Scolaris Content Display Scolaris Content Display

Esteroides inhalados y riesgo de neumonía para la enfermedad pulmonar obstructiva crónica

Collapse all Expand all

References

Referencias de los estudios incluidos en esta revisión

Anzueto 2009 {published and unpublished data}

Anzueto A, Ferguson GT, Feldman G, Chinsky K, Seibert A, Emmett A, et al. Effect of fluticasone propionate/salmeterol (250/50) on COPD exacerbations and impact on patient outcomes. Journal of Chronic Obstructive Pulmonary Disease 2009;6(5):320‐9.
Dalal AA, Blanchette CM, Petersen H, St. Charles M. Cost‐effectiveness of fluticasone propionate/salmeterol (250/50mcg) compared to salmeterol (50mcg) in patients with chronic obstructive pulmonary disease: economic evaluation of study SCO100250 [Abstract]. American Thoracic Society International Conference; 2009 May 15‐20; San Diego. 2009:A1502 [Poster K54].
DiSantostefano RL, Li H, Rubin DB, Stempel DA. Which patients with chronic obstructive pulmonary disease benefit from the addition of an inhaled corticosteroid to their bronchodilator? A cluster analysis. BMJ Open 2013;3(4):e001838. [DOI: 10.1136/bmjopen‐2012‐001838]
GlaxoSmithKline. A Randomized, Double‐Blind, Parallel‐Group, 52‐Week Study to Compare the Effect of Fluticasone Propionate/Salmeterol DISKUS 250/50mcg BID with Salmeterol DISKUS 50mcg BID on the Annual Rate of Moderate/Severe Exacerbations in Subjects with Chronic Obstructive Pulmonary Disease (COPD). SCO100250. www.gsk‐clinicalstudyregister.com/ (accessed 25 May 2013).
GlaxoSmithKline. A randomized, double‐blind, parallel‐group, 52‐week study to compare the effect of fluticasone propionate/salmeterol DISKUS 250/50mcg bid with salmeterol DISKUS 50mcg bid on the annual rate of moderate/severe exacerbations in subjects with chronic obstructive pulmonary disease (COPD). www.gsk‐clinicalstudyregister.com/ (accessed 8 April 2013).

Bourbeau 1998 {published data only}

Bourbeau J, Rouleau MY, Boucher S. Randomised controlled trial of inhaled corticosteroids in patients with chronic obstructive pulmonary disease. Thorax 1998;53(6):477‐82.

Bourbeau 2007 {published data only}

Bourbeau J, Christodoulopoulos P, Maltais F, Yamauchi Y, Olivenstein R, Hamid Q. Effect of salmeterol/fluticasone propionate on airway inflammation in COPD: a randomised controlled trial. Thorax 2007;62(11):938‐43.

Burge 2000 {published and unpublished data}

Bale G, Martinez‐Camblor P, Burge PS, Soriano JB. Long‐term mortality follow‐up of the ISOLDE participants: causes of death during 13 years after trial completion. Respiratory Medicine 2008;102(10):1468‐72.
Bale GA, Burge PS, Burge C, Moore VC. The ISOLDE Study 11‐13 years on: what were the causes of death? [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:Poster H42.
Briggs AH, Lozano‐Ortega G, Spencer S, Bale G, Spencer MD, Burge PS. Estimating the cost‐effectiveness of fluticasone propionate for treating chronic obstructive pulmonary disease in the presence of missing data. Value In Health 2006;9(4):227‐35.
Burge PS, Calverley PM, Jones PW, Spencer S, Anderson JA. Prednisolone response in patients with chronic obstructive pulmonary disease: results from the ISOLDE study. Thorax 2003;58(8):654‐8.
Burge PS, Calverley PM, Jones PW, Spencer S, Anderson JA, Maslen TK. Randomised, double blind, placebo controlled study of fluticasone propionate in patients with moderate to severe chronic obstructive pulmonary disease: the ISOLDE trial. British Medical Journal 2000;320(7245):1297‐303.
Burge S, Sondhi S, Williams MK, Wilson K, Efthimiou J. An economic evaluation of fluticasone propionate in patients with moderate to severe chronic obstructive pulmonary disease (COPD) from the isolde trial. American Journal of Respiratory and Critical Care Medicine 2000;161 Suppl 3:A491.
Calverley PM, Burge PS, Spencer S, Anderson JA, Jones PW. Bronchodilator reversibility testing in chronic obstructive pulmonary disease. Thorax 2003;58(8):659‐64.
Calverley PM, Spencer S, Willits L, Burge PS, Jones PW. Withdrawal from treatment as an outcome in the ISOLDE study of COPD. Chest 2003;124(4):1350.
D'Urzo AD, Calverley PMA. Withdrawal of treatment in the ISOLDE study [letter]. Chest 2004;125(6):2368.
GlaxoSmithKline F. A multi‐centre, double‐blind, placebo‐controlled, parallel group study of the efficacy and tolerability of long‐term inhaled fluticasone propionate 500 mcg twice daily via a Volumatic Spacer device in patients with non‐asthmatic chronic obstructive pulmonary disease, including an acute oral corticosteroid trial. FLTB3054. www.gsk‐clinicalstudyregister.com/ (accessed 20 May 2013).
Jarad NA, Wedzicha JA, Burge PS, Calverley PM. An observational study of inhaled corticosteroid withdrawal in stable chronic obstructive pulmonary disease. ISOLDE Study Group. Respiratory Medicine 1999;93(3):161‐6.
Jebrak G. Corticosteroid therapy and chronic obstructive broncho‐pneumopathies: the ISOLDE study. Presse Medicale 2000;29(16):1461‐2.
Jones PW, Willits LR, Burge PS, Calverley PMA. Disease severity and the effect of fluticasone propionate on chronic obstructive pulmonary disease exacerbations. European Respiratory Journal 2003;21(1):68‐73.
Marchand E. Withdrawal from and study design of the ISOLDE trial [comment]. Chest 2004;126(0012‐3692):659.
Spencer S, Calverley PM, Burge PS, Jones PW. Health status deterioration in patients with chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2001;163(1):122‐8.
Spencer S, Calverley PM, Burge PS, Jones PW. Impact of preventing exacerbations on deterioration of health status in COPD. European Respiratory Journal 2004;23(5):698‐702.

Calverley 2003b {published and unpublished data}

Borgstrom L, Asking L, Olsson H, Peterson S. Lack of interaction between disease severity and therapeutic response with budesonide/formoterol in a single inhaler [Abstract]. American Thoracic Society 100th International Conference; 2004 May 21‐26; Orlando. 2004:C22 Poster 505.
Calverley PM, Bonsawat W, Cseke Z, Zhong N, Peterson S, Olsson H. Maintenance therapy with budesonide and formoterol in chronic obstructive pulmonary disease. European Respiratory Journal 2003;22(6):912‐9.
Calverley PMA. Effect of budesonide/formoterol on severe exacerbations and lung function in moderate to severe COPD. Thorax 2002;57 Suppl III:iii44.
Calverley PMA, Cseke Z, Peterson S. Budesonide/formoterol reduces the use of oral corticosteroids in the treatment of COPD [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P436.
Calverley PMA, Kuna P, Olsson H. COPD exacerbations are reduced by budesonide/formoterol in a single inhaler [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P1587.
Calverley PMA, Olsson H, Symbicort International COPD Study Group. Budesonide/formoterol in a single inhaler sustains improvements in lung function over 12 months compared with mono‐components and placebo in patients with COPD [abstract]. American Thoracic Society 99th International Conference; 2003 May 16‐21; Seattle. 2003:B024 Poster 418.
Calverley PMA, Peterson S. Combining budesonide/formoterol in a single inhaler reduces exacerbation frequency in COPD [abstract]. American Thoracic Society 99th International Conference; 2003 May 16‐21; Seattle. 2003:D092 Poster 211.
Calverley PMA, Stahl E, Jones PW. Budesonide/formoterol improves the general health status of patients with COPD [Abstract]. American Thoracic Society International Conference; 2005 May 20‐25; San Diego. 2005:B93 Poster 303.
Calverley PMA, Szafranski W, Andersson A. Budesonide/formoterol is a well‐tolerated long term maintenance therapy for COPD. European Respiratory Journal 2005;26 Suppl 49:Poster 1917.
Calverley PMA, Thompson NC, Olsson H. Budesonide/formoterol in a single inhaler sustains lung function improvements in COPD [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P435.
Halpin D, Ståhl E, Lundback B, Anderson F, Peterson S. Treatment costs and number needed to treat (NNT) with budesonide/formoterol to avoid one exacerbation of COPD [Abstract]. American Thoracic Society 100th International Conference; 2004 May 21‐26; Orlando. 2004:D22 Poster 525.
Halpin DMG, Larsson T, Calverley PMA. How many patients with COPD must be treated with budesonide/formoterol compared with formoterol alone to avoid 1 day of oral steroid use? [Abstract]. American Thoracic Society International Conference; 2005 May 20‐25; San Diego. 2005:B93 Poster 314.
Jones PW, Ståhl E. Budesonide/formoterol in a single inhaler improves health status in patients with COPD [abstract]. American Thoracic Society 99th International Conference; 2003 May 16‐21; Seattle. 2003:B024 Poster 419.
Jones PW, Ståhl E. Budesonide/formoterol sustains clinically relevant improvements in health status in COPD [Abstract]. European Respiratory Journal 2005;26(Suppl 49):Abstract No. 1352.
Jones PW, Ståhl E. Reducing exacerbations leads to a better health‐related quality of life in patients with COPD. European Respiratory Society 13th Annual Congress; 2003 Sep 28‐29; Vienna. 2003:P1586.
Lofdahl CG. Reducing the impact of COPD exacerbations: clinical efficacy of budesonide/formoterol. European Respiratory Review 2004;13(88):14‐21.
Lofdahl CG, Andreasson E, Svensson K, Ericsson A. Budesonide/formoterol in a single inhaler improves health status in patients with COPD without increasing healthcare costs [Abstract]. European Respiratory Journal 2003;22(Suppl 45):P433.
Lofdahl CG, Ericsson A, Svensson K, Andreasson E. Cost effectiveness of budesonide/formoterol in a single inhaler for COPD compared with each mono‐component used alone. Pharmacoeconomics 2005;23(4):365‐75.

Calverley 2003 TRISTAN {published and unpublished data}

Calverley P, Pauwels R, Vestbo J, Jones P, Pride N, Gulsvik A, et al. Combined salmeterol and fluticasone in the treatment of chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 2003;361(9356):449‐56.
Calverley PMA, Pauwels R, Vestbo J, Jones P, Pride N, Gulsvik A, et al. Safety of salmeterol/fluticasone propionate combination in the treatment of chronic obstructive pulmonary disease. European Respiratory Journal 2002;20(Suppl 38):242s [P1572].
Calverley PMA, Pauwels RA, Vestbo J, Jones PW, Pride NB, Gulsvik A, et al. Clinical improvements with salmeterol / fluticasone propionate combination in differing severities of COPD [A035] [Poster D50]. http://www.abstracts2view.com (accessed 20 April 2012).
Calverley PMA, Pauwels RA, Vestbo J, Jones PW, Pride NB, Gulsvik A, et al. Salmeterol/fluticasone propionate combination for one year provides greater clinical benefit than its individual components [A98] [Poster 306]. http://www.abstracts‐on‐line.com/abstracts/ATS (accessed 20 April 2012).
Hunjan MK, Chandler F. Numbers needed to treat (NNT) to avoid an exacerbation in patients with chronic obstructive pulmonary disease (COPD) using salmeterol/fluticasone propionate combination (SFC) and associated costs [Abstract]. American Thoracic Society 100th International Conference; 2004 May 21‐26; Orlando. 2004:D22 Poster 503.
Hunjan MK, Williams DT. Costs of avoiding exacerbations in patients with chronic obstructive pulmonary disease (COPD) treated with salmeterol/fluticasone propionate combination (Seretide) and salmeterol. European Respiratory Journal 2004;24 Suppl 48:291s.
Hunjan MK, Williams DT. Salmeterol/fluticasone propionate combination is clinically effective in avoiding exacerbations in patients with moderate/severe COPD. European Respiratory Journal 2004;24 Suppl 48:513s.
Jones PW, Edin HM, Anderson J. Salmeterol/fluticasone propionate combination improves health status in COPD patients.. http://www.abstracts‐on‐line.com/abstracts/ATS (accessed 21 April 2012).
Jones PW, Ståhl E. Budesonide/formoterol sustains clinically relevant improvements in health status in COPD [Abstract]. European Respiratory Journal 2005;26 Suppl 49:Abstract No. 1352.
Jones PW, Vestbo J, Pauwels RA, Calverley PMA, Anderson JA, Spencer MD. Informative drop out in COPD studies. Investigation of health status of withdrawals in the TRISTAN study. European Respiratory Society 13th Annual Congress; 2003 Sep 28‐29; Vienna. 2003:P1593.
Nitschmann S. Inhalational combination therapy in chronic obstructive lung disease. TRISTAN study. German Internist 2004;45(6):727‐8.
Pauwels RA, Calverly PMA, Vestbo J, Jones PW, Pride N, Gulsvik A, et al. Reduction of exacerbations with salmeterol/fluticasone combination 50/500 mcg bd in the treatment of chronic obstructive pulmonary disease. European Respiratory Journal 2002;20 Suppl 38:240 [P1569].
Pauwels RA, Vestbo J, Calverley PMA, Jones PW, Pride NB, Gulsvik A. Characterization of exacerbations in the TRISTAN study of salmeterol / fluticasone propionate (SFC) combination in moderate to severe COPD. http://www.abstracts2view.com (accessed 20 April 2012).
SFCB3024. A multi‐centre, randomised, double‐blind, parallel group, placebo‐controlled study to compare the efficacy and safety of the salmeterol/FP combination product at a strength of 50/500mcg bd with salmeterol 50mcg bd alone and FP 500mcg bd alone, delivered via the DISKUS™/ACCUHALER™, in the treatment of subjects with chronic obstructive pulmonary disease (COPD) for 12 months. http://ctr.gsk.co.uk (accessed 21 April 2012).
Spencer M, Briggs AH, Grossman RF, Rance L. Development of an economic model to assess the cost effectiveness of treatment interventions for chronic obstructive pulmonary disease. Pharmacoeconomics 2005;23(6):619‐37.
Spencer MD, Karia N, Anderson J. The clinical significance of treatment benefits with the salmeterol/fluticasone propionate 50/500mcg combination in COPD. European Respiratory Journal 2004;24 Suppl 48:290s.
Vestbo J, Calverley PMA, Pauwels R, Jones P, Pride N, Gulsvik A, et al. Absence of gender susceptibility to the combination of salmeterol and fluticasone in the treatment of chronic obstructive pulmonary disease. European Respiratory Journal 2002;20 Suppl 38:240 [P1570].
Vestbo J, Pauwels R, Anderson JA, Jones P, Calverley P. Early onset of effect of salmeterol and fluticasone propionate in chronic obstructive pulmonary disease. Thorax 2005;60(4):301‐4.
Vestbo J, Pauwels RA, Calverley PMA, Jones PW, Pride NB, Gulsvik A. Salmeterol/fluticasone propionate combination produces improvement in lung function detectable within 24 hours in moderate to severe COPD. http://www.abstracts2view.com (accessed 20 April 2012).
Vestbo J, Soriano JB, Anderson JA, Calverley P, Pauwels R, Jones P. Gender does not influence the response to the combination of salmeterol and fluticasone propionate in COPD. Respiratory Medicine 2004;98(11):1045‐50.

Calverley 2007 TORCH {published and unpublished data}

Allegra L. TORCH study: An invitation to clinical considerations. GIMT ‐ Giornale Italiano delle Malattie del Torace 2007;61(1):15‐23.
Briggs AH, Glick HA, Lozano‐Ortega G, Spencer M, Calverley PM, Jones PW, et al. Is treatment with ICS and LABA cost‐effective for COPD? Multinational economic analysis of the TORCH study. European Respiratory Journal 2010;35(3):532‐9.
Briggs AH, Lozano‐Ortega G, Spencer S, Bale G, Spencer MD, Burge PS. Estimating the cost‐effectiveness of fluticasone propionate for treating chronic obstructive pulmonary disease in the presence of missing data. Value in Health 2006;9(4):227‐35.
Calverley P, Celli B, Anderson J, Ferguson G, Jenkins C, Jones P, et al. Salmeterol/fluticasone propionate combination (SFC) improves survival in COPD over three years: on‐treatment analysis from the TORCH study [Abstract]. American Thoracic Society International Conference; 2009 May 15‐20; San Diego. 2009:A6191[Poster #219].
Calverley P, Celli B, Anderson JA, Ferguson GT, Jenkins C, Jones PW, et al. The TOwards a Revolution in COPD Health (TORCH) study: salmeterol/fluticasone propionate improves survival in COPD over three years [Abstract]. Respirology 2006;11 Suppl 5:A149 [PS‐3‐8].
Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. New England Journal of Medicine 2007;356(8):775‐89.
Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW et al and TORCH Investigators. Cardiovascular events in patients with COPD: TORCH study results. Thorax 2010;65(8):719‐25.
Calverley PM, Celli B, Anderson JA, Ferguson GT, Jenkins C, Jones PW, et al. The TOwards a Revolution in COPD Health (TORCH) study: fluticasone propionate /salmeterol improves survival in COPD over three years. Chest 2006;130(4):122s.
Calverley PMA, Celli B, Andersen JA, Ferguson GT, Jenkins C, Jones PW, et al. The TORCH (TOwards a Revolution in COPD Health) study salmeterol/fluticasone propionate (SFC) improves survival in COPD over three years. European Respiratory Journal 2006;28 Suppl 50:34.
Celli B, Calverley PM, Anderson JA, Ferguson GT, Jenkins C, Jones P, et al. The TOwards a Revolution in COPD HEalth (TORCH) Study: salmeterol/fluticasone propionate reduces the rate of exacerbations over three years. Respirology 2006;11 Suppl 5:A140.
Celli B, Calverley PM, Anderson JA, Ferguson GT, Jenkins C, Jones PW, et al. The TOwards a Revolution in COPD Health (TORCH) study: fluticasone propionate/salmeterol reduces the rate of exacerbations over 3 years [Abstract]. Chest 2006;130(4):177s.
Celli B, Calverley PMA, Anderson JA, Ferguson GT, Jenkins C, Jones PW, et al. The TORCH (TOwards a Revolution in COPD Health) study salmeterol/fluticasone propionate (SFC) improves health status reduces exacerbations and improves lung function over three years [Abstract]. European Respiratory Journal 2006;28 Suppl 50:34s [E312].
Celli B, Emmett A, Crater G, Kalberg C. Fluticasone propionate/salmeterol (FSC) improves the inspiratory to total lung capacity ratio (IC/TLC) and exercise endurance time in patients with COPD. European Respiratory Society 16th Annual Congress; 2006 Sep 2‐6; Munich. 2006:A320 (Poster 323).
Celli B, Ferguson GT, Anderson JA, Jenkins CR, Jones PW, Vestbo J, et al. Salmeterol/fluticasone propionate (SFC) improves lung function and reduces the rate of decline over three years in the TORCH survival study [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:A763.
Celli B, Vestbo J, Jenkins CR, Jones PW, Ferguson GT, Calverley PMA, et al. Sex differences in mortality and clinical expressions of patients with chronic obstructive pulmonary disease: the TORCH experience. American Journal of Respiratory and Critical Care Medicine 2011;183(3):317‐22.
Celli BR, Thomas NE, Anderson JA, Ferguson GT, Jenkins CR, Jones PW, et al. Effect of pharmacotherapy on rate of decline of lung function in chronic obstructive pulmonary disease: results from the TORCH study. American Journal of Respiratory and Critical Care Medicine 2008;178(4):332‐8.
Celli D, Emmett A, Crater G, Kalberg C. Salmeterol/fluticasone propionate (SFC) improves the inspiratory to total lung capacity ratio (IC/TLC) and exercise endurance time in patients with COPD. European Respiratory Journal 2006;28 Suppl 50:764 [4395].
Corhay JL, Louis R. Clinical study of the month: the TORCH study (TOwards a Revolution in COPD Health) [L'étude clinique du mois. L'étude TORCH (TOwards a Revolution in COPD Health): vers une revolution de la santé des patients souffrant de BPCO]. Revue Medicale de Liege 2007;62(4):230‐4.
Ferguson GT, Calverley PM, Anderson JA, Celli B, Jenkins C, Jones PW, et al. The TOwards a Revolution in COPD Health (TORCH) study: fluticasone propionate/salmeterol is well tolerated in patients with COPD over 3 years [Abstract]. Chest 2006;130(4):178s.
Ferguson GT, Calverley PM, Anderson JA, Celli B, Jenkins CR, Jones PW, et al. Effect of salmeterol/fluticasone propionate (SFC) on bone mineral density (BMD) and eye disorders over three years in the TORCH trial [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:A763.
Ferguson GT, Calverley PM, Anderson JA, Jenkins CR, Celli B, et al. Prevalence and progression of osteoporosis in patients with COPD results from the towards a revolution in COPD health study. Chest 2009;136(6):1456‐65.
Ferguson GT, Calverley PMA, Anderson JA, et al. The TORCH (TOwards a Revolution in COPD Health) study: salmeterol/fluticasone propionate (SFC) improves survival in COPD over three years. European Respiratory Journal 2006;28 Suppl 50:34s.
Glick H, Briggs A, Lozano‐Ortega G, Spencer M, Vestbo J, Calverley P. Is treatment with ICS/LABA combination good value for money in COPD? Evidence from the TORCH study [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:[C97].
Jenkins C, Celli B, Anderson J, Ferguson G, Calverley P, Jones P, et al. Salmeterol/fluticasone propionate (SFC) is efficacious in GOLD stage II COPD patients: analysis from the TORCH study [Abstract]. American Thoracic Society International Conference; May 15‐20; San Diego. 2009:A6186 [Poster #213].
Jenkins CR, Celli B, Anderson JA, Ferguson GT, Jones PW, Vestbo J, et al. Seasonality and determinants of moderate and severe COPD exacerbations in the TORCH study. European Respiratory Journal 2012;39:38‐45.
Jenkins CR, Celli B, Anderson JA, Ferguson GT, Jones PW, Vestbo J, et al. The TORCH survival study: consistent efficacy results seen in geographic regions in a multi‐national study [Abstract]. American Thoracic Society International Conference; May 18‐23; San Francisco. 2007:[C97].
Jenkins CR, Jones PW, Calverley PM, Celli B, Anderson JA, Ferguson GT, et al. Efficacy of salmeterol/fluticasone propionate by GOLD stage of chronic obstructive pulmonary disease: analysis from the randomised, placebo‐controlled TORCH study. Respiratory Research 2009;10:59.
Jones PW, Anderson JA, Calverley PM, Celli BR, Ferguson GT, Jenkins C, et al. Health status in the TORCH study of COPD: treatment efficacy and other determinants of change. Respiratory Research 2011;12:71.
Jones PW, Calverley PM, Celli B, Anderson JA, Ferguson GT, Jenkins C, et al. The TOwards a Revolution in COPD Health (TORCH) study: fluticasone propionate/salmeterol improves and sustains health status in COPD over 3 years. Chest 2006;130(4):177s.
Jones PW, Vestbo J, Anderson JA, Celli B, Ferguson GT, Jenkins CR, et al. Informative withdrawal in a COPD study. An example from the TORCH Study [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:[C97].
Keene ON, Vestbo J, Anderson JA, Calverley PM, Celli B, Ferguson GT, et al. Methods for therapeutic trials in COPD: Lessons from the TORCH trial. European Respiratory Journal 2009;34(5):1018‐23.
McDonough C, Blanchard AR. TORCH study results: pharmacotherapy reduces lung function decline in patients with chronic obstructive pulmonary disease. Hospital Practice 2010;38(2):92‐3.
McGarvey LP, John M, Anderson JA, Zvarich M, Wise RA, Committee TCE et al. Ascertainment of cause‐specific mortality in COPD: operations of the TORCH Clinical Endpoint Committee. Thorax 2007;62(5):411‐5.
Mehta RS, Kathman SJ, Daley‐Yates PT, Cahn T, Beerahee M, Kunka RL, et al. Pharmacokinetics and pharmacodynamics in COPD patients following long‐term twice‐daily treatment with salmeterol/fluticasone propionate (SFC) 50/500mg and the individual components [Abstract]. American Thoracic Society International Conference; 2007 May 18‐23; San Francisco. 2007:Poster #A41.
SCO30003. A multi‐centre, randomised, double‐blind, parallel group, placebo‐controlled study to investigate the long‐term effects of salmeterol/fluticasone propionate (SERETIDE®/VIANI®/ADVAIR®) 50/500mcg bd, salmeterol 50mcg bd and fluticasone propionate 500mcg bd, all delivered via the DISKUS®/ACCUHALER® inhaler, on the survival of subjects with chronic obstructive pulmonary disease (COPD) over 3 years of treatment. www.ctr.gsk.co.uk (accessed 21 April 2012).
Sacchetta A. Long term therapy and outcome of chronic obstructive pulmonary disease with or without co‐morbidity: The TORCH Study. Italian Journal of Medicine 2008;2(3):11‐5.
Vestbo J, Anderson JA, Calverley PM, Celli B, Ferguson GT, Jenkins C, et al. Bias due to withdrawal in long‐term randomised trials in COPD: Evidence from the TORCH study. Clinical Respiratory Journal 2011;5(1):44‐9.
Vestbo J, TORCH Study Group. The TORCH (TOwards a Revolution in COPD Health) survival study protocol. European Respiratory Journal 2004;24(2):206‐10.

Calverley 2010 {published data only}

Calverley PMA, Kuna P, Monso E, Costantini M, Petruzzelli Sd, Sergio F, et al. Beclomethasone/formoterol in the management of COPD: a randomised controlled trial. Respiratory Medicine 2010;104:1858‐68.

Choudhury 2005 {published data only}

Choudhury AB, Dawson CM, Kilvington HE, Eldridge HE, James WY, Wedzicha JA, et al. Withdrawal of inhaled corticosteroids in people with chronic obstructive pulmonary disease (COPD) in primary care ‐ a randomised controlled trial [Abstract]. European Respiratory Journal 2005;26 Suppl 49:Abstract No. 1328.
Choudhury AB, Dawson CM, Kilvington HE, Eldridge S, James WY, Wedzicha JA, et al. Withdrawal of inhaled corticosteroids in people with COPD in primary care: a randomised controlled trial. Respiratory Research 2007;8:93.

Dal Negro 2003 {published data only}

Dal Negro R, Micheletto C, Trevsian F, Tognella S. Salmeterol and fluticasone 50ug/250ug BiD versus salmeterol 50ug bid and versus placebo in the long term treatment of COPD. American Thoracic Society 98th International Conference; May 17‐22; Georgia. 2002.
Dal Negro RW, Pomari C, Tognella S, Micheletto C. Salmeterol & fluticasone 50 mcg/250 mcg bid in combination provides a better long‐term control than salmeterol 50 microg bid alone and placebo in COPD patients already treated with theophylline. Pulmonary Pharmacology and Therapeutics 2003;16(4):241‐6.

Dransfield 2013 {published data only}

Dransfield MT, Bourbeau J, Jones PW, Hanania NA, Mahler DA, Vestbo J, et al. Once‐daily inhaled fluticasone furoate and vilanterol versus vilanterol only for prevention of exacerbation of COPD: two replicate double‐blind, parallel‐group, randomised controlled trials. The Lancet: Respiratory Medicine 2013;1(3):210‐23.

Ferguson 2008 {published data only}

Dalal AA, Blanchette CM, Petersen H, Manavi K, St. Charles M. Cost‐effectiveness of fluticasone propionate/salmeterol (250/50 mcg) compared to salmeterol (50 mcg) in patients with COPD: economic evaluation of a randomized, double‐blind, parallel‐group, multi‐centre trial (Study SCO40043) [Abstract]. Chest 2008;134(4):106003s.
Ferguson G, Anzueto A, Fei R, Emmett A, Crater G, Knobil K, et al. A randomized, double‐blind trial comparing the effect of fluticasone/salmeterol 250/50 to salmeterol on COPD exacerbations in patients with COPD. Chest 2007;132(4):530b‐531.
Ferguson GT, Anzueto A, Fei R, Emmett A, Knobil K, Kalberg C. Effect of fluticasone propionate/salmeterol (250/50 mcg) or salmeterol (50 mcg) on COPD exacerbations. Respiratory Medicine 2008;102:1099‐108.
SCO40043. A randomized, double‐blind, parallel‐group, 52‐week study to compare the effect of fluticasone propionate/salmeterol DISKUS® 250/50mcg bid with salmeterol DISKUS® 50mcg bid on the annual rate of moderate/severe exacerbations in subjects with chronic obstructive pulmonary disease (COPD). http://ctr.gsk.co.uk (accessed 8 April 2008).

