Skip to main content
Log in

Physiologically Based Pharmacokinetic Modeling Framework to Predict Neonatal Pharmacokinetics of Transplacentally Acquired Emtricitabine, Dolutegravir, and Raltegravir

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objective

Little is understood about neonatal pharmacokinetics immediately after delivery and during the first days of life following intrauterine exposure to maternal medications. Our objective was to develop and evaluate a novel, physiologically based pharmacokinetic modeling workflow for predicting perinatal and postnatal disposition of commonly used antiretroviral drugs administered prenatally to pregnant women living with human immunodeficiency virus.

Methods

Using previously published, maternal-fetal, physiologically based pharmacokinetic models for emtricitabine, dolutegravir, and raltegravir built with PK-Sim/MoBi®, placental drug transfer was predicted in late pregnancy. The total drug amount in fetal compartments at term delivery was estimated and subsequently integrated as initial conditions in different tissues of a whole-body, neonatal, physiologically based pharmacokinetic model to predict drug concentrations in the neonatal elimination phase after birth. Neonatal elimination processes were parameterized according to published data. Model performance was assessed by clinical data.

Results

Neonatal physiologically based pharmacokinetic models generally captured the initial plasma concentrations after delivery but underestimated concentrations in the terminal phase. The mean percentage error for predicted plasma concentrations was − 71.5%, − 33.8%, and 76.7% for emtricitabine, dolutegravir, and raltegravir, respectively. A sensitivity analysis suggested that the activity of organic cation transporter 2 and uridine diphosphate glucuronosyltransferase 1A1 during the first postnatal days in term newborns is ~11% and ~30% of that in adults, respectively.

Conclusions

These findings demonstrate the general feasibility of applying physiologically based pharmacokinetic models to predict washout concentrations of transplacentally acquired drugs in newborns. These models can increase the understanding of pharmacokinetics during the first postnatal days and allow the prediction of drug exposure in this vulnerable population.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

taken from an in vivo study of Hirt et al. [17], IMPAACT P1026 [10], and Clarke et al. [16]. a Emtricitabine 400-mg single dose in pregnant women with an average gestational age of 39 weeks at delivery. Empty circles represent individual concentration data in the maternal plasma taken from an in vivo study of Hirt et al. [17]; the line represents the predicted mean concentrations in the maternal plasma; the shaded area represents the predicted 5th–95th percentile range of the prediction. b Emtricitabine 400-mg single dose in pregnant women with an average gestational age of 39 weeks at delivery. Empty circles represent individual concentration data in the umbilical vein taken from an in vivo study of Hirt et al. [17] The line represents the predicted mean concentrations in the umbilical vein. The shaded area represents the predicted 5th–95th percentile range of the prediction. c Emtricitabine 400-mg single dose in pregnant women with an average gestational age of 39 weeks at delivery. The line represents the predicted mean amount of emtricitabine in the fetus. The marks represent the delivery time after the last dose. d Dolutegravir 50 mg once a day in pregnant women with an average gestational age of 38 weeks at delivery. Empty circles represent individual concentration data in the maternal plasma taken from an in vivo study of IMPAACT P1026; [10] the line represents the predicted mean concentration in the maternal plasma; the shaded area represents the predicted 5th–95th percentile range of the prediction. e Dolutegravir 50 mg once a day in pregnant women with an average gestational age of 38 weeks at delivery. Empty circles represent individual concentration data in the umbilical vein taken from an in vivo study of IMPAACT P1026; [10] the line represents the predicted mean concentration in the umbilical vein. The shaded area presents the predicted 5th–95th percentile range of the prediction. f Dolutegravir 50 mg once a day in pregnant women with an average gestational age of 38 weeks at delivery; the line represents the predicted mean amount of dolutegravir in the fetus. The marks represent the delivery time after the last dose. g Raltegravir 400 mg twice a day in pregnant women with an average gestational age of 38 weeks at delivery. Empty circles represent individual concentration data in the maternal plasma taken from an in vivo study of Clarke et al. [16]; the line represents the predicted mean concentrations in the maternal plasma; the shaded area represents the predicted 5th–95th percentile range of the prediction. h Raltegravir 400 mg twice a day in pregnant women with an average gestational age of 38 weeks at delivery. Empty circles represent individual concentration data in the umbilical vein taken from an in vivo study of Clarke et al. [16]; the line represents the predicted mean concentrations in the umbilical vein. The shaded area represents the predicted 5th–95th percentile range of the prediction. i Raltegravir 400 mg twice a day in pregnant women with an average gestational age of 38 weeks at delivery. The line represents the predicted mean amount of raltegravir in the fetus. The marks represent the delivery time after the last dose. conc concentration

