Scolaris Content Display Scolaris Content Display

Vilanterol dan fluticasone furoate untuk rawatan asma

Contraer todo Desplegar todo

Referencias

Allen 2013 {published data only}

Allen A, Schenkenberger I, Trivedi R, Cole J, Hicks W, Gul N, et al. Inhaled fluticasone furoate/vilanterol does not affect hypothalamic‐pituitary‐adrenal axis function in adolescent and adult asthma: randomised, double‐blind, placebo‐controlled study. Clinical Respiratory Journal 2013;7(4):397‐406. CENTRAL
NCT01086410. Safety study of the effects of inhaled fluticasone furoate/GW642444 on the hypothalamic‐pituitary‐adrenal (HPA) axis. https://clinicaltrials.gov/ct2/show/NCT01086410 (accessed 17/6/15). CENTRAL

Bateman 2014 {published data only}

Bateman ED, O'Byrne PM, Busse WW, Lotvall J, Bleecker ER, Andersen L, et al. Once‐daily fluticasone furoate (FF)/vilanterol reduces risk of severe exacerbations in asthma versus FF alone. Thorax 2014;69(4):312‐9. CENTRAL
NCT01086384. Asthma Exacerbation Study. http://clinicaltrials.gov/ct2/show/results/NCT01086384 (accessed 17/6/15). CENTRAL

Bernstein 2014 {published data only}

Bernstein DI, Bateman ED, Woodcock A, Toler WT, Forth R, Jacques L, et al. Efficacy and safety of once‐daily fluticasone furoate/vilanterol (FF/VI) and FF over 12 weeks in patients with persistent asthma (Abstract). American Journal of Respiratory and Critical Care Medicine 2014;189:A6671. [CENTRAL: 1035521; CRS: 4900126000023032]CENTRAL
EUCTR2012‐002797‐32‐DE. Efficacy/safety study of fluticasone furoate/vilanterol combination and fluticasone furoate in adult and adolescent asthmatics. http://apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2012‐002797‐32‐DE (accessed 18/6/15). CENTRAL
NCT01686633. A randomized, double‐blind, parallel group, multicenter study of fluticasone furoate/vilanterol 200/25 mcg inhalation powder, fluticasone furoate/vilanterol 100/25 mcg inhalation powder, and fluticasone furoate 100 mcg inhalation powder in the treatment of persistent asthma in adults and adolescents. http://clinicaltrials.gov/show/NCT01686633 (accessed 18/6/15). CENTRAL

Bleecker 2012 {published data only}

HZA106827: A randomised, double‐blind, placebo‐controlled (with rescue medication), parallel group multicentre study of Fluticasone Furoate/GW642444 Inhalation Powder and Fluticasone Furoate Inhalation Powder alone in the treatment of persistent asthma in adults and adolescents. http://www.gsk‐clinicalstudyregister.com/study/106827#ps (accessed 17/6/15). CENTRAL
Study HZA106827: Efficacy/safety study of fluticasone furoate/vilanterol (GW642444) in adult and adolescent asthmatics. http://clinicaltrials.gov/show/NCT01165138 (accessed 17/6/15). CENTRAL
Bleecker ER, Lotvall J, O'Bryne PM, Woodcock A, Busse WW, Forth R, et al. Efficacy of fluticasone furoate (FF) as a monotherapy and in combination with vilanterol (VI) over 12 weeks in patients with persistent asthma [Abstract]. European Respiratory Society 22nd Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:370s [P2091]. CENTRAL
Bleecker ER, Lotvall J, O'Byrne PM, Woodcock A, Busse WW, Kerwin EM, et al. Fluticasone furoate‐vilanterol 100‐25 mcg compared with fluticasone furoate 100 mcg in asthma: a randomized trial. Journal of Allergy and Clinical Immunology: In Practice 2014;2(5):553‐61. [CENTRAL: 1017358; CRS: 4900126000021864; EMBASE: 2014708281; PUBMED: 25213048]CENTRAL
Svedsater H, Jacques L, Goldfrad C, Bleecker ER, O'Byrne PM, Woodcock A. Ease of use of a two‐strip dry powder inhaler (DPI) to deliver fluticasone furoate/vilanterol (FF/VI) and FF alone in asthma. European Respiratory Journal 2013;42(Suppl 57):128‐9. [CRS: 4900132000000863; EMBASE: 71842452]CENTRAL

Busse 2013 {published data only}

Busse WW, O'Byrne PM, Bleecker ER, Lotvall J, Woodcock A, Andersen L, et al. Safety and tolerability of the novel inhaled corticosteroid (ICS) fluticasone furoate (FF) in combination with the long‐acting beta2 agonist (LABA) vilanterol (VI) administered once daily (OD) in patients with asthma [Abstract]. European Respiratory Society 22nd Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:370s [P2092]. CENTRAL
Busse WW, O'Byrne PM, Bleecker ER, Lotvall J, Woodcock A, Andersen L, et al. Safety and tolerability of the novel inhaled corticosteroid fluticasone furoate in combination with the beta2 agonist vilanterol administered once daily for 52 weeks in patients >=12 years old with asthma: a randomised trial. Thorax 2013;68(6):513‐20. CENTRAL
NCT01018186. Fluticasone furoate/GW642444 inhalation powder long‐term safety study. http://clinical trials.gov/show/NCT01018186 (accessed 17/6/15). CENTRAL

Hojo 2015 {published data only}

Hojo M, Iikura M, Hirashima J, Suzuki M, Izumi S, Sugiyama H. A comparison of anti‐inflammatory effect of once‐daily fluticasone furoate/vilanterol 200/25 with twice‐daily fluticasone propionate/salmeterol 500/50 in severe asthmatics. American Journal of Respiratory and Critical Care Medicine 2015;191(Meeting Abstracts):A4280. [CENTRAL: 1101091; CRS: 4900132000009922; EMBASE: 72052155]CENTRAL

Kempsford 2012 {published data only}

Kempsford R, Oliver A, Tombs L, Bal J. The efficacy of inhaled fluticasone furoate (FF) and vilanterol (VI) administered in combination in asthma is comparable when administered in the morning or evening [Abstract]. European Respiratory Society 22nd Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:370s [P2090]. CENTRAL
Kempsford RD, Oliver A, Bal J, Tombs L, Quinn D. The efficacy of once‐daily fluticasone furoate/vilanterol in asthma is comparable with morning or evening dosing. Respiratory Medicine 2013;107(12):1873‐80. CENTRAL
NCT01287065. A randomised, repeat‐dose, placebo‐controlled, double‐blind study to evaluate and compare the efficacy of fluticasone furoate/vilanterol inhalation powder, when administered either in the morning or in the evening, in male and female asthmatic subjects, 2010. http://clinicaltrials.gov/show/NCT01287065(accessed 9/8/16). CENTRAL

Lee 2014 {published data only}

Evaluate the safety, efficacy and dose response of GSK573719 in combination with fluticasone furoate in subjects with asthma (ILA115938). https://clinicaltrials.gov/ct2/show/NCT01573624accessed Dec 2015. CENTRAL
Lee LA, Yang S, Kerwin E, Trivedi R, Edwards LD, Pascoe S. The effect of fluticasone furoate/umeclidinium in adult patients with asthma: a randomized, dose‐ranging study. Respiratory Medicine 2014;109(1):54‐62. [CENTRAL: 1020064; CRS: 4900126000022534; EMBASE: 2014869631; PUBMED: 25452139]CENTRAL

Lin 2013 {published data only}

Jiangtao L, Crawford J, Jacques L, Stone S. Efficacy and safety of once‐daily fluticasone furoate/vilanterol 200/25 mcg compared with twice‐daily fluticasone propionate 500 mcg in asthma patients of Asian ancestry [Abstract]. Respirology (Carlton, Vic.) 2013;18(Suppl 4):111 [PS107]. CENTRAL
Lin J, Kang J, Lee SH, Wang C, Zhou X, Crawford J, et al. Fluticasone furoate/vilanterol 200/25 mcg in Asian asthma patients: a randomized trial. Respiratory Medicine 2015;109(1):44‐53. [CENTRAL: 1042748; CRS: 4900126000023443; EMBASE: 2014615181; PUBMED: 25524507]CENTRAL
NCT01498653. Evaluating the efficacy and safety of fluticasone furoate/vilanterol trifenatate in the treatment of asthma in adolescent and adult subjects of Asian ancestry. http://clinicaltrials.gov/show/NCT01498653 (accessed 17/6/15). CENTRAL

NCT01134042 {published data only}

NCT01134042. Study HZA106829: Efficacy/safety study of fluticasone furoate/vilanterol (GW642444) in adult and adolescent asthmatics, 2010. http://clinicaltrials.gov/show/NCT01134042(accessed 9/8/16). CENTRAL
O'Byrne PM, Bleecker ER, Bateman ED, Busse WW, Woodcock A, Forth R, et al. Once‐daily fluticasone furoate alone or combined with vilanterol in persistent asthma. European Respiratory Journal 2014;43(3):773‐82. CENTRAL

NCT01453023 {published data only}

NCT01453023. Inhaled fluticasone furoate/vilanterol safety and tolerability, PK and PD study. http://clinicaltrials.gov/show/NCT01453023 (accessed 17/6/15). CENTRAL
Oliver A, VanBuren S, Allen A, Hamilton M, Tombs L, Inamdar A, et al. Tolerability of fluticasone furoate/vilanterol combination therapy in children aged 5 to 11 years with persistent asthma. Clinical Therapeutics 2014;36(6):928‐39.e1. CENTRAL

Oliver 2012 {published data only}

NCT01128569. Randomised study comparing the effects of inhaled fluticasone furoate (FF)/vilanterol (VI; GW642444M) combination and FF on an allergen induced asthmatic response. http://clinicaltrials.gov/show/NCT01128569 (accessed 17/6/15). CENTRAL
Oliver A, Quinn D, Goldfrad C, van Hecke B, Ayer J, Boyce M. Combined fluticasone furoate/vilanterol reduces decline in lung function following inhaled allergen 23h after dosing in adult asthma: a randomised, controlled trial. Clinical and Translational Allergy 2012;2(1):11. CENTRAL
Oliver A, Quinn D, Goldfrad C, van Hecke B, Ayer J, Boyce M. The effect of fluticasone furoate alone and in combination with vilanterol on the early asthmatic response 23 hours after dosing In patients with mild persistent asthma: results from a 28‐day randomised, controlled, cross‐over study [Abstract]. American Journal of Respiratory and Critical Care Medicine 2012;185(Meeting Abstracts):A2758. CENTRAL

Oliver 2013 {published data only}

NCT01128595. Randomised study comparing the effects of inhaled FF/GW642444M combination, FF and GW642444M on an allergen induced asthmatic response. http://clinicaltrials.gov/show/NCT01128595 (accessed 17/6/15). CENTRAL
Oliver A, Bjermer L, Quinn D, Saggu P, Thomas P, Yarnall K, et al. Efficacy of fluticasone furoate (FF) and vilanterol (VI), separately and in combination (FF/VI), in an allergen challenge model [Abstract]. European Respiratory Society 22nd Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:386s [P2161]. CENTRAL
Oliver A, Bjermer L, Quinn D, Saggu P, Thomas P, Yarnall K, et al. Modulation of allergen‐induced bronchoconstriction by fluticasone furoate and vilanterol alone or in combination. Allergy 2013;68(9):1136–42. CENTRAL
Oliver A, Quinn D, Saggu P, Thomas P, Yarnall K, Lootvall J, et al. Fluticasone furoate and vilanterol suppress allergen‐induced bronchial hyper‐responsiveness to methacholine [Abstract]. Allergy 2012;67:452. CENTRAL

Woodcock 2013 {published data only}

NCT01147848. HZA113091 Efficacy and safety of fluticasone furoate/vilanterol (GW642444) in adults and adolescents. http://clinicaltrials.gov/show/NCT01147848 (accessed 18/6/15). CENTRAL
Woodcock A, Bleecker ER, Lotvall J, O'Byrne PM, Bateman ED, Medley H, et al. Efficacy and safety of fluticasone furoate (FF)/vilanterol (VI) compared with fluticasone propionate/salmeterol combination (FP/SAL) in adults and adolescents with persistent asthma [Abstract]. European Respiratory Society 22nd Annual Congress; 2012 Sep 1‐5; Vienna. 2012; Vol. 40, issue Suppl 56:313s [P1795]. CENTRAL
Woodcock A, Bleecker ER, Lötvall J, O'Byrne PM, Bateman ED, Medley H, et al. Efficacy and safety of fluticasone furoate/vilanterol compared with fluticasone propionate/salmeterol combination in adult and adolescent patients with persistent asthma: a randomized trial. Chest 2013;144(4):1222‐9. CENTRAL

References to studies excluded from this review

Calverley 2014 {published data only}

Calverley PMA. Inhaled corticosteroids as a cause of CAP. European Respiratory Monograph 2014;63:234‐42. CENTRAL

Gross 2013 {published data only}

Gross AS, Goldfrad C, Hozawa S, James M, Clifton CS, Sugiyama Y, et al. Ethnic sensitivity assessment of fluticasone furoate (FF)/vilanterol (VI) in asthma patients in Japan and Korea: a pre‐specified subgroup analysis. Respirology 2013;18:157. CENTRAL

Gross 2015 {published data only}

Gross AS, Goldfrad C, Hozawa S, James MH, Clifton CS, Sugiyama Y, et al. Ethnic sensitivity assessment of fluticasone furoate/vilanterol in East Asian asthma patients from randomized double‐blind multicentre Phase IIb/III trials. BMC Pulmonary Medicine 2015;15(1):165. [CENTRAL: 1108375; CRS: 4900132000012363; EMBASE: 20151066568; PUBMED: 26704701]CENTRAL

Hozawa 2016 {published data only}

Hozawa S, Terada M, Haruta Y, Hozawa M. Comparison of early effects of budesonide/formoterol maintenance and reliever therapy with fluticasone furoate/vilanterol for asthma patients requiring step‐up from inhaled corticosteroid monotherapy. Pulmonary Pharmacology & Therapeutics 2016;37:15‐23. [CENTRAL: 1135191; CRS: 4900132000015316; EMBASE: 20160310267; PUBMED: 26850307]CENTRAL

Ishiura 2015 {published data only}

Ishiura Y, Fujimura M, Shiba Y, Ohkura N, Hara J, Kasahara K. A comparison of the efficacy of once‐daily fluticasone furoate/vilanterole with twice‐daily fluticasone propionate/salmeterol in asthma‐COPD overlap syndrome. Pulmonary Pharmacology & Therapeutics 2015;35:28‐33. [CENTRAL: 1101036; CRS: 4900132000009842; EMBASE: 20160065157; PUBMED: 26497109]CENTRAL

