Pharmacokinetic evaluation of the penetration of antituberculosis agents in rabbit pulmonary lesions

Maria C Kjellsson, Laura E Via, Anne Goh, Danielle Weiner, Kang Min Low, Steven Kern, Goonaseelan Pillai, Clifton E Barry 3rd, Véronique Dartois, Maria C Kjellsson, Laura E Via, Anne Goh, Danielle Weiner, Kang Min Low, Steven Kern, Goonaseelan Pillai, Clifton E Barry 3rd, Véronique Dartois

Abstract

Standard antituberculosis (anti-TB) therapy requires the use of multiple drugs for a minimum of 6 months, with variable outcomes that are influenced by a number of microbiological, pathological, and clinical factors. This is despite the availability of antibiotics that have good activity against Mycobacterium tuberculosis in vitro and favorable pharmacokinetic profiles in plasma. However, little is known about the distribution of widely used antituberculous agents in the pulmonary lesions where the pathogen resides. The rabbit model of TB infection was used to explore the hypothesis that standard drugs have various abilities to penetrate lung tissue and lesions and that adequate drug levels are not consistently reached at the site of infection. Using noncompartmental and population pharmacokinetic approaches, we modeled the rate and extent of distribution of isoniazid, rifampin, pyrazinamide, and moxifloxacin in rabbit lung and lesions. Moxifloxacin reproducibly showed favorable partitioning into lung and granulomas, while the exposure of isoniazid, rifampin, and pyrazinamide in lesions was markedly lower than in plasma. The extent of penetration in lung and lesions followed different trends for each drug. All four agents distributed rapidly from plasma to tissue with equilibration half-lives of less than 1 min to an hour. The models adequately described the plasma concentrations and reasonably captured actual lesion concentrations. Though further refinement is needed to accurately predict the behavior of these drugs in human subjects, our results enable the integration of lesion-specific pharmacokinetic-pharmacodynamic (PK-PD) indices in clinical trial simulations and in in vitro PK-PD studies with M. tuberculosis.

Figures

Fig 1
Fig 1
Plasma concentration-time profiles of healthy rabbits following multiple (10 to 12) daily doses. (A) INH, 30 mg/kg; (B) RIF, 24 mg/kg; (C) PZA, 125 mg/kg; (D) MXF, 20 mg/kg. Average plasma concentrations with standard deviations (n = 4) are indicated.
Fig 2
Fig 2
Histograms of AUC ratios between tissue (lung or lesion) and plasma for each of the four study drugs. Predicted AUC ratios from the population analysis (PopPK) are compared with those computed using noncompartmental analysis (NCA) of data from the single-dose study with all four drugs administered together (dark gray bars). Individual AUC ratios from the population analysis were obtained from the effect compartment population PK model, based on all four lesion PK studies described in this work (light gray bars).
Fig 3
Fig 3
Composite photo of granuloma types sampled in this study. (A) Nonnecrotizing, predominantly cellular granuloma, with central epithelioid macrophages and no necrosis visible at dissection; cellular granulomas were typically ≤2 mm in diameter; (B) three or four coalescing nonnecrotizing granulomas that could not be divided into separate lesions during dissection; (C) necrotizing granuloma with central acellular region visible at dissection; (D) cavitating necrotic lesion which has become suppurative and macroscopically demonstrates a semiliquid core that oozes from the lesion during dissection. Histologically, the section shows infiltrating neutrophils and macrophages in the suppurative caseum. Bars, 500 μm.
Fig 4
Fig 4
Schematic picture describing the models for plasma pharmacokinetics and tissue penetration. The model for PZA included one absorption compartment. ka, absorption rate constant; V, oral volume of distribution; CL, clearance; V2, peripheral volume of distribution; Q, intercompartmental clearance; F, relative bioavailability for combination therapy; PC, penetration coefficient from plasma into lung or lesion; kpl-lung/lesion, distribution rate constant. Bioavailability (F) was fixed to 1 for all drugs but RIF, where the relative F was estimated for all single-dose studies and fixed to 1 for combination-therapy studies.
Fig 5
Fig 5
Observed plasma concentrations of INH, RIF, PZA, and MXF versus individual model-predicted concentrations. Solid line, line of identity.
Fig 6
Fig 6
Predicted concentration-time profiles for each individual animal are shown as solid lines. Observed concentrations are indicated by open circles. Data are shown for INH, RIF, PZA, and MXF in lung tissue and lesions.
Fig 7
Fig 7
Visual predictive check showing the predicted main trend (dashed lines) and 90% prediction interval (shaded areas) of plasma, lung, and lesion concentrations for INH at 50 mg/kg, MXF at 25 mg/kg, PZA at 125 mg/kg, and RIF at 30 mg/kg. Actual observations at the relevant doses are overlaid (open circles) and connected with a solid line.
Fig 8
Fig 8
Box plots of the ratios of individually predicted AUCs in lung and lesion tissue relative to plasma AUC for each drug. The box width is the interquartile range, with the horizontal line marking the median value. The whiskers indicate the maximum/minimum of ratios within 1.5 times the upper/lower quartiles. Observed data points falling outside this range are represented by open circles.

