Advertisement
Original Article| Volume 12, ISSUE 2, P130-140, March 2013

Download started.

Ok

Inhaled aztreonam lysine vs. inhaled tobramycin in cystic fibrosis: A comparative efficacy trial

Open ArchivePublished:September 17, 2012DOI:https://doi.org/10.1016/j.jcf.2012.07.006

      Abstract

      Background

      Open-label, parallel-group, international trial comparing aztreonam for inhalation solution (AZLI) and tobramycin nebulizer solution (TNS) for cystic fibrosis patients with airway Pseudomonas aeruginosa.

      Methods

      273 patients (≥6 years); randomized to three 28-day courses (AZLI 75 mg [three‐times/day] or TNS 300 mg [twice/day]); 28 off-days separated each course.

      Results

      268 patients were treated (AZLI/TNS: 136/132). Mean baseline FEV1 was 52% predicted. Mean relative changes after 1 course (AZLI: 8.35%; TNS: 0.55%; p<0.001) and mean actual changes across 3 courses (AZLI: 2.05%; TNS: −0.66%; p=0.002) indicated AZLI statistical superiority vs. TNS. AZLI-treated patients had fewer respiratory hospitalizations (p=0.044) and respiratory events requiring additional antipseudomonal antibiotics (p=0.004); both treatments were well tolerated. 133 patients received 1 to 3 courses of AZLI treatment in the open-label extension-period (28-day courses separated by 28 days off-treatment); lung function improvements were comparable regardless of whether patients had received TNS or AZLI in the preceding comparative period.

      Conclusions

      AZLI demonstrated statistical superiority in lung function and a reduction in acute pulmonary exacerbations compared to TNS over 3 treatment courses (ClinicalTrials.gov: NCT00757237).

      Keywords

      1. Introduction

      Inhaled antipseudomonal antibiotics are standard of care for cystic fibrosis (CF) patients with chronic Pseudomonas aeruginosa (PA) airway infection [
      • Flume P.A.
      • O'Sullivan B.P.
      • Robinson K.A.
      • Goss C.H.
      • Mogayzel Jr., P.J.
      • Willey-Courand D.B.
      • et al.
      Cystic fibrosis pulmonary guidelines. Chronic medications for maintenance of lung health.
      ,
      • Heijermann H.
      • Westerman E.
      • Conway S.
      • Touw D.
      • Döring G.
      • Working Group Consensus
      Inhaled medication and inhalation devices for lung disease in patients with cystic fibrosis: a European consensus.
      ]. Relatively high delivered drug dose maximizes airway concentrations, and low systemic absorption reduces side effects. Three inhaled antipseudomonal antibiotics with different mechanisms of action are available for treating CF patients: tobramycin nebulizer solution (TNS; an aminoglycoside), colistimethate sodium (colistin; a polymyxin; not licensed in the US), and aztreonam for inhalation solution (AZLI; a monobactam) [
      • US prescribing information
      TOBI®, tobramycin inhalation solution, USP.
      ,
      • Summary of Product Characteristics (SPC)
      ,
      • Summary of Product Characteristics
      , ,
      • Prescribing Information U.S.
      ,
      • Summary of Product Characteristics (SPC)
      ]. TNS has proven efficacy for CF patients, and established the standard of care of every other month inhaled antibiotic treatment for suppression of chronic airway infection [
      • Ramsey B.W.
      • Pepe M.S.
      • Quan J.M.
      • Otto K.L.
      • Montgomery A.B.
      • Williams-Warren J
      • et al.
      Cystic fibrosis inhaled tobramycin study group. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis.
      ]. Since AZLI was recently approved, it is appropriate to evaluate it against standard of care, given the ongoing need for more effective and better tolerated inhaled antibiotic treatments.
      AZLI is a lyophilized formulation of the monobactam antibiotic aztreonam, specifically designed for inhalation therapy with lysine as an excipient [
      • Prescribing Information U.S.
      ,
      • Summary of Product Characteristics (SPC)
      ,
      Prescribing information: AZACTAM® (aztreonam) for injection, USP.
      ]. In previous placebo-controlled clinical trials, efficacy and safety of AZLI were demonstrated in patients with CF and PA airway infection who previously received TNS treatment and in patients with relatively little previous TNS exposure. Additionally, therapeutic benefits observed in the placebo-controlled trials were sustained across 18 months of open-label treatment [
      • McCoy K.S.
      • Quittner A.L.
      • Oermann C.M.
      • Gibson R.L.
      • Retsch-Bogart G.Z.
      • Montgomery A.B.
      Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis.
      ,
      • Retsch-Bogart G.Z.
      • Quittner A.L.
      • Gibson R.L.
      • Oermann C.M.
      • McCoy K.S.
      • Montgomery A.B.
      • et al.
      Efficacy and safety of inhaled aztreonam lysine for airway Pseudomonas in cystic fibrosis.
      ,
      • Oermann C.M.
      • Retsch-Bogart G.Z.
      • Quittner A.L.
      • Gibson R.L.
      • McCoy K.S.
      • Montgomery A.B.
      • et al.
      An 18-month study, AIR-CF3, of the safety and improvement in pulmonary function and respiratory symptoms with repeated courses of aztreonam for inhalation solution in patients with cystic fibrosis and airway Pseudomonas aeruginosa.
      ].
      The trial described herein compared the efficacy and safety of AZLI to TNS, across three 28‐day treatment courses. An optional open‐label extension period for eligible patients included 3 additional AZLI treatment courses.

      2. Methods

      2.1 Study design

      This open-label, randomized, parallel-group, active-comparator study was conducted at 91 CF‐centers in Europe and the US (August 2008–May 2010). Eligible patients were randomized (1:1) to three 28-days on/28 days off treatment courses of AZLI (Cayston®, Gilead Sciences Inc.) 75 mg three‐times daily or TNS (TOBI®, Novartis Pharmaceuticals Corp.) 300 mg twice daily (Online Fig. 1) [
      • US prescribing information
      TOBI®, tobramycin inhalation solution, USP.
      ,
      • Summary of Product Characteristics (SPC)
      ,
      • Prescribing Information U.S.
      ,
      • Summary of Product Characteristics (SPC)
      ]. The 26‐week active-comparator period was followed by an optional, 24‐week, open‐label, single‐arm, extension period of 3 AZLI treatment-cycles for eligible patients at European study sites (completed: Nov 2010).
      The study was conducted in accordance with principles of the Declaration of Helsinki (as amended in Edinburgh, Tokyo, Venice, Hong Kong, and South Africa), International Conference on Harmonisation guidelines, Good Clinical Practice principles, and laws/regulations of each study location. Institutional Review Boards/Ethics Committees approved the study for each site. Patients/parents provided written informed consent/assent prior to undergoing study procedures.

      2.2 Participants

      Eligible patients (≥6 years of age) had a documented CF diagnosis, PA-positive sputum culture within the previous 3 months, and FEV175% predicted at screening. Additional TNS use was prohibited during the active-comparator period. Additional antipseudomonal antibiotics could be administered for symptoms consistent with the diagnosis of acute pulmonary exacerbation: decreased exercise tolerance, increased cough, increased sputum/chest congestion, or decreased appetite [
      • Rosenfeld M.
      • Emerson J.
      • Williams-Warren J.
      • Pepe M.
      • Smith A.
      • Montgomery A.B.
      • et al.
      Defining a pulmonary exacerbation in cystic fibrosis.
      ]. Patients receiving additional antipseudomonal antibiotics at any point after randomization could continue study treatments.

