Meropenem Versus Piperacillin-Tazobactam for Definitive Treatment of Bloodstream Infections Caused by AmpC β-Lactamase-Producing Enterobacter spp, Citrobacter freundii, Morganella morganii, Providencia spp, or Serratia marcescens: A Pilot Multicenter Randomized Controlled Trial (MERINO-2)

Adam G Stewart, David L Paterson, Barnaby Young, David C Lye, Joshua S Davis, Kellie Schneider, Mesut Yilmaz, Rumeysa Dinleyici, Naomi Runnegar, Andrew Henderson, Sophia Archuleta, Shirin Kalimuddin, Brian M Forde, Mark D Chatfield, Michelle J Bauer, Jeffrey Lipman, Tiffany Harris-Brown, Patrick N A Harris, MERINO Trial Investigators and the Australasian Society for Infectious Disease Clinical Research Network (ASID-CRN), Po Ying Chia, Gail Cross, Jyoti Somani, Gabriel Yan, Adam G Stewart, David L Paterson, Barnaby Young, David C Lye, Joshua S Davis, Kellie Schneider, Mesut Yilmaz, Rumeysa Dinleyici, Naomi Runnegar, Andrew Henderson, Sophia Archuleta, Shirin Kalimuddin, Brian M Forde, Mark D Chatfield, Michelle J Bauer, Jeffrey Lipman, Tiffany Harris-Brown, Patrick N A Harris, MERINO Trial Investigators and the Australasian Society for Infectious Disease Clinical Research Network (ASID-CRN), Po Ying Chia, Gail Cross, Jyoti Somani, Gabriel Yan

Abstract

Background: Carbapenems are recommended treatment for serious infections caused by AmpC-producing gram-negative bacteria but can select for carbapenem resistance. Piperacillin-tazobactam may be a suitable alternative.

Methods: We enrolled adult patients with bloodstream infection due to chromosomal AmpC producers in a multicenter randomized controlled trial. Patients were assigned 1:1 to receive piperacillin-tazobactam 4.5 g every 6 hours or meropenem 1 g every 8 hours. The primary efficacy outcome was a composite of death, clinical failure, microbiological failure, and microbiological relapse at 30 days.

Results: Seventy-two patients underwent randomization and were included in the primary analysis population. Eleven of 38 patients (29%) randomized to piperacillin-tazobactam met the primary outcome compared with 7 of 34 patients (21%) in the meropenem group (risk difference, 8% [95% confidence interval {CI}, -12% to 28%]). Effects were consistent in an analysis of the per-protocol population. Within the subcomponents of the primary outcome, 5 of 38 (13%) experienced microbiological failure in the piperacillin-tazobactam group compared to 0 of 34 patients (0%) in the meropenem group (risk difference, 13% [95% CI, 2% to 24%]). In contrast, 0% vs 9% of microbiological relapses were seen in the piperacillin-tazobactam and meropenem arms, respectively. Susceptibility to piperacillin-tazobactam and meropenem using broth microdilution was found in 96.5% and 100% of isolates, respectively. The most common AmpC β-lactamase genes identified were bla CMY-2, bla DHA-17, bla CMH-3, and bla ACT-17. No ESBL, OXA, or other carbapenemase genes were identified.

Conclusions: Among patients with bloodstream infection due to AmpC producers, piperacillin-tazobactam may lead to more microbiological failures, although fewer microbiological relapses were seen.

Clinical trials registration: NCT02437045.

Keywords: Enterobacterales; ampC β-lactamase; carbapenem; clinical trial; piperacillin-tazobactam.

© The Author(s) 2021. Published by Oxford University Press on behalf of Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
MERINO-2 patient recruitment, randomization, and flow through study. aPatients could meet >1 exclusion criteria. A total of 235 were excluded because >72 hours had elapsed since initial blood culture; 337, based on microbiology criteria; 48, allergy to trial drug; 205, polymicrobial infection; 46, not expected to survive >96 hours; 3, pregnant or breastfeeding; 12, no intent to cure; 16, <18 years old (<21 years in Singapore); and 4, previously enrolled. For 337 patients, microbiological exclusions based on susceptibility testing were as follows: 105 were nonsusceptible to third-generation cephalosporins, 120 were nonsusceptible to either meropenem or piperacillin-tazobactam and 205 were polymicrobial infections. Other exclusions included patient requiring ongoing antibiotic therapy (other than study drug) with activity against gram-negative bacilli (n = 21), Pitt bacteremia score >4 (n = 67), central nervous system source of infection (n = 25).
Figure 2.
Figure 2.
Piperacillin-tazobactam and meropenem minimum inhibitory concentration by intervention arm. Abbreviations: EUCAST, European Committee on Antimicrobial Susceptibility Testing; MIC, minimum inhibitory concentration.
Figure 3.
Figure 3.
Scatterplot comparison of piperacillin-tazobactam and meropenem susceptibility testing by disk diffusion testing (x-axis; zone diameter) and broth microdilution (y-axis; minimum inhibitory concentration). The red dashed line represents the European Committee on Antimicrobial Susceptibility Testing susceptible breakpoint and the blue dashed line represents the Clinical and Laboratory Standards Institute susceptible breakpoint. Abbreviation: BMD, broth microdilution.

