Ceftobiprole versus daptomycin in Staphylococcus aureus bacteremia: a novel protocol for a double-blind, Phase III trial

Kamal Hamed, Marc Engelhardt, Mark E Jones, Mikael Saulay, Thomas L Holland, Harald Seifert, Vance G Fowler Jr, Kamal Hamed, Marc Engelhardt, Mark E Jones, Mikael Saulay, Thomas L Holland, Harald Seifert, Vance G Fowler Jr

Abstract

Although Staphylococcus aureus is a common cause of bacteremia, treatment options are limited. The need for new therapies is particularly urgent for methicillin-resistant S. aureus bacteremia (SAB). Ceftobiprole is an advanced-generation, broad-spectrum cephalosporin with activity against both methicillin-susceptible and -resistant S. aureus. This is a Phase III, randomized, double-blind, active-controlled, parallel-group, multicenter, two-part study to establish the efficacy and safety of ceftobiprole compared with daptomycin in the treatment of SAB, including infective endocarditis. Anticipated enrollment is 390 hospitalized adult patients, aged ≥18 years, with confirmed or suspected complicated SAB. The primary end point is overall success rate. Target completion of the study is in the second half of 2021. Clinicaltrials.gov identifier: NCT03138733.

Keywords: Phase III; Staphylococcus aureus; bacteremia; ceftobiprole; daptomycin; double-blind; infective endocarditis; noninferiority; protocol; randomized.

Conflict of interest statement

Financial & competing interests disclosure

The study is funded in part with federal funds from the Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response and Biomedical Advanced Research and Development Authority (BARDA), under Contract No. HHSO100201600002C, and in part by Basilea Pharmaceutica International Ltd, Basel, Switzerland. K Hamed, M Engelhardt, ME Jones and M Saulay are employees of Basilea Pharmaceutica International Ltd. TL Holland is a scientific advisory board member for Motif Bio, and consultant for Basilea Pharmaceutica, Genentech, Motif Bio, Roivant and Theravance. H Seifert has received grants or research support from Accelerate, Cubist, Entasis, German Research Foundation (DFG), German Centre for Infection Research (DZIF), Novartis and Tetraphase; is a consultant for Basilea Pharmaceutica, Entasis, Genentech, MSD, Roche, Shionogi and Tetraphase; and has received payments for lectures from Gilead and Merck/MSD. VG Fowler Jr has received grants or research support from Advanced Liquid Logics, Affinergy, Basilea Pharmaceutica, Cerexa/Forest/Actavis/Allergan, ContraFect, Cubist/Merck, Genentech, Karius, Locus, Medical BioSurfaces, MedImmune, Novartis, Pfizer, Regeneron and Theravance; is a consultant for Achaogen, Affinergy, Affinium, Basilea Pharmaceutica, Bayer, Cerexa, ContraFect, Cubist, Debiopharm, Destiny, Durata, Galderma, Genentech, Janssen, Medicines Co., MedImmune, NovaDigm, Novartis, Pfizer, Regeneron, Tetraphase, Theravance, Trius and xBiotech; is Merck Co-Chair for V710 Vaccine; and has received educational fees from Cerexa, Cubist, Debiopharm, Durata, Green Cross and Theravance. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Medical writing support, under the direction of the authors, was provided by Stephanie Carter of Spirit Medical Communications Group Ltd, funded by Basilea Pharmaceutica International Ltd.

