PBPK Models in Postpartum Women and Breastfed Infants: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Physiologically based pharmacokinetic (PBPK) modelling is a bottom-up approach to predict pharmacokinetics in specific populations based on medicine-specific and population-specific data, such as physiology - A contribution from the ConcePTION Project. 

  • pharmacokinetics
  • physiologically based pharmacokinetic (PBPK) modelling and simulation
  • pregnancy
  • lactation
  • breastfeeding
  • infant
  • pharmacotherapy

1. Introduction

The European Medicines Agency defines a physiologically based pharmacokinetic (PBPK) model as “a mathematical model that simulates the concentration of a drug over time in tissue(s) and blood, by taking into account the rate of the drug’s absorption into the body, distribution in tissues, or metabolism and excretion (ADME) on the basis of interplay between physiological, physicochemical and biochemical determinants” [1]. In other words, PBPK modelling allows a bottom-up prediction of pharmacokinetics based on the integration of population-specific data regarding physiology with medicine-specific data regarding disposition.
PBPK modelling has already been embraced by the pharmaceutical industry and regulatory agencies, e.g., to predict drug–drug or drug–food interactions, or to support an initial dose finding in first-in-human and paediatric trials [1][2][3]. In addition, properly applied PBPK models have the potential to reduce the number of subjects in early phase clinical trials and the time and resources required in paediatric medicine development [4].
Despite the evolutions in physiologically based modelling and expanding insights into pharmacokinetics, there is still limited, poorly standardized and scattered knowledge regarding the physiology of special populations. These special population models require integration of underlying (patho)physiological data, collected through systematic searches and observed data, and subsequently converted to mathematical equations to develop and evaluate these tools [2]. Using structured integration, the predictive performance of PBPK modelling in special adult populations (like hepatic dysfunction, chronic kidney disease (CKD) or pregnancy and lactation) improved [2][5][6]. Along the same line, neonatal (and preterm) PBPK models were developed, but with still insufficient reflections on the specific physiology of breastfed versus formula-fed infants [7][8]. To fully operationalize the potentials of postpartum, lactation and breastfed infant PBPK models and generate more confidence in their applications, further collection of physiological data based on systematic review approaches specific in these populations is warranted [2][9].

2. Postpartum Maternal Weight Retention

Weight and body composition are crucial covariates of pharmacokinetics. Maternal weight changes during pregnancy and postpartum are therefore relevant to be incorporated as specific equations in PBPK models of these populations.
Around 30% of gestational weight gain (GWG) can be accounted for by the growth of the foetus, placenta and amniotic fluid. This suggests that maternal factors, such as the increased circulating volume and fat mass, cause the majority of weight gain during pregnancy [10]. Guidelines for ranges of GWG were published by the Institute of Medicine (IOM), based on pre-pregnancy BMI categories, to prevent postpartum weight retention (PPWR) and the consequences in mothers. Underweight women (BMI < 18.5 kg/m2) are recommended to gain 12.5 to 18 kg during their pregnancy, normal weight women (BMI 18.5–24.9 kg/m2) 11.5 to 16 kg, overweight women (BMI 25.0–29.9 kg/m2) 7 to 11.5 kg and obese women (BMI ≥ 30.0 kg/m2) 5 to 9 kg [11]. A technical advisory group has recently been established to develop new GWG recommendations [12].
Still, pregnancy is a risk factor for increasing body weight and developing obesity in women of a reproductive age because of their GWG and PPWR [13]. Furthermore, obesity has become a worldwide issue as approximately 50% of women of reproductive age are overweight or obese [10]. This worldwide increasing pre-pregnancy weight further adds to the postpartum maternal weight patterns and its variability and effect on pharmacokinetics.
Multiple studies described excessive GWG in approximately a third of pregnant women and an overall increase in GWG in several industrialized countries. A systematic review on excessive GWG reported GWG of 19 kg in normal weight women (3 kg above the IOM recommendations), 16 kg in women with overweight (4.5 kg above) and 14 kg in women with obesity (5 kg above) [13]. Excessive GWG can lead to multiple maternal and neonatal complications, such as an increased risk of caesarean section and foetal macrosomia and an increased risk for high PPWR [10][14][15].
