Dose Selection for Phase III Clinical Evaluation of Gepotidacin (GSK2140944) in the Treatment of Uncomplicated Urinary Tract Infections

Nicole E Scangarella-Oman, Mohammad Hossain, Jennifer L Hoover, Caroline R Perry, Courtney Tiffany, Aline Barth, Etienne F Dumont, Nicole E Scangarella-Oman, Mohammad Hossain, Jennifer L Hoover, Caroline R Perry, Courtney Tiffany, Aline Barth, Etienne F Dumont

Abstract

Antibiotics are the current standard-of-care treatment for uncomplicated urinary tract infections (uUTIs). However, increasing rates of bacterial antibiotic resistance necessitate novel therapeutic options. Gepotidacin is a first-in-class triazaacenaphthylene antibiotic that selectively inhibits bacterial DNA replication by interaction with the bacterial subunits of DNA gyrase (GyrA) and topoisomerase IV (ParC). Gepotidacin is currently in clinical development for the treatment of uUTIs and other infections. In this article, we review data for gepotidacin from nonclinical studies, including in vitro activity, in vivo animal efficacy, and pharmacokinetic (PK) and pharmacokinetic/pharmacodynamic (PK/PD) models that informed dose selection for phase III clinical evaluation of gepotidacin. Based on this translational package of data, a gepotidacin 1,500-mg oral dose twice daily for 5 days was selected for two ongoing, randomized, multicenter, parallel-group, double-blind, double-dummy, active-comparator phase III clinical studies evaluating the safety and efficacy of gepotidacin in adolescent and adult female participants with uUTIs (ClinicalTrials.gov identifiers NCT04020341 and NCT04187144).

Keywords: acute cystitis; acute uncomplicated cystitis; antibacterial; dose selection; gepotidacin; pharmacodynamics; pharmacokinetics; uncomplicated urinary tract infection.

Conflict of interest statement

The authors declare a conflict of interest. All authors report employment with, and stock/share ownership in, GlaxoSmithKline plc. at the time of study. M.H. is currently employed by Adios Pharmaceuticals, Cambridge, MA. C.T. is currently employed by Gan and Lee Pharmaceuticals, Bridgewater, NJ. A.B. is currently employed by Global Blood Therapeutics, San Francisco, CA. E.F.D. is currently employed by Boston Pharmaceuticals, Cambridge, MA.

All authors report employment with, and stock/share ownership in, GlaxoSmithKline plc at the time of the study.

Figures

FIG 1
FIG 1
Frequency distribution of GEP MICs (micrograms per milliliter) against 1,010 E. coli isolates, including those resistant to levofloxacin (25.9% resistant isolates), fosfomycin (0.3%), nitrofurantoin (0.6%), or trimethoprim-sulfamethoxazole (37.7%) or positive for the ESBL phenotype (17.7%) (15). The ESBL phenotype was based on a ceftriaxone MIC of ≥2 μg/mL, according to CLSI guidelines (15, 33). (Based on data from reference .)
FIG 2
FIG 2
Reductions in bacterial burdens of four strains of multidrug-resistant E. coli after 4 days of treatment in rat kidneys (A) and time course of the efficacy of gepotidacin (GEP) at 1,500 mg q12h in rat kidneys (B) (25). Rats were infected by injection of E. coli directly into each kidney. At 2 h postinfection, the administration of gepotidacin was initiated using a continuous i.v. infusion into preimplanted jugular catheters. Infusion rates were controlled by preprogrammed infusion pumps, and flow rates varied over time to create systemic PK profiles in rats that mimicked systemic PK profiles measured in humans (both corrected for differences in protein binding); mean PK profiles from human oral doses of 800 mg and/or 1,500 mg q12h were tested. *, P < 0.05; **, P < 0.01 (representing a statistically significant reduction versus the 2-h baseline controls). Note that gepotidacin had previously demonstrated MICs of 2 to 4 μg/mL against the four strains of multidrug-resistant E. coli. LLQ, lower limit of quantification; LVX, levofloxacin; PO, oral; q12h, every 12 h; q24h, every 24 h. (Redrawn from reference .)
FIG 3
FIG 3
Relationship between gepotidacin exposure and change in log10 CFU per milliliter from the baseline of the gepotidacin-resistant subpopulation for E. coli NCTC 13441 on day 10. (Redrawn from reference .)
FIG 4
FIG 4
Median gepotidacin urine concentration-time plot following oral administration of gepotidacin at 1,500 mg BID from the phase IIa uUTI study (ClinicalTrials.gov identifier NCT03568942) in 22 participants with uUTIs (8). The lower limit of quantification, represented by the dashed line, was 1.00 μg/mL. Data are plotted by the planned relative midpoint time for each interval. Notes that based on trough predose plasma concentrations of gepotidacin and statistical analysis, plasma steady state was achieved by day 3. One participant received nine doses of the study treatment. The second dose of the study treatment was not administered due to a study treatment administration protocol deviation by the site. (Redrawn from reference .)