References

    1. Acocella G. 1978. Clinical pharmacokinetics of rifampicin. Clin. Pharmacokinet. 3:108–127
    1. Barbour A, Scaglione F, Derendorf H. 2010. Class-dependent relevance of tissue distribution in the interpretation of anti-infective pharmacokinetic/pharmacodynamic indices. Int. J. Antimicrob. Agents 35:431–438
    1. Barclay WR, Ebert RH, Le Roy GV, Manthei RW, Roth LJ. 1953. Distribution and excretion of radioactive isoniazid in tuberculous patients. JAMA 151:1384–1388
    1. Bareggi SR, Cerutti R, Pirola R, Riva R, Cisternino M. 1987. Clinical pharmacokinetics and metabolism of pyrazinamide in healthy volunteers. Arzneimittelforschung 37:849–854
    1. Barry CE, et al. 2009. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat. Rev. Microbiol. 7:845–855
    1. Canetti G, Parrot R, Porven G, Le Lirzin M. 1969. Rifomycin levels in the lung and tuberculous lesions in man. Acta Tuberc. Pneumol. Belg. 60:315–322 (In French.)
    1. Carceles CM, Serrano JM, Marin P, Escudero E, Fernandez-Varon E. 2006. Pharmacokinetics of moxifloxacin in rabbits after intravenous, subcutaneous and a long-acting poloxamer 407 gel formulation administration. J. Vet. Med. A Physiol. Pathol. Clin. Med. 53:300–304
    1. Chan K, Wong CL. 1988. Pharmacokinetics of pyrazinamide in plasma and CSF of rabbits following intravenous and oral administration. Eur. J. Drug Metab. Pharmacokinet. 13:195–199
    1. Chang KC, Leung CC, Yew WW, Ho SC, Tam CM. 2004. A nested case-control study on treatment-related risk factors for early relapse of tuberculosis. Am. J. Respir. Crit. Care Med. 170:1124–1130
    1. Conte JE, Jr, Golden JA, Duncan S, McKenna E, Zurlinden E. 1999. Intrapulmonary concentrations of pyrazinamide. Antimicrob. Agents Chemother. 43:1329–1333
    1. Conte JE, Jr, et al. 2002. Effects of gender, AIDS, and acetylator status on intrapulmonary concentrations of isoniazid. Antimicrob. Agents Chemother. 46:2358–2364
    1. Dannenberg AM., Jr 2006. Pathogenesis of human pulmonary tuberculosis. ASM Press, Washington, DC
    1. Dannenberg AM., Jr 2006. Stages in the pathogenesis of human and rabbit tuberculosis, p 22–33 InPathogenesis of human pulmonary tuberculosis. ASM Press, Washington, DC
    1. Ellard GA, Humphries MJ, Allen BW. 1993. Cerebrospinal fluid drug concentrations and the treatment of tuberculous meningitis. Am. Rev. Respir. Dis. 148:650–655
    1. Ellard GA, Humphries MJ, Gabriel M, Teoh R. 1987. Penetration of pyrazinamide into the cerebrospinal fluid in tuberculous meningitis. Br. Med. J. (Clin. Res. ed.) 294:284–285
    1. Eum SY, et al. 2010. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest 137:122–128
    1. Fernandez-Varon E, et al. 2005. Pharmacokinetic-pharmacodynamic integration of moxifloxacin in rabbits after intravenous, intramuscular and oral administration. J. Vet. Pharmacol. Ther. 28:343–348
    1. Firsov AA, et al. 2000. Comparative pharmacodynamics of moxifloxacin and levofloxacin in an in vitro dynamic model: prediction of the equivalent AUC/MIC breakpoints and equiefficient doses. J. Antimicrob. Chemother. 46:725–732
    1. Gobburu JV, et al. 2001. Pharmacokinetic-pharmacodynamic modeling of rivastigmine, a cholinesterase inhibitor, in patients with Alzheimer's disease. J. Clin. Pharmacol. 41:1082–1090
    1. Goutelle S, et al. 2009. Population modeling and Monte Carlo simulation study of the pharmacokinetics and antituberculosis pharmacodynamics of rifampin in lungs. Antimicrob. Agents Chemother. 53:2974–2981
    1. Gumbo T. 2010. New susceptibility breakpoints for first-line antituberculosis drugs based on antimicrobial pharmacokinetic/pharmacodynamic science and population pharmacokinetic variability. Antimicrob. Agents Chemother. 54:1484–1491
