City, UT
| | | |
Collapse
|
23
|
Modi N. The implications of routine milk fortification for the short and long-term health of preterm babies. Semin Fetal Neonatal Med 2021; 26:101216. [PMID: 33714708 DOI: 10.1016/j.siny.2021.101216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fortification refers to the practice of enriching human milk feeds for very preterm babies with macronutrients, minerals and vitamins. Though standard of care in some parts of the world, adoption of fortification is not universal. Fortification entered into use on the assumption that human milk macronutrient content, principally protein, is insufficient to support the growth and development of very preterm babies. However, because of the substantial variability in human milk composition, routine fortification risks exposing some babies to very high protein intakes, which may be dangerous. Some clinicians fear fortification with cow-milk derived products will increase the risk of necrotising enterocolitis, leading them to favour commercial fortifiers made from pooled human milk over cow milk based products, a practice that has additional ethical implications. Randomised controlled trials of multi-nutrient fortification to-date are inadequate. No trial has had power to detect important functional effects; the majority are methodologically weak and focus primarily upon short-term growth. Evidence to guide practice is inadequate. There is an urgent need for collaboration to conduct high-quality research to end these long-standing uncertainties.
Collapse
Affiliation(s)
- Neena Modi
- Imperial College London, Chelsea and Westminster Hospital campus, 369 Fulham Road, London, SW10 9NH, UK.
| |
Collapse
|
24
|
Hung SC, Lin TI, Suen JL, Liu HK, Wu PL, Wu CY, Yang YCSH, Yang SN, Yang YN. Phthalate Exposure Pattern in Breast Milk within a Six-Month Postpartum Time in Southern Taiwan. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18115726. [PMID: 34073581 PMCID: PMC8198263 DOI: 10.3390/ijerph18115726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 11/22/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a common plasticizer, has been detected in breast milk in many countries; however, whether phthalate metabolite concentration and the detection rate in breast milk change postpartum is still unknown. We measured phthalate metabolite concentrations in breast milk in the first 6 months postpartum in women enrolled in the E-Da hospital from January to July 2017. A total of 56 breastfeeding mothers and 66 samples were included in this study. We analyzed the samples’ concentration of eight phthalate metabolites using liquid chromatography mass spectrometry. The concentration of mono-2-ethylhexyl phthalate (MEHP) was significantly higher in the first month, and then decreased over time. The detection rate of ono-isobutyl phthalate (MiBP) and mono-n-butyl phthalate (MBP) was low in the first month and then increased over time. Compared with a previous study published in 2011, the levels of MEHP and MiBP in breast milk were much lower in the present study, suggesting an increased awareness of the health risks of phthalate exposure after a food scandal occurred in Taiwan. This study provides information for evaluating newborns’ exposure to different kinds of phthalate through human milk in the postpartum period.
Collapse
Affiliation(s)
- Shen-Che Hung
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Ting-I Lin
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Hsien-Kuan Liu
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Pei-Ling Wu
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Chien-Yi Wu
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan;
| | - San-Nan Yang
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Yung-Ning Yang
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan; (S.-C.H.); (T.-I.L.); (H.-K.L.); (P.-L.W.); (C.-Y.W.); (S.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Correspondence:
| |
Collapse
|
25
|
Yeung CHT, Ito S, Autmizguine J, Edginton AN. Incorporating Breastfeeding-Related Variability with Physiologically Based Pharmacokinetic Modeling to Predict Infant Exposure to Maternal Medication Through Breast Milk: a Workflow Applied to Lamotrigine. AAPS JOURNAL 2021; 23:70. [PMID: 34002327 DOI: 10.1208/s12248-021-00599-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022]
Abstract
Current methods to assess risk in infants exposed to maternal medication through breast milk do not specifically account for infants most vulnerable to high drug exposure. A workflow applied to lamotrigine incorporated variability in infant anatomy and physiology, milk intake volume, and milk concentration to predict infant exposure. An adult physiologically based pharmacokinetic model of lamotrigine was developed and evaluated. The model was scaled to account for growth and maturation of a virtual infant population (n=100). Daily infant doses were simulated using milk intake volume and concentration models described by a nonlinear equation of weight-normalized intake across infant age and a linear function on the relationship of observed milk concentrations and maternal doses, respectively. Average infant plasma concentration at steady state was obtained through simulation. Models were evaluated by comparing observed to simulated infant plasma concentrations from breastfeeding infants based on a 90% prediction interval (PI). Upper AUC ratio (UAR) was defined as a novel risk metric. Twenty-five paired (milk concentrations measured) and 18 unpaired (milk concentrations unknown) infant plasma samples were retrieved from the literature. Forty-four percent and 11% of the paired and unpaired infant plasma concentrations were outside of the 90% PI, respectively. Over all ages (0-7 months), unpaired predictions captured more observed infant plasma concentrations within 90% PI than paired. UAR was 0.18-0.44 when mothers received 200 mg lamotrigine, suggesting that infants can receive 18-44% of the exposure per dose as compared to adults. UARs determined for further medications could reveal trends to better classify at-risk mother-infant pairs.
