We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

In vitro pharmacokinetic/pharmacodynamic models in anti-infective drug development: focus on TB

    Pavan K Vaddady

    Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 874 Union Avenue, Suite 5p, TN 38163, USA

    ,
    Richard E Lee

    Department of Chemical Biology & Therapeutics, St Jude Children’s Research Hospital, TN, USA

    &
    Published Online:https://doi.org/10.4155/fmc.10.224

    For rapid anti-tuberculosis (TB) drug development in vitro pharmacokinetic/pharmacodynamic (PK/PD) models are useful in evaluating the direct interaction between the drug and the bacteria, thereby guiding the selection of candidate compounds and the optimization of their dosing regimens. Utilizing in vivo drug-clearance profiles from animal and/or human studies and simulating them in an in vitro PK/PD model allows the in-depth characterization of antibiotic activity of new and existing antibacterials by generating time–kill data. These data capture the dynamic interplay between mycobacterial growth and changing drug concentration as encountered during prolonged drug therapy. This review focuses on important PK/PD parameters relevant to anti-TB drug development, provides an overview of in vitro PK/PD models used to evaluate the efficacy of agents against mycobacteria and discusses the related mathematical modeling approaches of time–kill data. Overall, it provides an introduction to in vitro PK/PD models and their application as critical tools in evaluating anti-TB drugs.