Kempsford 2011 {published data only}

Kempsford R, Allen A, Bal J, Rubin D, Tombs L. The effect of ketoconazole on the pharmacokinetics (PK) and pharmacodynamics (PD) of inhaled fluticasone furoate (FF) and vilanterol (VI) administered in combination in healthy subjects [Abstract]. European Respiratory Society 21st Annual Congress; 2011 Sep 24‐28; Amsterdam. 2011; Vol. 38, issue 55:137s [P820]. CENTRAL

Kempsford 2011a {published data only}

Kempsford R, Allen A, Bareille P, Bishop H, Hamilton M, Cheesbrough A. The safety, tolerability, pharmacodynamics and pharmacokinetics of inhaled fluticasone furoate (FF) and vilanterol (VI) are unaffected by administration in combination [Abstract]. European Respiratory Society 21st Annual Congress; 2011 Sep 24‐28; Amsterdam. 2011; Vol. 38, issue 55:138s [P824]. CENTRAL

Kempsford 2012a {published data only}

Kempsford R, Allen A, Kelly K, Saggu P, Crim C. A repeat dose, double‐blind, placebo‐controlled "Thorough QT/QTc Study" to assess the cardiac safety of fluticasone furoate (FF) and vilanterol (VI) administered in combination [Abstract]. American Thoracic Society International Conference; 2012 May 18‐23; San Francisco. 2012; Vol. 185, issue Meeting Abstracts:A2841. CENTRAL

Nakahara 2013 {published data only}

Nakahara N, Wakamatsu A, Kempsford R, Allen A, Yamada M, Nohda S, et al. The safety, pharmacokinetics and pharmacodynamics of a combination of fluticasone furoate and vilanterol in healthy Japanese subjects. International Journal of Clinical Pharmacology and Therapeutics 2013;51(8):660‐71. CENTRAL

NCT00603746 {published data only}

Busse WW, Bleecker ER, Bateman ED, Lotvall J, Forth R, Davis AM. Fluticasone furoate demonstrates efficacy in patients with asthma symptomatic on medium doses of inhaled corticosteroid therapy: an 8‐week, randomised, placebo‐controlled trial. Thorax 2012;67(1):35‐41. CENTRAL

NCT01181895 {published data only}

Lotvall J, Bateman ED, Busse WW, O'Byrne PM, Woodcock A, Toler WT, et al. Comparison of vilanterol, a novel long‐acting beta2 agonist, with placebo and a salmeterol reference arm in asthma uncontrolled by inhaled corticosteroids. Journal of Negative Results in Biomedicine 2014;13(1):9. CENTRAL
NCT01181895. Study B2C112060: A study of the efficacy and safety of vilanterol inhalation powder in adults and adolescents with persistent asthma. EUCTR2010‐020412‐11‐DE http://apps.who.int/trialsearch/Default.aspx(accessed 9/8/16). CENTRAL
NCT01181895. Study B2C112060: A study of the efficacy and safety of vilanterol inhalation powder in adults and adolescents with persistent asthma. https://clinicaltrials.gov/ct2/show/NCT01181895 (accessed 18/6/15). CENTRAL

NCT01213849 {published data only}

An open‐label, randomised, 3‐way crossover single dose study to demonstrate dose proportionality of fluticasone furoate (FF) and equivalence of vilanterol (VI) when administered as FF/VI inhalation powder from the novel dry powder inhaler in healthy subjects. http://clinicaltrials.gov/show/NCT01213849 (accessed 18/6/15). CENTRAL

NCT01435902 {published data only}

NCT01435902. A randomized, double‐blind, double‐dummy, crossover comparison of fluticasone furoate/vilanterol 100/25 mcg once daily versus fluticasone propionate 250 mcg twice daily in asthmatic adolescent and adult subjects with exercise‐induced bronchoconstriction. http://clinicaltrials.gov/show/NCT01435902 (accessed 18/6/15). CENTRAL

NCT01485445 {published data only}

NCT01485445. An open‐label, randomised, replicate, six‐way crossover, single dose study to determine the bioequivalence of fluticasone furoate (FF) inhalation powder (single strip configuration) compared with FF inhalation powder (two strip configuration) and compared with FF/vilanterol (VI) inhalation powder administered via the novel dry powder inhaler. http://clinicaltrials.gov/show/NCT01485445 (accessed 18/6/15). CENTRAL

NCT01573767 {published data only}

An open‐label, non‐randomized pharmacokinetic and safety study of repeat doses of fluticasone furoate and GW642444M combination in healthy subjects and in subjects with severe renal impairment. EUCTR2010‐020826‐17‐CZ http://apps.who.int/trialsearch/Default.aspx(accessed 9/8/16). CENTRAL
NCT01573767 A dose‐ranging study of vilanterol (VI) inhalation powder in children aged 5‐11 years with asthma on a background of Inhaled corticosteroid therapy, 2012. http://clinicaltrials.gov/show/NCT01573767(accessed 9/8/16). CENTRAL
NCT01573767. A dose‐ranging study of Vilanterol (VI) inhalation powder in children aged 5‐11 years with asthma. http://apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2011‐003337‐34‐DE (accessed 18/6/15). CENTRAL
Oliver AJ, Covar RA, Goldfrad CH, Klein RM, Pedersen SE, Sorkness CA, et al. Randomised trial of once‐daily vilanterol in children with asthma on inhaled corticosteroid therapy. Respiratory Research 2016;17(1):37. [CENTRAL: 1139867; CRS: 4900132000017699; EMBASE: 20160268109; PUBMED: 27044326]CENTRAL

NCT01711463 {published data only}

NCT01711463. A randomized, double‐blind, placebo‐controlled, four‐way crossover study to evaluate and compare the pharmacodynamics and pharmacokinetics of fluticasone furoate/vilanterol in different dose combination (50/25mcg, 100/25mcg and 200/25mcg) after single and repeat dose administration from a novel dry powder device in healthy Chinese subjects. http://clinicaltrials.gov/show/NCT01711463 (accessed 18/6/15). CENTRAL

NCT02712047 2016 {published data only}

NCT02712047. A randomised, placebo‐controlled, double‐blind, two period crossover study to characterise the exhaled nitric oxide time profile as a biomarker of airway inflammation in adult asthma patients following repeat administration of inhaled fluticasone furoate (FF)/vilanterol (VI) 100/25 mcg, 2016. https://clinicaltrials.gov/show/NCT02712047(accessed 9/8/16). [CRS: 4900132000023253]CENTRAL

Oliver 2014 {published data only}

EUCTR2012‐000741‐12‐Outside EU/EEA. A study to see if it is safe to give a new asthma reliever drug (called vilanterol) to 5 to 11 year old children with asthma. http://apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2012‐000741‐12‐Outside‐EU/EEA (accessed 18/6/15). CENTRAL
NCT01453296. Pharmacokinetics and pharmacodynamics of GW642444 in paediatric subjects. http://clinicaltrials.gov/ct2/show/NCT01453296 (accessed18/6/15). CENTRAL
Oliver A, Vanburen S, Allen A, Hamilton M, Tombs L, Kempsford R, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of vilanterol, a novel inhaled long‐acting beta‐agonist, in children aged 5‐11 years with persistent asthma: a randomized trial. Clinical Pharmacology in Drug Development 2014;3(3):215‐21. CENTRAL

Sterling 2012 {published data only}

NCT00980200. Efficacy and safety study in subjects with asthma. http://clinicaltrials.gov/show/ NCT00980200 (accessed 18/6/15). CENTRAL
Sterling R, Lim J, Frith L, Snowise NG, Jacques L, Haumann B. Efficacy and optimal dosing interval of the long‐acting beta2 agonist, vilanterol, in persistent asthma: a randomised trial. Respiratory Medicine 2012;106(8):1110‐5. CENTRAL

Woepse 2013 {published data only}

Woepse M, Dale P, Garrill K, Svedsater H, Walker R. Qualitative assessment of a two‐strip dry powder inhaler for chronic obstructive pulmonary disease and asthma [abstract]. Allergy 2013;68(suppl 97):675. CENTRAL

References to studies awaiting assessment

Bardsley 2017 {published data only}

Bardsley G, Daley‐Yates P, Baines A, Riddell K, Joshi S, Bareille PF, et al. The duration of anti‐inflammatory action of fluticasone furoate (FF) assessed via exhaled nitric oxide (FeNO) in asthmatics following administration of FF/vilanterol (VI). European Respiratory Journal 2017;50:OA276. CENTRAL

Bernstein 2017 {published data only}

Bernstein D, Forth R, Yates L, Andersen L, Jacques L. Comparison of once daily fluticasone furoate/vilanterol with twice daily fluticasone propionate/ salmeterol in patients with controlled asthma. Chest OS ‐ Chest. Conference: CHEST 2017 Annual Meeting. Canada. 152 (4 Supplement 1) (pp A186), 2017. Date of Publication: October 2017. 2017, (4):A186. CENTRAL

Braithwaite 2016 {published data only}

Braithwaite I, Williams M, Power S, Pilcher J, Weatherall M, Baines A, et al. Randomised, double‐blind, placebo‐controlled, cross‐over single dose study of the bronchodilator duration of action of combination fluticasone furoate/vilanterol inhaler in adult asthma. Respiratory Medicine 2016;119:115‐21. CENTRAL

Furuhashi 2017 {published data only}

Furuhashi K, Fujisawa T, Hashimoto D, Kamiya Y, Niwa M, Karayama M, et al. Comparison of the clinical effects between once‐daily fluticasone furoate/vilanterole combination and twice‐daily budesonide/formoterol combinations in stable asthma. American Journal of Respiratory and Critical Care Medicine 2017;195:A3198. CENTRAL

Ishiura 2017 {published data only}

Ishiura Y, Fujimura M, Shiba Y, Ohkura N, Hara J, Abo M, et al. A comparison of the efficacy of once‐daily fluticasone furoate/vilanterole with twice‐daily fluticasone propionate/salmeterol in elderly asthmatics. Drug Research 2017, (1):38‐44. CENTRAL

Lee 2017 {published data only}

Lee L, Kerwin E, Collison K, Nelsen L, Wu W, Yang S, et al. The effect of umeclidinium on lung function and symptoms in patients with fixed airflow obstruction and reversibility to salbutamol: a randomised, 3‐phase study. Respiratory Medicine 2017;131:148‐57. CENTRAL

NCT01498679 {published data only}

Lin J, Tang H, Chen P, Wang H, Kim MK, Crawford J, et al. Efficacy and safety evaluation of once‐daily fluticasone furoate/vilanterol in Asian patients with asthma uncontrolled on a low‐ to mid‐strength inhaled corticosteroid or low‐dose inhaled corticosteroid/long‐acting beta2‐agonist. Allergy and Asthma Proceedings 2016;37(4):302‐10. CENTRAL
NCT01498679. A randomised, double‐blind, placebo‐controlled, parallel group, multicentre study to evaluate the efficacy and safety of fluticasone furoate/vilanterol trifenatate (FF/VI) inhalation powder delivered once daily for 12 Weeks in the treatment of asthma in adolescent and adult subjects of Asian ancestry currently treated with low to mid‐strength inhaled corticosteroid or low‐strength combination therapy. http://clinicaltrials.gov/show/NCT01498679 (accessed 18/6/15). CENTRAL

NCT01573624 {published data only}

NCT01573624. A multi‐center, randomized, double‐blind, dose‐ranging study to evaluate GSK573719 in combination with fluticasone furoate, fluticasone furoate alone, and an active control of fluticasone furoate/vilanterol combination in subjects with asthma. http://clinicaltrials.gov/show/NCT01573624 (accessed 18/6/15). CENTRAL

NCT01706198 {published data only}

EUCTR2011‐005553‐31‐GB. Study to evaluate the effectiveness of fluticasone furoate/vilanterol delivered once daily via a novel dry powder inhaler (NDPI) compared with existing asthma maintenance therapy alone in subjects with asthma. http://apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2011‐005553‐31‐GB (accessed 18/6/15). CENTRAL
NCT01706198. A 12‐month, open label, randomised, effectiveness study to evaluate fluticasone furoate (FF, GW685698)/Vilanterol (VI, GW642444) inhalation powder delivered once daily via a novel dry powder inhaler compared with usual maintenance therapy in subjects with asthma. http://clinicaltrials.gov/show/NCT01706198 (accessed 18/6/15). []CENTRAL
Woodcock A, Bakerly ND, New JP, Gibson JM, Wu W, Vestbo J, et al. The Salford Lung Study protocol: a pragmatic, randomised phase III real‐world effectiveness trial in asthma. BMC Pulmonary Medicine 2015;15(1):160. [CENTRAL: 1128497; CRS: 4900132000012287; EMBASE: 20151033372; PUBMED: 26651333]CENTRAL

NCT01837316 {published data only}

NCT01837316. A randomized, double‐blind, placebo‐controlled cross‐over study to determine the bronchodilator effect of a single dose of fluticasone furoate (FF)/vilanterol (VI) 100/25 mcg combination administered in the morning in adult patients with asthma. http://clinicaltrials.gov/show/NCT01837316 (accessed 18/6/15). CENTRAL

NCT02094937 {published data only}

NCT02094937. A study to compare the efficacy and safety of fluticasone furoate (FF) 100 mcg once daily with fluticasone propionate (FP) 250 mcg twice daily (BD) and FP 100 mcg BD in well‐controlled asthmatic Japanese subjects. http://clinicaltrials.gov/ct2/show/NCT02094937 (accessed 19/6/15). CENTRAL

NCT02301975 {published data only}

NCT02301975. An efficacy and safety study of fluticasone furoate/vilanterol 100/25 microgram (mcg) inhalation powder, fluticasone propionate/salmeterol 250/50 mcg inhalation powder, and fluticasone propionate 250 mcg inhalation powder in adults and adolescents with persistent asthma. https://clinicaltrials.gov/ct2/show/NCT02301975 (accessed 19/6/15). CENTRAL

NCT02301975 2015 {published data only}

Bernstein D, Andersen L, Forth R, Jacques L, Yates L. Once‐daily fluticasone furoate/vilanterol versus twice‐daily fluticasone propionate/salmeterol in patients with asthma well controlled on ICS/LABA. Journal of Asthma 2017:1‐10. CENTRAL
NCT02301975. A randomized, double‐blind, double‐dummy, parallel group, multicenter study of once daily fluticasone furoate/vilanterol 100/25 mcg inhalation powder, twice daily fluticasone propionate/salmeterol 250/50 mcg inhalation powder, and twice daily fluticasone propionate 250 mcg inhalation powder in the treatment of persistent asthma in adults and adolescents already adequately controlled on twice‐daily inhaled corticosteroid and long‐acting beta2 agonist, 2015. https://clinicaltrials.gov/show/NCT02301975(accessed 9/8/16). [CRS: 4900132000009126]CENTRAL

