1,775
Views
3
CrossRef citations to date
0
Altmetric
Editorial

The role of PK/PD–based strategies to preserve new molecules against multi-drug resistant gram-negative strains

, ORCID Icon &
Pages 219-225 | Received 10 Jun 2020, Accepted 15 Jun 2020, Published online: 06 Jul 2020

References

  • Theuretzbacher U. Global antimicrobial resistance in Gram-negative pathogens and clinical need. Curr Opin Microbiol. 2017;39:106–12.
  • Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302(21):2323–9.
  • Shorr AF. Review of studies of the impact on Gram-negative bacterial resistance on outcomes in the intensive care unit. Crit Care Med. 2009;37(4):1463–9.
  • Jernigan JA, Hatfield KM, Wolford H, Nelson RE, Olubajo B, Reddy SC, et al. Multidrug-Resistant Bacterial Infections in US Hospitalized Patients, 2012-2017. N Engl J Med. 2020;382(14):1309–19.
  • Karaiskos I, Lagou S, Pontikis K, Rapti V, Poulakou G. The “Old” and the “New” antibiotics for MDR gram-negative pathogens: for whom, when, and how. Front Public Health. 2019;7:151.
  • Molina J, Peñalva G, Gil-Navarro MV, Praena J, Lepe JA, Pérez-Moreno MA, et al. Long-term impact of an educational antimicrobial stewardship program on hospital-acquired candidemia and multidrug-resistant bloodstream infections: a quasi-experimental study of interrupted time-series analysis. Clin Infect Dis. 2017;65(12):1992–9.
  • Moyá B, Zamorano L, Juan C, Ge Y, Oliver A. Affinity of the new cephalosporin CXA-101 to penicillin-binding proteins of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2010;54(9):3933–7.
  • Periti P, Nicoletti P. Classification of betalactam antibiotics according to their pharmacodynamics. J Chemother. 1999;11(5):323–30.
  • Caballero VR, Abuhussain SA, Kuti JL, Nicolau DP. Efficacy of human-simulated exposures of ceftolozane-tazobactam alone and in combination with amikacin or colistin against multidrug-resistant pseudomonas aeruginosa in an in vitro pharmacodynamic model. Antimicrob Agents Chemother. 2018;62(5):e02384–17.
  • Gómez-Junyent J, Benavent E, Sierra Y, El Haj C, Soldevila L, Torrejón B, et al. Efficacy of ceftolozane/tazobactam, alone and in combination with colistin, against multidrug-resistant Pseudomonas aeruginosa in an in vitro biofilm pharmacodynamic model. Int J Antimicrob Agents. 2019;53(5):612–9.
  • Mojica MF, Ouellette CP, Leber A, Becknell MB, Ardura MI, Perez F, et al. Successful treatment of bloodstream infection due to Metallo-β-Lactamase-producing stenotrophomonas maltophilia in a renal transplant patient. Antimicrob Agents Chemother. 2016;60(9):5130–4.
  • Shaw E, Rombauts A, Tubau F, Padullés A, Càmara J, Lozano T, et al. Clinical outcomes after combination treatment with ceftazidime/avibactam and aztreonam for NDM-1/OXA-48/CTX-M-15-producing Klebsiella pneumoniae infection. J Antimicrob Chemother. 2018;73(4):1104–6.
  • Kollef MH, Nováček M, Kivistik Ü, Réa-Neto Á, Shime N, Martin-Loeches I, et al. Ceftolozane-tazobactam versus meropenem for treatment of nosocomial pneumonia (ASPECT-NP): a randomised, controlled, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis. 2019;19(12):1299–311.
  • Lewis SJ, Mueller BA. Antibiotic dosing in patients with acute kidney injury: “Enough but not too much”. J Intensive Care Med. 2016;31(3):164–76.
  • Bidell MR, Lodise TP. Suboptimal clinical response rates with newer antibiotics among patients with moderate renal impairment: review of the literature and potential pharmacokinetic and pharmacodynamic considerations for observed findings. Pharmacotherapy. 2018;38(12):1205–15.
