Abacavir Exposure in Children Cotreated for Tuberculosis with Rifampin and Superboosted Lopinavir-Ritonavir

Helena Rabie, Tjokosela Tikiso, Janice Lee, Lee Fairlie, Renate Strehlau, Raziya Bobat, Afaaf Liberty, Helen McIlleron, Isabelle Andrieux-Meyer, Mark Cotton, Marc Lallemant, Paolo Denti, Helena Rabie, Tjokosela Tikiso, Janice Lee, Lee Fairlie, Renate Strehlau, Raziya Bobat, Afaaf Liberty, Helen McIlleron, Isabelle Andrieux-Meyer, Mark Cotton, Marc Lallemant, Paolo Denti

Abstract

In children requiring lopinavir coformulated with ritonavir in a 4:1 ratio (lopinavir-ritonavir-4:1) and rifampin, adding ritonavir to achieve a 4:4 ratio with lopinavir (LPV/r-4:4) overcomes the drug-drug interaction. Possible drug-drug interactions within this regimen may affect abacavir concentrations, but this has never been studied. Children weighing <15 kg needing rifampin and LPV/r-4:4 were enrolled in a pharmacokinetic study and underwent intensive pharmacokinetic sampling on 3 visits: (i) during the intensive and (ii) continuation phases of antituberculosis treatment with LPV/r-4:4 and (iii) 1 month after antituberculosis treatment completion on LPV/r-4:1. Pharmacometric modeling and simulation were used to compare exposures across weight bands with adult target exposures. Eighty-seven children with a median (interquartile range) age and weight of 19 (4 to 64) months and 8.7 (3.9 to 14.9) kg, respectively, were included in the abacavir analysis. Abacavir pharmacokinetics were best described by a two-compartment model with first-order elimination and transit compartment absorption. After allometric scaling adjusted for the effect of body size, maturation could be identified: clearance was predicted to be fully mature at about 2 years of age and to reach half of this mature value at about 2 months of age. Abacavir bioavailability decreased 36% during treatment with rifampin and LPV/r-4:4 but remained within the median adult recommended exposure, except for children in the 3- to 4.9-kg weight band, in which the exposures were higher. The observed predose morning trough concentrations were higher than the evening values. Though abacavir exposure significantly decreased during concomitant administration of rifampin and LPV/r-4:4, it remained within acceptable ranges. (This study is registered in ClinicalTrials.gov under identifier NCT02348177.).

Keywords: NONMEM; abacavir; children; lopinavir; population pharmacokinetics; rifampin.

Copyright © 2020 Rabie et al.

