Evaluation of Plazomicin, Tigecycline, and Meropenem Pharmacodynamic Exposure against Carbapenem-Resistant Enterobacteriaceae in Patients with Bloodstream Infection or Hospital-Acquired/Ventilator-Associated Pneumonia from the CARE Study (ACHN-490-007)

Joseph L Kuti, Aryun Kim, Daniel J Cloutier, David P Nicolau, Joseph L Kuti, Aryun Kim, Daniel J Cloutier, David P Nicolau

Abstract

Introduction: CARE was a Phase 3, randomized study evaluating the efficacy and safety of plazomicin-based combination therapy compared with colistin-based combination therapy for the treatment of patients with bloodstream infections or hospital-acquired/ventilator-associated pneumonia due to carbapenem-resistant Enterobacteriaceae (CRE). Adjunctive therapies included either tigecycline or meropenem. We sought to understand the contribution of tigecycline and meropenem to plazomicin-treated-patient outcomes by determining their observed pharmacodynamic exposures against baseline pathogens.

Methods: Blood samples collected for plazomicin therapeutic monitoring were assayed for tigecycline and meropenem concentrations. Population pharmacokinetic models were constructed for each antibiotic. Using the individual Bayesian posterior or a covariate-based model, concentration time profiles were simulated to estimate the pharmacodynamic exposures for each patient. Pharmacodynamic thresholds for plazomicin, tigecycline, and meropenem were a total area under the curve to minimum inhibitory concentration ratio (AUC/MIC) ≥ 85, free (f) AUC/MIC ≥ 0.9, and free time above the MIC (fT > MIC) of ≥ 40%, respectively.

Results: Fifteen plazomicin-treated patients were included (12 received tigecycline, 4 received meropenem, 1 received both). Microbiological response was observed in 13 (86.7%) and clinical efficacy was achieved in 11 (73.3%). Plazomicin achieved its pharmacodynamic target in all 15 patients. Meropenem fT > MIC was 0% in all 4 patients, and tigecycline fAUC/MIC was ≥ 0.9 in 9 (75%) patients. Overall, 6 (40%) of 15 patients had a tigecycline or meropenem exposure below the requisite thresholds. Microbiological response and clinical efficacy were observed in 100% (6/6) and 83.3% (5/6) of patients with low threshold attainment by tigecycline and meropenem dosing regimens, respectively.

Conclusions: Plazomicin successfully achieved its requisite pharmacodynamic exposure, and these data suggest that optimization of tigecycline and meropenem therapy was not required for the combination to achieve microbiological response and clinical efficacy against serious CRE infections.

Trial registration: ClinicalTrials.gov number, NCT01970371.

Funding: Achaogen, Inc.

Keywords: Aminoglycoside; Carbapenem; Glycylcycline; Pharmacodynamics; Pharmacokinetics.

Figures

Fig. 1
Fig. 1
Observed versus (left) population predicted and (right) individual predicted tigecycline concentrations from the final model
Fig. 2
Fig. 2
Observed versus Bayesian posterior individual predicted concentrations for a patient 4 and b patient 15, who received meropenem in the CARE Study. While precision was reasonable for patient 4, individual Bayesian parameter estimates for patient 15 resulted in predicted concentrations significantly different from observed