    1. Gumbo T, et al. 2004. Selection of a moxifloxacin dose that suppresses drug resistance in Mycobacterium tuberculosis, by use of an in vitro pharmacodynamic infection model and mathematical modeling. J. Infect. Dis. 190:1642–1651
    1. Hu Y, Coates AR, Mitchison DA. 2003. Sterilizing activities of fluoroquinolones against rifampin-tolerant populations of Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 47:653–657
    1. Jeena PM, Bishai WR, Pasipanodya JG, Gumbo T. 2011. In silico children and the glass mouse model: clinical trial simulations to identify and individualize optimal isoniazid doses in children with tuberculosis. Antimicrob. Agents Chemother. 55:539–545
    1. Jonsson EN, Karlsson MO. 1999. Xpose—an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput. Methods Programs Biomed. 58:51–64
    1. Kaplan G, et al. 2003. Mycobacterium tuberculosis growth at the cavity surface: a microenvironment with failed immunity. Infect. Immun. 71:7099–7108
    1. Kaplan G, Tsenova L. 2011. Pulmonary tuberculosis in the rabbit, p 107–128 In Leong FJ, Dartois V, Dick T. (ed), A color atlas of comparative pathology of pulmonary tuberculosis. CRC Press, Boca Raton, FL
    1. Kim DH, et al. 2008. Treatment outcomes and long-term survival in patients with extensively drug-resistant tuberculosis. Am. J. Respir. Crit. Care Med. 178:1075–1082
    1. Kislitsyna NA. 1985. Comparative evaluation of rifampicin and isoniazid penetration into the pathological foci of the lungs in tuberculosis patients. Probl. Tuberk., p 55–57 (In Russian.)
    1. Kislitsyna NA, Kotova NI. 1980. Rifampicin and isoniazid concentration in the blood and resected lungs in tuberculosis with combined use of the preparations. Probl. Tuberk., p 63–65 (In Russian.)
    1. Lee SY, Fan HW, Sutherland C, DeRyke AC, Nicolau DP. 2007. Antibacterial effects of moxifloxacin and levofloxacin simulating epithelial lining fluid concentrations against community-acquired methicillin-resistant Staphylococcus aureus. Drugs R D 8:69–77
    1. Leong FJ, Eum SY, Via LE, Barry CER. 2011. Pathology of tuberculosis in the human lung, p 53–81 In Dartois V, Dick T, Leong FJ. (ed), A color atlas of comparative pathology of pulmonary tuberculosis. CRC Press, Boca Raton, FL
    1. Lindbom L, Pihlgren P, Jonsson EN. 2005. PsN-toolkit—a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput. Methods Programs Biomed. 79:241–257
    1. Lindbom L, Ribbing J, Jonsson EN. 2004. Perl-speaks-NONMEM (PsN)—a Perl module for NONMEM related programming. Comput. Methods Programs Biomed. 75:85–94
    1. Liu P, Derendorf H. 2003. Antimicrobial tissue concentrations. Infect. Dis. Clin. North Am. 17:599–613
    1. Loos U, et al. 1985. Pharmacokinetics of oral and intravenous rifampicin during chronic administration. Klin. Wochenschr. 63:1205–1211
    1. Mitchison DA. 1985. The action of antituberculosis drugs in short-course chemotherapy. Tubercle 66:219–225
    1. Mitchison DA. 1973. Plasma concentrations of isoniazid in the treatment of tuberculosis. In Davies DS, Prichard BNC. (ed), Biological effects of drugs in relation to their plasma concentrations. Macmillan, New York, NY
    1. Mitchison DA, Coates AR. 2004. Predictive in vitro models of the sterilizing activity of anti-tuberculosis drugs. Curr. Pharm. Des. 10:3285–3295
    1. Mouton JW, et al. 2008. Tissue concentrations: do we ever learn? J. Antimicrob. Chemother. 61:235–237
    1. Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivisto KT. 2003. Pharmacokinetic interactions with rifampicin: clinical relevance. Clin. Pharmacokinet. 42:819–850