Fukuchi 2013 {published data only}

Fukuchi Y, Samoro R, Fassakhov R, Taniguchi H, Ekelund J, Carlsson LG, et al. Budesonide/formoterol via Turbuhaler® versus formoterol via Turbuhaler® in patients with moderate to severe chronic obstructive pulmonary disease: Phase III multinational study results. Respirology2013; Vol. 18:866‐73.
Ichinose M, Samoro R, Fassakhov R, Oguri M, Ekelund J, Carlsson LG, et al. Budesonide/formoterol vs formoterol, both via Turbuhaler®, in patients with moderate to severe COPD: Phase III study results [Abstract]. European Respiratory Society Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:374s [P2108].

GSK FCO30002 2005 {unpublished data only}

FCO30002. A multi‐centre, randomised, placebo‐controlled, double‐blind comparison with 3 parallel groups to investigate the efficacy and safety of inhaled glucocorticoid fluticasone (500 μg bd via Diskus™) vs. oral glucocorticoid therapy vs. placebo in subjects with chronic obstructive airway disease (COPD) under therapy with Salmeterol (50 μg bd). GlaxoSmithKline Clinical Trial Register (accessed 15 April 2012).

GSK FLTA3025 2005 {unpublished data only}

FLTA3025. A randomized, double‐blind, parallel‐group, comparative trial of inhaled fluticasone propionate 250mcg BID, 500mcg BID, and placebo BID via the DISKUS in subjects with chronic obstructive pulmonary disease (COPD). GlaxoSmithKline Clinical Trial Register (accessed 18 April 2012).

GSK SCO100470 2006 {unpublished data only}

SCO100470. A multi‐centre, randomised, double‐blind, parallel group, 24‐week study to compare the effect of the salmeterol/fluticasone propionate combination product 50/250mcg, with salmeterol 50mcg both delivered twice daily via the DISKUS/ACCUHALER inhaler on lung function and dyspnoea in subjects with Chronic Obstructive Pulmonary Disease (COPD). http:ctr.gsk.co.uk (accessed 20 April 2012).

GSK SCO104925 2008 {unpublished data only}

Evaluation of Novel Endpoints in Subjects with Chronic Obstructive Pulmonary Disease(COPD) in a Randomized, Double‐Blind, Placebo‐Controlled Study of Treatment with FluticasonePropionate/Salmeterol 500/50mcg combination and its individual components, FluticasonePropionate 500mcg and Salmeterol 50mcg. http://www.gsk‐clinicalstudyregister.com/ 2008 (accessed 19 April 2012).
SCO104925. Evaluation of Novel Endpoints in Subjects with Chronic Obstructive Pulmonary Disease(COPD) in a Randomized, Double‐Blind, Placebo‐Controlled Study of Treatment with FluticasonePropionate/Salmeterol 500/50mcg combination and its individual components, FluticasonePropionate 500mcg and Salmeterol 50mcg. http:ctr.gsk.co.uk (accessed 19 April 2012).

GSK SCO30002 2005 {unpublished data only}

SCO30002. A multi‐centre, randomised, double‐blind, parallel group, placebo‐controlled study to compare the efficacy and safety of inhaled salmeterol/fluticasone propionate combination product 25/250 µg two puffs bd and fluticasone propionate 250µg two puffs bd alone, all administered via metered dose inhalers (MDI), in the treatment of subjects with chronic obstructive pulmonary disease (COPD) for 52 weeks. GlaxoSmithKline Clinical Trial Register (accessed 21 April 2012).

GSK SCO40041 2008 {unpublished data only}

NCT00355342. A Randomized, Double‐Blind, Parallel‐Group Clinical Trial Evaluating the Effect of the Fluticasone Propionate/Salmeterol Combination Product 250/50mcg BID Via DISKUS Versus Salmeterol 50mcg BID Via DISKUS on Bone Mineral Density in Subjects With Chronic Obstructive Pulmonary Disease (COPD). http://clinicaltrials.gov/show/NCT00355342 (accessed 19 April 2012).

Hanania 2003 {published data only}

Hanania NA, Darken P, Horstman D, Reisner C, Lee B, Davis S, et al. The efficacy and safety of fluticasone propionate (250 mcg)/salmeterol (50 mcg) combined in the Diskus inhaler for the treatment of COPD. Chest 2003;124(3):834‐43.
Hanania NA, Knobil K, Watkins M, Wire P, Yates J, Darken P. Salmeterol and fluticasone propionate therapy administered by a single Diskus in patients with COPD. Chest 2002:S129.
Hanania NA, Ramsdell J, Payne K, Davis S, Horstman D, Lee B, et al. Improvements in airflow and dyspnoea in COPD patients following 24 weeks treatment with salmeterol 50mcg and fluticasone propionate 250mcg alone or in combination via the Diskus. American Journal of Respiratory and Critical Care Medicine 2001;163 Suppl 5:A279.
Horstman D, Darken P, Davis S, Lee B. Improvements in FEV1 and symptoms in poorly reversible COPD patients following treatment with salmeterol 50mcg/fluticasone propionate 250mcg combination [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P434.
Mahler DA, Darken P, Brown CP, Knobil K. Predicting lung function responses to combination therapy in chronic obstructive pulmonary disease (COPD) [Abstract]. National COPD Conference; 2003 Nov 14‐15; Arlington. 2003:1081.
Mahler DA, Darken P, Brown CP, Knobil K. Predicting lung function responses to salmeterol/fluticasone propionate combination therapy in COPD [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P429.
SFCA3007. A randomized, double‐blind, placebo‐controlled, parallel‐group trial evaluating the safety and efficacy of the DISKUS formulations of salmeterol (SAL) 50mcg BID and fluticasone propionate (FP) 250mcg BID individually and in combination as salmeterol 50mcg/fluticasone propionate 250mcg BID (SFC 50/250) compared to placebo in COPD subjects. http://ctr.gsk.co.uk (accessed 20 April 2012).
Spencer M, Wire P, Lee B, Chang CN, Darken P, Horstman D. Patients with COPD using salmeterol/fluticasone propionate combination therapy experience improved quality of life. European Respiratory Journal 2003;22 Suppl 45:51s.
Spencer MD, Karia N, Anderson J. The clinical significance of treatment benefits with the salmeterol/fluticasone propionate 50/500mcg combination in COPD. European Respiratory Journal 2004;24 Suppl 48:290s.

Hattotuwa 2002 {published data only}

Gizycki MJ, Hattotuwa KL, Barnes N, Jeffery PK. Effects of fluticasone propionate on inflammatory cells in COPD: an ultrastructural examination of endobronchial biopsy tissue. Thorax 2002;57(9):799‐803.
Hattotuwa KL, Gizycki MJ, Ansari TW, Jeffery PK, Barnes NC. The effects of inhaled fluticasone on airway inflammation in chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2002;165(12):1592‐9.

Kardos 2007 {published and unpublished data}

Kardos P, Wencker M. Combination therapy with salmeterol and fluticasone propionate (SFC) is more effective than salmeterol (SAL) alone in reducing exacerbations of COPD. European Respiratory Journal 2005;26 Suppl 49:Abstract 1944.
Kardos P, Wencker M, Glaab T, Vogelmeier C. Impact of salmeterol/fluticasone propionate versus salmeterol on exacerbations in severe chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2007;175(2):144‐9.
SCO30006. A randomised, double‐blind, parallel‐group study to investigate the protective effect of the combination of fluticasone and salmeterol (500/50 µg bid via the DISKUS) compared with salmeterol (50 µg bid via the DISKUS) on the incidence of moderate to severe exacerbations in patients with severe chronic obstructive pulmonary disease (COPD) (GOLD III/IV). http:ctr.gsk.co.uk (accessed 21 April 2012).
Vogelmeier C. Combination therapy with salmeterol and fluticasone propionate (SFC) improves quality of life (QoL) more than salmeterol (SAL) alone in COPD. 42nd Nordic Lung Conference; 2005 Jun 9‐11; Trondheim. 2005; Vol. 13 Suppl 22.
Vogelmeier CF, Wencker M, Glaab TH, Kardos P. Number needed to treat (NNT) to reduce exacerbations in severe COPD comparing salmeterol/fluticasone propionate (SFC) with salmeterol (SAL) treatment.. American Thoracic Society International Conference; May 19‐21; San Diego. 2006:A110.

Kerwin 2013 {published data only}

Kerwin EM, Scott‐Wilson C, Sanford L, Rennard S, Agusti A, Barnes N, et al. A randomised trial of fluticasone furoate/vilanterol (50/25 mug; 100/25 mug) on lung function in COPD. Respiratory Medicine2013; Vol. 107, issue 4:560‐9. [0954‐6111]
Kerwin EM, Scott‐Wilson C, Sanford L, Rennard SI, Agusti A, Barnes N, et al. Lung function effects and safety of fluticasone furoate (FF)/vilanterol (VI) in patients with COPD: Low‐mid dose assessment [Abstract]. European Respiratory Society Annual Congress; Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:545s [3082].

Lapperre 2009 {published and unpublished data}

Kunz LIZ, Strebus J, Budulac SE, Lapperre TS, Sterk PJ, Postma DS, et al. Inhaled Steroids Modulate Extracellular Matrix Composition in Bronchial Biopsies of COPD Patients: A Randomized, Controlled Trial. PloS one2013; Vol. 8, issue 5:e63430.
Lapperre TS, Snoeck‐Stroband JB, Gosman MM, Jansen DF, van Schadewijk A, Thiadens HA, et al. Effect of fluticasone with and without salmeterol on pulmonary outcomes in chronic obstructive pulmonary disease: a randomized trial. Annals of Internal Medicine 2009;151(8):517‐27.

Laptseva 2002 {published data only}

Laptseva IM, Laptseva EA, Borshchevsky VV, Gurevich G, Kalechits O. Inhaled budesonide in the management of chronic obstructive pulmonary disease. European Respiratory Journal 2002;20 Suppl 38:244s.

Mahler 2002 {published and unpublished data}

Mahler DA, Darken P, Brown CP, Knobil K. Predicting lung function responses to combination therapy in chronic obstructive pulmonary disease (COPD). http://www.abstracts2view.com (accessed 15 April 2012).
Mahler DA, Wire P, Horstman D, Chang CN, Yates J, Fischer T, et al. Effectiveness of fluticasone propionate and salmeterol combination delivered via the Diskus device in the treatment of chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2002;166(8):1084‐91.
SFCA3006. A randomized, double‐blind, placebo‐controlled, parallel‐group trial evaluating the safety and efficacy of the DISKUS formulations of salmeterol (SAL) 50mcg BID and fluticasone propionate (FP) 500mcg BID individually and in combination as salmeterol 50mcg/fluticasone propionate 500mcg BID (SFC 50/500) compared to placebo in COPD subjects. http://ctr.gsk.co.uk (accessed 17 April 2012).
Spencer M, Wire P, Lee B, Chang CN, Darken P, Horstman D. Patients with COPD using salmeterol/fluticasone propionate combination therapy experience improved quality of life. European Respiratory Journal 2003;22 Suppl 45:51.
Spencer MD, Anderson JA. Salmeterol/fluticasone combination produces clinically important benefits in dyspnoea and fatigue [Abstract]. American Thoracic Society 2005 International Conference; 2005 May 20‐25; San Diego. 2005:B93 [Poster 308].
Spencer MD, Karia N, Anderson J. The clinical significance of treatment benefits with the salmeterol/fluticasone propionate 50/500mcg combination in COPD. European Respiratory Journal 2004;24 Suppl 48:290.

Martinez 2013 {published data only}

Martinez F, Boscia J, Feldman G, Scott‐Wilson C, Kilbride S, Fabbri L, et al. Lung function effects and safety of fluticasone furoate (FF)/vilanterol (VI) in patients with COPD: mid‐high dose assessment [Abstract]. European Respiratory Society Annual Congress, Vienna, Austria, September 1‐5. 2012; Vol. 40, issue Suppl 56:527s [P2887].
Martinez FJ, Boscia J, Feldman G, Scott‐Wilson C, Kilbride S, Fabbri L, et al. Fluticasone furoate/vilanterol (100/25; 200/25 mug) improves lung function in COPD: a randomised trial. Respiratory Medicine 2013;107(4):550‐9. [0954‐6111]

Mirici 2001 {published data only}

Mirici A, Bektas Y, Ozbakis G, Erman Z. Effect of inhaled corticosteroids on respiratory function tests and airway inflammation in stable chronic obstructive pulmonary disease. Clinical Drug Investigation 2001;21(12):835‐42.

Ozol 2005 {published data only}

Ozol D, Aysan T, Solak ZA, Mogulkoc N, Veral A, Sebik F. The effect of inhaled corticosteroids on bronchoalveolar lavage cells and IL‐8 levels in stable COPD patients. Respiratory Medicine 2005;99(12):1494‐500.

Paggiaro 1998 {published data only}

FLIT97. A multi‐centre, randomised, double‐blind, parallel group study of the efficacy and safety of inhaled fluticasone propionate 1000 µg daily with placebo in chronic obstructive pulmonary disease. http://ctr.gsk.co.uk (accessed 20 April 2012).
Paggiaro PL, Dahle R, Bakran I, Frith L, Hollingworth K, Efthimiou J. Multicentre randomised placebo‐controlled trial of inhaled fluticasone propionate in patients with chronic obstructive pulmonary disease. International COPD Study Group. Lancet 1998;351(9105):773‐80.

Pauwels 1999 {published data only}

Johnell O, Pauwels R, Löfdahl C‐G, Laitinen LA, Postma DS, Pride NB, et al. Bone mineral density in patients with chronic obstructive pulmonary disease treated with budesonide Turbuhaler. European Respiratory Journal 2002;19(6):1058‐63.
Lofdahl CG, Postma DS, Laitinen LA, Ohlsson SV, Pauwels RA, Pride NB. The European Respiratory Society study on chronic obstructive pulmonary disease (EUROSCOP): recruitment methods and strategies. Respiratory Medicine 1998;92(3):467‐72.
Pauwels RA, Lofdahl CG, Laitinen LA, Schouten JP, Postma DS, Pride NB, et al. Long‐term treatment with inhaled budesonide in persons with mild chronic obstructive pulmonary disease who continue smoking. European Respiratory Society Study on Chronic Obstructive Pulmonary Disease. New England Journal of Medicine 1999;340(25):1948‐53.
Pauwels RA, Lofdahl CG, Pride NB, Postma DS, Laitinen LA, Ohlsson SV. European Respiratory Society study on chronic obstructive pulmonary disease (EUROSCOP): hypothesis and design. European Respiratory Journal 1992;5(10):1254‐61.

Renkema 1996 {published data only}

Renkema TE, Schouten JP, Koeter GH, Postma DS. Effects of long‐term treatment with corticosteroids in COPD. Chest 1996;109(5):1156‐62.
van Grunsven PM, van Schayck CP, Derenne JP, Kerstjens HA, Renkema TE, Postma DS, et al. Long term effects of inhaled corticosteroids in chronic obstructive pulmonary disease: a meta‐analysis. Thorax 1999;54(1):7‐14.

Rennard 2009 {published data only}

Bleecker ER, Meyers DA, Bailey WC, Sims AM, Bujac SR, Goldman M, et al. ADRB2 polymorphisms and budesonide/formoterol responses in COPD. Chest2012; Vol. 142, issue 2:320‐8.
Rennard SI, Tashkin DP, McElhattan J, Goldman M, Ramachandran S, Martin UJ, et al. Efficacy and tolerability of budesonide/formoterol in one hydrofluoroalkane pressurized metered‐dose inhaler in patients with chronic obstructive pulmonary disease. Results from a 1‐year randomized controlled clinical trial. Drugs 2009;69(5):549‐65.

Schermer 2009 {published data only}

Chavannes NH, Schermer TR, Wouters EF, Akkermans RP, Dekhuijzen RP, Muris JW, et al. Predictive value and utility of oral steroid testing for treatment of COPD in primary care: the COOPT study. International Journal of COPD 2009;4:431‐6.
Schermer T, Chavannes N, Dekhuijzen R, Wouters E, Muris J, Akkermans R, et al. Fluticasone and N‐acetylcysteine in primary care patients with COPD or chronic bronchitis. Respiratory Medicine 2009;103(4):542‐51.

Senderovitz 1999 {published data only}

Senderovitz T, Vestbo J, Frandsen J, Maltbaek N, Norgaard M, Nielsen C, et al. Steroid reversibility test followed by inhaled budesonide or placebo in outpatients with stable chronic obstructive pulmonary disease. Respiratory Medicine 1999;93(10):715‐8.

Shaker 2009 {published and unpublished data}

Shaker SB, Dirksen A, Ulrik CS, Hestad M, Stavngaard T, Laursen LC, et al. The effect of inhaled corticosteroids on the development of emphysema in smokers assessed by annual computed tomography. COPD: Journal of Chronic Obstructive Pulmonary Disease 2009;6(2):104‐11.
Shaker SB, Stavngaard T, Laursen LC, Stoel BC, Dirksen A. Rapid fall in lung density following smoking cessation in COPD. COPD: Journal of Chronic Obstructive Pulmonary Disease 2011;8(1):2‐7.

Sharafkhaneh 2012 {published data only}

Sharafkhaneh A, Southard JG, Goldman M, Uryniak T, Martin UJ. Effect of budesonide/formoterol pMDI on COPD exacerbations: a double‐blind, randomized study. Respiratory Medicine 2012;106(2):257‐68.
Sharafkhaneh A, Uryniak T, Martin U. Long‐term effects of budesonide/formoterol pressurized metered‐dose inhaler on Chronic Obstructive Pulmonary Disease (COPD) symptoms and health status in patients with COPD [Abstract]. Chest 2011;140(4):528A.

Szafranski 2003 {published data only}

Anderson P. Budesonide/formoterol in a single inhaler (Symbicort) provides early and sustained improvement in lung function in moderate to severe COPD. Thorax 2002;57 Suppl 3:iii43.
Borgstrom L, Asking L, Olsson H, Peterson S. Lack of interaction between disease severity and therapeutic response with budesonide/formoterol in a single inhaler [Abstract].. American Thoracic Society 100th International Conference; May 21‐26; Orlando. 2004:C22 [Poster 505].
Calverley P, Pauwels Dagger R, Löfdahl CG, Svensson K, Higenbottam T, Carlsson LG, et al. Relationship between respiratory symptoms and medical treatment in exacerbations of COPD. European Respiratory Journal 2005;26(3):406‐13.
Calverley PMA. Effect of budesonide/formoterol on severe exacerbations and lung function in moderate to severe COPD. Thorax 2002;BTS Winter Meeting 2002:S145.
Calverley PMA, Szafranski W, Andersson A. Budesonide/formoterol is a well‐tolerated long term maintenance therapy for COPD. European Respiratory Journal 2005;26 Suppl 49:Poster 1917.
Calverley PMA, Thompson NC, Olsson H. Budesonide/formoterol in a single inhaler sustains lung function improvements in COPD [Abstract]. European Respiratory Journal 2003;22 Suppl 45:P435.
Campbell LM, Szafranski W. Budesonide/formoterol in a single inhaler (Symbicort) provides sustained relief from symptoms in moderate to severe COPD. Thorax. 2002; Vol. BTS Winter Meeting 2002:S143.
Campbell LW, Szafranski W. Budesonide/formoterol in a single inhaler (Symbicort) reduces severe exacerbations in patients with moderate‐severe COPD. Thorax. 2002; Vol. BTS Winter Meeting 2002:S141.
Dahl R, Cukier A, Olsson H. Budesonide/formoterol in a single inhaler reduces severe and mild exacerbations in patients with moderate to severe COPD. European Respiratory Journal 2002;20 Suppl 38:242 [P1575].
Egede F, Menga G. Budesonide/formoterol in a single inhaler provides sustained relief from symptoms and night‐time awakenings in moderate‐severe COPD: results from symptoms and night‐time awakenings in moderate to severe COPD: results from a 1‐year study. European Respiratory Journal 2002;20 Suppl 38:242 [P1574].
Halpin D, Stahl E, Lundback B, Anderson F, Peterson S. Treatment costs and number needed to treat (NNT) with budesonide/formoterol to avoid one exacerbation of COPD [Abstract]. American Thoracic Society 100th International Conference; May 21‐26; Orlando. 2004:D22 Poster 525.
Jones PW, Stahl E, Svensson K. Improvement in health status in patients with moderate to severe COPD after treatment with budesonide/formoterol in a single inhaler. European Respiratory Journal 2002;20 Suppl 38:250 [P1613].
Korsgaard J, Sansores R. Budesonide/formoterol (single inhaler) provides sustained relief from shortness of breath and chest tightness in a 1‐year study of patients with moderate to severe COPD. European Respiratory Journal 2002;20 Suppl 38:242 [P1577].
Lange P, Saenz C. Budesonide/formoterol in a single inhaler is well tolerated in patients with moderate to severe COPD: results of a 1 year study. European Respiratory Journal 2002;20 Suppl 38:242 [P1573].
Lofdahl CG. Reducing the impact of COPD exacerbations: clinical efficacy of budesonide/formoterol. European Respiratory Review 2004;13(88):14‐21.
Milanowski J, Nahabedian S. Budesonide/formoterol in a single inhaler acts rapidly to improve lung function and relieve symptoms in patients with moderate to severe COPD. European Respiratory Journal 2002;20 Suppl 38:242 [P1576].
Szafranski W, Cukier A, Ramirez A, Menga G, Sansores R, Nahabedian S, et al. Efficacy and safety of budesonide/formoterol in the management of chronic obstructive pulmonary disease. European Respiratory Journal 2003;21:74‐81.
Szfranski W, Ramirez A, Peterson S. Budesonide/formoterol in single inhaler provides sustained improvements in lung function in patients with moderate to severe COPD [Abstract]. European Respiratory Society Annual Congress; Sep 14‐18; Stockholm. 2002.

Tashkin 2008 SHINE {published and unpublished data}

AstraZeneca. A 6‐month double‐blind, double‐dummy, randomized, parallel group, multicenter efficacy & safety study of SYMBICORT® pMDI 2 x 160/4.5 μg & 80/4.5 μg bid compared to formoterol TBH, budesonide pMDI (& the combination) & placebo in COPD Patients (SHINE) (study number D589900002). www.astrazenecaclinicaltrials.com (accessed 18 April 2012).
Bleecker ER, Meyers DA, Bailey WC, Sims AM, Bujac SR, Goldman M, et al. Effect of ß2‐Adrenergic Receptor Gene Polymorphism Gly16Arg on Response to Budesonide/Formoterol Pressurized Metered‐Dose Inhaler in Chronic Obstructive Pulmonary Disease [Abstract]. American Journal of Respiratory and Critical Care Medicine 2011;183:A4086.
Tashkin DP, Rennard SI, Martin P, Ramachandran S, Martin UJ, Silkoff PE. Efficacy and safety of budesonide and formoterol in one pressurized metered‐dose inhaler in patients with moderate to very severe chronic obstructive pulmonary disease: results of a 6‐month randomized clinical trial. Drugs 2008;68(14):1975‐2000.

van Grunsven 2003 {published data only}

van Grunsven P, Schermer T, Akkermans R, Albers M, van den Boom G, van Schayck O, et al. Short‐ and long‐term efficacy of fluticasone propionate in subjects with early signs and symptoms of chronic obstructive pulmonary disease. Results of the DIMCA study. Respiratory Medicine 2003;97(12):1303‐12.

Verhoeven 2002 {published and unpublished data}

Verhoeven GT, Garrelds IM, Hoogsteden HC, Zijlstra FJ. Effects of fluticasone propionate inhalation on levels of arachidonic acid metabolites in patients with chronic obstructive pulmonary disease. Mediators of Inflammation 2001;10(1):21‐6.
Verhoeven GT, Hegmans JP, Mulder PG, Bogaard JM, Hoogsteden HC, Prins JB. Effects of fluticasone propionate in COPD patients with bronchial hyper‐responsiveness. Thorax 2002;57(8):694‐700.
Verhoeven GT, Wijkhuijs AJ, Hooijkaas H, Hoogsteden HC, Sluiter W. Effect of an inhaled glucocorticoid on reactive oxygen species production by bronchoalveolar lavage cells from smoking COPD patients. Mediators of Inflammation 2000;9(2):109‐13.

Vestbo 1999 {published data only}

Vestbo J, Sorensen T, Lange P, Brix A, Torre P, Viskum K. Long‐term effect of inhaled budesonide in mild and moderate chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 1999;353(9167):1819‐23. [MEDLINE: 99285938]

Yildiz 2004 {published data only}

Yildiz F, Basyigit I, Yildirim E, Boyaci H, Ilgazli A. Does addition of inhaled steroids to combined bronchodilator therapy affect health status in patients with COPD?. Respirology 2004;9(3):352‐5.

Referencias de los estudios excluidos de esta revisión

GSK FLIP63 2005 {published data only}

GlaxoSmithKline F. A single‐centre, double‐blind study to investigate the effects of inhaled fluticasone propionate (FP) 1500mcgdaily on inflammatory processes in the lungs of patients with chronic obstructive airways disease [FLIP63]. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SAM30022 2005 {published data only}

GSK SAM30022. A phase IV, multi‐centre, double‐blind, double‐dummy, parallel group, randomised study comparing Seretide (25/50 2 puffs bd) via the Evohaler (MDI‐HFA) with Beclometasone dipropionate (200mcg 2 puffs bd) via the MDI‐CFC in adolescents and adults experiencing moderate symptoms of reversible airways obstruction. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SAM40004 2006 {published data only}

GSK SAM40004. A multi‐centre, randomised, double‐blind, placebo‐controlled parallel group study to compare the effect on airway inflammation and remodelling of treatment with salmeterol/fluticasone propionate combination product (50/100μg strength) bd via the Accuhaler inhaler, or fluticasone propionate 100μg bd via the Accuhaler inhaler or placebo via the Accuhaler inhaler for 16 weeks, followed by double‐blind treatment for 52 weeks with the salmeterol/fluticasone propionate combination product (50/100μg strength) bd via the Accuhaler inhaler or fluticasone propionate 100μg bd via the Accuhaler inhaler, in adults with reversible airways obstruction (SIRIAS ‐ Seretide in Inflammation and Remodelling In Asthma Study). www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SAS40015 2007 {published data only}

GSK SAS40015. A multi‐centre, randomised, double‐blind, double‐dummy, parallel‐group, 12‐week, active control comparison of the salmeterol/fluticasone propionate combination product (50/100 mcg strength) bd via the DISKUS/ACCUHALER inhaler with fluticasone propionate (100 mcg strength) bd via the DISKUS/ACCUHALER inhaler plus oral montelukast 10 mg od in adolescents and adults with reversible airways obstruction [SAS40015]. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO100540 2006 {published data only}

GSK SCO100540. A multi‐centre, randomised, double‐blind, parallel group study to investigate the efficacy and safety of the Salmeterol/fluticasone propionate combination at a strength of 50/500µg BD, compared with placebo via Accuhaler, added to usual chronic obstructive pulmonary disease (COPD) therapy, in subjects with COPD for 24 weeks [SCO100540]. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO100646 2008 {published data only}

GSK SCO100646. Clinical Evaluation of SFC for Chronic Obstructive Pulmonary Disease (Chronic Bronchitis, Emphysema) ‐Assessment of the Effect of Addition of Fluticasone Propionate to Salmeterol Xinafoate 50 mcg after Switching under Double‐blinded Conditions to SFC 50/250 mcg in Chronic Obstructive Pulmonary Disease. Study number SCO100646. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO30005 2006 {published data only}

GSK SCO30005. A 13‐week, double‐blind, parallel‐group, multi‐centre study to compare the bronchial anti‐inflammatory activity of the combination of salmeterol/ fluticasone propionate (SERETIDE™/ADVAIR™/VIANI™) 50/500 mcg twice daily compared with placebo twice daily in patients with Chronic Obstructive Pulmonary Disease [SCO30005]. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO40030 2005 {published data only}

GSK SCO40030. A Randomized, Double‐Blind, Placebo‐Controlled, Parallel Group Clinical Trial Evaluating the Effect of the Fluticasone Propionate/Salmeterol Combination Product 250/50mcg BID via DISKUS and Salmeterol 50mcg BID via DISKUS on Lung Hyperinflation in Subjects with Chronic Obstructive Pulmonary Disease (COPD). [SCO40030]. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO40034 2009 {published data only}

GSK SCO40034. A multi‐centre, randomised, double‐blind, double dummy, parallel group 12‐week exploratory study to compare the effect of the fluticasone/salmeterol propionate combination product (SERETIDE™) 50/500mcg bd via the DISKUS™/ACCUHALER™ inhaler with tiotropium bromide 18 mcg od via the Handihaler inhalation device on efficacy and safety in patients with Chronic Obstructive Pulmonary Disease (COPD). www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

GSK SCO40036 2009 {published data only}

GSK SCO40036. Multicentre, Randomised, Double‐Blind, Double Dummy, Parallel Group, 104‐week Study to Compare the Effect of the Salmeterol/Fluticasone Propionate Combination Product (SERETIDE*) 50/500mcg Delivered Twice Daily via the DISKUS*/ACCUHALER* Inhaler with Tiotropium Bromide 18 mcg Delivered Once Daily via the HandiHaler Inhalation Device on the Rate of Health Care Utilisation Exacerbations in Subjects with Severe Chronic Obstructive Pulmonary Disease (COPD). www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).
GSK SCO40036. Multicentre, randomised, double‐blind, double‐dummy, parallel group, 104‐week study to compare the effect of the salmeterol/fluticasone propionate combination product (SERETIDE*) 50/500mcg delivered twice daily via the DISKUS*/ACCUHALER* inhaler with tiotropium bromide 18 mcg delivered once daily via the HandiHaler inhalation device on the rate of health care utilisation exacerbations in subjects with severe chronic obstructive pulmonary disease (COPD). www.gsk‐clinicalstudyregister.com (accessed 8 April 2012).

