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

In the last decade, infections caused by multi-drug resistant (MDR) Gram-negative (GN) strains have been increasing worldwide, especially in intensive care units (ICUs).Citation1 More than one third of hospital-acquired infections in ICUs are caused by carbapenem resistant Enterobacterales (CRE), Pseudomonas aeruginosa (PA) and Acinetobacter baumannii (AB) with a strong impact on hospital/ICU length of stay, mortality rate, and costs.Citation2,Citation3 In the 2012-2017 period, infections caused by Extended spectrum β-lactamase (ESBL) producing strains increased by 53% in US hospitals.Citation4 In the last few years, due to the lack of new/effective molecules, old ones such as colistin and fosfomycin, with a narrow therapeutic window and/or unpredictable PK, have come back into use.Citation5

Recently, the availability of new drugs or new associations based on old or new β-lactam molecules with either a novel or a classic β-lactamase inhibitor (BL/BLI) have enabled clinicians to tackle MDR GN infections again. However, it is of primary importance to use these new therapeutic opportunities according to antimicrobial stewardship programs - which necessarily take into consideration pharmacokinetic (PK) and pharmacodynamic (PD) indexes - to limit the emergence of resistance against them.Citation6

The emergence of resistance during antimicrobial therapy is particularly at risk in the following circumstances:

  • when treating critically ill septic patients (due to the frequency of MDR strains and the changes in sepsis-related pharmacokinetics);

  • when treating pulmonary infections (a difficult-to-reach infection site)

  • when renal function is altered (this commonly occurs for these BL/BLI which are eliminated via the kidneys).

Here we will focus on the PK/PD data available for the latest 5 β-lactam drugs which target MDR GN strains, and we will provide some suggestions for better treatment and to prevent the emergence of resistance.

Ceftolozane/Tazobactam (TOL/TAZ)—the combination of a new fourth-generation cephalosporin with a former β-lactamase sulfone inhibitor has demonstrated strong activity against ESBL+ Enterobacterales and MDR PA.Citation7 Both the molecules have a time-dependent activity, therefore the percentage time above MIC of the unbound drug (%fT > MIC) is the major PK/PD parameter that better describes their activity against MDR GN ( and ).Citation8–12 Among the new molecules/new associations, both ceftolozane and tazobactam are, together with meropenem vaborbactam (MER/VAB), those which best penetrate the epithelial lining fluid (ELF). After doubling the suggested standard dose and with 1 hour infusion, a higher concentration in ELF can be obtained and this can be effective against pneumonia caused by MDR PA. Recently, a non-inferiority phase 3 randomized controlled trial showed that high dosage TOL/TAZ is as efficient as meropenem in treating ventilation-associated pneumonia (VAP) caused by GN strains.Citation13 Dose adjustment of TOL/TAZ is suggested in the presence of renal dysfunction.Citation14 However, whether this reduction is advisable has been questioned after the results obtained in trials where there was a worse outcome in patients with reduced Creatinine Clearance (CrCl), for whom the BL dose had been decreased.Citation15 TOL/TAZ extended infusion regimens have been proposed as a potential administration modality to improve the Probability Target Attainment (PTA) against infections due to MDR-PA with higher MIC values.Citation16

Table 1. Pharmacokinetic (PK), Percentage of binding protein, PK/PD, Dosage, and market approval for Ceftolozane/Tazobactam (TOL/TAZ), Ceftazidime-Avibactam (CAZ/AVI), Meropenem-vaborbactam (MER/VAB) and Cefiderocol.

Table 2. Spectrum of activity and MIC breakpoints of Ceftolozane/Tazobactam (TOL/TAZ), Ceftazidime-Avibactam (CAZ/AVI), Meropenem-vaborbactam (MER/VAB) and Cefiderocol.and Imipenem/Relebactam (IMI/REL).

