Characterizing Absorption Properties of Dispersible Pretomanid Tablets Using Population Pharmacokinetic Modelling

Yuanxi Zou, Jerry Nedelman, Antonio Lombardi, Frances Pappas, Mats O Karlsson, Elin M Svensson, Yuanxi Zou, Jerry Nedelman, Antonio Lombardi, Frances Pappas, Mats O Karlsson, Elin M Svensson

Abstract

Background and introduction: The dispersible tablet formulation (DTF) of pretomanid has been developed to facilitate future use in children. This work aimed to assess the pharmacokinetics (PK) and relative bioavailability of the DTF compared to the marketed formulation (MF) and the potential influence of dose.

Methods: Pretomanid DTF was investigated in a single-dose, randomized, four-period, cross-over study, with 7 days of washout between doses. Forty-eight healthy volunteers were enrolled and randomized into one of two panels to receive doses either in the fasted state or after a high-fat meal. Each volunteer received doses of 10, 50, and 200 mg DTF, and 200 mg MF pretomanid. Blood samples for pharmacokinetic assessment were drawn following a rich schedule up to 96 h after each single dose. The study data from the panel receiving the high-fat meal were analyzed using a nonlinear mixed-effects modeling approach, and all data were characterized with noncompartmental methods.

Results: A one-compartment model with first-order elimination and absorption through a transit compartment captured the mean and variability of the observed pretomanid concentrations with acceptable precision. No significant difference in bioavailability was found between formulations. The mean absorption time for the DTF was typically 137% (86-171%) of that for the MF. The bioavailability was found to be dose dependent with a small positive and larger negative correlation under fed and fasted conditions, respectively.

Conclusion: Using data from a relative bioavailability study in healthy adult volunteers, a mathematical model has been developed to inform dose selection for the investigation of pretomanid in children using the new dispersible tablet formulation. Under fed conditions and at the currently marketed adult dose of 200 mg, the formulation type was found to influence the absorption rate, but not the bioavailability. The bioavailability of the DTF was slightly positively correlated with doses when administered with food.

Clinical trial registration: ClinicalTrials.gov Identifier: NCT04309656, first posted on 16 March 2020.

Conflict of interest statement

The authors declare that they have no competing interests. JN, AL, and FP are employees of TB Alliance, the organization developing pretomanid. ES is involved in development of multiple novel anti-tuberculosis compounds through the UNITE4TB consortium.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Mean plasma concentrations versus time after dose per dose and formulation of pretomanid at fed condition. Treatment A, 200 mg marketed formulation; B, 200 mg dispersible tablet formulation; C, 50 mg dispersible tablet formulation; D, 10 mg dispersible tablet formulation
Fig. 2
Fig. 2
Visual predictive checks of the final model on the CL-011 study data (fed conditions only) by formulations and doses from time after dose to 96 h. The concentrations are plotted on log scale. The open circles represent the observed samples. The solid and dashed black lines represent the 50th and 10th/90th percentiles of the observations in the data set. The red- and blue-colored areas represent the 95% model-predicted confidence intervals for the corresponding percentiles
Fig. 3
Fig. 3
Histogram of the simulated population of the AUC0–last from 300 simulations, compared with the observed AUC (red vertical lines), under fed state in the CL-011 study. The distribution of simulated AUC is represented by the 2.5th, 50th, and 97.5th percentiles in grey lines