Figures

FIG 1
FIG 1
(Top) Visual predictive check of abacavir concentration versus time after dose, stratified by PK visit. PK visit 1 is the intensive phase of antituberculosis treatment with LPV/r-4:4, PK visit 2 is the continuation phase of antituberculosis treatment with LPV/r-4:4, and PK visit 3 represents 1 month after antituberculosis treatment completion on LPV/r-4:1. The solid and dashed lines represent the 50th, 5th, and 95th percentiles of the observed data, while the shaded areas represent the model-predicted 95% confidence intervals for the same percentiles. The dots are the observed concentrations. The yellow ticks on the x axis are bin boundaries. (Bottom) Proportion of LLOQ values versus time after dose. The solid blue line represents the observed proportion, while the blue shaded area is the 90% confidence interval for the same proportion, as predicted by the model.
FIG 2
FIG 2
Maturation function of abacavir clearance versus postmenstrual age (bottom x axis) or postnatal age (top x axis; assuming an average gestation of 9 months), after adjusting for weight. The solid vertical blue line represents birth, while the dashed vertical lines represent 1 year and 2 years of postnatal age. The red ticks on the lower x axis represent the postmenstrual age values available in our data.
FIG 3
FIG 3
Simulated steady-state abacavir AUC0–12 versus body weight. (Left) Exposures during cotreatment with superboosted LPV/r-4:4 and rifampin; (right) exposures during cotreatment with unboosted LPV/r-4:1. The box indicates the interquartile range, while the whiskers denote the 2.5th and the 97.5th percentiles. The green box plots show the exposures of children weighing from 3 to 24.9 kg receiving the current pediatric dosing, as shown in Table 4, while the red box plots show the predicted exposure in adults weighing 25 to 39.9 kg and 40 to 59.9 kg, with children receiving a dose of 300 mg twice daily and adults receiving a dose of 600 mg once daily. The adult AUC0–24 was divided by 2 to obtain a value comparable to the AUC0–12 for comparison to the children’s exposures. The red horizontal dashed line represents the recommended median adult exposure (6.02 mg·h/liter). The weights of the children in this study population were mostly in the range of 4 to 16 kg; all the results outside this weight range (boxes with faint color) were extrapolated using maturation and allometric scaling. To improve the readability of the chart, the y axis was cut; the 97.5th percentile predicted AUC for children in the 3-kg weight band reached 67 mg·h/liter.
FIG 4
FIG 4
Summary of model-predicted abacavir AUC0–12 versus weight bands in each pharmacokinetic visit. The box indicates the interquartile range, while the whiskers denote the 2.5th and the 97.5th percentiles. Each dot represents an individual AUC. PK visit 1 is the intensive phase of antituberculosis treatment with LPV/r-4:4, PK visit 2 is the continuation phase of antituberculosis treatment with LPV/r-4:4, and PK visit 3 represents 1 month after the completion of antituberculosis treatment with LPV/r-4:1. The red horizontal solid line represents the reference median AUC, while the broken lines represent adult AUC values from the literature: Yuen et al. (7) (dashed orange line), Moyle et al. (17) (long-dash black line), McDowell et al. (6) (dot-dash green line), and Weller et al. (43) (dotted blue line).

