Infant Exposure to Dolutegravir Through Placental and Breast Milk Transfer: A Population Pharmacokinetic Analysis of DolPHIN-1

Laura Dickinson, Stephen Walimbwa, Yashna Singh, Julian Kaboggoza, Kenneth Kintu, Mary Sihlangu, Julie-Anne Coombs, Thokozile R Malaba, Josaphat Byamugisha, Henry Pertinez, Alieu Amara, Joshua Gini, Laura Else, Christie Heiberg, Eva Maria Hodel, Helen Reynolds, Landon Myer, Catriona Waitt, Saye Khoo, Mohammed Lamorde, Catherine Orrell, DolPHIN-1 Study Group, Ritah Nakijoba, Isabella Kyohairwe, Johnson Magoola, Emmanuel Ssempija, Laura Dickinson, Stephen Walimbwa, Yashna Singh, Julian Kaboggoza, Kenneth Kintu, Mary Sihlangu, Julie-Anne Coombs, Thokozile R Malaba, Josaphat Byamugisha, Henry Pertinez, Alieu Amara, Joshua Gini, Laura Else, Christie Heiberg, Eva Maria Hodel, Helen Reynolds, Landon Myer, Catriona Waitt, Saye Khoo, Mohammed Lamorde, Catherine Orrell, DolPHIN-1 Study Group, Ritah Nakijoba, Isabella Kyohairwe, Johnson Magoola, Emmanuel Ssempija

Abstract

Background: Rapid reduction in human immunodeficiency virus (HIV) load is paramount to prevent peripartum transmission in women diagnosed late in pregnancy. We investigated dolutegravir population pharmacokinetics in maternal plasma, umbilical cord, breast milk, and infant plasma samples from DolPHIN-1 participants (NCT02245022) presenting with untreated HIV late in pregnancy (28-36 weeks gestation).

Methods: Pregnant women from Uganda and South Africa were randomized (1:1) to daily dolutegravir (50 mg/d) or efavirenz-based therapy. Dolutegravir pharmacokinetic sampling (0-24 hours) was undertaken 14 days after treatment initiation and within 1-3 weeks after delivery, with matched maternal and cord samples at delivery. Mothers were switched to efavirenz, and maternal and infant plasma and breast milk samples were obtained 24, 48, or 72 hours after the switch. Nonlinear mixed-effects modeling was used to describe dolutegravir in all matrices and to evaluate covariates.

Results: A total of 28 women and 22 infants were included. Maternal dolutegravir was described by a 2-compartment model linked to a fetal and breast milk compartment. Cord and breast milk to maternal plasma ratios were 1.279 (1.209-1.281) and 0.033 (0.021-0.050), respectively. Infant dolutegravir was described by breast milk-to-infant and infant elimination rate constants. No covariate effects were observed. The median predicted infant dolutegravir half-life and median time to protein-adjusted 90% inhibitory concentration (0.064 mg/L) for those above this threshold were 37.9 (range, 22.1-63.5) hours and 108.9 (18.6-129.6) hours (4.5 [0.8-5.4] days) (n = 13), respectively.

Conclusions: Breastfeeding contributed relatively little to infant plasma exposure, but a median of 4.5 days of additional prophylaxis to some of the breastfed infants was observed after cessation of maternal dolutegravir (3-15 days postpartum), which waned with time postpartum as transplacental dolutegravir cleared.

Keywords: breast milk; dolutegravir; infant pharmacokinetics; population pharmacokinetics; pregnancy.

© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Dolutegravir visual predictive check for maternal plasma at third trimester (n = 28) (A), maternal plasma at delivery (n = 18) (B), maternal plasma postpartum (n = 27) (C), umbilical cord (n = 16) (D), and breast milk (n = 27) (E), along with the proportion below the lower limit of quantification (LLQ). Maternal plasma postpartum and breast milk samples were obtained after the final maternal dolutegravir dose. Lines represent percentiles of the observed data (fifth, 50th, and 95th percentiles), and shaded areas, 95% confidence intervals of the simulated data. Observed concentration-time data for maternal plasma (250 concentrations for the third trimester, 18 at delivery, and 265 postpartum), umbilical cord (16 concentrations), and breast milk (80 concentrations) are superimposed (open circles).
Figure 2.
Figure 2.
Dolutegravir prediction-corrected visual predictive check for infant plasma (n = 22) after the final maternal dolutegravir dose, given a median of 7 days postpartum. Lines represent percentiles of the observed data (fifth, 50th, and 95th percentiles), and shaded areas, 95% confidence intervals of the simulated data. Observed concentration time data are superimposed (65 concentrations [open circles]).
Figure 3.
Figure 3.
Schematic of the population pharmacokinetic model to simultaneously describe dolutegravir in maternal plasma, umbilical cord, and breast milk. Maternal plasma, cord and breast milk individual model estimates were then fixed to describe dolutegravir pharmacokinetics in infant plasma using a sequential approach. Abbreviations: CL/F, apparent oral clearance; ka, absorption rate constant; kBM-INF, breast milk–to–infant transfer rate constant; kBM-M, breast milk–to–mother transfer rate constant; kF-M, fetus-to-mother transfer rate constant; kINF, infant elimination rate constant; kM-BM, mother–tobreast milk transfer rate constant; kM-F, mother-to-fetus transfer rate constant; Q/F, intercompartmental clearance; RSE, relative standard error; VBM, volume of the breast milk compartment; Vc/F, apparent volume of distribution of the central compartment; VINF/F, infant apparent volume of distribution; Vp/F, volume of the peripheral compartment.