Figures

Figure 1.. Study design.
Figure 1.. Study design.
†Patients in Part 1 (N = 80) will receive active treatment for 21–28 days. For Part 2, maximum duration of therapy may be extended to 42 days, pending the outcome of the interim safety analysis and implementation of a protocol amendment. ‡Patients in the ceftobiprole group with Gram-negative infections receive placebo to maintain blinding. §EOT visit to be conducted within 72 h of last study-drug administration. EOT: End of treatment; IE: Infective endocarditis; PTE: Post-treatment evaluation; R: Randomization; SAB: Staphylococcus aureus bacteremia.
Figure 2.. Random effects meta-analysis of clinical…
Figure 2.. Random effects meta-analysis of clinical outcome (all-cause mortality) in untreated patients with Staphylococcus aureus bacteremia.
Seven original articles [36–42] were identified that described outcomes in case series of SAB in patients who did not receive antibacterial treatments. These studies were included in a random effects meta-analysis, which suggests an all-cause mortality of 76%. The lower bound of the 95% CI for this mortality estimate is 0.71, suggesting a conservative estimate of 71% mortality for patients with SAB who did not receive antibacterial treatment. CIs were calculated using the Clopper Pearson formula. SAB: Staphylococcus aureus bacteremia.
Figure 3.. Random effects meta-analysis of clinical…
Figure 3.. Random effects meta-analysis of clinical outcome (all-cause mortality) in patients with Staphylococcus aureus bacteremia who receive antibacterial treatment.
Three original articles [8,43,44] were identified that described outcomes of SAB patients treated with antibacterial treatment in randomized controlled trials published since 1985. These studies were included in a random effects meta-analysis, which suggests an all-cause mortality of 16%. The upper bound of the 95% CI for this mortality estimate is 0.20, suggesting a conservative estimate of 20% mortality for patients with SAB who receive antibacterial treatment. CIs were calculated using the Clopper Pearson formula. SAB: S. aureus bacteremia.

References

    1. Tong SY, Davis JS, Eichenberger E, Holland TL, Fowler VG., Jr Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management . Clin. Microbiol. Rev. 28(3), 603–661 (2015).
    1. Asgeirsson H, Thalme A, Weiland O. Staphylococcus aureus bacteraemia and endocarditis – epidemiology and outcome: a review . Infect. Dis. (Lond.) 50(3), 175–192 (2018).
    2. • A recent review of the epidemiology and burden of disease assocated with Staphylococcus aureus bacteremia (SAB).

    1. Bassetti M, Peghin M, Trecarichi EM. et al. Characteristics of Staphylococcus aureus bacteraemia and predictors of early and late mortality. PLoS ONE 12(2), e0170236 (2017).
    1. Austin ED, Sullivan SS, Nacesic N. et al. Reduced mortality of Staphylococcus aureus bacteremia in a retrospective cohort study of 2139 patients: 2007–2015. Clin. Infect. Dis. (2019) (Epub ahead of print).
    1. Van Hal SJ, Jensen SO, Vaska VL, Espedido BA, Paterson DL, Gosbell IB. Predictors of mortality in Staphylococcus aureus bacteremia. Clin. Microbiol. Rev. 25(2), 362–386 (2012).
    1. Thwaites GE, Edgeworth JD, Gkrania-Klotsas E. et al. Clinical management of Staphylococcus aureus bacteraemia. Lancet Infect. Dis. 11(3), 208–222 (2011).
    2. • Discusses the evidence behind the key clinical decisions in the management of SAB, the priorities for immediate and long-term treatment and the direction of future research.

    1. Holland TL, Chambers HF, Boucher HW. et al. Considerations for clinical trials of Staphylococcus aureus bloodstream infection in adults. Clin. Infect. Dis. 68(5), 865–872 (2019).
    2. •• Reviews the challenges in designing and executing interventional clinical trials for SAB, and provides recommendations for improvements in trial design.

    1. Fowler VG, Jr, Boucher HW, Corey GR. et al. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus . N. Engl. J. Med. 355(7), 653–665 (2006).
    2. •• Documents the design and execution of the open-label trial establishing daptomycin as a treatment option for SAB.

