Novel Bacterial Topoisomerase Inhibitor Gepotidacin Demonstrates Absence of Fluoroquinolone-Like Arthropathy in Juvenile Rats

Cindy Fishman, Jessica M Caverly Rae, Lorraine M Posobiec, Susan B Laffan, Steven A Lerman, Neil Pearson, Salim Janmohamed, Etienne Dumont, Deatra Nunn-Floyd, Dinesh J Stanislaus, Cindy Fishman, Jessica M Caverly Rae, Lorraine M Posobiec, Susan B Laffan, Steven A Lerman, Neil Pearson, Salim Janmohamed, Etienne Dumont, Deatra Nunn-Floyd, Dinesh J Stanislaus

Abstract

Fluoroquinolone use in children is limited due to its potential toxicity and negative effects on skeletal development, but the actual effects/risks of fluoroquinolones on bone growth and the mechanisms behind fluoroquinolone-driven arthropathy remain unknown. Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibiotic with a unique mechanism of action that is not anticipated to have the same risks to bone growth as those of fluoroquinolones. Gepotidacin is in phase III clinical development for uncomplicated urinary tract infections (ClinicalTrials.gov identifiers NCT04020341 and NCT04187144) and urogenital gonorrhea (ClinicalTrials.gov identifier NCT04010539) in adults and adolescents ≥12 years of age. To inform arthropathy and other potential toxicity risks of gepotidacin in pediatric studies, this nonclinical study assessed oral gepotidacin toxicity in juvenile rats from postnatal day (PND) 4 to PND 32/35 (approximately equivalent to human ages from newborn to 11 years), using both in-life assessments (tolerability, toxicity, and toxicokinetics) and terminal assessments (necropsy with macroscopic and microscopic skeletal femoral head and/or stifle joint examinations). Gepotidacin doses of ≤300 mg/kg of body weight/day were well tolerated from PND 4 to PND 21, and higher doses of ≤1,250 mg/kg/day were well tolerated from PND 22 when the dose levels were escalated to maintain systemic exposure levels up to PND 35, with no observed treatment-related clinical signs, effects on mean body weight gain, or macroscopic findings on articular surfaces. A dose of 1,000 mg/kg/day was not tolerated during the dosing period from PND 4 to 21, with effects on body weight gain, fecal consistency, and body condition. Microscopic effects on articular surfaces were evaluated after 32 days of gepotidacin treatment at the highest tolerated dose. After 32 days of treatment with the highest tolerated gepotidacin dose of 300/1,250 mg/kg/day (systemic concentrations [area under the curve {AUC} values] of 93.7 μg · h/mL [males] and 121 μg · h/mL [females]), no skeletal effects on articular surfaces of the femoral head or stifle joint were observed. The absence of treatment-related clinical signs and arthropathy in juvenile rats provides evidence to support the potential future use of gepotidacin in children.

Keywords: antibiotic resistance; arthropathy; children; drug safety; drug toxicity; fluoroquinolone; gepotidacin; juvenile rat; nonclinical study; pediatric; preclinical study.

Conflict of interest statement

The authors declare a conflict of interest. C.F., S.B.L., D.J.S., N.P., S.J., and D.N.-F. are employees of GSK. J.M.C.R., L.M.P., S.A.L., and E.D. are former employees of GSK.