References

    1. Ennis SS, Guo H, Raman L, Tambyah PA, Chen SL, Tiong HY. 2018. Premenopausal women with recurrent urinary tract infections have lower quality of life. Int J Urol 25:684–689. doi:10.1111/iju.13698.
    1. Wagenlehner F, Wullt B, Ballarini S, Zingg D, Naber KG. 2018. Social and economic burden of recurrent urinary tract infections and quality of life: a patient Web-based study (GESPRIT). Expert Rev Pharmacoecon Outcomes Res 18:107–117. doi:10.1080/14737167.2017.1359543.
    1. Flores-Mireles AL, Walker JN, Caparon M, Hultgren SJ. 2015. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol 13:269–284. doi:10.1038/nrmicro3432.
    1. Geerlings SE. 2016. Clinical presentations and epidemiology of urinary tract infections. Microbiol Spectr 4:UTI-0002-2012. doi:10.1128/microbiolspec.UTI-0002-2012.
    1. Gupta K, Hooton TM, Naber KG, Wullt B, Colgan R, Miller LG, Moran GJ, Nicolle LE, Raz R, Schaeffer AJ, Soper DE, Infectious Diseases Society of America, European Society for Microbiology and Infectious Diseases. 2011. International clinical practice guidelines for the treatment of acute uncomplicated cystitis and pyelonephritis in women: a 2010 update by the Infectious Diseases Society of America and the European Society for Microbiology and Infectious Diseases. Clin Infect Dis 52:e103–e120. doi:10.1093/cid/ciq257.
    1. Tacconelli E, Cataldo MA, Dancer SJ, De Angelis G, Falcone M, Frank U, Kahlmeter G, Pan A, Petrosillo N, Rodríguez-Baño J, Singh N, Venditti M, Yokoe DS, Cookson B, European Society of Clinical Microbiology. 2014. ESCMID guidelines for the management of the infection control measures to reduce transmission of multidrug-resistant Gram-negative bacteria in hospitalized patients. Clin Microbiol Infect 20(Suppl 1):1–55. doi:10.1111/1469-0691.12427.
    1. Orsi GB, Falcone M, Venditti M. 2011. Surveillance and management of multidrug-resistant microorganisms. Expert Rev Anti Infect Ther 9:653–679. doi:10.1586/eri.11.77.
    1. Overcash JS, Tiffany CA, Scangarella-Oman NE, Perry CR, Tao Y, Hossain M, Barth A, Dumont EF. 2020. Phase 2a pharmacokinetic, safety, and exploratory efficacy evaluation of oral gepotidacin (GSK2140944) in female participants with uncomplicated urinary tract infection (acute uncomplicated cystitis). Antimicrob Agents Chemother 64:e00199-20. doi:10.1128/AAC.00199-20.
    1. Ironmonger D, Edeghere O, Bains A, Loy R, Woodford N, Hawkey PM. 2015. Surveillance of antibiotic susceptibility of urinary tract pathogens for a population of 5.6 million over 4 years. J Antimicrob Chemother 70:1744–1750. doi:10.1093/jac/dkv043.
    1. Lee DS, Lee SJ, Choe HS. 2018. Community-acquired urinary tract infection by Escherichia coli in the era of antibiotic resistance. Biomed Res Int 2018:7656752. doi:10.1155/2018/7656752.
    1. Stapleton AE, Wagenlehner FME, Mulgirigama A, Twynholm M. 2020. Escherichia coli resistance to fluoroquinolones in community-acquired uncomplicated urinary tract infection in women: a systematic review. Antimicrob Agents Chemother 64:e00862-20. doi:10.1128/AAC.00862-20.
    1. World Health Organization. 2001. WHO global strategy for containment of antimicrobial resistance. World Health Organization, Geneva, Switzerland. .
    1. Gibson EG, Bax B, Chan PF, Osheroff N. 2019. Mechanistic and structural basis for the actions of the antibacterial gepotidacin against Staphylococcus aureus gyrase. ACS Infect Dis 5:570–581. doi:10.1021/acsinfecdis.8b00315.