    1. Sharma M, Riederer K, Chase P, Khatib R. High rate of decreasing daptomycin susceptibility during the treatment of persistent Staphylococcus aureus bacteremia. Eur. J. Clin. Microbiol. Infect. Dis. 27(6), 433–437 (2008).
    1. Gasch O, Camoez M, Dominguez MA. et al. Emergence of resistance to daptomycin in a cohort of patients with methicillin-resistant Staphylococcus aureus persistent bacteraemia treated with daptomycin. J. Antimicrob. Chemother. 69(2), 568–571 (2014).
    1. Zhang S, Sun X, Chang W, Dai Y, Ma X. Systematic review and meta-analysis of the epidemiology of vancomycin-intermediate and heterogeneous vancomycin-intermediate Staphylococcus aureus isolates. PLoS ONE 10(8), e0136082 (2015).
    1. Liapikou A, Cilloniz C, Torres A. Ceftobiprole for the treatment of pneumonia: a European perspective. Drug Des. Devel. Ther. 9, 4565–4572 (2015).
    1. Morosini MI, Diez-Aguilar M, Canton R. Mechanisms of action and antimicrobial activity of ceftobiprole. Rev. Esp. Quimioter. 32(Suppl. 3), 3–10 (2019).
    1. Giacobbe DR, De Rosa FG, Del Bono V. et al. Ceftobiprole: drug evaluation and place in therapy. Expert Rev. Anti. Infect. Ther. 17(9), 689–698 (2019).
    1. Pfaller MA, Flamm RK, Mendes RE. et al. Ceftobiprole activity against Gram-positive and -negative pathogens collected from the United States in 2006 and 2016. Antimicrob. Agents Chemother. 63(1), pii:e01566–e01518 (2019).
    2. •• Demonstrates that cetfobiprole remains highly active in vitro against a large number of pathogens associated with serious infections compared with results from a decade ago.

    1. Zevtera 500 mg powder for concentrate for solution for infusion (2019).
    1. Ceftobiprole in the treatment of patients with Staphylococcus aureus bacteremia – NCT03138733 (2019).
    1. Hassoun A, Linden PK, Friedman B. Incidence, prevalence, and management of MRSA bacteremia across patient populations-a review of recent developments in MRSA management and treatment. Crit. Care 21(1), 211 (2017).
    1. Habib G, Lancellotti P, Antunes MJ. et al. 2015 ESC Guidelines for the management of infective endocarditis: the Task Force for the Management of Infective Endocarditis of the European Society of Cardiology (ESC). Endorsed by: European Association for Cardio-Thoracic Surgery (EACTS), the European Association of Nuclear Medicine (EANM). Eur. Heart J. 36(44), 3075–3128 (2015).
    1. Holland TL, Arnold C, Fowler VG., Jr Clinical management of Staphylococcus aureus bacteremia: a review. JAMA 312(13), 1330–1341 (2014).
    2. • Summarizes the management strategies available for SAB, offers recommendations on the use of echocardiography and identifies the need for high-quality trials in SAB.

    1. Casapao AM, Davis SL, Barr VO. et al. Large retrospective evaluation of the effectiveness and safety of ceftaroline fosamil therapy. Antimicrob. Agents Chemother. 58(5), 2541–2546 (2014).
    1. Pani A, Colombo F, Agnelli F. et al. Off-label use of ceftaroline fosamil: a systematic review. Int. J. Antimicrob. Agents 54(5), 562–571 (2019).
    1. Chambers HF. Evaluation of ceftobiprole in a rabbit model of aortic valve endocarditis due to methicillin-resistant and vancomycin-intermediate Staphylococcus aureus . Antimicrob. Agents Chemother. 49(3), 884–888 (2005).
    1. Entenza JM, Veloso TR, Vouillamoz J, Giddey M, Majcherczyk P, Moreillon P. In vivo synergism of ceftobiprole and vancomycin against experimental endocarditis due to vancomycin-intermediate Staphylococcus aureus . Antimicrob. Agents Chemother. 55(9), 3977–3984 (2011).
    1. Fernandez J, Abbanat D, Shang W. et al. Synergistic activity of ceftobiprole and vancomycin in a rat model of infective endocarditis caused by methicillin-resistant and glycopeptide-intermediate Staphylococcus aureus . Antimicrob. Agents Chemother. 56(3), 1476–1484 (2012).
    1. Tattevin P, Basuino L, Bauer D, Diep BA, Chambers HF. Ceftobiprole is superior to vancomycin, daptomycin, and linezolid for treatment of experimental endocarditis in rabbits caused by methicillin-resistant Staphylococcus aureus . Antimicrob. Agents Chemother. 54(2), 610–613 (2010).
    2. • Documents the superiority of ceftobiprole versus standard therapies in treating experimental endocarditis caused by methicillin-resistant Staphylococcus aureus in rabbits.