Figures

FIG 1
FIG 1
Round 1 mean area under the curve from time zero to the last measurable concentration (AUC0–t) values in males (A) and females (B) on postnatal day (PND) 13 and PND 21 by gepotidacin dose showing a marked decrease in exposure at PND 21 compared to PND 13. The days on which samples were collected are marked in black text. aFor the purpose of calculating the AUC0–t, the concentrations at time zero were assigned with the concentrations at 24 h. Female parameters for a 1-mg/kg/day gepotidacin dose on PND 13 and male and female parameters for 1- and 10-mg/kg/day gepotidacin doses on PND 21 could not be calculated due to limited measurable data.
FIG 2
FIG 2
Round 2 mean area under the curve from time zero to the last measurable concentration (AUC0–t) values in males (A) and females (B) on postnatal day (PND) 13, PND 22, and PND 35 by gepotidacin dose with dose escalation on PND 22 to maintain systemic exposure as much as possible across the age range. The days on which samples were collected are marked in black text. aFor the purpose of calculating the AUC0–t, the concentrations at time zero were assigned with the concentrations at 24 h.
FIG 3
FIG 3
Round 3 toxicokinetics of mean area under the curve from time zero to the last measurable concentration (AUC0–t) values in males (A) and females (B) on postnatal day (PND) 22 and PND 32 by gepotidacin dose with dose escalation on PND 21 to maintain exposure levels and separation between dose groups as much as possible at the potential maximum tolerated doses. The days on which samples were collected are marked in black text. Additional lines show approximate AUC clinical exposures for uncomplicated urinary tract infection (uUTI) and urogenital gonorrhea (GC) indications (40 μg · h/mL [orange] and 85 μg · h/mL [green], respectively). aFor the purpose of calculating the AUC0–t, the concentrations at time zero were assigned with the concentrations at 24 h.
FIG 4
FIG 4
Normal microscopic appearance of articular cartilage of the distal femur (A) and proximal tibia (B) from control juvenile rats (left) and high-dose gepotidacin (300/1,250 mg/kg/day)-treated juvenile rats (right) dosed from postnatal day (PND) 4 to PND 32, with necropsy on PND 33. Arrows indicate articular cartilage. A subtle tiling artifact in the white background (of the top right panel) was contained within the digital image file and considered related to computer stitching of images by the Aperio Scanscope instrument (Leica Biosystems, Deer Park, IL, USA).
FIG 5
FIG 5
Study design. All rat pups were dosed directly by oral gavage with a gepotidacin suspension (1% aqueous methylcellulose, with a viscosity of 400 cP [2% solution in water]) at a dose volume of 5 mL/kg based on daily body weight. In-life observations included clinical observations and recording of body weight. PND, postnatal day.