Ceftazidime-Avibactam (CAZ/AVI) is a combination of a third-generation cephalosporin and avibactam, a novel diazabicyclo-octane β-lactamase inhibitor.Citation17 The presence of avibactam enables ceftazidime to recover its activity versus ESBL and Class A, C and some D (OXA 48), but not Class B (metallo-β-lactamase, MBL) carbapenemasesCitation18 (). For ceftazidime, the more important PK/PD index is represented by fT > MIC, while for avibactam both fT > MIC and AUC/MIC are important.Citation19 Indeed, for Enterobacterales and PA, the optimal PK/PD target is 50% fT > MIC for ceftazidime and a concentration threshold >1 mg/L (with consequent AUC0-24, 48 mg/l·h) for avibactam.Citation18,Citation20 However, avibactam concentrations >4 mg/L are necessary in order to reduce the bacterial load of KPC-producing Klebsiella pneumoniae (KP), due to low efficacy of this BLI on KPC 3 and 4.Citation20,Citation21 Ceftazidime and avibactam enter the ELF to the same degree, with average lung AUC values being 30% of those obtained in plasma.Citation22 While these concentrations have been shown to generally exceed those effective in experimental pneumonia models, to date there is no evidence that similar exposures can be obtained among critically-ill patients, in whom concentration in the ELF can be decreased during pneumonia for several reasons, including changes in pulmonary circulation.Citation23 Consistently, Shields et al., when they analyzed the risk factors for CAZ-AVI failure in patients with CRE infections, found positive associations with healthcare-associated pneumonia (HAP) and renal replacement therapy (RRT) (OR 3.09, 95% CI 1.03-9.34 and 4.78, 95% CI 1.03–22.2, respectively).Citation24 The latter was also a risk factor for the emergence of resistance (OR 26.67, 95% CI 26.67– 317.1) (OR 3.09, 95% CI 1.03–9.34). It cannot be excluded that resistance development and treatment failure in the population analyzed by Shields were due to inappropriate dosing. It has been proposed that patients receiving CAZ/AVI should be ‘aggressively dosed’ in order to obtain optimal exposures at least for the first 48 hours of treatment, irrespective of their renal function.Citation23

Table 3. Pharmacokinetic (PK)/Pharmocodynamic (PD) PK/PD, Dosage adjustment, Infusion needs for Ceftolozane/Tazobactam (TOL/TAZ), Ceftazidime-Avibactam (CAZ/AVI), Meropenem-vaborbactam (MER/VAB) and Cefiderocol.

Meropenem-vaborbactam (MER/VAB) is a combination of a broad spectrum carbapenem and vaborbactam, a boronic acid β-lactamase inhibitor with no intrinsic antibacterial activity. Vaborbactam is unique in that it is the first cyclic boronic acid inhibitor with high affinity for serine residues, allowing it to serve as a competitive inhibitor, by forming a covalent bond with beta-lactamases, without undergoing hydrolysis.Citation25 Vaborbactam enables meropenem to be effective against Enterobacterales which produce KPC and other class A serine carbapenemases, but not against class D or class B carbapenemases.Citation26 MER/VAB potency is highly reduced in KPC-producing KP overexpressing the major multidrug efflux pump AcrAB-TolC and lacking both ompK35 and ompK36 porins, used by MER/VAB to cross the outer membrane of KP.Citation27 Moreover, this combination does not have any further activity than the meropenem alone towards PA, since PA resistance is principally related to outer membrane impermeability, upregulation of efflux systems, and production of class B or class D β-lactamases.Citation5 Meropenem is a time-dependent antimicrobial agent and benefits from prolonged infusions in order to keep a concentration > MIC for 40% of the dosing interval (fT > MIC> 40%), a value which is necessary to kill bacteria.Citation28,Citation29 In order to be effective, vaborbactam needs to maintain a concentration ≥8 mg/L in plasma and its PK/PD efficacy is connected to the ratio of vaborbactam AUC and the MIC, due to its rapid target binding and its slow reversibility.Citation30 In vitro data seem to indicate that selection of resistance is less common with MER/VAB than with other newBL/BLI, but more robust clinical data are needed to confirm this. Moreover, caution should be used in the treatment of susceptible CRE when OXA-based enzymes have not been excluded.Citation23

Imipenem/Relebactam (IMI/REL) is a combination of the carbapenem imipenem and relebactam, a diazabicyclooctane beta-lactamase inhibitor with a similar structure of avibactam but with an additional piperidine ring.Citation31 Relebactam has been shown to be efficient against serine β-lactamase class A and C and some class D. It has no effect on class B and OXA48 class D. Imipenem (always administered with cilastatin) is a time-dependent antibiotic and a prolonged infusion is necessary in order to obtain at least a target of fT > MIC 69% for bacteriostasis.Citation31 In order to be effective, relebactam needs to have a concentration of 4 mg/L and the PK/PD index predicting efficacy is fAUC/MIC, with a target value of 7.5.Citation32 IMI/REL has recently been approved for the treatment of cUTIs and IAIs. While IMI/REL shows potential restoration of activity in isolates resistant to imipenem/cilastatin, much less data are available on its role in clinical practice, including its stability against the development of resistance. In relebactam, desulfation (and consequent inactivation) occurs more slowly than in avibactam, but relebactam shows a reduced inhibitory potency than avibactam (about 1/8).Citation33 This combination, unlike the other BLI/BLI, is administered as a 30 min infusion. The optimal duration of administration has not yet been validated in the clinical setting.