    Bibliography

    • World Health Organization. Global tuberculosis control. Epidemiology, strategy, financing. WHO report 2009. WHO/HTM/TB/2009.411, Geneva, Switzerland (2009).
    • Meibohm B, Derendorf H. Pharmacokinetic/pharmacodynamic studies in drug product development. J. Pharm. Sci.91,18–31 (2002).
    • Suryawanshi S, Zhang L, Pfister M, Meibohm B. The current role of model-based drug development. Expert Opin. Drug Discov.5,311–321 (2010).
    • Zhang L, Pfister M, Meibohm B. Concepts and challenges in quantitative pharmacology and model-based drug development. AAPS J.10,552–559 (2008).
    • Budha NR, Lee RE, Meibohm B. Biopharmaceutics, pharmacokinetics and pharmacodynamics of anti-tuberculosis drugs. Curr. Med. Chem.15,809–825 (2008).
    • Boshoff HI, Barry CE 3rd. Tuberculosis – metabolism and respiration in the absence of growth. Nat. Rev. Microbiol.3,70–80 (2005).
    • Gloede J, Scheerans C, Derendorf H, Kloft C. In vitro pharmacodynamic models to determine the effect of antibacterial drugs. J. Antimicrob. Chemother.65,186–201 (2010).
    • Katsube T, Yano Y, Yamano Y, Munekage T, Kuroda N, Takano M. Pharmacokinetic–pharmacodynamic modeling and simulation for bactericidal effect in an in vitro dynamic model. J. Pharm. Sci.97,4108–4117 (2008).
    • White RL. What in vitro models of infection can and cannot do. Pharmacotherapy21,292S–301S (2001).
    • 10  Schmidt S, Barbour A, Sahre M, Rand Kh, Derendorf H. PK/PD: new insights for antibacterial and antiviral applications. Curr. Opin. Pharmacol.8,549–556 (2008).
    • 11  Mouton JW, Theuretzbacher U, Craig WA, Tulkens PM, Derendorf H, Cars O. Tissue concentrations: do we ever learn? J. Antimicrob. Chemother.61,235–237 (2008).
    • 12  Clinical and Laboratory Standards Institute. Approved standard M7-A7. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically (2007).
    • 13  Mueller M, De La Pena A, Derendorf H. Issues in pharmacokinetics and pharmacodynamics of anti-infective agents: kill curves versus MIC. Antimicrob. Agents Chemother.48,369–377 (2004).
    • 14  Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin. Infect. Dis.26,1–10 (1998).
    • 15  Macgowan A, Bowker K. Developments in PK/PD: optimising efficacy and prevention of resistance. A critical review of PK/PD in in vitro models. Int. J. Antimicrob. Agents19,291–298 (2002).
    • 16  Garrett ER, Wright OK, Miller GH, Smith KL. Quantification and prediction of the biological activities of chloramphenicol analogs by microbial kinetics. J. Med. Chem.9,203–208 (1966).
    • 17  Li RC, Zhu M, Schentag JJ. Achieving an optimal outcome in the treatment of infections. The role of clinical pharmacokinetics and pharmacodynamics of antimicrobials. Clin. Pharmacokinet.37,1–16 (1999).
    • 18  Nolting A, Dalla Costa T, Rand KH, Derendorf H. Pharmacokinetic–pharmacodynamic modeling of the antibiotic effect of piperacillin in vitro. Pharm. Res.13,91–96 (1996).
    • 19  Schentag JJ. Pharmacokinetic and pharmacodynamic surrogate markers: studies with fluoroquinolones in patients. Am. J. Health Syst. Pharm.56,S21–S24 (1999).
    • 20  Schentag JJ. Antimicrobial action and pharmacokinetics/pharmacodynamics: the use of AUIC to improve efficacy and avoid resistance. J. Chemother.11,426–439 (1999).
    • 21  Jayaram R, Gaonkar S, Kaur P et al. Pharmacokinetics-pharmacodynamics of rifampin in an aerosol infection model of tuberculosis. Antimicrob. Agents Chemother.47,2118–2124 (2003).
    • 22  Jayaram R, Shandil RK, Gaonkar S et al. Isoniazid pharmacokinetics–pharmacodynamics in an aerosol infection model of tuberculosis. Antimicrob. Agents Chemother.48,2951–2957 (2004).
    • 23  Shandil RK, Jayaram R, Kaur P et al. Moxifloxacin, ofloxacin, sparfloxacin and ciprofloxacin against Mycobacterium tuberculosis. Evaluation of in vitro and pharmacodynamic indices that best predict in vivo efficacy. Antimicrob. Agents Chemother.51,576–582 (2007).
    • 24  Grasso S, Meinardi G, De Carneri I, Tamassia V. New in vitro model to study the effect of antibiotic concentration and rate of elimination on antibacterial activity. Antimicrob Agents Chemother.13,570–576 (1978).
    • 25  Keil S, Wiedemann B. Mathematical corrections for bacterial loss in pharmacodynamic in vitro dilution models. Antimicrob. Agents Chemother.39,1054–1058 (1995).
    • 26  White CA, Toothaker RD, Smith AL, Slattery JT. Correction for bacterial loss in in vitro dilution models. Antimicrob. Agents Chemother.31,1859–1860 (1987).
    • 27  Budha NR, Lee RB, Hurdle JG, Lee RE, Meibohm B. A simple in vitro PK/PD model system to determine time-kill curves of drugs against mycobacteria. Tuberculosis89,378–385 (2009).
    • 28  Lowdin E, Odenholt I, Bengtsson S, Cars O. Pharmacodynamic effects of sub-MICs of benzylpenicillin against Streptococcus pyogenes in a newly developed in vitro kinetic model. Antimicrob. Agents Chemother.40,2478–2482 (1996).