References

    1. European Medicines Agency. 2018. Pharmacovigilance risk assessment: quinolone and fluoroquinolone medicinal products for systemic and inhalation use. European Medicines Agency, Amsterdam, The Netherlands. . Accessed 21 May 2021.
    1. Kocsis B, Domokos J, Szabo D. 2016. Chemical structure and pharmacokinetics of novel quinolone agents represented by avarofloxacin, delafloxacin, finafloxacin, zabofloxacin and nemonoxacin. Ann Clin Microbiol Antimicrob 15:34. 10.1186/s12941-016-0150-4.
    1. Ingham B, Brentnall DW, Dale EA, McFadzean JA. 1977. Arthropathy induced by antibacterial fused N-alkyl-4-pyridone-3-carboxylic acids. Toxicol Lett 1:21–26. 10.1016/0378-4274(77)90016-9.
    1. Burkhardt JE, Hill MA, Carlton WW, Kesterson JW. 1990. Histologic and histochemical changes in articular cartilages of immature beagle dogs dosed with difloxacin, a fluoroquinolone. Vet Pathol 27:162–170. 10.1177/030098589002700303.
    1. Machida M, Kusajima H, Aijima H, Maeda A, Ishida R, Uchida H. 1990. Toxicokinetic study of norfloxacin-induced arthropathy in juvenile animals. Toxicol Appl Pharmacol 105:403–412. 10.1016/0041-008x(90)90144-j.
    1. Gough A, Johnson R, Campbell E, Hall L, Tylor J, Carpenter A, Black W, Basrur PK, Baragi VM, Sigler R, Metz A. 1996. Quinolone arthropathy in immature rabbits treated with the fluoroquinolone, PD 117596. Exp Toxicol Pathol 48:225–232. 10.1016/S0940-2993(96)80003-0.
    1. Linseman DA, Hampton LA, Branstetter DG. 1995. Quinolone-induced arthropathy in the neonatal mouse. Morphological analysis of articular lesions produced by pipemidic acid and ciprofloxacin. Fundam Appl Toxicol 28:59–64. 10.1006/faat.1995.1146.
    1. Patel K, Goldman JL. 2016. Safety concerns surrounding quinolone use in children. J Clin Pharmacol 56:1060–1075. 10.1002/jcph.715.
    1. US Food and Drug Administration. 2018. FDA drug safety communication: FDA updates warnings for oral and injectable fluoroquinolone antibiotics due to disabling side effects. US Food and Drug Administration, Silver Spring, MD. . Accessed 28 February 2022.
    1. European Medicines Agency. 2018. Fluoroquinolone and quinolone antibiotics: PRAC recommends new restrictions on use following review of disabling and potentially long-lasting side effects. European Medicines Agency, Amsterdam, The Netherlands. . Accessed 1 March 2022.
    1. US Food and Drug Administration. 2018. FDA warns about increased risk of ruptures or tears in the aorta blood vessel with fluoroquinolone antibiotics in certain patients. US Food and Drug Administration, Silver Spring, MD. . Accessed 28 February 2022.
    1. Pradhan KM, Arora NK, Jena A, Susheela AK, Bhan MK. 1995. Safety of ciprofloxacin therapy in children: magnetic resonance images, body fluid levels of fluoride and linear growth. Acta Paediatr 84:555–560. 10.1111/j.1651-2227.1995.tb13694.x.
    1. Adefurin A, Sammons H, Jacqz-Aigrain E, Choonara I. 2011. Ciprofloxacin safety in paediatrics: a systematic review. Arch Dis Child 96:874–880. 10.1136/adc.2010.208843.
    1. Forsythe CT, Ernst ME. 2007. Do fluoroquinolones commonly cause arthropathy in children? CJEM 9:459–462. 10.1017/s1481803500015517.
    1. Goldman J, Kearns G. 2011. 18th Expert Committee on the Selection and Use of Essential Medicines. Fluoroquinolone use in paediatrics: focus on safety and place in therapy. .
    1. Stahlmann R, Forster C, Shakibaei M, Vormann J, Gunther T, Merker HJ. 1995. Magnesium deficiency induces joint cartilage lesions in juvenile rats which are identical to quinolone-induced arthropathy. Antimicrob Agents Chemother 39:2013–2018. 10.1128/AAC.39.9.2013.
    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. 10.1128/AAC.02820-15.
    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. 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. 10.1038/nature09197.
    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.
    1. Mushtaq S, Vickers A, Sadouki Z, Cole M, Fifer H, Donascimento V, Day M, De Pinna E, Jenkins C, Godbole G, Woodfor 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 Eur Congr Clin Microbiol Infect Dis, Amsterdam, The Netherlands.
    1. GSK. 2019. A study to evaluate efficacy and safety of gepotidacin in the treatment of uncomplicated urinary tract infection (UTI). .