      2.3 Study endpoints

      Co-primary efficacy endpoints were designed to satisfy registrational requirements based on European Medicines Agency and the US Food and Drug Administration guidance: non‐inferiority of AZLI for relative change in FEV1% predicted at Day 28 and superiority of AZLI for actual change in FEV1% predicted across 3 treatment cycles. Selected secondary and tertiary endpoints were: time-to-need for IV antipseudomonal antibiotics for respiratory events, change from baseline on cystic fibrosis questionnaire—revised (CFQ-R) respiratory symptoms scale (RSS), number of respiratory hospitalizations, additional antipseudomonal antibiotic use, weight change from baseline, change in sputum PA density (colony forming units per gram sputum [CFU/g]), and responses to the treatment satisfaction questionnaire for medication (TSQM) [
      • Quittner A.L.
      • Buu A.
      • Messer M.A.
      • Modi A.C.
      • Watrous M.
      Development and validation of the cystic fibrosis questionnaire in the United States: a health related quality of life measure for cystic fibrosis.
      ,
      • Atkinson M.J.
      • Sinha A.
      • Hass S.L.
      • Colman S.S.
      • Kumar R.N.
      • Brod M.
      • et al.
      Validation of a general measure of treatment satisfaction, the treatment satisfaction questionnaire for medication (TSQM), using a national panel study of chronic disease.
      ]. An independent, blinded data adjudication committee determined respiratory hospitalizations and respiratory events requiring additional antipseudomonal antibiotics.

      2.4 Statistical analysis

      Statistical analyses were performed on the intent-to-treat (ITT) population: randomized patients receiving ≥1 dose of AZLI/TNS. The primary non‐inferiority endpoint was assessed with an analysis of covariance (ANCOVA) model with terms for treatment, baseline FEV1% predicted (continuous variable), and inhaled tobramycin use in previous year (≥84, <84 days). If the 95% confidence interval (CI; 2-sided) lower boundary for the treatment difference (AZLI–TNS) was >4% (pre‐specified non‐inferiority margin), non-inferiority of AZLI to TNS was concluded. Superiority was concluded if the lower limit of the 95% CI was above zero (i.e., p<0.05).
      The primary superiority endpoint was the average least‐square means from Weeks 4, 12, and 20 visits, based on a mixed-effect model repeated measures (MMRM) analysis method outlined by Siddiqui, which included terms for treatment, baseline FEV1% predicted (continuous variable), inhaled tobramycin use (≥84, <84 days), visit, and treatment/visit interaction [
      • Siddiqui O.
      • Hung H.M.
      • O'Neill R.
      MMRM vs. LOCF: a comprehensive comparison based on simulation study and 25 NDA datasets.
      ].

      3. Results

      3.1 Disposition

      Of 273 randomized patients, 268 received treatment with AZLI (n=136) or TNS (n=132); 233 patients (85.3%) completed the active-comparator period (Fig. 1). Mean use of distributed vials was 94.0% (AZLI) and 94.2% (TNS). Of 169 eligible patients, 133 (78.7%) entered the open‐label extension period (AZLI: 68; TNS: 65), and 118 patients completed 3 AZLI courses (88.7%).
      Figure thumbnail gr1
      Fig. 1Patient disposition. Efficacy and safety analyses for the active-comparator period were conducted on the intent to treat (ITT) population, which included 268 randomized and treated patients (AZLI: 136; TNS: 132).

      3.2 Patient characteristics

      Patient characteristics were balanced between treatment groups (Table 1). Mean age was 25.5 years, with 78.0% of patients ≥18 years of age (n=209/268). FEV1 was ≤50% predicted for 43.7% of patients (n=117). Inhaled tobramycin use ≥84 days in the previous year (equivalent to at least three 28-day courses) was reported for 85.1% of patients (n=228). The 40 patients (AZLI: 21, TNS: 19) with inhaled tobramycin use <84 days during the previous year were a heterogeneous group: 25 used no tobramycin in the previous year, some used inhaled tobramycin more than 1 year prior, and many had used inhaled colistin. The incidence of multi-drug resistant PA for cohorts with <84 or ≥84 days of inhaled tobramycin use in the previous year was comparable (34.4% vs. 30.6%, respectively), indicating significant prior antibiotic exposure in both groups. Characteristics of the 133 European patients who entered the extension period were similar to those of the overall group.
      Table 1Baseline demographic and clinical characteristics.
      AZLI-treated n=136TNS-treated n=132Overall n=268
      Region; n (%)
       Europe92 (67.6)82 (62.1)174 (64.9)
       United States44 (32.4)50 (37.9)94 (35.1)
      Age, years; mean (SD)25.8 (9.1)25.1 (9.0)25.5 (9.0)
      Age group; n (%)
       6 to 12 years8 (5.9)5 (3.8)13 (4.9)
       >12 to <18 years20 (14.7)26 (19.7)46 (17.2)
       ≥18 years108 (79.4)101 (76.5)209 (78.0)
      Male gender; n (%)68 (50.0)66 (50.0)134 (50.0)
      White race; n (%)130 (95.6)131 (99.2)261 (97.4)
      Body mass index, kg/m2; mean (SD)20.2 (3.0)20.5 (2.8)20.4 (2.9)
      CFTR genotype;
      n=117, 111, 228 for 3 columns.
      n (%)
       Homozygous for ∆50864 (54.7)60 (54.1)124 (54.4)
       Heterozygous for ∆50836 (30.8)30 (27.0)66 (28.9)
       Unidentified or other17 (14.5)21 (18.9)38 (16.7)
      FEV1% predicted at screening; n (%)
       >50% to ≤75%76 (55.9)75 (56.8)151 (56.3)
       ≤50%60 (44.1)57 (43.2)117 (43.7)
       <25%6 (4.4)6 (4.5)12 (4.5)
      Inhaled tobramycin use in previous year;
      Enrollment included ≤40 patients with <84days inhaled tobramycin use in the previous year. One patient was mistakenly stratified to ≥84days and 2 patients were mistakenly stratified to <84days.
      n (%)
       <84 days21 (15.4)19 (14.4)40 (14.9)
       ≥84 days115 (84.6)113 (85.6)228 (85.1)
      Inhaled colistin use in previous year;
      Colistin is not licensed in the US.
      n (%)
      50 (36.8%)53 (40.2%)103 (38.4%)
      Azithromycin use at baseline; n (%)85 (62.5)89 (67.4)174 (64.9)
      Dornase alfa use at baseline; n (%)92 (67.6)91 (68.9)183 (68.3)
      Hypertonic saline use at baseline; n (%)44 (32.4)46 (34.8)90 (33.6)
      Baseline values; mean (SD)
       FEV1% predicted52.3 (15.6)52.2 (14.6)52.3 (15.1)
       FEV1 (L)1.8 (0.6)1.8 (0.6)1.8 (0.6)
       Log10 PA CFU per g sputum
      n=102, 103, and 205 for 3 columns.
      6.4 (2.1)5.9 (2.7)6.2 (2.4)
       CFQ-R respiratory symptoms score
      n=131, 131, and 262 for 3 columns.
      62.9 (20.4)58.0 (20.8)60.4 (20.7)
      MIC of aztreonam for all PA isolates at baseline
      n=115, 110, and 225 for 3 columns.
       MIC50; μg/mL222
       MIC90; μg/mL3212864
       Minimum MIC; μg/mL111
       Maximum MIC; μg/mL>20482048>2048
       Isolates tested; n208198406
      MIC of tobramycin for all PA isolates at baseline
      n=115, 110, and 225 for 3 columns.
       MIC50; μg/mL222
       MIC90; μg/mL646464
       Minimum MIC; μg/mL0.120.120.12
       Maximum MIC; μg/mL>1024>1024>1024
       Isolates tested; n208198406
      Multi-drug resistant PA;
      n=115, 110, and 225 for 3 columns.
      Resistance assessed using CF Foundation definition: resistance to all antibiotics tested in 2 of the 3 drug classes (aminoglycosides, beta-lactams, quinolones) [18]. CFQ-R = cystic fibrosis questionnaire—revised; CFTR = cystic fibrosis transmembrane conductance regulator; CFU = colony forming units; FEV1 = forced expiratory volume in 1s; MIC = minimum inhibitory concentration; PA = Pseudomonas aeruginosa.
      n (%)
      35 (30.4)35 (31.8)70 (31.1)
      a n=117, 111, 228 for 3 columns.
      b Enrollment included ≤40 patients with <84 days inhaled tobramycin use in the previous year. One patient was mistakenly stratified to ≥84 days and 2 patients were mistakenly stratified to <84 days.
      c Colistin is not licensed in the US.
      d n=102, 103, and 205 for 3 columns.
      e n=131, 131, and 262 for 3 columns.
      f n=115, 110, and 225 for 3 columns.
      g Resistance assessed using CF Foundation definition: resistance to all antibiotics tested in 2 of the 3 drug classes (aminoglycosides, beta-lactams, quinolones)
      • Saiman L.
      • Siegel J.
      • the CF Foundation Consensus Conference on Infection Control Participants
      Infection control recommendations for patients with cystic fibrosis: microbiology, important pathogens, and infection control practices to prevent patient-to-patient transmission.
      . CFQ-R = cystic fibrosis questionnaire—revised; CFTR = cystic fibrosis transmembrane conductance regulator; CFU = colony forming units; FEV1 = forced expiratory volume in 1 s; MIC = minimum inhibitory concentration; PA = Pseudomonas aeruginosa.