NCT02446418 2015 {published data only}

NCT02446418. A 6‐month, open label, randomised, efficacy study to evaluate fluticasone furoate (FF, GW685698)/vilanterol (VI, GW642444) inhalation powder delivered once daily via the dry powder inhaler ELLIPTA compared with usual ICS/LABA maintenance therapy delivered by dry powder inhaler in subjects with persistent asthma, 2015. https://clinicaltrials.gov/show/NCT02446418 (accessed 9/8/2016). [CRS: 4900132000009127]CENTRAL

NCT02730351 2016 {published data only}

NCT02730351. A randomised, double‐blind, double‐dummy, crossover comparison of fluticasone furoate/vilanterol 100/25 mcg once daily versus fluticasone propionate 250 mcg twice daily in adolescent and adult subjects with asthma and exercise‐induced bronchoconstriction, 2016. https://clinicaltrials.gov/show/NCT02730351 (accessed 9/8/2016). [CRS: 4900132000023254]CENTRAL

NCT02753712 2016 {published data only}

NCT02753712. A two‐arm, randomised, assessor‐blind, parallel group study to evaluate the effect of fluticasone/formoterol breath actuated inhaler (BAI) and Relvar Ellipta DPI on ventilation heterogeneity in subjects with partially controlled or uncontrolled asthma. https://clinicaltrials.gov/show/NCT02753712 (accessed 9/8/2016). [CRS: 4900132000023255]CENTRAL

New 2014 (NCT01551758) {published data only}

New JP, Bakerly ND, Leather D, Woodcock A. Obtaining real‐world evidence: the Salford Lung Study. [][Erratum appears in Thorax. 2015 Oct;70(10):1008; PMID: 26371090]. Thorax 2014;69(12):1152‐4. [CENTRAL: 1035662; CRS: 4900126000023176; EMBASE: 2014630882; PUBMED: 24603195]CENTRAL

Woodcock 2014 (NCT01706198) {published data only}

Anonymous. Erratum: effectiveness of fluticasone furoate plus vilanterol on asthma control in clinical practice: an open‐label, parallel group, randomised controlled trial. Lancet 2017;390(10109):e40. CENTRAL
Woodcock A, Bakerly ND, New JP, Gibson JM, Wu W, Vestbo J, et al. The Salford Lung Study protocol: a phase III real‐world effectiveness trial in asthma (Abstract). American Journal of Respiratory and Critical Care Medicine 2014;189:A1407. [CENTRAL: 1035657; CRS: 4900126000023169]CENTRAL
Woodcock A, Vestbo J, Bakerly ND, New J, Gibson JM, McCorkindale S, et al. Effectiveness of fluticasone furoate plus vilanterol on asthma control in clinical practice: an open‐label, parallel group, randomised controlled trial. Lancet 2017, (10109):2247‐55. CENTRAL

Barnes 1998

Barnes PJ. Current issues for establishing inhaled corticosteroids as the antiinflammatory agents of choice in asthma. Journal of Allergy and Clinical Immunology 1998;101(4):S427‐33.

Bisgaard 2006

Bisgaard H, Le Roux P, Bjamer D, Dymek A, Vermeulen JH, Hultquist C. Budesonide/formoterol maintenance plus reliever therapy: a new strategy in pediatric asthma. Chest 2006;130(6):1733‐43.

Bleecker 2011

Bleecker ER, Bateman ED, Busse WW, Lotvall J, Woodcock A, Frith L, et al. Consistently favourable safety profile of fluticasone furoate (FF), a once‐daily (OD) inhaled corticosteroid (ICS), across a range of treatment steps in patients with uncontrolled asthma. American Thoracic Society International Conference; 2011 May 13‐18; Denver. 2011.

Braman 2006

Braman SS. The global burden of asthma. Chest 2006;130(1 Suppl):4S‐12S.

BTS/SIGN 2014

BTS/SIGN 2014. British guideline on the management of asthma. A national clinical guideline. Revised 2014. http://www.sign.ac.uk/pdf/SIGN141.pdf (accessed 19/6/15)).

Cazzola 2011

Cazzola M, Calzetta L, Matera MG. b2‐adrenoceptor agonists:current and future direction. British Journal of Pharmacology 2011;163(1):4‐17.

CDC 2012

Centers for Disease Control and Prevention. CDC vital signs. Asthma in the US. www.cdc.gov/vitalsigns/asthma/ (accessed 10/12/2012).

CDCP 2011

Centers for Disease Control and Prevention. Trends in asthma prevalence, health care, and mortality in the United States, 2001‐2010. CDC. http://www.cdc.gov/nchs/data/databriefs/db94.htm (accessed 10/12/2012).

Clayton 2005

Clayton S. Paediatric asthma: overcoming barriers to an improved quality of life. British Journal of Nursing2005; Vol. 14, issue 2:80–5.

Eid 2010

Eid NS, Noonan MJ, Chipps B, Parasuraman B, Miller C, O’Brien CD. Once‐ vs twice‐daily budesonide/formoterol in 6‐ to 15‐year‐old patients with stable asthma. Pediatrics 2010;126(3):e565‐75.

Fuso 2013

Fuso L, Mores S, Valente M, Melerba M, Montuschi P. Long‐acting beta‐agonists and their association with inhaled corticosteroids in COPD. Current Medicinal Chemistry 2013;20(12):1477‐95.

GINA 2015

Global Initiative for Asthma (GINA). From the global strategy for asthma management and prevention, global initiative for asthma (GINA) 2015. http://www.ginasthma.org/documents/4 (accessed 19/6/15).

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1 [updated March 2011]. The Cochrane Collaboration, 2011. www.cochrane‐handbook.org.

Kempsford 2013

Kempsford R, Norris V, Siederer S. Vilanterol trifenatate, a novel inhaled long‐acting beta2 adrenoceptor agonist, is well tolerated in healthy subjects and demonstrates prolonged bronchodilation in subjects with asthma and COPD. Pulmonary Pharmacology and Therapeutics 2013;26(2):256‐64.

Krishnan 2006

Krishnan V, Diette GB, Rand CS, Bilderback AL, Merriman B, Hansel NN, et al. Mortality in patients hospitalized for asthma exacerbations in the United States. American Journal of Respiratory and Critical Care Medicine 2006;174(6):633‐8.

Kuna 2006

Kuna P, Creemers JP, Vondra V, Black PN, Lindqvist A, Nihlen U, et al. Once‐daily dosing with budesonide/formoterol compared with twice‐daily budesonide/formoterol and once‐daily budesonide in adults with mild to moderate asthma. Respiratory Medicine 2006;100(12):2151‐9.

Lotvall 2012

Lotvall J, Bateman ED, Bleecker ER, Busse WW, Woodcock A, Followse R, et al. 24‐Hour duration of the novel LABA vilanterol trifenatate in asthma patients treated with inhaled corticosteroids. European Respiratory Journal 2012;40(3):570–9.

Montuschi 2008

Montuschi P. Leukotrienes, antileukotrienes and asthma. Mini Reviews in Medicinal Chemistry 2008;8(7):647‐56.

Montuschi 2010

Montuschi P, Peters‐Golden ML. Leukotriene modifiers for asthma treatment. Clinical and Experimental Allergy 2010;40(12):1732‐41.

Montuschi 2011

Montuschi P, Barnes PJ. New perspectives in pharmacological treatment of mild persistent asthma. Drug Discovery Today 2011;16(23‐24):1084‐91.

Mortaz 2008

Mortaz E, Rad MV, Johnson M, Raats D, Nijkamp FP, Folkerts G. Salmeterol with fluticasone enhances the suppression of IL‐8 release and increases the translocation of glucocorticoid receptor by human neutrophils stimulated with cigarette smoke. Journal of Molecular Medicine 2008;86(9):1045–56.

NHS 2011

NHS. NHS 2011 HES online hospital episode statistics. www.hesonline.nhs.uk (accessed 10/12/2012).

NIH 2007

National Heart, Lung, and Blood Institute. Guidelines for the diagnosis and management of asthma (EPR 3). http://www.nhlbi.nih.gov/health‐pro/guidelines/current/asthma‐guidelines (accessed 10/12/2012).

Nocker 1996

Nocker RE, Schoonbrood DF, van de Graaf EA, Hack CE, Lutter R, Jansen HM, et al. Interleukin‐8 in airway inflammation in patients with asthma and chronic obstructive pulmonary disease. International Archives of Allergy and Immunology 1996;109(2):183‐91.

Ordonez 1998

Ordonez G, Phelan P, Olinsky A, Robertson CF. Preventable factors in hospital admissions for asthma. Archives of Disease in Childhood 1998;78(2):143‐7.

Patel 2008

Patel SP, Jarvelin MR, Little M. Systematic review of worldwide variations of the prevalence of wheezing symptoms in children. Environmental Health 2008;7:57.

Powell 2003

Powell H, Gibson PG. Inhaled corticosteroid doses in asthma: an evidence‐based approach. Medical Journal of Australia 2003;178(5):223‐5.

Remington 2005

Remington TL, DiGiovine B. Long‐acting beta 2‐agonists: anti‐inflammatory properties and synergy with corticosteroids in asthma. Current Opinion in Pulmonary Medicine 2005;11(1):74‐8.

Review Manager 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Usmani 2005

Usmani OS, Ito K, Maneechotesuwan K, Ito M, Johnson M, Barnes PJ, et al. Glucocorticoid receptor nuclear translocation in airway cells after inhaled combination therapy. Amercian Journal of Respiratory and Critical Care Medicine 2005;172(6):704‐12.

Weiner 1995

Weiner P, Weiner M, Azgad Y. Long term clinical comparison of single versus twice daily administration of inhaled budesonide in moderate asthma. Thorax 1995;50(12):1270‐3.

WHO 2007

Bousquet J, Khaltaev N (editors). Vital signs. World Health Organization. Global surveillance, prevention and control of chronic respiratory diseases: a comprehensive approach. WHO Press Geneva Centers for Disease Control and Prevention. www.cdc.gov/vitalsigns/asthma/(accessed September 2013).

WHO 2011

World Health Organization. Health topics: asthma. http://www.who.int/topics/asthma/en/ (accessed 13 September 2011).

Woodcock 2011

Woodcock A, Bateman ED, Busse WW, Lötvall J, Snowise NG, Forth R, et al. Efficacy in asthma of once‐daily treatment with fluticasone furoate: a randomized, placebo controlled trial. Respiratory Research 2011;12(1):132.

Wu 2007

Wu F, Takaro TK. Childhood asthma and environmental interventions. Environmental Health Perspectives 2007;115(6):971–5.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Allen 2013

Methods

Randomised double‐blind multi‐centre trial

Participants

Total sample

N = 185 participants, 177 completed study

FF/VI 100/25 mcg, n = 56 (54 completed study)

FF/VI 200/25 mcg, n = 56 (55 completed study)

Placebo, n = 58 (55 completed study)

Prednisolone, n = 15 (13 completed study)

Age

FF/VI 100/25 mcg, mean 34.4 (SD 15.63)

FF/VI 200/25 mcg, mean 34.0 (SD 13.74)

Placebo, mean 36.1 (SD 15.42)

Prednisolone, mean 37.5 (SD 14.19)

Males

FF/VI 100/25 mcg, 25 (45%)

FF/VI 200/25 mcg, 33 (59%)

Placebo, 31 (53%)

Prednisolone, 9 (60%)

Baseline FEV1 (% predicted)

FF/VI 100/25 mcg, mean 79.9 (SD 12.58)

FF/VI 200/25 mcg, mean 77.5 (SD 13.22)

Placebo, mean 77.0 (SD 11.88)

Prednisolone, mean 78.6 (SD 13.17)

Inclusion criteria

  • Outpatient with ability to comply with study requirements and complete two 24‐hour clinic visits

  • Clinical diagnosis of asthma for ≥ 12 weeks

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL

  • FEV1 ≥ 50% of predicted

Exclusion criteria

  • History of life‐threatening asthma

  • Respiratory infection or oral candidiasis

  • Asthma exacerbation

  • Uncontrolled disease or clinical abnormality

  • Allergies to study drugs, study drugs' excipients, medications related to study drugs

  • Taking another investigational medication or prohibited medication

Interventions

Arm 1: FF/VI dose 100/25 mcg inhalation powder once‐daily for 6 weeks' treatment + 1 oral placebo capsule each day on the last 7 days of the study

Arm 2: FF/VI 200/25 mcg inhalation powder once‐daily for 6 weeks' treatment + 1 oral placebo capsule each day on the last 7 days of the study

Arm 3: placebo inhalation powder once‐daily for 6 weeks' treatment + 1 oral placebo capsule each day on the last 7 days of the study

Arm 4: placebo inhalation powder once‐daily for 6 weeks' treatment + 1 oral prednisolone 10 mg capsule each day on the last 7 days of the study

Outcomes

Primary outcome

  • Ratio from baseline of serum cortisol weighted mean (0‐24 hours) at day ‐1/1 (baseline) and day 42

Secondary outcomes

  • Ratio from baseline of serum cortisol area under concentration‐time curve (AUC) (0‐24 hours) at day ‐1/1 (baseline) and day 42

  • Ratio from baseline of serum cortisol trough (0‐24 hours) at day ‐1/1 (baseline) and day 42

  • Ratio from baseline of 0 to 24 hours urinary free cortisol excretion at day ‐1/1 (baseline) and day 42

  • Plasma FF and VI PK concentration

  • AUC(0‐t) and AUC(0‐24) for FF at day 42

  • Cmax for FF at day 42

  • Tmax and Tlast of FF at day 42

  • AUC(0‐t) for VI at day 42

  • Cmax for VI at day 42

  • Tmax and Tlast of VI at day 42

  • Number of participants with any AE or SAE during treatment period

  • Change from baseline in basophil, eosinophil, lymphocyte, monocyte and segmented neutrophil values at day 42/early withdrawal

  • Change from baseline in eosinophil, total neutrophil, platelet and white blood cell (WBC) count values at day 42/early withdrawal

  • Change from baseline in haemoglobin values at day 42/early withdrawal

  • Change from baseline in haematocrit values at day 42/early withdrawal

  • Change from baseline in ALT, ALP, AST, CK and GGT values at day 42/early withdrawal

  • Change from baseline in albumin and total protein values at day 42/early withdrawal

  • Change from baseline in direct bilirubin, indirect bilirubin, total bilirubin and creatinine values at day 42/early withdrawal

  • Change from baseline in chloride, CO2 content/bicarbonate, glucose, potassium, sodium, and urea/BUN values at day 42/early withdrawal

  • Change from baseline in SBP and DBP at days 14, 28 and 42, and maximum post baseline