    1. Canepari P, Boaretti M, Lleo MM, Satta G. Lipoteichoic acid as a new target for activity of antibiotics: mode of action of daptomycin (LY146032). Antimicrob. Agents Chemother. 34(6), 1220–1226 (1990).
    1. CUBICIN Prescribing Information (2018).
    1. Cubicin Summary of Product Characteristics (2019).
    1. Steenbergen JN, Alder J, Thorne GM, Tally FP. Daptomycin: a lipopeptide antibiotic for the treatment of serious Gram-positive infections. J. Antimicrob. Chemother. 55(3), 283–288 (2005).
    1. Li JS, Sexton DJ, Mick N. et al. Proposed modifications to the Duke criteria for the diagnosis of infective endocarditis. Clin. Infect. Dis. 30(4), 633–638 (2000).
    1. Liu C, Bayer A, Cosgrove SE. et al. Clinical practice guidelines by the infectious diseases society of america for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: executive summary. Clin. Infect. Dis. 52(3), 285–292 (2011).
    1. Timbrook TT, Caffrey AR, Luther MK, Lopes V, Laplante KL. Association of higher daptomycin dose (7 mg/kg or greater) with improved survival in patients with methicillin-resistant Staphylococcus aureus bacteremia. Pharmacotherapy 38(2), 189–196 (2018).
    1. Angeletti S. Matrix assisted laser desorption time of flight mass spectrometry (MALDI-TOF MS) in clinical microbiology. J. Microbiol. Methods 138, 20–29 (2017).
    1. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing; 23rd Informational supplement M100-S23. Clinical and Laboratory Standards Institute, PA, USA: (2013).
    1. Macneal W, Frisbee F. One hundred patients with Staphylococcus septicemia receiving bacteriophage service. Am. J. Med. Sci. 191, 179–195 (1936).
    1. Lowenstein P. Staphylococcus septicemia . Am. J. M. Sc. 181, 196–203 (1936).
    1. Mendell TH. Staphylococcic septicemia: a review of thirty-five cases, with six recoveries, twenty-nine deaths and sixteen autopsies . AMA. Arch. Intern. Med. (Chic.) 63(6), 1068–1083 (1939).
    1. Neuhof H, Aufses A, Hirshfeld S. Pyogenic sepsis. Surg. Gynecol. Obstet. LVIII, 886–896 (1934).
    1. Rosenow ECJ, Brown AE. Septicemia: a review of cases, 1934–1936 inclusive. Proc. Staff Meet. Mayo Clin. 13, 89–93 (1938).
    1. Scott WJM. The principles of the treatment of septicemia. JAMA 105, 1246–1249 (1935).
    1. Skinner D, Keefer C. Significance of bacteremia caused by Staphylococcus aureus . Arch. Intern. Med. (Chic.) 68, 851–875 (1941).
    1. Davis JS, Sud A, O'sullivan MVN. et al. Combination of vancomycin and beta-lactam therapy for methicillin-resistant Staphylococcus aureus bacteremia: a pilot multicenter randomized controlled trial. Clin. Infect. Dis. 62(2), 173–180 (2016).
    1. Van Der Auwera P, Klastersky J, Thys JP, Meunier-Carpentier F, Legrand JC. Double-blind, placebo-controlled study of oxacillin combined with rifampin in the treatment of staphylococcal infections. Antimicrob. Agents Chemother. 28(4), 467–472 (1985).

Source: PubMed

3
구독하다