      3.3 Efficacy — active-comparator period

      Mean relative changes from baseline FEV1% predicted at Day 28 were 8.35% (AZLI) and 0.55% (TNS; Fig. 2A ). The treatment difference (AZLI–TNS) was 7.80% (95% CI=3.86%, 11.73%; p<0.001). The lower CI boundary was >0, indicating AZLI was superior to TNS.
      Figure thumbnail gr2
      Fig. 2Change in FEV1% predicted. A. Adjusted mean relative change from baseline FEV1% predicted (MMRM analysis). The difference between AZLI and TNS after the first treatment course (Day 28) was the non‐inferiority co‐primary efficacy endpoint, shown by the arrow; results shown in the box were based on an ANCOVA analysis, as described in the section. AZLI–TNS treatment differences were 6.77 at Week 2 (p<0.001); 7.88 at Week 4 (p<0.001); 6.04 at Week 12 (p=0.007), and 5.89 at Week 20 (p=0.004). B. Adjusted mean actual change from baseline FEV1% predicted (MMRM analysis). The difference between AZLI and TNS across 3 treatment courses was the superiority co-primary efficacy endpoint, as described in the box. AZLI–TNS treatment differences were 3.24 at Week 2 (p<0.001); 3.35 at Week 4 (p<0.001); 2.61 at Week 12 (p=0.02), and 2.15 at Week 20 (p=0.03). C. Adjusted mean relative change from baseline FEV1% predicted, with censoring for additional antibiotic use. The MMRM analysis shown in panel A was repeated with censoring of subsequent FEV1% predicted values after a course of IV/inhaled antipseudomonal antibiotics was prescribed for treatment of respiratory symptoms. AZLI–TNS treatment differences (trt. diff.) are shown at Week 2 (p<0.001), Week 4 (p<0.001), Week 12 (p=0.002) and Week 20 (p=0.005). Patients censored: AZLI-treated 52, TNS-treated 76. D. Adjusted mean relative change from baseline FEV1% predicted, for patients participating in the open-label AZLI extension period. The MMRM analysis shown in panel A was repeated for this patient population. TNS/AZLI patients received TNS during the active-comparator period and AZLI during the open-label extension period.
      Mean actual changes from baseline FEV1% predicted across 3 treatment courses were 2.05% (AZLI) and −0.66% (TNS). The treatment difference (AZLI–TNS) was 2.70% (p=0.002; Fig. 2B), again demonstrating AZLI superiority over TNS. Both mean relative change and mean actual change from baseline FEV1% predicted were significantly larger for AZLI‐treated than for TNS‐treated patients after each on-treatment course (Fig. 2A, B).
      Across 3 treatment cycles in the active-comparator period, mean relative changes in FEV1% predicted were 3.11% (AZLI) and −0.29% (TNS). The treatment difference (AZLI–TNS) was statistically significant (3.4%, p=0.02). The treatment difference for actual change in FEV1% predicted across all active-comparator study visits favored AZLI and approached statistical significance (AZLI: 0.85%, TNS: −0.5%; treatment difference 1.35%, p=0.066).
      AZLI–TNS differences in mean relative change from baseline FEV1% predicted at Day 28 were statistically significant for several subgroups (Online Fig. 2).
      Average change from baseline CFQ-R RSS across 3 treatment cycles was significantly larger for AZLI-treated than for TNS‐treated patients (Table 2). Decreases in sputum PA density after each course were similar for AZLI and TNS-treated patients. AZLI-treated patients demonstrated weight gains above baseline at each study visit, while TNS-treated patients' body weight remained at or below baseline. Differences favoring AZLI were statistically significant at Weeks 4, 12, and 16, but not at Week 24. Scores on the TSQM effectiveness and global satisfaction scales were significantly higher for AZLI‐treated than TNS-treated patients.
      Table 2Selected secondary and tertiary efficacy endpoints.
      EndpointAZLI-treated n=136TNS-treated n=132p-Value
      Time to need for IV antipseudomonal antibiotics for respiratory events
      Kaplan–Meier method; p-value based on log rank test.
       Median number of days (95% CI)NE (177, NE)151 (113, NE)0.003
       Event rate at Week 24 (end of active-comparator period), %3654
      Time-to-first respiratory hospitalization
      Kaplan–Meier method; p-value based on log rank test.
       Median number of days (95% CI)NE (NE, NE)NE (NE, NE)0.111
       Event rate at Week 24 (end of active-comparator period), %2431
      CFQ-R respiratory symptoms scale, change from baseline score,
      Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      adjusted mean (SE)
       Week 4 (after course 1; AZLI: n=131; TNS: n=131)8.2 (1.7)2.6 (1.7)0.005
       Average across 3 courses (Weeks 4, 12, 20; AZLI: n=131; TNS: n=131)6.3 (1.5)2.2 (1.5)0.019
      Number of respiratory hospitalizations
      p-Value is for comparison of the number of events for each patient adjusted for time on study through Week 24 based on a negative binomial regression model.
      40580.044
      Number of respiratory events requiring IV and/or additional inhaled antipseudomonal antibiotics