Cefiderocol is a fourth-generation cephalosporin containing a Cl-cathecol side chain with a chemical structure related to ceftazidime and cefepime but with less susceptibility to beta-lactamases such as AMP C, KPC, NDM and OXA carbapenemases and ESBLs. Cefiderocol is therefore active against KPC, and NDM carbapenemases, MDR PA, AB and Stenotrophomonas maltophilia. This molecule creates a complex with ferric iron and promotes a siderophore-like transport across the outer membrane of GN, behaving like a ‘Trojan Horse’.Citation34 Once in the periplasmic space, its separation from ferric iron enables the molecule to impair peptidoglycan synthesis by binding to penicillin-binding proteins (PBPs).Citation35–37 Cefiderocol elimination is directly related to renal function.Citation38,Citation39 A 90% PTA can be obtained with a dose of 2 g x 3 in 3 hours of infusion, with a target of 75% fT > MIC.Citation40 This antimicrobial has been approved for cUTIs, but it could be effective in HAP, where MDR Gram-negative bacilli are responsible, since an adequate concentration in the ELF has been shown in healthy volunteers. Clinical trials testing the possible use of the molecule for pneumonia are ongoing.Citation23,Citation41

Reduced in vitro activity of cefiderocol has been seen towards PA strains that show a decrease in the production of iron transport system components or mutations on the outer membrane for the iron transport system.Citation42,Citation43 At present it is not known whether in patients with low free iron concentrations the development of resistance to this molecule is likely. A 90% PTA for bloodstream and urinary GN bacteria with cefiderocol MICs ≤ 4 mg/L is obtainable for the dose of 2 g IV every 8 hours as a 3-hr infusion in patients with normal renal function.Citation40 Suggestions for a 6-hr interval for patients with increased renal function has been proposed. Cefiderocol may be the first monotherapy for treating metallo beta-lactamases.

Suggestions for conserving new beta-lactams in severely ill patients

New or recently introduced beta-lactam molecules are available to treat infections caused by MDR GN bacteria. Since a rapid emergence of resistance has been described/reported for some of them, the judicious use of these drugs with the aim of preserving them from resistance is a high priority.

For some of these beta-lactam molecules their true specific clinical role together with the possible contribution of an associated therapy have not yet been defined. Moreover, ideal dosing strategies, especially for fragile/severely ill patients (such as ICU patients), are still lacking. Rapid bedside diagnostic tests to identify pathogens and detect their mechanisms of resistance will hopefully be available in the near future, together with real-world clinical data, which will help to provide evidence concerning their efficacy and risk of developing resistance.

Three Tables are reported below that compare these 5 new antibiotics according to their PK characteristics, spectrum of activity, PK/PD targets, susceptibility breakpoints and clinical use. Some suggestions for their use according to PK/PD parameters, with the aim of optimizing clinical outcome and containing the risk of resistance, are also reported.

All 5 molecules belong to the beta-lactam antibiotic class and share some characteristics. Being hydrophilic molecules, there is the risk that if prescribed according to the manufacturer’s dose they are under-dosed in critically ill septic patients, in whom permeability and volume of distribution are increased.Citation44 The risk of underexposure (which affects clinical cure and favors resistances) must be considered in ICU patients in whom renal function is altered. Renal function can be not only reduced, but also increased in a certain subset of ICU patients; moreover, kidney function is not static, and changes (both increasing and decreasing) can occur very rapidly.Citation15 Therefore, increased doses during increased renal clearances should be proposed, while even in the case of reduced renal function some authors suggest not reducing doses, at least for the first 48 hrs of treatment.Citation15 A keen revision of dosing according to CrCl formulas validated for ICU patients must also be applied. Finally, data on the probability of reaching PK/PD targets in ICU patients undergoing RRT are also necessary.

In order to limit the development of resistance there is the need for specific PK/PD studies, on specific subsets of patients, that can validate both the use of these molecules in deep-seated infections and, for some of them, the value of association therapy to limit the possibility of emergence of resistance. Finally, with the aim of customizing/individualizing antimicrobial therapy, the use of bedside therapeutic drug monitoring (TDM), possibly in a simplified, clinically affordable modality,Citation45 should be encouraged as part of routine care in the near future.

Collectively, these efforts may help to preserve these new drugs for a longer period.

Conflicts of interest

The authors report no conflict of interest.

Additional information

Notes on contributors

Chiara Adembri

Doctor Iacopo Cappellini MD, PhD, Anaesthesiologist, expert in Critical Care Medicine and infections in the critically ill patient.

Iacopo Cappellini

Prof Chiara Adembri MD, PhD, Anaesthesiologist, expert in antimicrobial therapy and infections in the critically ill patient.

Andrea Novelli

Prof Andrea Novelli, MD, pharmacologist, expert in antimicrobial therapy and PK/PD applied to antimicrobial therapy.

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.