Bone and Joint Tissue Penetration of the Staphylococcus-Selective Antibiotic Afabicin in Patients Undergoing Elective Hip Replacement Surgery

Annick Menetrey, Annick Janin, John Pullman, J Scott Overcash, Amina Haouala, François Leylavergne, Laurent Turbe, Frederick Wittke, Valérie Nicolas-Métral, Annick Menetrey, Annick Janin, John Pullman, J Scott Overcash, Amina Haouala, François Leylavergne, Laurent Turbe, Frederick Wittke, Valérie Nicolas-Métral

Abstract

Afabicin (formerly Debio 1450, AFN-1720) is a prodrug of afabicin desphosphono (Debio 1452, AFN-1252), a novel antibiotic in development which targets the staphylococcal enoyl-acyl carrier protein reductase (FabI) and exhibits selective potent antibacterial activity against staphylococcal species, including methicillin-resistant Staphylococcus aureus As part of clinical development in bone and joint infections, a distribution study in bone was performed in 17 patients who underwent elective hip replacement surgery. Patients received 3 doses of 240 mg afabicin orally (every 12 h) at various time points before surgery. Afabicin desphosphono concentrations were measured by liquid chromatography-tandem mass spectrometry in plasma, cortical bone, cancellous bone, bone marrow, soft tissue, and synovial fluid collected during surgery at 2, 4, 6, or 12 h after the third afabicin dose. The study showed good penetration of afabicin desphosphono into bone tissues, with mean area under the curve ratios for cortical bone-, cancellous bone-, bone marrow-, soft tissue-, and synovial fluid-to-total plasma concentrations of 0.21, 0.40, 0.32, 0.35, and 0.61, respectively. When accounting for the free fraction in plasma (2%) and synovial fluid (9.4%), the mean ratio was 2.88, which is indicative of excellent penetration and which showed that the afabicin desphosphono concentration was beyond the MIC90 of S. aureus over the complete dosing interval. These findings, along with preclinical efficacy data, clinical efficacy data for skin and soft tissue staphylococcal infection, the availability of both intravenous and oral formulations, and potential advantages over broad-spectrum antibiotics for the treatment of staphylococcal bone or joint infections, support the clinical development of afabicin for bone and joint infections. (This study has been registered at ClinicalTrials.gov under identifier NCT02726438.).

Keywords: Debio 1450; Staphylococcus aureus; afabicin; drug development; drug penetration; joint infections; osteomyelitis; pharmacokinetics.

Copyright © 2019 Menetrey et al.

Figures

FIG 1
FIG 1
Profile of the afabicin desphosphono concentration in plasma after the third oral dose of afabicin (240 mg, q12h) in patients undergoing hip replacement surgery. The results are presented as the geometric mean ± SD obtained using data from nominal time points (number of patients = 14).
FIG 2
FIG 2
Individual afabicin desphosphono concentrations in plasma (A), cortical bone (B), cancellous bone (C), synovial fluid (D), bone marrow (E), and soft tissue (F) after the third oral dose of afabicin (240 mg, q12h) in patients undergoing hip replacement surgery. Data are from 14 patients, with the following exceptions: for 1 patient, the plasma sample was missing at the time of resection and the plasma concentration was extrapolated by linear regression between the two adjacent concentration data from the full PK profile in plasma. Synovial fluid was obtained for 8 patients only. For one patient no results except for plasma were reportable at the time of resection, and for another, only plasma, soft tissue, and bone marrow were collected at the time of resection.
FIG 3
FIG 3
Osteoarticular tissue- and fluid-to-plasma concentration ratios obtained for vancomycin, linezolid, ertapenem, daptomycin, dalbavancin, and afabicin. Mean ratios (median ratios for ertapenem) were obtained from published data from studies with designs similar to that of this study, i.e., dosing in patients undergoing elective replacement surgery and analysis of the total concentration of the drugs in homogenized bone tissues or synovial fluid collected during surgery (20–25) at steady state for linezolid and afabicin and after a single dose for the other antibiotics. Vancomycin, linezolid, and ertapenem were quantified in serum, whereas daptomycin, dalbavancin, and afabicin desphosphono were quantified in plasma.