Fig. 3

taken from Hirt et al. [17]. b Dolutegravir plasma concentration in newborns; maternal dose of 50 mg once a day. Observed data were taken from IMPAACT P1026 [10]. c Raltegravir plasma concentration in newborns; maternal dose of 400 mg twice a day. Observed data were taken from Clarke et al. [16] conc concentration

Fig. 4

taken from Hirt et al. [17] and black circles represent median observed data. b Emtricitabine plasma concentration in newborns with differing unbound fractions; maternal dose of 400 mg single dose. Empty circles represent observed data taken from Hirt et al. [17] and black circles represent median observed data. c Dolutegravir plasma concentration in newborns with differing uridine diphosphate glucuronosyltransferase 1A1 and cytochrome P450 3A4 activities; maternal dose of 50 mg once a day. Empty circles represent observed data taken from IMPAACT P1026 [10] and black circles represent median observed data. d Dolutegravir plasma concentration in newborns with differing unbound fractions; maternal dose of 50 mg once a day. Empty circles represent observed data taken from IMPAACT P1026 [10] and black circles represent median observed data. e Raltegravir plasma concentration in newborns with differing uridine diphosphate glucuronosyltransferase 1A1 activity and renal elimination; maternal dose of 400 mg twice a day. Empty circles represent observed data taken from Clarke et al. [16] and black circles represent median observed data. f Raltegravir plasma concentration in newborns with differing unbound fractions; maternal dose of 50 mg once a day. Empty circles represent observed data taken from Clarke et al. [16] and black circles represent median observed data. conc concentration

Fig. 5

taken from Hirt et al. [17] and black circles represent median observed data. b Dolutegravir plasma concentration in newborns with differing dosing; maternal dose of 50 mg once a day. Empty circles represent observed data taken from IMPAACT P1026 [10] and black circles represent median observed data. c Raltegravir plasma concentration in newborns with differing dosing; maternal dose of 400 mg twice a day. Empty circles represent observed data taken from Clarke et al. [16] and black circles represent median observed data. conc concentration

Similar content being viewed by others

References

  1. Pariente G, Leibson T, Carls A, Adams-Webber T, Ito S, Koren G. Pregnancy-associated changes in pharmacokinetics: a systematic review. PLoS Med. 2016;13(11):e1002160.

    PubMed  PubMed Central  Google Scholar 

  2. Wang J, Avant D, Green D, Seo S, Fisher J, Mulberg AE, et al. A survey of neonatal pharmacokinetic and pharmacodynamic studies in pediatric drug development. Clin Pharmacol Ther. 2015;98(3):328–35.

    CAS  PubMed  Google Scholar 

  3. Van Den Anker J, Reed MD, Allegaert K, Kearns GL. Developmental changes in pharmacokinetics and pharmacodynamics. J Clin Pharmacol. 2018;58(Suppl. 10):S10-25.

    PubMed  Google Scholar 

  4. Ku LC, Smith PB. Dosing in neonates: special considerations in physiology and trial design. Pediatr Res. 2015;77(1–1):2–9.

    PubMed  Google Scholar 

  5. Claassen K, Thelen K, Coboeken K, Gaub T, Lippert J, Allegaert K, et al. Development of a physiologically-based pharmacokinetic model for preterm neonates: evaluation with in vivo data. Curr Pharm Des. 2015;21(39):5688–98.

    CAS  PubMed  Google Scholar 

  6. Michelet R, Bocxlaer JV, Vermeulen A. PBPK in preterm and term neonates: a review. Curr Pharm Des. 2017;23(38):5943–54.

    CAS  PubMed  Google Scholar 

  7. Abduljalil K, Pan X, Pansari A, Jamei M, Johnson TN. Preterm physiologically based pharmacokinetic model. Part II: applications of the model to predict drug pharmacokinetics in the preterm population. Clin Pharmacokinet. 2020;59(4):501–18.