Failure to lose this GWG is a risk factor for obesity and associated morbidities later in life, such as type II diabetes mellitus and cardiovascular diseases. Unfortunately, the majority of women fail to lose their pregnancy weight with a mean weight retention of 4.5 kg 12 months postpartum [13]. Furthermore, and next to GWG, increases in body fat percentage and waist circumference have been described from 12 weeks of gestation (29 ± 0.5% and 78 ± 1 cm, respectively) to 6 weeks postpartum (31 ± 0.5% and 81 ± 1 cm, respectively), with a decrease to 6 months after childbirth (30 ± 1% and 80 ± 1 cm, respectively) [16]. These postpartum maternal weight patterns, their variability and covariates involved are also relevant to inform postpartum maternal PBPK models.
In women with excessive GWG, a faster weight loss during the first 6 weeks postpartum has been described compared to women with GWG within and below the guidelines; although, the weight retention thereafter was higher compared to women with GWG within the guidelines (5 kg vs. 2 kg) [10]. Similarly, overweight and obese women showed a faster weight loss from 2 weeks postpartum compared to normal weight women. At 6 weeks postpartum, women with obesity showed a lower mean weight retention compared to normal weight women (2 kg vs. 6 kg) but had a higher fat percentage (39% vs. 31%) and a higher waist circumference (98 cm vs. 82 cm) [10][13]. From 6 weeks to 6 months after delivery, postpartum weight loss was slower in overweight women compared to normal weight women with a stagnation of PPWR in overweight women after 6 months. A slight increase in PPWR was seen in women with obesity between 6 weeks and 6 months postpartum, with a small monthly increase thereafter [13]. In terms of long-term maternal weight retention, excessive GWG in women leads to PPWR of 3.1 kg and 4.7 kg after 3 years and 15 years postpartum, respectively [14]. Excessive GWG and PPWR may result in a cycle of BMI increases that can be repeated in future pregnancies [15]. Around 50% of women with excessive GWG have a higher BMI during their next pregnancy compared to their pre-pregnancy BMI of the previous pregnancy. This PPWR means that they have an increased risk of pregnancy and birth-related complications in the next pregnancy, including pregnancy induced hypertension and gestational diabetes [10].
The incidence of gestational diabetes mellitus (GDM) is rising, together with the worldwide increase in obesity [17]. Maternal and foetal outcomes are influenced by GDM, with an increased risk of complications during pregnancy, perinatal period and postpartum, such as pre-eclampsia, preterm delivery and foetal macrosomia. In 70–85% of patients diagnosed with GDM, the management is sufficient with physical activity, lifestyle modifications and GWG management. However, up to 15–30% of patients require pharmacotherapy, such as insulin and oral hypoglycaemic agents. In addition, women with GDM have a 10-fold risk of developing type II diabetes mellitus in the future, have a higher risk of being overweight and developing metabolic syndrome. Therefore, mothers are recommended to exercise to prevent GDM before the pregnancy or early in the pregnancy and women who experienced GDM should be monitored for the development of type II diabetes mellitus [17][18].
Highly relevant to lactation-related PBPK model construction, PPWR is also inversely associated with breastfeeding, since it requires more energy (roughly 500 kcal/day) and possible fat mobilization [19][20]. Weight loss after 3 and 6 months postpartum in exclusively breastfeeding women was 0.7 and 0.5 kg/month, respectively, which resulted in losing more than 85% of the gestational weight gain [21]. Longitudinal data found that breastfeeding, and more specifically exclusive breastfeeding, is connected to a lower weight gain for 8 to 10 years postpartum and might be associated with long-term weight control [19]. It is reported that the initiation of exclusive breastfeeding and duration of breastfeeding, in particular 3 to 6 months, is required to show an influence on maternal weight and that little influence is seen when mothers breastfeed for more than 6 months. However, the evidence on the effects of lactation on maternal weight is inconsistent and conflicting [10][13][20]. In addition, as many women are questioning the energy restriction during breastfeeding for fear of decreased milk production, women often retain or increase their postpartum weight while breastfeeding [20].
To better explore the variability in exposure in different postpartum subpopulations, systematic searches on the specific characteristics of these populations to develop accurate mathematical functions should be used as an approach. This is of potential relevance for both maternal, as well as lactation-related exposure in infants.