    1. GSK. 2020. Comparative study to evaluate efficacy and safety of gepotidacin to nitrofurantoin in treatment of uncomplicated urinary tract infection (UTI). .
    1. GSK. 2019. A study to evaluate efficacy and safety of gepotidacin compared with ceftriaxone plus azithromycin in the treatment of uncomplicated urogenital gonorrhea. .
    1. Uivarosi V. 2013. Metal complexes of quinolone antibiotics and their applications: an update. Molecules 18:11153–11197. 10.3390/molecules180911153.
    1. Sengupta P. 2013. The laboratory rat: relating its age with human’s. Int J Prev Med 4:624–630.
    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. 10.1128/AAC.01263-21.
    1. Fishman C, Caverly Rae JM, Posobiec LM, Laffan SB, Lerman SA, Pearson N, Janmohamed S, Dumont E, Nunn-Floyd D, Stanislaus DJ. 2022. Novel bacterial topoisomerase inhibitor gepotidacin demonstrates absence of fluoroquinolone-like arthropathy in juvenile rats. Abstr Eur Soc Clin Microbiol Infect Dis-Am Soc Microbiol Joint Conf Drug Dev, Dublin, Ireland. .
    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. 10.3109/00498254.2015.1112933.
    1. Ginsberg G, Hattis D, Miller R, Sonawane B. 2004. Pediatric pharmacokinetic data: implications for environmental risk assessment for children. Pediatrics 113:973–983. 10.1542/peds.113.S3.973.
    1. Zoetis T, Tassinari MS, Bagi C, Walthall K, Hurtt ME. 2003. Species comparison of postnatal bone growth and development. Birth Defects Res B Dev Reprod Toxicol 68:86–110. 10.1002/bdrb.10012.
    1. Burkhardt JE, Walterspiel JN, Schaad UB. 1997. Quinolone arthropathy in animals versus children. Clin Infect Dis 25:1196–1204. 10.1086/516119.
    1. Kato M. 2008. Chondrotoxicity of quinolone antimicrobial agents. J Toxicol Pathol 21:123–131. 10.1293/tox.21.123.
    1. Pfister K, Mazur D, Vormann J, Stahlmann R. 2007. Diminished ciprofloxacin-induced chondrotoxicity by supplementation with magnesium and vitamin E in immature rats. Antimicrob Agents Chemother 51:1022–1027. 10.1128/AAC.01175-06.
    1. Cavusoglu I, Kahveci Z, Noyan S, Minbay Z. 2000. Quinolone arthropathy induced by ofloxacin in juvenile rats: a light microscopic study. Turk J Med Sci 30:441–447.
    1. Lozo E, Riecke K, Schwabe R, Vormann J, Stahlmann R. 2002. Synergistic effect of ofloxacin and magnesium deficiency on joint cartilage in immature rats. Antimicrob Agents Chemother 46:1755–1759. 10.1128/AAC.46.6.1755-1759.2002.
    1. Vormann J, Forster C, Zippel U, Lozo E, Gunther T, Merker H, Stahlmann R. 1997. Effects of magnesium deficiency on magnesium and calcium content in bone and cartilage in developing rats in correlation to chondrotoxicity. Calcif Tissue Int 61:230–238. 10.1007/s002239900328.
    1. Sabharwal V, Marchant CD. 2006. Fluoroquinolone use in children. Pediatr Infect Dis J 25:257–258. 10.1097/01.inf.0000205799.35780.f3.
    1. Bidell MR, Lodise TP. 2016. Fluoroquinolone-associated tendinopathy: does levofloxacin pose the greatest risk? Pharmacotherapy 36:679–693. 10.1002/phar.1761.
    1. Dirks RC, Warden SJ. 2011. Models for the study of tendinopathy. J Musculoskelet Neuronal Interact 11:141–149.
    1. Bolon B. 2017. Mini-review: toxic tendinopathy. Toxicol Pathol 45:834–837. 10.1177/0192623317711614.
    1. Kato M, Takada S, Kashida Y, Nomura M. 1995. Histological examination on Achilles tendon lesions induced by quinolone antibacterial agents in juvenile rats. Toxicol Pathol 23:385–392. 10.1177/019262339502300315.
    1. Kashida Y, Kato M. 1997. Characterization of fluoroquinolone-induced Achilles tendon toxicity in rats: comparison of toxicities of 10 fluoroquinolones and effects of anti-inflammatory compounds. Antimicrob Agents Chemother 41:2389–2393. 10.1128/AAC.41.11.2389.
    1. European Medicines Agency. 2020. ICH guideline S11 on nonclinical safety testing in support of development of paediatric pharmaceuticals—step 5. European Medicines Agency, Amsterdam, The Netherlands. . Accessed 3 August 2022.
    1. GSK. 2022. Use of animals. GSK, Brentford, Middlesex, United Kingdom. .
    1. Posobiec LM, Laffan SB. 2021. Dose range finding approach for rodent preweaning juvenile animal studies. Birth Defects Res 113:409–426. 10.1002/bdr2.1856.

Source: PubMed

3
S'abonner