References

    1. Landersdorfer CB, Bulitta JB, Kinzig M, Holzgrabe U, Soergel F. 2009. Penetration of antibacterials into bone: pharmacokinetic, pharmacodynamic and bioanalytical considerations. Clin Pharmacokinet 48:89–124. doi:10.2165/00003088-200948020-00002.
    1. Sheehy SH, Atkins BA, Bejon P, Byren I, Wyllie D, Athanasou NA, Berendt AR, McNally MA. 2010. The microbiology of chronic osteomyelitis: prevalence of resistance to common empirical anti-microbial regimens. J Infect 60:338–343. doi:10.1016/j.jinf.2010.03.006.
    1. Kremers HM, Nwojo ME, Ransom JE, Wood-Wentz CM, Melton LJ, Huddleston PM. 2015. Trends in the epidemiology of osteomyelitis: a population-based study, 1969 to 2009. J Bone Joint Surg Am 97:837–845. doi:10.2106/JBJS.N.01350.
    1. Tong SY, Davis JS, Eichenberger E, Holland TL, Fowler VG. 2015. Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clin Microbiol Rev 28:603–661. doi:10.1128/CMR.00134-14.
    1. Tande AJ, Patel R. 2014. Prosthetic joint infection. Clin Microbiol Rev 27:302–345. doi:10.1128/CMR.00111-13.
    1. Sana TG, Monack DM. 2016. Microbiology: the dark side of antibiotics. Nature 534:624–625. doi:10.1038/nature18449.
    1. Pavia AT, Shipman LD, Wells JG, Puhr ND, Smith JD, McKinley TW, Tauxe RV. 1990. Epidemiologic evidence that prior antimicrobial exposure decreases resistance to infection by antimicrobial-sensitive Salmonella. J Infect Dis 161:255–260. doi:10.1093/infdis/161.2.255.
    1. Kelly CP, Pothoulakis C, LaMont JT. 1994. Clostridium difficile colitis. N Engl J Med 330:257–262. doi:10.1056/NEJM199401273300406.
    1. Mathews CJ, Weston VC, Jones A, Field M, Coakley G. 2010. Bacterial septic arthritis in adults. Lancet 375:846–855. doi:10.1016/S0140-6736(09)61595-6.
    1. Hawser S, Gueny M, Rochat I, Morrissey I, Magnet S, Dieppois G. 2018. Activity of Debio 1452 against Staphylococcus spp. collected in 2015/2016, abstr P1824 Abstr 28th Eur Congr Clin Microbiol Infect Dis.
    1. Barbier M, Menetrey A, Haouala A, Dieppois G, Wittke F, Vuagniaux G. 2016. Efficacy of the FabI inhibitor Debio 1450 for the treatment of Staphylococcus aureus-induced osteomyelitis in an experimental rat model, abstr O496 Abstr 26th Eur Congr Clin Microbiol Infect Dis.
    1. Barbier M, Menetrey A, Haouala A, Bravo J, Wittke F, Jacqueline C, Vuagniaux G. 2016. Efficacy of the FabI inhibitor afabicin for the treatment of Staphylococcus aureus-induced acute osteomyelitis in rabbit, abstr 4495 Abstr ASM Microbe, Boston, MA. American Society for Microbiology, Washington, DC.
    1. Dyon-Tafani V, Dieppois G, Cara A, Monteix A, Valour F, Josse J, Ferry T, Laurent F. 2018. Activity of Debio 1452 on intracellular Staphylococcus aureus in osteoblasts in vitro, abstr 5462 Abstr ASM Microbe, Atlanta, GA. American Society for Microbiology, Washington, DC.
    1. Nowakowska J, Kuehn J, Vincent C, Wittke FL, Fresne-Languille S, Carton T, Leuillet S, Le Vacon F, Vuagniaux G. 2018. Staphylococcal-selective antibiotic preserves the human gut microbiota, abstr 2471 Abstr 28th Eur Congr Clin Microbiol Infect Dis.
    1. Fluhler E, Vazvaei F, Singhal P, Vinck P, Li W, Bhatt J, de Boer T, Chaudhary A, Tangiuchi M, Rezende V, Zhong D. 2014. Repeat analysis and incurred sample reanalysis: recommendation for best practices and harmonization from the Global Bioanalysis Consortium Harmonization Team. AAPS J 16:1167–1174. doi:10.1208/s12248-014-9644-1.
    1. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz RJ, Kaplan SL, Karchmer AW, Levine DP, Murray BE, Rybak JM, Talan DA, Chambers HF, Infectious Diseases Society of America. 2011. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin Infect Dis 52:e18–e55. doi:10.1093/cid/ciq146.
    1. Berbari EF, Kanj SS, Kowalski TJ, Darouiche RO, Widmer AF, Schmitt SK, Hendershot EF, Holtom PD, Huddleston PM, Petermann GW, Osmon DR. 2015. Infectious Diseases Society of America (IDSA) clinical practice guidelines for the diagnosis and treatment of native vertebral osteomyelitis in adults. Clin Infect Dis 61:e26–e46. doi:10.1093/cid/civ482.
