Model-Based Meta-Analysis of Relapsing Mouse Model Studies from the Critical Path to Tuberculosis Drug Regimens Initiative Database

Alexander Berg, James Clary, Debra Hanna, Eric Nuermberger, Anne Lenaerts, Nicole Ammerman, Michelle Ramey, Dan Hartley, David Hermann, Alexander Berg, James Clary, Debra Hanna, Eric Nuermberger, Anne Lenaerts, Nicole Ammerman, Michelle Ramey, Dan Hartley, David Hermann

Abstract

Tuberculosis (TB), the disease caused by Mycobacterium tuberculosis (Mtb), remains a leading infectious disease-related cause of death worldwide, necessitating the development of new and improved treatment regimens. Nonclinical evaluation of candidate drug combinations via the relapsing mouse model (RMM) is an important step in regimen development, through which candidate regimens that provide the greatest decrease in the probability of relapse following treatment in mice may be identified for further development. Although RMM studies are a critical tool to evaluate regimen efficacy, making comprehensive "apples to apples" comparisons of regimen performance in the RMM has been a challenge in large part due to the need to evaluate and adjust for variability across studies arising from differences in design and execution. To address this knowledge gap, we performed a model-based meta-analysis on data for 17 unique regimens obtained from a total of 1592 mice across 28 RMM studies. Specifically, a mixed-effects logistic regression model was developed that described the treatment duration-dependent probability of relapse for each regimen and identified relevant covariates contributing to interstudy variability. Using the model, covariate-normalized metrics of interest, namely, treatment duration required to reach 50% and 10% relapse probability, were derived and used to compare relative regimen performance. Overall, the model-based meta-analysis approach presented herein enabled cross-study comparison of efficacy in the RMM and provided a framework whereby data from emerging studies may be analyzed in the context of historical data to aid in selecting candidate drug combinations for clinical evaluation as TB drug regimens.

Keywords: Mycobacterium; model-based meta-analysis; modeling and simulation; relapsing mouse model; tuberculosis.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

FIG 1
FIG 1
Relapse proportion by treatment duration across stratified by regimen and study. Each study is grouped with lines representing the relapse-time course in a particular study. Points represent individual time points in a particular study. “RMZ/RM_BIDM” denotes a version of the RMZ/RM regimen where moxifloxacin was administered at 100 mg/kg twice daily for a total daily dose of 200 mg/kg. M, moxifloxacin; R, rifampin; Z, pyrazinamide.
FIG 2
FIG 2
Visual predictive check for the final model stratified by regimen. Black dots and solid lines represent the observed relapse proportion. Blue lines represent the median prediction from the final model. The red shaded area represents the 90% prediction interval. “RMZ/RM_BIDM” denotes a version of the RMZ/RM regimen where moxifloxacin was administered at 100 mg/kg twice daily for a total daily dose of 200 mg/kg. M, moxifloxacin; R, rifampin; Z, pyrazinamide.
FIG 3
FIG 3
Relapse probability versus treatment duration by regimen. Blue lines and areas represent the median and 95% confidence interval, respectively, of relapse probability versus treatment duration profiles for each regimen as derived by bootstrap (N = 500 runs). Black lines and areas represent HRZE/HR as the clinical standard of care regimen. For comparative purposes, all regimens are presented as normalized to the median covariate values. “RMZ/RM_BIDM” denotes a version of the RMZ/RM regimen where moxifloxacin was administered at 100 mg/kg twice daily for a total daily dose of 200 mg/kg. E, ethambutol; H, isoniazid; M, moxifloxacin; R, rifampin; Z, pyrazinamide.
FIG 4
FIG 4
Forest plots of T50 and T10 relapse probability by regimen. Median estimates and 95% confidence intervals from bootstrapped data sets (n = 500 runs) for (A) T50 and (B) T10 metrics. Regimens are in descending rank orders for metrics based on median value, with red and blue coloring indicating regimens for which the confidence intervals are completely above or below the median value for HRZE/HR (included as the clinical standard of care regimen). For comparative purposes, all regimens are presented as normalized to the median covariate values. “RMZ/RM_BIDM” denotes a version of the RMZ/RM regimen where moxifloxacin was administered at 100 mg/kg twice daily for a total daily dose of 200 mg/kg. E, ethambutol; H, isoniazid; M, moxifloxacin; R, rifampin; Z, pyrazinamide.
FIG 5
FIG 5
Simulations of the model with a range of covariate effects. Simulations of covariate effects from range of data for (A) inoculum (log10 CFU/mL) and (B) baseline bacterial burden (log10 CFU/mL).