3. Human Milk Intake and Composition

Daily human milk volume and composition evolve during the lactation period as well as during a single feeding moment.
Many approaches have been described to measure human milk intake in infants and to subsequently calculate infant medicine exposure through breastfeeding. Test-weighing and deuterium oxide dose-to-the-mother technique are the most often used to measure milk intake. The first method is the conventional technique to estimate milk intake by weighing the infant before and after each feeding. This procedure is used during the first weeks of life and is easy and direct with minimal interference with the lactation process, but it is time-consuming since the observer has to be present for up to 48 h. Studies found that this method is accurate but imprecise, as it underestimates or overestimates the human milk intake of a feeding by up to 15 mL in 95% of cases, which means that this is an unreliable method to be implemented in clinical practices. An alternative method for human milk intake estimation in clinical practice could be test-weighing after every feed over a 24 h period; however, these findings were not validated [22][23].
The deuterium oxide dose-to-the mother technique, on the other hand, is a non-invasive method that uses a stable, non-radioactive isotope to assess the human milk intake. The deuterium oxide is orally consumed by the mother, whereafter the disappearance in the mother’s and the appearance in the child’s saliva are monitored. The consumed amount of deuterium (0.1%) in these studies is far under the toxicity threshold (15%) and is stated to be non-toxic for mother and infant, since no adverse effects in mother or infant have been described. One of the benefits of this method is the possibility to measure the human milk intake in not only exclusively breastfed (EBF) but also partially breastfed (PBF) infants. However, this technique is fairly slow and measures the volume of human milk intake over a period of 14 days [24][25][26].
Yeung et al. recently quantified the daily human milk intake from birth to 1 year of life for input in term and preterm PBPK models [27]. Following a comprehensive search on terms related to premature and term infants, breastfeeding and volume, a regression equation for the weight-normalized human milk intake (WHMI), consistent with the observed data and similar to already existing linear regression models, has been described in term infants up to 6 months of postnatal age:
W H M I   mL / kg / day = 160.39 × 0.232 0.232 0.00252 × e 0.00252 t e 0.232 t ,
where t = days. The maximum WHMI for infants, exclusively breastfed up to 6 months of age, was set at 153 mL/kg/day at day 20, with a weighted mean feeding frequency of 7.7 feeds/day. This equation is similar to the human milk intake of breastfed preterm infants [27].
The peak volume of intake during the first month indicates that the greatest risk of medicine exposure in breastfed infants occurs at 2–4 weeks. The lower medicine metabolism and excretion in infants, together with the high medicine dose relative to their weight explains the risk for higher medicine exposure and toxic effects during the first months, especially in the preterm population [24][28]. Furthermore, infants are more likely breastfed and mainly exclusively breastfed during the first months of life, which might result in higher medicine exposure in younger infants. These findings are consistent with a review on adverse medicine reactions in breastfed infants, as almost 80% of cases of adverse reactions in infants were observed in the first 2 months, with approximately two-thirds of the cases in the first month of life [29]. Moreover, early postpartum is a specific lactation interval of interest as it covers both the shift from colostrum to mature milk, as well as a fast progressive increase in the daily milk intake (mL/kg/day), an important determinant of compound dosing through human milk (mg/mL × mL).
The daily frequency of feeds stays rather stable with the age of the EBF term infant, with no difference between boys and girls [30]. Oras et al. described that the feeding frequency in EBF preterm infants is higher as described in the term population; however, large variations are seen in the frequency of breastfeeding sessions [30][31]. Strong differences in breastfeeding frequency can be explained by biological and cultural perspectives, considering fewer feeding frequencies are described in studies in urban communities or developed countries, whereas higher frequencies are reported in studies in rural communities or in developing countries [27][31]. However, it is suggested that the feeding frequency has a minimal impact on medicine exposure in breastfed infants [32].