    1. Fetterly GL, Ong CM, Bhavnani SM, Loutit JS, Porter SB, Morello LG, Ambrose PG, Nicolau DP. 2005. Pharmacokinetics of oritavancin in plasma and skin blister fluid following administration of a 200-milligram dose for 3 days or a single 800-milligram dose. Antimicrob Agents Chemother 49:148–152. doi:10.1128/AAC.49.1.148-152.2005.
    1. Sun HK, Ong CT, Umer A, Harper D, Troy S, Nightingale CH, Nicolau DP. 2005. Pharmacokinetic profile of tigecycline in serum and skin blister fluid of healthy subjects after multiple intravenous administrations. Antimicrob Agents Chemother 49:1629–1632. doi:10.1128/AAC.49.4.1629-1632.2005.
    1. Graziani AL, Lawson LA, Gibson GA, Steinberg MA, MacGregor RR. 1988. Vancomycin concentrations in infected and noninfected human bone. Antimicrob Agents Chemother 32:1320–1322. doi:10.1128/AAC.32.9.1320.
    1. Roy ME, Peppers MP, Whiteside LA, LaZear RM. 2014. Vancomycin concentration in synovial fluid: direct injection into the knee vs. intravenous infusion. J Arthroplasty 29:564–568. doi:10.1016/j.arth.2013.08.017.
    1. Rana B, Butcher I, Grigoris P, Murnaghan C, Seaton RA, Tobin CM. 2002. Linezolid penetration into osteo-articular tissues. J Antimicrob Chemother 50:747–750. doi:10.1093/jac/dkf207.
    1. Boselli E, Breilh D, Djabarouti S, Bel JC, Saux MC, Allaouchiche B. 2007. Diffusion of ertapenem into bone and synovial tissues. J Antimicrob Chemother 6:893–896. doi:10.1093/jac/dkm296.
    1. Montange D, Berthier F, Leclerc G, Serre A, Jeunet L, Berard M, Muret P, Vettoretti L, Leroy J, Hoen B, Chirouze C. 2014. Penetration of daptomycin into bone and synovial fluid in joint replacement. Antimicrob Agents Chemother 58:3991–3996. doi:10.1128/AAC.02344-14.
    1. Dunne MW, Puttagunta S, Sprenger CR, Rubino C, Van Wart S, Baldassarre J. 2015. Extended-duration dosing and distribution of dalbavancin into bone and articular tissue. Antimicrob Agents Chemother 59:1849–1855. doi:10.1128/AAC.04550-14.
    1. Landersdoerfer CB, Kinzig M, Bulitta JB. 2009. Bone penetration of amoxicillin and clavulanic acid evaluation by population pharmacokinetics and Monte-Carlo simulation. Antimicrob Agents Chemother 53:2569–2578. doi:10.1128/AAC.01119-08.
    1. Stolle LB, Plock N, Joukhadar C, Arpi M, Emmertsen KJ, Buerger C, Riegels-Nielsen P, Kloft C. 2008. Pharmacokinetics of linezolid in bone tissue investigated by in vivo microdialysis. Scand J Infect Dis 40:24–29. doi:10.1080/00365540701509873.
    1. Housman ST, Bhalodi AA, Shepard A, Nugent J, Nicolau DP. 2015. Vancomycin tissue pharmacokinetics in patients with lower-limb infections via in vivo microdialysis. J Am Podiatr Med Assoc 105:381–388. doi:10.7547/14-033.
    1. Traunmuller F, Schintler MV, Metzler J, Spendel S, Mauric O, Popovic M, Konz KH, Scharnagl E, Joukhadar C. 2010. Soft tissue and bone penetration abilities of daptomycin in diabetic patients with bacterial foot infections. J Antimicrob Chemother 65:1252–1257. doi:10.1093/jac/dkq109.
    1. Traunmuller F, Schintler MV, Spendel S, Popovic M, Mauric O, Scharnagl E, Joukhadar C. 2010. Linezolid concentrations in infected soft tissue and bone following repetitive doses in diabetic patients with bacterial foot infections. Int J Antimicrob Agents 36:84–86. doi:10.1016/j.ijantimicag.2010.03.007.
    1. Bader JC, Lakota EA, Bravo J, Dieppois G, Nicolas-Métral V, Miesel L, Lin KY, Ambrose PG, Bhavnani SM. 2017. Pharmacokinetic-pharmacodynamic analyses for Debio 1450 (afabicin), a staphylococcal-specific antibiotic, using data from a murine-thigh infection model, abstr 2176. Abstr ASM Microbe, New Orleans, LA. American Society for Microbiology, Washington, DC.
    1. EMA. 2011. Guideline on bioanalytical method validation. EMEA/CHMP/EWP/192217/2009, revision 1, corrections 2 EMA, London, United Kingdom.
    1. Singh JK, Solanki A, Maniyar RC, Banerjee D, Shirsath VS. 2006. Rapid equilibrium dialysis (RED): an in-vitro high-throughput screening technique for plasma protein binding using human and rat plasma. J Chromatogr A 1102:125–134. doi:10.4172/jbb.S14-005.

Source: PubMed

3
Tilaa