Collapse
Affiliation(s)
- Cindy H T Yeung
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Julie Autmizguine
- Department of Pediatrics & Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada.,Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
26
|
Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur J Pharm Sci 2021; 162:105812. [PMID: 33753215 DOI: 10.1016/j.ejps.2021.105812] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
The absorption of oral drugs is frequently plagued by significant variability with potentially serious therapeutic consequences. The source of variability can be traced back to interindividual variability in physiology, differences in special populations (age- and disease-dependent), drug and formulation properties, or food-drug interactions. Clinical evidence for the impact of some of these factors on drug pharmacokinetic variability is mounting: e.g. gastric pH and emptying time, small intestinal fluid properties, differences in pediatrics and the elderly, and surgical changes in gastrointestinal anatomy. However, the link of colonic factors variability (transit time, fluid composition, microbiome), sex differences (male vs. female) and gut-related diseases (chronic constipation, anorexia and cachexia) to drug absorption variability has not been firmly established yet. At the same time, a way to decrease oral drug pharmacokinetic variability is provided by the pharmaceutical industry: clinical evidence suggests that formulation approaches employed during drug development can decrease the variability in oral exposure. This review outlines the main drivers of oral drug exposure variability and potential approaches to overcome them, while highlighting existing knowledge gaps and guiding future studies in this area.
Collapse
|
27
|
Nauwelaerts N, Deferm N, Smits A, Bernardini C, Lammens B, Gandia P, Panchaud A, Nordeng H, Bacci ML, Forni M, Ventrella D, Van Calsteren K, DeLise A, Huys I, Bouisset-Leonard M, Allegaert K, Annaert P. A comprehensive review on non-clinical methods to study transfer of medication into breast milk - A contribution from the ConcePTION project. Biomed Pharmacother 2021; 136:111038. [PMID: 33526310 DOI: 10.1016/j.biopha.2020.111038] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022] Open
Abstract
Breastfeeding plays a major role in the health and wellbeing of mother and infant. However, information on the safety of maternal medication during breastfeeding is lacking for most medications. This leads to discontinuation of either breastfeeding or maternal therapy, although many medications are likely to be safe. Since human lactation studies are costly and challenging, validated non-clinical methods would offer an attractive alternative. This review gives an extensive overview of the non-clinical methods (in vitro, in vivo and in silico) to study the transfer of maternal medication into the human breast milk, and subsequent neonatal systemic exposure. Several in vitro models are available, but model characterization, including quantitative medication transport data across the in vitro blood-milk barrier, remains rather limited. Furthermore, animal in vivo models have been used successfully in the past. However, these models don't always mimic human physiology due to species-specific differences. Several efforts have been made to predict medication transfer into the milk based on physicochemical characteristics. However, the role of transporter proteins and several physiological factors (e.g., variable milk lipid content) are not accounted for by these methods. Physiologically-based pharmacokinetic (PBPK) modelling offers a mechanism-oriented strategy with bio-relevance. Recently, lactation PBPK models have been reported for some medications, showing at least the feasibility and value of PBPK modelling to predict transfer of medication into the human milk. However, reliable data as input for PBPK models is often missing. The iterative development of in vitro, animal in vivo and PBPK modelling methods seems to be a promising approach. Human in vitro models will deliver essential data on the transepithelial transport of medication, whereas the combination of animal in vitro and in vivo methods will deliver information to establish accurate in vitro/in vivo extrapolation (IVIVE) algorithms and mechanistic insights. Such a non-clinical platform will be developed and thoroughly evaluated by the Innovative Medicines Initiative ConcePTION.