      p-Value is for comparison of the number of events for each patient adjusted for time on study through Week 24 based on a negative binomial regression model.
      841210.004
      Patients requiring IV and/or additional inhaled antipseudomonal antibiotics for respiratory events,
      p-Value based on Fisher's exact test.
      n (%)
      52 (38.2)76 (57.6)0.002
      Time to need for IV or additional inhaled antipseudomonal antibiotics for respiratory events
      Kaplan–Meier method; p-value based on log rank test.
       Median number of days (95% CI)NE (177, NE)117 (102, 169)<0.001
       Event rate at Week 24 (end of active-comparator period), %3858
      Days of oral, IV, and/or additional inhaled antipseudomonalantibiotic use,
      p-Value based on Wilcoxon rank sum test.
      mean (SD)
      21.3 (24.8)28.4 (31.5)0.060
      Log10 PA CFU/g sputum, change from baseline,
      Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      adjusted mean (SE)
       Week 4 (after course 1; AZLI: n=88; TNS: n=94)0.60 (0.23)0.34 (0.23)0.330
       Average across 3 courses (Weeks 4, 12, 20; AZLI: n=97; TNS: n=97)0.55 (0.19)0.32 (0.19)0.295
      Weight, relative change from baseline at Week 24 (end of active‐comparator period),
      Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      %, adjusted mean (SE)
      0.58 (0.41)0.06 (0.43)0.289
      TSQM scales,
      Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      Week 4 (after course 1), adjusted mean (SE)
       Effectiveness (AZLI: n=131; TNS: n=126)69.5 (1.9)56.9 (2.0)<0.001
       Side-effects (AZLI: n=130; TNS: n=126)91.6 (1.6)89.2 (1.7)0.204
       Convenience (AZLI: n=129; TNS: n=125)72.2 (2.1)67.2 (2.1)0.041
       Global satisfaction (AZLI: n=130; TNS: n=126)68.9 (2.1)61.8 (2.2)0.004
      TSQM scales,
      Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      Week 20 (after course 3), adjusted mean (SE)
       Effectiveness (AZLI: n=118; TNS: n=110)73.4 (2.3)57.9 (2.5)<0.001
       Side-effects (AZLI: n=118; TNS: n=107)94.3 (1.6)92.5 (1.7)0.328
       Convenience (AZLI: n=118; TNS: n=107)68.0 (2.2)66.1 (2.4)0.460
       Global satisfaction (AZLI: n=118; TNS: n=109)75.8 (4.6)61.7 (4.9)0.010
      CFQ-R = cystic fibrosis questionnaire—revised; CFU = colony forming units; CI = confidence interval; FEV1 = forced expiratory volume in 1 s; IV = intravenous; NE = not estimable (median time exceeded study duration); PA = Pseudomonas aeruginosa; TSQM = treatment satisfaction questionnaire for medicine.
      a Kaplan–Meier method; p-value based on log rank test.
      b Analyzed with ANCOVA models, with terms for treatment, baseline FEV1% predicted, and previous TNS use. CFQ-R respiratory symptoms, PA density, and weight models included the corresponding values at baseline (Week 0).
      c p-Value is for comparison of the number of events for each patient adjusted for time on study through Week 24 based on a negative binomial regression model.
      d p-Value based on Fisher's exact test.
      e p-Value based on Wilcoxon rank sum test.
      Fewer AZLI‐treated patients received additional IV and/or inhaled antipseudomonal antibiotics for respiratory events (AZLI: 38% vs. TNS: 58%; p=0.002; Table 2). Significantly fewer respiratory events requiring IV and/or additional inhaled antipseudomonal antibiotics (AZLI: 84, TNS: 121; p=0.004) and fewer respiratory hospitalizations (AZLI: 40, TNS: 58; p=0.044) occurred in AZLI-treated patients.
      Using Kaplan–Meier methods, median time-to-need for IV antipseudomonal antibiotics for respiratory events was significantly longer for AZLI‐treated than for TNS‐treated patients (p=0.003; Table 2; Fig. 3).
      Figure thumbnail gr3
      Fig. 3Kaplan–Meier estimates for the time to need for IV antipseudomonal antibiotics for a respiratory event. An independent, blinded committee reviewed IV antipseudomonal antibiotic use in the active-comparator period and identified those that were respiratory events. Patients without events were considered as right‐censored; time to censoring was the number of days from baseline (Day 0) to the date of completion of the active-comparator period or early withdrawal. Time to need was the number of days from baseline (Day 0) to the date of need. The log‐rank test was used to compare AZLI and TNS among all ITT patients.
      To account for the confounding effect of additional antibiotic use on lung function, a post-hoc MMRM analysis of relative change from baseline FEV1% predicted was performed, censoring FEV1% predicted values for individual patients subsequent to their receiving a course of IV/inhaled antipseudomonal antibiotics (Fig. 2C). Compared with the primary analysis, FEV1% predicted responses were lower in TNS-treated patients at the end of each treatment cycle, and the magnitude of the treatment differences (AZLI–TNS) were better maintained over the 3 treatment courses.