    CAS  PubMed  Google Scholar 

  8. Stek AM, Best BM, Luo W, Capparelli E, Burchett S, Hu C, et al. Effect of pregnancy on emtricitabine pharmacokinetics. HIV Med. 2012;13(4):226–35.

    CAS  PubMed  Google Scholar 

  9. Colbers AP, Hawkins DA, Gingelmaier A, Kabeya K, Rockstroh JK, Wyen C, et al. The pharmacokinetics, safety and efficacy of tenofovir and emtricitabine in HIV-1-infected pregnant women. AIDS. 2013;27(5):739–48.

    CAS  PubMed  Google Scholar 

  10. Mulligan N, Best BM, Wang J, Capparelli EV, Stek A, Barr E, et al. Dolutegravir pharmacokinetics in pregnant and postpartum women living with HIV. AIDS. 2018;32(6):729–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bollen P, Freriksen J, Konopnicki D, Weizsacker K, Hidalgo Tenorio C, Molto J, et al. The effect of pregnancy on the pharmacokinetics of total and unbound dolutegravir and its main metabolite in women living with human immunodeficiency virus. Clin Infect Dis. 2020. https://doi.org/10.1093/cid/ciaa006.

    Article  Google Scholar 

  12. Watts DH, Stek A, Best BM, Wang J, Capparelli EV, Cressey TR, et al. Raltegravir pharmacokinetics during pregnancy. J Acquir Immune Defic Syndr. 2014;67(4):375–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Blonk MI, Colbers AP, Hidalgo-Tenorio C, Kabeya K, Weizsacker K, Haberl AE, et al. Raltegravir in HIV-1-infected pregnant women: pharmacokinetics, safety, and efficacy. Clin Infect Dis. 2015;61(5):809–16.

    CAS  PubMed  Google Scholar 

  14. Liu XI, Momper JD, Rakhmanina N, Van Den Anker JN, Green DJ, Burckart GJ, et al. Physiologically based pharmacokinetic models to predict maternal pharmacokinetics and fetal exposure to emtricitabine and acyclovir. J Clin Pharmacol. 2020;60(2):240–55.

    CAS  PubMed  Google Scholar 

  15. Liu XI, Momper JD, Rakhmanina NY, Green DJ, Burckart GJ, Cressey TR, et al. Prediction of maternal and fetal pharmacokinetics of dolutegravir and raltegravir using physiologically based pharmacokinetic modeling. Clin Pharmacokinet. 2020;59(11):1433–50.

    CAS  PubMed  Google Scholar 

  16. Clarke DF, Acosta EP, Rizk ML, Bryson YJ, Spector SA, Mofenson LM, et al. Raltegravir pharmacokinetics in neonates following maternal dosing. J Acquir Immune Defic Syndr. 2014;67(3):310–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Hirt D, Urien S, Rey E, Arrive E, Ekouevi DK, Coffie P, et al. Population pharmacokinetics of emtricitabine in human immunodeficiency virus type 1-infected pregnant women and their neonates. Antimicrob Agents Chemother. 2009;53(3):1067–73.

    CAS  PubMed  Google Scholar 

  18. Waitt C, Orrell C, Walimbwa S, Singh Y, Kintu K, Simmons B, et al. Safety and pharmacokinetics of dolutegravir in pregnant mothers with HIV infection and their neonates: a randomised trial (DolPHIN-1 study). PLoS Med. 2019;16(9):e1002895.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Lippert J, Burghaus R, Edginton A, Frechen S, Karlsson M, Kovar A, et al. Open Systems Pharmacology community: an open access, open source, open science approach to modeling and simulation in pharmaceutical sciences. CPT Pharmacometrics Syst Pharmacol. 2019;8(12):878–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dallmann A, Solodenko J, Ince I, Eissing T. Applied concepts in PBPK modeling: how to extend an Open Systems Pharmacology model to the special population of pregnant women. CPT Pharmacometrics Syst Pharmacol. 2018;7(7):419–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. US Food and Drug Administration. Emtriva™ (emtricitabine) capsules. 2003. https://www.accessdata.fda.gov/drugsatfda_docs/label/2003/21500_emtriva_lbl.pdf. Accessed 10 Jun 2020.