References

    1. World Health Organization. 2019. Updated recommendations on first-line and second-line antiretroviral regimens and post-exposure prophylaxis and recommendations on early infant diagnosis of HIV. World Health Organization, Geneva, Switzerland.
    1. Bergshoeff A, Burger D, Verweij C, Farrelly L, Flynn J, Le Prevost M, Walker S, Novelli V, Lyall H, Khoo S, Gibb D, PENTA-13 Study Group. 2005. Plasma pharmacokinetics of once- versus twice-daily lamivudine and abacavir: simplification of combination treatment in HIV-1-infected children (PENTA-13). Antivir Ther 10:239–246.
    1. Paediatric European Network for Treatment of AIDS (PENTA). 2010. Pharmacokinetic study of once-daily versus twice-daily abacavir and lamivudine in HIV type-1-infected children aged 3-36 months. Antivir Ther 15:297–305. doi:10.3851/IMP1532.
    1. Western Cape Health Department. 2018. The Western Cape consolidated guidelines for HIV treatment: prevention of mother- to- child transmission of HIV (PMTCT), children, adolescents and adults (amended version). Western Cape Health Department, Cape Town, South Africa.
    1. Nahirya-Ntege P, Musiime V, Naidoo B, Bakeera-Kitaka S, Nathoo K, Munderi P, Mugyenyi P, Kekitiinwa A, Bwakura-Dangarembizi MF, Crawley J, ARROW Trial Team. 2011. Low incidence of abacavir hypersensitivity reaction among African children initiating antiretroviral therapy. Pediatr Infect Dis J 30:535–537. doi:10.1097/INF.0b013e3182076864.
    1. McDowell JA, Chittick GE, Stevens CP, Edwards KD, Stein DS. 2000. Pharmacokinetic interaction of abacavir (1592U89) and ethanol in human immunodeficiency virus-infected adults. Antimicrob Agents Chemother 44:1686–1690. doi:10.1128/aac.44.6.1686-1690.2000.
    1. Yuen GJ, Weller S, Pakes GE. 2008. A review of the pharmacokinetics of abacavir. Clin Pharmacokinet 47:351–371. doi:10.2165/00003088-200847060-00001.
    1. Zhao W, Piana C, Danhof M, Burger D, Della Pasqua O, Jacqz-Aigrain E. 2013. Population pharmacokinetics of abacavir in infants, toddlers and children. Br J Clin Pharmacol 75:1525–1535. doi:10.1111/bcp.12024.
    1. Lindsey JC, Hughes MD, Violari A, Eshleman SH, Abrams EJ, Bwakura-Dangarembizi M, Barlow-Mosha L, Kamthunzi P, Sambo PM, Cotton MF, Moultrie H, Khadse S, Schimana W, Bobat R, Zimmer B, Petzold E, Mofenson LM, Jean-Philippe P, Palumbo P, P1060 Study Team. 2014. Predictors of virologic and clinical response to nevirapine versus lopinavir/ritonavir-based antiretroviral therapy in young children with and without prior nevirapine exposure for the prevention of mother-to-child HIV transmission. Pediatr Infect Dis J 33:846–854. doi:10.1097/INF.0000000000000337.
    1. Violari A, Lindsey JC, Hughes MD, Mujuru HA, Barlow-Mosha L, Kamthunzi P, Chi BH, Cotton MF, Moultrie H, Khadse S, Schimana W, Bobat R, Purdue L, Eshleman SH, Abrams EJ, Millar L, Petzold E, Mofenson LM, Jean-Philippe P, Palumbo P. 2012. Nevirapine versus ritonavir-boosted lopinavir for HIV-infected children. N Engl J Med 366:2380–2389. doi:10.1056/NEJMoa1113249.
    1. Kumar GN, Jayanti VK, Johnson MK, Uchic J, Thomas S, Lee RD, Grabowski BA, Sham HL, Kempf DJ, Denissen JF, Marsh KC, Sun E, Roberts SA. 2004. Metabolism and disposition of the HIV-1 protease inhibitor lopinavir (ABT-378) given in combination with ritonavir in rats, dogs, and humans. Pharm Res 21:1622–1630. doi:10.1023/b:pham.0000041457.64638.8d.
    1. Perloff MD, Von Moltke LL, Marchand JE, Greenblatt DJ. 2001. Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line. J Pharm Sci 90:1829–1837. doi:10.1002/jps.1133.
    1. Gordon LA, Malati CY, Hadigan C, McLaughlin M, Alfaro RM, Calderón MM, Kovacs JA, Penzak SR. 2016. Lack of an effect of ritonavir alone and lopinavir-ritonavir on the pharmacokinetics of fenofibric acid in healthy volunteers. Pharmacotherapy 36:49–56. doi:10.1002/phar.1682.
    1. Mugundu G, Hariparsad N, Desai P. 2010. Impact of ritonavir, atazanavir and their combination on the CYP3A4 induction by efavirenz in primary human hepatocytes. Drug Metab Lett 4:45–50. doi:10.2174/187231210790980453.