GSK SFCB3019 2004 {published data only}

GSK SFCB3019. A multi‐centre randomized, double‐blind, double‐dummy, parallel‐group comparison of the salmeterol/fluticasone propionate combination product (50/500mcg strength) BD via one DISKUS/Accuhaler inhaler with salmeterol 50mcg BD via one DISKUS/Accuhaler and fluticasone propionate 500mcg BD via another DISKUS/Accuhaler and with fluticasone propionate 500mcg BD via one DISKUS/Accuhaler in adolescents and adults with reversible airways obstruction. www.gsk‐clinicalstudyregister.com (accessed 20 April 2012).

Lung Health Study 2000 {published data only}

Lung Health Study Research Group. Effect of inhaled triamcinolone on the decline in pulmonary function in chronic obstructive pulmonary disease. New England Journal of Medicine 2000;343(26):1902‐9.

van der Valk 2002 {published data only}

Monninkhof E, van der Valk P, van der Palen J, Zielhuis G, van Hervaarden C, Van Herwaarden C. Effect of discontinuation of inhaled steroids on health‐related quality of life in patients with COPD. American Journal of Respiratory and Critical Care Medicine 2002;165 Suppl 8:A226.
van der Valk P, Monninkhof E, van der Palen J, Zielhuis G, van Herwaarden C, van der Phalen J. Effect of discontinuation of inhaled corticosteroids in patients with chronic obstructive pulmonary disease: the COPE study. American Journal of Respiratory and Critical Care Medicine 2002;166(1073‐449X):1358‐63.
van der Valk P, Monninkhof EM, van der Palen JAM, Zielhuis GA, van Herwaarden CLA. Effect of discontinuation of inhaled fluticasone propionate on exacerbations in patients with COPD. European Respiratory Journal 2001;18 Suppl 33:183s.

Weir 1999 {published data only}

Weir DC, Bale GA, Bright P, Sherwood Burge P. A double‐blind placebo‐controlled study of the effect of inhaled beclomethasone dipropionate for 2 years in patients with nonasthmatic chronic obstructive pulmonary disease. Clinical and Experimental Allergy 1999;29(Suppl 2):125‐8.

Wouters 2005 {published data only}

Wouters EF. COPD and Seretide: a multi‐centre intervention and characterisation (COSMIC) study: a rationale and baseline characteristics [abstract]. European Respiratory Society Annual Congress; Sep 14‐18; Stockholm. 2002:P1571.
Wouters EF, Postma DS, Fokkens B, Hop WC, Prins J, Kuipers AF, et al. Withdrawal of fluticasone propionate from combined salmeterol/fluticasone treatment in patients with COPD causes immediate and sustained disease deterioration: a randomised controlled trial. Thorax 2005;60:480‐7.
Wouters EF, Postma DS, Fokkens B, Hop WC, Prins J, Kuipers AFTCC, et al. [Withdrawal of fluticasone propionate from combined salmeterol/fluticasone treatment in patients with COPD causes immediate and sustained disease deterioration: a randomised controlled trial]. [Portuguese]. Revista Portuguesa de Pneumologia 2005;11(6):587‐9.

Referencias de los estudios en espera de evaluación

Ohar 2013 {published data only}

Ohar JA, Crater G, Emmett A, Ferro T, Morris A, Raphiou I, et al. Effects of Fluticasone Propionate/Salmeterol Combination 250/50mcg BID (ADVAIR DISKUS™) vs. Salmeterol 50mcg BID (SEREVENT DISKUS™) on Chronic Obstructive Pulmonary Disease (COPD) Exacerbation Rate, Following Acute Exacerbation or Hospitalization. American Journal of Respiratory and Critical Care Medicine. 2013; Vol. 187(Meeting Abstracts):A2439.

Referencias de los estudios en curso

Vestbo 2013 {published data only}

Vestbo J, Anderson J, Brook RD, Calverley PMA, Celli BR, Crim C, et al. The study to understand mortality and morbidity in COPD (SUMMIT) study protocol. European Respiratory Journal 2013;41(5):1017‐22. [0903‐1936]

Agarwal 2010

Agarwal R, Aggarwal AN, Gupta D, Jindal SK. Inhaled corticosteroids vs placebo for preventing COPD exacerbations. Chest 2010;137(2):318‐25.

Agusti 2013

Agusti A, de Teresa L, De Backer W, Zvarich MT, Locantore N, Barnes N, et al. A comparison of the efficacy and safety of once‐daily fluticasone furoate/vilanterol with twice‐daily fluticasone propionate/salmeterol in moderate to very severe COPD. The European Respiratory Journal. 2013/10/12 2013 Oct 10 [Epub ahead of print]. [1399‐3003: (Electronic)]

ATS/ERS 2004

American Thoracic Society / European Respiratory Society Task Force. Standards for the diagnosis and management of patients with COPD [Internet]. Version 1.2. http://www.thoracic.org/go/copd. New York: American Thoracic Society, (accessed 4 May 2012). [URL: http://www.thoracic.org/go/copd]

Blais 2010

Blais L, Forget A, Ramachandran S. Relative effectiveness of budesonide/formoterol and fluticasone propionate/salmeterol in a 1‐year, population‐based, matched cohort study of patients with chronic obstructive pulmonary disease (COPD): effect on COPD‐related exacerbations, emergency department visits and hospitalizations, medication utilization, and treatment adherence. Clinical Therapeutics 2010;32(7):1320‐8.

BTS 2009

British Thoracic Society, Community Acquired Pneumonia in Adults Guideline Group. Guidelines for the management of community acquired pneumonia in adults: update 2009. Thorax 2009;64(Suppl III):iii1–iii55.

Bucher 1997

Bucher HC, Guyatt GH, Griffith LE, Walter SD. The results of direct and indirect treatment comparisons in meta‐analysis of randomized controlled trials. Journal of Clinical Epidemiology1997; Vol. 50, issue 6:683‐91. [0895‐4356: (Print)]

Cates 2013

Cates C. Inhaled corticosteroids in COPD: quantifying risks and benefits. Thorax 2013;68(6):499‐500.

Cipriani 2013

Cipriani A, Higgins JPT, Geddes JR, Salanti GS. Conceptual and technical challenges in network meta‐analysis. Annals of Internal Medicine 2013;159(2):130‐7.

Drummond 2008

Drummond MB, Dasenbrook EC, Pitz MW, Murphy DJ, Fan E. Inhaled corticosteroids in patients with stable chronic obstructive pulmonary disease: a systematic review and meta‐analysis. JAMA 2008;300(20):2407‐16. [1538‐3598: (Electronic)]

Effing 2007

Effing T, Monninkhof EEM, van der Valk PPDLPM, Zielhuis GGA, Walters EH, van der Palen JJ, et al. Self‐management education for patients with chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2007, Issue 4. [DOI: 10.1002/14651858.CD002990.pub2]

Ernst 2007

Ernst P, Gonzalez AV, Brassard P, Suissa S. Inhaled corticosteroid use in chronic obstructive pulmonary disease and the risk of hospitalization for pneumonia. American Journal of Respiratory and Critical Care Medicine 2007;176:162‐6.

Glenny 2005

Glenny AM, Altman DG, Song F, Sakarovitch C, Deeks JJ, D'Amico R, et al. Indirect comparisons of competing interventions. Health Technology Assessment 2005;9(26):1‐134, iii‐iv. [1366‐5278: (Print)]

GOLD 2013

GOLD. Global Strategy for the Diagnosis, Management and Prevention of COPD, Global Initiative for Chronic Obstructive Lung Disease (GOLD) 2013. http://www.goldcopd.com(accessed 22 November 13).

Halpin 2011

Halpin DM, Gray J, Edwards SJ, Morais J, Singh D. Budesonide/formoterol vs. salmeterol/fluticasone in COPD: a systematic review and adjusted indirect comparison of pneumonia in randomised controlled trials. International Journal of Clinical Practice 2011;65(7):764‐74. [1742‐1241: (Electronic)]

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.2 [updated September 2009]. The Cochrane Collaboration, 2009. www.cochrane‐handbook.org.

Janson 2013 [PATHOS]

Janson C, Larsson K, Lisspers KH, Stallberg B, Stratelis G, Goike H, et al. Pneumonia and pneumonia related mortality in patients with COPD treated with fixed combinations of inhaled corticosteroid and long acting beta2 agonist: observational matched cohort study (PATHOS). BMJ. 2013/05/31 2013; Vol. 346:f3306. [1756‐1833: (Electronic)]

Johnson 1998

Johnson M. Development of fluticasone propionate and comparison with other inhaled corticosteroids. Journal of Allergy and Clinical Immunology 1998;101(4):S434‐9.

Lacasse 2006

Lacasse Y, Goldstein R, Lasserson TJ, Martin S. Pulmonary rehabilitation for chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2006, Issue 4. [DOI: 10.1002/14651858.CD003793.pub2]

Marzoratti 2013

Marzoratti L, Iannella HA, Waterer GW. Inhaled corticosteroids and the increased risk of pneumonia. Therapeutic Advances in Respiratory Disease. 2013/03/01 2013; Vol. 7, issue 4:225‐34. [1753‐4666: (Electronic)]

Nannini 2012

Nannini LJ, Lasserson TJ, Poole P. Combined corticosteroid and long‐acting beta2‐agonist in one inhaler versus long‐acting beta2‐agonists for chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2012, Issue 9. [DOI: 10.1002/14651858.CD006829.pub2]

Nannini 2013

Nannini LJ, Poole P, Milan SJ, Holmes R, Normansell R. Combined corticosteroid and long‐acting beta2‐agonist in one inhaler versus placebo for chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2013, Issue 11. [DOI: 10.1002/14651858.CD003794.pub4]

NICE 2011

National Institute for Health and Clinical Excellence. Chronic obstructive pulmonary disease, costing report, implementing NICE guidance. http://guidance.nice.org.uk/CG101/CostingReport/pdf/English (accessed 4 February 2012). [URL: http://guidance.nice.org.uk/CG101/CostingReport/pdf/English]

Partridge 2009

Partridge MR, Schuermann W, Beckman O, Persson T, Polanowski T. Effect on lung function and morning activities of budesonide/formoterol versus salmeterol/fluticasone in patients with COPD. Therapeutic Advances in Respiratory Disease 2009;3(4):147‐57.

Puhan 2011

Puhan MA, Gimeno‐Santos E, Scharplatz M, Troosters T, Walters EH, Steurer J. Pulmonary rehabilitation following exacerbations of chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2011, Issue 10. [DOI: 10.1002/14651858.CD005305.pub3]

Roberts 2011

Roberts M, Mapel D, Petersen H, Blanchette C, Ramachandran S. Comparative effectiveness of budesonide/formoterol and fluticasone/salmeterol for COPD management. Journal of Medical Economics 2011;14(6):769‐76.

Rossios 2011

Rossios C, To Y, To M, Ito M, Barnes PJ, Adcock IM, Johnson M, Ito K. Long‐acting fluticasone furoate has a superior pharmacological profile to fluticasone propionate in human respiratory cells. European Journal of Pharmacology 2011;670(1):244‐51.

Ruiz 2011

Ruiz Garcia V, Compte Torrero L. [Risk of pneumonia with inhaled corticosteroids for stable chronic obstructive pulmonary disease patients]. Medicina Clinica. 2011/03/15 2011; Vol. 137, issue 7:302‐4. [0025‐7753: (Print)]

Sin 2009

Sin DD, Tashkin D, Zhang X, Radner F, Sjobring U, Thoren A, et al. Budesonide and the risk of pneumonia: a meta‐analysis of individual patient data. Lancet 2009;374(9691):712‐9. [1474‐547X: (Electronic)]

Singanayagam 2010

Singanayagam A, Chalmers J, Hill A. Inhaled corticosteroids and risk of pneumonia: evidence for and against the proposed association. Quarterly Journal of Medicine 2010;103:379‐85.

Singh 2009

Singh S, Amin AV, Loke YK. Long‐term use of inhaled corticosteroids and the risk of pneumonia in chronic obstructive pulmonary disease: a meta‐analysis. Archives of Internal Medicine 2009;169(3):219‐29. [1538‐3679: (Electronic)]

Song 2008

Song F, Harvey I, Lilford R. Adjusted indirect comparison may be less biased than direct comparison for evaluating new pharmaceutical interventions. Journal of Clinical Epidemiology 2008;61(5):455‐63. [0895‐4356: (Print)]

Suissa 2013

Suissa S, Patenaude V, Lapi F, Ernst P. Inhaled corticosteroids in COPD and the risk of serious pneumonia. Thorax 2013;68:1029‐36.

Van der Meer 2001

Van der Meer RM, Wagena E, Ostelo RWJG, Jacobs AJE, van Schayck O. Smoking cessation for chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2001, Issue 1. [DOI: 10.1002/14651858.CD002999]

WHO 2012

World Health Organization. Chronic respiratory diseases. http://www.who.int/respiratory/en/ (accessed 7 February 2012).

Yang 2009

Yang IA, Fong K, Sim EHA, Black PN, Lasserson TJ. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD002991.pub2]

Yang 2012

Yang IA, Clarke MS, Sim EH, Fong KM. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. The Cochrane Database of Systematic Reviews. 2012/07/13 2012; Vol. 7:CD002991. [1469‐493X: (Electronic)]

Yawn 2013

Yawn BP, Li Y, Tian H, Zhang J, Arcona S, Kahler KH. Inhaled corticosteroid use in patients with chronic obstructive pulmonary disease and the risk of pneumonia: a retrospective claims data analysis. International Journal of Chronic Obstructive Pulmonary Disease. 2013/07/10 2013; Vol. 8:295‐304. [1178‐2005: (Electronic)]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Anzueto 2009

Methods

Design: randomised, double‐blind, parallel‐group study

12‐Month treatment period

Conducted at 98 research sites in the United States and Canada

Participants

Participants: 797 people were randomly assigned to fluticasone/salmeterol combination (394) and salmeterol alone (403)

Baseline characteristics:

Male %: flut/sal 51, sal 57

Mean age (SD), years: flut/sal 65.4 (9.1), sal 65.3 (8.8)

Smoking history (mean (SD) pack‐years): flut/sal 57.8 (32.7), sal 56.5 (27.8)

Mean % predicted pre‐FEV1 (SD): 34.1 (11.1), 33.9 (10.6)

Inclusion criteria: ≥ 40 years of age with a diagnosis of COPD (chronic bronchitis and/or emphysema), a cigarette smoking history ≥ 10 pack‐years, a prealbuterol FEV1/FVC ≤ 0.70, an FEV1 ≤ 50% of predicted normal and a documented history of at least one COPD exacerbation the year before the study that required treatment with antibiotics or oral corticosteroids and/or hospitalisation
Exclusion criteria: Individuals were excluded if they had a current diagnosis of asthma, a respiratory disorder other than COPD, historical or current evidence of a clinically significant uncontrolled disease or a COPD exacerbation that was not resolved at screening

Interventions

Run‐in: Four‐week run‐in period during which participants received open‐label FSC 250/50 via DISKUS twice daily

Treatments:

1. Fluticasone/salmeterol 250/50 mcg twice daily

2. Salmeterol 50 mcg twice daily

Inhaler device: Diskus

Co‐treatment: Concurrent use of inhaled long‐acting bronchodilators was not allowed during the study period. Oral corticosteroids and antibiotics were allowed for short‐term treatment of a COPD exacerbation

Outcomes

Primary: annual rate of moderate/severe exacerbations

Secondary: time to first moderate/severe exacerbation, annual rate of exacerbations requiring oral corticosteroids, predose FEV1, time to onset of each moderate/severe exacerbation, diary records of dyspnoea scores, night‐time awakenings due to COPD and use of supplemental albuterol

Notes

Funding: GSK

Clinicaltrials.gov: NCT00115492

Definition of pneumonia: confirmed by chest x‐ray

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Treatments were assigned in blocks using a centre‐based randomisation schedule. As bronchodilator response to FSC 250/50 is generally larger in individuals with COPD who demonstrate FEV1 reversibility to albuterol, assignment to blinded study medication was stratified on the basis of participants' FEV1 response to albuterol at screening to provide a similar distribution of albuterol‐responsive and non‐responsive participants in each treatment group

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (assumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Withdrawal rates were very high compared with the numbers of events for different outcomes

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported and could be included

Bourbeau 1998

Methods

Design: double‐blind, randomised, parallel‐group trial of high‐dose inhaled budesonide versus placebo

Six‐month treatment period (originally intended as 12 months)

Conducted at a single centre in Canada

Participants

Participants: 79 people were randomly assigned to budesonide (39) and placebo (40)
Baseline characteristics:

Male %: bud 84.6, placebo 72.5

Mean age (SD), years: bud 66 (8), placebo 66 (8)

Smoking history (mean (SD) pack‐years): bud 52 (27), placebo 50 (28)

Mean % predicted pre‐FEV1 (SD): bud 36 (12), placebo 37 (10)

Inclusion criteria: 40 years of age or older; smokers or ex‐smokers; absence of an exacerbation in
respiratory symptoms during the two months before the study; pre‐bronchodilator FEV1 less than 65% of predicted and FEV1/forced vital capacity (FVC) less than 0.65; postbronchodilator FEV1 less than 80%; regular treatment with at least one bronchodilator
Exclusion criteria: history of allergic asthma during childhood or as an adult; inhaled corticosteroids in the previous month or oral corticosteroids in the previous two months; any other active lung disease; diabetes, active peptic ulcer disease, uncontrolled high blood pressure or congestive heart failure; disease other than COPD that might interfere with quality of life

Interventions

Run‐in: All participants were assessed in a single‐blind manner with a two‐week course of oral placebo followed by two weeks of prednisone 40 mg daily. The prednisone was subsequently tapered and was discontinued completely during the third week. Those who did not respond were randomly assigned

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: Turbohaler

Co‐treatment: All medications needed for the well‐being of participants were permitted, except inhaled corticosteroids other than budesonide. In cases of treatment failure, rescue medication with beta2‐agonists or systemic steroids was available

Outcomes

Primary: change in FEV1

Secondary: pre‐bronchodilator and postbronchodilator FEV1 and FVC, pre‐bronchodilator six‐minute walking test, dyspnoea with exercise, quality of life questionnaires, morning and evening PEFR, symptom scores and adverse events

Notes

Funding: Astra Pharma Inc

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central computer‐generated list of random numbers

Allocation concealment (selection bias)

Low risk

Identification of individual treatment assignments was possible only in cases of emergency by breaking the sealed envelope kept by the investigator. The envelopes had to be kept with the case record forms and had to be returned unbroken at the end of the study

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

To ensure that outcomes were measured similarly in the treatment groups, both participants and investigators were blinded to the study treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

To ensure that outcomes were measured similarly in the treatment groups, both participants and investigators were blinded to the study treatment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Uneven dropout. Much higher in placebo group (25% vs 7.7% in the ICS group)

Selective reporting (reporting bias)

High risk

Key outcomes missing (mortality, adverse events). No reply from study author

Bourbeau 2007

Methods

Design: randomised, double‐blind, parallel‐group, placebo‐controlled trial

Three‐month treatment period

Conducted at two respiratory centres in Canada

Participants

Participants: 41 people were randomly assigned to fluticasone (20) and placebo (21)
Baseline characteristics:

Male %: flut 84.6, placebo 72.5

Mean age (SD), years: flut 66 (8), placebo 66 (8)

Smoking history (mean (SD) pack‐years): flut 52 (27), placebo 50 (28)

Mean % predicted FEV1 (SD): flut 36 (12), placebo 37 (10)

Inclusion criteria: Age > 40 and < 75 years; smoking history (> 10 pack‐years); postbronchodilator FEV1 > 25% of predicted
value and FEV1/forced vital capacity (FVC) < 0.70
Exclusion criteria: history of asthma, atopy (as assessed by an allergy skin prick test during screening) or any other active lung disease. Individuals receiving home oxygen or with raised carbon dioxide tension (.44 mm Hg), α1‐antitrypsin deficiency, recent exacerbation (in the last 4 weeks), uncontrolled medical condition or hypersensitivity to inhaled corticosteroids and bronchodilators were not eligible for the study

Interventions

Run‐in: unclear duration

Treatments:

1. Fluticasone 500 mcg twice daily

2. Placebo twice daily

Participants were also randomly assigned to fluticasone/salmeterol combination, but this was not included in the present review because no salmeterol arm was available for comparison

Inhaler device: Diskus

Co‐treatment: short‐acting bronchodilators, short‐ and long‐acting anticholinergics or theophylline was allowed throughout the study. Oral corticosteroids and/or antibiotics could be given only in short courses for exacerbation treatment

Outcomes

Primary: treatment difference in the numbers of CD8+ T lymphocytes and CD68+ macrophages on bronchial biopsies

Secondary: numbers of neutrophils and eosinophils in bronchial biopsies

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central computer‐generated list of random numbers

Allocation concealment (selection bias)

Low risk

Set up by a data management/randomisation company (GEREQ, Montreal, Quebec). A procedure was established by GEREQ, which was in possession of the treatment code, to ensure that the treatment code would be broken only in accordance with the protocol and the criteria set up for unbinding of the study

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Observers and participants were blinded to drug treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Observers were blinded to whether the biopsies were performed post‐treatment or pretreatment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout uneven but low in both groups (5% ICS and 14% placebo)

Selective reporting (reporting bias)

High risk

Key outcomes not reported. No reply from study author

Burge 2000

Methods

Design: double‐blind, placebo‐controlled study

Three‐year treatment period

Conducted at 18 hospitals in the UK

Participants

Participants: 740 people were randomly assigned to fluticasone (372) and placebo (370)Baseline characteristics:

Male %: flut 75.0, placebo 74.1

Mean age (SD), years: flut 63.7 (7.1), placebo 63.8 (7.1)

Smoking history (mean (SD) pack‐years): flut 44 (30), placebo 44 (34)

Mean % predicted FEV1 (SD): flut 50.3 (14.9), placebo 50.0 (14.9)

Inclusion criteria: current or former smokers 40 to 75 years of age with non­asthmatic chronic obstructive pulmonary disease. Baseline FEV1 after bronchodilator was at least 0.8 L but less than 85% of predicted normal, and the ratio of FEV1 to forced vital capacity was less than 70%. Previous use of inhaled and oral corticosteroids was permitted
Exclusion criteria: Individuals were excluded if their FEV1 response to 400 mcg salbutamol exceeded 10% of predicted normal, if they had a life expectancy of less than five years from concurrent disease or if they used beta blockers

Interventions

Run‐in: eight‐week run­in period after withdrawal from any oral or inhaled corticosteroids

Treatments:

1. Fluticasone propionate 500 mcg daily

2. Placebo twice daily

Inhaler device: metered‐dose inhaler with a spacer device

Co‐treatment: Nasal and ophthalmic corticosteroids, theophyllines and all other bronchodilators were allowed during the study

Outcomes

Primary: decline (mL/y) in FEV1 after bronchodilator

Secondary: frequency of exacerbations, changes in health status, withdrawals due to respiratory disease, morning serum cortisol concentrations and adverse events

Notes

Funding: GlaxoWellcome Research and Development

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated allocation (block size of six)

Allocation concealment (selection bias)

Low risk

Participants were randomly assigned sequentially from a list comprising treatment numbers only

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Three‐year double‐blind phase using an identical placebo inhaler

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Very high dropout in both groups (43% and 53%)

Selective reporting (reporting bias)

Unclear risk

No outcomes appear to be missing, but protocol could not be located to ensure that all were reported. Author attempted contact with GSK statistician, but no data were provided in time for publication

Calverley 2003 TRISTAN

Methods

Design: multi‐centre, randomised, double‐blind, parallel‐group, placebo‐controlled study

12‐Month treatment period

Conducted at 196 hospitals in 25 countries

Participants

Participants: 1465 people were randomly assigned to fluticasone/salmeterol combination (358), fluticasone (374), salmeterol (372) and placebo (361)
Baseline characteristics:

Male %: flut/salm 75, flut 70, salm 70, placebo 75

Mean age (SD), years: flut/salm 62.7 (8.7), flut 63.5 (8.5), salm 63.2 (8.6), placebo 63.4 (8.6)

Smoking history (mean (SD) pack‐years): flut/salm 42.0 (22.4), flut 41.5 (20.7), salm 43.7 (21.9), placebo 43.4 (22.4)

Mean % predicted FEV1 (SD): flut/salm 44.8 (14.7), flut 45.0 (13.6), salm 44.3 (13.8), placebo 44.2 (13.7)

Inclusion criteria: All participants had a baseline FEV1 before bronchodilation that was 25% to 70% of that predicted, an increase of less than 10% of predicted FEV1 30 minutes after inhalation of 400 mcg salbutamol and a pre‐bronchodilator FEV1/forced vital capacity (FVC) ratio of 70% or less. Participants also had a history of at least 10 pack‐years of smoking, chronic bronchitis, at least one episode of acute COPD symptom exacerbation per year in the previous 3 years and at least one exacerbation in the year immediately before trial entry that required treatment with oral corticosteroids, antibiotics or both
Exclusion criteria: We excluded individuals who had respiratory disorders other than COPD, required regular oxygen treatment or had received systemic corticosteroids, high doses of inhaled corticosteroids (> 1000 mcg daily beclomethasone dipropionate, budesonide or flunisolide, or > 500 mcg daily fluticasone) or antibiotics in the 4 weeks before the 2 week run‐in period before the trial began

Interventions

Run‐in: During the 2‐week run‐in, participants stopped taking regular inhaled corticosteroids or long‐acting beta2‐agonists

Treatments:

1. Fluticasone/salmeterol 500/50 mcg twice daily

2. Fluticasone 500 mcg twice daily

3. Salmeterol 50 mcg twice daily

4. Placebo twice daily

Inhaler device: multi‐dose dry powder inhaler (Diskus or Accuhaler)

Co‐treatment: Inhaled salbutamol was used as relief medication throughout the study, and regular treatment with anticholinergics, mucolytics and theophylline was allowed. All non‐COPD medications could be continued if the dose remained constant when possible, and if their use would not be expected to affect lung function

Outcomes

Primary: FEV1 at least 6 and 12 hours after study medication

Secondary: pretreatment FVC and post‐treatment FEV1 and FVC, daily record card symptoms, morning and evening PEF, use of relief medication, night‐time awakenings, acute exacerbations, health‐related quality of life (SGRQ), adverse events and electrocardiology

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A randomisation schedule generated by the participant allocation for clinical trials (PACT) programme to assign participants to study treatment groups

Allocation concealment (selection bias)

Unclear risk

Every participating centre was supplied with a list of participant numbers (assigned to participants at their first visit) and a list of treatment numbers. Participants who satisfied the eligibility criteria were assigned the next sequential treatment number from the list