      3.4 Efficacy — extension period

      Extension period patients (n=133; AZLI/AZLI: 68; TNS/AZLI: 65), demonstrated improvements in FEV1% predicted during each AZLI treatment cycle (Fig. 2D). For AZLI/AZLI-treated patients, active-comparator period treatment responses were sustained across the extension period. TNS-treated patients had minimal improvements in FEV1% predicted during the active-comparator period, but after switching to AZLI in the extension period, their lung function improvements were comparable to the AZLI/AZLI group.
      Increases in body weight were observed for AZLI treated patients (Online Fig. 3). AZLI/AZLI-treated patients gained weight across the active-comparator period (mean relative change from study baseline: 0.55%, 0.86%, and 0.94%, after courses 1, 2 and 3) and the 3 extension-period courses (1.30%, 1.76%, and 1.93%, respectively). TNS/AZLI-treated patients lost weight during the 3 TNS treatment courses (mean relative change from study baseline: −0.83%, −0.82%, and −0.20%, respectively) and had increases in weight after switching to AZLI for the 3 extension-period courses (0.64%, 1.43%, and 1.43%, respectively).

      3.5 Safety

      Adverse event incidences in the active-comparator period were comparable between treatment groups, with cough and productive cough most commonly reported (Table 3). Severe adverse events were experienced by 22 AZLI-treated (16.2%) and 11 TNS-treated (8.3%) patients (p=0.063; Fisher's exact test), most commonly cough (AZLI: n=7/22; TNS: n=4/11) and productive cough (AZLI: n=4/22; TNS: n=5/11). Adverse events considered by investigators as treatment‐related were reported for 22.8% of AZLI-treated (n=31/136) and 12.9% of TNS‐treated (n=17/132) patients (p=0.039), most commonly respiratory incidents (AZLI: n=19/31; TNS: n=14/17).
      Table 3Adverse events occurring during the active-comparator period for ≥10% patients in either treatment group.
      Incidence of adverse event, n (%)
      Adverse event
      Adverse events are coded with Medical Dictionary for Regulatory Activities (MedDRA) Version 11.1. Multiple occurrences per patient were counted once.
      AZLI-treated n=136TNS-treated n=132
      Cough96 (70.6)104 (78.8)
      Productive cough70 (51.5)79 (59.8)
      Pyrexia43 (31.6)40 (30.3)
      Oropharyngeal pain36 (26.5)37 (28.0)
      Dyspnea32 (23.5)36 (27.3)
      Hemoptysis31 (22.8)21 (15.9)
      Rales30 (22.1)35 (26.5)
      Headache29 (21.3 )27 (20.5)
      Nasal congestion29 (21.3)26 (19.7)
      Rhinorrhea25 (18.4 )33 (25.0)
      Exercise tolerance decreased25 (18.4)27 (20.5)
      Fatigue24 (17.6)25 (18.9)
      Decreased appetite19 (14.0)19 (14.4)
      Abdominal pain18 (13.2)8 (6.1)
      Respiratory tract congestion16 (11.8)19 (14.4)
      Wheezing16 (11.8)20 (15.2)
      Chest discomfort14 (10.3)13 (9.8)
      Nausea14 (10.3)10 (7.6)
      Vomiting14 (10.3)14 (10.6)
      Pulmonary function test decreased11 (8.1)17 (12.9)
      Breath sounds abnormal8 (5.9)15 (11.4)
      a Adverse events are coded with Medical Dictionary for Regulatory Activities (MedDRA) Version 11.1. Multiple occurrences per patient were counted once.
      Fifteen patients discontinued treatment due to adverse events (AZLI: 9; TNS: 1) or unspecified safety and/or tolerability reasons (TNS: 5), most commonly productive cough (AZLI: 3) and hemoptysis (AZLI: 3). Fourteen of these 15 patients subsequently discontinued the study for safety or tolerability reasons (AZLI: 3; TNS: 5), consent withdrawn (AZLI: 3, TNS: 1), or investigator decision (AZLI: 2; Fig. 1).
      As was observed for the active-comparator period, cough (n=92/133; 69.2%) and productive cough (n=62/133; 46.2%) were the most commonly reported adverse events in the extension period. Severe adverse events were experienced by 15/133 (11.3%) patients, most commonly cough (n=7/133), productive cough (n=5/133), and pyrexia (n=5/133). One life-threatening event was reported (lung infiltration unrelated to treatment). Two patients died due to complications of CF disease; both deaths were considered unrelated to treatment.
      Adverse events considered by investigators as treatment‐related were reported for 6.8% of patients (n=9/133), most commonly respiratory incidents (n=6/133).

      3.6 Microbiology

      In the active-comparator period, no changes (≥4-fold) were observed in MIC50 values of any antibiotics for all PA isolates in both treatment groups and the proportion of AZLI-treated patients with PA isolates with an aztreonam MIC>8 μg/mL (parenteral breakpoint [
      • Clinical and Laboratory Standards Institute
      Performance standards for antimicrobial susceptibility testing; twentieth informational supplement.
      ]) increased from 33.9% to 49.1%. The percentage of patients with multi‐drug resistant PA was comparable at baseline and Week 24. No clinically concerning changes were observed in PA susceptibility to other aminoglycoside, quinolone, or beta‐lactam antibiotics. The prevalence of other respiratory pathogens remained similar for both treatment groups.
      During the extension period, AZLI/AZLI patients had no changes (≥4-fold) observed in MIC50 values of any antibiotics for all PA isolates, except for 3 intermittent increases in aztreonam and 1 intermittent increase in ticarcillin/clavulanate. TNS/AZLI-treated patients had no changes (≥4‐fold) during either period in MIC50 values of any antibiotics for all PA isolates.
      The proportion of AZLI/AZLI-treated patients with PA isolates with an aztreonam MIC>8 μg/mL increased from 35.2% to 50.0% during the study. The proportion of TNS/AZLI‐treated patients with PA isolates with a tobramycin MIC>4 μg/mL increased from 37.3% to 46.0% during the active-comparator phase, while the proportion with PA isolates with an aztreonam MIC>8 μg/mL increased from 38.3% to 49.0% during the extension phase. The percentage of patients with multi‐drug resistant PA was comparable at study baseline and study end. The prevalence of other respiratory pathogens remained similar for both treatment groups. Burkholderia cepacia complex was not isolated during the study.