    1. Fahmi OA, Ripp SL. 2010. Evaluation of models for predicting drug–drug interactions due to induction. Expert Opin Drug Metab Toxicol 6:1399–1416. doi:10.1517/17425255.2010.516251.
    1. Fahmi OA, Kish M, Boldt S, Obach RS. 2010. Cytochrome P450 3A4 mRNA is a more reliable marker than CYP3A4 activity for detecting pregnane X receptor-activated induction of drug-metabolizing enzymes. Drug Metab Dispos 38:1605–1611. doi:10.1124/dmd.110.033126.
    1. Waters LJ, Moyle G, Bonora S, D'Avolio A, Else L, Mandalia S, Pozniak A, Nelson M, Gazzard B, Back D, Boffito M. 2007. Abacavir plasma pharmacokinetics in the absence and presence of atazanavir/ritonavir or lopinavir/ritonavir and vice versa in HIV-infected patients. Antivir Ther 12:825–830.
    1. B-Lajoie M-R, Drouin O, Bartlett G, Nguyen Q, Low A, Gavriilidis G, Easterbrook P, Muhe L. 2016. Incidence and prevalence of opportunistic and other infections and the impact of antiretroviral therapy among HIV-infected children in low- and middle-income countries: a systematic review and meta-analysis. Clin Infect Dis 62:1586–1594. doi:10.1093/cid/ciw139.
    1. Dresser GK, Spence JD, Bailey DG. 2000. Pharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clin Pharmacokinet 38:41–57. doi:10.2165/00003088-200038010-00003.
    1. Mahatthanatrakul W, Nontaput T, Ridtitid W, Wongnawa M, Sunbhanich M. 2007. Rifampin, a cytochrome P450 3A inducer, decreases plasma concentrations of antipsychotic risperidone in healthy volunteers. J Clin Pharm Ther 32:161–167. doi:10.1111/j.1365-2710.2007.00811.x.
    1. Johnson LF, Davies M-A, Moultrie H, Sherman GG, Bland RM, Rehle TM, Dorrington RE, Newell M-L. 2012. The effect of early initiation of antiretroviral treatment in infants on pediatric AIDS mortality in South Africa a model-based analysis. Pediatr Infect Dis J 31:474–480. doi:10.1097/INF.0b013e3182456ba2.
    1. Rabie H, Denti P, Lee J, Masango M, Coovadia A, Pillay S, Liberty A, Simon F, McIlleron H, Cotton MF, Lallemant M. 2019. Lopinavir–ritonavir super-boosting in young HIV-infected children on rifampicin-based tuberculosis therapy compared with lopinavir–ritonavir without rifampicin: a pharmacokinetic modelling and clinical study. Lancet HIV 6:e32–e42. doi:10.1016/S2352-3018(18)30293-5.
    1. Jackson A, Moyle G, Dickinson L, Back D, Khoo S, Taylor J, Gedela K, Abongomera G, Gazzard B, Boffito M. 2012. Pharmacokinetics of abacavir and its anabolite carbovir triphosphate without and with darunavir/ritonavir or raltegravir in HIV-infected subjects. Antivir Ther 17:19–24. doi:10.3851/IMP1910.
    1. Mulenga V, Musiime V, Kekitiinwa A, Cook AD, Abongomera G, Kenny J, Chabala C, Mirembe G, Asiimwe A, Owen-Powell E, Burger D, McIlleron H, Klein N, Chintu C, Thomason MJ, Kityo C, Walker AS, Gibb DM, CHAPAS-3 Trial Team. 2016. Abacavir, zidovudine, or stavudine as paediatric tablets for African HIV-infected children (CHAPAS-3): an open-label, parallel-group, randomised controlled trial. Lancet Infect Dis 16:169–179. doi:10.1016/S1473-3099(15)00319-9.
    1. Hughes W, McDowell JA, Shenep J, Flynn P, Kline MW, Yogev R, Symonds W, Lou Y, Hetherington S. 1999. Safety and single-dose pharmacokinetics of abacavir (1592U89) in human immunodeficiency virus type 1-infected children. Antimicrob Agents Chemother 43:609–615. doi:10.1128/AAC.43.3.609.
    1. Cella M, Gorter de Vries F, Burger D, Danhof M, Della Pasqua O. 2010. A model-based approach to dose selection in early pediatric development. Clin Pharmacol Ther 87:294–302. doi:10.1038/clpt.2009.234.
    1. Van Heeswijk RPG, Bourbeau M, Seguin I, Giguere P, Garber GE, Cameron DW. 2005. Absence of circadian variation in the pharmacokinetics of lopinavir/ritonavir given as a once daily dosing regimen in HIV-1-infected patients. Br J Clin Pharmacol 59:398–404. doi:10.1111/j.1365-2125.2005.02337.x.