    1. Bax BD, Chan PF, Eggleston DS, Fosberry A, Gentry DR, Gorrec F, Giordano I, Hann MM, Hennessy A, Hibbs M, Huang J, Jones E, Jones J, Brown KK, Lewis CJ, May EW, Saunders MR, Singh O, Spitzfaden CE, Shen C, Shillings A, Theobald AJ, Wohlkonig A, Pearson ND, Gwynn MN. 2010. Type IIA topoisomerase inhibition by a new class of antibacterial agents. Nature 466:935–940. doi:10.1038/nature09197.
    1. Biedenbach DJ, Bouchillon SK, Hackel M, Miller LA, Scangarella-Oman NE, Jakielaszek C, Sahm DF. 2016. In vitro activity of gepotidacin, a novel triazaacenaphthylene bacterial topoisomerase inhibitor, against a broad spectrum of bacterial pathogens. Antimicrob Agents Chemother 60:1918–1923. doi:10.1128/AAC.02820-15.
    1. Mushtaq S, Vickers A, Sadouki Z, Cole M, Fifer H, Donascimento V, Day M, De Pinna E, Jenkins C, Godbole G, Woodford N. 2019. In vitro activities of gepotidacin, a novel triazaacenaphthylene and topoisomerase IV DNA gyrase inhibitor, against Gram-negative bacteria and Staphylococcus saprophyticus, poster P1849. Abstr 29th ECCMID, Amsterdam, The Netherlands.
    1. . 2019. NCT04020341: a study to evaluate efficacy and safety of gepotidacin in the treatment of uncomplicated urinary tract infection (UTI). .
    1. . 2020. NCT04187144: comparative study to evaluate efficacy and safety of gepotidacin to nitrofurantoin in treatment of uncomplicated urinary tract infection (UTI). .
    1. Clinical and Laboratory Standards Institute. 2012. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically; approved standard, 9th ed, M07-A9. Clinical and Laboratory Standards Institute, Wayne, PA.
    1. Bulitta JB, Hope WW, Eakin AE, Guina T, Tam VH, Louie A, Drusano GL, Hoover JL. 2019. Generating robust and informative nonclinical in vitro and in vivo bacterial infection model efficacy data to support translation to humans. Antimicrob Agents Chemother 63:e02307-18. doi:10.1128/AAC.02307-18.
    1. Leekha S, Terrell CL, Edson RS. 2011. General principles of antimicrobial therapy. Mayo Clin Proc 86:156–167. doi:10.4065/mcp.2010.0639.
    1. Barber AE, Norton JP, Wiles TJ, Mulvey MA. 2016. Strengths and limitations of model systems for the study of urinary tract infections and related pathologies. Microbiol Mol Biol Rev 80:351–367. doi:10.1128/MMBR.00067-15.
    1. O’Brien VP, Dorsey DA, Hannan TJ, Hultgren SJ. 2018. Host restriction of Escherichia coli recurrent urinary tract infection occurs in a bacterial strain-specific manner. PLoS Pathog 14:e1007457. doi:10.1371/journal.ppat.1007457.
    1. Hannan TJ, Mysorekar IU, Hung CS, Isaacson-Schmid ML, Hultgren SJ. 2010. Early severe inflammatory responses to uropathogenic E. coli predispose to chronic and recurrent urinary tract infection. PLoS Pathog 6:e1001042. doi:10.1371/journal.ppat.1001042.
    1. Hoover JL, Singley CM, Elefante P, Rittenhouse S. 2019. Efficacy of human exposures of gepotidacin (GSK2140944) against Escherichia coli in a rat pyelonephritis model. Antimicrob Agents Chemother 63:e00086-19. doi:10.1128/AAC.00086-19.
    1. Hossain M, Tiffany CA, McDonald M, Dumont EF. 2014. Safety and pharmacokinetics of repeat escalating oral doses of GSK2140944: a novel bacterial topoisomerase inhibitor, poster F-276. Abstr 54th Intersci Conf Antimicrob Agents Chemother, Washington, DC.
    1. European Medicines Agency. 2016. Guideline on the use of pharmacokinetics and pharmacodynamics in the development of antimicrobial medicinal products (EMA/CHMP/594085/2015). European Medicines Agency, London, United Kingdom. .