      4. Discussion

      Comparative efficacy trials can demonstrate the value of newly developed therapies compared with existing therapies. For CF patients, inhaled antibiotics comparisons can be assessed by lung function, measurements of pulmonary exacerbations, respiratory symptoms, surrogate markers of health (e.g., weight gain/loss), adverse events, and changes in microbial antibiotic sensitivity. Crossover designs, such as the open-label extension in this study, can also show incremental benefits of a new therapy.
      This study demonstrated superiority of AZLI to TNS in lung function improvement after 28 days and superiority across 3 treatment courses. Compared with TNS, AZLI also reduced the total number of pulmonary exacerbations, delayed time to need for additional antibiotic therapies, improved respiratory symptoms, and increased weight in a contemporary CF population that was considered stable at baseline. Importantly, patients with prior TNS experience responded to AZLI treatment during the active-comparator portion of the study, and patients treated with TNS in the active-comparator portion of the study showed incremental improvement in the cross-over period, achieving the same benefit in lung function responses as patients initially randomized to AZLI.
      Sputum PA density decreased equally for TNS and AZLI groups in the current study, suggesting a similar antibacterial effect for the 2 treatment groups. Given confounding factors (e.g., patient ability to produce sputum throughout a clinical trial; regional intra-pulmonary variability of bacterial density) these data must be interpreted with caution, in light of the marked differences in clinical benefit favoring AZLI treatment.
      AZLI was well tolerated with an adverse-event profile consistent with previous clinical trials. Drug‐related adverse events were reported for significantly more AZLI‐treated than TNS-treated patients. In this unblinded study, investigators may have attributed more adverse events to a less familiar antibiotic (AZLI) than to a drug used for over a decade (TNS). Also, patients with a history of intolerance to TNS were excluded from study enrollment. Concerning changes in PA susceptibilities were not observed for either treatment group.
      The AZLI–TNS treatment effect for FEV1% predicted at Day 28 in this study (+7.80%) was comparable to the treatment effects observed in previous phase 3 AZLI‐placebo studies (+10.2%; +6.6%) [
      • McCoy K.S.
      • Quittner A.L.
      • Oermann C.M.
      • Gibson R.L.
      • Retsch-Bogart G.Z.
      • Montgomery A.B.
      Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis.
      ,
      • Retsch-Bogart G.Z.
      • Quittner A.L.
      • Gibson R.L.
      • Oermann C.M.
      • McCoy K.S.
      • Montgomery A.B.
      • et al.
      Efficacy and safety of inhaled aztreonam lysine for airway Pseudomonas in cystic fibrosis.
      ]. The 0.55% improvement in FEV1% predicted observed at Day 28 of TNS treatment in this study was much smaller than the 9.7% TNS‐placebo treatment effect observed at Day 28 in early TNS studies, but is comparable to the effect seen in a more recent study with TNS-experienced patients [
      • Ramsey B.W.
      • Pepe M.S.
      • Quan J.M.
      • Otto K.L.
      • Montgomery A.B.
      • Williams-Warren J
      • et al.
      Cystic fibrosis inhaled tobramycin study group. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis.
      ,
      • McCoy K.S.
      • Quittner A.L.
      • Oermann C.M.
      • Gibson R.L.
      • Retsch-Bogart G.Z.
      • Montgomery A.B.
      Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis.
      ].
      On average, decreases in FEV1 of 1 to 4% predicted per year are observed for CF patients [
      • Konstan M.W.
      • Morgan W.J.
      • Butler S.M.
      • Pasta D.J.
      • Craib M.L.
      • Silva S.J.
      • et al.
      Risk factors for rate of decline in forced expiratory volume in one second in children and adolescents with cystic fibrosis.
      ,
      • Sanders D.B.
      • Bittner R.C.
      • Rosenfeld M.
      • Redding G.J.
      • Goss C.H.
      Pulmonary exacerbations are associated with subsequent FEV1 decline in both adults and children with cystic fibrosis.
      ,
      • Cystic Fibrosis Foundation. Patient Registry
      Annual data report to the center directors.
      ]. Pulmonary exacerbations are associated with step-wise loss of lung function and deleterious effects on quality of life. These events also incur significant costs, whether treated in-hospital or with home IV-antibiotics [
      • Montgomery A.B.
      • Lewis S.
      • Higuchi K.
      • Marshall B.
      • Oermann C.
      Hospitalization risk of current standard of care (SOC) vs. aztreonam for inhalation solution (AZLI) in patients with cystic fibrosis (CF) [abstract 806].
      ]. AZLI‐treated patients experienced a prolonged time-to-need for IV antipseudomonal antibiotics for a respiratory event compared with TNS-treated patients, and had significantly fewer respiratory hospitalizations and respiratory events requiring IV and/or additional inhaled antipseudomonal antibiotics. Patients receiving additional antipseudomonal antibiotics for exacerbations were allowed to continue in the study; these additional treatments likely supplemented their FEV1 responses over the course of the trial. While FEV1 responses at the end of each off-treatment cycle were similar for both treatment groups, significantly more IV/inhaled antibiotics were used for TNS‐treated patients in order to maintain lung function; 38% of AZLI‐treated patients vs. 58% of TNS-treated patients received additional therapies during the active‐comparator portion of the study. After adjusting for the use of additional antipseudomonal antibiotics, the AZLI–TNS treatment differences were better maintained across 3 treatment cycles. Although FEV1 was below baseline for both groups at Week 24, the AZLI treatment effects in each of the extension phase courses were comparable in magnitude to those observed for the first 3 cycles. Similarly, favorable treatment responses to repeated AZLI courses were observed during a prior 18‐month study [
      • Oermann C.M.
      • Retsch-Bogart G.Z.
      • Quittner A.L.
      • Gibson R.L.
      • McCoy K.S.
      • Montgomery A.B.
      • et al.
      An 18-month study, AIR-CF3, of the safety and improvement in pulmonary function and respiratory symptoms with repeated courses of aztreonam for inhalation solution in patients with cystic fibrosis and airway Pseudomonas aeruginosa.
      ]. Additional evidence of improved patient well-being associated with AZLI treatment was the increase above baseline values for body weight observed during AZLI treatment compared with the decrease during TNS treatment.
      The open-label design is a limitation of this study; differences in taste, administration frequency, and approved nebulizers did not allow blinding the study. A double‐dummy design would impose an excessive treatment burden on study participants. In this open-label study, AZLI‐treated patients with TNS experience may have been more likely to report improvements in respiratory symptoms or treatment satisfaction with AZLI than with TNS. A further limitation is that only 4.9% of the patients enrolled in this study were 6 to 12 years of age (n=13/268). This reflects the contemporary CF population; children with CF are generally not chronically infected with PA and they do not tend to have diminished lung function. Results from an ongoing clinical trial assessing the safety of AZLI in children with CF and chronic PA infection will provide more data for the pediatric population (Clinicaltrials.gov: NCT01404234).
      Another perceived limitation is the inclusion of TNS-experienced patients. Since TNS is standard of care for CF patients with chronic PA infection, enrolling only TNS-naïve patients would not be feasible. Further, in a comparative efficacy study, it is necessary to enroll the group of patients receiving existing standard of care therapy, to allow generalization of the results to the overall patient population. Although treatment paradigms vary by both country and center, several lines of evidence suggest that the population in this study was representative of the general CF population with chronic PA infection. During 2010, 68.3% patients with chronic PA infection in the US registry received inhaled tobramycin [
      • Cystic Fibrosis Foundation. Patient Registry
      Annual data report to the center directors.
      ]. Further, 82% of patients in a recent international open‐label, prospective, randomized trial had received inhaled tobramycin prior to screening [
      • Konstan M.W.
      • Flume P.A.
      • Kappler M.
      • Chiron R.
      • Higgins M.
      • Brockhaus F.
      • et al.
      Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: the EAGER trial.
      ].
      In the current study, AZLI (75 mg; 3 times a day) and TNS (300 mg; 2 times a day) doses were used in accordance with the respective EMA and FDA approved labels. Dosing regimens have been established during the development of each drug on the basis of efficacy results in clinical trials [
      • US prescribing information
      TOBI®, tobramycin inhalation solution, USP.
      ,
      • Summary of Product Characteristics (SPC)
      ,
      • Prescribing Information U.S.
      ,
      • Summary of Product Characteristics (SPC)
      ]. The difference in the approved dosing frequencies, in part, reflects the difference in the mechanisms of action for these 2 antibiotics. The antimicrobial activity of aztreonam is time dependent, relating to the elapsed time above a specific drug concentration in the airway. The activity of tobramycin is concentration dependent, thus its antibiotic effects will depend on the peak drug concentration achieved in the airway. The availability of 2 inhaled antibiotics approved for use in CF patients with chronic PA infection now provides the opportunity to prospectively study the benefit of a continuous alternating therapy regimen of 2 antibiotics compared to the standard of care alternate-month monotherapy. Eliminating off-treatment periods has the potential to better preserve lung function, control respiratory symptoms, and reduce the rates of acute pulmonary exacerbations and hospitalizations.
      In conclusion, AZLI demonstrated statistical superiority to TNS over the first comparative treatment course, and maintained superiority to TNS across 3 treatment courses in a patient cohort that was representative of the current CF population receiving standard of care treatment for chronic PA infection. AZLI-treated patients had fewer respiratory hospitalizations than TNS‐treated patients and less frequently used IV or additional inhaled antipseudomonal antibiotics for respiratory events. This study demonstrates that AZLI provides an effective and well tolerated treatment option for CF patients with chronic airway PA infection.