    1. Kline MW, Blanchard S, Fletcher CV, Shenep JL, McKinney RE, Brundage RC, Culnane M, Van Dyke RB, Dankner WM, Kovacs A, McDowell JA, Hetherington S. 1999. A phase I study of abacavir (1592U89) alone and in combination with other antiretroviral agents in infants and children with human immunodeficiency virus infection. Pediatrics 103:e47. doi:10.1542/peds.103.4.e47.
    1. Chittick GE, Gillotin C, McDowell JA, Lou Y, Edwards KD, Prince WT, Stein DS. 1999. Abacavir: absolute bioavailability, bioequivalence of three oral formulations, and effect of food. Pharmacotherapy 19:932–942. doi:10.1592/phco.19.11.932.31568.
    1. Ouellet D, Hsu A, Granneman GR, Carlson G, Cavanaugh J, Guenther H, Leonard JM. 1998. Pharmacokinetic interaction between ritonavir and clarithromycin. Clin Pharmacol Ther 64:355–362. doi:10.1016/S0009-9236(98)90065-0.
    1. Hsu A, Granneman GR, Witt G, Locke C, Denissen J, Molla A, Valdes J, Smith J, Erdman K, Lyons N, Niu P, Decourt JP, Fourtillan JB, Girault J, Leonard JM. 1997. Multiple-dose pharmacokinetics of ritonavir in human immunodeficiency virus-infected subjects. Antimicrob Agents Chemother 41:898–905. doi:10.1128/AAC.41.5.898.
    1. Bienczak A, Cook A, Wiesner L, Olagunju A, Mulenga V, Kityo C, Kekitiinwa A, Owen A, Walker AS, Gibb DM, McIlleron H, Burger D, Denti P. 2016. The impact of genetic polymorphisms on the pharmacokinetics of efavirenz in African children. Br J Clin Pharmacol 82:185–198. doi:10.1111/bcp.12934.
    1. Panhard X, Goujard C, Legrand M, Taburet AM, Diquet B, Mentré F, COPHAR 1-ANRS Study Group. 2005. Population pharmacokinetic analysis for nelfinavir and its metabolite M8 in virologically controlled HIV-infected patients on HAART. Br J Clin Pharmacol 60:390–403. doi:10.1111/j.1365-2125.2005.02456.x.
    1. Lemmer B, Nold G. 1991. Circadian changes in estimated hepatic blood flow in healthy subjects. Br J Clin Pharmacol 32:627–629. doi:10.1111/j.1365-2125.1991.tb03964.x.
    1. Moyle G, Boffito M, Fletcher C, Higgs C, Hay PE, Song IH, Lou Y, Yuen GJ, Min SS, Guerini EM. 2009. Steady-state pharmacokinetics of abacavir in plasma and intracellular carbovir triphosphate following administration of abacavir at 600 milligrams once daily and 300 milligrams twice daily in human immunodeficiency virus-infected subjects. Antimicrob Agents Chemother 53:1532–1538. doi:10.1128/AAC.01000-08.
    1. Boeckmann AJ, Beal SL, Sheiner LB. 1998. NONMEM user’s guide—part V. Introductory guide p 48 NONMEM Project Group, University of California at San Francisco, San Francisco, CA.
    1. Keizer RJ, Karlsson MO, Hooker A. 2013. Modeling and simulation workbench for NONMEM: tutorial on Pirana, PsN, and Xpose. CPT Pharmacometrics Syst Pharmacol 2:e50. doi:10.1038/psp.2013.24.
    1. Beal SL. 2001. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn 28:481–504. doi:10.1023/A:1012299115260.
    1. Anderson BJ, Holford N. 2008. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 48:303–332. doi:10.1146/annurev.pharmtox.48.113006.094708.
    1. World Health Organization. 2007. WHO child growth standards: head circumference-for-age, arm circumference-for-age, triceps skinfold-for-age and subscapular skinfold-for-age: methods and development. World Health Organization, Geneva, Switzerland.
    1. Dosne A-G, Bergstrand M, Harling K, Karlsson MO. 2016. Improving the estimation of parameter uncertainty distributions in nonlinear mixed effects models using sampling importance resampling. J Pharmacokinet Pharmacodyn 43:583–596. doi:10.1007/s10928-016-9487-8.
    1. Svensson EM, Yngman G, Denti P, McIlleron H, Kjellsson MC, Karlsson MO. 2018. Evidence-based design of fixed-dose combinations: principles and application to pediatric anti-tuberculosis therapy. Clin Pharmacokinet 57:591–599. doi:10.1007/s40262-017-0577-6.
    1. Weller S, Radomski KM, Lou Y, Daniel S, Stein DS. 2000. Population pharmacokinetics and pharmacodynamic modeling of abacavir trial with human immunodeficiency virus-infected subjects population pharmacokinetics and pharmacodynamic modeling of abacavir (1592U89) from a dose-ranging, double-blind, randomized. Antimicrob Agents Chemother 44:2052–2060. doi:10.1128/aac.44.8.2052-2060.2000.

Source: PubMed

3
Abonner