Lung penetration, bronchopulmonary pharmacokinetic/pharmacodynamic profile and safety of 3 g of ceftolozane/tazobactam administered to ventilated, critically ill patients with pneumonia

Luzelena Caro, David P Nicolau, Jan J De Waele, Joseph L Kuti, Kajal B Larson, Elaine Gadzicki, Brian Yu, Zhen Zeng, Adedayo Adedoyin, Elizabeth G Rhee, Luzelena Caro, David P Nicolau, Jan J De Waele, Joseph L Kuti, Kajal B Larson, Elaine Gadzicki, Brian Yu, Zhen Zeng, Adedayo Adedoyin, Elizabeth G Rhee

Abstract

Objectives: Ceftolozane/tazobactam is approved for hospital-acquired/ventilator-associated bacterial pneumonia at double the dose (i.e. 2 g/1 g) recommended for other indications. We evaluated the bronchopulmonary pharmacokinetic/pharmacodynamic profile of this 3 g ceftolozane/tazobactam regimen in ventilated pneumonia patients.

Methods: This was an open-label, multicentre, Phase 1 trial (clinicaltrials.gov: NCT02387372). Mechanically ventilated patients with proven/suspected pneumonia received four to six doses of 3 g of ceftolozane/tazobactam (adjusted for renal function) q8h. Serial plasma samples were collected after the first and last doses. One bronchoalveolar lavage sample per patient was collected at 1, 2, 4, 6 or 8 h after the last dose and epithelial lining fluid (ELF) drug concentrations were determined. Pharmacokinetic parameters were estimated by non-compartmental analysis and pharmacodynamic analyses were conducted to graphically evaluate achievement of target exposures (plasma and ELF ceftolozane concentrations >4 mg/L and tazobactam concentrations >1 mg/L; target in plasma: ≥30% and ≥20% of the dosing interval, respectively).

Results: Twenty-six patients received four to six doses of study drug; 22 were included in the ELF analyses. Ceftolozane and tazobactam Tmax (6 and 2 h, respectively) were delayed in ELF compared with plasma (1 h). Lung penetration, expressed as the ratio of mean drug exposure (AUC) in ELF to plasma, was 50% (ceftolozane) and 62% (tazobactam). Mean ceftolozane and tazobactam ELF concentrations remained >4 mg/L and >1 mg/L, respectively, for 100% of the dosing interval. There were no deaths or adverse event-related study discontinuations.

Conclusions: In ventilated pneumonia patients, 3 g of ceftolozane/tazobactam q8h yielded ELF exposures considered adequate to cover ceftolozane/tazobactam-susceptible respiratory pathogens.

© The Author(s) 2020. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy.

Figures

Figure 1.
Figure 1.
Arithmetic mean (±SD) total concentration–time profiles in plasma [first dose (n = 25 patients) and last dose (n = 24 patients)] and ELF (last dose; n = 22 patients) for (a) ceftolozane and (b) tazobactam. Note: curves are slightly offset around each timepoint in order to achieve separation of error bars.
Figure 2.
Figure 2.
Individual unbound plasma concentration–time profiles for first (n = 25) and last (n = 24) doses, with target concentrations shown as horizontal lines, among patients included in the plasma PK analyses, for (a) ceftolozane first dose, (b) ceftolozane last dose, (c) tazobactam first dose and (d) tazobactam last dose.

