Bedaquiline and Pyrazinamide Treatment Responses Are Affected by Pulmonary Lesion Heterogeneity in Mycobacterium tuberculosis Infected C3HeB/FeJ Mice

Scott M Irwin, Brendan Prideaux, Edward R Lyon, Matthew D Zimmerman, Elizabeth J Brooks, Christopher A Schrupp, Chao Chen, Matthew J Reichlen, Bryce C Asay, Martin I Voskuil, Eric L Nuermberger, Koen Andries, Michael A Lyons, Véronique Dartois, Anne J Lenaerts, Scott M Irwin, Brendan Prideaux, Edward R Lyon, Matthew D Zimmerman, Elizabeth J Brooks, Christopher A Schrupp, Chao Chen, Matthew J Reichlen, Bryce C Asay, Martin I Voskuil, Eric L Nuermberger, Koen Andries, Michael A Lyons, Véronique Dartois, Anne J Lenaerts

Abstract

BALB/c and Swiss mice are routinely used to validate the effectiveness of tuberculosis drug regimens, although these mouse strains fail to develop human-like pulmonary granulomas exhibiting caseous necrosis. Microenvironmental conditions within human granulomas may negatively impact drug efficacy, and this may not be reflected in non-necrotizing lesions found within conventional mouse models. The C3HeB/FeJ mouse model has been increasingly utilized as it develops hypoxic, caseous necrotic granulomas which may more closely mimic the pathophysiological conditions found within human pulmonary granulomas. Here, we examined the treatment response of BALB/c and C3HeB/FeJ mice to bedaquiline (BDQ) and pyrazinamide (PZA) administered singly and in combination. BALB/c mice consistently displayed a highly uniform treatment response to both drugs, while C3HeB/FeJ mice displayed a bimodal response composed of responsive and less-responsive mice. Plasma pharmacokinetic analysis of dissected lesions from BALB/c and C3HeB/FeJ mice revealed that PZA penetrated lesion types from both mouse strains with similar efficiency. However, the pH of the necrotic caseum of C3HeB/FeJ granulomas was determined to be 7.5, which is in the range where PZA is essentially ineffective under standard laboratory in vitro growth conditions. BDQ preferentially accumulated within the highly cellular regions in the lungs of both mouse strains, although it was present at reduced but still biologically relevant concentrations within the central caseum when dosed at 25 mg/kg. The differential treatment response which resulted from the heterogeneous pulmonary pathology in the C3HeB/FeJ mouse model revealed several factors which may impact treatment efficacy, and could be further evaluated in clinical trials.

Keywords: C3HeB/FeJ; bedaquiline; mouse models; pyrazinamide; tuberculosis.

Conflict of interest statement

The authors declare the following competing financial interest(s): K.A. is an employee of Janssen Pharmaceutica, the company that developed bedaquiline.