    • 29  Blaser J, Stone BB, Zinner SH. Two compartment kinetic model with multiple artificial capillary units. J. Antimicrob. Chemother.15,131–137 (1985).
    • 30  Garrison MW, Vance-Bryan K, Larson TA et al. Assessment of effects of protein binding on daptomycin and vancomycin killing of Staphylococcus aureus by using an in vitro pharmacodynamic model. Antimicrob. Agents Chemother.34,1925–1931 (1990).
    • 31  Reeves DS. Advantages and disadvantages of an in vitro model with two compartments connected by a dialyser: results of experiments with ciprofloxacin. J. Antimicrob. Chemother.15,159–167 (1985).
    • 32  Gumbo T, Louie A, Deziel MR, Parsons LM, Salfinger M, Drusano GL. 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 (2004).
    • 33  Gumbo T, Louie A, Deziel MR, Drusano GL. Pharmacodynamic evidence that ciprofloxacin failure against tuberculosis is not due to poor microbial kill but to rapid emergence of resistance. Antimicrob. Agents Chemother.49,3178–3181 (2005).
    • 34  Gumbo T, Louie A, Liu W et al. Isoniazid’s bactericidal activity ceases because of the emergence of resistance, not depletion of Mycobacterium tuberculosis in the log phase of growth. J. Infect. Dis.195,194–201 (2007).
    • 35  Gumbo T, Louie A, Liu W et al. Isoniazid bactericidal activity and resistance emergence: integrating pharmacodynamics and pharmacogenomics to predict efficacy in different ethnic populations. Antimicrob. Agents Chemother.51,2329–2336 (2007).
    • 36  Gumbo T, Louie A, Deziel MR et al. Concentration-dependent Mycobacterium tuberculosis killing and prevention of resistance by rifampin. Antimicrob. Agents Chemother.51,3781–3788 (2007).
    • 37  Gumbo T, Dona CS, Meek C, Leff R. Pharmacokinetics–pharmacodynamics of pyrazinamide in a novel in vitro model of tuberculosis for sterilizing effect: a paradigm for faster assessment of new anti-tuberculosis drugs. Antimicrob. Agents Chemother.53,3197–3204 (2009).
    • 38  Srivastava S, Musuka S, Sherman C, Meek C, Leff R, Gumbo T. Efflux-pump-derived multiple drug resistance to ethambutol monotherapy in Mycobacterium tuberculosis and the pharmacokinetics and pharmacodynamics of ethambutol. J. Infect. Dis.201,1225–1231 (2010).
    • 39  Vogelman BS, Craig WA. Post-antibiotic effects. J. Antimicrob. Chemother.15,37–46 (1985).
    • 40  McDonald PJ, Craig WA, Kunin CM. Persistent effect of antibiotics on Staphylococcus aureus after exposure for limited periods of time. J. Infect. Dis.135,217–223 (1977).
    • 41  Mouton JW, Dudley MN, Cars O, Derendorf H, Drusano GL. Standardization of pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs. Int. J. Antimicrob. Agents19,355–358 (2002).
    • 42  Chan CY, Au-Yeang C, Yew WW, Leung CC, Cheng AF. In vitro post-antibiotic effects of rifapentine, isoniazid and moxifloxacin against Mycobacterium tuberculosis. Antimicrob. Agents Chemother.48,340–343 (2004).
    • 43  Levison ME. Pharmacodynamics of antimicrobial drugs. Infect. Dis. Clin. North. Am.18,451–465 (2004).
    • 44  Odenholt-Tornqvist I, Lowdin E, Cars O. Pharmacodynamic effects of subinhibitory concentrations of b-lactam antibiotics in vitro. Antimicrob. Agents Chemother.35,1834–1839 (1991).
    • 45  Ginsburg AS, Lee J, Woolwine SC, Grosset JH, Hamzeh FM, Bishai WW. Modeling in vivo pharmacokinetics and pharmacodynamics of moxifloxacin therapy for Mycobacterium tuberculosis infection by using a novel cartridge system. Antimicrob. Agents Chemother.49,853–856 (2005).
    • 46  National Committee for Clinical Laboratory Standards. Performance standards for antimicrobial susceptibility testing: twentieth informational supplement M100–M120 (2010).
    • 47  Drusano GL, Liu W, Fregeau C, Kulawy R, Louie A. Differing effects of combination chemotherapy with meropenem and tobramycin on cell kill and suppression of resistance of wild-type Pseudomonas aeruginosa PAO1 and its isogenic MexAB efflux pump-overexpressed mutant. Antimicrob. Agents Chemother.53,2266–2273 (2009).
    • 48  Hurdle JG, Lee RB, Budha NR et al. A microbiological assessment of novel nitrofuranylamides as anti-tuberculosis agents. J. Antimicrob. Chemother.62,1037–1045 (2008).
    • 49  Blaser J. In vitro model for simultaneous simulation of the serum kinetics of two drugs with different half-lives. J. Antimicrob. Chemother.15,125–130 (1985).
    • 50  Cha R, Rybak MJ. Linezolid and vancomycin, alone and in combination with rifampin, compared with moxifloxacin against a multidrug-resistant and a vancomycin-tolerant Streptococcus pneumoniae strain in an in vitro pharmacodynamic model. Antimicrob. Agents Chemother.47,1984–1987 (2003).
    • 51  Craig WA. Proof of concept: performance testing in models. Clin. Microbiol. Infect.10,12–17 (2004).
    • 52  Leonard SN, Kaatz GW, Rucker LR, Rybak MJ. Synergy between gemifloxacin and trimethoprim/sulfamethoxazole against community-associated methicillin-resistant Staphylococcus aureus. J. Antimicrob. Chemother.62,1305–1310 (2008).