Furthermore, fresh human milk holds enzymatic activities, and these enzymes may also affect the medicine absorption or enterohepatic recirculation patterns [33][34]. Breastfed infants have an increased enterohepatic recirculation and a decreased bilirubin clearance, as human milk has considerably more ß-glucuronidase activity than formula milk (419 units/mL vs. 6 units/mL). This difference in ß-glucuronidase activity is more pronounced in the first days postpartum, with a decrease of almost 70% in the third week [33]. Similarly, amylase levels are highest in the initial phase but remain constant in mature human milk up to 27 months postpartum [35]. Conversely, the levels of bile salt-stimulated lipase (BSSL) activity in human milk increases from colostrum to the third week of lactation (30.8 ± 1.09 U/mL vs. 42.6 ± 1.03 U/mL, respectively) and decreases after 6 weeks to levels similar to those in colostrum at 12 weeks (30.8 ± 1.23 U/mL) [36].
Finally, pH values, protein and fat content change in human milk over time, which can have an effect on medicine kinetics in milk [37][38]. The pH values of milk are decreasing from colostrum to 14 days postpartum (pH 7.45 vs. pH 7.04, respectively) but are rising again after 90 days with pH values of 7.40 at 300 days postpartum [38]. Human milk comprises more than 400 different proteins with high levels of whey protein in colostrum. The curve of protein content in human milk decreases during lactation and flattens after 7 months. Overall, mothers who delivered at full term express lower protein levels in their human milk than mothers who delivered preterm [37][39]. Human milk fat concentration is highly variable and increases with a longer breastfeeding duration and increases during each breastfeeding moment [40]. The initial milk of a feed, the foremilk, can contain less than half of the milk fat concentration of the hindmilk, the last milk of the feed [37]. Moreover, a diurnal variation has been described with a higher milk fat concentration during the evening than the morning (43 ± 9 g/L vs. 37 ± 10 g/L, respectively) [30].
Systematic searches to develop specific mathematical functions are needed to adequately explore the variability in human milk volume and composition.

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics15112618

References

  1. European Medicines Agency: Committee for Medicinal Products for Human Use (CHMP). Guideline on the Reporting of Physiologically Based Pharmacokinetic (PBPK) Modelling and Simulation; European Medicines Agency: London, UK, 2018.
  2. Smits, A.; de Cock, P.; Vermeulen, A.; Allegaert, K. Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulation in Neonatal Drug Development: How Clinicians Can Contribute. Expert. Opin. Drug Metab. Toxicol. 2019, 15, 25–34.
  3. FDA. Physiologically Based Pharmacokinetic Analyses—Format and Content: Guidance for Industry; FDA: Rockville, MD, USA, 2018; pp. 1–6.
  4. Leong, R.; Vieira, M.L.T.; Zhao, P.; Mulugeta, Y.; Lee, C.S.; Huang, S.M.; Burckart, G.J. Regulatory Experience with Physiologically Based Pharmacokinetic Modeling for Pediatric Drug Trials. Clin. Pharmacol. Ther. 2012, 91, 926–931.
  5. Murphy, W.A.; Adiwidjaja, J.; Sjöstedt, N.; Yang, K.; Beaudoin, J.J.; Spires, J.; Siler, S.Q.; Neuhoff, S.; Brouwer, K.L.R. Considerations for Physiologically Based Modeling in Liver Disease: From Nonalcoholic Fatty Liver (NAFL) to Nonalcoholic Steatohepatitis (NASH). Clin. Pharmacol. Ther. 2022, 113, 275–297.
  6. Huang, W.; Isoherranen, N. Novel Mechanistic PBPK Model to Predict Renal Clearance in Varying Stages of CKD by Incorporating Tubular Adaptation and Dynamic Passive Reabsorption. CPT Pharmacomet. Syst. Pharmacol. 2020, 9, 571–583.
  7. Claassen, K.; Thelen, K.; Coboeken, K.; Gaub, T.; Lippert, J.; Allegaert, K.; Willmann, S. Development of a Physiologically-Based Pharmacokinetic Model for Preterm Neonates: Evaluation with In Vivo Data. Curr. Pharm. Des. 2015, 21, 5688–5698.
  8. Edginton, A.N.; Schmitt, W.; Willmann, S. Development and Evaluation of a Generic Physiologically Based Pharmacokinetic Model for Children. Clin. Pharmacokinet. 2006, 45, 1013–1034.