Figures

Figure 1
Figure 1
PZA activity in BALB/c and C3HeB/FeJ mice. Plots represent log10 CFU determinations from the lungs of individual BALB/c (A, B) or C3HeB/FeJ (C, D) mice. Eight weeks following LDA infection, mice were treated for 3, 5, or 8 weeks with 150 (A, C) or 300 mg/kg (B, D) PZA via oral gavage. Pre-Rx represents untreated mice aerosol infected for 8 weeks. Ovals with solid lines represent the PZA less-responsive subpopulation, and dashed lines represent the PZA responsive subpopulation of C3HeB/FeJ mice. Pre-Rx and defined time points were determined to be statistically different (*; P < 0.01) using a one-way ANOVA. Responsive and less-responsive populations were determined to be statistically different (P < 0.001) using a two-way ANOVA.
Figure 2
Figure 2
BDQ exhibited dose-dependent activity between 10 and 25 mg/kg. Plots represent log10 CFU determinations from the lungs of individual BALB/c (A, B) or C3HeB/FeJ (C, D) mice. Eight weeks following LDA infection, mice were treated with BDQ 10 mg/kg (A, C) or 25 mg/kg (B, D) via oral gavage. BALB/c mice were treated for either 2 or 4 weeks. C3HeB/FeJ mice were treated for either 2, 4, or 8 weeks. Pre-Rx represents untreated mice aerosol infected for 8 weeks. Ovals with solid lines represent the BDQ less-responsive subpopulation, and dashed lines represent the BDQ responsive subpopulation of C3HeB/FeJ mice. Pre-Rx and defined time points were determined to be statistically different (*; P < 0.01) using a one-way ANOVA. Responsive and less-responsive populations were determined to be statistically different (P < 0.001) using a two-way ANOVA.
Figure 3
Figure 3
BDQ/PZA combination therapy had activity against both subpopulations in C3HeB/FeJ mice. Plots represent log10 CFU determinations from the lungs of individual BALB/c (A) or C3HeB/FeJ (B) mice. Pre-Rx represents untreated mice 8 weeks following aerosol infection. Mice were treated with BDQ 10 mg/kg and PZA 150 mg/kg in combination via oral gavage. BALB/c mice were treated for either 2 or 4 weeks. C3HeB/FeJ mice were treated for either 2, 4, or 8 weeks. Ovals with solid lines represent the less-responsive subpopulation, and dashed lines represent the responsive subpopulation of C3HeB/FeJ mice. Pre-Rx and defined time points were determined to be statistically different (*; P < 0.01) using a one-way ANOVA. Responsive and less-responsive populations were determined to be statistically different (P < 0.001) using a two-way ANOVA.
Figure 4
Figure 4
Plasma and lung tissue concentration–time profiles of PZA and POA in BALB/c and C3HeB/FeJ mice following a single 150 mg/kg PZA oral dose. Data points indicate mean values (n = 5 mice per time point) with SD error bars.
Figure 5
Figure 5
Plasma and lung tissue concentration–time profiles of BDQ and M2 in BALB/c and C3HeB/FeJ mice following a single 25 mg/kg BDQ oral dose. Data points indicate mean values (n = 5 mice per time point) with SD error bars.
Figure 6
Figure 6
Arithmetic mean and SD for BALB/c and C3HeB/FeJ plasma and lung tissue population PK parameters for PZA and POA. Panel A shows a comparison of absorption (ka), clearance (CL), and volume of distribution (V) parameters for PZA and POA in both mouse strains. Panel B shows a comparison of half-life (t1/2) and penetration (PC) coefficient for PZA and POA in both mouse strains. Histogram height represents the arithmetic mean, with error bars representing mean ± SD. The tissue equilibrium half-life was calculated as t1/2 = ln(2)/k, where k is the corresponding tissue penetration rate. All values were calculated from the corresponding GM and GSD listed in Supplemental Table 1.
Figure 7
Figure 7
Arithmetic mean and SD for BALB/c and C3HeB/FeJ plasma and lung tissue population PK parameters for BDQ and M2. Panel A shows a comparison of absorption (ka), clearance (CL), and volume of distribution (V) parameters for BDQ and M2 in both mouse strains. Panel B shows a comparison of half-life (t1/2) and penetration (PC) coefficient for BDQ and M2 in both mouse strains. Histogram height represents the arithmetic mean, with error bars representing mean ± SD. The tissue equilibrium half-life was calculated as t1/2 = ln(2)/k where k is the corresponding tissue penetration rate. All values were calculated from the corresponding GM and GSD listed in Supplemental Table 2.
Figure 8
Figure 8
MALDI-MS time course images of PZA and POA in BALB/c mice acquired 0.08–8 h following a single oral 150 mg/kg dose of PZA. Cellular, non-necrotizing lesions (type III) are outlined in white. Images depict a single lung lobe obtained from a representative mouse (n = 4 mice per time point). A serial section was stained with hematoxylin and eosin (H&E) for comparison.
Figure 9
Figure 9
MALDI-MS time course images of PZA and POA in C3HeB/FeJ mice acquired 0.08–8 h following a single oral 150 mg/kg dose of PZA. Caseous necrotic granulomas (type I) are outlined in white. Images depict a single lung lobe obtained from a representative mouse (n = 4 mice per time point). A serial section was stained with hematoxylin and eosin (H&E) for comparison.
Figure 10
Figure 10
MALDI-MS time course images of BDQ and M2 in BALB/c mice acquired 8–168 h following a single oral 25 mg/kg dose of BDQ. Cellular, non-necrotizing lesions (type III) are outlined in white. Images depict a single lung lobe obtained from a representative mouse (n = 4 mice per time point). A serial section was stained with hematoxylin and eosin (H&E) for comparison.
Figure 11
Figure 11
MALDI-MS time course images of BDQ and M2 in C3HeB/FeJ mice acquired 8–168 h following a single oral 25 mg/kg dose of BDQ. Caseous necrotic granulomas (type I) are outlined in white. Images depict a single lung lobe obtained from a representative mouse (n = 4 mice per time point). A serial section was stained with hematoxylin and eosin (H&E) for comparison.
Figure 12
Figure 12
Reduced penetration of BDQ into caseous necrotic granulomas. Light micrograph of a caseous necrotic granuloma (A; arrow) and a corresponding MALDI-MS image (B) acquired with a saturated intensity scale showing no detectable BDQ within the central caseum of the granuloma. Representative MALDI-MS image of a single lung lobe obtained from a C3HeB/FeJ mouse (n = 5). Mice were given a single 25 mg/kg oral dose of BDQ 1 h prior to necropsy.
Figure 13
Figure 13
Differential penetration of BDQ and M2 (N-desmethyl-BDQ) into caseous necrotic granulomas. MALDI-MS images of BDQ (red) and M2 (green) acquired 24 h (A) and 72 h (B) following a single oral 25 mg/kg dose of BDQ. Images represent a single lung lobe obtained from C3HeB/FeJ mice.
Figure 14
Figure 14
BDQ accumulated to a greater degree within THP-1 cells. THP-1 cells were exposed to 1 mM PZA, 0.4 mM POA, or 0.5 mM BDQ. Intracellular-to-extracellular ratios were calculated based upon LC/MS/MS quantification of drug levels following washing and cell lysis.