References

    1. Global tuberculosis report 2021. Geneva: World Health Organization. Published online 2021. licence: CC BY-NC-SA 3.0 IGO.
    1. WHO updated definitions of extensively drug-resistant tuberculosis. 2021. . Accessed 6 Jan 2022.
    1. Manjunatha U, Boshoff HIM, Barry CE. The mechanism of action of PA-824: novel insights from transcriptional profiling. Commun Integr Biol. 2009;2(3):215. doi: 10.4161/CIB.2.3.7926.
    1. FDA Approves New Drug for Treatment-Resistant Forms of Tuberculosis That Affects the Lungs | FDA.; 2019. . Accessed 21 Nov 2021
    1. Dovprela (previously Pretomanid FGK) | European Medicines Agency. Published online 2020. . Accessed 21 Nov 2021
    1. Jenkins HE, Tolman AW, Yuen CM, et al. Incidence of multidrug-resistant tuberculosis disease in children: systematic review and global estimates. Lancet. 2014;383(9928):1572–1579. doi: 10.1016/S0140-6736(14)60195-1.
    1. Dodd PJ, Sismanidis C, Seddon JA. Global burden of drug-resistant tuberculosis in children: a mathematical modelling study. Lancet Infect Dis. 2016;16(10):1193–1201. doi: 10.1016/S1473-3099(16)30132-3.
    1. Jenkins HE, Yuen CM. The burden of multidrug-resistant tuberculosis in children. Int J Tuberc Lung Dis. 2018;22(5):S3–S6. doi: 10.5588/ijtld.17.0357.
    1. Keam SJ. Pretomanid: first approval. Drugs. 2019;79:1797–1803. doi: 10.1007/s40265-019-01207-9.
    1. Salinger DH, Subramoney V, Everitt D, Nedelman JR. Population pharmacokinetics of the antituberculosis agent pretomanid. Antimicrob Agents Chemother. 2019 doi: 10.1128/AAC.00907-19.
    1. FDA. Pretomanid Tablets label. Published online 2019. . Accessed 28 Nov 2021
    1. Savic RM, Jonker DM, et al. Implementation of a transit compartment model for describing drug absorption in pharmacokinetic studies. J Pharmacokinet Pharmacodyn. 2007;34:711–726. doi: 10.1007/s10928-007-9066-0.
    1. Anderson BJ, Holford NHG. Mechanistic basis of using body size and maturation to predict clearance in humans. Drug Metab Pharmacokinet. 2009;24(1):25–36. doi: 10.2133/dmpk.24.25.
    1. John JA, Draper NR. An alternative family of transformations. Appl Stat. 1980;29(2):190. doi: 10.2307/2986305.
    1. Box GEP, Cox DR. An analysis of transformations. Source J R Stat Soc Ser B. 1964;26(2):211–52. . Accessed 6 Jan 2022.
    1. Collins JJ, Fanciulli M, Hohlfeld RG, Finch DC, Sandri GVH, Shtatland ES. A random number generator based on the logit transform of the logistic variable. Comput Phys. 1998;6(6):630. doi: 10.1063/1.168442.
    1. Karlsson MO, Freiberga S, Yngman G, Nordgren R, Ueckert S. Extensive and automatic assumption assessment of pharmacometric models. PAGE 27 abstract 8754. Published 2018. . Accessed 6 May 2021.
    1. Lindbom L, Pihlgren P, Jonsson N. PsN-Toolkit—a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed. 2005;79(3):241–257. doi: 10.1016/j.cmpb.2005.04.005.
    1. Jonsson EN, Karlsson MO. Xpose—an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed. 1998;58(1):51–64. doi: 10.1016/S0169-2607(98)00067-4.
    1. Keizer RJ, van Benten M, Beijnen JH, Schellens JHM, Huitema ADR. Piraña and PCluster: a modeling environment and cluster infrastructure for NONMEM. Comput Methods Programs Biomed. 2011;101(1):72–79. doi: 10.1016/j.cmpb.2010.04.018.
    1. Karlsson MO, Holford N. A tutorial on visual predictive checks. . Accessed 6 May 2021.
    1. Acharya C, Hooker AC, Türkyılmaz GY, Jönsson 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. doi: 10.1016/j.cmpb.2016.01.013.
    1. FDA, Cder, Purdief. Guidance for Industry Bioavailability and Bioequivalence Studies Submitted in NDAs or INDs-General Considerations; 2014. . Accessed 21 Nov 2019.
    1. Winter H, Ginsberg A, Egizi E, et al. Effect of a high-calorie, high-fat meal on the bioavailability and pharmacokinetics of PA-824 in healthy adult subjects. Antimicrob Agents Chemother. 2013;57(11):5516. doi: 10.1128/AAC.00798-13.
    1. Karlsson MO, Beal SL, Sheiner LB. Three new residual error models for population PK/PD analyses. J Pharmacokinet Biopharm. 1995;23(6):651–672. doi: 10.1007/BF02353466.
    1. Ahmad Z, Peloquin CA, Singh RP, et al. PA-824 exhibits time-dependent activity in a murine model of tuberculosis. Antimicrob Agents Chemother. 2011;55(1):239. doi: 10.1128/AAC.00849-10.
    1. Dubois A, Gsteiger S, Pigeolet E, Mentré F. Bioequivalence tests based on individual estimates using non-compartmental or model-based analyses: Evaluation of estimates of sample means and type i error for different designs. Pharm Res. 2010;27(1):92–104. doi: 10.1007/s11095-009-9980-5.
    1. Holford N, Heo YA, Anderson B. A pharmacokinetic standard for babies and adults. J Pharm Sci. 2013;102(9):2941–2952. doi: 10.1002/jps.23574.

Source: PubMed

3
Sottoscrivi