Articles
Ceftolozane–tazobactam versus meropenem for treatment of nosocomial pneumonia (ASPECT-NP): a randomised, controlled, double-blind, phase 3, non-inferiority trial

https://doi.org/10.1016/S1473-3099(19)30403-7Get rights and content

Summary

Background

Nosocomial pneumonia due to antimicrobial-resistant pathogens is associated with high mortality. We assessed the efficacy and safety of the combination antibacterial drug ceftolozane–tazobactam versus meropenem for treatment of Gram-negative nosocomial pneumonia.

Methods

We conducted a randomised, controlled, double-blind, non-inferiority trial at 263 hospitals in 34 countries. Eligible patients were aged 18 years or older, were undergoing mechanical ventilation, and had nosocomial pneumonia (either ventilator-associated pneumonia or ventilated hospital-acquired pneumonia). Patients were randomly assigned (1:1) with block randomisation (block size four), stratified by type of nosocomial pneumonia and age (<65 years vs ≥65 years), to receive either 3 g ceftolozane–tazobactam or 1 g meropenem intravenously every 8 h for 8–14 days. The primary endpoint was 28-day all-cause mortality (at a 10% non-inferiority margin). The key secondary endpoint was clinical response at the test-of-cure visit (7–14 days after the end of therapy; 12·5% non-inferiority margin). Both endpoints were assessed in the intention-to-treat population. Investigators, study staff, patients, and patients' representatives were masked to treatment assignment. Safety was assessed in all randomly assigned patients who received study treatment. This trial was registered with ClinicalTrials.gov, NCT02070757.

Findings

Between Jan 16, 2015, and April 27, 2018, 726 patients were enrolled and randomly assigned, 362 to the ceftolozane–tazobactam group and 364 to the meropenem group. Overall, 519 (71%) patients had ventilator-associated pneumonia, 239 (33%) had Acute Physiology and Chronic Health Evaluation II scores of at least 20, and 668 (92%) were in the intensive care unit. At 28 days, 87 (24·0%) patients in the ceftolozane–tazobactam group and 92 (25·3%) in the meropenem group had died (weighted treatment difference 1·1% [95% CI −5·1 to 7·4]). At the test-of-cure visit 197 (54%) patients in the ceftolozane–tazobactam group and 194 (53%) in the meropenem group were clinically cured (weighted treatment difference 1·1% [95% CI −6·2 to 8·3]). Ceftolozane–tazobactam was thus non-inferior to meropenem in terms of both 28-day all-cause mortality and clinical cure at test of cure. Treatment-related adverse events occurred in 38 (11%) of 361 patients in the ceftolozane–tazobactam group and 27 (8%) of 359 in the meropenem group. Eight (2%) patients in the ceftolozane–tazobactam group and two (1%) in the meropenem group had serious treatment-related adverse events. There were no treatment-related deaths.

Interpretation

High-dose ceftolozane–tazobactam is an efficacious and well tolerated treatment for Gram-negative nosocomial pneumonia in mechanically ventilated patients, a high-risk, critically ill population.

Funding

Merck & Co.

Introduction

Nosocomial pneumonia, which is often associated with mechanical ventilation, is one of the most common hospital-acquired infections and is associated with high mortality.1, 2 Crude mortality estimates range from 20% to 50%;3 infections caused by multidrug-resistant bacteria are associated with a particularly high mortality risk.4 Rising multidrug resistance among Gram-negative pathogens, including Pseudomonas aeruginosa and Enterobacteriaceae (Enterobacterales), is widely recognised as a major public health issue globally.5, 6, 7 Resistant pathogens are especially problematic in critically ill patients, who are at high risk of adverse clinical outcomes,7 and in whom up to 20–30% of cases of ventilator-associated pneumonia due to P aeruginosa are caused by multidrug-resistant strains.8 New treatment options for nosocomial pneumonia are therefore urgently needed.

Previous large phase 3 trials9, 10, 11 in patients with nosocomial pneumonia have been unable to show non-inferiority of several novel drugs (eg, tigecycline, doripenem, and ceftobiprole) to established therapies. However, underdosing of these novel drugs might have contributed to the negative results.12, 13, 14 Many patients with nosocomial pneumonia are critically ill, and the pharmacokinetic and pharmacodynamic profiles for antimicrobials in such patients are frequently complex, potentially leading to rapid drug elimination and changes in volume of distribution.12 Additionally, drug concentrations in the lungs are often lower than those in plasma,12 and causative pathogens in nosocomial pneumonia often have reduced antibacterial susceptibility.15, 16, 17 The combination of these factors could lead to insufficient drug concentrations at the infection site, and thus antibacterial dosing regimens in patients with nosocomial pneumonia should be carefully optimised.12

Research in context

Evidence before this study

Nosocomial pneumonia is one of the most common and serious hospital-acquired infections (crude mortality 20–50%). We searched PubMed with terms including “hospital-acquired pneumonia”, “ventilator-associated pneumonia”, “phase 3”, and “randomised” for randomised, controlled trials published in any language between July 1, 2009, and July 1, 2019, that assessed antibacterial agents for the treatment of nosocomial pneumonia. Full details of the search are provided in the appendix (p 33). Previous clinical trials showed higher mortality at 28 days in patients with ventilated hospital-acquired pneumonia than in those with ventilator-associated pneumonia. Nosocomial pneumonia is frequently caused by Gram-negative pathogens, including Pseudomonas aeruginosa and Enterobacteriaceae. Selection of appropriate antibacterial therapy is increasingly complicated by the rising incidence of multidrug resistance among these key causative pathogens, and this problem is widely recognised as a major, global public health issue. New safe and effective antibacterial drugs are thus urgently needed, but phase 3 trials of novel drugs tigecycline, doripenem, and ceftobiprole were unsuccessful. Ceftolozane–tazobactam, a novel combination of a potent anti-pseudomonal cephalosporin and a β-lactamase inhibitor, is approved for complicated urinary tract and intra-abdominal infections. Its profile suggests that it would also be an efficacious treatment for Gram-negative nosocomial pneumonia.