References

    1. Canetti G. (1955) The tubercle bacillus in the pulmonary lesion of man; histobacteriology and its bearing on the therapy of pulmonary tuberculosis, American rev. ed., Springer Publishing Co., New York, NY.
    1. Hunter R. L.; Actor J. K.; Hwang S. A.; Karev V.; Jagannath C. (2014) Pathogenesis of post primary tuberculosis: immunity and hypersensitivity in the development of cavities. Ann. Clin. Lab. Sci. 44, 365–387.
    1. Lenaerts A.; Barry C. E. 3rd; Dartois V. (2015) Heterogeneity in tuberculosis pathology, microenvironments and therapeutic responses. Immunol Rev. 264, 288–307. 10.1111/imr.12252.
    1. Bumann D. (2015) Heterogeneous host-pathogen encounters: act locally, think globally. Cell Host Microbe 17, 13–19. 10.1016/j.chom.2014.12.006.
    1. Driver E. R.; Ryan G. J.; Hoff D. R.; Irwin S. M.; Basaraba R. J.; Kramnik I.; Lenaerts A. J. (2012) Evaluation of a mouse model of necrotic granuloma formation using C3HeB/FeJ mice for testing of drugs against Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 56, 3181–3195. 10.1128/AAC.00217-12.
    1. Irwin S. M.; Driver E.; Lyon E.; Schrupp C.; Ryan G.; Gonzalez-Juarrero M.; Basaraba R. J.; Nuermberger E. L.; Lenaerts A. J. (2015) Presence of multiple lesion types with vastly different microenvironments in C3HeB/FeJ mice following aerosol infection with Mycobacterium tuberculosis. Dis. Models &amp; Mech. 8, 591–602. 10.1242/dmm.019570.
    1. Irwin S. M.; Gruppo V.; Brooks E.; Gilliland J.; Scherman M.; Reichlen M. J.; Leistikow R.; Kramnik I.; Nuermberger E. L.; Voskuil M. I.; Lenaerts A. J. (2014) Limited activity of clofazimine as a single drug in a mouse model of tuberculosis exhibiting caseous necrotic granulomas. Antimicrob. Agents Chemother. 58, 4026–4034. 10.1128/AAC.02565-14.
    1. Yano T.; Kassovska-Bratinova S.; Teh J. S.; Winkler J.; Sullivan K.; Isaacs A.; Schechter N. M.; Rubin H. (2011) Reduction of clofazimine by mycobacterial type 2 NADH:quinone oxidoreductase: a pathway for the generation of bactericidal levels of reactive oxygen species. J. Biol. Chem. 286, 10276–10287. 10.1074/jbc.M110.200501.
    1. Lanoix J. P.; Lenaerts A. J.; Nuermberger E. L. (2015) Heterogeneous disease progression and treatment response in a C3HeB/FeJ mouse model of tuberculosis. Dis. Models &amp; Mech. 8, 603–610. 10.1242/dmm.019513.
    1. Koul A.; Vranckx L.; Dendouga N.; Balemans W.; Van den Wyngaert I.; Vergauwen K.; Gohlmann H. W.; Willebrords R.; Poncelet A.; Guillemont J.; Bald D.; Andries K. (2008) Diarylquinolines are bactericidal for dormant mycobacteria as a result of disturbed ATP homeostasis. J. Biol. Chem. 283, 25273–25280. 10.1074/jbc.M803899200.
    1. Veziris N.; Ibrahim M.; Lounis N.; Andries K.; Jarlier V. (2011) Sterilizing activity of second-line regimens containing TMC207 in a murine model of tuberculosis. PLoS One 6, e17556.10.1371/journal.pone.0017556.
    1. Williams K.; Minkowski A.; Amoabeng O.; Peloquin C. A.; Taylor D.; Andries K.; Wallis R. S.; Mdluli K. E.; Nuermberger E. L. (2012) Sterilizing activities of novel combinations lacking first- and second-line drugs in a murine model of tuberculosis. Antimicrob. Agents Chemother. 56, 3114–3120. 10.1128/AAC.00384-12.