  • Change from baseline in pulse rate at days 14, 28 and 42, and maximum post baseline

Notes

Data collected from 17 locations in Germany (6), Poland (7) and USA (4)

Funded by GlaxoSmithKline

Study duration: 6 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline, Stevenage, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System (GlaxoSmithKline)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. Placebo inhalers and capsules were identical in appearance to active treatments

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind. Placebo inhalers and capsules were identical in appearance to active treatments

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of the 8 withdrawals included in NCT01086410 report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Bateman 2014

Methods

Randomised double‐blind parallel‐group trial

Participants

Total sample

N = 2020 participants, 1748 completed study

FF/VI 100/25, n = 1009 (885 completed study)

FF 100, n = 1011 (863 completed study)

Age

FF/VI 100/25, mean 41.1 (SD 17.10)

FF 100, mean 42.3 (SD 16.82)

Males

FF/VI 100/25, 348 (34%)

FF 100, 321 (32%)

Baseline FEV1 (% predicted)

FF/VI 100/25, mean 24.4 (SD 12.71)

FF 100, mean 24.3 (SD 12.10)

Inclusion criteria

  • Clinical diagnosis of asthma

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL and greater approximately 10 to 40 minutes following 2 to 4 inhalations of albuterol

  • FEV1 50% to 90% of predicted

  • Currently using ICS therapy

  • History of ≥ 1 asthma exacerbations requiring treatment with oral/systemic corticosteroids or emergency department visit or in‐patient hospitalisation in previous year

Exclusion criteria

  • History of life‐threatening asthma in previous 5 years (requiring intubation and/or associated with hypercapnia, hypoxic seizure or respiratory arrest)

  • Respiratory infection or oral candidiasis

  • Uncontrolled disease or clinical abnormality

  • Allergies

  • Taking another investigational medication or prohibited medication

Interventions

Arm 1: FF/VI dose 100/25 mcg inhalation powder inhaled orally once‐daily in the evening

Arm 2: FF dose 100 mcg inhalation powder inhaled orally once‐daily in the evening

Outcomes

Primary outcome

  • Number of participants with ≥ 1 SAE

Secondary outcomes

  • Number of SAEs

  • Change from baseline in evening pre‐dose trough FEV1 at week 36

Notes

Data collected from 182 locations in Argentina (9), Australia (6), Germany (28), Japan (14), Mexico (6), Philippines (5), Poland (15), Romania (6), Russian Federation (16), Ukraine (13) and USA (64)

Funded by GlaxoSmithKline

Study duration: variable (≥ 24 to 78 weeks)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline, Stevenage, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System (GlaxoSmithKline)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Described as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information on 271 participants failing to complete study included in trial report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Bernstein 2014

Methods

Randomised double‐blind parallel‐group multi‐centre study

Participants

Total sample

N = 2019 screened, N = 1039 randomised, 956 completed study

FF/VI 100/25, n = 346 (314 completed study)

FF 100, n = 347 (296 completed study)

FF/VI 200/25, n = 346 (321 completed study)

Age

FF/VI 100/25, mean 45.9 (SD 16.14)

FF 100, mean 44.7 (SD 15.89)

FF/VI 200/25, mean 46.6 (SD 14.72)

Males

FF/VI 100/25, 141 (40.75%)

FF 100, 148 (42.65%)

FF/VI 200/25, 122 (35.26%)

Baseline FEV1 (% predicted)

Not reported

Inclusion criteria

  • > 12 years of age

  • FEV1 40% to 80% of predicted

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL and greater approximately 10 to 40 minutes following 4 inhalations of salbutamol/albuterol

  • Received ICS for > 12 weeks before study

Exclusion criteria

  • History of life‐threatening asthma within past 5 years

  • Unresolved upper or lower respiratory tract, sinus or middle ear infection in past 4 weeks that led to a change in asthma management

  • Asthma exacerbation requiring oral corticosteroids in 12 weeks before visit 1, or exacerbation resulting in overnight hospitalisation and additional asthma treatment in 6 months before visit 1

  • Evidence of atelectasis (segmental or larger), bronchopulmonary dysplasia, chronic obstructive pulmonary disease or concurrent respiratory disease

  • Any clinically significant, uncontrolled condition or disease state

  • Chronic stable hepatitis B or C if screening alanine transaminase (ALT) is > 2 > upper limit of normal (ULN)

  • Chronic co‐infection with hepatitis B and hepatitis C

  • Clinical visual evidence of candidiasis

  • Use of any investigational drug within 30 days before visit 1, or within 5 half‐lives (t½), whichever is longer of the 2

  • Allergies or adverse reactions to drug or milk protein: any adverse reaction to any beta2‐agonist, sympathomimetic drug or intranasal, inhaled or systemic corticosteroid therapy

  • Taking another investigational medication or prohibited medication

  • Current smoker or smoking history of 10 pack‐years or used inhaled tobacco products within the past 3 months

  • Shift workers

Interventions

Randomised 1:1:1

Arm 1: FF/VI dose 100/25 mcg inhalation powder inhaled orally once‐daily in the evening

Arm 2: FF 100 mcg inhalation powder inhaled orally once‐daily in the evening

Arm 3: FF/VI dose 200/25 mcg inhalation powder inhaled orally once‐daily in the evening

Outcomes

Primary endpoint

Weighted mean (WM) serial FEV1 0 to 24 hours post dose at week 12

Secondary endpoints

  • Change from baseline in trough FEV1

  • Change from baseline in % rescue‐free 24‐hour periods

  • Change from baseline in % symptom‐free 24‐hour periods

  • Change from baseline in morning and evening PEF

Adverse events were assessed throughout the study

Notes

Data collected from 137 locations in Argentina (13), Chile (7), Germany (12), Mexico (4), Netherlands (7), Poland (8), Romania (13), Russian Federation (19), Sweden (5), Ukraine (12) and USA (37)

Funded by GlaxoSmithKline

Study duration: 12 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised through a validated computerised system (RandAll Version 2.5, GlaxoSmithKline)

Allocation concealment (selection bias)

Low risk

The Registration and Medication Ordering System was used to register and randomise participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Described as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information on 108 participants failing to complete study included in NCT01686633 report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Bleecker 2012

Methods

Randomised double‐blind placebo‐controlled parallel‐group multi‐centre trial

Participants

Total sample

N = 609 participants, 515 completed study

FF/VI 100/25, n = 201 (179 completed study)

FF 100, n = 205 (185 completed study)

Placebo, n = 203 (151 completed study)

Age

FF/VI 100/25, mean 40.7 (SD 16.38)

FF 100, mean 40.4 (SD 16.78)

Placebo, mean 38.1 (SD 16.49)

Males

FF/VI 100/25, 85 (42%)

FF 100, 79 (39%)

Placebo, 92 (45%)

Baseline FEV1 (% predicted)

Not reported

Inclusion criteria

  • Outpatients 12 years of age or older

  • Male and female; female participants of childbearing potential must be willing to use birth control

  • Pre‐bronchodilator FEV1 40% to 90% of predicted normal

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL

  • Current asthma therapy includes ICS use for ≥ 12 weeks before first visit

Exclusion criteria

  • History of life‐threatening asthma during past 10 years

  • Respiratory infection or oral candidiasis

  • Asthma exacerbation requiring OCS or overnight hospitalisation with additional asthma treatment

  • Uncontrolled disease or clinical abnormality

  • Allergies to study drugs or to excipients

  • Taking another investigational or prohibited medication

  • Night shift workers

  • Current smokers or participants with a smoking history of 10 or more pack‐years

Interventions

Arm 1: FF/VI dose 100/25 mcg inhalation powder inhaled orally once‐daily for 12 weeks

Arm 2: FF dose 100 mcg inhalation powder inhaled orally once‐daily for 12 weeks

Arm 3: Placebo inhalation powder inhaled orally once‐daily for 12 weeks

Outcomes

Primary outcomes

  • Mean change from baseline in clinic visit trough (pre‐bronchodilator and pre‐dose) FEV1 at week 12

  • Change from baseline in weighted mean serial FEV1 over 0 to 24 hours post dose at week 12

Secondary outcomes

  • Mean change from baseline in % of rescue‐free 24‐hour periods during 12‐week treatment period

  • Change from baseline in % of symptom‐free 24‐hour periods during 12‐week treatment period

  • Change from baseline in total AQLQ (+12) score at week 12/early withdrawal

  • Number of participants who withdrew owing to lack of efficacy

  • Serial FEV1 over 0 to 1 hour post dose at randomisation

  • Clinic visit 12‐hour post‐dose FEV1 at week 12

  • Weighted mean serial FEV1 over 0 to 24 hours post dose at baseline

  • Weighted mean serial FEV1 over 0 to 4 hours post dose at baseline and at week 12

  • Number of participants with bronchodilator effect

  • Mean change from baseline in daily AM PEF averaged over 12‐week treatment period

  • Mean change from baseline in daily PM PEF averaged over 12‐week treatment period

  • Change from baseline in ACT score at week 12

  • Number of participants with indicated global assessment of change responses at week 4, week 8 and week 12/early withdrawal

  • Number of indicated unscheduled asthma‐related healthcare visits during treatment period

  • Number of participants who used the inhaler correctly or incorrectly at baseline, week 2 and week 4

  • Number of participants with indicated reason for incorrect inhaler use who required additional instruction indicated number of times at baseline, week 2 and week 4

Notes

Data collected from 12 sites in Poland (1), Ukraine (10) and USA (1)

Funded by GlaxoSmithKline

Study duration: 12 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Details not reported

Allocation concealment (selection bias)

Unclear risk

Details not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Described as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of 94 participants withdrawn from the study are included in NCT01165138 report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Busse 2013

Methods

Randomised double‐blind double‐dummy parallel‐group multi‐centre trial

Participants

Total sample

N = 503 participants, 393 completed study

FF/VI 100/25, n = 201 (161 completed study)

FF/VI 200/25, n = 202 (161 completed study)

FP 500 mcg twice‐daily, n = 100 (71 completed study)

Age

FF/VI 100/25, mean 39.7 (SD 15.85)

FF/VI 200/25, mean 38.5 (SD 15.64)

FP 500 mcg twice‐daily, 38.6 (SD 15.97)

Males

FF/VI 100/25, 71 (35%)

FF/VI 200/25, 78 (39%)

FP 500 mcg twice‐daily, 38 (38%)

Baseline FEV1 (% predicted)

FF/VI 100/25, mean 74.2 (SD 13.48)

FF/VI 200/25, mean 74.1 (SD 14.13)

FP 500 mcg twice‐daily, mean 75.2 (SD 12.46)

Inclusion criteria

  • Clinical diagnosis of asthma

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL and greater approximately 10 to 40 minutes following 2 to 4 inhalations of albuterol

  • FEV1 ≥ 50% of predicted

  • Currently using moderate‐ to high‐dose ICS therapy

Exclusion criteria

  • History of life‐threatening asthma

  • Respiratory infection or oral candidiasis

  • Asthma exacerbation

  • Uncontrolled disease or clinical abnormality

  • Allergies

  • Taking another investigational or prohibited medication

Interventions

Arm 1: FF/VI 100/25 mcg once‐daily

Arm 2: FF/VI 200/25 mcg once‐daily

Arm 3: FP 500 mcg twice‐daily

Outcomes

Primary outcome measures

  • Number of participants with any AE or SAE during treatment period

  • Number of participants with severe asthma exacerbations during treatment period

  • Change from baseline in albumin and total protein at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in ALP, ALT, AST, CK and GGT at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in direct bilirubin, indirect bilirubin, total bilirubin and creatinine at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in chloride, CO2 content/bicarbonate, glucose, potassium, sodium and urea/BUN at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in % of basophils, eosinophils, haematocrit, lymphocytes, monocytes and segmented neutrophils in the blood at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in eosinophil count, total ANC, platelet count and WBC count at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in haematocrit at week 12, week 28 and week 52/early withdrawal

  • Change from baseline in haemoglobin at week 12, week 28 and week 52/early withdrawal

  • Number of participants with indicated shift from baseline to high, normal or no change, and with low post‐baseline values for urinary cortisol excretion

  • Ratio of 24‐hour urinary cortisol excretion at week 12 to baseline, week 28 to baseline and week 52 to baseline

  • Number of participants with evidence of oral candidiasis during treatment period

  • Maximum change from baseline in SBP and minimum change from baseline in DBP

  • Maximum change from baseline in pulse rate

  • Number of participants with indicated change from baseline in LOCS III posterior subcapsular opacity (P) at week 28 and week 52

  • Number of participants with indicated change from baseline in IOP at week 28 and week 52

  • Change from baseline in horizontal cup‐to‐disc ratio at week 28 and week 52

  • Number of participants with indicated change from baseline in LOCS III cortical opacity (C) at week 28 and week 52

  • Change from baseline in LOCS III nuclear colour (NC) at week 28 and week 52

  • Change from baseline in LOCS III nuclear opalescence (NO) at week 28 and week 52

  • Change from baseline in LogMAR visual acuity at week 28 and week 52

  • Maximum change from baseline in QTcB and QTcF

  • Mean 24‐hour Holter heart rate for participants with 16 or more hours of recorded data

  • Maximum 24‐hour Holter heart rate for participants with 16 or more hours of recorded data

Notes

Data collected from 46 sites in Germany (15), Thailand (4), Ukraine (9) and USA (18)

Funded by GlaxoSmithKline

Study duration: 52 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via an automated telephone‐based registration and medication ordering system (Registration and Medication Ordering System (RAMOS))

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of 110 participants withdrawn from the study are provided in the ClinicalTrials.gov NCT01018186 report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Hojo 2015

Methods

Randomised cross‐over trial

Participants

Total sample: 32 adults

Age: participants over 20 years of age with severe asthma. Mean age, 62.2 years (SD 13.3)

Baseline: % FEV1 mean, 70 (SD 11.9%); ACT mean, 20.3 (SD 2.79)

ppb at time of entry to trial suggested relatively poor asthma control status

Interventions

Sequence 1: FF/VI 100/25 once‐daily vs FP/salmeterol 500/50 twice‐daily

Sequence 2: FP/salmeterol 500/50 twice‐daily vs FF/VI 100/25 once‐daily

Participants randomised to receive 4 weeks of treatment followed by 4‐week washout period, then second 4 weeks of treatment with the remaining intervention

Outcomes

Fractional exhaled nitric oxide (FeNO) measured by NIOX‐MINO

Asthma control test

Morning PEF

Respiratory resistance/reactance measured by Forced Oscillation Technique Mostgraph‐01

Notes

Reported as conference abstract. Minimal information available

Study duration: Each treatment period ran for 4 weeks with a 4‐week washout period between treatment periods

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Details not reported

Allocation concealment (selection bias)