  22. Reese MJ, Savina PM, Generaux GT, Tracey H, Humphreys JE, Kanaoka E, et al. In vitro investigations into the roles of drug transporters and metabolizing enzymes in the disposition and drug interactions of dolutegravir, a HIV integrase inhibitor. Drug Metab Dispos. 2013;41(2):353–61.

    CAS  PubMed  Google Scholar 

  23. Kassahun K, Mcintosh I, Cui D, Hreniuk D, Merschman S, Lasseter K, et al. Metabolism and disposition in humans of raltegravir (MK-0518), an anti-AIDS drug targeting the human immunodeficiency virus 1 integrase enzyme. Drug Metab Dispos. 2007;35(9):1657–63.

    CAS  PubMed  Google Scholar 

  24. Dallmann A, Ince I, Meyer M, Willmann S, Eissing T, Hempel G. Gestation-specific changes in the anatomy and physiology of healthy pregnant women: an extended repository of model parameters for physiologically based pharmacokinetic modeling in pregnancy. Clin Pharmacokinet. 2017;56(11):1303–30.

    CAS  PubMed  Google Scholar 

  25. Dallmann A, Ince I, Solodenko J, Meyer M, Willmann S, Eissing T, et al. Physiologically based pharmacokinetic modeling of renally cleared drugs in pregnant women. Clin Pharmacokinet. 2017;56(12):1525–41.

    CAS  PubMed  Google Scholar 

  26. Reznicek J, Ceckova M, Cerveny L, Muller F, Staud F. Emtricitabine is a substrate of MATE1 but not of OCT1, OCT2, P-gp, BCRP or MRP2 transporters. Xenobiotica. 2017;47(1):77–85.

    CAS  PubMed  Google Scholar 

  27. Cheung KWK, Van Groen BD, Spaans E, Van Borselen MD, De Bruijn A, Simons-Oosterhuis Y, et al. A comprehensive analysis of ontogeny of renal drug transporters: mRNA analyses, quantitative proteomics, and localization. Clin Pharmacol Ther. 2019;106(5):1083–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Badee J, Qiu N, Collier AC, Takahashi RH, Forrest WF, Parrott N, et al. Characterization of the ontogeny of hepatic UDP-glucuronosyltransferase enzymes based on glucuronidation activity measured in human liver microsomes. J Clin Pharmacol. 2019;59(Suppl. 1):S42-55.

    CAS  PubMed  Google Scholar 

  29. Acharya C, Hooker AC, Turkyilmaz GY, Jonsson S, Karlsson MO. A diagnostic tool for population models using non-compartmental analysis: the ncappc package for R. Comput Methods Programs Biomed. 2016;127:83–93.

    PubMed  Google Scholar 

  30. Codaccioni M, Brochot C. Assessing the impacts on fetal dosimetry of the modelling of the placental transfers of xenobiotics in a pregnancy physiologically based pharmacokinetic model. Toxicol Appl Pharmacol. 2020;409:115318.

    CAS  PubMed  Google Scholar 

  31. Lommerse J, Clarke D, Kerbusch T, Merdjan H, Witjes H, Teppler H, et al. Maternal-neonatal raltegravir population pharmacokinetics modeling: implications for initial neonatal dosing. CPT Pharmacometrics Syst Pharmacol. 2019;8(9):643–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Yamamoto K, Fukushima S, Mishima Y, Hashimoto M, Yamakawa K, Fujioka K, et al. Pharmacokinetic assessment of alprazolam-induced neonatal abstinence syndrome using physiologically based pharmacokinetic model. Drug Metab Pharmacokinet. 2019;34(6):400–2.

    CAS  PubMed  Google Scholar 

  33. Li H, Lampe JN. Neonatal cytochrome P450 CYP3A7: a comprehensive review of its role in development, disease, and xenobiotic metabolism. Arch Biochem Biophys. 2019;673:108078.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Miyagi SJ, Milne AM, Coughtrie MW, Collier AC. Neonatal development of hepatic UGT1A9: implications of pediatric pharmacokinetics. Drug Metab Dispos. 2012;40(7):1321–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Moss DM, Kwan WS, Liptrott NJ, Smith DL, Siccardi M, Khoo SH, et al. Raltegravir is a substrate for SLC22A6: a putative mechanism for the interaction between raltegravir and tenofovir. Antimicrob Agents Chemother. 2011;55(2):879–87.