  • Natesan S, Pai MP, Lodise TP. Determination of alternative ceftolozane/tazobactam dosing regimens for patients with infections due to Pseudomonas aeruginosa with MIC values between 4 and 32 mg/L. J Antimicrob Chemother. 2017;72(10):2813–6.
  • Keepers TR, Gomez M, Celeri C, Nichols WW, Krause KM. Bactericidal activity, absence of serum effect, and time-kill kinetics of ceftazidime-avibactam against β-lactamase-producing enterobacteriaceae and pseudomonas aeruginosa. Antimicrob Agents Chemother. 2014;58(9):5297–305.
  • Karaiskos I, Galani I, Souli M, Giamarellou H. Novel β-lactam-β-lactamase inhibitor combinations: expectations for the treatment of carbapenem-resistant Gram-negative pathogens. Expert Opin Drug Metab Toxicol. 2019;15(2):133–49.
  • Nichols WW, Stone GG, Newell P, Broadhurst H, Wardman A, MacPherson M, et al. Ceftazidime-avibactam susceptibility breakpoints against enterobacteriaceae and pseudomonas aeruginosa. Antimicrob Agents Chemother. 2018;62(11):e02590-17. doi:10.1128/AAC.02590-17.
  • MacGowan A, Tomaselli S, Noel A, Bowker K. The pharmacodynamics of avibactam in combination with ceftaroline or ceftazidime against β-lactamase-producing Enterobacteriaceae studied in an in vitro model of infection. J Antimicrob Chemother. 2017;72(3):762–9.
  • Nichols WW, Newell P, Critchley IA, Riccobene T, Das S. Avibactam pharmacokinetic/pharmacodynamic targets. Antimicrob Agents Chemother. 2018;62(6):e02446-17. doi:10.1128/AAC.02446-17.
  • Nicolau DP, Siew L, Armstrong J, Li J, Edeki T, Learoyd M, et al. Phase 1 study assessing the steady-state concentration of ceftazidime and avibactam in plasma and epithelial lining fluid following two dosing regimens. J Antimicrob Chemother. 2015;70(10):2862–9.
  • Lasko MJ, Nicolau DP. Carbapenem-Resistant Enterobacterales: Considerations for Treatment in the Era of New Antimicrobials and Evolving Enzymology. Curr Infect Dis Rep. 2020;22(3):6.
  • Shields RK, Nguyen MH, Chen L, Press EG, Kreiswirth BN, Clancy CJ. Pneumonia and renal replacement therapy are risk factors for ceftazidime-avibactam treatment failures and resistance among patients with carbapenem-resistant enterobacteriaceae infections. Antimicrob Agents Chemother. 2018;62(5):e02497–17.
  • Novelli A, Del Giacomo P, Rossolini GM, Tumbarello M. Meropenem/vaborbactam: a next generation β-lactam β-lactamase inhibitor combination. Expert Rev anti Infect Ther. 2020;:1–13.
  • Castanheira M, Huband MD, Mendes RE, Flamm RK. Meropenem-vaborbactam tested against contemporary gram-negative isolates collected worldwide during 2014, including carbapenem-resistant, KPC-producing, multidrug-resistant, and extensively drug-resistant enterobacteriaceae. Antimicrob Agents Chemother. 2017;61(9):e00567-17. doi:10.1128/AAC.00567-17.
  • Ho S, Nguyen L, Trinh T, MacDougall C. Recognizing and overcoming resistance to new beta-lactam/beta-lactamase inhibitor combinations. Curr Infect Dis Rep. 2019;21(10).
  • Grupper M, Kuti JL, Nicolau DP. Continuous and prolonged intravenous β-lactam dosing: implications for the clinical laboratory. Clin Microbiol Rev. 2016;29(4):759–72.
  • Adembri C, Novelli A. Pharmacokinetic and pharmacodynamic parameters of antimicrobials: potential for providing dosing regimens that are less vulnerable to resistance. Clin Pharmacokinet. 2009;48(8):517–28.
  • Ambrose PG, Lomovskaya O, Griffith DC, Dudley MN, VanScoy B. β-Lactamase inhibitors: what you really need to know. Curr Opin Pharmacol. 2017;36:86–93.