Unclear risk

Details not reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Details not reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Details not reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Details not reported

Selective reporting (reporting bias)

Unclear risk

Details not reported

Kempsford 2012

Methods

Randomised double‐blind cross‐over trial

Participants

Total sample

N = 26 participants

Age: mean, 38.1 (SD 11.30)

Males: 18 (69%)

Baseline FEV1 (% predicted): not reported

Inclusion criteria

  • Participants with documented history of persistent asthma, with exclusion of other significant pulmonary disease

  • Male or female between 18 and 70 years of age inclusive

  • A female participant is eligible to participate if she is of non‐childbearing potential. Females on HRT and whose menopausal status is in doubt will be required to use a contraception method if they wish to continue their HRT during the study. Otherwise, they must discontinue HRT to allow confirmation of post‐menopausal status before study enrolment. Childbearing potential and agrees to use one of the protocol contraception methods

  • All participants must be using ICS, with or without SABA, for ≥ 12 weeks before screening

  • Participants with screening pre‐bronchodilator FEV1 ≥ 60% of predicted

  • During screening visit, participants must demonstrate the presence of reversible airway disease

  • All participants must be able to replace all their current asthma treatments with albuterol/salbutamol aerosol inhaler at screening for use as needed for run‐in period and throughout the duration of the study. Participants must be able to withhold albuterol/salbutamol for ≥ 6 hours before study visits

  • Participants who are current non‐smokers, who have not used any inhaled tobacco products in the 12‐month period preceding the screening visit

  • Body weight ≥ 50 kg and BMI within the range of 19.0 to 29.9 kg/m2 (inclusive)

  • No evidence of significant abnormality on the 12‐lead ECG performed at screening

  • AST and ALT < 2 × ULN; alkaline phosphatase and bilirubin ≤ 1.5 × ULN (isolated bilirubin > 1.5 × ULN is acceptable if bilirubin is fractionated and direct bilirubin is < 35%

  • Capable of giving written informed consent

  • Able to satisfactorily use novel DPI

Exclusion criteria

  • History of life‐threatening asthma within past 5 years

  • Culture‐documented or suspected bacterial or viral infection that was not resolved within 4 weeks of screening and led to a change in asthma management or, in the opinion of the investigator, is expected to affect participant's asthma status or ability to participate in the study

  • Any asthma exacerbation requiring OCS within 12 weeks of screening or resulting in overnight hospitalisation with additional treatment for asthma within 6 months before screening

  • Participant with any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation

  • Participant will not be eligible if he/she has clinical visual evidence of oral candidiasis at screening

  • Pregnant females

  • Lactating females

  • Participant has participated in a clinical trial and has received an investigational product within 30 days before first dosing day in the current study

  • Exposure to ≥ 4 new chemical entities within 12 months before first dosing day

  • Any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist, sympathomimetic drug or intranasal, inhaled or systemic corticosteroid therapy

  • History of severe milk protein allergy

  • History of drug or other allergy that, in the opinion of the investigator or the GSK medical monitor, contraindicates participation

  • Use of prescription or non‐prescription drugs within 7 days (or 14 days if the drug is a potential enzyme inducer) or 5 half‐lives (whichever is longer) before first dose of study medication, unless in the opinion of the Investigator and the GSK medical monitor, the medication will not interfere with study procedures or compromise participant safety

  • Participants who have taken high doses of an ICS within 8 weeks of screening visit or OCS within 12 weeks of screening visit

  • Participants who have changed their ICS treatment within the past 4 weeks before screening or can be expected to do so during the study

  • History of regular alcohol consumption within 6 months of the study

  • Positive test for hepatitis B or hepatitis C within 3 months of screening

  • Positive breath carbon monoxide (CO) test

  • Positive pre‐study drug/alcohol screen

  • Positive test for HIV antibody

  • When participation in the study would result in donation of blood or blood products in excess of 500 mL within a 56‐day period

  • No participant is permitted to perform night shift work for 1 week before screening until completion of study treatment periods

  • Unwillingness or inability to follow procedures outlined in the protocol

Interventions

Sequence 1: placebo, FF/VI 100/25 mcg AM, FF/VI 100/25 mcg PM

Sequence 2: placebo, FF/VI 100/25 mcg PM, FF/VI 100/25 mcg AM

Sequence 3: FF/VI 100/25 mcg AM, FF/VI 100/25 mcg PM, placebo

Sequence 4: FF/VI 100/25 mcg AM, placebo, FF/VI 100/25 mcg PM

Sequence 5: FF/VI 100/25 mcg PM, placebo, FF/VI 100/25 mcg AM

Sequence 6: FF/VI 100/25 mcg PM, FF/VI 100/25 mcg AM, placebo

Participants received all treatments once a day in the evening from a DPI for 14 days. Each 14‐day treatment period was followed by a 14 to 21‐day washout period

Outcomes

Primary outcome

  • Weighted mean FEV1 over 0 to 24 hours post dose on day 14

Secondary outcomes

  • Pre‐treatment PEF (AM and PM) at days 1 to 12

  • AM and PM pre‐treatment trough FEV1 at day 14

  • Number of participants with any AE or SAE

Notes

Data collected from 1 site in New Zealand

Funded by GlaxoSmithKline

Study duration: Each treatment period ran for 14 days, with a 14 to 21‐day washout period between treatment periods

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by GSK
Quantitative sciences using validated internal software

Allocation concealment (selection bias)

Low risk

Investigator or designee received medication assignment information and randomised participants using sequentially numbered containers

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Details of the 2 withdrawals are included in the trial report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Lee 2014

Methods

Randomised double‐blind 3‐period cross‐over incomplete block study

Participants

Total sample

706 screened, N = 421 participants, 323 completed study

Age

Mean, 47.5 (SD 13.84)

Males

132 (31%)

Baseline FEV1(% predicted)

1.847 L (62.31%)

Inclusion criteria

  • Outpatient

  • > 18 years of age

  • Diagnosis of asthma for > 6 months

  • Pre‐bronchodilator FEV1 40% to 80% of predicted

  • Demonstrated reversibility by ≥ 12% and ≥ 200 mL of FEV1 within 40 minutes following albuterol

  • Need for regular controller therapy for minimum of 8 weeks

  • Stable dose of ICS for > 4 weeks

Exclusion criteria

  • History of life‐threatening asthma

  • Respiratory infection not resolved

  • Asthma exacerbation

  • Concurrent respiratory disease

  • Current smoker

  • Uncontrolled disease

  • Positive hepatitis B surface antigen or positive hepatitis C antibody and/or HIV

  • Visual clinical evidence of oropharyngeal candidiasis

  • Drug or milk protein allergies

  • Concomitant medications affecting course of asthma

  • Use of any other investigational medication within 30 days or 5 drug half‐lives (whichever is longer)

  • Previous use of GSK573719

  • Any disease preventing use of anticholinergics

  • Any condition that impairs compliance with study protocol, including visit schedule and completion of daily diaries

  • Any participant with a history of alcohol or substance abuse

Interventions

Arm 1: FF 100 mcg once daily for 14 days

Arm 2: FF/VI 100/25 mcg once daily for 14 days

Arm 3: FF/UMEC 100/variable dose (15.6, 31.25, 62.5, 125, 250 mcg) once daily for 14 days

Outcomes

Primary outcome measure

  • Change from baseline in trough FEV1

Secondary outcome measures

  • Mean change from baseline in daily AM/PM PEF

  • Mean change from baseline in rescue albuterol use

Notes

32 centres ‐ Argentina (6), Chile (7), Russia (11), Thailand (4) and USA (4)

Funded by GlaxoSmithKline

Study duration: Each treatment period ran for 14 days with a 12 to 14‐day washout period between treatment periods

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

In a 3‐period cross‐over study, participants were randomised to a sequence of 3 of 7 treatments using SAS‐generated codes in a validated computerised system (RandAll Version 2.5, GlaxoSmithKline)

Allocation concealment (selection bias)

Low risk

The Registration and Medication Ordering System was used to register and randomise participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinding in all 3 conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blinding in all 3 conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information on 98 participants who failed to complete the study included in the trial report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Lin 2013

Methods

Randomised double‐blind double‐dummy parallel‐group trial

Participants

Total sample

N = 309 participants, 255 completed study

FF/VI 200/25, n = 155 (136 completed study)

FP 500, n = 154 (119 completed study)

Age

FF/VI 200/25, mean 46.9 (SD 12.93)

FP 500, n = 48.8 (SD 13.41)

Males

FF/VI 100/25. 59 (38%)

FP 500. n = 64 (42%)

Baseline FEV1 (% predicted)

FF/VI 100/25, mean 67.51 (SD 13.249)

FP 500, n = 67.55 (SD 13.432)

Inclusion criteria

  • Informed consent: All participants must be able and willing to give written informed consent to take part in the study

  • Type of participant: outpatients, of Asian ancestry, 12 years of age or older at visit 1 (or 18 years of age or older if local regulations or regulatory status of the study medication permits enrolment of adults only) with a diagnosis of asthma as defined by the Global Initiative for Asthma (GINA, 2009) ≥ 12 weeks before visit 1

  • Gender: male or eligible female, defined as non‐childbearing potential or childbearing potential and using an acceptable method of birth control consistently and correctly

  • Severity of disease: best FEV1 40% to 90% of predicted normal value at visit 1 screening visit. Predicted values will be based upon NHANES III using the Asian adjustment

  • Reversibility of disease: demonstrated ≥ 12% and ≥ 200 mL reversibility of FEV1 within 10 to 40 minutes following 2 to 4 inhalations of albuterol/salbutamol inhalation aerosol (or 1 nebulised treatment with albuterol/salbutamol solution) at screening visit

  • Current antiasthma therapy: All participants must be using an ICS, with or without LABA, for ≥ 12 weeks before visit 1

  • SABA: All participants must be able to replace their current SABA with albuterol/salbutamol inhaler at visit 1 for use as needed for the duration of the study. Participants must be able to withhold albuterol/salbutamol for ≥ 4 hours before study visits

Exclusion criteria

  • History of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 10 years

  • Respiratory infection: culture‐documented or suspected bacterial or viral infection of upper or lower respiratory tract, sinus or middle ear that is not resolved within 4 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, is expected to affect participant's asthma status or ability to participate in the study

  • Asthma exacerbation: any asthma exacerbation requiring OCS within 12 weeks of visit 1 or resulting in overnight hospitalisation with additional treatment for asthma within 6 months before visit 1

  • Concurrent respiratory disease: Participant must not have current evidence of pneumonia, pneumothorax, atelectasis, pulmonary fibrotic disease, bronchopulmonary dysplasia, chronic bronchitis, emphysema, COPD or other respiratory abnormalities other than asthma

  • Other concurrent diseases/abnormalities: Participant must not have a clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation, or would confound interpretation of efficacy results if the condition/disease was exacerbated during the study

  • Oropharyngeal examination: Participant will not be eligible for the run‐in if he/she has clinical visual evidence of candidiasis at visit 1

  • Allergies: drug allergy: any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist, sympathomimetic drug or intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to constituents of the new powder inhaler; milk protein allergy: history of severe milk protein allergy

  • Concomitant medications: use of protocol‐defined prohibited medications within prohibited time intervals before screening (visit 1) or during the study

  • Tobacco use: current smoker or smoking history of 10 pack‐years (e.g. 20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products within the past 3 months (i.e. cigarettes, cigars, smokeless or pipe tobacco)

  • Affiliation with investigator's site: Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or is an employee of the participating investigator

  • Previous participation: Participant may not have been randomised previously to treatment in another phase III FF/VI combination product study

  • Compliance: Participant will not be eligible if he/she or his/her parent or legal guardian has any infirmity, disability, disease or geographical location that seems likely (in the opinion of the Investigator) to impair compliance with any aspect of this study protocol, including visit schedule and completion of daily diaries

Interventions

  • Arm 1: FF/VI 200/25 mcg once‐daily

  • Arm 2: FP 500 mcg twice‐daily

Outcomes

Primary outcome

  • Mean change from baseline in daily PM PEF averaged over 12‐week treatment period

Secondary outcomes

  • Mean change from baseline in daily AM PEF averaged over 12‐week treatment period

  • Mean change from baseline in % of rescue‐free 24‐hour periods during 12‐week treatment period

  • Mean change from baseline in % of symptom‐free 24‐hour periods during 12‐week treatment period

  • Change from baseline in total AQLQ score at week 12

Notes

Data collected from 24 sites in China (12), Republic of Korea (10) and Philippines (2)

Funded by GlaxoSmithKline

Study duration: 12 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via an automated telephone‐based registration and medication ordering system (Registration and Medication Ordering System (RAMOS))

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of attrition bias included in trial report. Three participants (1 FF/VI; 2 FP) reported a total of 5 serious adverse events and were withdrawn from the study

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

NCT01134042

Methods

Randomised double‐blind parallel‐group multi‐centre trial

Participants

Total sample

N = 586 participants, 476 completed study

FF 200 mcg once‐daily, n = 198 (146 completed study)

FF/VI 200/25 mcg once‐daily, n = 197 (169 completed study)

FP 500 mcg twice‐daily, n = 195 (161 completed study)

Age

FF 200 mcg once‐daily, mean 44.6 years (SD 14.33)

FF/VI 200/25 mcg once‐daily, mean 46.6 (SD 15.05)

FP 500 mcg twice‐daily, mean 47.3 (SD 14.06 )

Males

FF 200 mcg once‐daily, 81 (42%)

FF/VI 200/25 mcg once‐daily, 81 (42%)

FP 500 mcg twice‐daily, 79 (41%)

Baseline FEV1 (% predicted)

FF 200 mcg once‐daily, mean 66.66 (SD 12.388)

FF/VI 200/25 mcg once‐daily, mean 66.59 (SD 12.614)

FP 500 mcg twice‐daily, mean 67.57 (SD 12.185)

Inclusion criteria

  • Outpatient ≥ 12 years of age

  • Both genders; females of childbearing potential must be willing to use birth control method

  • Pre‐bronchodilator FEV1 40% to 90% of predicted

  • Reversibility FEV1 ≥ 12% and ≥ 200 mL

  • Current asthma therapy that includes an ICS for ≥ 12 weeks before first visit

Exclusion criteria

  • History of life‐threatening asthma

  • Respiratory infection or oral candidiasis

  • Asthma exacerbation within 12 weeks

  • Concurrent respiratory disease or other disease that would confound study participation or affect participant safety

  • Allergies to study drugs, study drug excipients, medications related to study drugs

  • Taking another investigational medication or medication prohibited for use during this study

Interventions

Arm 1: FF 200 mcg once‐daily

Arm 2: FF/VI 200/25 mcg once‐daily

Arm 3: FP 500 mcg twice‐daily

Outcomes

Primary outcomes

  • Change from baseline in clinic visit trough (pre‐bronchodilator and pre‐dose) FEV1 at the end of the 24‐week treatment period