References

    1. Meyers K, Qian H, Wu Y, et al. . Early initiation of ARV during pregnancy to move towards virtual elimination of mother-to-child-transmission of HIV-1 in Yunnan, China. PLoS One 2015; 10:e0138104.
    1. World Health Organization. Policy brief: update of recommendations on first- and second-line antiretroviral regimens. 2019. Available at: . Accessed 10 August 2020.
    1. Hill A, Clayden P, Thorne C, Christie R, Zash R. Safety and pharmacokinetics of dolutegravir in HIV-positive pregnant women: a systematic review. J Virus Erad 2018; 4:66–71.
    1. Zash R, Jacobson DL, Diseko M, et al. . Comparative safety of dolutegravir-based or efavirenz-based antiretroviral treatment started during pregnancy in Botswana: an observational study. Lancet Glob Health 2018; 6:e804–10.
    1. Cottrell ML, Hadzic T, Kashuba AD. Clinical pharmacokinetic, pharmacodynamic and drug-interaction profile of the integrase inhibitor dolutegravir. Clin Pharmacokinet 2013; 52:981–94.
    1. Llibre JM, Pulido F, García F, García Deltoro M, Blanco JL, Delgado R. Genetic barrier to resistance for dolutegravir. AIDS Rev 2015; 17:56–64.
    1. Waitt C, Orrell C, Walimbwa S, 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:e1002895.
    1. Kintu K, Malaba TR, Nakibuka J, et al. ; DolPHIN-2 Study Group . Dolutegravir versus efavirenz in women starting HIV therapy in late pregnancy (DolPHIN-2): an open-label, randomised controlled trial. Lancet HIV 2020; 7:e332–9.
    1. Meintjes G, Black J, Conradie F, et al. . Southern African HIV Clinicians Society adult antiretroviral therapy guidelines: update on when to initiate antiretroviral therapy. South Afr J HIV Med 2015; 16:428.
    1. The Republic of Uganda Ministry of Health. Consolidated guidelines for the prevention and treatment of HIV in Uganda. 2016. Available at: . Accessed 05 January 2021.
    1. Gini J, Penchala SD, Amara A, et al. . Validation and clinical application of a novel LC-MS method for quantification of dolutegravir in breast milk. Bioanalysis doi:10.4155/bio-2018-0085. Published 19 November 2018.
    1. Penchala SD, Fawcett S, Else L, et al. . The development and application of a novel LC-MS/MS method for the measurement of dolutegravir, elvitegravir and cobicistat in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1027:174–80.
    1. Beal SL, Sheiner LB, Boeckmann AJ, Bauer RJ.. NONMEM users guide 2018. Gaithersburg, Maryland: ICON Development Solutions, 1989–2018.
    1. Min S, Song I, Borland J, et al. . Pharmacokinetics and safety of S/GSK1349572, a next-generation HIV integrase inhibitor, in healthy volunteers. Antimicrob Agents Chemother 2010; 54:254–8.
    1. Anderson PO, Valdés V. Variation of milk intake over time: clinical and pharmacokinetic implications. Breastfeed Med 2015; 10:142–4.
    1. Salman S, Sy SK, Ilett KF, Page-Sharp M, Paech MJ. Population pharmacokinetic modeling of tramadol and its O-desmethyl metabolite in plasma and breast milk. Eur J Clin Pharmacol 2011; 67:899–908.
    1. Wilson JT. Determinants and consequences of drug excretion in breast milk. Drug Metab Rev 1983; 14:619–52.
    1. James RJ, James A, Drewett RF, Cheetham TD. Milk intake and feeding behavior in the first week of life and its relationship to cord blood ghrelin, leptin, and insulin concentrations. Pediatr Res 2007; 62:695–9.
    1. Zhang J, Hayes S, Sadler BM, et al. . Population pharmacokinetics of dolutegravir in HIV-infected treatment-naive patients. Br J Clin Pharmacol 2015; 80:502–14.
    1. Bollen P, Colbers A, Schalkwijk S. A comparison of the pharmacokinetics of dolutegravir during pregnancy and postpartum. Presented at: 18th International Workshop on Clinical Pharmacology of Antiviral Therapy; 14–17 June 2017; Chicago, IL. Abstract O_07.
    1. Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol 2015; 39:512–9.
    1. Mulligan N, Best BM, Wang J, et al. ; IMPAACT P1026s Protocol Team . Dolutegravir pharmacokinetics in pregnant and postpartum women living with HIV. AIDS 2018; 32:729–37.
    1. Kobbe R, Schalkwijk S, Dunay G, et al. . Dolutegravir in breast milk and maternal and infant plasma during breastfeeding. AIDS 2016; 30:2731–3.
    1. Min S, Sloan L, DeJesus E, et al. . Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of dolutegravir as 10-day monotherapy in HIV-1-infected adults. AIDS 2011; 25:1737–45.
    1. Badée J, Fowler S, de Wildt SN, Collier AC, Schmidt S, Parrott N. The ontogeny of UDP-glucuronosyltransferase enzymes, recommendations for future profiling studies and application through physiologically based pharmacokinetic modelling. Clin Pharmacokinet 2019; 58:189–211.
    1. Miyagi SJ, Collier AC. The development of UDP-glucuronosyltransferases 1A1 and 1A6 in the pediatric liver. Drug Metab Dispos 2011; 39:912–9.
    1. Badée J, Qiu N, Collier AC, 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):42–55.
    1. Begg EJ, Atkinson HC, Duffull SB. Prospective evaluation of a model for the prediction of milk:plasma drug concentrations from physicochemical characteristics. Br J Clin Pharmacol 1992; 33:501–5.
    1. Berle JO, Spigset O. Antidepressant use during breastfeeding. Curr Womens Health Rev 2011; 7:28–34.
    1. Fogel JM, Mwatha A, Richardson P, et al. . Impact of maternal and infant antiretroviral drug regimens on drug resistance in HIV-infected breastfeeding infants. Pediatr Infect Dis J 2013; 32:e164–9.
    1. Inzaule SC, Weidle PJ, Yang C, et al. . Prevalence and dynamics of the K65R drug resistance mutation in HIV-1-infected infants exposed to maternal therapy with lamivudine, zidovudine and either nevirapine or nelfinavir in breast milk. J Antimicrob Chemother 2016; 71:1619–26.
    1. Zeh C, Weidle PJ, Nafisa L, et al. . HIV-1 drug resistance emergence among breastfeeding infants born to HIV-infected mothers during a single-arm trial of triple-antiretroviral prophylaxis for prevention of mother-to-child transmission: a secondary analysis. PLoS Med 2011; 8:e1000430.

Source: PubMed

3
Iratkozz fel