Collapse
Affiliation(s)
- Nina Nauwelaerts
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| | - Neel Deferm
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| | - Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, UZ Leuven, Neonatology, Herestraat 49, 3000, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Belgium.
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | | | - Peggy Gandia
- Laboratoire de Pharmacocinétique et Toxicologie, Centre Hospitalier Universitaire de Toulouse, France.
| | - Alice Panchaud
- Service of Pharmacy Service, Lausanne University Hospital and University of Lausanne, Switzerland; Institute of Primary Health Care (BIHAM), University of Bern, Switzerland
| | - Hedvig Nordeng
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, University of Oslo, PB. 1068 Blindern, 0316, Oslo, Norway.
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | - Domenico Ventrella
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, BO, Italy.
| | | | - Anthony DeLise
- Novartis Pharmaceuticals Corporation, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, NJ, 07936, USA.
| | - Isabelle Huys
- KU Leuven, Department of Clinical Pharmacology and Pharmacotherapy, ON II Herestraat 49 - bus, 521 3000, Leuven, Belgium.
| | - Michele Bouisset-Leonard
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Werk Klybeck Postfach, Basel, CH-4002, Switzerland.
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Belgium; KU Leuven, Department of Clinical Pharmacology and Pharmacotherapy, ON II Herestraat 49 - bus, 521 3000, Leuven, Belgium; Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands.
| | - Pieter Annaert
- KU Leuven Drug Delivery and Disposition Lab, Department of Pharmaceutical and Pharmacological Sciences, O&N II Herestraat, 49 3000, Leuven, Belgium.
| |
Collapse
|
28
|
Integration of physiological changes during the postpartum period into a PBPK framework and prediction of amoxicillin disposition before and shortly after delivery. J Pharmacokinet Pharmacodyn 2020; 47:341-359. [DOI: 10.1007/s10928-020-09706-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022]
|
29
|
Acute Maternal Fasting or Fluid Abstention Does Not Significantly Affect the Macronutrient Composition of Human Milk: Clinical and Clinical Research Relevance. CHILDREN-BASEL 2020; 7:children7060060. [PMID: 32531924 PMCID: PMC7346150 DOI: 10.3390/children7060060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/28/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
There are guidelines on lactation following maternal analgo-sedative exposure, but these do not consider the effect of maternal fasting or fluid abstention on human milk macronutrient composition. We therefore performed a structured search (PubMed) on ‘human milk composition’ and screened title, abstract and full paper on ‘fasting’ or ‘abstention’ and ‘macronutrient composition’ (lactose, protein, fat, solids, triglycerides, cholesterol). This resulted in six papers and one abstract related to religious fasting (n = 129 women) and observational studies in lactating women (n = 23, healthy volunteers, fasting). These data reflect two different ‘fasting’ patterns: an acute (18–25 h) model in 71 (healthy volunteers, Yom Kippur/Ninth of Av) women and a chronic repetitive fasting (Ramadan) model in 81 women. Changes were most related to electrolytes and were moderate and mainly in the chronic repetitive fasting model, with no clinical significant changes in macronutrients during acute fasting. We therefore conclude that neither short-term fasting nor fluid abstention (18–25 h) affect human milk macronutrient composition, so that women can be reassured when this topic was raised during consulting. Besides the nutritional relevance, this also matters, as clinical research samples—especially estimating analgo-sedative exposure by lactation—are commonly collected after maternal procedural sedation and maternal fasting. Based on these results, it is reasonable to assume stable human milk composition when such data are used in physiology-based pharmacokinetic (PBPK) models.