    • 53  Zinner SH, Blaser J, Stone BB, Groner MC. Use of an in vitro kinetic model to study antibiotic combinations. J. Antimicrob Chemother.15,221–226 (1985).
    • 54  Tam VH, Louie A, Deziel MR, Liu W, Drusano GL. The relationship between quinolone exposures and resistance amplification is characterized by an inverted U: a new paradigm for optimizing pharmacodynamics to counterselect resistance. Antimicrob. Agents Chemother.51,744–747 (2007).
    • 55  Dong Y, Zhao X, Domagala J, Drlica K. Effect of fluoroquinolone concentration on selection of resistant mutants of Mycobacterium bovis BCG and Staphylococcus aureus. Antimicrob. Agents Chemother.43,1756–1758 (1999).
    • 56  Homma T, Hori T, Sugimori G, Yamano Y. Pharmacodynamic assessment based on mutant prevention concentrations of fluoroquinolones to prevent the emergence of resistant mutants of Streptococcus pneumoniae. Antimicrob. Agents Chemother.51,3810–3815 (2007).
    • 57  Berger-Bachi B, Strassle A, Kayser FH. Characterization of an isogenic set of methicillin-resistant and susceptible mutants of Staphylococcus aureus. Eur. J. Clin. Microbiol.5,697–701 (1986).
    • 58  Wootton M, Howe RA, Hillman R, Walsh–TR, Bennett PM, MacGowan AP. A modified population analysis profile (PAP) method to detect hetero-resistance to vancomycin in Staphylococcus aureus in a UK hospital. J. Antimicrob. Chemother.47,399–403 (2001).
    • 59  Czock D, Keller F. Mechanism-based pharmacokinetic–pharmacodynamic modeling of antimicrobial drug effects. J. Pharmacokinet. Pharmacodyn.34,727–751 (2007).
    • 60  Dahl SG, Aarons L, Gundert-Remy U et al. Incorporating physiological and biochemical mechanisms into pharmacokinetic–pharmacodynamic models: a conceptual framework. Basic Clin. Pharmacol. Toxicol.106,2–12 (2009).
    • 61  Balaban NQ, Merrin J, Chait R, Kowalik L, Leibler S. Bacterial persistence as a phenotypic switch. Science305,1622–1625 (2004).
    • 62  Nielsen EI, Viberg A, Lowdin E, Cars O, Karlsson MO, Sandstrom M. Semimechanistic pharmacokinetic/pharmacodynamic model for assessment of activity of antibacterial agents from time-kill curve experiments. Antimicrob. Agents Chemother.51,128–136 (2007).
    • 63  Li Y, Nguyen MH, Cheng S et al. A pharmacokinetic/pharmacodynamic mathematical model accurately describes the activity of voriconazole against candida spp. in vitro. Int. J. Antimicrob. Agents31,369–374 (2008).
    • 64  Russell DG. Who puts the tubercle in tuberculosis? Nat. Rev. Microbiol.5,39–47 (2007).
    • 65  Monack DM, Mueller A, Falkow S. Persistent bacterial infections: the interface of the pathogen and the host immune system. Nat. Rev. Microbiol.2,747–765 (2004).
    • 66  Eum SY, Kong JH, Hong MS et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest137,122–128 (2010).
    • 67  Dalhoff A. Differences between bacteria grown in vitro and in vivo. J. Antimicrob. Chemother.15,175–195 (1985).
    • 68  Watson DL, Prideaux JA. Comparisons of Staphylococcus aureus grown in vitro or in vivo. Microbiol. Immunol.23,543–547 (1979).
    • 69  Manganelli R, Dubnau E, Tyagi S, Kramer FR, Smith I. Differential expression of 10 sigma factor genes in Mycobacterium tuberculosis. Mol. Microbiol.31,715–724 (1999).
    • 70  Nuermberger E, Grosset J. Pharmacokinetic and pharmacodynamic issues in the treatment of mycobacterial infections. Eur. J. Clin. Microbiol. Infect. Dis.23,243–255 (2004).
    • 71  Holdiness MR. Clinical pharmacokinetics of the anti-tuberculosis drugs. Clin. Pharmacokinet.9,511–544 (1984).
    • 72  Johnson JL, Hadad DJ, Boom WH et al. Early and extended early bactericidal activity of levofloxacin, gatifloxacin and moxifloxacin in pulmonary tuberculosis. Int. J. Tuberc. Lung Dis.10,605–612 (2006).
    • 73  McGee B, Dietze R, Hadad DJ et al. Population pharmacokinetics of linezolid in adults with pulmonary tuberculosis. Antimicrob. Agents Chemother.53,3981–3984 (2009).
    • 74  Peloquin CA. Tuberculosis. In: Pharmacotherapy: A Pathophysiologic Approach (7th Edition). DiPiro JT TR, Yee GC, Matzke GR, Wells BG, Posey LM (Eds). McGraw-Hill Medical, NY, USA, 1839–1856 (2008).
    • 75  Petri WA. Chemotherapy of tuberculosis, Mycobacterium avium complex disease, and leprosy. In: Goodman & Gilman’s The Pharmacological Basis of Therapeutics (11th Edition). Brunton LL, Lazo JS, Parker KL (Eds). McGraw-Hill Medical, NY, USA, 1203–1223 (2005).
    • 76  World Health Organization Treatment of tuberculosis. Guidelines for national programs. WHO/HTM/TB/2009.420. World Health Organization, Geneva, Switzerland (2009).
    • 77  Jureen P, Angeby K, Sturegard E et al. Wild-type minimal inhibitory concentration distributions for aminoglycoside and cyclic polypeptide antibiotics used for the treatment of Mycobacterium tuberculosis. J. Clin. Microbiol.48,1853–1458 (2010).