    1. Diacon A. H.; Donald P. R.; Pym A.; Grobusch M.; Patientia R. F.; Mahanyele R.; Bantubani N.; Narasimooloo R.; De Marez T.; van Heeswijk R.; Lounis N.; Meyvisch P.; Andries K.; McNeeley D. F. (2012) Randomized pilot trial of eight weeks of bedaquiline (TMC207) treatment for multidrug-resistant tuberculosis: long-term outcome, tolerability, and effect on emergence of drug resistance. Antimicrob. Agents Chemother. 56, 3271–3276. 10.1128/AAC.06126-11.
    1. Diacon A. H.; Pym A.; Grobusch M.; Patientia R.; Rustomjee R.; Page-Shipp L.; Pistorius C.; Krause R.; Bogoshi M.; Churchyard G.; Venter A.; Allen J.; Palomino J. C.; De Marez T.; van Heeswijk R. P.; Lounis N.; Meyvisch P.; Verbeeck J.; Parys W.; de Beule K.; Andries K.; Mc Neeley D. F. (2009) The diarylquinoline TMC207 for multidrug-resistant tuberculosis. N. Engl. J. Med. 360, 2397–2405. 10.1056/NEJMoa0808427.
    1. (1981) Controlled trial of four thrice-weekly regimens and a daily regimen all given for 6 months for pulmonary tuberculosis. Lancet 317, 171–174. 10.1016/S0140-6736(81)90057-X.
    1. Combs D. L.; O’Brien R. J.; Geiter L. J. (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. Ahmad Z.; Fraig M. M.; Bisson G. P.; Nuermberger E. L.; Grosset J. H.; Karakousis P. C. (2011) Dose-dependent activity of pyrazinamide in animal models of intracellular and extracellular tuberculosis infections. Antimicrob. Agents Chemother. 55, 1527–1532. 10.1128/AAC.01524-10.
    1. Ibrahim M.; Andries K.; Lounis N.; Chauffour A.; Truffot-Pernot C.; Jarlier V.; Veziris N. (2007) Synergistic activity of R207910 combined with pyrazinamide against murine tuberculosis. Antimicrob. Agents Chemother. 51, 1011–1015. 10.1128/AAC.00898-06.
    1. Diacon A. H.; Dawson R.; von Groote-Bidlingmaier F.; Symons G.; Venter A.; Donald P. R.; van Niekerk C.; Everitt D.; Winter H.; Becker P.; Mendel C. M.; Spigelman M. K. (2012) 14-day bactericidal activity of PA-824, bedaquiline, pyrazinamide, and moxifloxacin combinations: a randomised trial. Lancet 380, 986–993. 10.1016/S0140-6736(12)61080-0.
    1. Zhang Y.; Wade M. M.; Scorpio A.; Zhang H.; Sun Z. (2003) Mode of action of pyrazinamide: disruption of Mycobacterium tuberculosis membrane transport and energetics by pyrazinoic acid. J. Antimicrob. Chemother. 52, 790–795. 10.1093/jac/dkg446.
    1. Shi W.; Zhang X.; Jiang X.; Yuan H.; Lee J. S.; Barry C. E. 3rd; Wang H.; Zhang W.; Zhang Y. (2011) Pyrazinamide inhibits trans-translation in Mycobacterium tuberculosis. Science 333, 1630–1632. 10.1126/science.1208813.
    1. Zhang S.; Chen J. Z.; Shi W. L.; Liu W.; Zhang W. H.; Zhang Y. (2013) Mutations in panD encoding aspartate decarboxylase are associated with pyrazinamide resistance in Mycobacterium tuberculosis. Emerging Microbes Infect. 2, e34.10.1038/emi.2013.38.
    1. Tasneen R.; Li S. Y.; Peloquin C. A.; Taylor D.; Williams K. N.; Andries K.; Mdluli K. E.; Nuermberger E. L. (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. Global Alliance for Tuberculosis Drug Development. A Phase 2 Open Label Partially Randomized Trial to Evaluate the Efficacy, Safety and Tolerability of Combinations of Bedaquiline, Moxifloxacin, PA-824 and Pyrazinamide in Adult Subjects With Drug-Sensitive or Multi Drug-Resistant Pulmonary Tuberculosis. (NC-005), in . , Bethesda, MD.