References

    1. Harding E. 2020. WHO global progress report on tuberculosis elimination. Lancet Respir Med 8:19. 10.1016/S2213-2600(19)30418-7.
    1. World Health Organization. 2018. Latent TB Infection: updated and consolidated guidelines for programmatic management. .
    1. World Health Organization, Global Tuberculosis Programme. 2020. Global tuberculosis report. .
    1. European Medicines Agency. 2013. Committee for Medicinal Products for Human Use. Summary of Opinion: Deltyba. .
    1. Food and Drug Administration. 2019. Pretomanid product insert. Silver Spring (MD). .
    1. Food and Drug Administration. 2012. Sirturo (bedaquiline) product insert. Silver Spring (MD). .
    1. Combs DL, O'Brien RJ, Geiter LJ. 1990. USPHS Tuberculosis Short-Course Chemotherapy Trial 21: effectiveness, toxicity, and acceptability. The report of final results. Ann Intern Med 112:397–406. 10.7326/0003-4819-76-3-112-6-397.
    1. Critical Path Institute. 2017. Critical path to TB drug regimens. Available from: .
    1. Imperial MZ, Nahid P, Phillips PPJ, Davies GR, Fielding K, Hanna D, Hermann D, Wallis RS, Johnson JL, Lienhardt C, Savic RM. 2018. A patient-level pooled analysis of treatment-shortening regimens for drug-susceptible pulmonary tuberculosis. Nat Med 24:1708–1715. 10.1038/s41591-018-0224-2.
    1. Gumbo T, Lenaerts AJ, Hanna D, Romero K, Nuermberger E. 2015. Nonclinical models for antituberculosis drug development: a landscape analysis. J Infect Dis 211 Suppl 3:S83–S95. 10.1093/infdis/jiv183.
    1. Franzblau SG, DeGroote MA, Cho SH, Andries K, Nuermberger E, Orme IM, Mdluli K, Angulo-Barturen I, Dick T, Dartois V, Lenaerts AJ. 2012. Comprehensive analysis of methods used for the evaluation of compounds against Mycobacterium tuberculosis. Tuberculosis (Edinb) 92:453–488. 10.1016/j.tube.2012.07.003.
    1. De Groote MA, Gruppo V, Woolhiser LK, Orme IM, Gilliland JC, Lenaerts AJ. 2012. Importance of confirming in vivo efficacy data of novel antibacterial drug regimens against various strains of Mycobacterium tuberculosis. Antimicrob Agents Chemother 56:731–738. 10.1128/AAC.05701-11.
    1. De Groote MA, Gilliland JC, Wells CL, Brooks EJ, Woolhiser LK, Gruppo V, Peloquin CA, Orme IM, Lenaerts AJ. 2011. Comparative studies evaluating mouse models used for efficacy testing of experimental drugs against M. tuberculosis. Antimicrob Agents Chemother 55:1237–1247. 10.1128/AAC.00595-10.
    1. Lenaerts AJ, Chapman PL, Orme IM. 2004. Statistical limitations to the Cornell model of latent tuberculosis infection for the study of relapse rates. Tuberculosis (Edinb) 84:361–364. 10.1016/j.tube.2004.03.002.
    1. Mourik BC, Svensson RJ, de Knegt GJ, Bax HI, Verbon A, Simonsson USH, de Steenwinkel JEM. 2018. Improving treatment outcome assessment in a mouse tuberculosis model. Sci Rep 8:5714. 10.1038/s41598-018-24067-x.
    1. Mudde SE, Alsoud RA, van der Meijden A, Upton AM, Lotlikar MU, Simonsson USH, Bax HI, de Steenwinkel JEM. 2021. Predictive modeling to study the treatment-shortening potential of novel tuberculosis drug regimens, toward bundling of preclinical data. J Infect Dis jiab101.
    1. Nuermberger EL. 2017. Preclinical efficacy testing of new drug candidates. Microbiol Spectr 5. 10.1128/microbiolspec.TBTB2-0034-2017.
    1. Nuermberger EL, Yoshimatsu T, Tyagi S, Williams K, Rosenthal I, O'Brien RJ, Vernon AA, Chaisson RE, Bishai WR, Grosset JH. 2004. Moxifloxacin-containing regimens of reduced duration produce a stable cure in murine tuberculosis. Am J Respir Crit Care Med 170:1131–1134. 10.1164/rccm.200407-885OC.
    1. Nuermberger EL, Yoshimatsu T, Tyagi S, O'Brien RJ, Vernon AN, Chaisson RE, Bishai WR, Grosset JH. 2004. Moxifloxacin-containing regimen greatly reduces time to culture conversion in murine tuberculosis. Am J Respir Crit Care Med 169:421–426. 10.1164/rccm.200310-1380OC.
    1. Li SY, Irwin SM, Converse PJ, Mdluli KE, Lenaerts AJ, Nuermberger EL. 2015. Evaluation of moxifloxacin-containing regimens in pathologically distinct murine tuberculosis models. Antimicrob Agents Chemother 59:4026–4030. 10.1128/AAC.00105-15.
    1. Wallis RS, Wang C, Meyer D, Thomas N. 2013. Month 2 culture status and treatment duration as predictors of tuberculosis relapse risk in a meta-regression model. PLoS One 8:e71116. 10.1371/journal.pone.0071116.