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Salmeterol and fluticasone combination, salmeterol, fluticasone and placebo were packaged in identical inhaler devices

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Adverse event information was obtained at each clinic visit by recording spontaneously reported complaints from participants and asking general questions about medical troubles and concomitant medications

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawal rates varied from 25% in the combination inhaler group to 39% in the placebo group. These numbers are substantially higher than the numbers of events reported for any of the outcomes. The unknown outcome of participant withdrawal from the study could have a major impact on the result

Selective reporting (reporting bias)

Low risk

All outcomes reported. Checked with study authors

Calverley 2003b

Methods

Design: multi‐centre, randomised, double‐blind, parallel‐group, placebo‐controlled study

12‐Month treatment period

Conducted at 109 centres in 15 countries

Participants

Participants: 1022 people were randomly assigned to budesonide/formoterol combination (254), budesonide (257), formoterol (255) and placebo (256)
Baseline characteristics:

Male %: bud/form 78, bud 74, form 75, placebo 75

Mean age (range), years: bud/form 64 (42 to 86), bud 64 (41 to 85), form 63 (41 to 84), placebo 65 (43 to 85)

Smoking history (mean pack‐years): bud/form 33, bud 39, form 36, placebo 30

Mean % predicted FEV1 (SD): bud/form 36 (10), bud 36 (10), form 36 (10), placebo 36 (10)

Inclusion criteria: GOLD‐defined COPD (stages III and IV); ≥ 40 years; COPD symptoms > 2 years; smoking history ≥ 10 pack‐years; FEV1/FVC ≤ 70% pre‐bronchodilator; FEV1 ≤ 50% predicted; use of short‐acting beta2‐agonists as reliever medication; ≥ 1 COPD exacerbation requiring oral corticosteroids/antibiotics 2 to 12 months before first clinic visit
Exclusion criteria: history of asthma/rhinitis before 40 years of age; any relevant cardiovascular disorders; exacerbation of COPD requiring medical intervention within 4 weeks of run‐in/during run‐in phase; non‐allowed medications: oxygen therapy; ICS (aside from study medication), disodium cromoglycate, leukotriene‐antagonists, 5‐lipoxygenase inhibitors, bronchodilators (other than study medication and terbutaline 0.5 mg as needed), antihistamines, medication containing ephedrine, beta‐blocking agents

Interventions

Run‐in: All participants received 30 mg oral prednisolone twice daily and 2 × 4.5 mg formoterol twice daily (2 weeks)

Treatments:

1. Budesonide/formoterol 320/9 mcg twice daily

2. Budesonide 400 mcg twice daily

3. Formoterol 9 mcg twice daily

4. Placebo twice daily

Inhaler device: Turbuhaler

Co‐treatment: terbutaline 0.5 mg as needed, courses of oral corticosteroids (maximum 3 weeks per course) and antibiotics in the event of exacerbations, parenteral steroids and/or nebulised treatment (single injections/inhalations) at emergency visits

Outcomes

Primary: time to first exacerbation and change in postmedication FEV1

Secondary: number of exacerbations, time to and number of oral corticosteroid–treated episodes, change in postdose FEV1, slow VC, morning and evening PEF, quality of life (SGRQ), symptoms, use of reliever medication and adverse events

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised trial (no other details—funded by AstraZeneca and presumed to adhere to usual methods)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind (assume participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

An intention‐to‐treat analysis was used but dropout was high in all groups (ranging from 29% to 44%)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the industry document were reported in full. Checked with study authors

Calverley 2007 TORCH

Methods

Design: multi‐centre, double‐blind, placebo‐controlled, randomised, parallel‐group study

Three‐year treatment period

Conducted at 444 centres in 42 countries

Participants

Participants: 6184 people were randomly assigned to fluticasone/salmeterol combination (1546), fluticasone (1551), salmeterol (1542) and placebo (1545)
Baseline characteristics:

Male %: flut/salm 75, flut 75, salm 76, placebo 76

Mean age (SD), years: flut/salm 65.0 (8.3), flut 65.0 (8.4), salm 65.1 (8.2), placebo 65.0 (8.2)

Smoking history (mean (SD) pack‐years): flut/salm 47.0 (26.5), flut 49.2 (28.6), salm 49.3 (27.7), placebo 48.6 (26.9)

Mean % predicted FEV1 (SD): flut/salm 44.3 (12.3), flut 44.1 (12.3), salm 43.6 (12.6), placebo 44.1 (12.3)

Inclusion criteria: current or former smokers with at least a 10‐pack‐year history. Eligible individuals were 40 to 80 years of age and had received a diagnosis of COPD, with a pre‐bronchodilator forced expiratory volume in one second (FEV1) of less than 60% of predicted value, an increase in FEV1 with the use of 400 mcg of albuterol of less than 10% of the predicted value for that individual and a ratio of pre‐bronchodilator FEV1 to forced vital capacity (FVC) equal to or less than 0.70
Exclusion criteria: diagnosis of asthma, current respiratory disorders other than COPD (e.g. lung cancer, sarcoidosis, tuberculosis, lung fibrosis); chest x‐ray indicating diagnosis other than COPD that might interfere with the study (chest x‐ray to be taken up to six months before entry to the treatment period); lung volume reduction surgery and/or lung transplant, requirement for long‐term oxygen therapy (LTOT is defined as oxygen therapy prescribed for 12 or more hours per day) at start of study; receiving long‐term oral corticosteroid therapy defined as continuous use for longer than 6 weeks; serious, uncontrolled disease (including serious psychological disorders) likely to interfere with the study and/or likely to cause death within the 3‐year study duration; received any other investigational drugs in the last 4 weeks before entry to visit one; evidence of alcohol, drug or solvent abuse; known or suspected hypersensitivity to inhaled corticosteroids, bronchodilators or lactose; known deficiency of α1‐antitrypsin

Interventions

Run‐in: Before the 2 week run‐in period, all use of corticosteroids and inhaled long‐acting bronchodilators was stopped, but participants could continue other medications for COPD

Treatments:

1. Fluticasone/salmeterol 500/50 mcg twice daily

2. Fluticasone 500 mcg twice daily

3. Salmeterol 50 mcg twice daily

4. Placebo twice daily

Inhaler device: multi‐dose dry powder inhaler (Diskus or Accuhaler)

Co‐treatment: all COPD medications except corticosteroids and inhaled long‐acting bronchodilators

Outcomes

Primary: time to death from any cause by three years

Secondary: frequency of exacerbations, health status (SGRQ), postbronchodilator spirometry and adverse events

Notes

Funding: GSK

Clinicaltrials.gov ID: NCT00268216

Definition of pneumonia: Study authors state there was no prospective definition because the finding was unexpected

NOTE: Participants previously enrolled into TRISTAN (SFCB3024) may be recruited to this trial 4 weeks after stopping their previous study medication

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random assignment with permuted blocks with stratification

Allocation concealment (selection bias)

Low risk

Medication was allocated with the use of three numbers as follows.

  1. Each participant who was screened was allocated a participant number. This number was unique to each participant and was assigned from a list provided to the site, in chronological order

  2. Each participant who satisfied the randomisation criteria was assigned a unique treatment number from the interactive voice response (IVR) system, which is part of the system for central allocation of drug (SCAD). Once a treatment number had been assigned to a participant, it could not be assigned to any other participant. Neither the participant nor the investigator knew to which treatment arm a participant had been allocated

  3. At each treatment visit, the participant was provided with a treatment pack. Each pack number was unique and corresponded to the study medication pack dispensed to the participant at the visit

 

A specialist IVR system company, ClinPhone, managed this system. At the randomisation visit (visit two), the principal investigator or designee contacted the IVR system through an automated 24‐hour telephone number; upon provision of a unique personal identification number (PIN) and answers to a series of questions, the site was provided with the participant's treatment number as well as a pack number

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Neither the subject nor the investigator knew to which treatment arm a participant had been allocated. At each treatment visit, each participant was issued with a treatment pack containing DISKUS/ACCUHALER inhalers. The inhalers contained one of the four treatments (salmeterol/fluticasone propionate combination product, fluticasone propionate, salmeterol or placebo) in accordance with the randomisation schedule. The inhalers were labelled in accordance with all applicable regulatory requirements. Each treatment pack and study treatment inhaler was labelled with the protocol number; storage and dosing instructions were provided by GW Research and Development

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The investigator is responsible for the detection and documentation of events meeting the definition of an AE or SAE as provided in this protocol. However, no prospective definition of pneumonia is provided in the study protocol

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Outcome all‐cause mortality: Participants who prematurely discontinue study drug will be followed up for 156 weeks from randomisation for assessment of survival. The risk of bias due to incomplete outcome data for mortality is low

General

Withdrawal rates were high but fairly even (with the exception of the placebo group, which had an even higher withdrawal rate; placebo 44%, LABA 36%, ICS 38% and ICS/LABA 34%). However, for participants who withdrew from the study prematurely, all data on exacerbations, health status and lung function available at the time of a participant's withdrawal from the study were included in the analysis. All efficacy analyses were performed according to the intention‐to‐treat principle

Selective reporting (reporting bias)

Low risk

All collected data reported. Checked with study authors

Calverley 2010

Methods

Design: double‐blind, double‐dummy, randomised, active‐controlled, parallel‐group study

11‐Month treatment period (48 weeks)

Conducted at 76 centres in 8 countries across Europe

Participants

Participants: 481 people were randomly assigned to budesonide/formoterol combination (242) and formoterol (239)
Baseline characteristics:

Male %: bud/form 81.5, form 81.1

Mean age (SD), years: bud/form 64.1 (9.1), form 63.7 (8.8)

Smoking history (mean (SD) pack‐years): bud/form 37.8 (14.6), form 39.7 (19.1)

Mean % predicted FEV1 (SD): bud/form 42.3 (6.0), form 42.5 (5.9)

Inclusion criteria: hospital outpatients with severe stable COPD according to the GOLD guidelines; 40 years of age with a diagnosis of symptomatic COPD for > 2 years, at least a 20‐pack‐year smoking history, a postbronchodilator FEV1 between 30% and 50% of predicted normal and at least 0.7 L absolute value and a predose FEV1/forced vital capacity (FVC) of 0.7; at least one exacerbation requiring medical intervention (oral corticosteroid and/or antibiotic treatment and/or need for a visit to an emergency department and/or hospitalisation) within 2 to 12 months before the screening visit and the need to be clinically stable for the 2 months before study entry; change in FEV1 < 12% of predicted normal value 30 minutes following inhalation of 200 mg of salbutamol pMDI
Exclusion criteria: history of asthma, allergic rhinitis or other atopic disease, variability of symptoms from day to day and frequent symptoms at night and early morning (suggestive of asthma); receiving long‐term oxygen therapy or having a lower respiratory tract infection or having been hospitalised for an acute COPD exacerbation within two months before screening or during the run‐in period. Treatments with oral, injectable or depot corticosteroids and antibiotics, long‐acting antihistamines and changes in the dose of an oral modified‐release theophylline in the two months before screening and during the run‐in period were excluded

Interventions

Run‐in: During the 4‐week run‐in period, all non‐permitted COPD treatments were discontinued and eligible participants were treated with combination ipratropium/salbutamol (20/100 mg, two inhalations three times daily)

Treatments:

1. Budesonide/formoterol 400/12 mcg twice daily

2. Formoterol 12 mcg twice daily

Inhaler device: dry powder inhaler

Co‐treatment: not described

Outcomes

Primary: change in predose morning FEV1 and mean rate of COPD exacerbations per participant per year

Secondary: FVC, PEF, SGRQ total score, six‐minute walking test, BMI, BODE index, safety evaluations including ECG

Notes

Funding: Chiesi Farmaceutici

Clinicaltrials.gov ID: NCT00476099

Definition of pneumonia: not defined

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation scheme followed a balanced‐block centre‐stratified design and was prepared via a computerised system

Allocation concealment (selection bias)

Low risk

Participants were centrally assigned, in each centre, to one of the three treatment arms at the end of the run‐in period through an Interactive voice/web response System (IXRS)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

On each study day, participants took both active medications and matched placebo twice daily, to maintain blinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

On each study day, participants took both active medications and matched placebo twice daily, to maintain blinding. In cases of emergency, unbinding of the treatment code was done through IXRS

Incomplete outcome data (attrition bias)
All outcomes

Low risk

12.3% withdrew from the combination group and 14.2% from the formoterol group. Judged to be relatively low and even between groups, and for the intention‐to‐treat population, last observation carried forward was used

Selective reporting (reporting bias)

Low risk

All outcomes stated in the prospectively registered protocol were reported in full

Choudhury 2005

Methods

Design: randomised, double‐blind, placebo‐controlled trial

12‐Month treatment period in total, but participants/clinicians could stop study inhalers and return to usual (prerandomisation) steroid inhalers at any point during the study. These participants remained in the study, continued completing their diary cards and were followed up

Conducted at 31 general practices in East London and Essex, UK

Participants

Participants: 260 people were randomly assigned to fluticasone (128) and placebo (132)
Baseline characteristics:

Male %: flut 48, placebo 56

Mean age (SD), years: flut 67.6 (8.9), placebo 67.3 (9.0)

Smoking history (mean (SD) pack‐years): flut 40.0 (24.2), placebo 38.8 (22.3)

Mean % predicted FEV1 (SD): flut 53.2 (18.2), placebo 55.0 (17.1)

Inclusion criteria: Investigators searched the medical record database at each practise to identify people 40 years of age and older, with a history of smoking, who had been prescribed ICS for a minimum of six months. We invited people who fulfilled these criteria to attend a recruitment interview. Individuals with lung function consistent with international guidelines for the diagnosis of COPD were invited to join the study: postbronchodilator FEV1 less than 80% predicted, FEV1/FVC ratio less than 70% and a pre‐bronchodilator to postbronchodilator change in FEV1 of less than 15%. Participants with an FEV1 greater than 15% but with a volume change of less than 200 mL were also included
Exclusion criteria: At interview, we excluded people if they were on long‐term oral corticosteroids, were not taking their prescribed ICS for at least four days a week or had other chronic active lung disease or lung cancer

Interventions

Run‐in: two‐week run‐in period before randomisation, when participants stopped their regular ICS

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo twice daily

Inhaler device: Accuhaler

Co‐treatment: General practitioners were advised to manage exacerbations according to usual guidance with antibiotics and/or oral steroids. Decisions about stopping study inhalers and returning to usual (prerandomisation) steroid inhalers were made by the general practitioner and the participant. Participants who did return to their usual steroid inhaler after randomisation remained in the study, continued completing their diary cards and were followed up for one year

Outcomes

Primary: COPD exacerbation frequency

Secondary: time to first exacerbation (from diary cards and medical records), reported symptoms, peak expiratory flow rate and reliever inhaler use (from diary cards) and lung function and health=related quality of life (at follow‐up visits)

Notes

Funding: The British Lung Foundation and Newham National Health Service Trust Research and Development funded the study. GlaxoSmithKline provided the study inhalers free of charge but was not involved in study design, data collection or analysis or interpretation of results

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were allocated with minimisation to intervention or control using the programme MINIM v1.3. Minimisation factors were age, smoking status, pretrial weekly dose of ICS, self reported COPD exacerbation frequency and percentage predicted FEV1

Allocation concealment (selection bias)

Low risk

Inhalers were given an alphanumerical code to conceal allocation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study nurses and regular clinicians were blind to allocation throughout the study. Inhalers were given an alphanumerical code to conceal allocation

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Study nurses performed the measurements and were blind to allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Decisions about stopping study inhalers and returning to usual (prerandomisation) steroid inhalers were made by the general practitioner and the participant. Participants who did return to their usual steroid inhaler after randomisation remained in the study, continued completing their diary cards and were followed up for one year

Selective reporting (reporting bias)

High risk

Key outcomes were not reported (e.g. total mortality, breakdown of all and serious adverse events). No reply from study authors

Dal Negro 2003

Methods

Design: randomised, double‐blind, parallel‐group, pilot study

12‐Month treatment period

Conducted at a single centre in Italy

Participants

Participants: 12 people were randomly assigned to fluticasone/salmeterol combination (6) and salmeterol alone (6)
Baseline characteristics:

Male %: flut/salm 83.3, salm 100

Age range (mean not reported), years: flut/salm 53 to 77, salm 55 to 78

Smoking history (mean (SD) pack‐years): flut/salm 40.1 (6.3), salm 43.1 (5.3)

Mean % predicted FEV1 (SD): flut/salm 50 (2.0), salm 48 (5.6)

Inclusion criteria: basal FEV1 < 80% predicted normal value, but > 800 mL; FEV1/FVC ratio < 70% predicted; FEV1 change of < 12% as a percentage of predicted normal
value following salbutamol 400 mg; regular treatment with oral theophylline 200 mg bid and short‐acting beta2‐adrenergics prn for a period of at least six months; current smokers or ex‐smokers with a smoking history of at least 10 pack‐years
Exclusion criteria: current evidence of asthma or other pulmonary diseases; regular treatment with ICS; unstable respiratory disease requiring oral/parenteral corticosteroids within four weeks before the beginning of the study; changes in COPD medication in the last four weeks before entering the run‐in period; upper or lower respiratory tract infection within four weeks before the screening visit; unstable angina or unstable arrhythmias; recent myocardial infarction or hearth failure; insulin‐dependent diabetes mellitus; neuropsychiatric disorders; concurrent use of medications that affected COPD (e.g. beta‐blockers) or interacted with methylxanthine products such as macrolides or fluoroquinolones; known or suspected hypersensitivity to ICS, beta2‐agonist or lactose; evidence of alcohol abuse

Interventions

Run‐in: Patients entered a two‐week run‐in period during which they assumed their regular treatment with theophylline and salbutamol as required

Treatments:

1. Fluticasone/salmeterol 250/50 mcg twice daily

2. Salmeterol 50 mcg twice daily

Participants were also randomly assigned to placebo (6), but this group was not included in the current review because no group received fluticasone monotherapy

Inhaler device: Diskus

Co‐treatment: theophylline and salbutamol as required

Outcomes

Unclear which outcome was primary

Exacerbations per year, FEV1, morning PEF, daily symptom scores, use of rescue medication and adverse events

Notes

Funding: unclear

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

At the end of the run‐in, eligible participants will be randomly assigned to receive one of the three double‐blind treatments (no other details and does not appear to be industry funded)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Assigned to receive one of the three double‐blind treatments, all via Diskus

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropout

Selective reporting (reporting bias)

High risk

Several key outcomes not reported (mortality, adverse events). Difficulty contacting study authors

Dransfield 2013

Methods

Design: two replicate double‐blind parallel‐group trials

12‐Month treatment period

Study one was conducted at 167 centres in 15 countries (Argentina, Australia, Canada, Chile, Estonia, Germany, Italy, Mexico, Netherlands, Peru, Phillipines, South Africa, Sweden, the United Kingdom and the United States). Study two was conducted at 183 centres in 15 countries

Participants

Participants: 1622 people were randomly assigned in study one and 1633 in study two to vilanterol 25 (818), fluticasone/vilanterol combination 50/25 (820), 100/25 (806) and 200/25 (811)
Baseline characteristics:

Male %: Vil 58.0, F/Vil 50/25 58.0, F/Vil 100/25 56.2, F/Vil 200/25 57.6

Mean age, years: Vil 63.6, F/Vil 50/25 63.7, F/Vil 100/25 63.8, F/Vil 200/25 63.7

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1: Vil 45.2, F/Vil 50/25 45.4, F/Vil 100/25 46.1, F/Vil 200/25 45.2

Inclusion criteria: Eligible patients were 40 years of age or older and had a history of COPD, a smoking history of 10 or more pack‐years, a ratio of forced expiratory volume in one second (FEV1) to forced vital capacity of 0.70 or less after bronchodilators (and an FEV1 of 70% or less of predicted) and a documented history of one or more moderate or severe disease exacerbations in the year before screening
Exclusion criteria: Appendix listing the exclusion criteria and the drugs that were and were not permissible for the study duration could not be located

Interventions

Run‐in: four weeks during which participants received open‐label combination fluticasone propionate (250) and salmeterol (50) twice daily to establish adherence to treatment and a stable baseline

Treatments:

1. Fluticasone furoate/vilanterol 50/25 mcg daily

2. Fluticasone furoate/vilanterol 100/25 mcg daily

3. Fluticasone furoate/vilanterol 200/25 mcg daily

4. Vilanterol 25 daily

Inhaler device: dry powder inhaler

Co‐treatment: Appendix listing the exclusion criteria and the drugs that were and were not permissible for the study duration could not be located

Outcomes

Primary: annual rate of moderate and severe exacerbations

Secondary: time to first moderate or severe exacerbation; annual rate of exacerbations requiring oral corticosteroids; predose AM FEV1

Notes

Funding: GSK

Clinicaltrials.gov ID: NCT01009463 and NCT01017952

Definition of pneumonia: confirmed by chest x‐ray

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The central randomisation schedule was generated by the GSK statistics group, which used a validated computerised system (RandAll; GSK, London, UK)

Allocation concealment (selection bias)

Unclear risk

The statistician entered the parameters for the randomisation but was masked to treatment assignment and did not have access to the master randomisation schedule until the study was unmasked at database lock. The Registration and Medication Ordering System (RAMOS; GSK, London, UK) was used to register and randomly assign participants and assign drugs

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and investigators were masked to allocation, and the Ellipta dry powder inhalers were identical in appearance. Every effort was made to ensure that the statistics and programming department of GSK was not unmasked to any treatment allocations ahead of the database freeze

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants and investigators were masked to allocation, and the Ellipta dry powder inhalers were identical in appearance. Every effort was made to ensure that the statistics and programming department of GSK was not unmasked to any treatment allocations ahead of the database freeze

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout quite high but even between groups (ranging from 23% to 31%). A generalised linear model was used for the intention‐to‐treat analyses of each study, including all randomly assigned participants receiving at least one dose of study drug (representing 100% of those randomly assigned)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the prospectively registered protocol were reported in the published paper

Ferguson 2008

Methods

Design: randomised, double‐blind, parallel‐group study

12‐Month study period

Conducted at 94 research sites in the United States and Canada

Participants

Participants: 782 people were randomly assigned to fluticasone/salmeterol combination (394) and salmeterol alone (388)
Baseline characteristics:

Male %: flut/salm 58, salm 52

Mean age (SD), years: flut/salm 64.9 (9.0), salm 65.0 (9.1)

Smoking history (mean (SD) pack‐years): flut/salm 58.5 (30.6), salm 54.4 (25.7)

Mean % predicted FEV1 (SD): flut/salm 32.8 (11.0), salm 32.8 (10.1)

Inclusion criteria: 40 years of age or older with a diagnosis of COPD, a cigarette smoking history of greater than or equal to 10 pack‐years, a prealbuterol FEV1/FVC of 0.70 or less, an FEV1 of 50% of predicted normal or less and a history of 1 or more exacerbations of COPD in the year before the study, which required treatment with oral corticosteroids or antibiotics, or hospitalisation
Exclusion criteria: diagnosis of asthma, a significant lung disease other than COPD, a clinically significant and uncontrolled medical disorder including but not limited to cardiovascular, endocrine or metabolic, neurological, psychiatric, hepatic, renal, gastric and neuromuscular diseases, or had a COPD exacerbation that was not resolved at screening

Interventions

Run‐in: four‐week run‐in period during which participants received open‐label fluticasone/salmeterol 250/50 via Diskus twice daily

Treatments:

1. Fluticasone/salmeterol 250/50 mcg twice daily

2. Salmterol 50 mcg twice daily

Inhaler device: Diskus

Co‐treatment: As‐needed albuterol was provided for use throughout the study. Concurrent inhaled long‐acting bronchodilators (beta2‐agonist and anticholinergic), ipratropium/albuterol combination products, oral beta‐agonists, inhaled corticosteroids and theophylline preparations were not allowed during the treatment period. Oral corticosteroids and antibiotics were allowed for the short‐term treatment of COPD exacerbations

Outcomes

Primary: annual rate of moderate to severe exacerbations

Secondary: time to first moderate to severe exacerbation, annual rate of exacerbations requiring oral corticosteroids, predose FEV1, diary records of dyspnoea, night‐time awakenings due to COPD and use of supplemental albuterol

Notes

Funding: GSK (ID SCO40043)

Clinicaltrials.gov ID: NCT00144911

Definition of pneumonia: no objective definition given

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centre‐based randomisation schedule

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropout high and fairly even (30% vs 38%). Method of imputation described only for the primary outcome (‘Endpoint was defined as the last scheduled measurement of predose AM FEV1 during the 52‐week treatment period’)

Selective reporting (reporting bias)

Low risk

Checked GSK documents—all stated and expected outcomes are reported

Fukuchi 2013

Methods

Design: randomised, double‐blind, parallel‐group study

Three‐month study period

Conducted at 163 centres in 9 countries (India, Japan, Republic of Korea, Phillipines, Poland, Russian Federation, Taiwan, Ukraine and Vietnam)

Participants

Participants: 1293 people were randomly assigned to budesonide/formoterol combination (636) and formoterol alone (657)
Baseline characteristics:

Male %: bud/form 87.6, form 90.3

Mean age (range), years: bud/form 64.5 (40 to 89), form 65.6 (40 to 87)

Smoking history (mean (range) pack‐years): bud/form 43.4 (10 to 160), form 44.7 (0 to 300)

Mean % predicted FEV1 (range): bud/form 40.9 (12 to 79), form 40.8 (8‐84)

Inclusion criteria: male and female individuals, 40 years of age or older with a diagnosis of moderate to severe COPD for at least 2 years (pre‐bronchodilator forced expiratory volume in one second (FEV1) 50% of predicted normal or less, postbronchodilator FEV1/forced vital capacity (FVC) < 70%), current or previous smoking history of 10 or more pack‐years and having at least one COPD exacerbation in 12 months
Exclusion criteria: history or current clinical diagnosis of asthma or atopic disease such as allergic rhinitis; significant or unstable ischaemic heart disease, arrhythmia, cardiomyopathy, heart failure, uncontrolled hypertension or any other relevant cardiovascular disorder; experiencing a COPD exacerbation during the run‐in period or within 4 weeks before randomisation that required hospitalisation and/or a course of oral or parenteral steroids; and requiring regular oxygen therapy

Interventions

Run‐in: 1‐ to 2‐week run‐in period during which participants received open‐label formoterol 4.5 mg two inhalations twice daily and all other COPD medications were discontinued, with the exception of salbutamol 100 mg/actuation via pMDI

Treatments:

1. Budesonide/formoterol combination 320/9 mcg twice daily

2. Formoterol 9 mcg twice daily

Inhaler device: Turbuhaler

Co‐treatment: Salbutamol 100 mg/actuation was available as reliever medication throughout the treatment period. During the randomised treatment period, participants were not permitted to take any other medication for their COPD, including beta2‐agonists, anticholinergics, leukotriene receptor antagonists, medications containing ephedrine or xanthine‐containing derivatives or inhaled corticosteroids

Outcomes

Primary: change in predose FEV1 from baseline to treatment period

Secondary: 1 hour postdose FEV1, predose and one hour postdose FVC, COPD symptoms, time to first COPD exacerbation, number of COPD exacerbations, health‐related quality of life on the SGRQ, morning and evening PEF, adverse events and vital signs

Notes

Funding: AstraZeneca (ID: D589DC00007)

Clinicaltrials.gov ID: NCT01069289

Definition of pneumonia: pneumonia, brochopneumonia and bacterial pneumonia counted as serious adverse events. Diagnosis not given

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned 1:1 to study treatment. No details of sequence generation methods, but presumed to adhere to usual AstraZeneca methods

Allocation concealment (selection bias)

Unclear risk

No details

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

From clinicaltrials.gov: masking: double‐blind (participant, caregiver, investigator, outcomes assessor)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

From clinicaltrials.gov: masking: double‐blind (participant, caregiver, investigator, outcomes assessor)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout low and even between groups

Selective reporting (reporting bias)

Low risk

All outcomes were fully reported on clinical.trials.gov and in the published report

GSK FCO30002 2005

Methods

Design: multi‐centre, randomised, placebo‐controlled, double‐blind trial

Three‐month treatment period

Conducted at 32 centres in Germany

Participants

Participants: 140 people were randomly assigned to salmeterol plus fluticasone (68) and salmeterol plus placebo (69)
Baseline characteristics:

Male %: flut+salm 60.6, salm+placebo 71.2

Mean age (SD), years: flut+salm 61 (8), salm+placebo 63 (10)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: documented history of COPD; male and female subjects 40 to 79 years of age; smokers and ex‐smokers with a smoking history of ≥ 10 pack‐years; FEV1 40% to 80% of predicted, FEV1/FVC < 70% at visit 1 or 2; low reversibility of airway obstruction at visit 1 or 2: increase in FEV1 (normal value) < 10% at 30 minutes after inhalation of 200 mcg salbutamol; symptomatic COPD during run‐in as documented in the participant diary: on ≥ 5 days, symptom score was > 5 and/or salbutamol inhalation; ability to correctly use the Mini‐Wright Peak‐Flow‐Meter and the Diskus™ inhaler
Exclusion criteria: long‐term oxygen therapy; use of inhaled or systemic corticosteroids during the 8 weeks before study entry; acute exacerbation, antibiotic treatment or hospital stay within 4 weeks before study entry; use of beta blockers within two weeks before study entry

Interventions

Run‐in: 2 weeks during which all participants received salmeterol 50 mcg twice daily as bronchodilator treatment and salbutamol as rescue medication

Treatments:

1. Salmeterol 50 mcg plus fluticasone 500 mcg twice daily (and placebo tablets for 2 weeks)

2. Salmeterol 50 mcg plus placebo inhaler twice daily (and placebo tablets for 2 weeks)

A third group receiving oral prednisolone in combination with salmeterol was not included in this review

Inhaler device: Diskus

Co‐treatment: salbutamol as rescue medication

Outcomes

Primary: change in FEV1

Secondary: Participants' self assessment of exercise capacity and morning serum cortisol concentrations

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised (presumed to adhere to usual GSK methodology)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout relatively low and even between groups (17.6% vs 14.5%). Full analysis set (FAS) and per‐protocol analyses were reported, but only the FAS was extracted

Selective reporting (reporting bias)

Unclear risk

All outcomes stated were reported. Pneumonia‐related outcomes were not reported. GSK data request was not successful at the time of publication

GSK FLTA3025 2005

Methods

Design: randomised, double‐blind, parallel‐group, comparative trial

Six‐month treatment period

Conducted at 55 centres in the United States

Participants

Participants: 640 people were randomly assigned to fluticasone 250 (216), fluticasone 500 (218) and placebo (206)
Baseline characteristics:

Male %: flut250 72.2, flut500 66.1, placebo 68.0

Mean age (SD), years: flut250 65.2 (8.7), flut500 63.3 (10.0), placebo 64.8 (9.5)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: Male or female individuals were eligible if they were diagnosed with COPD; were at least 40 years of age; had a current or prior history of at least 20 pack‐years of cigarette smoking; had a history of cough productive of sputum on most days for at least 3 months of the year, for at least 2 years that was not attributable to another disease process; had a baseline FEV1 < 65% of predicted normal but > 0.70 L or FEV1 ≤ 0.70 L and > 40% of predicted normal and FEV1/forced vital capacity (FVC) ratio of < 0.70; had a score of ≥ 2 on the Modified Medical Research Council (MMRC) Dyspnea Scale at screening and a score of ≥ 4 on the CBSQ at randomisation and had not received systemic corticosteroids or high‐dose inhaled corticosteroid therapy for at least 6 months before screening
Exclusion criteria: current diagnosis of asthma, concurrent participation in a pulmonary rehabilitation programme, respiratory disease other than COPD or other significant concurrent disease, an abnormal and clinically significant ECG at screening and the occurrence of a moderate or severe COPD exacerbation during the run‐in period

Interventions

Run‐in: 2‐week placebo run‐in period

Treatments:

1. Fluticasone propionate 250 mcg twice daily

2. Fluticasone propionate 500 mcg twice daily

3. Placebo twice daily

Inhaler device: Diskus

Co‐treatment: Concurrent use of the following respiratory medications was not allowed: beta‐agonists (other than salbutamol), cromolyns, corticosteroids (oral, inhaled and intranasal), anti‐leukotrienes and ipratropium. Concurrent use of theophylline was allowed. Use of antibiotics for the treatment of up to three COPD exacerbations was allowed

Outcomes

Primary: morning predose FEV1

Secondary: Chronic Bronchitis Symptoms Questionnaire (CBSQ), Transition Dyspnoea Index (TDI, exacerbations of COPD, participant‐recorded daily morning PEF rate, supplemental salbutamol use, night‐time awakenings and quality of life (CDRQ)

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised trial (GSK funded, likely to be computerised randomisation schedule)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded trial (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout high but even across groups. The intent‐to‐treat (ITT) population consisted of all randomly assigned participants who received at least 1 dose of study medication. The ITT population was the primary population for all efficacy and safety analyses

Selective reporting (reporting bias)

Low risk

All outcomes were reported in the results summary

GSK SCO100470 2006

Methods

Design: multi‐centre, randomised, double‐blind, double‐dummy, parallel‐group design

6 month treatment period

Conducted at 135 centres in 20 countries (Australia (10), Bulgaria (5), Croatia (1), Czech Republic (8), France (14), Germany (18), Greece (4), Italy (16), Latvia (5), Lithuania (2), Netherlands (12), Philippines (3), Poland (5), Romania (3), Russian Federation (8), Slovakia (4), Slovenia (4), Sweden (4), Thailand (4) and United Kingdom (5))

Participants

Participants: 1050 people were randomly assigned to fluticasone/salmeterol combination (518) and salmeterol alone (532)
Baseline characteristics:

Male %: flut/salm 78.4, salm 77.3

Mean age (SD), years: flut/salm 63.5 (9.4), salm 63.7 (9.0)

Smoking history (mean pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: male or female, 40 to 80 years of age with an established history of GOLD stage II COPD; poor reversibility of airflow obstruction (defined as ≤ 10% increase in FEV1 as a percentage of normal predicted value); minimum score of ≥ 2 on the Modified Medical Research Council Dyspnoea Scale and a smoking history of at least 10 pack‐years. In addition, participants had to achieve a composite symptom score of ≥ 120 (out of 400 maximum score, measured using visual analogue scales) on at least 4 of the last 7 days of the run‐in period, and to have a Baseline Dyspnoea Index (BDI) score of ≤ 7 units at visit 2
Exclusion criteria: Participants would be excluded if they had asthma or atopic disease, had a lung disease likely to confound the drug response other than COPD, had a recent exacerbation (within 4 weeks of screening or during run‐in); were receiving long‐term oxygen therapy or pulmonary rehabilitation or had taken tiotropium bromide, inhaled corticosteroids or anti‐leukotriene medication within 14 days of visit one

Interventions

Run‐in: run‐in mentioned, unclear duration

Treatments:

1. Salmeterol/fluticasone propionate 50/250 mcg twice daily

2. Salmeterol 50 mcg twice daily

Inhaler device: Diskus accuhaler

Co‐treatment: not reported

Outcomes

Primary: FEV1, Transitional Dyspnoea Index (TDI)

Secondary: change from baseline in trough FEV1, change from baseline in trough FVC and FVC/FEV1 ratio, TDI focal score, change from baseline in postdose FEV1, FVC and FVC/FEV1 ratio, change from baseline in mean morning PEF, change from baseline in St George's Respiratory Questionnaire

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised (assumed to adhere to usual GSK methodology)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described—only results summary available

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout low and even between groups (11.4% vs 13.9%). The ITT (intent‐to‐treat) population (all participants randomly assigned and confirmed as having received at least one dose of double‐blind study medication) was the primary population for analysis of all efficacy and health outcome variables; the safety population (identical to the ITT population) was used for analysis of all safety variables

Selective reporting (reporting bias)

Low risk

All stated outcomes were reported and no expected outcomes were missing

GSK SCO104925 2008

Methods

Design: multi‐centre, randomised, double‐blind, placebo‐controlled, parallel‐group study

Three‐month treatment period

Conducted at 11 centres (4 centres in the Russian Federation, 4 centres in the United States, 2 centres in Chile and 1 centre in Estonia)

Participants

Participants: 161 people were randomly assigned to fluticasone (42), placebo (42), fluticasone/salmeterol combination (39) and salmeterol (38)
Baseline characteristics:

Male %: flut 69.0, placebo 76.2, flut/salm 82.1, salm 78.9

Mean age (SD), years: flut 64.2 (11.2), placebo 65.2 (8.6), flut/salm 63.6 (7.8), salm 64.0 (9.3)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: Males or females of non‐childbearing potential 40 years of age or older were eligible to participate if they had an established clinical history of COPD, evidence of bronchitis as a component of the COPD disease and a current or prior history of at least 10 pack‐years of cigarette smoking. Participants had a measured postalbuterol FEV1/FVC ≤ 70% at visit 1 (screening) and a measured postalbuterol FEV1 ≥ 30% and ≤ 70% of predicted normal
Exclusion criteria: no other criteria reported

Interventions

Run‐in: not reported

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo twice daily

3. Fluticasone/salmeterol combination 500/50 mcg twice daily

4. Salmeterol 50 mcg twice daily

Inhaler device: not reported

Co‐treatment: not reported

Outcomes

Primary: predose resistance difference between 5 Hz and 15 Hz (R5 to R15) as measured by IOS

Secondary: predose‐ and 2 hours postdose low‐frequency reactance area (AX); 2 hours postdose R5 to R15; postalbuterol computed tomography (CT) parameters of area of airway wall and area of airway lumen

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to treatment (assumed to adhere to GSK protocol)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawal uneven between groups but all less than 20%. ITT analysis used

Selective reporting (reporting bias)

Low risk

All outcomes reported in detail

GSK SCO30002 2005

Methods

Design: multi‐centre, randomised, double‐blind, parallel‐group, placebo‐controlled study

12‐Month treatment period

Conducted at 49 centres in Italy and 7 in Poland

Participants

Participants: 256 people were randomly assigned to fluticasone (131) and placebo (125)
Baseline characteristics:

Male %: flut 83, placebo 80

Mean age (SD), years: flut 64.6 (8.7), placebo 65.7 (9.0)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: male or female individuals aged > 40 years with an established clinical history of COPD; participants who demonstrated at visit 1 a pre‐bronchodilator baseline FEV1/VC < 88% for men and < 89% for women of predicted normal values and FEV1 ≤ 70% of predicted normal value, but > 800 mL; participants who demonstrated at visit 1, poor reversibility of airflow obstruction, defined as an increase in FEV1 < 10% of the normal predicted FEV1 value (or < 200 mL from baseline), 30 minutes after inhalation of 400 mcg salbutamol via MDI; current smokers or ex‐smokers with a smoking history of at least 10 pack‐years
Exclusion criteria: as above

Interventions

Run‐in: two‐week run‐in during which all inhaled corticosteroids and long‐acting beta2‐agonists were discontinued

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo twice daily

Inhaler device: metered‐dose inhaler

Co‐treatment: not reported

Outcomes

Primary: time to first moderate or severe exacerbation

Secondary: number and severity of exacerbations, withdrawals due to exacerbations, clinic FEV1, VC, FEV1/VC, daily record card symptoms, PEFR, distance walked in the six‐minute walk test (SWT), perceived breathlessness before and after SWT, quality of life (SGRQ), use of relief medication, adverse events, SAEs on therapy

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to treatment. No details given but assumed to adhere to GSK methodology

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout high at 26% and 32% for ICS and placebo, respectively. The safety population/Intent‐to‐treat (ITT) population consisted of all randomly assigned participants who took study medication (all of those randomly assigned)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the GSK summary were reported in detail

GSK SCO40041 2008

Methods

Design: randomised, double‐blind, parallel‐group trial

Three‐year treatment period

Conducted at 31 centres in the United States

Participants

Participants: 186 people were randomly assigned to fluticasone/salmeterol combination (92) and salmeterol alone (94)
Baseline characteristics:

Male %: flut/salm 59.8, salm 62.8

Mean age (SD), years: flut/salm 65.4 (8.4), salm 65.9 (9.5)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: male/female participants with an established clinical history of COPD (including a history of exacerbations), a baseline (pre‐bronchodilator) FEV1 < 70% of the predicted normal value, a baseline (pre‐bronchodilator) FEV1/FVC ratio of 70%, at least one evaluable native hip and a smoking history of ≥ 10 pack‐years
Exclusion criteria: no information

Interventions

Run‐in: not reported

Treatments:

1. Fluticasone propionate/salmeterol 250/50 mcg twice daily

2. Salmeterol 50 mcg twice daily

Inhaler device: Diskus

Co‐treatment: 'permitted COPD therapy' unclear

Outcomes

Primary: change in bone mineral density at the lumbar spine

Secondary: change in bone mineral density at the hip, adverse events, serious adverse events, fatal SAEs

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to treatment (no specific information but assumed to adhere to GSK methods)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawal very high in both groups (39% and 41%)

Selective reporting (reporting bias)

Low risk

All outcomes described in the GSK summary were reported

Hanania 2003

Methods

Design: randomised, double‐blind, multi‐centre, placebo‐controlled study

Six‐month treatment period

Conducted at 76 investigative sites in the United States

Participants

Participants: 723 people were randomly assigned to fluticasone/salmeterol combination (178), fluticasone (183), salmeterol (177) and placebo (185)
Baseline characteristics:

Male %: flut/salm 61, flut 66, salm 58, placebo 68

Mean age (range), years: flut/salm 63 (40 to 87), flut 63 (40 to 84), salm 64 (42 to 87), placebo 65 (40 to 81)

Smoking history (median (range) pack‐years): flut/salm 53 (20 to 220), flut 60 (20 to 162), salm 57 (20 to 224), placebo 56 (20 to 165)

Mean % predicted FEV1 (SD): flut/salm 41 (11), flut 42 (11), salm 42 (12), placebo 42 (12)

Inclusion criteria: Participants were > 40 years of age, were current or former smokers with a > 20 pack‐year history and had received a diagnosis of COPD, as defined by the American Thoracic Society. Inclusion criteria required a baseline FEV1/FVC ratio of < 70% and a baseline FEV1 of < 65% of predicted normal, but > 0.70 L (or if < 0.70 L, then > 40% of predicted normal). Participants were required to have symptoms of chronic bronchitis and moderate dyspnoea
Exclusion criteria: current diagnosis of asthma; use of oral corticosteroids within the past six weeks; abnormal clinically significant ECG; long‐term oxygen therapy; moderate or severe exacerbation during the run‐in period; and any significant medical disorder that would place the individual at risk, interfere with evaluations or influence study participation

Interventions

Run‐in: two‐week, single‐blind run‐in period during which participants received placebo and albuterol and discontinued use of corticosteroids and bronchodilators with the exception of stable regimens of theophylline

Treatments:

1. Fluticasone propionate 250 mcg twice daily

2. Salmeterol 50 mcg twice daily

3. Fluticasone/salmeterol 250/50 mcg twice daily

4. Placebo twice daily

Inhaler device: Diskus

Co‐treatment: Participants were given as‐needed albuterol and were not allowed corticosteroids or bronchodilators, with the exception of stable regimens of theophylline

Outcomes

Primary: predose and 2 hours postdose FEV1

Secondary: morning PEF, dyspnoea (TDI), supplemental albuterol use, health status (CRDQ), Chronic Bronchitis Symptom Questionnaire, exacerbations, adverse events, ECG, vital signs and clinical laboratory evaluations

Notes

Funding: GSK (ID: SFCA3007)

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was stratified by reversibility (no other info but GSK sponsored—likely to adhere to GSK methods)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind trial (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout relatively high but even across groups. To account for participant withdrawals, endpoint was used as the primary time point and was defined as the last on‐treatment post‐baseline assessment excluding any data from the discontinuation visit

Selective reporting (reporting bias)

Low risk

Compared with GSK result summary and protocol—no evidence of publication bias

Hattotuwa 2002

Methods

Design: double‐blind, placebo‐controlled study

Three‐month study period

Conducted at the London Chest Hospital

Participants

Participants: 36 people were randomly assigned to fluticasone (17) and placebo (19)
Baseline characteristics:

Male %: flut 81.3, placebo 92.9

Mean age (SD), years: flut 64.7 (6.2), placebo 64.7 (6.5)

Smoking history (mean (SD) pack‐years): flut 64.9 (50.3), placebo 60.3 (46.6)

Mean % predicted FEV1 (SD): flut 46.2 (13.6), placebo 45.5 (16.1)

Inclusion criteria: male or female, 40 to 75 years of age, current smokers or ex‐smokers with more than 20 pack‐years of smoking, non‐atopic and with an FEV1 25% to 80% of predicted, which improved by less than 15% over baseline and 200 mL after 200 mcg inhaled salbutamol
Exclusion criteria: Individuals with severe concurrent medical problems, psychological impairment on immunosuppressive treatment or with a chest infection within 8 weeks were excluded. Participants who were already taking inhaled steroids had the drug withdrawn and had to be stable for at least 8 weeks before the first biopsy

Interventions

Run‐in: After recruitment, participants had a run‐in period of 8 weeks to ensure that they were stable before the first bronchoscopy

Treatments:

1. Fluticasone 500 mcg twice daily

2. Placebo twice daily

Inhaler device: multi‐dose dry powder Accuhaler

Co‐treatment: All reliever medications (inhaled beta2‐agonist, anticholinergics and theophylline) were continued as before

Outcomes

Primary: bronchoscopy

Secondary: spirometry (FEV1 and VC), biopsy processing and counts, symptom scores

Notes

Funding: Glaxo‐Wellcome and Departmental Funds

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to FP or P in a double‐blind manner using a random numbers table

Allocation concealment (selection bias)

Low risk

Randomly assigned in a double‐blind manner

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Seven of the randomly assigned participants did not appear in the analyses (18.9%) but were counted in the adverse event data relevant for this review

Selective reporting (reporting bias)

High risk

Serious adverse events not reported. No reply from study authors at time of publication

Kardos 2007

Methods

Design: randomised, double‐blind, parallel‐group study

10‐Month study period

Conducted at 95 respiratory centres in Germany

Participants

Participants: 994 people were randomly assigned to fluticasone/salmeterol combination (507) and salmeterol alone (487)
Baseline characteristics:

Male %: flut/salm 74.0, salm 77.6

Mean age (SD), years: flut/salm 63.8 (8.3), salm 64.0 (8.2)

Smoking history (mean pack‐years): flut/salm 36.8, salm 37.0

Mean % predicted FEV1 (SD): flut/salm 40.4 (8.9), salm 40.3 (8.5)

Inclusion criteria: outpatients with postbronchodilator FEV1 < 50% predicted, FEV1/FVC of 70% predicted or less, age 40 years or older, smoking history of 10 or more pack‐years and a documented history of two or more moderate to severe exacerbations during the year before the study
Exclusion criteria: Individuals with COPD exacerbations, hospital admissions or change in COPD therapy during the 4 weeks before visit one or during the 4‐week run‐in period were excluded. Those with asthma, significant lung diseases other than COPD and need for long‐term oxygen therapy or long‐term systemic steroid use were also excluded

Interventions

Run‐in: four weeks

Treatments:

1. Salmeterol/fluticasone 50/500 mcg twice daily

2. Salmeterol 50 mcg twice daily

Inhaler device: Diskus

Co‐treatment: Inhaled salbutamol was used as reliever medication, and regular treatment with short‐acting bronchodilators, antioxidants/mucolytics, short‐acting oral beta2‐agonists and theophylline was permitted

Outcomes

Primary: number of exacerbations

Secondary: Time to first exacerbation, pre‐bronchodilator PEF, postbronchodilator FEV1, SGRQ, symptoms and breathlessness, reliever medication use and use of other COPD medications were recorded on diary cards

Notes

Funding: GSK

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Consecutive numbers assigned to participants determined the blinded treatment based on a centrally generated list with blocks of six

Allocation concealment (selection bias)

Low risk

Randomisation list was centrally generated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind treatment (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Similar withdrawal rates in each group. ITT included 99.6% of the randomly assigned population (4 participants were excluded as the result of a randomisation error)

Selective reporting (reporting bias)

Unclear risk

Unable to locate prospective trial registration to check that all outcomes were reported. Study author contacted and forwarded request to GSK—no data were provided in time for publication

Kerwin 2013

Methods

Design: randomised, multi‐centre, placebo‐controlled, parallel‐group trial

Six‐month treatment period

Conducted at 221 centres in nine countries (Chile, Estonia, Germany, Japan, Korea, Phillipines, Poland, Russian Federation and the United States)

Participants

Participants: 1030 people were randomly assigned to placebo (207), fluticasone furoate 100 mcg (206), vilanterol 25 mcg (205), fluticasone furoate 50 mcg and vilanterol combination (206) and fluticasone furoate 100 mcg and vilanterol combination (206)
Baseline characteristics:

Male %: placebo 68, flut100 64, vil 68, flut50/vil 66, flut100/vil 67

Mean age (SD), years: placebo 62.1, flut100 62.7, vil 63.4, flut50/vil 62.8, flut100/vil 62.3

Smoking history (mean (SD) pack‐years): placebo 45.6, flut100 46.2, vil 47.6, flut50/vil 44.2, flut100/vil 46.6

Mean % predicted FEV1 (SD): placebo 42.4, flut10041.5, vil 44.5, flut50/vil 42.5, flut100/vil 42.3

Inclusion criteria: over 40 years of age and have a clinical diagnosis of COPD, a smoking history of 10 pack‐years, a postbronchodilator FEV1/forced vital capacity (FVC) ratio of less than or equal to 0.70, a postbronchodilator FEV1 less than or equal to 70% predicted (NHANES III) and a score of greater than or equal to 2 on the Modified Medical Research Council Dyspnoea Scale (mMRC). A history of COPD exacerbations was not required for individuals to be eligible to enter the study. Albuterol reversibility was assessed at the screening visit, and both reversible and non‐reversible individuals were eligible to enter the study
Exclusion criteria: current diagnosis of asthma or other non‐COPD respiratory disorders; lung volume reduction surgery within 12 months of visit one; poorly controlled COPD, defined as acute worsening of COPD requiring patient‐managed therapy with corticosteroids or antibiotics or treatment prescribed by a physician within 6 weeks before visit 1; hospitalisation due to poorly controlled COPD within 12 weeks before visit 1; lower respiratory tract infection that required the use of antibiotics within 6 weeks before visit 1; the need for long‐term oxygen therapy or nocturnal oxygen therapy (greater than or equal to 12 hours/d)

Interventions

Run‐in: 2‐week, single‐blind run‐in period during which participants received placebo once daily in the morning via a dry powder inhaler that contains two strips

Treatments:

1. Fluticasone furoate 100 mcg daily

2. Vilanterol 25 mcg daily

3. Fluticasone 50 mcg/ vilanterol 25 mcg daily

4. Fluticasone 100 mcg/ vilanterol 25 mcg daily

5. Placebo

Inhaler device: dry powder inhaler

Co‐treatment: Albuterol, ipratropium (provided that the person was on a stable dose from the screening visit throughout the study), mucolytics, antibiotics for short‐term treatment, cough suppressants for short‐term treatment, intranasal decongestants and corticosteroids, flu and pneumonia vaccines, MAOIs, medications for other disorders as long as the dose remained constant whenever possible and their use were not expected to affect lung function

Outcomes

Primary: weighted mean FEV1 (0 to 4 hours postdose) on day 168 to assess bronchodilation by FF/VI and VI (vs placebo) and FF/VI vs FF; and the change from baseline in trough (23 to 24 hours postdose) FEV1 on day 169 to assess the 24‐hour effect of VI and to determine the contribution of FF to lung function (i.e. FF/VI vs VI)

Secondary: CRQ self administered standardised dyspnoea domain on day 168, various FEV1 parameters, PEF, symptom measures, adverse events, exacerbations and pneumonia events

Notes

Funding: GSK (ID: HZC112206)

Clinicaltrials.gov ID: NCT01053988

Definition of pneumonia: presumptive diagnosis or radiographically confirmed

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated using a validated computerised system (RandAll; GlaxoSmithKline, London, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomly assigned using the Registration and Medication Ordering System (RAMOS; GlaxoSmithKline, London UK) to register and randomly assign the participant and to receive medication assignment information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Protocol stated that the study medication was double‐blind for participant and investigator

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated in protocol. Some details in supplementary material about outcome assessors but unclear who was blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout was quite high but even across groups (26.7% to 33.3%). ITT population used

Selective reporting (reporting bias)

Low risk

All stated outcomes were available in full in the published report and on clinicaltrials.gov

Lapperre 2009

Methods

Design: double‐blind, parallel, 4‐group, placebo‐controlled, randomised design

2.5‐Year treatment period

Conducted at 2 centres in the Netherlands

Participants

Participants: 55 people were randomly assigned to fluticasone (26) and placebo (29)
Baseline characteristics:

Male %: flut 88.5, placebo 83.3

Mean age (SD), years: flut 62 (8), placebo 59 (8)

Smoking history (mean (range) pack‐years): flut 44 (31 to 55), placebo 42 (34 to 54)

Mean % predicted FEV1 (SD): flut 57 (9.9), placebo 54 (8.3)

Inclusion criteria: 45 to 75 years of age, were current or former smokers, had smoked for 10 or more pack‐years and had lung function levels compatible with Global Initiative for Chronic Obstructive Lung Disease (GOLD) stages II and III
Exclusion criteria: asthma and receipt of ICS within 6 months before random assignment. Important co‐morbid conditions

Interventions

Run‐in: not reported

Treatments:

1. Fluticasone 500 mcg twice daily

2. Placebo twice daily

Inhaler device: Diskus dry powder inhaler

Co‐treatment: short‐acting bronchodilators

Outcomes

Primary: inflammatory cell counts in bronchial biopsies and induced sputum

Secondary: postbronchodilator spirometry, hyperresponsiveness to methacholine PC20, dyspnoea score by the MRC scale, health status by the SGRQ and the Clinical COPD Questionnaire

Notes

Funding: Netherlands Asthma Foundation, both centres and GSK

Clinicaltrials.gov ID: NCT00158847

Definition of pneumonia: confirmed by chest x‐ray

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

At entry, an independent randomisation centre provided participant and medication numbers by using a minimisation procedure that balanced treatment groups for centre, sex, smoking status, FEV1/IVC 60% and methacholine PC20 (the provocative concentration of methacholine that causes a 20% decrease in FEV1) 2 mg/mL)

Allocation concealment (selection bias)

Low risk

An independent randomisation centre provided participant and medication numbers

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study medications were individually numbered, and we used Diskus dry powder inhalers (GlaxoSmithKline, Zeist, The Netherlands) with 60 doses per inhaler; all active treatment medications and placebo were identical in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawal rates were very high compared with the numbers of events for the different outcomes. Per‐protocol analysis was used

Selective reporting (reporting bias)

Low risk

Data for several outcomes not available from the published report, but study authors provided data upon request

Laptseva 2002

Methods

Design: randomised, double‐blind, parallel‐group study

Six‐month treatment period

Participants

Participants: 49 people were randomly assigned to budesonide (25) and placebo (24)
Baseline characteristics: none reported—abstract only

Inclusion criteria: individuals between 40 and 65 years of age, FEV1 40% to 60% of predicted normal, FEV1/VC < 55%, bronchodilator reversibility < 15%
Exclusion criteria: not reported

Interventions

Run‐in: not reported

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo

Inhaler device: not reported

Co‐treatment: All participants received anticholinergic drug and methylxanthine or short‐acting beta2 agent

Outcomes

Number and severity of exacerbations, FEV1, FVC, diary card symptoms, PEFR

Notes

Abstract only

Funding: not reported

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No details provided

Selective reporting (reporting bias)

High risk

Only abstract available. Outcomes could not be used. Could not find contact information for study authors

Mahler 2002

Methods

Design: randomised, double‐blind, placebo‐controlled, parallel‐group, multi‐centre trial

6 ‐month treatment period

Conducted at 65 centres

Participants

Participants: 674 people were randomly assigned to fluticasone (168), placebo (181), fluticasone/salmeterol combination (165) and salmeterol alone (160)
Baseline characteristics:

Male %: flut 61, placebo 75, flut/salm 62, salm 64

Mean age (range), years: flut 64.4 (42 to 82), placebo 64 (44 to 90), flut/salm 61.9 (40 to 86), salm 63.5 (40 to 84)

Smoking history (median (range) pack‐years): flut 54 (20 to 200), placebo 60 (20 to 165), flut/salm 55 (15 to 150), salm 52.5 (20 to 193)

Mean % predicted FEV1 (no SD reported): flut 41, placebo 41, flut/salm 41, salm 40