    CAS  PubMed  Google Scholar 

  36. Bunglawala F, Rajoli RKR, Mirochnick M, Owen A, Siccardi M. Prediction of dolutegravir pharmacokinetics and dose optimization in neonates via physiologically based pharmacokinetic (PBPK) modelling. J Antimicrob Chemother. 2020;75(3):640–7.

    CAS  PubMed  Google Scholar 

  37. Ota Y, Maruo Y, Matsui K, Mimura Y, Sato H, Takeuchi Y. Inhibitory effect of 5beta-pregnane-3alpha,20beta-diol on transcriptional activity and enzyme activity of human bilirubin UDP-glucuronosyltransferase. Pediatr Res. 2011;70(5):453–7.

    CAS  PubMed  Google Scholar 

  38. Shibuya A, Itoh T, Tukey RH, Fujiwara R. Impact of fatty acids on human UDP-glucuronosyltransferase 1A1 activity and its expression in neonatal hyperbilirubinemia. Sci Rep. 2013;3:2903.

    PubMed  PubMed Central  Google Scholar 

  39. Moss DM, Siccardi M, Murphy M, Piperakis MM, Khoo SH, Back DJ, et al. Divalent metals and pH alter raltegravir disposition in vitro. Antimicrob Agents Chemother. 2012;56(6):3020–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. US Food and Drug Administration. Clinical pharmacology review: dolutegravir, GSK1349572. 2012. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/204790Orig1s000ClinPharmR.pdf. Accessed 29 Jun 2020.

  41. Laufer R, Paz OG, Di Marco A, Bonelli F, Monteagudo E, Summa V, et al. Quantitative prediction of human clearance guiding the development of Raltegravir (MK-0518, isentress) and related HIV integrase inhibitors. Drug Metab Dispos. 2009;37(4):873–83.

    CAS  PubMed  Google Scholar 

  42. Moss DM, Siccardi M, Back DJ, Owen A. Predicting intestinal absorption of raltegravir using a population-based ADME simulation. J Antimicrob Chemother. 2013;68(7):1627–34.

    CAS  PubMed  Google Scholar 

  43. Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94(6):1259–76.

    CAS  PubMed  Google Scholar 

  44. Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238–57.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiaomei I. Liu.

Ethics declarations

Funding

Overall support for the International Maternal Pediatric Adolescent AIDS Clinical Trials Network (IMPAACT) was provided by the National Institute of Allergy and Infectious Diseases with co-funding from the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) and the National Institute of Mental Health, all components of the National Institutes of Health (NIH), under Award Numbers UM1AI068632 (IMPAACT LOC), UM1AI068616 (IMPAACT SDMC), and UM1AI106716 (IMPAACT LC), and by NICHD contract number HHSN275201800001I. The NIH awards numbers 5T32HD087969-03 and 5T32HD087969-02 also support this project.

Conflicts of interest/Competing Interests

Xiaomei I. Liu, Jeremiah D. Momper, Natella Y. Rakhmanina, Dionna J. Green, Gilbert J. Burckart, Tim R. Cressey, Mark Mirochnick, Brookie M. Best, John N. van den Anker, and André Dallmann have no conflicts of interest with respect to the research, authorship, and/or publication of this article. André Dallmann is an employee of Bayer AG, a company that is part of the Open Systems Pharmacology member team and involved in the Open Systems Pharmacology software development used in this study. The results from this study will be presented in part at the American College of Clinical Pharmacology Annual Meeting, September 2020.

Ethics approval

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Authors’ contributions

Not applicable.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X.I., Momper, J.D., Rakhmanina, N.Y. et al. Physiologically Based Pharmacokinetic Modeling Framework to Predict Neonatal Pharmacokinetics of Transplacentally Acquired Emtricitabine, Dolutegravir, and Raltegravir. Clin Pharmacokinet 60, 795–809 (2021). https://doi.org/10.1007/s40262-020-00977-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-020-00977-w

Navigation