  • Change from baseline in weighted mean serial FEV1 over 0 to 24 hours post dose at week 24

Secondary outcomes

  • Change from baseline in percentage of rescue‐free and symptom‐free 24‐hour periods at the end of the 24‐week treatment period

  • Change from baseline in total AQLQ (+12) score at week 12 and at week 24/early withdrawal

  • Clinic visit 12‐hour post‐dose FEV1 at week 24

  • Change from baseline in weighted mean serial FEV1 over 0 to 4 hours post dose at week 24

  • Mean change from baseline in daily trough AM and PM PEF averaged over first 12 weeks and 24 weeks of 24‐week treatment period

  • Number of participants who withdrew owing to lack of efficacy during 24‐week treatment period

  • Change from baseline in ACT scores at week 12 and at week 24

  • Number of participants with indicated global assessment of change questionnaire responses at weeks 4, 12 and 24

  • Number of indicated unscheduled asthma‐related healthcare visits during treatment period

Notes

Data collected from 71 sites in Germany (10), Japan (12), Poland (8), Romania (7), Russian Federation (11) and USA (23)

Funded by GlaxoSmithKline

Study duration: 24 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated, computerised system (RandAll, GlaxoSmithKline, Stevenage, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System (GlaxoSmithKline)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trial reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information on 110 participants not completing the study is reported at http://clinicaltrials.gov/show/NCT01134042

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

NCT01453023

Methods

Randomised double‐blind cross‐over trial

Participants

Total sample

N = 26 participants, 23 completed study

Age: mean 8.1 years (SD 1.97)

Males: 15 (58%)

Asthma severity, no. (%)

Mild (well controlled with GINA step 2 low‐dose ICS), 21 (84%)

Moderate (well controlled with GINA step 3 medium‐dose ICS), 4 (16%)

Inclusion criteria

  • Healthy as determined by a study physician on the basis of medical history, physical examination, laboratory testing and electrocardiogram (ECG), with no significant medical condition apart from asthma, eczema or rhinitis. Participant with a clinical abnormality or laboratory parameters outside the reference range for this study may be included if the investigator and the GSK medical monitor agree that the finding is not likely to introduce additional risk factors nor interfere with study procedures

  • Male and pre‐menarcheal female participants 5 to less than 12 years of age on last planned treatment day are eligible for this study. A pre‐menarcheal female is defined as any female who has not begun menses and is considered Tanner stage 2 or less

  • Diagnosis of asthma ≥ 6 months before screening

  • Stable asthma therapy (FP, total daily dose ≤ 400 mcg or equivalent) and SABA inhaler for ≥ 4 weeks before screening

  • Participants must be controlled on existing asthma treatment at screening, which will be continued during run‐in, washout and run‐out periods (but not during active treatment periods). Control is defined as Childhood ACT score > 19 and PEF > 75% predicted

  • Participants must demonstrate an ability to accept and effectively use a demonstration inhaler from demonstration kits provided

  • Participants must weigh ≥ 20 kg

  • Participant and parent/guardian are able to understand and comply with protocol requirements, instructions and protocol stated restrictions. Parent or guardian must have the ability to read, write and record diary information collected throughout the study. Parent or guardian must have the ability to manage study drug administration and PEF assessments

  • One or more parents/guardians have signed and dated the written informed consent before admission to the study. This will be accompanied by informed assent from the participant for children 7 to 11 years of age

Exclusion criteria

  • Participants with a history of life‐threatening asthma, an asthma exacerbation requiring systemic corticosteroids or emergency room attendance (within 3 months) or hospitalisation (within 6 months) before screening

  • Participants with any medical condition or circumstance making the volunteer unsuitable for participation in the study

  • Culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear, not resolved within 4 weeks of screening and leading to a change in asthma management, or, in the opinion of the investigator, is likely to affect participants' asthma status or ability to participate in the study

  • Clinical visual evidence of oral candidiasis at screening

  • Participants currently receiving (or received within 4 weeks of screening) asthma therapies including theophyllines, long‐acting inhaled beta‐agonists or oral beta‐agonists, or who have changed their asthma medication within 4 weeks of screening

  • Significant abnormality of rate, interval, conduction or rhythm in the 12‐lead ECG, as determined by the investigator, in conjunction with age and gender of the child and assessment provided by the remote analysis service

  • QTcF > 450 milliseconds or ECG not suitable for QT measurement (e.g. poorly defined termination of the T wave)

  • Aspartarte aminotransferase, alanine aminotransferase, alkaline phosphatase and bilirubin > 1.5 times ULN (isolated bilirubin > 1.5 times ULN is acceptable if bilirubin is fractionated and direct bilirubin is less than 35%)

  • Known or suspected sensitivity to any constituents of the novel DPI (i.e. lactose or magnesium stearate) (e.g. history of severe milk protein allergy)

  • Any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist, sympathomimetic drug or intranasal, inhaled or systemic corticosteroid therapy

  • Use of prescription or non‐prescription drugs, including vitamins and herbal and dietary supplements (including St John's Wort) within 7 days or 5 half‐lives (whichever is longer) before first dose of study medication, unless in the opinion of the investigator and the GSK medical monitor the medication will not interfere with study procedures nor compromise participant safety

  • Consumption of red wine, Seville oranges, grapefruit or grapefruit juice and/or pummelos, exotic citrus fruits, grapefruit hybrids or fruit juices from 7 days before first dose of study medication

  • Individual has participated in a clinical trial and has received an investigational product within 30 days, 5 half‐lives or twice the duration of the biological effect of the investigational product (whichever is longer)

  • Exposure to more than 4 new chemical entities within 12 months before first dosing day

  • When participation in the study would result in donation of blood or blood products in excess of the lesser of 50 mL or 3 mL per kilogram within a 56‐day period

  • Parent/guardian has a history of psychiatric disease, intellectual deficiency, substance abuse or other condition (e.g. inability to read, comprehend and write) that will limit the validity of consent to participate in this study

  • Unwillingness or inability of participant or parent/guardian to follow procedures outlined in the protocol

  • Participant who is mentally or legally incapacitated

  • Children who are wards of the state or government

  • Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, sub‐investigator or study co‐ordinator, or is an employee of the participating investigator

Interventions

Sequence 1: FF 100 mcg/VI 25 mcg in period 1 and FF 100 mcg in period 2

Sequence 2: FF 100 mcg in period 1 and FF 100 mcg/VI 25 mcg in period 2

With a washout period ≥ 7 days

Outcomes

Primary outcomes

  • Number of participants with AE or SAE during treatment period

  • Basophil, eosinophil, lymphocyte, monocyte, total neutrophil, platelet and white blood cell count values at day 14 of treatment period

  • Haemoglobin and MCHC values at day 14 of treatment period

  • Reticulocyte and RBC values at day 14 of treatment period

  • Haematocrit values at day 14 of treatment period

  • MCV value at day 14 of treatment period

  • MCH values at day 14 of treatment period

  • ALT, ALP, AST and GGT values at day 14 of treatment period

  • Albumin and total protein values at day 14 of treatment period

  • Calcium chloride, CO2 content/bicarbonate, glucose, potassium, sodium and urea/BUN values at day 14 of treatment period

  • Total bilirubin, direct bilirubin, creatinine and uric acid values at day 14 of treatment period

  • PEF at days 1 and 14 of treatment period

  • Change from baseline in SBP and DBP at days 1 and 14 of treatment period

  • Change from baseline in heart rate at days 1 and 14 of treatment period

  • Maximum QTcF at days 1 and 14 of treatment period

Secondary outcomes

  • AUC(0‐t) and AUC(0‐4) of FF on day 14 of treatment period

  • Cmax of FF at day 14 of treatment period

  • Tmax and Tlast of FF at day 14 of treatment period

  • AUC(0‐t) and AUC(0‐4) of VI at day 14 of treatment period

  • Cmax of VI at day 14 of treatment period

  • Tmax and Tlast of VI at day 1 of treatment period

  • Blood glucose and potassium values at day 14 of treatment period

  • Serum cortisol weighted mean (0 to 12 hours) at day 14 of treatment period

  • Average oropharyngeal cross‐sectional area at days 1 and 14 of treatment period

  • Distance of assessment at days 1 and 14 of treatment period

  • Oropharyngeal volume at days 1 and 14 of treatment period

  • Average flow rate and PIFR at days 1 and 14 of treatment period

  • Inhalation time at days 1 and 14 of treatment period

  • Inhaled volume at days 1 and 14 of treatment period

  • Peak pressure drop at days 1 and 14 of treatment period

  • TED at day 14 of treatment period

  • Ex‐throat dose (ETD) and ETD < 2 microns at day 14 of treatment period

Notes

Data collected from 1 site in California, USA

Funded by GlaxoSmithKline

Study duration: 2 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline, Stevenage, UK)

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System (GlaxoSmithKline)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trial reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information on the 3 participants not completing the study is provided at http://clinicaltrials.gov/show/NCT01453023

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Oliver 2012

Methods

Randomised double‐blind cross‐over trial

Participants

Total sample

N = 52 participants, 50 completed

Age: mean, 35.4 (SD 8.63)

Males: 34 years (65%)

Pre‐bronchodilator FEV1% predicted: mean, 89.71 (SD 8.848)

Inclusion criteria

  • BMI within the range 18.5 to 35.0 kg/m2

  • Females of non‐childbearing potential

  • Documented history of bronchial asthma, first diagnosed ≥ 6 months before screening visit and currently treated only with intermittent SABA therapy by inhalation

  • Pre‐bronchodilator FEV1 > 70% of predicted at screening

  • Participants who are current non‐smokers

  • Methacholine challenge PC20 < 8 mg/mL at screening

  • Screening allergen challenge demonstrates that participant experiences an early asthmatic response

Exclusion criteria

  • Current or chronic history of liver disease, or known hepatic or biliary abnormalities

  • Participant hypertensive at screening

  • Respiratory tract infection and/or exacerbation of asthma within 4 weeks before first dose of study medication

  • History of life‐threatening asthma

  • Symptomatic with hay fever at screening or predicted to have symptomatic hay fever

  • Unable to abstain from short‐acting beta‐agonists

  • Unable to abstain from antihistamines

  • Unable to abstain from other medications, including NSAIDs, antidepressant drugs and antiasthma, anti‐rhinitis or hay fever medication

  • Participant has participated in a study with a new molecular entity during previous 3 months or has participated in 4 or more clinical studies in previous 12 months

  • Undergoing allergen desensitisation therapy

Interventions

Sequence 1: placebo, FF 100 mcg, FF/VI 100/25 mcg

Sequence 2: placebo, FF/VI 100/25 mcg, FF 100 mcg

Sequence 3: FF 100 mcg, FF/VI 100/25 mcg, placebo

Sequence 4: FF 100 mcg, placebo, FF/VI 100/25 mcg

Sequence 5: FF/VI 100/25 mcg, placebo, FF 100 mcg

Sequence 6: FF/VI 100/25 mcg, FF 100 mcg, placebo

Following the run‐in period, participants were randomised to 1 of 6 treatment sequences of placebo, FF 100 mcg once‐daily and FF/VI 100/25 mcg once‐daily. The 3 treatment periods were separated by a washout period of ≥ 21 days (from day 28 dose) and maximum of 35 days

Outcomes

Primary outcome

  • Weighted mean change from baseline in FEV1 from 0 to 2 hours, following 22 to 23‐hour post‐treatment allergen challenge on day 29 of each treatment period

Secondary outcomes

  • Maximum % decrease from baseline in FEV1 from 0 to 2 hours, following 22 to 23‐hour post‐treatment allergen challenge on day 29 of each treatment period

  • Minimum FEV1 absolute change from baseline from 0 to 2 hours, following 22 to 23‐hour post‐treatment allergen challenge on day 29 of each treatment period

  • Number of participants with treatment‐emergent AEs

Notes

Data collected from 4 sites in Germany (1), New Zealand (1) and UK (2)

Funded by GlaxoSmithKline

Study duration: 4 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised by RandAll (GlaxoSmithKline validated internal randomisation software) to 1 of 6 treatment sequences, each comprising 3 treatment periods

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System (GlaxoSmithKline)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trial reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of attrition bias included in trial report. Two participants withdrew: 1 withdrew consent and 1 experienced an SAE

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Oliver 2013

Methods

Randomised double‐blind cross‐over trial

Participants

Total sample

N = 27 participants

Age: mean, 30.8 years (SD 7.46)

Males: 19 (70%)

Pre‐bronchodilator FEV1: mean % pred, 92.3 (range 71.3 to 119.8)

Inclusion criteria

  • BMI within the range 18.5 to 35.0 kg/m2

  • Females of non‐childbearing potential

  • Documented history of bronchial asthma, first diagnosed ≥ 6 months before screening visit and currently treated only with intermittent SABA therapy by inhalation

  • Pre‐bronchodilator FEV1 > 70% of predicted at screening

  • Participants who are current non‐smokers

  • Methacholine challenge PC20 < 8 mg/mL at screening

  • Screening allergen challenge demonstrates that participant experiences an early asthmatic response

Exclusion criteria

  • Current or chronic history of liver disease, or known hepatic or biliary abnormalities

  • Participant hypertensive at screening

  • Respiratory tract infection and/or exacerbation of asthma within 4 weeks before first dose of study medication

  • History of life‐threatening asthma

  • Symptomatic with hay fever at screening or predicted to have symptomatic hay fever

  • Unable to abstain from short‐acting beta‐agonists

  • Unable to abstain from antihistamines

  • Unable to abstain from other medications, including NSAIDs, antidepressant drugs and antiasthma, antirhinitis or hay fever medication

  • Participant has participated in a study with a new molecular entity during previous 3 months or has participated in 4 or more clinical studies in previous 12 months

  • Undergoing allergen desensitisation therapy

Interventions

Sequence 1: VI 25 mcg, placebo, FF 100 mcg, FF/VI 100/25 mcg

Sequence 2: FF/VI 100/25 mcg, FF 100 mcg, placebo, VI 25 mcg

Sequence 3: placebo, FF/VI 100/25 mcg, VI 25 mcg, FF 100 mcg

Sequence 4: FF 100 mcg, VI 25 mcg, FF/VI 100/25 mcg, placebo

Participants meeting all inclusion criteria and no exclusion criteria during screening visit, conducted 14 to 42 days before first dose of study medication, entered a 14‐day run‐in period. Participants were then randomised to 4 treatment periods, each lasting 21 days and all separated by a nominal washout period of 21 to 35 days

Outcomes

Primary outcomes

  • LAR: absolute change from baseline in minimum FEV1 between 4 and 10 hours following 1‐hour post‐treatment allergen challenge on day 21 of each treatment period