      5. Study investigators

      In addition to the authors, the other members of the AZLI Active Comparator Study Team were:
      Austria:
      Helmuth Ellemunter and Isidor Huttegger
      Belgium:
      Frans de Baets, Kristin Desager, Lieven DuPont and Anne Malfroot
      France:
      Marc Albertini, Chantal Belleguic, Christophe Delacourt, Philippe Domblides, Jean Francois Duhamel, Marcel Guillot, Gerard Lenoir, Sylvie Leroy, Jean Claude Pautard and Natascha Remus
      Germany:
      Thomas Biedermann, Eckard Hamelmann, Hans-Eberhardt Heuer, Uwe Mellies, Jens Schreiber, Doris Staab, Jens-Oliver Steiss, Helmut Teschler and Hubert Wirtz
      Ireland:
      Gerard Canny, Charles Gallagher, Peter Greally and Mary Herzig
      Italy:
      Sergio Bernasconi, Carla Colombo, Rolando Gagliardini, Vincenzina Lucidi, Francesca Pardo, Giovanna Pisi, Serena Quattrucci and Valeria Raia
      Netherlands:
      Harry Heijerman and Johannes Hendriks
      Portugal:
      Adelina Amorim and Celeste Barreto
      Spain:
      Carmen Antelo, Jose Villa Asensi, Gloria Garcia Hernandez, Luis Maiz and Francisco Peres‐Frias
      Switzerland:
      Annette Boehler and Alexander Moeller
      United Kingdom:
      Mary Carroll, Steven Conway, Stuart Elborn, Charles Haworth, Ian Ketchell, Christopher Taylor and Martin Walshaw
      United States:
      Frank Accurso, Steven Boas, Aaron Chidekel, Rubin Cohen, Paul Comber, Cori Daines, Robert Fink, David Geller, Gavin Graff, Denis Hadjiliadis, Jeffrey Hoag, Theodore Liou, Craig Nakamura, Edward Naureckas, David Nichols, Bruce Nickerson, Peggy Radford, Santiago Reyes, Dion Roberts, Mark Rolfe, Howard Schmidt, Pedro Sepulveda, Terry Spencer, Arvey Stone, Bruce Trapnell, Pierre Vauthy and Isabel Virella‐Lowell
      Sub-investigators for this study included:
      Austria:
      Johannes Eder, Watraud Eder, Katalin Kovacs and Udo Langenhorst
      Belgium:
      Elke Dewachter, Valerie Dufresne, Michel Dumonceaux, Monique Lequesne, Alain Michils, Pascal Van Bleyenbergh and Sabine Van Daele
      Denmark:
      Marianne Skov
      France:
      Laurence Bassinet, Michele Berlioz, Graziella Brinchault, Francoise Ceccato, Frederique Chedevergne, Francois Counil, Benoit Desrues, Severine Gaillot, Isabelle Halphen, Vincent Boisserie Lacroix, Muriel Laurans, Stephanie Mechain-Bui, Lahouari Meziane, Thierry Perez, Alice Picard, Cinthia Rames, Bernard Romeo, Isabelle Sermet, Stephanie Vrielynck and Benoît Wallaert
      Germany:
      Cordula Conrad-Kabbe, Norbert Grammann, Jorg Grobe-Onnebrink, Andrea Guttler, Andrea Jobst, Swante Klahr, Cordula Koerner-Rettberg, Sabine Matena, Susanne Nahrig, Hans Christoph Runge, Carsten Schwarz, Wolfgang Sextro, Albrecht Tacke, Kathrin Christina Weber, Diane Wieczorek and Monika Wolf
      Ireland:
      Peter Barry, Sanjay Chotirmall, Basil Elnazir, Cedric Gunaratnam, Edina Moylett and Trevor Nicolson
      Italy:
      Federico Alghisi, Giuseppe Cimino, Natalia Cirilli, Marco Cipolli, Laura Claut, Diana Costantini, Annarita Costantino, Luisa De Cristofaro, Fabiola De Gregorio, Violetta Di Pierantonio, Valeria Di Stefano, Nadia Faelli, Francesco Longo, Lucia Spicuzza, Gabriella Traverso, Maria Candida Tripodi, Patrizia Troiani and Giovanna Vitaliti
      Netherlands:
      Geert Jan Wesseling
      Portugal:
      Pilar Azeved, Joao Bento, José Cavaco and Luisa Pereira
      Spain:
      Maria Isabel Gonzalez Alvarez, Pilar Caro Auvilena, Isabel Barrio, Maria Castro Codesal, Lucrecia Suarez Cortina, Marta Ruiz de Valbuena, Adelaida Lamas Ferreiro, Adolfo Sequeiros Gonzalez, Carmen Martinez, Maria Teresa Martinez, Estela Perez-Ruiz, Antonio Salcedo Posadas and Elena Urgelles
      Switzerland:
      Klaus Haemmarle, Markus Hoefer, Demet Inci and Renate Spinas
      United Kingdom:
      Ina Aldag, Helen Barker, Gary Connett, Jane Davies, Christine Etherington, Andres Floto, Ben Green, Khin Ma Gyi, Mark Ledson, Anand Palamarthy, Felicity Perrin, Jacqueline Rendall and Dennis Wat
      United States:
      James Acton, Gabriel Aljadeff, Lisa Allwein, Raouf Amin, Bruce Barnett, Sandy Bartosi, Michael Biggin, Debra Boyer, Mark Brown, Terry Byars, Holly Carveth, Eduardo Cepeda-Davila, Barbara Chatfield, Michael Daines, Ruth DeVoogd, Karim Djekidel, Carolyn Donovan, Henry Dorkin, Lorrie Duan, Dawn Ericson, Monica Federico, Peter Fornos, Fadel Fuiz, James Goodwin, Gerald Gong, Roni Grad, Tarik Haddad, Anas Hadeh, Robert Heinle, Laura Herpel, Peter Hiatt, David Hicks, Douglas Kyle Hogarth, Douglas Holsclaw, Angelica Honsberg, Ashley Jones, Marion Jones, Diane Kitch, Lucille Lester, Susan Lester, Jeffrey Lewis, Floyd Livingston, Thomas Martin, Fernando Martinez, Martin Martinez, Margo Moore, Gary Mueller, Angala Narasimhan, Imre Noth, Adrian O'Hagan, Gary Onady, Raj Padman, Reynold Panettieri, Kathleen Peeke, Terri Phillips, Diane Rhodes, Jonathan Rosen, Frederick Royce, Milene Saavedra, Scott Sagel, Connie St. Clair, Gregory Sawicki, Scott Schroeder, Edward S. Schulman, Michael Schwartz, Amrapali Shah, Michael Sherman, Thomas C. Smith, Patrick Sobande, James Stark, Antine Stenbit, Mary Strek, Alumet Uluer, Robert Vender, William Walsh, Ronald Williams, Elizabeth Woods, James Woodward, Jamie Wooldridge, Nozomi Yagi, Robert Young and Anchalee Yuengsrigul

      Conflict of interest statement

      DB had consultancies with Gilead Sciences, Insmed and Aradigm, and received a research grant from Novartis. MF had a consultancy with Gilead Sciences. RF had consultancies with Gilead Sciences, MPEX, Gruenenthal, and Novartis, and participated in speaker's bureaus for Gilead Sciences, Astra Zeneca, Astellas, and Roche. ML participated in speaker's bureaus for Chiesi Farmaceutici and Mediolanum. CMO had a consultancy with Gilead Sciences and received a research grant from Gilead Sciences. SAL, MB, and ABM are employees and/or shareholders of Gilead Sciences. All other authors have no perceived conflicts of interests.

      Role of the Funding Source

      This study was sponsored by Gilead Sciences.
      BMA, TP, MB, and ABM participated in study design. BMA, TP, DB, MF, RF, RC, ML, CK, NM, and CMO were clinical investigators for the study. SAL oversaw statistical analyses. SAL, MB, ABM, and CMO wrote and edited the draft manuscript. All authors revised the manuscript and approved the final version for submission. The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication.

      Acknowledgments

      We thank the patients and their families. Study conduct was managed by Laura Haas and Sheila Leitzinger (Gilead Sciences, Inc.). Statistical analyses were performed by Lixin Shao, (Gilead Sciences, Inc.). Writing assistance was provided by Kate Loughney, under the sponsorship of Gilead Sciences. Independent Data Review Committee for Respiratory Events: Marcia Katz (Baylor University, Houston TX) and Patrick Flume (Medical University of South Carolina, Charleston SC). Cystic Fibrosis Foundation Therapeutics Data and Safety Monitoring Board: Lynne M Quittell (Children's Hospital of New York; Columbia University, New York NY), Richard A Kronmal (University of Washington, Seattle WA), Natalie Neu (Columbia University, New York, NY), and Margaret (Lou) Guill (Dartmouth–Hitchcock Medical Center, Lebanon NH).