References

    1. Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev 2009; 22:161–82, table of contents.
    1. Meini S, Tascini C, Cei M, et al. . AmpC β-lactamase-producing Enterobacterales: what a clinician should know. Infection 2019; 47:363–75.
    1. Lindberg F, Westman L, Normark S. Regulatory components in Citrobacter freundii ampC beta-lactamase induction. Proc Natl Acad Sci U S A 1985; 82:4620–4.
    1. Tamma PD, Doi Y, Bonomo RA, et al. ; Antibacterial Resistance Leadership Group. A primer on AmpC β-lactamases: necessary knowledge for an increasingly multidrug-resistant world. Clin Infect Dis 2019; 69:1446–55.
    1. Mizrahi A, Delerue T, Morel H, et al. ; Saint-Joseph/Avicenna Study Group. Infections caused by naturally AmpC-producing Enterobacteriaceae: can we use third-generation cephalosporins? A narrative review. Int J Antimicrob Agents 2020; 55:105834.
    1. Choi SH, Lee JE, Park SJ, et al. . Emergence of antibiotic resistance during therapy for infections caused by Enterobacteriaceae producing AmpC beta-lactamase: implications for antibiotic use. Antimicrob Agents Chemother 2008; 52:995–1000.
    1. Chow JW, Fine MJ, Shlaes DM, et al. . Enterobacter bacteremia: clinical features and emergence of antibiotic resistance during therapy. Ann Intern Med 1991; 115:585–90.
    1. Siebert JD, Thomson RB Jr, Tan JS, Gerson LW. Emergence of antimicrobial resistance in gram-negative bacilli causing bacteremia during therapy. Am J Clin Pathol 1993; 100:47–51.
    1. Vardakas KZ, Tansarli GS, Rafailidis PI, Falagas ME. Carbapenems versus alternative antibiotics for the treatment of bacteraemia due to Enterobacteriaceae producing extended-spectrum β-lactamases: a systematic review and meta-analysis. J Antimicrob Chemother 2012; 67:2793–803.
    1. Elshamy AA, Aboshanab KM. A review on bacterial resistance to carbapenems: epidemiology, detection and treatment options. Future Sci OA 2020; 6:FSO438.
    1. Harris PNA, Tambyah PA, Lye DC, et al. ; MERINO Trial Investigators and the Australasian Society for Infectious Disease Clinical Research Network (ASID-CRN). Effect of piperacillin-tazobactam vs meropenem on 30-day mortality for patients with E coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance: a randomized clinical trial. JAMA 2018; 320:984–94.
    1. Cheng L, Nelson BC, Mehta M, et al. . Piperacillin-tazobactam versus other antibacterial agents for treatment of bloodstream infections due to AmpC beta-lactamase-producing Enterobacteriaceae. Antimicrob Agents Chemother 2017; 61:e00276-17.
    1. Bush K, Macalintal C, Rasmussen BA, et al. . Kinetic interactions of tazobactam with beta-lactamases from all major structural classes. Antimicrob Agents Chemother 1993; 37:851–8.
    1. Tan SH, Ng TM, Chew KL, et al. . Outcomes of treating AmpC-producing Enterobacterales bacteraemia with carbapenems vs. non-carbapenems. Int J Antimicrob Agents 2020; 55:105860.
    1. McKamey L, Venugopalan V, Cherabuddi K, et al. . Assessing antimicrobial stewardship initiatives: clinical evaluation of cefepime or piperacillin/tazobactam in patients with bloodstream infections secondary to AmpC-producing organisms. Int J Antimicrob Agents 2018; 52:719–23.
    1. Holsen MR, Wardlow LC, Bazan JA, et al. . Clinical outcomes following treatment of Enterobacter species pneumonia with piperacillin/tazobactam compared to cefepime or ertapenem. Int J Antimicrob Agents 2019; 54:824–8.
    1. Harris PN, Wei JY, Shen AW, et al. . Carbapenems versus alternative antibiotics for the treatment of bloodstream infections caused by Enterobacter, Citrobacter or Serratia species: a systematic review with meta-analysis. J Antimicrob Chemother 2016; 71:296–306.
    1. Kohlmann R, Bähr T, Gatermann SG. Species-specific mutation rates for ampC derepression in Enterobacterales with chromosomally encoded inducible AmpC β-lactamase. J Antimicrob Chemother 2018; 73:1530–6.
    1. Henderson A, Paterson DL, Chatfield MD, et al. . Association between minimum inhibitory concentration, beta-lactamase genes and mortality for patients treated with piperacillin/tazobactam or meropenem from the MERINO study [manuscript published online ahead of print 27 October 2020]. Clin Infect Dis 2020. doi:10.1093/cid/ciaa1479.
    1. Drozdinsky G, Neuberger A, Rakedzon S, et al. . Treatment of bacteremia caused by Enterobacter spp.: should the potential for ampC induction dictate therapy? A retrospective study. Microb Drug Resist 2021; 27:410–4.
    1. Moraz M, Bertelli C, Prod’hom G, et al. . Piperacillin/tazobactam selects an ampC derepressed E. cloacae complex mutant in a diabetic osteoarticular infection. Clin Microbiol Infect 2021; 27:475–7.
    1. Akata K, Muratani T, Yatera K, et al. . Induction of plasmid-mediated AmpC β-lactamase DHA-1 by piperacillin/tazobactam and other β-lactams in Enterobacteriaceae. PLoS One 2019; 14:e0218589.
    1. Richter DC, Frey O, Röhr A, et al. . Therapeutic drug monitoring-guided continuous infusion of piperacillin/tazobactam significantly improves pharmacokinetic target attainment in critically ill patients: a retrospective analysis of four years of clinical experience. Infection 2019; 47:1001–11.
    1. Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical outcomes with extended or continuous versus short-term intravenous infusion of carbapenems and piperacillin/tazobactam: a systematic review and meta-analysis. Clin Infect Dis 2013; 56:272–82.
    1. Rhodes NJ, Liu J, O’Donnell JN, et al. . Prolonged infusion piperacillin-tazobactam decreases mortality and improves outcomes in severely ill patients: results of a systematic review and meta-analysis. Crit Care Med 2018; 46:236–43.

Source: PubMed

3
Předplatit