Inclusion criteria: baseline FEV1/FVC of 70% or less and baseline FEV1 of less than 65% of predicted but more than 0.70 L. Participants were required to have daily cough productive of sputum for 3 months of the year for 2 consecutive years and dyspnoea
Exclusion criteria: Specific exclusion criteria were current diagnosis of asthma, oral corticosteroid use within the past 6 weeks, abnormal clinically significant electrocardiogram, long‐term oxygen therapy, moderate or severe exacerbation during the run‐in and any clinically significant medical disorder

Interventions

Run‐in: 2‐week, single‐blind, run‐in period during which participants received placebo via Diskus on an as‐needed basis and discontinued use of corticosteroids and bronchodilators, with the exception of stable regimens of theophylline

Treatments:

1. Fluticasone 500 mcg twice daily

2. Placebo twice daily

3. Fluticasone/Salmeterol combination 500/50 mcg twice daily

4. Salmeterol 50 mcg twice daily

Inhaler device: Diskus

Co‐treatment: as needed albuterol and stable regimens of theophylline

Outcomes

Primary: change in predose FEV1 values and change in 2‐hour postdose FEV1 values

Secondary: morning and evening PEF, supplemental albuterol use, dyspnoea as assessed by the TDI, Chronic Bronchitis Symptom Questionnaire, exacerbations defined by treatment, health status on the Chronic Respiratory Disease Questionnaire, adverse events, 24‐hour Holter monitoring and vital signs

Notes

Funding: GSK (protocol number SFCA3006)

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was stratified by reversibility and investigative site to ensure balance between treatment groups at each site and in terms of the number of reversible participants (no other details, industry‐sponsored)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

17 participants at one investigative site were not evaluable because of poor study practices, and of the remaining 674 participants, 645 had an evaluable baseline assessment. A total of 234 participants were discontinued from the study (between 28% and 40% across groups)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported in detail. Only one secondary outcome was not available

Martinez 2013

Methods

Design: multi‐centre, randomised, stratified (by smoking status), placebo‐controlled, double‐blind, parallel‐group study

6 month treatment period

Conducted at 145 study centres in 8 countries (Czech Republic, Germany, Japan, Poland, Romania, Russian Federation, Ukraine and the United States)

Participants

Participants: 1224 people were randomly assigned to fluticasone furoate 100 mcg (204), fluticasone furoate 200 mcg (203), placebo (205), fluticasone furoate 100 mcg and vilanterol combination (204), fluticasone furoate 100 mcg and vilanterol combination (205) and vilanterol (203)
Baseline characteristics:

Male %: flut100 74, flut200 74, placebo 74, flut100/vil 71, flut200/vil 67, vil 74

Mean age (SD), years: flut100 61.8 (8.3), flut200 61.8 (9.0), placebo 61.9 (8.1), flut100/vil 61.9 (8.8), flut200/vil 61.1 (8.6), vil 61.2 (8.6)

Smoking history (mean (SD) pack‐years): flut100 39.8 (21.3), flut200 43.5 (22.5), placebo 45.7 (25.8), flut100/vil 42.8 (23.9), flut200/vil 41.5 (23.4), vil 42.0 (23.3)

Mean % predicted FEV1 (SD): flut100 48.4 (12.2), flut200 47.1 (12.0), placebo 48.3 (12.7), flut100/vil 48.1 (12.9), flut200/vil 47.1 (12.8), vil 48.5 (12.9)

Inclusion criteria: clinical diagnosis of COPD, 40 years of age or older, smoking history of 10 or more pack‐years, postbronchodilator FEV1/ forced vital capacity (FVC) ratio of 0.70 or less, postbronchodilator FEV1 70% predicted or less (NHANES III) and score of 2 or higher on the Modified Medical Research Council Dyspnoea Scale (mMRC). No prior history of COPD exacerbations was required for individuals to be eligible to enter the study. Reversibility to albuterol was assessed at the screening visit; both reversible and non‐reversible individuals were eligible to enter the study
Exclusion criteria: any respiratory disorder other than COPD; lung volume reduction surgery within 12 months of screening; acute worsening (participant‐managed corticosteroid or antibiotic treatment or physician prescription) of COPD within six weeks of screening, hospitalisation for COPD over 12 weeks or lower respiratory tract infection that required the use of antibiotics in the 6 weeks before screening; the need for long‐term oxygen therapy or nocturnal oxygen therapy (12 or more hours/d)

Interventions

Run‐in: 2‐week, single‐blind run‐in period during which participants received placebo once daily in the morning via a dry powder inhaler (DPI) that contained two strips

Treatments:

1. Fluticasone furoate 100 mcg daily

2. Fluticasone furoate 200 mcg daily

3. Placebo

4. Fluticasone furoate 100 mcg/vilanterol 25 mcg daily

5. Fluticasone furoate 200 mcg/vilanterol 25 mcg daily

6. Vilanterol 25 mcg

Inhaler device: dry powder inhaler

Co‐treatment: albuterol, ipratropium (provided that the person was on a stable dose from the screening visit throughout the study), mucolytics, antibiotics for short‐term treatment, cough suppressants for short‐term treatment, intranasal decongestants and corticosteroids, flu and pneumonia vaccines, MAOIs, medications for other disorders as long as the dose remained constant whenever possible and when their use was not expected to affect lung function

Outcomes

Primary: weighted mean FEV1 (zero to four hours postdose) on day 168 to assess bronchodilation by FF/VI and VI (vs placebo) and FF/VI versus FF; change from baseline in trough (23 to 24 hours postdose) FEV1 on day 169 to assess the 24‐hour effect of VI and to determine the contribution of FF to lung function (i.e. FF/VI vs VI)

Secondary: CRQ self administered standardised dyspnoea domain on day 168, various FEV parameters, PEF, symptom measures, adverse events, exacerbations and pneumonia events

Notes

Funding: GSK (ID: HZC112207)

Clinicaltrials.gov ID: NCT01054885

Definition of pneumonia: presumptive diagnosis or radiographically confirmed

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated using a validated computerised system (RandAll; GlaxoSmithKline, London, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomly assigned using the Registration and Medication Ordering System (RAMOS; GlaxoSmithKline, London, UK) to register and randomly assign the participant and to receive medication assignment information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Protocol stated that the study medication was double‐blind for participant and investigator

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated in protocol. Some details in supplementary material about outcome assessors but unclear who was blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout in all groups was less than 30% (range 20.1% to 29.4%) and an ITT analysis was used

Selective reporting (reporting bias)

Low risk

All stated outcomes were available in full in the published report and in supplementary tables

Mirici 2001

Methods

Design: randomised, placebo‐controlled, double‐blind, parallel‐group study

Three‐month treatment period

Conducted at a single centre in Turkey

Participants

Participants: 50 people were randomly assigned to budesonide (25) and placebo (25)
Baseline characteristics:

Male %: bud 70, placebo 80

Mean age (SD), years: bud 51.8 (9.5), placebo 54.5 (10.3)

Smoking history (mean (SD) pack‐years): bud 21.7 (12.5), placebo 31.3 (19.1)

Mean % predicted FEV1 (SD): bud 64.1 (6.5), placebo 59.9 (8.2)

Inclusion criteria: FEV1 < 70%, FEV1 reversibility after inhalation of terbutaline from a Turbuhaler of less than 15% of pre‐bronchodilator FEV1. All participants were smokers who refused or failed a programme to quit smoking
Exclusion criteria: long‐term treatment with oral or inhaled corticosteroids within 6 months of study entry, respiratory tract infection in the previous 3 months, pregnancy or lactation and the presence of other serious systemic disease

Interventions

Run‐in: not reported

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: Turbuhaler

Co‐treatment: Beta2‐agonists of all kinds, theophylline and mucolytics were allowed. Inhaled corticosteroids other than study medication and oral or parenteral corticosteroids were not allowed

Outcomes

Spirometry and sputum cell analysis

Notes

Funding: unclear

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation sequence was computer‐generated at the Research Centre of the Faculty of Medicine

Allocation concealment (selection bias)

Low risk

Randomisation was masked and case numbers were allocated in consecutive order

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind. All inhalers had the same appearance and did not have drug labels

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All spirometric indices and sputum cell analyses were performed at baseline and after treatment, blind to the clinical details

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Equal and fairly low dropout per group, values not imputed

Selective reporting (reporting bias)

High risk

Key expected outcomes were not reported (mortality and adverse events). Named outcomes were well reported. No reply from authors at the time of publication

Ozol 2005

Methods

Design: randomised, double‐blind, placebo‐controlled design

Six‐month treatment period

Conducted at a single centre in Turkey

Participants

Participants: 26 people were randomly assigned to budesonide (13) and placebo (13)
Baseline characteristics:

Male %: bud 84.6, placebo 53.8

Mean age (SD), years: bud 64.9 (6.1), placebo 65.9 (8.1)

Smoking history (mean (SD) pack‐years): bud 45.6 (22.2), placebo 44.4 (23.0)

Mean % predicted FEV1 (SD): bud 61.1 (9.7), placebo 57.3 (11.2)

Inclusion criteria: (1) FEV1/FVC <70% and FEV1 50% of predicted value, (2) reversibility with inhaled beta2‐agonists (400 mg salbutamol) of less than 200 mL or less than 12% of predicted FEV1, (3) stable COPD defined as no acute exacerbation within preceding 3 months, (4) no history of systemic disease or other pulmonary disease, (5) no therapy with inhaled or systemic corticosteroids within 3 months before entry into the study and (6) no history of asthma or atopy
Exclusion criteria: no additional information

Interventions

Run‐in: not reported

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: dry powder inhaler

Co‐treatment: All participants were receiving therapy with inhaled salbutamol and ipratropium bromide. For 9 participants, sustained‐released theophyline was also given

Outcomes

Primary: unclear

Secondary: FVC, FEV1, diary card data, inflammatory measures

Notes

Funding: unclear

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned by a computer‐generated, blinded randomisation list

Allocation concealment (selection bias)

Low risk

‘Blinded’ randomisation list

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Cells were counted by our pathologist, who was also blinded. Not clear for other outcomes

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

‘The results presented are an analysis of 22 subjects (12 budesonide‐treated subjects and 10 placebo‐treated subjects) who completed the study’. Both dropout rates were low but uneven between groups (two were excluded from the placebo group and one from the budesonide group. One extra, presumed to have been randomly assigned to placebo (assuming equal group size at randomisation) was excluded for failure to take the medication consistently)

Selective reporting (reporting bias)

High risk

Key expected outcomes missing (mortality, serious adverse events). No reply from study authors by time of publication

Paggiaro 1998

Methods

Design: multi‐centre, randomised, placebo‐controlled trial

Six‐month treatment period

Conducted in 13 European countries, New Zealand and South Africa

Participants

Participants: 281 people were randomly assigned to fluticasone (142) and placebo (139)
Baseline characteristics:

Male %: flut 70, placebo 78

Mean age (no SD reported), years: flut 62, placebo 64

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): flut 59 (18), placebo 55 (17)

Inclusion criteria: current or ex‐smokers, 50 to 75 years of age with a history of smoking equivalent to at least 10 pack‐years and chronic bronchitis (a cough with excess sputum production for at least 3 months in at least 2 consecutive years with no other pathology). Participants also had to have a history of at least one exacerbation per year for the previous 3 years that required a visit to their doctor or hospital; high expectation, according to the investigator, of experiencing an exacerbation during the 6‐month treatment period; regular productive cough; predicted FEV1 of 35% to 90%, ratio of FEV1 to forced vital capacity of 70% or less and reversibility in FEV1 of less than 15% after inhalation of 400 mcg or 800 mcg salbutamol via a metered‐dose inhaler or Diskhaler
Exclusion criteria: Individuals with abnormal chest radiographs or who had received oral or depot steroids, inhaled steroids of more than 500 mcg daily or antibiotic therapy; had been admitted to hospital in the 4 weeks before the study; or were currently taking fluticasone propionate were excluded

Interventions

Run‐in: 2 week run‐in period during which usual inhaled steroids were stopped and participants received salbutamol as required

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo twice daily

Inhaler device: metered‐dose inhalers, with a spacer if desired

Co‐treatment: Participants could take short‐acting beta2‐agonists for relief of symptoms as required throughout the study. Other COPD medications, such as anticholinergics and xanthine derivatives, could be continued throughout the study without dose changes

Outcomes

Primary: COPD exacerbations

Secondary: FEV1, morning PEF, FVC, 6‐minute walk test, Borg score, diary card symptom scores, daily sputum volume, total adverse events, serum cortisol concentration

Notes

Funding: unclear (‘code was held by the sponsor company’s statisticians’)

Definition of pneumonia:Not provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random numbers were computer‐generated on PACT (version 2.7)

Allocation concealment (selection bias)

Low risk

All investigators were given a set of four or more sealed envelopes containing assignment codes, from which they assigned treatment, starting with the lowest number

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

'We did analysis by intention to treat of all patients who took at least one dose of study medication’

‘Only available data was analysed’. Dropout uneven

Selective reporting (reporting bias)

High risk

Key expected outcomes not reported (mortality and serious adverse events). No reply from study authors by time of publication

Pauwels 1999

Methods

Design: parallel‐group, double‐blind, placebo‐controlled, randomised, multi‐centre study

3 year treatment period

Conducted at 39 study centres in nine European countries (Belgium, Denmark, Finland, Italy, the Netherlands, Norway, Spain, Sweden and the United Kingdom)

Participants

Participants: 1277 people were randomly assigned to budesonide (634) and placebo (643)
Baseline characteristics:

Male %: bud 73.5, placebo 72.2

Mean age (SD), years: bud 52.5 (7.5), placebo 52.4 (7.7)

Smoking history (mean (SD) pack‐years): bud 39.4 (20.1), placebo 39.2 (20.2)

Mean % predicted FEV1 (SD): bud 76.8 (12.4), placebo 76.9 (13.2)

Inclusion criteria: Persons 30 to 65 years of age were eligible if they were currently smoking at least five cigarettes per day and had smoked cigarettes for at least 10 years or had a smoking history of at least 5 pack‐years. FEV1 after use of a bronchodilator had to be between 50% and 100% of predicted normal value, and ratio of pre‐bronchodilator FEV1 to slow vital capacity had to be less than 70%. Increase in FEV1 after inhalation of 1 mg of terbutaline from a dry powder inhaler had to be less than 10% of predicted normal value. Change in FEV1 between the end of the first three‐month period of the run‐in phase and the end of the second had to be less than 15%
Exclusion criteria: Participants with history of asthma, allergic rhinitis or allergic eczema and those who had used oral glucocorticoids for longer than four weeks during the preceding six months were excluded. Use of inhaled glucocorticoids other than the study medication, beta‐blockers, cromones or long‐acting inhaled beta2‐adrenergic agonists was not allowed

Interventions

Run‐in: three‐month smoking cessation programme. For participants who did not stop smoking, this phase was followed by a three‐month period during which compliance with inhaled medication was assessed with the use of a placebo containing dry powder inhaler with a hidden mechanical counter. Participants who continued smoking and were at least 75% compliant with the recommended treatment regimen were randomly assigned

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: 1, Pulmicort; 2, dry powder turbuhaler

Co‐treatment: Use of inhaled glucocorticoids other than the study medication, beta‐blockers, cromones or long‐acting inhaled beta2‐adrenergic agonists was not allowed

Outcomes

Primary: change over time in postdose FEV1

Secondary: serious adverse events, mortality, glucocorticoid‐related adverse effects, bone density, non‐serious adverse events

Notes

Funding: funded by a grant from Astra Draco, Lund, Sweden

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

‘Randomly assigned’. No specific details given but industry‐sponsored

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participant and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Central evaluator who was unaware of the treatment received and was analysed according to a standardised computerised protocol

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data on randomly assigned participants were analysed on an intention‐to‐treat basis. Withdrawal rates under 30% and even in both groups

Selective reporting (reporting bias)

High risk

Several missing outcomes. Could not locate protocol to check that all prospectively registered outcomes were reported. Contacted second study author; no reply by time of publication

Renkema 1996

Methods

Design: parallel‐group, double‐blind, randomised, placebo‐controlled study

2‐year study period

Conducted at a single centre in the Netherlands

Participants

Participants: 39 people were randomly assigned to budesonide (21) and placebo (18)
Baseline characteristics:

Male %: bud 100, placebo 100

Mean age (SD), years: bud 56 (8), placebo 54 (10)

Smoking history, cigarettes/year (SD): bud 635 (530), placebo 729 (495)

Mean % predicted FEV1 (SD): bud 67 (15), placebo 60 (18)

Inclusion criteria: clinical diagnosis of COPD based on history (persistent dyspnoea, mainly on exertion, without sudden attacks of dyspnoea); FEV1 less than 80% of predicted value; residual volume (RV) greater than 100% of predicted value; specific compliance expressed as percentage of predicted value greater than 100% after bronchodilation; when, however, air trapping (calculated as thoracic gas volume measured by body plethysmography minus functional residual capacity measured with an indicator gas) was greater than 1.5 L Csp was allowed to be less than 100% of predicted; no signs of allergy (negative skin test results, total serum IgE < 200 IU/mL, eosinophils in peripheral blood < 250 × l03/mL); and stable phase of the disease
Exclusion criteria: Excluded were participants older than 70 years at entry, participants receiving continuous corticosteroid therapy and participants with severe concomitant disease, likely to interfere with the purpose of the study. All participants had alpha‐1 antitrypsin serum levels within the normal range. All participants were smokers or ex‐smokers. Smoking history was expressed as cigarette‐years

Interventions

Run‐in: three months with no corticosteroid medication

Treatments:

1. Budesonide 800 mcg twice daily (plus placebo tablet once daily)

2. Placebo twice daily (plus placebo tablet once daily)

Inhaler device: metered‐dose inhaler (MDI) through a 750‐mL spacer (Nebuhaler; ASTRA, Ryswylc, The Netherlands)

Co‐treatment: Throughout the study, participants were maintained on regimens of their usual bronchodilator medication, consisting of anticholinergics. beta‐agonists, theophylline or a combination of these drugs

Outcomes

Primary: FEV1

Secondary: compliance, symptom scores, fasting morning plasma cortisol levels

Notes

Funding: grants from the Netherlands Asthma Foundation, ASTRA BV Holland and AB DRACO Sweden

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

By computerised randomisation, stratified for smoking

Allocation concealment (selection bias)

Low risk

Allocated blindly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Uneven withdrawal rates, no description of imputation to account for dropout

Selective reporting (reporting bias)

High risk

Key expected outcomes not reported (mortality and adverse events). No reply from study authors by time of publication

Rennard 2009

Methods

Design: Randomised, double‐blind, double‐dummy, parallel‐group, active‐ and placebo‐controlled, multi‐centre study

12‐Month treatment period

237 sites in the US, Europe and Mexico

Participants

Participants: 1483 people were randomly assigned to budesonide/formoterol at high (494) and low dose (494), and to formoterol alone (495)
Baseline characteristics:

Male %: bud/form high 62.3, bud/form low 62.8, form 65.3

Mean age (SD), years: bud/form high 63.2 (8.9), bud/form low 63.6 (9.2), form 62.9 (9.1)

Smoking history (median pack‐years): bud/form high 40, bud/form low 40, form 40

Mean % predicted FEV1 (SD): bud/form high 38.6 (11.4), bud/form low 39.6 (10.9), form 39.3 (11.9)

Inclusion criteria: current smokers or ex‐smokers aged > 40 years with clinical diagnosis of COPD and symptoms for > 2 years were eligible for this study. Participants were required to have a history of at least one COPD exacerbation treated with a course of oral corticosteroids and/or antibacterials, with 1 to 12 months before screening and documented use of an inhaled short‐acting bronchodilator as rescue medication. Prebronchodilator FEV1 of < 50% of predicted normal and pre‐bronchodilator FEV1/FVC of < 70% were required at screening. Smoking history of at least 10 pack‐years, score ≥ 2 on the Modified Medical Research Council dyspnoea scale at the time of screening and breathlessness, cough and sputum scale score ≥ 2 per day for at least half of the 2‐week run‐in period
Exclusion criteria: Individuals were excluded if they had any of the following conditions: history of asthma, history of allergic rhinitis before 40 years of age, significant/unstable CV disorder, clinically significant respiratory tract disorder other than COPD and homozygous alpha1‐antitrypsin deficiency or any other clinically significant co‐morbidities. Individuals were also excluded if they needed additions or alterations to their usual COPD maintenance therapy or an increment in rescue therapy because of worsening symptoms within 30 days before screening. Oral or ophthalmic non‐cardioselective beta‐adrenoceptor antagonists, oral corticosteroids, pregnancy and breast‐feeding also were exclusionary

Interventions

Run‐in: 2 weeks during which previous inhaled corticosteroids were discontinued

Treatments:

1. Budesonide/formoterol 320/9 mcg twice daily

2. Budesonide/formoterol 160/9 mcg twice daily

3. Formoterol 9 mcg twice daily

Inhaler device: 1 and 2, pressurised metered‐dose inhaler. 3, dry powder inhaler

Co‐treatment: salbutamol allowed as reliever medication

Outcomes

Primary: predose FEV1 and one hour postdose FEV1

Secondary: morning and evening PEF, COPD exacerbations, quality of life (SGRQ), symptom scores, percentage of awakening‐free nights, any adverse event (AE), pneumonia‐related AEs, serious AEs, mortality, vital signs and cortisol levels

Notes

Funding: AstraZeneca

Clinicaltrials.gov ID: NCT00206167

Definition of pneumonia: reported by physicians based on the Medical Dictionary for Regulatory Activities (version 10.0)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Not described. Industry sponsored, presumed to follow usual AZ methods

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind [presumed participants and personnel/investigators]

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Safety was assessed by adverse event (AE) reporting. Pneumonia events were reported by physicians based on the Medical Dictionary for Regulatory Activities (version 10.0) pneumonia‐related preferred terms (pneumonia, bronchopneumonia, lobar pneumonia or pneumonia staphylococcal).

Incomplete outcome data (attrition bias)
All outcomes

High risk

The withdrawal rates were very high compared to the number of events for the different outcomes

Selective reporting (reporting bias)

Low risk

All outcomes reported [checked against protocol]. Only one secondary outcome not reported.

Schermer 2009

Methods

Design: randomised, double‐blind, double‐dummy, placebo‐controlled phase IV trial

Three‐year treatment period

Conducted at 44 general practices in the Netherlands

Participants

Participants: 190 people were randomly assigned to fluticasone (94) and placebo (96)
Baseline characteristics:

Male %: flut 73, placebo 68

Mean age (SD), years: flut 58.4 (9.9), placebo 59.6 (10.1)

Smoking history (mean (SD) pack‐years): flut 30.2 (18.2), placebo 26.5 (16.7)

Mean % predicted FEV1 (SD): flut 63.2 (17.1), placebo 65.7 (17.7)

Inclusion criteria: age 35 to 75 years; current or former smoker; chronic dyspnoea, sputum production and cough for at least three consecutive months per year during the previous two years; postbronchodilator forced expiratory volume in one second (FEV1) < 90% of predicted value and/or postbronchodilator FEV1/FVC (forced vital capacity) of predicted value < 88% for men and < 89% for women
Exclusion criteria: postbronchodilator FEV1 < 40% of predicted and/or a history of asthma, allergic rhinitis or allergic eczema

Interventions

Run‐in: Trial was preceded by the following (in chronological order): an optional smoking cessation attempt supported by the GP in trial candidates who were current smokers; a three‐month washout phase to eliminate possible carry‐over effects of a successful smoking cessation attempt or withdrawal of prior treatment with N‐acetylcysteine and/or inhaled corticosteroids; and a 14‐day pretreatment phase (30 mg oral prednisolone) to attain the highest possible baseline condition

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo twice daily

Inhaler device: Diskus dry powder inhaler. Unclear from trial report whether placebo was administered to match the fluticasone inhaler or the other active treatment, which was delivered as effervescent tablets dissolved in a glass of tap

Co‐treatment: not reported

Outcomes

Primary: rate of exacerbation and quality of life as measured by the interviewer‐administered version of the Chronic Respiratory Questionnaire (CRQ)

Notes

Funding: Dutch Council for Health Insurances, with complementary funding by the Netherlands Asthma Foundation (authors had received various GSK and other pharma research grants)

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An independent statistician generated a randomisation list based on a block size of three for treatment allocation to balance the three treatment arms by study centre.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Neither investigators nor patients were aware of the group assignment. Placebo described as ‘matching’

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Neither investigators nor patients were aware of the group assignment [presuming the investigators were those doing the outcome assessments]

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropout was high in both groups. The primary analyses were done on an intention‐to‐treat basis. Additional per protocol analyses were done on patients with a trial medication compliance rate >80%. Unclear how data were imputed or who was included in the ITT population

Selective reporting (reporting bias)

High risk

All outcomes stated in the protocol were reported but some key expected outcomes were missing (serious adverse events and pneumonia). No reply from author by time of publication.