  • LAR: absolute change from saline in weighted mean FEV1 between 4 and 10 hours following 1‐hour post‐treatment allergen challenge on day 21 of each treatment period

  • EAR: absolute change from baseline in minimum FEV1 between 0 and 2 hours following 1‐hour post‐treatment allergen challenge on day 21 of each treatment period

  • EAR: absolute change from baseline in weighted mean FEV1 between 0 and 2 hours following 1‐hour post‐treatment allergen challenge on day 21 of each treatment period

Secondary outcomes

  • Maximum % change from baseline in FEV1 between 0 and 2 hours following 1‐hour post‐treatment allergen challenge on day 21 of each treatment period

  • PC20 on day 22 of each treatment period

Notes

Data collected from 4 sites in Australia (1), New Zealand (1) and Sweden (2)

Funded by GlaxoSmithKline

Study duration: Each treatment period ran for 21 days with a 21 to 35‐day washout period between treatment periods

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation schedule based on a Williams square generated by the sponsor through validated internal software (RandAll, GlaxoSmithKline, London, UK)

Allocation concealment (selection bias)

Low risk

Automated telephone‐based interactive voice response system ‐ RAMOS (GlaxoSmithKline, London, UK) ‐ was used by investigators to register participants and obtain randomised treatment assignments in a blinded manner

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Reported as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Reported as double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Details of attrition bias included in trial report. Twenty‐seven participants were randomised, and 26 completed the study. One participant withdrew consent, and 4 protocol deviations were noted during period 1. Data for those participants were excluded from the analysis of relevant study periods

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

Woodcock 2013

Methods

Randomised double‐blind double‐dummy parallel‐group multi‐centre trial

Participants

Total sample

N = 806 participants, 715 completed study

FF/VI 100/25 mcg, n = 403 (358 completed study)

FP/SAL 250/50 mcg twice‐daily, n = 403 (357 completed study)

Age

FF/VI 100/25 mcg, mean 43.8 years (SD 15.86)

FP/SAL 250/50 mcg twice‐daily, mean 41.9 years (SD 16.90)

Males

FF/VI 100/25 mcg 159 (44%)

FP/SAL 250/50 mcg twice‐daily 158 (44%)

Baseline FEV1 (L)

FF/VI 100/25 mcg, mean 2.013 (SD 0.653)

FP/SAL 250/50 mcg twice‐daily, mean 2.043 (SD 0.6378)

Inclusion criteria

  • Clinical diagnosis of asthma

  • Reversibility ≥ 12% and ≥ 200 mL within 10 to 40 minutes following 2 to 4 inhalations of albuterol

  • FEV1 40% to 85% of predicted normal

  • Currently using ICS therapy

Exclusion criteria

  • History of life‐threatening asthma within previous 5 years (requiring intubation and/or associated with hypercapnia, respiratory arrest or hypoxic seizures)

  • Respiratory infection or oral candidiasis

  • Asthma exacerbation requiring OCS, or overnight hospitalisation requiring additional asthma treatment

  • Uncontrolled disease or clinical abnormality

  • Allergies

  • Taking another investigational medication or prohibited medication

  • Night shift workers

  • Current smokers or participants with smoking history of ≥ 10 pack‐years

Interventions

Arm 1: FF/VI 100/25 mcg once‐daily

Arm 2: FP/SAL 250/50 mcg twice‐daily

Outcomes

Primary outcome

  • Change from baseline in weighted mean 24‐hour serial FEV1 at day 168/week 24

Secondary outcomes

  • Serial FEV1 (0‐24 hours)

  • Number of participants with indicated time to onset of bronchodilator effect at day 1

  • Change from baseline in weighted mean serial FEV1 over 0 to 4 hours post first dose (at randomisation)

  • Change from baseline in weighted mean serial FEV1 over 0 to 4 hours at day 168

  • Number of participants obtaining ≥ 12% and ≥ 200 mL increase from baseline in FEV1

  • Change from baseline in trough FEV1 at day 168

  • Baseline FEV1 by completion status

  • Change from baseline in ACT scores at day 168

  • Number of healthcare contacts related to asthma or to treatment of asthma from baseline to day 168

  • Change from baseline in AQLQ total score for participants ≥ 12 years of age (AQLQ + 12)

  • Percentage of participants with "no problems" in EQ‐5D descriptive system dimensions at day 168/week 24

  • Change from baseline in EQ‐5D VAS score at day 168

Notes

Data collected from 63 sites in Argentina (10), Chile (7), Republic of Korea (7), Netherlands (8), Phillipines (6) and US (25).

Funded by GlaxoSmithKline

Study duration: 24 weeks

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule was generated by the sponsor through a validated computerised system (RandAll, GlaxoSmithKline)

Allocation concealment (selection bias)

Low risk

Participants were randomised via the Registration and Medication Ordering System

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Reported as double‐blind

'Neither the patients nor the investigator knew which study medication the patient was receiving'

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Reported as double‐blind

'Neither the patients nor the investigator knew which study medication the patient was receiving'

Incomplete outcome data (attrition bias)
All outcomes

Low risk

89% completed the study. Details of participant withdrawal included in trial report

Selective reporting (reporting bias)

Low risk

No apparent indication of reporting bias

AE: adverse event

ALP: alkaline phosphatase

ALT: alanine aminotransferase

AM: morning

ANC: absolute neutrophil count

AQLQ: asthma quality of life questionnaire

AST: aspartate aminotransferase

AUC: area under the curve

BMI: body mass index

BUN: blood urea nitrogen

CK: creatine kinase

Cmax: maximum serum concentration

CO: carbon monoxide

CO2: carbon dioxide

COPD: chronic obstructive pulmonary disease

DBP: diastolic blood pressure

DPI: dry powder inhaler

EAR: early asthmatic response

ECG: electrocardiogram

EQ‐5D: EuroQuality of Life 5D questionnaire

ETD: ex‐throat dose

FeNO: fractional exhaled nitric oxide

FEV1: forced expiratory volume in one second

FF: fluticasone furoate

FP: fluticasone propionate

GGT: gamma glutamyl transferase

HIV: human immunodeficiency virus

HRT: hormone replacement therapy

ICS: inhaled corticosteroid

IOP: intraocular pressure

LABA: long‐acting beta2‐agonist

LOCS III: Lens Opacities Classification System, Version III

LogMAR: logarithm of the minimum angle of resolution

MCH: mean corpuscular haemoglobin

MCHC: mean corpuscular haemoglobin concentration

MCV: mean corpuscular volume

NHANES: National Health and Nutrition Examination Survey

NIOX‐MINO: first point‐of‐care medical device for measuring fractional exhaled nitric oxide

NSAIDs: non‐steroidal anti‐inflammatory drugs

OCS: oral corticosteroid

OM: evening

PC20: provocative concentration of methacholine estimated to result in a 20% reduction in FEV1

PEF: peak expiratory flow

PEFR: peak expiatory flow rate

PIRF: peak inspiratory flow rate

PK: pharmacokinetics

ppb: parts per billion

QTcB: QT interval using Bazett's correction

QTcF: QT interval using Fridericia's correction

RAMOS: Registration and Medication Ordering System

RBC: red blood cell

SABA: short‐acting beta2‐agonist

SAE: serious adverse event

SAL: salmeterol

SAS: Statistical Analysis System (a software suite developed by SAS Institute)

SBP: systolic blood pressure

SD: standard deviation

TED: total emitted dose

Tlast: time of the last point with quantifiable concentration

Tmax: time to Cmax

ULN: upper limit of normal

UMEC: umeclidinium bromide

VAS: visual analogue scale

VI: vilanterol

WBC: white blood cell count

WM: weighted mean

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Calverley 2014

Study focuses on chronic obstructive pulmonary disease participants with community‐acquired pneumonia

Gross 2013

Pooled analysis of data from clinical trials

Gross 2015

Pooled analysis of data from clinical trials

Hozawa 2016

Comparison includes budesonide/formoterol maintenance and reliever therapy vs fluticasone furoate/vilanterol. Therefore, the comparison is not a direct evaluation of budesonide/formoterol maintenance vs fluticasone furoate/vilanterol

Ishiura 2015

Participants have a diagnosis of asthma/chronic obstructive pulmonary disease overlap syndrome, rather than asthma per se. COPD is included in the review's exclusion criteria

Kempsford 2011

Participants did not have a diagnosis of asthma (healthy participants)

Kempsford 2011a

Participants did not have a diagnosis of asthma (healthy participants)

Kempsford 2012a

Participants did not have a diagnosis of asthma (healthy participants)

Nakahara 2013

Report of 3 safety, pharmacokinetics and pharmacodynamics studies with healthy participants

NCT00603746

VI and FF are not used together in the intervention arm

NCT01181895

Inhaled steroid used in the trial was not specifically FF

NCT01213849

Dose proportionality study comparing 3 doses of FF/VI without an additional comparison arm in healthy participants

NCT01435902

Study was withdrawn before participants were enrolled

NCT01485445

Participants did not have a diagnosis of asthma (healthy participants)

NCT01573767

Trial focuses on VI and FP, not on VI and FF

NCT01711463

Participants did not have a diagnosis of asthma (healthy participants)

NCT02712047 2016

Study evaluating exhaled nitric oxide time profile as a biomarker of airway inflammation

Oliver 2014

Inhaled steroid used in the trial was not specifically FF

Sterling 2012

Inhaled steroid used in the trial was not specifically FF

Woepse 2013

Evaluation of DPI among participants with asthma and COPD

COPD: chronic obstructive pulmonary disease

DPI: dry powder inhaler

FF: fluticasone furoate

FP: fluticasone propionate

VI: vilanterol

Characteristics of ongoing studies [ordered by study ID]

NCT01498679

Trial name or title

A randomised, double‐blind, placebo‐controlled, parallel group, multi‐centre study to evaluate the efficacy and safety of FF/VI inhalation powder delivered once‐daily for 12 weeks in the treatment of asthma in adolescent and adult participants of Asian ancestry currently treated with low to mid‐strength ICS or low‐strength combination therapy

Methods

Randomised double‐blind placebo‐controlled parallel‐group multi‐centre study

Participants

Participants of Asian ancestry with asthma. 12 years of age or older

Inclusion criteria

  • Informed consent: All participants must be able and willing to give written informed consent to take part in the study

  • Type of participants: outpatients, of Asian ancestry, 12 years of age or older at visit 1 (or ≥ 18 years of age or older if local regulations or the regulatory status of study medication permit enrolment of adults only), with a diagnosis of asthma as defined by the Global Initiative for Asthma (GINA 2009) ≥ 12 weeks before visit 1

  • Gender: male or eligible female, defined as non‐childbearing potential or childbearing potential using a protocol‐defined acceptable method of birth control consistently and correctly. Female participants should not be enrolled if they are pregnant, lactating or plan to become pregnant during the time of study participation. A serum pregnancy test is required for females of childbearing potential at initial screening visit (visit 1) and at visit 5 or early withdrawal

  • Severity of disease: best FEV1 40% to 90% of predicted normal value at visit 1, screening visit. Predicted values will be based upon NHANES III using adjustment for Asians (Hankinson 2010)

  • Reversibility of disease: demonstrated ≥ 12% and ≥ 200 mL reversibility of FEV1 within 10 to 40 minutes following 2 to 4 inhalations of albuterol/salbutamol inhalation aerosol (or 1 nebulised treatment with albuterol/salbutamol solution) at screening visit

  • Current antiasthma therapy: All participants must be using an ICS, with or without LABA, for ≥ 12 weeks before visit 1, in accordance with protocol‐defined acceptable dose ranges

  • SABA: All participants must be able to replace their current SABA with albuterol/salbutamol inhaler at visit 1 for use as needed for the duration of the study. Participants must be able to withhold albuterol/salbutamol for ≥ 4 hours before study visits

Exclusion criteria

  • History of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 10 years

  • Respiratory infection: culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that was not resolved within 4 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, was expected to affect participant asthma status or ability of participant to participate in the study

  • Asthma exacerbation: any asthma exacerbation requiring OCS within 12 weeks of visit 1, or that resulted in overnight hospitalisation requiring additional treatment for asthma within 6 months before visit 1

  • Concurrent respiratory disease: Participant must not have current evidence of pneumonia, pneumothorax, atelectasis, pulmonary fibrotic disease, bronchopulmonary dysplasia, chronic bronchitis, emphysema, COPD or respiratory abnormalities other than asthma

  • Other concurrent diseases/abnormalities: Participant must not have any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of efficacy results if the condition/disease was exacerbated during the study

  • Oropharyngeal examination: Participant will not be eligible for the run‐in if he/she has clinical visual evidence of candidiasis at visit 1

  • Allergies: drug allergy: any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or to any intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to constituents of the new powder inhaler (i.e. lactose or magnesium stearate). Milk protein allergy: history of severe milk protein allergy

  • Concomitant medications: use of protocol‐defined prohibited medications before visit 1 or during the study, in accordance with the protocol

  • Tobacco use: current smoker or participants with smoking history of 10 pack‐years ( i.e. 20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products within the past 3 months (i.e. cigarettes, cigars, smokeless or pipe tobacco)

  • Affiliation with investigator site: Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or an employee of the participating investigator

  • Previous participation: Participant may not have previously been randomised to treatment in another phase III FF/VI combination product study (i.e. HZA113714, HZA106827, HZA106829, HZA106837, HZA106839, HZA106851, HZA113091)

  • Compliance: Participant will not be eligible if he/she or his/her parent or legal guardian has an infirmity, disability, disease or geographical location that seems likely (in the opinion of the Investigator) to impair compliance with any aspect of this study protocol, including visit schedule and completion of daily diaries

Interventions

FF/VI ICS/LABA combination vs placebo

Outcomes

Primary outcome measure

  • Mean change from baseline in PM PEF averaged over 12‐week treatment period

Secondary outcome measures

  • Mean change from baseline in daily AM PEF averaged over 12‐week treatment period

  • Change from baseline in percentage of rescue‐free 24‐hour periods during 12‐week treatment period

  • Change from baseline in percentage of symptom‐free 24‐hour periods during 12‐week treatment period

  • Change from baseline in total AQLQ score at week 12

Starting date

January 2012

Contact information

GlaxoSmithKline Research and Development Limited

Notes

NCT01573624

Trial name or title

A multi‐centee, randomised, double‐blind, dose‐ranging study to evaluate GSK573719 in combination with fluticasone furoate, fluticasone furoate alone and an active control of fluticasone furoate/vilanterol combination in participants with asthma