    1. Zhang Y.; Permar S.; Sun Z. (2002) Conditions that may affect the results of susceptibility testing of Mycobacterium tuberculosis to pyrazinamide. J. Med. Microbiol. 51, 42–49. 10.1099/0022-1317-51-1-42.
    1. Rouan M. C.; Lounis N.; Gevers T.; Dillen L.; Gilissen R.; Raoof A.; Andries K. (2012) Pharmacokinetics and pharmacodynamics of TMC207 and its N-desmethyl metabolite in a murine model of tuberculosis. Antimicrob. Agents Chemother. 56, 1444–1451. 10.1128/AAC.00720-11.
    1. Ahmad Z.; Nuermberger E. L.; Tasneen R.; Pinn M. L.; Williams K. N.; Peloquin C. A.; Grosset J. H.; Karakousis P. C. (2010) Comparison of the ’Denver regimen’ against acute tuberculosis in the mouse and guinea pig. J. Antimicrob. Chemother. 65, 729–734. 10.1093/jac/dkq007.
    1. Via L. E.; Savic R.; Weiner D. M.; Zimmerman M. D.; Prideaux B.; Irwin S. M.; Lyon E.; O’Brien P.; Gopal P.; Eum S.; Lee M.; Lanoix J. P.; Dutta N. K.; Shim T.; Cho J. S.; Kim W.; Karakousis P. C.; Lenaerts A.; Nuermberger E.; Barry C. E. 3rd; Dartois V. (2015) Host-Mediated Bioactivation of Pyrazinamide: Implications for Efficacy, Resistance, and Therapeutic Alternatives. ACS Infect. Dis. 1, 203–214. 10.1021/id500028m.
    1. Kjellsson M. C.; Via L. E.; Goh A.; Weiner D.; Low K. M.; Kern S.; Pillai G.; Barry C. E. 3rd; Dartois V. (2012) Pharmacokinetic evaluation of the penetration of antituberculosis agents in rabbit pulmonary lesions. Antimicrob. Agents Chemother. 56, 446–457. 10.1128/AAC.05208-11.
    1. Svensson E. M.; Aweeka F.; Park J. G.; Marzan F.; Dooley K. E.; Karlsson M. O. (2013) Model-based estimates of the effects of efavirenz on bedaquiline pharmacokinetics and suggested dose adjustments for patients coinfected with HIV and tuberculosis. Antimicrob. Agents Chemother. 57, 2780–2787. 10.1128/AAC.00191-13.
    1. Prideaux B.; Stoeckli M. (2012) Mass spectrometry imaging for drug distribution studies. J. Proteomics 75, 4999–5013. 10.1016/j.jprot.2012.07.028.
    1. Dartois V. (2014) The path of anti-tuberculosis drugs: from blood to lesions to mycobacterial cells. Nat. Rev. Microbiol. 12, 159–167. 10.1038/nrmicro3200.
    1. Prideaux B.; Dartois V.; Staab D.; Weiner D. M.; Goh A.; Via L. E.; Barry C. E. 3rd; Stoeckli M. (2011) High-sensitivity MALDI-MRM-MS imaging of moxifloxacin distribution in tuberculosis-infected rabbit lungs and granulomatous lesions. Anal. Chem. 83, 2112–2118. 10.1021/ac1029049.
    1. Dhillon J.; Andries K.; Phillips P. P.; Mitchison D. A. (2010) Bactericidal activity of the diarylquinoline TMC207 against Mycobacterium tuberculosis outside and within cells. Tuberculosis (Oxford, U. K.) 90, 301–305. 10.1016/j.tube.2010.07.004.
    1. Baik J.; Stringer K. A.; Mane G.; Rosania G. R. (2013) Multiscale distribution and bioaccumulation analysis of clofazimine reveals a massive immune system-mediated xenobiotic sequestration response. Antimicrob. Agents Chemother. 57, 1218–1230. 10.1128/AAC.01731-12.
    1. Barry V. C.; Belton J. G.; Conalty M. L.; Denneny J. M.; Edward D. W.; O’Sullivan J. F.; Twomey D.; Winder F. (1957) A new series of phenazines (rimino-compounds) with high antituberculosis activity. Nature 179, 1013–1015. 10.1038/1791013a0.