Senderovitz 1999

Methods

Design: placebo‐controlled, randomised, double‐blind, multi‐centre trial

Six‐month treatment period

Five centres in Denmark

Participants

Participants: unclear how many people were randomly assigned. 26 were evaluable in the budesonide (14) and placebo (12) groups
Baseline characteristics:

Male %: bud 57, placebo 50

Median age (range), years: bud 58.5 (51 to 74), placebo 62.5 (57 to 74)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): not reported

Inclusion criteria: outpatients 18 to 75 years of age with stable COPD were included. FEV1, forced vital capacity (FVC) < 0.7, postbronchodilator FEV1 < 70% of predicted, FEV1 > 40% of predicted and increase in FEV1 < 15% after inhalation of 0 to 5 mg terbutaline
Exclusion criteria: clinical evidence of asthma (e.g. pollen season–related symptoms, exercise‐induced symptoms only and significantly elevated levels of eosinophils and IgE), history of atopy (hay fever and/or atopic dermatitis), treatment with inhaled corticosteroids within the past 6 months, treatment with oral corticosteroids, cromoglycate or nedocromil within the past 4 weeks, other systemic disease making compliance and participation in the study difficult, pregnancy and breast‐feeding and an increase in FEV1 > 30% of baseline after 2 weeks of prednisolone treatment

Interventions

Run‐in: All participants received 2 weeks of treatment with oral prednisolone 37.5 mg daily. Reversible participants with 15% < AFEV1 < 30% of baseline and irreversible participants with AFEV1 < 15% were separately randomly assigned to inhaled budesonide 400 pg bid or placebo

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: Spirocort Turbuhaler

Co‐treatment: not reported

Outcomes

Primary: FEV1

Secondary: exacerbations, adverse events and symptom scores

Notes

Funding: not reported

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Reversible participants with 15% < AFEV1 < 30% of baseline and irreversible participants with AFEV1 < 15% were separately randomly assigned

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

26 of 37 were evaluable (12 in placebo group, 14 in active group—30% dropout overall). Number randomly assigned and number of dropouts not provided for each group. ITT not adopted

Selective reporting (reporting bias)

High risk

Several key expected outcomes not reported (mortality, adverse events, withdrawal per group). Study author contacted but not able to provide data

Shaker 2009

Methods

Design: randomised, double‐blinded, placebo‐controlled, parallel‐group, single‐centre study

Two‐ to four‐year treatment period

Conducted at a single centre in Denmark

Participants

Participants: 254 people were randomly assigned to budesonide (127) and placebo (127)
Baseline characteristics:

Male %: bud 62.2, placebo 54.3

Mean age (SD), years: bud 63.6 (7.5), placebo (63.6 (7.2)

Smoking history (mean (SD) pack‐years): bud 56 (23), placebo 56 (24)

Mean % predicted FEV1 (SD): bud 51 (11), placebo 53 (11)

Inclusion criteria: Individuals 50 to 80 years of age were eligible if they were current smokers with a clinical diagnosis of COPD for not less than 2 years. All participants should have a significant smoking history of at least 10 cigarettes per day during the past 6 months and a previous history of at least 20 pack‐years. Ex‐smokers were excluded. Baseline lung function criteria were as follows: FEV1 between 35% and 70% of predicted (pre‐bronchodilator), and FEV1/forced vital capacity (FEV1/FVC) ≤ 60%
Exclusion criteria: Reversibility of ≥ 12% and 200 mL in FEV1 from baseline values, 15 minutes after inhalation of 1 mg terbutaline or ≥ 15% and 300 mL after 2 weeks on oral prednisolone (25 mg), was an exclusion criterion. Individuals were also excluded if they had any severe concomitant disease; had an exacerbation within 30 days before the first visit; received oral steroids for longer than four weeks within six months of the first visit; or were on long‐term oxygen therapy

Interventions

Run‐in: two‐week run‐in period on oral prednisolone

Treatments:

1. Budesonide 400 mcg twice daily

2. Placebo twice daily

Inhaler device: Pulmicort Turbuhaler

Co‐treatment: Bronchodilators, mucolytics and short courses of oral corticosteroids (maximum 3 courses of 4 weeks' duration per year) and antibiotics were allowed during the study

Outcomes

Primary: 15th percentile density (PD15)

Secondary: change over time in the relative area of emphysema at a threshold of –910 Hounsfield units (RA‐910), FEV1 and diffusion capacity (DLCO) and in number of exacerbations, which was defined as a combination of 2 of the 3 following criteria: increased dyspnoea, increased sputum production and change in sputum colour

Notes

Funding: AstraZeneca

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were allocated to either group in a proportion of 1:1 by block randomisation using a random sequence generated by a computer programme at AstraZeneca

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. To maintain blinding, all Turbuhalers were of identical appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

High proportion of dropouts in both groups (43% intervention and 49% placebo)

Selective reporting (reporting bias)

Low risk

Could not locate protocol to check that all prospectively registered outcomes were reported, but study authors provided all relevant outcomes upon request

Sharafkhaneh 2012

Methods

Design: randomised, double‐blind, double‐dummy, parallel‐group, multi‐centre study

12‐Month treatment period

Conducted at 180 study sites in the United States (106 sites), Central and South America (53 sites) and South Africa (21 sites)

Participants

Participants: 1219 people were randomly assigned to high‐dose budesonide/formoterol (407), low‐dose budesonide/formoterol (408) and formoterol alone (404)
Baseline characteristics:

Male %: bud/form high 64.4, bud/form low 64.7, form 56.8

Mean age (SD), years: bud/form high 63.8 (9.4), bud/form low 62.8 (9.2), form 62.5 (9.4)

Smoking history (mean pack‐years): bud/form high 46, bud/form low 44, form 43

Mean % predicted FEV1 (SD): bud/form high 37.9 (11.8), bud/form low 37.6 (11.6), form 37.5 (12.4)

Inclusion criteria: Individuals were current smokers or ex‐smokers with a smoking history of 10 pack‐years, 40 years of age, with a clinical diagnosis of COPD with symptoms for >2 years. Participants were required to have a history of 1 COPD exacerbation requiring treatment with a course of systemic corticosteroids, antibiotics or both, within one to 12 months before screening (visit 1) and documented use of an inhaled short‐acting bronchodilator as rescue medication. At screening, a pre‐bronchodilator forced expiratory volume in one second (FEV1) of 50% of predicted normal and a pre‐bronchodilator FEV1/forced vital capacity (FVC) of < 70% also were required
Exclusion criteria: Exclusion criteria included current, previous (within past 60 days) or planned enrolment in a COPD pulmonary rehabilitation programme, treatment with oral corticosteroids and incidence of a COPD exacerbation or any other significant medical diagnosis between screening and randomisation visits

Interventions

Run‐in: 2‐week run‐in period

Treatments:

1. Budesonide/formoterol 320/9 mcg twice daily

2. Budesonide/formoterol 160/9 mcg twice daily

3. Formoterol DPI 9 mcg twice daily

Inhaler device: 1 and 2, pressurised metered‐dose inhaler. 3, dry powder inhaler

Co‐treatment: Rescue medication (albuterol pMDI 90 mg 2 inhalations) was provided for as needed use during screening and run‐in, and throughout the study

Outcomes

Primary: COPD exacerbations

Secondary: FEV1, FVC, morning and evening PEF, diary card symptoms, rescue medication use, BODE index, exercise capacity, health‐related quality of life (SGRQ), adverse events. Unclear which was primary

Notes

Funding: AstraZeneca ID: D589CC00003

Clinicaltrials.gov ID: NCT00419744

Definition of pneumonia: based on clinical judgement, not on radiological or microbial assessments

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Assignments were made sequentially by interactive voice response system in a a computer generated allocation schedule produced in advance.

Allocation concealment (selection bias)

Low risk

Assignments were made sequentially by interactive voice response system following a computer generated allocation schedule produced in advance.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

To maintain patient and investigator blinding, all active treatments were provided in blinded treatment kits. Patients in the budesonide/formoterol pMDI groups received a placebo DPI and those in the formoterol DPI group received a placebo pMDI.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

The withdrawal rates were relatively even but high, especially compared to the low event rates for the outcomes of interest

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported in detail

Szafranski 2003

Methods

Design: Randomised, double‐blind, placebo‐controlled, parallel‐group, multi‐centre study

12 month treatment period

Conducted in 89 centres from 11 countries

Participants

Participants: 812 people were randomly assigned to budesonide (198), placebo (205), budesonide/formoterol (208) and formoterol (201)
Baseline characteristics:

Male %: bud 80, placebo 83, bud/form 76, form 76

Mean age (range), years: bud 64 (40 to 90), placebo 65 (47 to 92), bud/form 64 (41 to 82), form 63 (40 to 90)

Smoking history (mean pack‐years (SD not reported)): bud 44, placebo 45, bud/form 44, form 45

Mean % predicted FEV1 (SD not reported): bud 37, placebo 36, bud/form 36, form 36

Inclusion criteria: Adults with moderate to severe asthma were included and were selected according to the following criteria: outpatients aged 0 to 40 years; COPD symptoms for 2 years; > 10 pack‐year smoking history; FEV1/FVC 70%; FEV1 50% predicted normal (stages IIB and III according to the GOLD classification); total symptom score of two per day during at least 7 days of the run‐in period; documented use of short‐acting inhaled bronchodilators for reliever medication; > 1 severe COPD exacerbation within 2 to 12 months before the first clinic visit
Exclusion criteria: History of asthma and/or seasonal allergic rhinitis before the age of 40; any relevant cardiovascular disorders as judged by the investigator; using beta‐blocking agents; current respiratory tract disorders other than COPD or any other significant diseases or disorders that may have put them at risk or that may have influenced the results of the study; requirement for regular use of oxygen therapy or an exacerbation during run‐in. Individuals for whom it would have been considered unethical to withdraw inhaled steroids were also excluded

Interventions

Run‐in: 2‐week run‐in period

Treatments:

1. Budesonide 400 mcg twice daily (Pulmicort)

2. Placebo twice daily

3. Budesonide/formoterol 320/9 mcg twice daily (Symbicort)

4. Formoterol 9 mcg twice daily (Oxis)

Inhaler device: as above

Co‐treatment: Only study medication was allowed during the treatment period, plus terbutaline 0.5 mg when needed as reliever medication

Outcomes

Primary: severe exacerbations and FEV1

Secondary: VC and PEF, health‐related quality of life, diary card data, reliever medication use, mild exacerbations, adverse events and clinical chemistry

Notes

Funding: AstraZeneca

Definition of pneumonia: Not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Total of 812 participants were randomly assigned (no other details, industry‐sponsored)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigator)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawal high and uneven between groups (formoterol 31.8%, placebo 43.9%, 31% ICS and 28% LABA/ICS). An intention‐to‐treat analysis was used, but imputation methods were unclear for some outcomes

Selective reporting (reporting bias)

Unclear risk

Could not locate trial registration to check protocol adherence—pneumonia outcomes not reported. Difficulty contacting study author to clarify

Tashkin 2008 SHINE

Methods

Design: randomised, double‐blind, double‐dummy, placebo‐controlled, parallel‐group, multi‐centre study

Six‐month treatment period

Conducted at 194 centres in the US, Czech Republic, the Netherlands, Poland and South Africa

Participants

Participants: 1704 people were randomly assigned to six groups, but the three ICS/LABA groups were merged for analysis. Participant numbers in the four categories were as follows: budesonide (275), placebo (300), budesonide/formoterol (845) and formoterol (284)
Baseline characteristics:

Male %: bud 67.6, placebo 69.0, bud/form 68.9, form 65.5

Mean age (SD), years: bud 63.4 (8.8), placebo 63.2 (9.6), bud/form 63.5, form 63.5 (9.5)

Smoking history (mean pack‐years (SD not reported)): bud 41, placebo 40, bud/form 41, form 40

Mean % predicted FEV1 (SD): bud 39.7 (12.0), placebo 41.3 (12.1), bud/form 39.4, form 39.6 (12.8)

Inclusion criteria: Current smokers or ex‐smokers older than 40 years of age with a clinical diagnosis of COPD and symptoms for longer than two years were eligible for this study. Participants were required to have a history of at least one COPD exacerbation treated with a course of oral corticosteroids and/or antibacterials with documented use of an inhaled short‐acting bronchodilator as rescue medication one to 12 months before screening. Prebronchodilator FEV1 < 50% of predicted normal and pre‐bronchodilator FEV1/FVC < 70% were required at screening. Smoking history of at least 10 pack‐years, score two or higher on the Modified Medical Research Council dyspnoea scale at the time of screening and a breathlessness, cough and sputum scale score of 2 or higher per day for at least half of the 2‐week run‐in period
Exclusion criteria: Individuals were excluded if they had any of the following conditions: history of asthma, history of allergic rhinitis before 40 years of age, significant/unstable CV disorder, clinically significant respiratory tract disorder other than COPD and homozygous alpha1‐antitrypsin deficiency or any other clinically significant co‐morbidities. Individuals were excluded if they needed additions or alterations to their usual COPD maintenance therapy or an increment in rescue therapy because of worsening symptoms within 30 days before screening. Oral or ophthalmic non‐cardioselective beta‐adrenoceptor antagonists, oral corticosteroids, pregnancy and breast‐feeding also were exclusionary

Interventions

Run‐in: 2‐week run‐in period

Treatments:

1. Budesonide 160 mcg × 2 inhalations (320 mcg) twice daily

2. Placebo twice daily

3. Budesonide/formoterol 160/4.5 mcg × 2 inhalations (320/9 mcg) twice daily OR

Budesonide/formoterol 80/4.5 mcg × 2 inhalations (160/9 mcg) twice daily OR

Budesonide 160 mcg × 2 inhalations (320 mcg) twice daily plus formoterol DPI 4.5 mcg × 2 inhalations (9 mcg) twice daily

4. Formoterol DPI 4.5 mcg × 2 inhalations (9 mcg) twice daily

Inhaler device: 1 and 3, pressurised metered‐dose inhaler. 4, dry powder inhaler. 2, unclear

Co‐treatment: Participants were allowed the following concomitant medications during the study period: ephedrine‐free antitussives and mucolytics, nasal corticosteroids, stable‐dose non‐nebulised ipratropium bromide, oral or ophthalmic cardioselective beta‐adrenoceptor antagonists or study‐provided salbutamol as rescue medication. The following medications were allowed for exacerbations after randomisation: oral and parenteral corticosteroids, short‐term use of xanthines, increased use of inhaled beta2‐adrenoceptor agonists and ipratropium bromide, nebulised beta2‐adrenoceptor agonists and ipratropium bromide

Outcomes

Primary: predose FEV1 and 1 hour postdose FEV1

Secondary: 12‐hour spirometry, predose and 1 hour postdose morning and evening PEF, dyspnoea, health‐related quality of life, COPD exacerbations, breathlessness diary and symptom scores, use of rescue medication, adverse events, serious adverse events and mortality

Notes

Funding: AstraZeneca

Clinicantrials.gov identifier: NCT00206154

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Eligible participants were randomly assigned in balanced blocks according to a computer‐generated randomisation scheme at each site

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. To maintain blinding, participants received both a pressurised metered‐dose inhaler (pMDI) and a dry powder inhaler (DPI) containing active treatment or placebo (PL), or combinations of active treatment and placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

ECG results were evaluated by a cardiologist in a blinded fashion through an independent ECG service provider. Unclear for other outcomes

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawal rates lower in combination groups (14.8% combined) than in other groups (22.9%, 25.7% and 21.5% for ICS, placebo and LABA, respectively). ITT analysis used with last observation carried forward for missing values

Selective reporting (reporting bias)

Low risk

Checked against protocol and contacted study authors. All stated outcomes reported in full

van Grunsven 2003

Methods

Design: randomised, placebo‐controlled, double‐blind trial

24‐Month treatment period

Conducted at 10 general practices in Holland

Participants

Participants: 48 people were randomly assigned to fluticasone (24) and placebo (24)
Baseline characteristics:

Male %: flut 50, placebo 54

Mean age (SD), years: flut 46 (10), placebo 47 (11)

Smoking history (mean (SD) pack‐years): flut 11.9 (9.5), placebo 5.8 (8.4)

Mean % predicted FEV1 (SD): flut 95 (18), placebo 98 (17)

Inclusion criteria: chronic cough and/or sputum production for at least three consecutive months and showed an annual decline in pre‐bronchodilator FEV1 of 40 to 80 mL
Exclusion criteria: previous diagnosis of a pulmonary condition; presence of a co‐morbid condition with reduced life expectancy; intolerance for inhaled beta2‐agonists; use of beta‐blocking agents; inability to use inhalation devices or peak flow meters

Interventions

Run‐in: general population screened, followed by 2‐year monitoring of participants with respiratory symptoms, then randomisation

Treatments:

1. Fluticasone propionate 250 mcg twice daily

2. Placebo twice daily

Inhaler device: Rotadisk dry powder inhaler

Co‐treatment: Apart from short‐acting ("rescue") bronchodilators in case of acute dyspnoea, participants were not allowed to use other pulmonary medication

Outcomes

Primary outcome: FEV1

Secondary outcomes: PC20, exacerbations, COOP/WONCA

Notes

Funding: GSK

Definition of pneumonia: not given

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised. No details but assumed to adhere to usual GSK methods

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout high but even between groups (25%). All participants with at least one follow‐up measurement for the primary outcome (postbronchodilator FEV1) were included in an intention to‐treat analysis

Selective reporting (reporting bias)

High risk

Key outcomes not reported (e.g. mortality). SAEs not given per arm. No reply from study authors by time of publication

Verhoeven 2002

Methods

Design: randomised, double‐blind, placebo‐controlled study

Six‐month treatment period

Single study centre in the Netherlands

Participants

Participants: 23 people were randomly assigned to fluticasone (10) and placebo (13)
Baseline characteristics:

Male %: flut 80, placebo 84.6

Mean age (range), years: flut 54 (42 to 65), placebo 56 (42 to 67)

Smoking history (mean (range) pack‐years): flut 25 (five to 50), placebo 26 (11 to 50)

Mean % predicted FEV1 (range): flut 66 (55 to 93), placebo 61 (34 to 72)

Inclusion criteria: chronic productive cough, FEV1 < 70% of predicted normal value, FEV1 reversibility of < 10% predicted after 750 mg terbutaline administered by metered‐dose inhalation, negative serological examination (Phadiatop test) and negative skin prick tests for standard inhaled allergens. Individuals with an FEV1/inspiratory vital capacity (IVC) ratio of < 0.70 were also included, provided their total lung capacity (TLC) was greater than the predicted value + 1.64 SD. Participants had to be current and persistent smokers 40 to 70 years of age
Exclusion criteria: history of asthma characterised by attacks of dyspnoea, chest tightness or wheezing; respiratory tract infection in the 4 weeks preceding the first visit; or suffering from serious or unstable concomitant disease

Interventions

Run‐in: two weeks

Treatments:

1. Fluticasone propionate 500 mcg twice daily

2. Placebo

Inhaler device: Diskhaler

Co‐treatment: Eligible participants using anti‐inflammatory treatment including non‐steroidal anti‐inflammatory drugs were asked to refrain from oral prescriptions for at least three months and from inhaled corticosteroids, sodium cromoglycate or nedocromil sodium for at least 6 weeks before the start of the study. Long‐acting beta2‐agonists, xanthine derivatives and antihistamine drugs also had to be stopped at least 6 weeks before the start of the study

Outcomes

Primary outcome unclear. Outcomes reported were use of secondary medication, compliance, FEV1, PC20, FEV1/FVC, cortisol levels and inflammatory markers

Notes

Funding: GlaxoWellcome (FLIL44/FMS40060)

Definition of pneumonia: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly allocated (no details, but industry‐funded)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts in either group

Selective reporting (reporting bias)

Low risk

Adverse event data not adequately reported but information supplied by the study author

Vestbo 1999

Methods

Design: double‐blind, parallel‐group, randomised clinical trial nested in a continuing epidemiological study, the Copenhagen City Heart Study (CCHS)

36‐Month treatment period

Conducted at a single centre in Denmark

Participants

Participants: 290 people were randomly assigned to budesonide (145) and placebo (145)
Baseline characteristics:

Male %: bud 85, placebo 90

Mean age (SD), years: bud 59 (8.3), placebo 59.1 (9.7)

Smoking history (mean (SD) pack‐years): not reported

Mean % predicted FEV1 (SD): bud 86.2 (20.6), placebo 86.9 (21.1)

Inclusion criteria: CCHS participant; 30 to 70 years of age; FEV1/vital capacity ratio 0.7 or less; FEV1 reversibility after inhalation of 1·0 mg terbutaline from Turbuhaler (Bricanyl, Lund, Sweden) of less than 15% of pre‐bronchodilator FEV1; FEV1 reversibility after 10 days of treatment with oral prednisolone 37.5 mg daily of less than 15% of pre‐bronchodilator FEV1; and informed consent. Pack‐years and other measures of cigarette smoking were not part of inclusion criteria
Exclusion criteria: Long‐term treatment (more than two episodes of longer than 4 weeks) with oral or inhaled steroids within 6 months of study entry. Other exclusion criteria were pregnancy or lactation, intention to become pregnant, other serious systemic disease that could influence the results of this study (investigators’ judgement), chronic alcohol or drug use and participation in other clinical studies of COPD within 1 month of inclusion

Interventions

Run‐in: not described

Treatments:

1. First six months: budesonide 800 mcg am and 400 mcg pm; following 30 months: 400 mcg twice daily

2. Placebo twice daily

Inhaler device: Turbuhaler

Co‐treatment: Continuous use of inhaled corticosteroids other than study medication was not allowed. Oral, inhaled or parenteral steroids could be used during exacerbations for up to three periods of four weeks each year. Treatment with beta2‐agonists of all kinds, theophylline, disodium cromoglycate and mucolytics was allowed but kept constant. Concomitant use of beta‐blockers during the study was not allowed

Outcomes

Primary: spirometric indices (FEV1, VC, FVC)

Secondary: respiratory symptoms (e.g. wheeze, wheeze without a cold, breathlessness at rest and at different grades of exertion, cough night and day, phlegm night and day, chest tightness), exacerbations, chronic mucus hypersecretion, adverse events

Notes

Funding: AstraZeneca

Definition of pneumonia: not given

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was masked and the randomisation sequence generated by computer at Astra. Study numbers were allocated in a consecutive order

Allocation concealment (selection bias)

Low risk

The randomisation code was held by Astra and was not available to the researchers until the study had been completed.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. All study inhalers (budesonide and placebo) had the same appearance.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropout higher in placebo group (35% vs 25% in budesonide group). ITT used.

Selective reporting (reporting bias)

Unclear risk

All stated outcomes reported but unable to check against trial registration. Difficulty contacting authors.

Yildiz 2004

Methods

Design: randomised, double‐blind, placebo‐controlled design

Three‐month treatment period

Conducted at a single centre in Turkey

Participants

Participants: 38 people randomly assigned to budesonide plus existing bronchodilator therapy (20) and placebo (18)
Baseline characteristics:

Male %: bud 100, placebo 100

Mean age (SD), years: bud 70 (7), placebo 64 (9)

Smoking history (mean (SD) pack‐years): bud 55 (31), placebo 47.5 (18)

Mean % predicted FEV1 (SD): bud 51 (22), placebo 40 (14)

Inclusion criteria: pre‐bronchodilator FEV1 between 30% and 80% of predicted and FEV1/FVC < 70% (stage II according to the GOLD classification), irreversible airway obstruction suggested by < 10% improvement in FEV1 after inhalation of 200 mg salbutamol, smoking history of more than 20 pack‐years and no exacerbation or respiratory tract infection in the previous four weeks
Exclusion criteria: history suggestive of asthma, clinical signs of right heart failure, recent hospitalisation or admission to the emergency department because of exacerbation, requirement for regular use of oxygen therapy or used inhaled or oral ICS in the past six weeks

Interventions

Run‐in: no information

Treatments:

1. 800 mcg budesonide twice daily plus existing bronchodilator therapy

2. Placebo twice daily plus existing bronchodilator therapy

Inhaler device: Miflonide inhaler, Novartis

Co‐treatment: All participants were receiving combined bronchodilator therapy consisting of inhaled long‐acting beta2‐agonist (Formoterol, Foradil Aerolizer, Novartis) plus inhaled anticholinergic (ipratropium bromide, Atrovent inhaler, Boehringer Ingelheim)

Outcomes

Primary: unclear which outcome was primary

Secondary: St. George's Respiratory Questionnaire, FEV1, arterial blood gas analysis, serious adverse events, exacerbations

Notes

Funding: unclear

Definition of pneumonia: not given

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

'Randomised'. No other details, funding unclear

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind (presumed participants and personnel/investigators)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawal rates were low (2/18 placebo, 0/20 ICS)

Selective reporting (reporting bias)

Low risk

All outcomes were reported except one secondary outcome—not deemed to reflect bias. Difficulty finding correct contact details for study author

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

GSK FLIP63 2005

Treatment period less than 12 weeks

GSK SAM30022 2005

Comparison not of interest (beclomethasone vs salmeterol/fluticasone combination)

GSK SAM40004 2006

Asthma, not COPD

GSK SAS40015 2007

Comparison not of interest (fluticasone plus oral montelukast vs salmeterol/fluticasone combination)

GSK SCO100540 2006

Comparison not of interest (salmeterol/fluticasone combination vs placebo)

GSK SCO100646 2008

Comparison not of interest (participants received salmeterol and salmeterol/fluticasone combination for varying amounts of time during the study)

GSK SCO30005 2006

Comparison not of interest (salmeterol/fluticasone combination vs placebo)

GSK SCO40030 2005

Treatment period less than 12 weeks

GSK SCO40034 2009

Comparison not of interest (salmeterol/fluticasone combination vs tiotropium)

GSK SCO40036 2009

Comparison not of interest (salmeterol/fluticasone combination vs tiotropium)

GSK SFCB3019 2004

Asthma, not COPD

Lung Health Study 2000

Comparison not of interest (triamcinolone acetonide used as inhaled steroid)

van der Valk 2002

ICS discontinuation study; all participants received treatment for three months before randomisation

Weir 1999

Comparison not of interest (beclomethasone used as inhaled steroid)

Wouters 2005

ICS discontinuation study; all participants received treatment for three months before randomisation

Characteristics of studies awaiting assessment [ordered by study ID]

Ohar 2013

Methods

Six‐month randomised controlled trial

Participants

Participants were > 40 years of age with a historical FEV1/FVC < 0.7. Six‐month history of hospitalisation attributed to AECOPD was also required

Interventions

Fluticasone/salmeterol combination or salmeterol alone within 14 days of an exacerbation event: < 10‐day hospitalisation for AECOPD, or AECOPD requiring treatment with OCS or OCS + antibiotics in an emergency department, or during a physician’s office visit (if the index event was office‐based)

Outcomes

Exacerbation rates

Notes

Abstract only

Characteristics of ongoing studies [ordered by study ID]

Vestbo 2013

Trial name or title

The Study to Understand Mortality and Morbidity in COPD (SUMMIT) study protocol (NCT01313676)

Methods

Design: multi‐centre, placebo‐controlled, double‐blind, randomised, parallel‐group trial

15‐ to 44‐month treatment period (duration of treatment phase depended on mortality rate in the study; the study will last until 1000 deaths have been recorded)

Participants

Inclusion criteria: male or female, 40 to 80 years of age
Current smokers or ex‐smokers with a smoking history of at least 10 pack‐years
Established history of COPD with FEV1/FVC ratio 0.70 and FEV1 greater than 50 and less than 70% of predicted normal
History of CVD or at increased risk for CVD
For participants older than 40 years of age, this is defined as any one of the following: established coronary artery disease, established peripheral vascular disease, previous stroke, previous myocardial infarction or diabetes mellitus with target organ disease
For participants older than 60 years of age, any one of the above or two of the following: treated for hypercholesterolaemia, treated for hypertension, treated for diabetes mellitus or treated for peripheral vascular disease
Exclusion criteria: current diagnosis of asthma or respiratory disorders other than COPD
Chest radiograph indicating diagnosis other than COPD
Undergone lung volume reduction surgery and/or lung transplant
Requirement for long‐term oxygen therapy at start of study (12 hours per day)
Receiving long‐term oral corticosteroid therapy
Current severe heart failure (NYHA class IV); individuals will also be excluded if they have a known ejection fraction of 30% or if they have an implantable cardioverter‐defibrillator
Any life‐threatening condition with life expectancy of three years, other than vascular disease or COPD, that might prevent the individual from completing the study
End‐stage chronic renal disease

Interventions

Run‐in: 4 to 10 days

Treatments:

1. Placebo;

2. Fluticasone furoate (100 mcg once daily)

3. Vilanterol (25 mcg)

4. Fluticasone furoate/vilanterol combination (100/25 mcg once daily)

Inhaler device: novel dry powder inhaler

Co‐treatment: All prior use of ICS and inhaled long‐acting bronchodilators will be discontinued at entry to the run‐in period

Outcomes

Primary: mortality

Secondary: decline in FEV1 and effect on a composite cardiovascular endpoint

Starting date

Recruitment commenced in March 2011 and was ongoing in March 2013

Contact information

J. Vestbo, Department of Respiratory Medicine J, Odense University Hospital, Sdr Ringvej 29, 5000 Odense C, Denmark
E‐mail: [email protected]

Notes

None

Data and analyses

Open in table viewer
Comparison 1. Fluticasone versus controls (all outcomes by treatment)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.78 [1.50, 2.12]

Analysis 1.1

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

1.1 Fluticasone versus placebo

11

6635

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.39, 2.44]

1.2 Fluticasone/LABA versus LABA

13

12869

Odds Ratio (M‐H, Fixed, 95% CI)

1.75 [1.41, 2.17]

2 Mortality, all‐cause Show forest plot

22

20861

Odds Ratio (M‐H, Fixed, 95% CI)

0.99 [0.87, 1.13]

Analysis 1.2

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

2.1 Fluticasone versus placebo

15

7857

Odds Ratio (M‐H, Fixed, 95% CI)

1.05 [0.88, 1.25]

2.2 Fluticasone/LABA versus LABA

14

13004

Odds Ratio (M‐H, Fixed, 95% CI)

0.94 [0.78, 1.12]

3 Mortality, due to pneumonia Show forest plot

18

19532

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.23 [0.70, 2.15]

Analysis 1.3

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

3.1 Fluticasone versus placebo

12

6665

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.20 [0.52, 2.77]

3.2 Fluticasone/LABA versus LABA

13

12867

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.25 [0.59, 2.65]

4 Non‐fatal, serious adverse events (all) Show forest plot

19

20381

Odds Ratio (M‐H, Fixed, 95% CI)

1.06 [0.99, 1.14]

Analysis 1.4

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

4.1 Fluticasone versus placebo

12

7377

Odds Ratio (M‐H, Fixed, 95% CI)

1.07 [0.95, 1.20]

4.2 Fluticasone/LABA versus LABA

14

13004

Odds Ratio (M‐H, Fixed, 95% CI)

1.06 [0.96, 1.16]

5 All pneumonia events Show forest plot

11

15377

Odds Ratio (M‐H, Fixed, 95% CI)

1.68 [1.49, 1.90]

Analysis 1.5

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

5.1 Fluticasone versus placebo

6

4971

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.33, 1.97]

5.2 Fluticasone/LABA versus LABA

9

10406

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [1.47, 2.01]

6 Withdrawals Show forest plot

26

21243

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.77, 0.86]

Analysis 1.6

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

6.1 Fluticasone versus placebo

18

8227

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.70, 0.84]

6.2 Fluticasone/LABA versus LABA

15

13016

Odds Ratio (M‐H, Fixed, 95% CI)

0.85 [0.79, 0.92]

Open in table viewer
Comparison 2. Subgroup analyses—fluticasone versus controls

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dose—Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.76 [1.48, 2.08]

Analysis 2.1

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 1 Dose—Non‐fatal, serious adverse pneumonia events.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 1 Dose—Non‐fatal, serious adverse pneumonia events.

