Single- and multiple-dose pharmacokinetics and absolute bioavailability of tedizolid

Shawn Flanagan, Edward Fang, Kelly A Muñoz, Sonia L Minassian, Philippe G Prokocimer, Shawn Flanagan, Edward Fang, Kelly A Muñoz, Sonia L Minassian, Philippe G Prokocimer

Abstract

Objectives: Tedizolid phosphate is a novel antibacterial under investigation for the treatment of gram-positive infections. This study was conducted to assess the pharmacokinetics, safety, and tolerability of intravenous tedizolid phosphate as well as the oral bioavailability of tedizolid phosphate.

Design: Double-blind, single-ascending dose, multiple-dose pharmacokinetics study, as well as tolerability and open-label crossover studies.

Setting: Single center in the United States (Covance Clinical Research Unit, Madison, WI) between September 2009 and January 2010.

Participants: Ninety healthy volunteers.

Intervention: Single intravenous (IV) doses of tedizolid phosphate 50 mg (lead-in) and 100-400 mg. Single oral and IV dose of tedizolid phosphate 200 mg in crossover fashion. Multiple IV doses of tedizolid phosphate 200 and 300 mg for up to 7 days.

Measurements and main results: A dose-dependent increase was observed in the maximum plasma concentration (1.2-5.1 μg/ml) and the area under the concentration-time curve (17.4-58.7 μg × hr/ml) of tedizolid (the microbiologically active moiety of tedizolid phosphate) after single IV doses of tedizolid phosphate 100-400 mg. Administration of IV tedizolid phosphate 200 mg once/day for 7 days resulted in minimal (28%) tedizolid accumulation. The absolute oral bioavailability of tedizolid after a single 200-mg dose of tedizolid phosphate was 91%; pharmacokinetic parameters of tedizolid were similar with oral and IV administration. Treatment-related adverse events occurred in 41% of subjects. Most adverse events were related to infusion site and became more frequent with multiple dosing. In an additional 3-day tolerability study, IV tedizolid phosphate 200 mg and placebo were similarly tolerated, based on visual infusion phlebitis scores.

Conclusion: These results from a population of healthy volunteers support once/day dosing of tedizolid phosphate 200 mg with both the oral and IV formulations, without the need for dose adjustment when switching administration routes.

Trial registration: ClinicalTrials.gov NCT00983255.

Keywords: intravenous; pharmacokinetics; tedizolid phosphate.

© 2014 Cubist Pharmaceuticals. Pharmacotherapy published by Wiley Periodicals, Inc. on behalf of Pharmacotherapy Publications, Inc.

Figures

Figure 1
Figure 1
Study design. IV = intravenous.
Figure 2
Figure 2
Mean plasma tedizolid phosphate or tedizolid concentrations during a 1-hour intravenous (IV) infusion of tedizolid phosphate 200 mg (n=8). These data are from the IV portion of the crossover bioavailability phase of the study. Error bars denote standard deviations.
Figure 3
Figure 3
Mean tedizolid plasma concentration after a single dose of intravenous (IV) tedizolid phosphate, plotted on a (A) linear or (B) log time scale (n=9, 9, 8, and 9 for 100-, 200-, 300-, and 400-mg doses, respectively). Postinfusion data from subjects with 1-hour infusions were offset by 1 hour to match postinfusion times of subjects having 2-hour infusions. Mean tedizolid plasma concentration after 7 days of IV tedizolid phosphate 200 mg/day, plotted on a (C) linear or (D) log time scale (n=8). Mean tedizolid plasma concentration after a single dose of IV or oral 200 mg tedizolid phosphate, plotted on a (E) linear or (F) log time scale (n=8). Error bars denote standard deviations.
Figure 4
Figure 4
(A) Maximum plasma concentration (Cmax) for tedizolid after single or first multiple dose of intravenous (IV) tedizolid phosphate. (B) Area under the concentration-time curve (AUC) for tedizolid after single (AUC0–∞) and multiple (AUC0–24) doses of IV tedizolid phosphate. There were 9, 26, 8, and 9 individuals in the 100-, 200-, 300-, and 400-mg cohorts, respectively. Error bars denote standard deviations.