    1. Noel AR, Bowker KE, Macgowan AP. 2005. Pharmacodynamics of moxifloxacin against anaerobes studied in an in vitro pharmacokinetic model. Antimicrob. Agents Chemother. 49:4234–4239
    1. O'Brien JK, Doerfler ME, Harkin TJ, Rom WN. 1998. Isoniazid levels in the bronchoalveolar lavage fluid of patients with pulmonary tuberculosis. Lung 176:205–211
    1. Pillai G, et al. 2006. Population pharmacokinetics of ibandronate in Caucasian and Japanese healthy males and postmenopausal females. Int. J. Clin. Pharmacol. Ther. 44:655–667
    1. Piotrovskii VK. 1987. The use of Weibull distribution to describe the in vivo absorption kinetics. J. Pharmacokinet. Biopharm. 15:681–686
    1. Prideaux B, et al. 2011. High-sensitivity MALDI-MRM-MS imaging of moxifloxacin distribution in tuberculosis-infected rabbit lungs and granulomatous lesions. Anal. Chem. 83:2112–2118
    1. Reed MB, et al. 2004. A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature 431:84–87
    1. Sacchettini JC, Rubin EJ, Freundlich JS. 2008. Drugs versus bugs: in pursuit of the persistent predator Mycobacterium tuberculosis. Nat. Rev. Microbiol. 6:41–52
    1. Sauermann R, et al. 2005. Antibiotic abscess penetration: fosfomycin levels measured in pus and simulated concentration-time profiles. Antimicrob. Agents Chemother. 49:4448–4454
    1. Savic RM, Jonker DM, Kerbusch T, Karlsson MO. 2007. Implementation of a transit compartment model for describing drug absorption in pharmacokinetic studies. J. Pharmacokinet. Pharmacodyn. 34:711–726
    1. Soman A, Honeybourne D, Andrews J, Jevons G, Wise R. 1999. Concentrations of moxifloxacin in serum and pulmonary compartments following a single 400 mg oral dose in patients undergoing fibre-optic bronchoscopy. J. Antimicrob. Chemother. 44:835–838
    1. Stass H, Dalhoff A, Kubitza D, Schuhly U. 1998. Pharmacokinetics, safety, and tolerability of ascending single doses of moxifloxacin, a new 8-methoxy quinolone, administered to healthy subjects. Antimicrob. Agents Chemother. 42:2060–2065
    1. Tunblad K, et al. 2008. The use of clinical irrelevance criteria in covariate model building with application to dofetilide pharmacokinetic data. J. Pharmacokinet. Pharmacodyn. 35:503–526
    1. Via LE, et al. 2008. Tuberculous granulomas are hypoxic in guinea pigs, rabbits, and nonhuman primates. Infect. Immun. 76:2333–2340
    1. Wagner C, Sauermann R, Joukhadar C. 2006. Principles of antibiotic penetration into abscess fluid. Pharmacology 78:1–10
    1. Wallis RS, et al. 1999. Drug tolerance in Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 43:2600–2606
    1. Walubo A, Chan K, Wong CL. 1991. The pharmacokinetics of isoniazid and hydrazine metabolite in plasma and cerebrospinal fluid of rabbits. Methods Find. Exp. Clin. Pharmacol. 13:199–204
    1. Walubo A, Chan K, Woo J, Chan HS, Wong CL. 1991. The disposition of antituberculous drugs in plasma of elderly patients. II. Isoniazid, rifampicin and pyrazinamide. Methods Find. Exp. Clin. Pharmacol. 13:551–556
    1. Wayne LG, Hayes LG. 1996. An in vitro model for sequential study of shiftdown of Mycobacterium tuberculosis through two stages of nonreplicating persistence. Infect. Immun. 64:2062–2069
    1. Wayne LG, Sohaskey CD. 2001. Nonreplicating persistence of Mycobacterium tuberculosis. Annu. Rev. Microbiol. 55:139–163
    1. WHO 2011. Tuberculosis MDR-TB & XDR-TB 2011 progress report. WHO, Geneva, Switzerland
    1. Ziglam HM, Baldwin DR, Daniels I, Andrew JM, Finch RG. 2002. Rifampicin concentrations in bronchial mucosa, epithelial lining fluid, alveolar macrophages and serum following a single 600 mg oral dose in patients undergoing fibre-optic bronchoscopy. J. Antimicrob. Chemother. 50:1011–1015

Source: PubMed

3
Subscribe