  9. Abduljalil, K.; Pansari, A.; Ning, J.; Jamei, M. Prediction of Drug Concentrations in Milk during Breastfeeding, Integrating Predictive Algorithms within a Physiologically-Based Pharmacokinetic Model. CPT Pharmacomet. Syst. Pharmacol. 2021, 10, 878–889.
  10. Bogaerts, A.; De Baetselier, E.; Ameye, L.; Dilles, T.; Van Rompaey, B.; Devlieger, R. Postpartum Weight Trajectories in Overweight and Lean Women. Midwifery 2017, 49, 134–141.
  11. Institute of Medicine (US) and National Research Council (US) Committee to Reexamine IOM Pregnancy Weight Guidelines; Rasmussen, K.; Yaktine, A. Weight Gain during Pregnancy: Reexamining the Guidelines; National Academies Press: Washington, DC, USA, 2009; ISBN 9780309131131.
  12. Technical Advisory Group on Gestational Weight Gain (TAG-GWG). Available online: https://www.who.int/groups/technical-advisory-group-on-gestational-weight-gain-(tag-gwg) (accessed on 26 July 2023).
  13. Bijlholt, M.; Ameye, L.; van Uytsel, H.; Devlieger, R.; Bogaerts, A. Evolution of Postpartum Weight and Body Composition after Excessive Gestational Weight Gain: The Role of Lifestyle Behaviors—Data from the INTER-ACT Control Group. Int. J. Environ. Res. Public Health 2021, 18, 6344.
  14. Nehring, I.; Schmoll, S.; Beyerlein, A.; Hauner, H.; Von Kries, R. Gestational Weight Gain and Long-Term Postpartum Weight Retention: A Meta-Analysis. Am. J. Clin. Nutr. 2011, 94, 1225–1231.
  15. McDowell, M.; Cain, M.A.; Brumley, J. Excessive Gestational Weight Gain. J. Midwifery Womens Health 2019, 64, 46–54.
  16. Versele, V.; Stas, L.; Aerenhouts, D.; Deliens, T.; Clarys, P.; Gucciardo, L.; Bogaerts, A.; Devlieger, R. Changes in Maternal and Paternal Body Composition during the Transition to Parenthood (TRANSPARENTS). Obesity 2022, 31, 225–233.
  17. Lende, M.; Rijhsinghani, A. Gestational Diabetes: Overview with Emphasis on Medical Management. Int. J. Environ. Res. Public Health 2020, 17, 9573.
  18. Chiefari, E.; Arcidiacono, B.; Foti, D.; Brunetti, A. Gestational Diabetes Mellitus: An Updated Overview. J. Endocrinol. Investig. 2017, 40, 899–909.
  19. Rosenbaum, D.L.; Gillen, M.M.; Markey, C.H. The Importance of Sleep and Parity in Understanding Changes in Weight and Breastfeeding Behavior among Postpartum Women. Appetite 2022, 170, 105889.
  20. Lambrinou, C.P.; Karaglani, E.; Manios, Y. Breastfeeding and Postpartum Weight Loss. Curr. Opin. Clin. Nutr. Metab. Care 2019, 22, 413–417.
  21. Antonakou, A.; Papoutsis, D.; Panou, I.; Chiou, A.; Matalas, A.L. Role of Exclusive Breastfeeding in Energy Balance and Weight Loss during the First Six Months Postpartum. Clin. Exp. Obs. Gynecol. 2013, 40, 485–488.
  22. Borschel, M.W.; Phd, R.D.; Kirksey, A.; Hannemann, R.E. Evaluation of Test-Weighing for the Assessment of Milk Volume Intake of Formula-Fed Infants and Its Application to Breast-Fed Infants. Am. J. Clin. Nutr. 1986, 43, 367–373.
  23. Savenije, O.E.M.; Brand, P.L.P. Accuracy and Precision of Test Weighing to Assess Milk Intake in Newborn Infants. Arch. Dis. Child. -Fetal Neonatal Ed. 2006, 91, F330-2.
  24. International Atomic Energy Agency. Stable Isotope Technique to Assess Intake of Human Milk in Breastfed Infants; IAEA Human Health Series No. 7; IAEA: Vienna, Austria, 2010; pp. 1–66.