References

    1. McCaig LF, McDonald LC, Mandal S, Jernigan DB. Staphylococcus aureus–associated skin and soft tissue infections in ambulatory care. Emerg Infect Dis. 2006;12:1715–23.
    1. Prokocimer P, De Anda C, Fang E, Mehra P, Das A. Tedizolid phosphate vs linezolid for treatment of acute bacterial skin and skin structure infections: the ESTABLISH-1 randomized trial. JAMA. 2013;309:559–69.
    1. U.S. Food and Drug Administration. Guidance for industry—acute bacterial skin and skin structure infections: developing drugs for treatment. Available from Guidances/ucm071185.pdf. Effective October 2013. Accessed November 13, 2013.
    1. Gerber JS, Coffin SE, Smathers SA, Zaoutis TE. Trends in the incidence of methicillin-resistant Staphylococcus aureus infection in children's hospitals in the United States. Clin Infect Dis. 2009;49:65–71.
    1. Diekema DJ, Pfaller MA, Schmitz FJ, et al. Survey of infections due to Staphylococcus species: frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, Latin America, Europe, and the Western Pacific region for the SENTRY Antimicrobial Surveillance Program, 1997–1999. Clin Infect Dis. 2001;32(Suppl 2):S114–32.
    1. Dryden MS. Complicated skin and soft tissue infection. J Antimicrob Chemother. 2010;65(Suppl 3):iii35–44.
    1. Prokocimer P, Bien P, Surber J, et al. Phase 2, randomized, double-blind, dose-ranging study evaluating the safety, tolerability, population pharmacokinetics, and efficacy of oral torezolid phosphate in patients with complicated skin and skin structure infections. Antimicrob Agents Chemother. 2011;55:583–92.
    1. Urbina O, Ferrandez O, Espona M, Salas E, Ferrandez I, Grau S. Potential role of tedizolid phosphate in the treatment of acute bacterial skin infections. Drug Des Devel Ther. 2013;7:243–65.
    1. Schaadt R, Sweeney D, Shinabarger D, Zurenko G. In vitro activity of TR-700, the active ingredient of the antibacterial prodrug TR-701, a novel oxazolidinone antibacterial agent. Antimicrob Agents Chemother. 2009;53:3236–9.
    1. Flanagan SD, Bien PA, Munoz KA, Minassian SL, Prokocimer PG. Pharmacokinetics of tedizolid following oral administration: single and multiple dose, effect of food, and comparison of two solid forms of the prodrug. Pharmacotherapy. 2014;24:240–50.
    1. Moran GJ, Fang E, Corey GR, Das AF, De Anda C, Prokocimer P. Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH 2): a randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis 2014 June 6. Epub ahead of print]
    1. Dunn K, O'Reilly A, Silke B, Rogers T, Bergin C. Implementing a pharmacist-led sequential antimicrobial therapy strategy: a controlled before-and-after study. Int J Clin Pharm. 2011;33:208–14.
    1. Tan JS, File TM., Jr Management of community-acquired pneumonia: a focus on conversion from hospital to the ambulatory setting. Am J Respir Med. 2003;2:385–94.
    1. Athanassa Z, Makris G, Dimopoulos G, Falagas ME. Early switch to oral treatment in patients with moderate to severe community-acquired pneumonia: a meta-analysis. Drugs. 2008;68:2469–81.
    1. Jackson A. Infection control—a battle in vein: infusion phlebitis. Nurs Times. 1998;94(68):71.
    1. Fang E, De Anda C, Das A, Prokocimer P. Berlin, Germany: Paper presented at: European Society of Clinical Microbiology and Infectious Diseases; 2013. Efficacy and safety results from the ESTABLISH-2 ABSSSI study comparing IV and oral tedizolid phosphate and linezolid.
    1. Huttunen KM, Raunio H, Rautio J. Prodrugs—from serendipity to rational design. Pharmacol Rev. 2011;63:750–71.
    1. Heimbach T, Oh DM, Li LY, et al. Absorption rate limit considerations for oral phosphate prodrugs. Pharm Res. 2003;20:848–56.

Source: PubMed

3
Sottoscrivi