Phase II studies of nebulised Arikace in CF patients with Pseudomonas aeruginosa infection

J P Clancy, L Dupont, M W Konstan, J Billings, S Fustik, C H Goss, J Lymp, P Minic, A L Quittner, R C Rubenstein, K R Young, L Saiman, J L Burns, J R W Govan, B Ramsey, R Gupta, Arikace Study Group, J Clancy, R Young, R Ahrens, M Aitken, J Billings, A Faro, C Goss, D Layish, N Lechtzin, M Light, S Miller, S Nasr, J Nick, Rc Rubenstein, A Sannuti, G Sawicki, J Taylor-Cousar, B Trapnell, J Wallace, P Minic, S Fustik, E Solyom, H Mazurek, Y Antipkin, A Feketeova, A Senatorova, E Csiszer, V Kostromina, B Takac, R Ujhelyi, A Sovtic, Anne Marie Buccat, Catherine Doherty, J P Clancy, L Dupont, M W Konstan, J Billings, S Fustik, C H Goss, J Lymp, P Minic, A L Quittner, R C Rubenstein, K R Young, L Saiman, J L Burns, J R W Govan, B Ramsey, R Gupta, Arikace Study Group, J Clancy, R Young, R Ahrens, M Aitken, J Billings, A Faro, C Goss, D Layish, N Lechtzin, M Light, S Miller, S Nasr, J Nick, Rc Rubenstein, A Sannuti, G Sawicki, J Taylor-Cousar, B Trapnell, J Wallace, P Minic, S Fustik, E Solyom, H Mazurek, Y Antipkin, A Feketeova, A Senatorova, E Csiszer, V Kostromina, B Takac, R Ujhelyi, A Sovtic, Anne Marie Buccat, Catherine Doherty

Abstract

Rationale: Arikace is a liposomal amikacin preparation for aerosol delivery with potent Pseudomonas aeruginosa killing and prolonged lung deposition.

Objectives: To examine the safety and efficacy of 28 days of once-daily Arikace in cystic fibrosis (CF) patients chronically infected with P aeruginosa.

Methods: 105 subjects were evaluated in double-blind, placebo-controlled studies. Subjects were randomised to once-daily Arikace (70, 140, 280 and 560 mg; n=7, 5, 21 and 36 subjects) or placebo (n=36) for 28 days. Primary outcomes included safety and tolerability. Secondary outcomes included lung function (forced expiratory volume at one second (FEV1)), P aeruginosa density in sputum, and the Cystic Fibrosis Quality of Life Questionnaire-Revised (CFQ-R).

Results: The adverse event profile was similar among Arikace and placebo subjects. The relative change in FEV1 was higher in the 560 mg dose group at day 28 (p=0.033) and at day 56 (28 days post-treatment, 0.093L±0.203 vs -0.032L±0.119; p=0.003) versus placebo. Sputum P aeruginosa density decreased >1 log in the 560 mg group versus placebo (days 14, 28 and 35; p=0.021). The Respiratory Domain of the CFQ-R increased by the Minimal Clinically Important Difference (MCID) in 67% of Arikace subjects (560 mg) versus 36% of placebo (p=0.006), and correlated with FEV1 improvements at days 14, 28 and 42 (p<0.05). An open-label extension (560 mg Arikace) for 28 days followed by 56 days off over six cycles confirmed durable improvements in lung function and sputum P aeruginosa density (n=49).

Conclusions: Once-daily Arikace demonstrated acute tolerability, safety, biologic activity and efficacy in patients with CF with P aeruginosa infection.

Trial registration: ClinicalTrials.gov NCT00558844 NCT00777296.

Keywords: Bacterial Infection; Cystic Fibrosis; Respiratory Infection.

Figures

Figure 1
Figure 1
Patient enrolment across the two Arikace studies. In the European study, 75 patients were screened, and 66 were randomised. In the US study, 56 patients were screened, and 46 were randomised. *Across all randomised subjects, seven subjects withdrew consent prior to dosing. Data shown is for the 105 subjects dosed at least once with Arikace or placebo.
Figure 2
Figure 2
Change in FEV1 (L) from baseline through day 56. Filled squares, solid line=Arikace 560 mg, *p=0.033 at day 28, *p=0.003 at day 56 (compared with placebo). Filled triangles, solid line=Arikace 280 mg, *p=0.009 at day 28 (compared with placebo). Open squares, dashed line=Arikace 140 mg. Open diamonds, dashed line=Arikace 70 mg. Open circles, dashed line=placebo. The values above the abscissa are the number of subjects in each dose cohort providing data at each time point (70 mg/140 mg/280 mg/560 mg/placebo).
Figure 3
Figure 3
Change in sputum density of Pseudomonas aeruginosa (log10 CFU/g) from baseline through day 35. Filled squares, solid line=Arikace 560 mg, *p=0.007 at day 28, *p=0.021 at day 35. Filled triangles, solid line=Arikace 280 mg. Open squares, dashed line=Arikace 140 mg. Open diamonds, dashed line=Arikace 70 mg. Open circles, dashed line=placebo. The values above the abscissa are the number of subjects in each dose cohort providing data at each time point (70 mg/140 mg/280 mg/560 mg/placebo).
Figure 4
Figure 4
Change in FEV1 (% predicted) from baseline through cycle 6 of Arikace. Each cycle consisted of 28 days of once daily Arikace (560 mg) followed by 56 days off study drug. Each shaded box is a treatment cycle. Study days (every 2 weeks) are as shown on the abscissa, with the number of subjects at each time point as noted immediately above the study days. *p<0.0001 for FEV1 at end of treatment following six cycles compared with baseline; **p=0.0001 for FEV1 at 56 days post-treatment following six cycles compared with baseline.
Figure 5
Figure 5
Change in sputum density of Pseudomonas aeruginosa (log10 CFU/g) from baseline through cycle 6 of Arikace. Each set of three bars is the change in sputum P aeruginosa density compared with baseline (day 1 of cycle 1) for days 1 (white), 14 (gray), and 28 (black) of each respective Arikace cycle. *p=0.003 for change in CFU across all of the Arikace treatment cycles relative to baseline (cycle 1, day 1).