1.1 Fluticasone propionate 500 mcg (250 mcg bid)

6

3857

Odds Ratio (M‐H, Fixed, 95% CI)

1.46 [0.91, 2.36]

1.2 Fluticasone propionate 1000 mcg (500 mcg bid)

9

10138

Odds Ratio (M‐H, Fixed, 95% CI)

1.78 [1.47, 2.16]

1.3 Fluticasone furoate 50 mcg

2

1366

Odds Ratio (M‐H, Fixed, 95% CI)

2.10 [0.73, 6.06]

1.4 Fluticasone furoate 100 mcg

3

2447

Odds Ratio (M‐H, Fixed, 95% CI)

1.61 [0.70, 3.70]

1.5 Fluticasone furoate 200 mcg

2

1696

Odds Ratio (M‐H, Fixed, 95% CI)

2.38 [0.87, 6.51]

2 Duration—Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [1.51, 2.12]

Analysis 2.2

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 2 Duration—Non‐fatal, serious adverse pneumonia events.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 2 Duration—Non‐fatal, serious adverse pneumonia events.

2.1 Duration ≤ one year

14

13078

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [1.39, 2.63]

2.2 Duration > one year

3

6426

Odds Ratio (M‐H, Fixed, 95% CI)

1.74 [1.42, 2.13]

3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events Show forest plot

12

17211

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.53, 2.17]

Analysis 2.3

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events.

3.1 FEV1 < 50% predicted

10

17133

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.55, 2.20]

3.2 FEV1 ≥ 50% predicted

2

78

Odds Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.53]

Open in table viewer
Comparison 3. Budesonide versus controls (all outcomes by treatment)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal, serious adverse pneumonia events Show forest plot

7

6472

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.00, 2.62]

Analysis 3.1

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

1.1 Budesonide versus placebo

3

867

Odds Ratio (M‐H, Fixed, 95% CI)

3.47 [1.11, 10.83]

1.2 Budesonide/formoterol versus formoterol

5

5605

Odds Ratio (M‐H, Fixed, 95% CI)

1.33 [0.78, 2.28]

2 Mortality, all‐cause Show forest plot

12

10009

Odds Ratio (M‐H, Fixed, 95% CI)

0.90 [0.65, 1.24]

Analysis 3.2

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

2.1 Budesonide versus placebo

8

3487

Odds Ratio (M‐H, Fixed, 95% CI)

0.85 [0.52, 1.37]

2.2 Budesonide/formoterol versus formoterol

7

6522

Odds Ratio (M‐H, Fixed, 95% CI)

0.94 [0.61, 1.46]

3 Mortality, due to pneumonia Show forest plot

3

1511

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.46 [0.07, 286.99]

Analysis 3.3

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

3.1 Budesonide versus placebo

2

292

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Budesonide/formoterol versus formoterol

1

1219

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.46 [0.07, 286.99]

4 Non‐fatal, serious adverse events (all) Show forest plot

12

10009

Odds Ratio (M‐H, Random, 95% CI)

1.01 [0.83, 1.22]

Analysis 3.4

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

4.1 Budesonide versus placebo

8

3487

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.69, 1.50]

4.2 Budesonide/formoterol versus formoterol

7

6522

Odds Ratio (M‐H, Random, 95% CI)

0.93 [0.78, 1.11]

5 All pneumonia events Show forest plot

6

7011

Odds Ratio (M‐H, Fixed, 95% CI)

1.12 [0.83, 1.51]

Analysis 3.5

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

5.1 Budesonide versus placebo

3

1378

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.50, 1.50]

5.2 Budesonide/formoterol versus formoterol

5

5633

Odds Ratio (M‐H, Fixed, 95% CI)

1.24 [0.87, 1.77]

6 Withdrawals Show forest plot

15

10150

Odds Ratio (M‐H, Fixed, 95% CI)

0.78 [0.71, 0.85]

Analysis 3.6

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

6.1 Budesonide versus placebo

11

3627

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.69, 0.93]

6.2 Budesonide/formoterol versus formoterol

7

6523

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.67, 0.86]

Open in table viewer
Comparison 4. Subgroup analyses—budesonide versus controls

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dose ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6472

Odds Ratio (M‐H, Fixed, 95% CI)

1.54 [0.96, 2.48]

Analysis 4.1

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 1 Dose ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 1 Dose ‐ Non‐fatal, serious adverse pneumonia events.

1.1 Budesonide 320 mcg (160 mcg bid)

3

1775

Odds Ratio (M‐H, Fixed, 95% CI)

0.68 [0.27, 1.71]

1.2 Budesonide 640 mcg (320 mcg bid)

6

4659

Odds Ratio (M‐H, Fixed, 95% CI)

2.02 [1.15, 3.57]

1.3 Budesonide 1280 mcg (640 mcg bid)

1

38

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Duration ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6471

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.00, 2.62]

Analysis 4.2

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 2 Duration ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 2 Duration ‐ Non‐fatal, serious adverse pneumonia events.

2.1 Duration ≤ one year

6

6217

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [0.83, 2.37]

2.2 Duration > one year

1

254

Odds Ratio (M‐H, Fixed, 95% CI)

3.53 [0.95, 13.15]

3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6471

Odds Ratio (M‐H, Fixed, 95% CI)

1.60 [0.99, 2.59]

Analysis 4.3

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events.

3.1 FEV1 < 50% predicted

6

6217

Odds Ratio (M‐H, Fixed, 95% CI)

1.39 [0.82, 2.34]

3.2 FEV1 ≥ 50% predicted

1

254

Odds Ratio (M‐H, Fixed, 95% CI)

3.53 [0.95, 13.15]

Open in table viewer
Comparison 5. Sensitivity analysis—risk of bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal serious adverse pneumonia events Show forest plot

16

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 Sensitivity analysis—risk of bias, Outcome 1 Non‐fatal serious adverse pneumonia events.

Comparison 5 Sensitivity analysis—risk of bias, Outcome 1 Non‐fatal serious adverse pneumonia events.

1.1 Fluticasone versus control

12

16338

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.52, 2.19]

1.2 Budesonide versus control

4

3515

Odds Ratio (M‐H, Fixed, 95% CI)

3.28 [1.22, 8.81]

Direct and indirect comparisons of fluticasone and budesonide covered in the review.
Figures and Tables -
Figure 1

Direct and indirect comparisons of fluticasone and budesonide covered in the review.

Study flow diagram.
Figures and Tables -
Figure 2

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Summary of pooled effects of trials comparing ICS versus placebo and combination versus LABA.
 Non‐fatal serious adverse pneumonia events were those requiring hospital admission. Data for all pneumonia events were not pooled because of heterogeneity.
Figures and Tables -
Figure 4

Summary of pooled effects of trials comparing ICS versus placebo and combination versus LABA.
Non‐fatal serious adverse pneumonia events were those requiring hospital admission. Data for all pneumonia events were not pooled because of heterogeneity.

Indirect comparisons of fluticasone and budesonide monotherapy.Non‐fatal serious adverse pneumonia events were defined as those requiring hospital admission.
Figures and Tables -
Figure 5

Indirect comparisons of fluticasone and budesonide monotherapy.

Non‐fatal serious adverse pneumonia events were defined as those requiring hospital admission.

Indirect comparisons of fluticasone and budesonide monotherapy—Sensitivity analysis removing (Calverley 2007 TORCH).
Figures and Tables -
Figure 6

Indirect comparisons of fluticasone and budesonide monotherapy—Sensitivity analysis removing (Calverley 2007 TORCH).

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 1.1

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.
Figures and Tables -
Analysis 1.2

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.
Figures and Tables -
Analysis 1.3

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).
Figures and Tables -
Analysis 1.4

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.
Figures and Tables -
Analysis 1.5

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 6 Withdrawals.
Figures and Tables -
Analysis 1.6

Comparison 1 Fluticasone versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 1 Dose—Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 2.1

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 1 Dose—Non‐fatal, serious adverse pneumonia events.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 2 Duration—Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 2.2

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 2 Duration—Non‐fatal, serious adverse pneumonia events.

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 2.3

Comparison 2 Subgroup analyses—fluticasone versus controls, Outcome 3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 3.1

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 1 Non‐fatal, serious adverse pneumonia events.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.
Figures and Tables -
Analysis 3.2

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 2 Mortality, all‐cause.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.
Figures and Tables -
Analysis 3.3

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 3 Mortality, due to pneumonia.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).
Figures and Tables -
Analysis 3.4

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 4 Non‐fatal, serious adverse events (all).

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.
Figures and Tables -
Analysis 3.5

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 5 All pneumonia events.

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 6 Withdrawals.
Figures and Tables -
Analysis 3.6

Comparison 3 Budesonide versus controls (all outcomes by treatment), Outcome 6 Withdrawals.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 1 Dose ‐ Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 4.1

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 1 Dose ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 2 Duration ‐ Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 4.2

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 2 Duration ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events.
Figures and Tables -
Analysis 4.3

Comparison 4 Subgroup analyses—budesonide versus controls, Outcome 3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events.

Comparison 5 Sensitivity analysis—risk of bias, Outcome 1 Non‐fatal serious adverse pneumonia events.
Figures and Tables -
Analysis 5.1

Comparison 5 Sensitivity analysis—risk of bias, Outcome 1 Non‐fatal serious adverse pneumonia events.

Summary of findings for the main comparison. Fluticasone for chronic obstructive pulmonary disease

Fluticasone for chronic obstructive pulmonary disease

Patient or population: patients with chronic obstructive pulmonary disease
Intervention: fluticasone (alone or with LABA co‐intervention)

Comparison: placebo or LABA monotherapy (dependent upon whether fluticasone was given with LABA in the intervention group)

Setting: community

Outcomes

Follow‐ups presented as weighted means

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Fluticasone

Non‐fatal, serious adverse pneumonia events (requiring hospital admission)
Follow‐up: 18 months

25 per 1000

43 per 1000
(37 to 51)

OR 1.78
(1.50 to 2.12)

19,504
(17 studies)

⊕⊕⊕⊕
high

Mortality, all‐cause
Follow‐up: 19 months

58 per 1000

58 per 1000
(51 to 65)

OR 0.99
(0.87 to 1.13)

20,861
(22 studies)

⊕⊕⊕⊕
high

Mortality, due to pneumonia
Follow‐up: 18 months

2 per 1000

3 per 1000
(2 to 5)

OR 1.23
(0.70 to 2.15)

19,532
(18 studies)

⊕⊕⊕⊝
moderate1

Non‐fatal, serious adverse events (all)
Follow‐up: 19 months

227 per 1000

237 per 1000
(225 to 251)

OR 1.06
(0.99 to 1.14)

20,381
(19 studies)

⊕⊕⊕⊕
high

All pneumonia events
Follow‐up: 22 months

72 per 1000

116 per 1000
(104 to 129)

OR 1.68
(1.49 to 1.90)

15,377
(11 studies)

⊕⊕⊕⊝
moderate2

Withdrawals
Follow‐up: 18 months

343 per 1000

297 per 1000
(286 to 310)

OR 0.81
(0.77 to 0.86)

21,243
(26 studies)

⊕⊕⊕⊕
high

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio.

Unless otherwise stated, subgroup differences between monotherapy studies (fluticasone versus placebo) and combination therapy studies (fluticasone/LABA versus LABA) were not significant.

GRADE Working Group grades of evidence.
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Wide confidence intervals include significant benefit and harm, based on very few events (‐1 for imprecision).
2More than half the studies did not report the outcome (‐1 for publication bias).

Figures and Tables -
Summary of findings for the main comparison. Fluticasone for chronic obstructive pulmonary disease
Summary of findings 2. Budesonide for chronic obstructive pulmonary disease

Budesonide for chronic obstructive pulmonary disease

Patient or population: patients with chronic obstructive pulmonary disease
Intervention: budesonide (alone or with LABA co‐intervention)

Comparison: placebo or LABA monotherapy (dependent upon whether fluticasone was given with LABA in the intervention group)

Setting: community

Outcomes

Follow‐ups presented as weighted means

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Budesonide

Non‐fatal, serious adverse pneumonia events (requiring hospital admission)
Follow‐up: 9 months

9 per 1000

15 per 1000
(9 to 24)

OR 1.62
(1.00 to 2.62)

6472
(7 studies)

⊕⊕⊕⊝
moderate1,2

Mortality, all‐cause
Follow‐up: 14 months

17 per 1000

16 per 1000
(11 to 21)

OR 0.90
(0.65 to 1.24)

10,009
(12 studies)

⊕⊕⊕⊝
moderate3

Mortality, due to pneumonia
Follow‐up: 12 months

0 per 1000

0 per 1000
(0 to 0)

OR 4.46
(0.07 to 286.99)

1511
(3 studies)

⊕⊝⊝⊝
very low2, 4

Non‐fatal, serious adverse events (all)
Follow‐up: 14 months

145 per 1000

146 per 1000
(124 to 172)

OR 1.01
(0.83 to 1.22)

10,009
(12 studies)

⊕⊕⊕⊝
moderate5

All pneumonia events
Follow‐up: 10 months

28 per 1000

31 per 1000
(23 to 41)

OR 1.12
(0.83 to 1.51)

7011
(6 studies)

⊕⊕⊕⊝
moderate1,2

Withdrawals
Follow‐up: 14 months

280 per 1000

232 per 1000
(216 to 248)

OR 0.78
(0.71 to 0.85)

10150
(15 studies)

⊕⊕⊕⊕
high

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio.

Unless otherwise stated, subgroup differences between monotherapy studies (budesonide versus placebo) and combination therapy studies (budesonide/LABA versus LABA) were not significant.

GRADE Working Group grades of evidence.
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Confidence intervals are quite wide but are not considered serious enough to downgrade.
2More than half the studies did not report the outcome (‐1 for publication bias).
3Confidence interval includes significant benefit and potential harm.
4Very wide confidence intervals. Only one death observed over the three studies (‐2 for imprecision).
5I2 = 59%, P value 0.002 (‐1 for inconsistency).

Figures and Tables -
Summary of findings 2. Budesonide for chronic obstructive pulmonary disease
Table 1. Fluticasone—summary of studies and baseline characteristics

Study ID

Duration (m)

N Rand

Funder

ICS dose (mcg)

% Male

Mean age

Pack‐years

% pred FEV1

Fluticasone versus placebo (n = 18)

Bourbeau 2007

3

41

GSK

1000

78

65

53

57

Burge 2000

36

740

GSK

1000

75

64

44

50

Calverley 2003 TRISTANa

12

763

GSK

1000

73

63

43

45

Calverley 2007 TORCHa

36

3097

GSK

1000

76

65

49

44

Choudhury 2005

12

260

Indep.

1000

52

67

39

54

GSK FLTA3025 2005

6

640

GSK

500, 1000

69

64

GSK SCO104925 2008a

3

84

GSK

1000

77

64

GSK SCO30002 2005

12

256

GSK

1000

82

65

Hanania 2003a

6

368

GSK

500

63

64

57

42

Hattotuwa 2002

3

36

GSK

1000

87

65

63

46

Kerwin 2013a,b

6

413

GSK

100

66

62

46

42

Lapperre 2009

30

55

GSK

1000

86

60

43

55

Mahler 2002a

6

349

GSK

1000

66

65

55

41

Martinez 2013a,b

6

612

GSK

100, 200

72

62

43

48

Paggiaro 1998

6

281

1000

74

63

57

Schermer 2009

36

190

Indep.

1000

71

59

28

64

van Grunsven 2003

24

48

GSK

500

52

47

9

97

Verhoeven 2002

6

23

GSK

1000

82

55

26

63

WM 22 m

459

72

62

43

54

Fluticasone/LABA combination versus LABA monotherapy (n = 15)

Anzueto 2009

12

797

GSK

500

54

65

57

34

Calverley 2003 TRISTANa

12

731

GSK

1000

73

63

43

45

Calverley 2007 TORCHa

36

3088

GSK

1000

76

65

49

44

Dal Negro 2003

12

12

500

92

42

50

Dransfield 2013b

12

3255

GSK

50, 100, 200

57

64

45

Ferguson 2008

12

782

GSK

500

55

65

56

33

GSK FCO30002 2005

3

140

GSK

1000

66

62

GSK SCO100470 2006

6

1050

GSK

500

78

64

GSK SCO104925 2008a

3

77

GSK

1000

77

64

GSK SCO40041 2008

36

186

GSK

500

61

66

Hanania 2003a

6

355

GSK

500

63

64

57

42

Kardos 2007

10

994

GSK

1000

76

64

37

40

Kerwin 2013a,b

6

617

GSK

100

67

63

46

43

Mahler 2002a

6

325

GSK

1000

66

65

55

41

Martinez 2013a,b

6

610

GSK

100, 200

72

62

43

48

WM 16 m

867

69

64

53

42

aMulti‐arm studies making both comparisons of interest (ICS vs placebo and ICS/LABA vs LABA).

bStudies using vilanterol as the LABA combination and monotherapy comparator, with fluticasone furoate.

Dose is given as the total received per day (i.e. 500 signifies 250 morning and evening).

WM = weighted mean.

Figures and Tables -
Table 1. Fluticasone—summary of studies and baseline characteristics
Table 2. Budesonide—summary of studies and baseline characteristics

Study ID

Duration (m)

N Rand

Funder

ICS dose (mcg)

% Male

Mean age

Pack‐years

% pred FEV1

Budesonide versus placebo (n = 13)

Bourbeau 1998

6

79

AZ

640

79

66

51

37

Calverley 2003ba

12

513

GSK

640

76

64

35

36

Laptseva 2002

6

49

NR

640

NR

NR

NR

NR

Mirici 2001

3

50

NR

640

75

53

27

62

Ozol 2005

6

26

NR

640

69

65

45

59

Pauwels 1999

36

1277

AZ

640

73

52

39

77

Renkema 1996

24

39

AZ

1280

100

55

NR

64

Senderovitz 1999

6

26

NR

640

54

61

NR

NR

Shaker 2009

36

254

AZ

640

58

64

56

52

Szafranski 2003a

12

403

AZ

640

79

64

45

36

Tashkin 2008 SHINEa

6

575

AZ

640

68

63

41

40

Vestbo 1999

36

290

AZ

640

88

59

NR

87

Yildiz 2004

3

38

?

1280

100

67

51

46

WM 23 m

278

77

61

43

54

Budesonide/LABA combination versus LABA monotherapy (n = 7)

Calverley 2003ba

12

509

GSK

640

76

64

35

36

Calverley 2010

11

481

Chiesi

640

81

64

39

42

Fukuchi 2013

3

1293

AZ

640

89

65

44

41

Rennard 2009

12

1483

AZ

320, 640

63

63

NR

39

Sharafkhaneh 2012

12

1219

AZ

320, 640

62

63

44

38

Szafranski 2003a

12

409

AZ

640

79

64

45

36

Tashkin 2008 SHINEa

6

1129

AZ

320, 640

68

63

41

40

WM 9 m

932

75

64

41

39

aMulti‐arm studies making both comparisons of interest (ICS vs placebo and ICS/LABA vs LABA).

Dose is given as the total received per day (i.e. 640 signifies 320 morning and evening).

WM = weighted mean.

Figures and Tables -
Table 2. Budesonide—summary of studies and baseline characteristics
Table 3. BDP equivalent doses

Drug

Daily dose (mcg)

BDP equivalent (mcg)

Budesonide

320

320

640

640

1280

1280

Fluticasone

500 (propionate)

1000

1000 (propionate)

2000

50 (furoate)

500

100 (furoate)

1000

200 (furoate)

2000

Figures and Tables -
Table 3. BDP equivalent doses
Table 4. Control group event rates

Monotherapy comparison—Placebo control events

Combination comparison—LABA control events

Fluticasone

Budesonide

Fluticasone

Budesonide

Pneumonia‐related serious adverse events

2.5%, 77/310

0.9%, 4/445

2.5%, 134/5420

0.9%, 19/2079

0.5% without TORCH

0.7% without TORCH

All‐cause mortality

7.6%, 282/3713

2.1%, 37/1763

5.1%, 254/5489

1.5%, 37/2534

2.4% without TORCH

1.2% without TORCH

All‐cause serious adverse events

25%, 882/3471

15%, 268/1763

21%, 1152/5489

14%, 356/2534

14% without TORCH

14% without TORCH

For the fluticasone control groups with and without the large 3‐year TORCH study.

Figures and Tables -
Table 4. Control group event rates
Comparison 1. Fluticasone versus controls (all outcomes by treatment)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.78 [1.50, 2.12]

1.1 Fluticasone versus placebo

11

6635

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.39, 2.44]

1.2 Fluticasone/LABA versus LABA

13

12869

Odds Ratio (M‐H, Fixed, 95% CI)

1.75 [1.41, 2.17]

2 Mortality, all‐cause Show forest plot

22

20861

Odds Ratio (M‐H, Fixed, 95% CI)

0.99 [0.87, 1.13]

2.1 Fluticasone versus placebo

15

7857

Odds Ratio (M‐H, Fixed, 95% CI)

1.05 [0.88, 1.25]

2.2 Fluticasone/LABA versus LABA

14

13004

Odds Ratio (M‐H, Fixed, 95% CI)

0.94 [0.78, 1.12]

3 Mortality, due to pneumonia Show forest plot

18

19532

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.23 [0.70, 2.15]

3.1 Fluticasone versus placebo

12

6665

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.20 [0.52, 2.77]

3.2 Fluticasone/LABA versus LABA

13

12867

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.25 [0.59, 2.65]

4 Non‐fatal, serious adverse events (all) Show forest plot

19

20381

Odds Ratio (M‐H, Fixed, 95% CI)

1.06 [0.99, 1.14]

4.1 Fluticasone versus placebo

12

7377

Odds Ratio (M‐H, Fixed, 95% CI)

1.07 [0.95, 1.20]

4.2 Fluticasone/LABA versus LABA

14

13004

Odds Ratio (M‐H, Fixed, 95% CI)

1.06 [0.96, 1.16]

5 All pneumonia events Show forest plot

11

15377

Odds Ratio (M‐H, Fixed, 95% CI)

1.68 [1.49, 1.90]

5.1 Fluticasone versus placebo

6

4971

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.33, 1.97]

5.2 Fluticasone/LABA versus LABA

9

10406

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [1.47, 2.01]

6 Withdrawals Show forest plot

26

21243

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.77, 0.86]

6.1 Fluticasone versus placebo

18

8227

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.70, 0.84]

6.2 Fluticasone/LABA versus LABA

15

13016

Odds Ratio (M‐H, Fixed, 95% CI)

0.85 [0.79, 0.92]

Figures and Tables -
Comparison 1. Fluticasone versus controls (all outcomes by treatment)
Comparison 2. Subgroup analyses—fluticasone versus controls

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dose—Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.76 [1.48, 2.08]

1.1 Fluticasone propionate 500 mcg (250 mcg bid)

6

3857

Odds Ratio (M‐H, Fixed, 95% CI)

1.46 [0.91, 2.36]

1.2 Fluticasone propionate 1000 mcg (500 mcg bid)

9

10138

Odds Ratio (M‐H, Fixed, 95% CI)

1.78 [1.47, 2.16]

1.3 Fluticasone furoate 50 mcg

2

1366

Odds Ratio (M‐H, Fixed, 95% CI)

2.10 [0.73, 6.06]

1.4 Fluticasone furoate 100 mcg

3

2447

Odds Ratio (M‐H, Fixed, 95% CI)

1.61 [0.70, 3.70]

1.5 Fluticasone furoate 200 mcg

2

1696

Odds Ratio (M‐H, Fixed, 95% CI)

2.38 [0.87, 6.51]

2 Duration—Non‐fatal, serious adverse pneumonia events Show forest plot

17

19504

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [1.51, 2.12]

2.1 Duration ≤ one year

14

13078

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [1.39, 2.63]

2.2 Duration > one year

3

6426

Odds Ratio (M‐H, Fixed, 95% CI)

1.74 [1.42, 2.13]

3 % FEV1 predicted normal—Non‐fatal, serious adverse pneumonia events Show forest plot

12

17211

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.53, 2.17]

3.1 FEV1 < 50% predicted

10

17133

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.55, 2.20]

3.2 FEV1 ≥ 50% predicted

2

78

Odds Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.53]

Figures and Tables -
Comparison 2. Subgroup analyses—fluticasone versus controls
Comparison 3. Budesonide versus controls (all outcomes by treatment)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal, serious adverse pneumonia events Show forest plot

7

6472

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.00, 2.62]

1.1 Budesonide versus placebo

3

867

Odds Ratio (M‐H, Fixed, 95% CI)

3.47 [1.11, 10.83]

1.2 Budesonide/formoterol versus formoterol

5

5605

Odds Ratio (M‐H, Fixed, 95% CI)

1.33 [0.78, 2.28]

2 Mortality, all‐cause Show forest plot

12

10009

Odds Ratio (M‐H, Fixed, 95% CI)

0.90 [0.65, 1.24]

2.1 Budesonide versus placebo

8

3487

Odds Ratio (M‐H, Fixed, 95% CI)

0.85 [0.52, 1.37]

2.2 Budesonide/formoterol versus formoterol

7

6522

Odds Ratio (M‐H, Fixed, 95% CI)

0.94 [0.61, 1.46]

3 Mortality, due to pneumonia Show forest plot

3

1511

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.46 [0.07, 286.99]

3.1 Budesonide versus placebo

2

292

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Budesonide/formoterol versus formoterol

1

1219

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.46 [0.07, 286.99]

4 Non‐fatal, serious adverse events (all) Show forest plot

12

10009

Odds Ratio (M‐H, Random, 95% CI)

1.01 [0.83, 1.22]

4.1 Budesonide versus placebo

8

3487

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.69, 1.50]

4.2 Budesonide/formoterol versus formoterol

7

6522

Odds Ratio (M‐H, Random, 95% CI)

0.93 [0.78, 1.11]

5 All pneumonia events Show forest plot

6

7011

Odds Ratio (M‐H, Fixed, 95% CI)

1.12 [0.83, 1.51]

5.1 Budesonide versus placebo

3

1378

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.50, 1.50]

5.2 Budesonide/formoterol versus formoterol

5

5633

Odds Ratio (M‐H, Fixed, 95% CI)

1.24 [0.87, 1.77]

6 Withdrawals Show forest plot

15

10150

Odds Ratio (M‐H, Fixed, 95% CI)

0.78 [0.71, 0.85]

6.1 Budesonide versus placebo

11

3627

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.69, 0.93]

6.2 Budesonide/formoterol versus formoterol

7

6523

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.67, 0.86]

Figures and Tables -
Comparison 3. Budesonide versus controls (all outcomes by treatment)
Comparison 4. Subgroup analyses—budesonide versus controls

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dose ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6472

Odds Ratio (M‐H, Fixed, 95% CI)

1.54 [0.96, 2.48]

1.1 Budesonide 320 mcg (160 mcg bid)

3

1775

Odds Ratio (M‐H, Fixed, 95% CI)

0.68 [0.27, 1.71]

1.2 Budesonide 640 mcg (320 mcg bid)

6

4659

Odds Ratio (M‐H, Fixed, 95% CI)

2.02 [1.15, 3.57]

1.3 Budesonide 1280 mcg (640 mcg bid)

1

38

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Duration ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6471

Odds Ratio (M‐H, Fixed, 95% CI)

1.62 [1.00, 2.62]

2.1 Duration ≤ one year

6

6217

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [0.83, 2.37]

2.2 Duration > one year

1

254

Odds Ratio (M‐H, Fixed, 95% CI)

3.53 [0.95, 13.15]

3 % FEV1 predicted normal ‐ Non‐fatal, serious adverse pneumonia events Show forest plot

7

6471

Odds Ratio (M‐H, Fixed, 95% CI)

1.60 [0.99, 2.59]

3.1 FEV1 < 50% predicted

6

6217

Odds Ratio (M‐H, Fixed, 95% CI)

1.39 [0.82, 2.34]

3.2 FEV1 ≥ 50% predicted

1

254

Odds Ratio (M‐H, Fixed, 95% CI)

3.53 [0.95, 13.15]

Figures and Tables -
Comparison 4. Subgroup analyses—budesonide versus controls
Comparison 5. Sensitivity analysis—risk of bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Non‐fatal serious adverse pneumonia events Show forest plot

16

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Fluticasone versus control

12

16338

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.52, 2.19]

1.2 Budesonide versus control

4

3515

Odds Ratio (M‐H, Fixed, 95% CI)

3.28 [1.22, 8.81]

Figures and Tables -
Comparison 5. Sensitivity analysis—risk of bias