  • Zhanel GG, Lawrence CK, Adam H, Schweizer F, Zelenitsky S, Zhanel M, et al. Imipenem-relebactam and meropenem-vaborbactam: two novel carbapenem-β-lactamase inhibitor combinations. Drugs. 2018;78(1):65–98.
  • Bhagunde P, Zhang Z, Racine F, Carr D, Wu J, Young K, et al. A translational pharmacokinetic/pharmacodynamic model to characterize bacterial kill in the presence of imipenem-relebactam. Int J Infect Dis. 2019;89:55–61.
  • Tooke CL, Hinchliffe P, Lang PA, Mulholland AJ, Brem J, Schofield CJ, et al. Molecular basis of class a beta-lactamase inhibition by relebactam. Antimicrob Agents Chemother. 2019;63(10):e00564-19. doi:10.1128/AAC.00564-19.
  • Tillotson GS. Trojan horse antibiotics-a novel way to circumvent gram-negative bacterial resistance? Infect Dis (Auckl). 2016;9:45–52.
  • Ito A, Kohira N, Bouchillon SK, West J, Rittenhouse S, Sader HS, et al. In vitro antimicrobial activity of S-649266, a catechol-substituted siderophore cephalosporin, when tested against non-fermenting Gram-negative bacteria. J Antimicrob Chemother. 2016;71(3):670–7.
  • Kohira N, West J, Ito A, Ito-Horiyama T, Nakamura R, Sato T, et al. In vitro antimicrobial activity of a siderophore cephalosporin, S-649266, against enterobacteriaceae clinical isolates, including carbapenem-resistant strains. Antimicrob Agents Chemother. 2016;60(2):729–34.
  • Ito A, Sato T, Ota M, Takemura M, Nishikawa T, Toba S, et al. In vitro antibacterial properties of cefiderocol, a novel siderophore cephalosporin, against gram-negative bacteria. Antimicrob Agents Chemother. 2018;62(1):e01454-17. doi:10.1128/AAC.01454-17
  • Katsube T, Echols R, Ferreira JCA, Krenz HK, Berg JK, Galloway C. Cefiderocol, a siderophore cephalosporin for gram-negative bacterial infections: pharmacokinetics and safety in subjects with renal impairment. J Clin Pharmacol. 2017;57(5):584–91.
  • Saisho Y, Katsube T, White S, Fukase H, Shimada J. Pharmacokinetics, safety, and tolerability of cefiderocol, a novel siderophore cephalosporin for gram-negative bacteria, in healthy subjects. Antimicrob Agents Chemother. 2018;62(3):e02163-17. doi:10.1128/AAC.02163-17
  • Katsube T, Wajima T, Ishibashi T, Ferreira JCA, Echols R. Pharmacokinetic/pharmacodynamic modeling and simulation of cefiderocol, a parenteral siderophore cephalosporin, for dose adjustment based on renal function. Antimicrob Agents Chemother. 2017;61(1):e01381-16. doi:10.1128/AAC.01381-16.
  • Katsube T, Saisho Y, Shimada J, Furuie H. Intrapulmonary pharmacokinetics of cefiderocol, a novel siderophore cephalosporin, in healthy adult subjects. J Antimicrob Chemother. 2019;74(7):1971–4.
  • Giamarellou H, Galani L, Baziaka F, Karaiskos I. Effectiveness of a double-carbapenem regimen for infections in humans due to carbapenemase-producing pandrug-resistant klebsiella pneumoniae. Antimicrob Agents Chemother. 2013;57(5):2388–90.
  • Souli M, Karaiskos I, Masgala A, Galani L, Barmpouti E, Giamarellou H. Double-carbapenem combination as salvage therapy for untreatable infections by KPC-2-producing Klebsiella pneumoniae. Eur J Clin Microbiol Infect Dis. 2017;36(7):1305–15.
  • Veiga RP, Paiva JA. Pharmacokinetics-pharmacodynamics issues relevant for the clinical use of beta-lactam antibiotics in critically ill patients. Crit Care. 2018;22(1):1–34.
  • Pai MP, Neely M, Rodvold KA, Lodise TP. Innovative approaches to optimizing the delivery of vancomycin in individual patients. Adv Drug Deliv Rev. 2014;77:50–7.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.