Collapse
|
30
|
Dos Santos e Silva RK, Sany de Moura MS, Carvalho MDL, Braz ZR, Moura NDS, Mendes AN, Rodrigues MP. Desejo de doar leite: relação com características maternas. AVANCES EN ENFERMERÍA 2020. [DOI: 10.15446/av.enferm.v38n2.82838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objetivo: relacionar o desejo de doar leite materno aos aspectos sociodemográficos, clínicos e obstétricos.Materiais e métodos: estudo transversal e analítico com abordagem quantitativa, realizado com 226 mulheres durante o puerpério imediato, em um Hospital de Referência do interior do Piauí, Brasil, entre dezembro de 2016 e junho de 2018. Na estatística analítica, realizaram-se os testes de Kolmogorov-Smirnov, Qui-quadrado de Pearson, Razão de verossimilhança e o teste t Student.Resultados: a caracterização sociodemográfica revelou que as participantes do estudo eram predominantemente casadas (n = 165; 73 %); pardas (n = 145; 64,1 %) e 79 (34,9 %) praticavam atividade física. 78,3 % (n = 177) das entrevistadas desejavam ser doadoras de leite materno, o que indica relação significativa com os anos de estudo (p = 0,038) e com as que receberam orientações sobre o assunto durante o pré-natal (p = 0,028).Conclusões: o desejo de doar leite 3 materno esteve significativamente relacionado às variáveis de maior escolaridade da mulher e às orientações recebidas no pré-natal sobre o assunto, demonstrando a relevância da educação em saúde dentro do contexto.
Collapse
|
31
|
Yeung CHT, Fong S, Malik PRV, Edginton AN. Quantifying breast milk intake by term and preterm infants for input into paediatric physiologically based pharmacokinetic models. MATERNAL AND CHILD NUTRITION 2020; 16:e12938. [PMID: 31965755 PMCID: PMC7083422 DOI: 10.1111/mcn.12938] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/13/2019] [Accepted: 12/15/2019] [Indexed: 12/18/2022]
Abstract
Despite the many benefits of breast milk, mothers taking medication are often uncertain about the risks of drug exposure to their infants and decide not to breastfeed. Physiologically based pharmacokinetic models can contribute to drug‐in‐milk safety assessments by predicting the infant exposure and subsequently, risk for toxic effects that would result from continuous breastfeeding. This review aimed to quantify breast milk intake feeding parameters in term and preterm infants using literature data for input into paediatric physiologically based pharmacokinetic models designed for drug‐in‐milk risk assessment. Ovid MEDLINE and Embase were searched up to July 2, 2019. Key study reference lists and grey literature were reviewed. Title, abstract and full text were screened in nonduplicate. Daily weight‐normalized human milk intake (WHMI) and feeding frequency by age were extracted. The review process retrieved 52 studies. A nonlinear regression equation was constructed to describe the WHMI of exclusively breastfed term infants from birth to 1 year of age. In all cases, preterm infants fed with similar feeding parameters to term infants on a weight‐normalized basis. Maximum WHMI was 152.6 ml/kg/day at 19.7 days, and weighted mean feeding frequency was 7.7 feeds/day. Existing methods for approximating breast milk intake were refined by using a comprehensive set of literature data to describe WHMI and feeding frequency. Milk feeding parameters were quantified for preterm infants, a vulnerable population at risk for high drug exposure and toxic effects. A high‐risk period of exposure at 2–4 weeks of age was identified and can inform future drug‐in‐milk risk assessments.
Collapse
Affiliation(s)
- Cindy H T Yeung
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Simon Fong
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Paul R V Malik
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| |
Collapse
|