    1. Prideaux B.; Via L. E.; Zimmerman M. D.; Eum S.; Sarathy J.; O’Brien P.; Chen C.; Kaya F.; Weiner D. M.; Chen P. Y.; Song T.; Lee M.; Shim T. S.; Cho J. S.; Kim W.; Cho S. N.; Olivier K. N.; Barry C. E. 3rd; Dartois V. (2015) The association between sterilizing activity and drug distribution into tuberculosis lesions. Nat. Med. 21, 1223–1227. 10.1038/nm.3937.
    1. Zhang Y.; Mitchison D. (2003) The curious characteristics of pyrazinamide: a review. Int. J. Tuberc. Lung Dis. 7, 6–21.
    1. Cho S. H.; Warit S.; Wan B.; Hwang C. H.; Pauli G. F.; Franzblau S. G. (2007) Low-oxygen-recovery assay for high-throughput screening of compounds against nonreplicating Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 51, 1380–1385. 10.1128/AAC.00055-06.
    1. Hu Y.; Coates A. R.; Mitchison D. A. (2006) Sterilising action of pyrazinamide in models of dormant and rifampicin-tolerant Mycobacterium tuberculosis. Int. J. Tuberc. Lung Dis. 10, 317–322.
    1. Peterson N. D.; Rosen B. C.; Dillon N. A.; Baughn A. D. (2015) Uncoupling Environmental pH and Intrabacterial Acidification from Pyrazinamide Susceptibility in Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 59, 7320–7326. 10.1128/AAC.00967-15.
    1. Lee R. E.; Hurdle J. G.; Liu J.; Bruhn D. F.; Matt T.; Scherman M. S.; Vaddady P. K.; Zheng Z.; Qi J.; Akbergenov R.; Das S.; Madhura D. B.; Rathi C.; Trivedi A.; Villellas C.; Lee R. B.; Rakesh; Waidyarachchi S. L.; Sun D.; McNeil M. R.; Ainsa J. A.; Boshoff H. I.; Gonzalez-Juarrero M.; Meibohm B.; Bottger E. C.; Lenaerts A. J. (2014) Spectinamides: a new class of semisynthetic antituberculosis agents that overcome native drug efflux. Nat. Med. 20, 152–158. 10.1038/nm.3458.
    1. Lenaerts A. J.; Gruppo V.; Marietta K. S.; Johnson C. M.; Driscoll D. K.; Tompkins N. M.; Rose J. D.; Reynolds R. C.; Orme I. M. (2005) Preclinical testing of the nitroimidazopyran PA-824 for activity against Mycobacterium tuberculosis in a series of in vitro and in vivo models. Antimicrob. Agents Chemother. 49, 2294–2301. 10.1128/AAC.49.6.2294-2301.2005.
    1. Kelly B. P.; Furney S. K.; Jessen M. T.; Orme I. M. (1996) Low-dose aerosol infection model for testing drugs for efficacy against Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 40, 2809–2812.
    1. Ryan G. J.; Shapiro H. M.; Lenaerts A. J. (2014) Improving acid-fast fluorescent staining for the detection of mycobacteria using a new nucleic acid staining approach. Tuberculosis (Oxford, U. K.) 94, 511–518. 10.1016/j.tube.2014.07.004.
    1. Gabrielsson J., and Weiner D. (2007) Pharmacokinetic and pharmacodynamic data analysis: concepts and applications, 4th ed., Swedish Pharmaceutical Press, Stockholm, Sweden.
    1. Bernillon P.; Bois F. Y. (2000) Statistical issues in toxicokinetic modeling: a bayesian perspective. Environ. Health Perspect 108 (Suppl. 5), 883–893. 10.2307/3454322.
    1. R. Core Team (2014) R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, Ed.) Vienna, Austria.
    1. Bois F. Y. (2009) GNU MCSim: Bayesian statistical inference for SBML-coded systems biology models. Bioinformatics 25, 1453–1454. 10.1093/bioinformatics/btp162.

Source: PubMed

3
購読する