References

    1. Gupta N, Limbago BM, Patel JB, et al. Carbapenem-resistant Enterobacteriaceae: epidemiology and prevention. Clin Infect Dis. 2011;53:60–67. doi: 10.1093/cid/cir202.
    1. Guh AY, Bulens SN, Mu Y, et al. Epidemiology of carbapenem-resistant Enterobacteriaceae in 7 US communities, 2012–2013. JAMA. 2015;314:1479–1487. doi: 10.1001/jama.2015.12480.
    1. Rice LB, Carias LL, Hutton RA, et al. The KQ element, a complex genetic region conferring transferable resistance to carbapenems, aminoglycosides, and fluoroquinolones in Klebsiella pneumoniae. Antimicrob Agents Chemother. 2008;52:3427–3429. doi: 10.1128/AAC.00493-08.
    1. Almaghrabi R, Clancy CJ, Doi Y, et al. Carbapenem-resistant Klebsiella pneumoniae strains exhibit diversity in aminoglycoside-modifying enzymes, which exert differing effects on plazomicin and other agents. Antimicrob Agents Chemother. 2014;58:4443–4451. doi: 10.1128/AAC.00099-14.
    1. Cox G, Ejim L, Stogios PJ, et al. Plazomicin retains antibiotic activity against most aminoglycoside modifying enzymes. ACS Infect Dis. 2018;4:980–987. doi: 10.1021/acsinfecdis.8b00001.
    1. Armstrong ES, Miller GH. Combating evolution with intelligent design: the neoglycoside ACHN-490. Curr Opin Microbiol. 2010;13:565–573. doi: 10.1016/j.mib.2010.09.004.
    1. Livermore DM, Mushtaq S, Warner M, et al. Activity of aminoglycosides, including ACHN-490, against carbapenem-resistant Enterobacteriaceae isolates. J Antimicrob Chemother. 2011;66:48–53. doi: 10.1093/jac/dkq408.
    1. McKinnell JA, Dwyer JP, Talbot GH, et al. Plazomicin for infections caused by carbapenem-resistant Enterbacteriaceae. N Engl J Med. 2019;380:8. doi: 10.1056/NEJMc1807634.
    1. Ambrose PG, Bhavnani SM, Rubino CM, et al. Pharmacokinetics-pharmacodynamics of antimicrobial therapy: it’s not just for mice anymore. Clin Infect Dis. 2007;44:79–86. doi: 10.1086/510079.
    1. Trang M, Seroogy JD, Van Wart SA, et al. Population pharmacokinetic analyses for plazomicin using pooled data from phase 1, 2, and 3 clinical studies. Antimicrob Agents Chemother. 2019;63:e02329-18. doi: 10.1128/AAC.02329-18.
    1. Neely MN, van Guilder MG, Yamada WM, et al. Accurate detection of outliers and subpopulations with Pmetrics, a nonparametric and parametric pharmacometric modeling and simulation package for R. Ther Drug Monit. 2012;34:467–476. doi: 10.1097/FTD.0b013e31825c4ba6.
    1. Bhavnani SM, Rubino CM, Hammel JP, et al. Pharmacological and patient-specific response determinants in patients with hospital-acquired pneumonia treated with tigecycline. Antimicrob Agents Chemother. 2012;56:1065–1072. doi: 10.1128/AAC.01615-10.
    1. Crandon JL, Ariano RE, Zelenitsky SA, et al. Optimization of meropenem dosage in the critically ill population based on renal function. Intensive Care Med. 2011;37:632–638. doi: 10.1007/s00134-010-2105-0.
    1. Grupper M, Kuti JL, Nicolau DP. Continuous and prolonged intravenous beta-lactam dosing: implications for the clinical laboratory. Clin Microbiol Rev. 2016;29:759–772. doi: 10.1128/CMR.00022-16.
    1. Rubino CM, Forrest A, Bhavnani SM, et al. Tigecycline population pharmacokinetics in patients with community- or hospital-acquired pneumonia. Antimicrob Agents Chemother. 2010;54:5180–5186. doi: 10.1128/AAC.01414-09.
    1. Bhavnani SM, Hammel JP, Trang M, et al. Pharmacokinetic–pharmacodynamic target attainment analyses to support plazomicin dose selection and recommendations for interpretive criteria for in vitro susceptibility testing for Enterobacteriaceae (poster no. 518). In: American Society for Microbiology Microbe 2018, Atlanta, GA; 2018.
    1. Daikos GL, Jackson GG, Lolans VT, et al. Adaptive resistance to aminoglycoside antibiotics from first-exposure down-regulation. J Infect Dis. 1990;162:414–420. doi: 10.1093/infdis/162.2.414.
    1. Daikos GL, Lolans VT, Jackson GG. First-exposure adaptive resistance to aminoglycoside antibiotics in vivo with meaning for optimal clinical use. Antimicrob Agents Chemother. 1991;35:117–123. doi: 10.1128/AAC.35.1.117.
    1. Karlowsky JA, Zelenitsky SA, Zhanel GG. Aminoglycoside adaptive resistance. Pharmacotherapy. 1997;17:549–555.
    1. Abdelraouf K, Kim A, Krause KM, et al. In vivo efficacy of plazomicin alone or in combination with meropenem or tigecycline against Enterobacteriaceae isolates exhibiting various resistance mechanisms in an immunocompetent murine septicemia model. Antimicrob Agents Chemother. 2018;62:e01074-18. doi: 10.1128/AAC.01074-18.
    1. Xie J, Roberts JA, Alobaid AS, et al. Population pharmacokinetics of tigecycline in critically ill patients with severe infections. Antimicrob Agents Chemother. 2017;61:e00345-17. doi: 10.1128/AAC.00345-17.
    1. Borsuk-De Moor A, Rypulak E, Potrec B, et al. Population pharmacokinetics of high-dose tigecycline in patients with sepsis or septic shock. Antimicrob Agents Chemother. 2018;62:e02273-17. doi: 10.1128/AAC.02273-17.
    1. Korth-Bradley JM, Troy SM, Matschke K, et al. Tigecycline pharmacokinetics in subjects with various degrees of renal function. J Clin Pharmacol. 2012;52:1379–1387. doi: 10.1177/0091270011416938.
    1. Passarell JA, Meagher AK, Liolios K, et al. Exposure-response analyses of tigecycline efficacy in patients with complicated intra-abdominal infections. Antimicrob Agents Chemother. 2008;52:204–210. doi: 10.1128/AAC.00813-07.
    1. Koomanachai P, Kim A, Nicolau DP. Pharmacodynamic evaluation of tigecycline against Acinetobacter baumannii in a murine pneumonia model. J Antimicrob Chemother. 2009;63:982–987. doi: 10.1093/jac/dkp056.
    1. Li C, Du X, Kuti JL, et al. Clinical pharmacodynamics of meropenem in patients with lower respiratory tract infections. Antimicrob Agents Chemother. 2007;51:1725–1730. doi: 10.1128/AAC.00294-06.
    1. Crandon JL, Luyt CE, Aubry A, et al. Pharmacodynamics of carbapenems for the treatment of Pseudomonas aeruginosa ventilator-associated pneumonia: associations with clinical outcome and recurrence. J Antimicrob Chemother. 2016;71:2534–2537. doi: 10.1093/jac/dkw200.
    1. Rodriguez-Avial I, Pena I, Picazo JJ, et al. In vitro activity of the next-generation aminoglycoside plazomicin alone and in combination with colistin, meropenem, fosfomycin or tigecycline against carbapenemase-producing Enterobacteriaceae strains. Int J Antimicrob Agents. 2015;46:616–621. doi: 10.1016/j.ijantimicag.2015.07.021.
    1. Thwaites M, Hall D, Stoneburner A, et al. Activity of plazomicin in combination with other antibiotics against multidrug-resistant Enterobactericeae. Diagn Microbiol Infect Dis. 2018;92:338–345. doi: 10.1016/j.diagmicrobio.2018.07.006.

Source: PubMed

3
Subskrybuj