      Appendix A. Supplementary data

      • Online Fig. 2

        Relative change from baseline FEV1% predicted at Day 28 for patient subgroups. The adjusted means are from an ANCOVA model that included terms for treatment, baseline FEV1% predicted, and previous inhaled tobramycin use. Missing data were imputed with the last observation carried forward method. The US patient subgroup did not include any patients with inhaled tobramycin use <84 days in the previous year. At the bottom of the figure, the SE bars for the TNS and AZLI data sets overlap for patients <18 years of age and for patients with prior inhaled tobramycin use <84 days. Moderate disease severity=FEV1>50% to ≤75% predicted at baseline and severe disease severity=FEV150% predicted at baseline.

      • Online Fig. 3

        Adjusted mean relative change in body weight (%) from study baseline by study visit for patients participating in the AZLI open-label extension period. Significant differences between the AZLI and TNS treatment groups were observed at Week 4 (p=0.004), Week 12 (p=0.003), and Week 16 (p=0.009).

      References

        • Flume P.A.
        • O'Sullivan B.P.
        • Robinson K.A.
        • Goss C.H.
        • Mogayzel Jr., P.J.
        • Willey-Courand D.B.
        • et al.
        Cystic fibrosis pulmonary guidelines. Chronic medications for maintenance of lung health.
        Am J Respir Crit Care Med. 2007; 176: 957-969
        • Heijermann H.
        • Westerman E.
        • Conway S.
        • Touw D.
        • Döring G.
        • Working Group Consensus
        Inhaled medication and inhalation devices for lung disease in patients with cystic fibrosis: a European consensus.
        J Cyst Fibros. 2009; 8: 295-315
        • US prescribing information
        TOBI®, tobramycin inhalation solution, USP.
        (Available from) (Cited, Feb 29 2012)
        • Summary of Product Characteristics (SPC)
        TOBI® 300 mg/5 mL nebuliser solution.
        (Available from:) (Cited, Feb 29 2012)
        • Summary of Product Characteristics
        Bramitob 300 mg/4 ml nebuliser solution.
        (Available from:) (Cited, Feb 29 2012)
        • Summary of Product Characteristics (SPC)
        Promixin® 1 MIU powder for nebuliser solution.
        (Available from:) (Cited, Feb 29 2012)
        • Prescribing Information U.S.
        Cayston® (aztreonam for inhalation solution).
        (Available from) (Cited, Sept 27 2011)
        • Summary of Product Characteristics (SPC)
        Cayston® 75 mg powder and solvent for nebuliser solution.
        (Available from) (Cited. Feb 29 2012)
        • Ramsey B.W.
        • Pepe M.S.
        • Quan J.M.
        • Otto K.L.
        • Montgomery A.B.
        • Williams-Warren J
        • et al.
        Cystic fibrosis inhaled tobramycin study group. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis.
        N Eng J Med. 1999; 340: 23-30
      1. Prescribing information: AZACTAM® (aztreonam) for injection, USP.
        (Available from) (Cited 29 Feb 2012)
        • McCoy K.S.
        • Quittner A.L.
        • Oermann C.M.
        • Gibson R.L.
        • Retsch-Bogart G.Z.
        • Montgomery A.B.
        Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis.
        Am J Respir Crit Care Med. 2008; 178: 921-928
        • Retsch-Bogart G.Z.
        • Quittner A.L.
        • Gibson R.L.
        • Oermann C.M.
        • McCoy K.S.
        • Montgomery A.B.
        • et al.
        Efficacy and safety of inhaled aztreonam lysine for airway Pseudomonas in cystic fibrosis.
        Chest. 2009; 135: 1223-1232
        • Oermann C.M.
        • Retsch-Bogart G.Z.
        • Quittner A.L.
        • Gibson R.L.
        • McCoy K.S.
        • Montgomery A.B.
        • et al.
        An 18-month study, AIR-CF3, of the safety and improvement in pulmonary function and respiratory symptoms with repeated courses of aztreonam for inhalation solution in patients with cystic fibrosis and airway Pseudomonas aeruginosa.
        Pediatr Pulmonol. 2010; 45: 1121-1134
        • Rosenfeld M.
        • Emerson J.
        • Williams-Warren J.
        • Pepe M.
        • Smith A.
        • Montgomery A.B.
        • et al.
        Defining a pulmonary exacerbation in cystic fibrosis.
        J Pediatr. 2001; 139: 359-365
        • Quittner A.L.
        • Buu A.
        • Messer M.A.
        • Modi A.C.
        • Watrous M.
        Development and validation of the cystic fibrosis questionnaire in the United States: a health related quality of life measure for cystic fibrosis.
        Chest. 2005; 128: 2347-2354
        • Atkinson M.J.
        • Sinha A.
        • Hass S.L.
        • Colman S.S.
        • Kumar R.N.
        • Brod M.
        • et al.
        Validation of a general measure of treatment satisfaction, the treatment satisfaction questionnaire for medication (TSQM), using a national panel study of chronic disease.
        Health Qual Life Outcomes. 2004; 2: 12
        • Siddiqui O.
        • Hung H.M.
        • O'Neill R.
        MMRM vs. LOCF: a comprehensive comparison based on simulation study and 25 NDA datasets.
        J Biopharm Stat. 2009; 19: 227-246
        • Saiman L.
        • Siegel J.
        • the CF Foundation Consensus Conference on Infection Control Participants
        Infection control recommendations for patients with cystic fibrosis: microbiology, important pathogens, and infection control practices to prevent patient-to-patient transmission.
        Infect Control Hosp Epidemiol. 2003; 24: S6-S52
        • Clinical and Laboratory Standards Institute
        Performance standards for antimicrobial susceptibility testing; twentieth informational supplement.
        CLSI, Wayne (PA)2010
        • Konstan M.W.
        • Morgan W.J.
        • Butler S.M.
        • Pasta D.J.
        • Craib M.L.
        • Silva S.J.
        • et al.
        Risk factors for rate of decline in forced expiratory volume in one second in children and adolescents with cystic fibrosis.
        J Pediatr. 2007; 151: 134-139
        • Sanders D.B.
        • Bittner R.C.
        • Rosenfeld M.
        • Redding G.J.
        • Goss C.H.
        Pulmonary exacerbations are associated with subsequent FEV1 decline in both adults and children with cystic fibrosis.
        Pediatr Pulmonol. 2011; 46: 393-400
        • Cystic Fibrosis Foundation. Patient Registry
        Annual data report to the center directors.
        2011
        • Montgomery A.B.
        • Lewis S.
        • Higuchi K.
        • Marshall B.
        • Oermann C.
        Hospitalization risk of current standard of care (SOC) vs. aztreonam for inhalation solution (AZLI) in patients with cystic fibrosis (CF) [abstract 806].
        Am J Respir Crit Care Med. 2009; 179: A1188
        • Konstan M.W.
        • Flume P.A.
        • Kappler M.
        • Chiron R.
        • Higgins M.
        • Brockhaus F.
        • et al.
        Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: the EAGER trial.
        J Cyst Fibros. 2010; 10: 54-61