    1. Lanoix JP, Chaisson RE, Nuermberger EL. 2016. Shortening tuberculosis treatment with fluoroquinolones: lost in translation? Clin Infect Dis 62:484–490. 10.1093/cid/civ911.
    1. Dorman SE, Nahid P, Kurbatova EV, Phillips PPJ, Bryant K, Dooley KE, Engle M, Goldberg SV, Phan HTT, Hakim J, Johnson JL, Lourens M, Martinson NA, Muzanyi G, Narunsky K, Nerette S, Nguyen NV, Pham TH, Pierre S, Purfield AE, Samaneka W, Savic RM, Sanne I, Scott NA, Shenje J, Sizemore E, Vernon A, Waja Z, Weiner M, Swindells S, Chaisson RE, Tuberculosis Trials Consortium. 2021. Four-month rifapentine regimens with or without moxifloxacin for tuberculosis. N Engl J Med 384:1705–1718. 10.1056/NEJMoa2033400.
    1. Saini V, Ammerman NC, Chang YS, Tasneen R, Chaisson RE, Jain S, Nuermberger E, Grosset JH. 2019. Treatment-shortening effect of a novel regimen combining clofazimine and high-dose rifapentine in pathologically distinct mouse models of tuberculosis. Antimicrob Agents Chemother 63:e00388-19. 10.1128/AAC.00388-19.
    1. Ammerman NC, Swanson RV, Bautista EM, Almeida DV, Saini V, Omansen TF, Guo H, Chang YS, Li SY, Tapley A, Tasneen R, Tyagi S, Betoudji F, Moodley C, Ngcobo B, Pillay L, Bester LA, Singh SD, Chaisson RE, Nuermberger E, Grosset JH. 2018. Impact of clofazimine dosing on treatment shortening of the first-line regimen in a mouse model of tuberculosis. Antimicrob Agents Chemother 62:e00636-18. 10.1128/AAC.00636-18.
    1. Tyagi S, Ammerman NC, Li S-Y, Adamson J, Converse PJ, Swanson RV, Almeida DV, Grosset JH. 2015. Clofazimine shortens the duration of the first-line treatment regimen for experimental chemotherapy of tuberculosis. Proc Natl Acad Sci USA 112:869–874. 10.1073/pnas.1416951112.
    1. Rosenthal IM, Zhang M, Williams KN, Peloquin CA, Tyagi S, Vernon AA, Bishai WR, Chaisson RE, Grosset JH, Nuermberger EL. 2007. Daily dosing of rifapentine cures tuberculosis in three months or less in the murine model. PLoS Med 4:e344. 10.1371/journal.pmed.0040344.
    1. Lanoix J-P, Betoudji F, Nuermberger E. 2016. Sterilizing activity of pyrazinamide in combination with first-line drugs in a C3HeB/FeJ mouse model of tuberculosis. Antimicrob Agents Chemother 60:1091–1096. 10.1128/AAC.02637-15.
    1. Tasneen R, Li S-Y, Peloquin CA, Taylor D, Williams KN, Andries K, Mdluli KE, Nuermberger EL. 2011. Sterilizing activity of novel TMC207- and PA-824-containing regimens in a murine model of tuberculosis. Antimicrob Agents Chemother 55:5485–5492. 10.1128/AAC.05293-11.
    1. Nuermberger E, Tyagi S, Tasneen R, Williams KN, Almeida D, Rosenthal I, Grosset JH. 2008. Powerful bactericidal and sterilizing activity of a regimen containing PA-824, moxifloxacin, and pyrazinamide in a murine model of tuberculosis. Antimicrob Agents Chemother 52:1522–1524. 10.1128/AAC.00074-08.
    1. Tasneen R, Betoudji F, Tyagi S, Li S-Y, Williams K, Converse PJ, Dartois V, Yang T, Mendel CM, Mdluli KE, Nuermberger EL. 2016. Contribution of oxazolidinones to the efficacy of novel regimens containing bedaquiline and pretomanid in a mouse model of tuberculosis. Antimicrob Agents Chemother 60:270–277. 10.1128/AAC.01691-15.
    1. Akaike H. 1973. Information theory and an extension of the maximum likelihood principle, p 267–281. In Petrov BN, Csáki F (ed), 2nd International Symposium on Information Theory, Tsahkadsor, Armenia, USSR, September 2–8, 1971, Budapest: Akadémiai Kiadó. Republished in 1992. p 610–24, Kotz S, Johnson NL (ed), Breakthroughs in Statistics, I, Springer-Verlag.
    1. R Core Team. 2017. R: a language and environment for statistical computing. R Vienna, Austria: Foundation for Statistical Computing. .
    1. RStudio Team. 2020. RStudio: Integrated Development for R. RStudio, PBC, Boston, MA. .
    1. Bates D, Maechler M, Bolker B, Walker S. 2015. Fitting linear mixed-effects models using lme4. J Stat Softw 67:1–48.
    1. Dardis C. 2015. LogisticDx: Diagnostic tests for models with a binomial response. R package Version 0.2. .
    1. Wickham H. 2016. ggplot2: elegant Graphics for Data Analysis, p. 276, 2nd ed. New York, NY: Springer.

Source: PubMed

3
Se inscrever