References

    1. Rommens JM, Iannuzzi MC, Kerem B, et al. Identification of the cystic fibrosis gene: chromosome walking and jumping. Science (New York, NY) 1989;245:1059–65
    1. Kerem B, Rommens JM, Buchanan JA, et al. Identification of the cystic fibrosis gene: genetic analysis. Science (New York, NY) 1989;245:1073–80
    1. Rowe SM, Miller S, Sorscher EJ. Cystic fibrosis. N Engl J Med 2005;352:1992–2001
    1. Strausbaugh SD, Davis PB. Cystic fibrosis: a review of epidemiology and pathobiology. Clin Chest Med 2007;28:279–88
    1. Li Z, Kosorok MR, Farrell PM, et al. Longitudinal development of mucoid Pseudomonas aeruginosa infection and lung disease progression in children with cystic fibrosis. JAMA 2005;293:581–8
    1. Dasenbrook EC, Merlo CA, Diener-West M, et al. Persistent methicillin-resistant Staphylococcus aureus and rate of FEV1 decline in cystic fibrosis. Am J Respir Crit Care Med 2008;178:814–21
    1. Dasenbrook EC, Checkley W, Merlo CA, et al. Association between respiratory tract methicillin-resistant Staphylococcus aureus and survival in cystic fibrosis. JAMA 2010;303:2386–92
    1. Ramsey BW, Pepe MS, Quan JM, et al. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group. N Engl J Med 1999;340:23–30
    1. Moss RB. Long-term benefits of inhaled tobramycin in adolescent patients with cystic fibrosis. Chest 2002;121:55–63
    1. McCoy KS, Quittner AL, Oermann CM, et al. Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis. Am J Respir Crit Care Med 2008;178:921–8
    1. Oermann CM, Assael B, Nakamura C, et al. Poster: Aztreonam for Inhalation Solution (AZLI) vs. Tobramycin Inhalation Solution (TIS): a 6-month Comparative Trial in Cystic Fibrosis (CF) Patients with Pseudomonas aeruginosa (PA). Pediatr Pulmonol 2010;45(Supplement 33)(abs 305):327
    1. Retsch-Bogart GZ, Quittner AL, Gibson RL, et al. Efficacy and safety of inhaled aztreonam lysine for airway pseudomonas in cystic fibrosis. Chest 2009;135:1223–32
    1. Schuster A, Haliburn C, Doring G, et al. Safety, efficacy and convenience of colistimethate sodium dry powder for inhalation (Colobreathe DPI) in patients with cystic fibrosis: a randomised study. Thorax 2013;68:344–50
    1. Oermann CM, Retsch-Bogart GZ, Quittner AL, et al. An 18-month study of the safety and efficacy of repeated courses of inhaled aztreonam lysine in cystic fibrosis. Pediatr Pulmonol 2010;45:1121–34
    1. Konstan MW, Flume PA, Kappler M, et al. Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: The EAGER trial. J Cyst Fibros 2011;10:54–61
    1. Assael BM, Pressler T, Bilton D, et al. Inhaled aztreonam lysine vs. inhaled tobramycin in cystic fibrosis: A comparative efficacy trial. J Cyst Fibros 2013;12:130–40
    1. Coates AL. Performance of PARI eFlow. J Aerosol Med Pulm Drug Deliv 2010;23:114; author reply -8
    1. Meers P, Neville M, Malinin V, et al. Biofilm penetration, triggered release and in vivo activity of inhaled liposomal amikacin in chronic Pseudomonas aeruginosa lung infections. J Antimicrob Chemother 2008;61:859–68
    1. Li Z, Zhang Y, Wurtz W, et al. Characterization of nebulized liposomal amikacin (Arikace) as a function of droplet size. J Aerosol Med Pulm Drug Deliv 2008;21:245–54
    1. Clancy JP. Clinical Trials of Lipid-Associated Aerosolized Amikacin: the ArikaceTM Story. Pediatr Pulmonol 2009;44:109–212
    1. Weers J, Metzheiser B, Taylor G, et al. A gamma scintigraphy study to investigate lung deposition and clearance of inhaled amikacin-loaded liposomes in healthy male volunteers. J Aerosol Med Pulm Drug Deliv 2009;22:131–8
    1. Chinoy MR, Fisher AB, Shuman H. Confocal imaging of time-dependent internalization and localization of NBD-PC in intact rat lungs. Am J Physiol 1994;266(6 Pt 1):L713–21
    1. Martini WZ, Chinkes DL, Barrow RE, et al. Lung surfactant kinetics in conscious pigs. Am J Physiol 1999;277(1 Pt 1):E187–95
    1. Babiker A, Andersson O, Lindblom D, et al. Elimination of cholesterol as cholestenoic acid in human lung by sterol 27-hydroxylase: evidence that most of this steroid in the circulation is of pulmonary origin. J Lipid Res 1999;40:1417–25
    1. Diczfalusy U, Lund E, Lutjohann D, et al. Novel pathways for elimination of cholesterol by extrahepatic formation of side-chain oxidized oxysterols. Scand J Clin Lab Invest Suppl 1996;226:9–17
    1. Neville M, Liu S, Artis C, et al. Functionality of Foamy Alveolar Macrophages After Inhalation of Aerosolized Liposomal Amikacin (ArikaceTM). Pediatric Pulmonology 2009;44:109–212

Source: PubMed

Подписаться