Added value of this study

This trial is the first randomised, controlled study to assess the efficacy and safety of ceftolozane–tazobactam for nosocomial pneumonia, an infection for which additional treatment options are urgently needed. Unlike most non-inferiority studies of other novel antibacterial agents in the same clinical setting, we enrolled only mechanically ventilated patients—specifically, those with ventilator-associated pneumonia or ventilated hospital-acquired pneumonia, who have higher mortality than non-ventilated patients with nosocomial pneumonia. Notably, on the basis of pharmacokinetic–pharmacodynamic modelling, we selected a dose of ceftolozane–tazobactam that was twice that approved for other indications. Ceftolozane–tazobactam was non-inferior to meropenem in both the primary endpoint of 28-day all-cause mortality and the key secondary endpoint of clinical response at the test-of-cure visit, irrespective of causative pathogens (most commonly Enterobacteriaceae and P aeruginosa). It also seemed to be well tolerated in this critically ill population, with a low incidence of treatment-related adverse events.

Implications of all the available evidence

High-dose ceftolozane–tazobactam can be used to treat nosocomial pneumonia caused by P aeruginosa (including multidrug-resistant strains), Enterobacteriaceae (including producers of extended-spectrum β-lactamases), and other Gram-negative pathogens.

Ceftolozane–tazobactam is a novel combination anti-bacterial consisting of ceftolozane (a potent anti-pseudomonal cephalosporin) and tazobactam (a β-lactamase inhibitor).18 It is approved for complicated urinary tract and intra-abdominal infections at a dose of 1·5 g (ie, 1 g ceftolozane and 0·5 g tazobactam) every 8 h.19 Ceftolozane–tazobactam is active in vitro against many important pathogens associated with nosocomial pneumonia, including multidrug-resistant pseudomonal species and Enterobacteriaceae that produce extended-spectrum β-lactamases (ESBLs),15, 19 and had good lung penetration in two phase 1 trials20, 21 (one of which was done in critically ill patients undergoing mechanical ventilation). These findings suggest that the combination would be efficacious against Gram-negative nosocomial pneumonia.

We therefore aimed to assess the efficacy and safety of ceftolozane–tazobactam compared with meropenem (an established, broad-spectrum, first-line treatment) in patients with nosocomial pneumonia. To ensure sufficient drug concentrations in patients' lungs, we used a new dosing regimen for ceftolozane–tazobactam (ie, double the dose approved for other indications).21, 22, 23

Section snippets

Study design and participants

Protocol MK-7625A-008 (ASPECT-NP) was a randomised, controlled, double-blind, phase 3, non-inferiority trial done at 263 hospitals in 34 countries. Eligible patients were aged 18 years or older, were intubated and mechanically ventilated, and had ventilator-associated pneumonia or ventilated hospital-acquired pneumonia. Pneumonia was diagnosed if patients had the following clinical and radiographic criteria within 24 h before the first dose of study drug: purulent tracheal secretions with at

Results

Between Jan 16, 2015, and April 27, 2018, 726 patients were enrolled and randomly assigned, 362 to the ceftolozane–tazobactam group and 364 to the meropenem group (intention-to-treat population; figure 1). Patients were enrolled at 119 of the 263 participating hospitals in 29 countries (appendix p 3). Premature unblinding occurred in six patients in each group (ie, one instance of protocol-allowed unblinding in the meropenem group and 11 instances of accidental unblinding that involved

Discussion

In this randomised, controlled trial, we showed that ceftolozane–tazobactam was non-inferior to meropenem in terms of 28-day mortality and clinical response in patients with nosocomial pneumonia, a clinically challenging infection that is associated with a high risk of treatment failure and death.1, 3, 8, 30 Both study drugs had similar efficacy across all secondary endpoints, including microbiological endpoints. The baseline pathogen distribution was consistent with previous reports, with most

Data sharing

Merck Sharp & Dohme's data sharing policy, including restrictions, is available online. Requests for access to the clinical study data can be submitted online or via email ([email protected]).

References (37)

  • SS Magill et al.

    Multistate point-prevalence survey of health care-associated infections

    N Engl J Med

    (2014)
  • AC Kalil 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)
  • A Rodrigo-Troyano et al.

    The respiratory threat posed by multidrug resistant Gram-negative bacteria

    Respirology

    (2017)
  • Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics

    (2017)
  • M Bassetti et al.

    Antimicrobial resistance in the next 30 years, humankind, bugs and drugs: a visionary approach

    Intensive Care Med

    (2017)
  • ST Micek et al.

    Pseudomonas aeruginosa nosocomial pneumonia: impact of pneumonia classification

    Infect Control Hosp Epidemiol

    (2015)
  • SS Awad et al.

    A phase 3 randomized double-blind comparison of ceftobiprole medocaril versus ceftazidime plus linezolid for the treatment of hospital-acquired pneumonia

    Clin Infect Dis

    (2014)
  • MH Kollef et al.

    A randomized trial of 7-day doripenem versus 10-day imipenem–cilastatin for ventilator-associated pneumonia

    Crit Care

    (2012)
  • Cited by (221)

    • Managing Antimicrobial Resistance in the Emergency Department

      2024, Emergency Medicine Clinics of North America
    View all citing articles on Scopus
    View full text