    1. Velkov T, Bergen PJ, Lora-Tamayo J, Landersdorfer CB, Li J. 2013. PK/PD models in antibacterial development. Curr Opin Microbiol 16:573–579. doi:10.1016/j.mib.2013.06.010.
    1. Nielsen EI, Cars O, Friberg LE. 2011. Pharmacokinetic/pharmacodynamic (PK/PD) indices of antibiotics predicted by a semimechanistic PKPD model: a step toward model-based dose optimization. Antimicrob Agents Chemother 55:4619–4630. doi:10.1128/AAC.00182-11.
    1. VanScoy BD, Lakota EA, Conde H, Fikes S, Bhavnani SM, Elefante PB, Scangarella-Oman NE, Ambrose PG. 2021. Gepotidacin pharmacokinetics-pharmacodynamics against Escherichia coli in the one-compartment and hollow-fiber in vitro infection model systems. Antimicrob Agents Chemother 65:e00122-21. doi:10.1128/AAC.00122-21.
    1. Bulik CC, Okusanya ÓO, Lakota EA, Forrest A, Bhavnani SM, Hoover JL, Andes DR, Ambrose PG. 2017. Pharmacokinetic-pharmacodynamic evaluation of gepotidacin against Gram-positive organisms using data from murine infection models. Antimicrob Agents Chemother 61:e00115-16. doi:10.1128/AAC.00115-16.
    1. Rizk ML, Zou L, Savic RM, Dooley KE. 2017. Importance of drug pharmacokinetics at the site of action. Clin Transl Sci 10:133–142. doi:10.1111/cts.12448.
    1. Clinical and Laboratory Standards Institute. 2021. Performance standards for antimicrobial susceptibility testing, 31st ed. CLSI supplement M100. Clinical and Laboratory Standards Institute, Wayne, PA.
    1. European Committee on Antimicrobial Susceptibility Testing. 2020. Breakpoint tables for interpretation of MICs and zone diameters. Version 10.0. .
    1. . 2018. NCT02853435. To assess bioavailability, food effect and pharmacokinetics of gepotidacin tablets: a phase I, single-dose, 2 part study in healthy subjects. .
    1. Barth A, Hossain M, Brimhall DB, Perry CR, Tiffany CA, Xu S, Dumont EF. 2022. Pharmacokinetics of oral formulations of gepotidacin (GSK2140944), a triazaacenaphthylene bacterial type II topoisomerase inhibitor, in healthy adult and adolescent participants. Antimicrob Agents Chemother 66:e01263-21. doi:10.1128/AAC.01263-21.
    1. Hossain M, Tiffany C, Raychaudhuri A, Nguyen D, Tai G, Alcorn H, Jr, Preston RA, Marbury T, Dumont E. 2020. Pharmacokinetics of gepotidacin in renal impairment. Clin Pharmacol Drug Dev 9:560–572. doi:10.1002/cpdd.807.
    1. Hossain M, Tiffany C, Tao Y, Barth A, Marbury TC, Preston RA, Dumont E. 2021. Pharmacokinetics of gepotidacin in subjects with normal hepatic function and hepatic impairment. Clin Pharmacol Drug Dev 10:588–597. doi:10.1002/cpdd.913.
    1. Negash K, Andonian C, Felgate C, Chen C, Goljer I, Squillaci B, Nguyen D, Pirhalla J, Lev M, Schubert E, Tiffany C, Hossain M, Ho M. 2016. The metabolism and disposition of GSK2140944 in healthy human subjects. Xenobiotica 46:683–702. doi:10.3109/00498254.2015.1112933.
    1. Scangarella-Oman NE, Hossain M, Hoover JL, Perry CR, Tiffany C, Gardiner D, Dumont EF. 2021. Dose selection for phase 3 studies evaluating gepotidacin (GSK2140944) in the treatment of uncomplicated urinary tract infections, abstr 478. Abstr 31st ECCMID.
    1. Zimmerman EI, Dumont E, Perry C, Tiffany C, Scangarella-Oman N, Raychaudhuri A, Hossain M. 2019. Plasma and urine pharmacokinetic analysis of gepotidacin (GSK2140944) following BID oral dosing in a phase IIa study for treatment of uncomplicated urinary tract infections, poster 1480. Abstr IDWeek 2019, Washington, DC.

Source: PubMed

3
Abonnieren