References

    1. Kumar A. Life-threatening infections in the critically ill. Crit Care Clin 2013; 29: xiii–xiv.
    1. Sader HS, Farrell DJ, Flamm RK. et al. Antimicrobial susceptibility of Gram-negative organisms isolated from patients hospitalized in intensive care units in United States and European hospitals (2009–2011). Diagn Microbiol Infect Dis 2014; 78: 443–8.
    1. Zilberberg MD, Shorr AF.. Prevalence of multidrug-resistant Pseudomonas aeruginosa and carbapenem-resistant Enterobacteriaceae among specimens from hospitalized patients with pneumonia and bloodstream infections in the United States from 2000 to 2009. J Hosp Med 2013; 8: 559–63.
    1. Maraolo AE, Cascella M, Corcione S. et al. Management of multidrug-resistant Pseudomonas aeruginosa in the intensive care unit: state of the art. Expert Rev Anti Infect Ther 2017; 15: 861–71.
    1. Talbot GH, Das A, Cush S. et al. Evidence-based study design for hospital-acquired bacterial pneumonia and ventilator-associated bacterial pneumonia. J Infect Dis 2019; 219: 1536–44.
    1. Micek ST, Kollef MH, Torres A. et al. Pseudomonas aeruginosa nosocomial pneumonia: impact of pneumonia classification. Infect Control Hosp Epidemiol 2015; 36: 1190–7.
    1. Kalil AC, Metersky ML, Klompas M. et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis 2016; 63: e61–111.
    1. Kumar A, Roberts D, Wood KE. et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med 2006; 34: 1589–96.
    1. Kethireddy S, Bilgili B, Sees A. et al. Culture-negative septic shock compared with culture-positive septic shock: a retrospective cohort study. Crit Care Med 2018; 46: 506–12.
    1. Rhodes A, Evans LE, Alhazzani W. et al. Surviving Sepsis Campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med 2017; 43: 304–77.
    1. Ferrer R, Martin-Loeches I, Phillips G. et al. Empiric antibiotic treatment reduces mortality in severe sepsis and septic shock from the first hour: results from a guideline-based performance improvement program. Crit Care Med 2014; 42: 1749–55.
    1. Kumar A, Ellis P, Arabi Y. et al. Initiation of inappropriate antimicrobial therapy results in a fivefold reduction of survival in human septic shock. Chest 2009; 136: 1237–48.
    1. Kang CI, Kim SH, Kim HB. et al. Pseudomonas aeruginosa bacteremia: risk factors for mortality and influence of delayed receipt of effective antimicrobial therapy on clinical outcome. Clin Infect Dis 2003; 37: 745–51.
    1. Luyt CE, Hekimian G, Koulenti D. et al. Microbial cause of ICU-acquired pneumonia: hospital-acquired pneumonia versus ventilator-associated pneumonia. Curr Opin Crit Care 2018; 24: 332–8.
    1. Tamma PD, Rodriguez-Bano J.. The use of noncarbapenem β-lactams for the treatment of extended-spectrum β-lactamase infections. Clin Infect Dis 2017; 64: 972–80.
    1. Zhanel GG, Chung P, Adam H. et al. Ceftolozane/tazobactam: a novel cephalosporin/β-lactamase inhibitor combination with activity against multidrug-resistant gram-negative bacilli. Drugs 2014; 74: 31–51.
    1. Goodlet KJ, Nicolau DP, Nailor MD.. Ceftolozane/tazobactam and ceftazidime/avibactam for the treatment of complicated intra-abdominal infections. Ther Clin Risk Manag 2016; 12: 1811–26.
    1. Shortridge D, Pfaller MA, Castanheira M. et al. Antimicrobial activity of ceftolozane–tazobactam tested against Enterobacteriaceae and Pseudomonas aeruginosa collected from patients with bloodstream infections isolated in United States hospitals (2013–2015) as part of the Program to Assess Ceftolozane-Tazobactam Susceptibility (PACTS) surveillance program. Diagn Microbiol Infect Dis 2018; 92: 158–63.
    1. Castanheira M, Duncan LR, Mendes RE. et al. Activity of ceftolozane–tazobactam against Pseudomonas aeruginosa and Enterobacteriaceae isolates collected from respiratory tract specimens of hospitalized patients in the United States during 2013 to 2015. Antimicrob Agents Chemother 2018; 62: e02125-17.
    1. ZERBAXA™ (Ceftolozane and Tazobactam) for Injection, Full Prescribing Information. Merck Sharp & Dohme Corp, Whitehouse Station, NJ, USA, 2014.
    1. Miller B, Hershberger E, Benziger D. et al. Pharmacokinetics and safety of intravenous ceftolozane–tazobactam in healthy adult subjects following single and multiple ascending doses. Antimicrob Agents Chemother 2012; 56: 3086–91.
    1. Xiao AJ, Miller BW, Huntington JA. et al. Ceftolozane/tazobactam pharmacokinetic/pharmacodynamic-derived dose justification for phase 3 studies in patients with nosocomial pneumonia. J Clin Pharmacol 2016; 56: 56–66.
    1. Chandorkar G, Huntington JA, Gotfried MH. et al. Intrapulmonary penetration of ceftolozane/tazobactam and piperacillin/tazobactam in healthy adult subjects. J Antimicrob Chemother 2012; 67: 2463–9.