  25. Tongchom, W.; Pongcharoen, T.; Judprasong, K.; Udomkesmalee, E.; Kriengsinyos, W.; Winichagoon, P. Human Milk Intake of Thai Breastfed Infants During the First 6 Months Using the Dose-to-Mother Deuterium Dilution Method. Food Nutr. Bull. 2020, 41, 343–354.
  26. Kushner, D.J.; Baker, A.; Dunstall, T.G. Pharmacological Uses and Perspectives of Heavy Water and Deuterated Compounds. Can. J. Physiol. Pharmacol. 1999, 77, 79–88.
  27. Yeung, C.H.T.; Fong, S.; Malik, P.R.V.; Edginton, A.N. Quantifying Breast Milk Intake by Term and Preterm Infants for Input into Paediatric Physiologically Based Pharmacokinetic Models. Matern. Child Nutr. 2020, 16, e12938.
  28. Victora, C.G.; Bahl, R.; Barros, A.J.D.; França, G.V.A.; Horton, S.; Krasevec, J.; Murch, S.; Sankar, M.J.; Walker, N.; Rollins, N.C.; et al. Breastfeeding in the 21st Century: Epidemiology, Mechanisms, and Lifelong Effect. Lancet 2016, 387, 475–490.
  29. Anderson, P.O.; Manoguerra, A.S.; Valdés, V. A Review of Adverse Reactions in Infants from Medications in Breastmilk. Clin. Pediatr. 2016, 55, 236–244.
  30. Kent, J.C.; Mitoulas, L.R.; Cregan, M.D.; Ramsay, D.T.; Doherty, D.A.; Hartmann, P.E. Volume and Frequency of Breastfeedings and Fat Content of Breast Milk throughout the Day. Pediatrics 2006, 117, e387–e395.
  31. Oras, P.; Blomqvist, Y.T.; Nyqvist, K.H.; Gradin, M.; Rubertsson, C.; Hellström-Westas, L.; Funkquist, E.-L. Breastfeeding Patterns in Preterm Infants Born at 28-33 Gestational Weeks. J. Hum. Lact. 2015, 31, 377–385.
  32. Pressly, M.A.; Schmidt, S.; Guinn, D.; Liu, Z.; Ceresa, C.; Samuels, S.; Madabushi, R.; Florian, J.; Fletcher, E.P. 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, S106–S116.
  33. Gourley, G.R.; Arend, R.A. Beta-Glucuronidase and Hyperbilirubinaemia in Breast-Fed and Formula-Fed Babies. Lancet 1986, 1, 644–646.
  34. Ren, Q.; Zhou, Y.; Zhang, W.; Tian, Y.; Sun, H.; Zhao, X.; Xu, Y.; Jiang, S. Longitudinal Changes in the Bioactive Proteins in Human Milk of the Chinese Population: A Systematic Review. Food Sci. Nutr. 2021, 9, 25–35.
  35. Hamosh, M. Enzymes in Human Milk. In Handbook of Milk Composition; Elsevier: Amsterdam, The Netherlands, 1995; pp. 388–427.
  36. Freed, L.M.; Berkow, S.E.; Hamosh, P.; York, C.M.; Mehta, N.R.; Hamosh, M. Lipases in Human Milk: Effect of Gestational Age and Length of Lactation on Enzyme Activity. J. Am. Coll. Nutr. 1989, 8, 143–150.
  37. Ballard, O.; Morrow, A.L. Human Milk Composition. Nutrients and Bioactive Factors. Pediatr. Clin. North. Am. 2013, 60, 49–74.
  38. Morriss, F.H.; Brewer, E.D.; Spedale, S.B.; Riddle, L.; Temple, D.M.; Caprioli, R.M.; West, M.S. Relationship of Human Milk PH During Course of Lactation to Concentrations of Citrate and Fatty Acids. Pediatrics 1986, 78, 458–464.
  39. Andreas, N.J.; Kampmann, B.; Mehring Le-Doare, K. Human Breast Milk: A Review on Its Composition and Bioactivity. Early Hum. Dev. 2015, 91, 629–635.
  40. Koletzko, B. Human Milk Lipids. Ann. Nutr. Metab. 2017, 69, 28–40.
More
This entry is offline, you can click here to edit this entry!
Video Production Service