    1. Kollef MH, Novacek M, Kivistik U. 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: 1299–311.
    1. Neuner EA, Gallagher JC.. Pharmacodynamic and pharmacokinetic considerations in the treatment of critically Ill patients infected with carbapenem-resistant Enterobacteriaceae. Virulence 2017; 8: 440–52.
    1. Roberts JA, Abdul-Aziz MH, Lipman J. et al. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis 2014; 14: 498–509.
    1. Blot SI, Pea F, Lipman J.. The effect of pathophysiology on pharmacokinetics in the critically ill patient—concepts appraised by the example of antimicrobial agents. Adv Drug Deliv Rev 2014; 77: 3–11.
    1. Rodvold KA, George JM, Yoo L.. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antibacterial agents. Clin Pharmacokinet 2011; 50: 637–64.
    1. Rodvold KA, Hope WW, Boyd SE.. Considerations for effect site pharmacokinetics to estimate drug exposure: concentrations of antibiotics in the lung. Curr Opin Pharmacol 2017; 36: 114–23.
    1. Yu B, Adedoyin A, Hershberger E. et al. Safety, tolerability, and pharmacokinetics of 3 g of ceftolozane/tazobactam in healthy adults: a randomized, placebo-controlled, multiple-dose study. Clin Pharmacol Drug Dev 2018; 7: 382–91.
    1. Rennard SI, Basset G, Lecossier D. et al. Estimation of volume of epithelial lining fluid recovered by lavage using urea as marker of dilution. J Appl Physiol (1985) 1986; 60: 532–8.
    1. Craig WA, Andes DR.. In vivo activities of ceftolozane, a new cephalosporin, with and without tazobactam against Pseudomonas aeruginosa and Enterobacteriaceae, including strains with extended-spectrum β-lactamases, in the thighs of neutropenic mice. Antimicrob Agents Chemother 2013; 57: 1577–82.
    1. CLSI. Performance Standards for Antimicrobial Susceptibility Testing—Twenty-Ninth Edition: M100 2019.
    1. Melchers MJ, Mavridou E, van Mil AC. et al. Pharmacodynamics of ceftolozane combined with tazobactam against Enterobacteriaceae in a neutropenic mouse thigh model. Antimicrob Agents Chemother 2016; 60: 7272–9.
    1. Martinkova J, Malbrain M, Havel E. et al. A pilot study on pharmacokinetic/pharmacodynamic target attainment in critically ill patients receiving piperacillin/tazobactam. Anaesthesiol Intensive Ther 2016; 48: 23–8.
    1. Rizk ML, Bhavnani SM, Drusano G. et al. Considerations for dose selection and clinical pharmacokinetics/pharmacodynamics for the development of antibacterial agents. Antimicrob Agents Chemother 2019; 63: e02309-18.
    1. Natesan SP, 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: 2813–16.
    1. Xiao AJ, Caro L, Popejoy MW. et al. PK/PD target attainment with ceftolozane/tazobactam using Monte Carlo simulation in patients with various degrees of renal function, including augmented renal clearance and end-stage renal disease. Infect Dis Ther 2017; 6: 137–48.
    1. Aiudi A, Miller B, Krishna G. et al. Pharmacokinetics, safety, and tolerability of ceftolozane/tazobactam in healthy Japanese, Chinese, and white subjects. Fundam Clin Pharmacol 2016; 30: 625–33.
    1. Rizk ML, Rhee EG, Jumes PA. et al. Intrapulmonary pharmacokinetics of relebactam, a novel β-lactamase inhibitor, dosed in combination with imipenem–cilastatin in healthy subjects. Antimicrob Agents Chemother 2018; 62: e01411-17.
    1. Wenzler E, Gotfried MH, Loutit JS. et al. Meropenem-RPX7009 concentrations in plasma, epithelial lining fluid, and alveolar macrophages of healthy adult subjects. Antimicrob Agents Chemother 2015; 59: 7232–9.
    1. Nicolau DP, Siew L, Armstrong J. 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: 2862–9.
    1. Riccobene TA, Pushkin R, Jandourek A. et al. Penetration of ceftaroline into the epithelial lining fluid of healthy adult subjects. Antimicrob Agents Chemother 2016; 60: 5849–57.
    1. Chandorkar G, Xiao A, Mouksassi MS. et al. Population pharmacokinetics of ceftolozane/tazobactam in healthy volunteers, subjects with varying degrees of renal function and patients with bacterial infections. J Clin Pharmacol 2015; 55: 230–9.
    1. Wooley M, Miller B, Krishna G. et al. Impact of renal function on the pharmacokinetics and safety of ceftolozane–tazobactam. Antimicrob Agents Chemother 2014; 58: 2249–55.
    1. Crass RL, Rodvold KA, Mueller BA. et al. Renal dosing of antibiotics: are we jumping the gun? Clin Infect Dis 2019; 68: 1596–602.
    1. Motos A, Kuti JL, Li Bassi G. et al. Is one sample enough? β-Lactam target attainment and penetration into epithelial lining fluid based on multiple bronchoalveolar lavage sampling time points in a swine pneumonia model. Antimicrob Agents Chemother 2019; 63: e01922-18..

Source: PubMed

3
Subscribe