Methods

Randomised double‐blind cross‐over study

Participants

Participants with asthma. 18 years of age or older

Inclusion criteria

  • Outpatient

  • 18 years of age or older at visit 1

  • Diagnosis of asthma

  • Male or eligible female

  • Pre‐bronchodilator FEV1 40% to 80% of predicted normal value at visit 1

  • Demonstrated reversibility by ≥ 12% and ≥ 200 mL of FEV1 within 40 minutes following albuterol at visit 1

  • Need for regular controller therapy (i.e. ICSs alone or in combination with a LABA, or leukotriene modifier, etc.) for a minimum of 8 weeks before visit 1

Exclusion criteria

  • History of life‐threatening asthma

  • Respiratory infection not resolved

  • Asthma exacerbation

  • Concurrent respiratory disease

  • Current smokers

  • Other uncontrolled disease or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation, or would confound interpretation of efficacy results if the condition/disease was exacerbated during the study

  • Positive hepatitis B surface antigen or positive hepatitis C antibody and/or HIV

  • Visual clinical evidence of oropharyngeal candidiasis

  • Drug or milk protein allergies

  • Concomitant medications affecting course of asthma

  • Use of any other investigational medication within 30 days or 5 drug half‐lives (whichever is longer)

  • Previous use of GSK573719

  • Any disease preventing use of anticholinergics

  • Any condition that impairs compliance with study protocol including visit schedule and completion of daily diaries

  • Any participant with a history of alcohol or substance abuse

  • Any affiliation with investigator's site

Interventions

FF 100 mcg vs FF/VI 100/25 mcg vs FF/GSK573719 100/15.6 to 250 mcg

Outcomes

Primary outcome measures

  • Change from baseline in trough FEV1

  • FEV1 value obtained 24 hours after morning dosing on day 14 of each treatment period

Secondary outcome measures

  • Mean change from baseline in daily AM/PM PEF

  • Mean change from baseline in rescue albuterol use

Starting date

April 2012

Contact information

GlaxoSmithKline Research and Development Limited; [email protected]

Notes

NCT01706198

Trial name or title

A 12‐month, open label, randomised, effectiveness study to evaluate fluticasone furoate (GW685698)/vilanterol (GW642444) inhalation powder delivered once‐daily via a novel dry powder inhaler compared with usual maintenance therapy in participants with asthma

Methods

Randomised parallel‐group study

Participants

Participants with asthma. 18 years of age or older

Inclusion criteria

Participants eligible for enrolment in the study must meet all of the following criteria

  • Informed consent: Participants must be able to provide informed consent and have their consent signed and dated

  • Type of participants: participants with documented GP diagnosis of asthma as their primary respiratory disease

  • Current antiasthma therapy: All participants must be prescribed maintenance therapy and must be receiving ICS with or without LABA (fixed combination or via separate inhalers) for ≥ 4 weeks before visit 2. Other background asthma medication such as antileukotrienes are permitted

  • All participants receiving ICS monotherapy or ICS/LABA combination (this can be a fixed‐dose combination or an ICS alone or LABA alone in separate inhalers) must have had symptoms in the past week before visit 2. Symptoms are defined by daytime symptoms more than twice per week, use of SABA bronchodilator more than twice per week, any limitation of activities or any nocturnal symptoms/awakening (Symptoms are based on participant's recall and are consistent with GINA and agree in principle with BTS/SIGN guidelines)

  • Participant questionnaires: Participants must be able to complete electronic participant questionnaires as well as questionnaires to be completed by phone or must provide a proxy (e.g. partner/relative/friend) who can do so on their behalf

  • Gender and age: male or female participants ≥ 18 years of age at visit 1. A female is eligible to enter and participate in the study if she is of: non‐childbearing potential (i.e. physiologically incapable of becoming pregnant, including any female who is post‐menopausal or surgically sterile). Surgically sterile females are defined as those with a documented hysterectomy and/or bilateral oophorectomy or tubal ligation. Post‐menopausal females are defined as amenorrhoeic for > 1 year with an appropriate clinical profile (e.g. age appropriate, history of vasomotor symptoms). However. in questionable cases, a blood sample with FSH > 40 MIU/mL and estradiol < 40 pg/mL (< 147 pmol/L) is confirmatory OR childbearing potential has a negative urine pregnancy test at visit 2, and participant agrees to one of the highly effective and acceptable contraceptive methods used consistently and correctly (i.e. in accordance with approved product label and instructions of the physician for the duration of the study ‐ visit 2 to end of study)

Exclusion criteria

Participants meeting any of the following criteria must not be enrolled in the study

  • Recent history of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 6 months

  • COPD: Participant must not have current evidence or GP diagnosis of COPD

  • Other diseases/abnormalities: participants with historical or current evidence of uncontrolled or clinically significant disease. Significant is defined as any disease that, in the opinion of the GP/investigator, would put the safety of the patient at risk through study participation, or that would affect the efficacy or safety analysis if the disease/condition was exacerbated during the study

  • Drug/food allergy: participants with a history of hypersensitivity to any of the study medications (e.g. beta2‐agonists, corticosteroid) or components of the inhalation powder (e.g. lactose, magnesium stearate). In addition, participants with a history of severe milk protein allergy that, in the opinion of the GP/investigator, contraindicates the participant's participation will also be excluded

  • Investigational medications: Participant must not have used any investigational drug within 30 days before visit 2 or within 5 half‐lives (t½) of prior investigational study (whichever is the longer of the 2) (if unsure, discuss with medical monitor before screening)

  • Long‐term user of systemic corticosteroids: participant who, in the opinion of the GP/investigator, is considered to be a long‐term user of systemic corticosteroids for respiratory or other indications (if unsure, discuss with the medical monitor before screening)

  • Participants who are using LABA without an ICS as asthma maintenance therapy

  • Participants who plan to move away from the geographical area where the study is being conducted during the study period and/or if participants have not consented to inclusion of their medical records in the electronic medical records database that is operational in the Salford area

Interventions

GW685698+GW642444 once‐daily via a novel dry powder inhaler vs existing maintenance therapy (ICS alone or in combination with a LABA)

Outcomes

Primary outcome measure

  • Percentage of participants who have an ACT total score ≥ 20 at week 24 (6th month) assessment

Secondary outcome measures

  • Percentage of participants with asthma control (ACT total score ≥ 20)

  • Mean change from baseline in ACT total score

  • Percentage of participants who have an increase from baseline of ≥ 3 in ACT total score

  • Asthma‐related secondary care contacts

  • Asthma‐related primary care contacts

  • All secondary care contacts

  • All primary care contacts

  • Mean annual rate of severe asthma exacerbations

  • Number of salbutamol inhalers (adjusted to equivalence of 200 actuations) collected by participants from study‐enrolled community pharmacies over the entire treatment period

  • Time to discontinuation or modification of initial therapy

  • Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) total score at week 52

  • Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) environmental stimuli domain score at week 52

Starting date

November 2012

Contact information

GlaxoSmithKline Research and Development Limited; [email protected]

Notes

NCT01837316

Trial name or title

A study to assess the bronchodilator effect of a single dose of fluticasone furoate (FF)/vilanterol (VI) 100/25 micrograms (mcg) combination when administered in adult participants with asthma

Methods

Randomised double‐blind placebo‐controlled cross‐over study

Participants

32 adult participants with moderately severe asthma

Inclusion criteria

  • Asthma: a doctor diagnosis of asthma

  • Age of participant: 18 to 65 years of age inclusive, at the time of signing the informed consent

  • Severity of disease: screening pre‐bronchodilator FEV1 ≥ 60% of predicted

  • Reversibility of disease: demonstrated presence of reversible airway disease at screening

  • Current therapy: on ICS with or without a SABA for ≥ 12 weeks before screening. Able to stop current short‐acting beta2‐agonists (SABAs) and replace with albuterol/salbutamol inhaler

  • Body weight and BMI: body weight ≥ 50 kg and BMI within the range 19.0 to 29.9 kg/m2 (inclusive)

  • Gender: male or female. A female participant is eligible to participate if she is of:

Non‐childbearing potential. Females on HRT and whose menopausal status is in doubt will be required to use one of the contraception methods if they wish to continue their HRT during the study. Otherwise, they must discontinue HRT to allow confirmation of post‐menopausal status before study enrolment.

Childbearing potential and agrees to use one of the contraception methods for an appropriate period of time (as determined by product label or investigator) before the start of dosing to sufficiently minimise risk of pregnancy at that point. Female participants must agree to use contraception until completion of the follow‐up visit.

  • Liver criteria: AST and ALT < 2 × ULN; alkaline phosphatase and bilirubin ≤ 1.5 × ULN (isolated bilirubin > 1.5 × ULN is acceptable if bilirubin is fractionated and direct bilirubin < 35%)

  • Consent: capable of giving written informed consent, which includes compliance with requirements and restrictions listed in the consent form

Exclusion criteria

  • History of life‐threatening asthma

  • Other significant pulmonary disease: pneumonia, pneumothorax, atelectasis, pulmonary fibrotic disease, bronchopulmonary dysplasia, chronic bronchitis, emphysema, COPD, respiratory abnormalities other than asthma

  • Respiratory infection: culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that is not resolved within 4 weeks of screening that; led to a change in asthma management OR, in the opinion of the Investigator, is expected to affect the participant's asthma status, OR the participant's ability to participate in the study

  • Asthma exacerbation: any asthma exacerbation requiring OCS within 12 weeks of screening or that resulted in overnight hospitalisation requiring additional treatment for asthma within 6 months before screening

  • Concomitant medications: use of medications, ICS prohibited for each study period from 24 hours before dosing to 72 hours after dosing; LABA, LTRA or LAMA prohibited for 12 weeks before screening; high doses of an ICS prohibited for 8 weeks before screening; OCS prohibited for 12 weeks before screening; potent CYP3A4 inhibitors prohibited within 4 weeks before dosing. The following medications may not be used during the study from first dosing to the end of period 2 inclusive: anticonvulsants, polycyclic antidepressants, beta‐adrenergic blocking agents, phenothiazines and MAO inhibitors

  • Other concurrent diseases/abnormalities: Participant has any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of study results if the condition/disease was exacerbated during the study

  • Oropharyngeal examination: Participant will not be eligible if he/she has clinical visual evidence of oral candidiasis at screening

  • Pregnant and lactating females: pregnant females as determined by positive serum human chorionic gonadotropin (hCG) test at screening or by positive urine hCG test before dosing. Lactating females

  • Allergies: milk protein allergy: history of severe milk protein allergy. Drug allergy: any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or to any intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to constituents of the DPI (i.e. lactose or magnesium stearate). Historical allergy: history of drug or other allergy that, in the opinion of the investigator or the GSK medical monitor, contraindicates participation

  • 12‐Lead ECG abnormality: significant abnormality in the 12‐lead ECG performed at screening

  • Tobacco use: current smokers or smoking history ≥ 10 pack‐years. Participant may not have used any inhaled tobacco products in the 12‐month period preceding the screening visit

  • Previous participation: exposure to more than 4 new chemical entities within 12 months before first dosing day

Interventions

After screening, participant will be randomised and will be assigned to 1 of 2 treatment sequences (AB or BA, where A is placebo and B is FF/VI 100/25 mcg). Between the 2 treatment periods, a washout period of 7 to 14 days will occur

Outcomes

Serial FEV1 measurements will be taken at 15 and 30 minutes, and at 1, 2, 4, 12, 24, 36, 48, 60 and 72 hours post dose. Safety assessments will include vital signs, ECGs, AE monitoring and laboratory safety tests; however, these will not constitute study endpoints. Results of the study will provide supporting information to prescribers on the bronchodilator effect of FF/VI over 72 hours

Starting date

Information on http://clinicaltrials.gov/ in October 2013 indicated that study was not yet recruiting. Estimated primary completion date: December 2013

Contact information

GlaxoSmithKline Research and Development Limited; [email protected]

Notes

NCT02094937

Trial name or title

A study to compare the efficacy and safety of fluticasone furoate (FF) 100 mcg once‐daily with fluticasone propionate (FP) 250 mcg twice‐daily and FP 100 mcg twice‐daily in well‐controlled asthmatic Japanese participants

Methods

Randomised double‐blind multi‐centre parallel‐group study

Participants

Exclusion criteria

  • History of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within past 10 years

  • Respiratory infection: culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that was not resolved within 8 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, is expected to affect the participant's asthma status or ability to participate in the study

  • Asthma exacerbation: any asthma exacerbation requiring systemic corticosteroids or injection within 12 weeks of visit 1, or that resulted in overnight hospitalisation requiring additional treatment for asthma within 6 months before visit 1

  • Concurrent respiratory disease: Participant must not have current evidence of pneumonia, pneumothorax, atelectasis, pulmonary fibrotic disease, bronchopulmonary dysplasia, chronic bronchitis, emphysema, COPD or any respiratory abnormalities other than asthma

  • Other concurrent diseases/abnormalities: Participant must not have a clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of efficacy results if the condition/disease was exacerbated during the study. The list of additional excluded conditions/diseases includes, but is not limited to, the following: congestive heart failure, known aortic aneurysm, clinically significant coronary heart disease, clinically significant cardiac arrhythmia, stroke within 3 months of visit 1, uncontrolled hypertension (≥ 2 measurements with systolic BP > 160 mmHg, or DBP > 100 mmHg), recent or poorly controlled peptic ulcer, haematologic, hepatic or renal disease, immunologic compromise, current malignancy (history of malignancy is acceptable only if participant has been in remission for 1 year before visit 1 (remission = no current evidence of malignancy and no treatment for malignancy in the 12 months before visit 1), tuberculosis (current or untreated) (participants with a history of tuberculosis infection who have completed an appropriate course of antituberculous treatment may be suitable for study entry provided there is no clinical suspicion of active or recurrent disease), Cushing's disease, Addison's disease, uncontrolled diabetes mellitus, uncontrolled thyroid disorder, recent history of drug or alcohol abuse

  • Oropharyngeal examination: Participant will not be eligible for the run‐in if he/she has clinical visual evidence of candidiasis at visit 1

  • Investigational medications: Participant must not have used any investigational drug within 30 days before visit 1 or within 5 half‐lives (t1/2) of the prior investigational study (whichever is longer of the 2)

  • Allergies: drug allergy: any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or to any intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to constituents of the investigational product (i.e. lactose or magnesium stearate); milk protein allergy: history of severe milk protein allergy

  • Concomitant medication: administration of prescription or over‐the‐counter medication that would significantly affect the course of asthma, or interact with study drug, such as anticonvulsants (barbiturates, hydantoins, carbamazepine); polycyclic antidepressants; beta‐adrenergic blocking agents; phenothiazines and MAO inhibitors. Immunosuppressive medications: Participant must not be using or require use of immunosuppressive medications during the study.

Note: Immunotherapy is permitted for treatment of allergies during the study, provided it was initiated ≥ 4 weeks before visit 1 and participants remain in the maintenance phase for the duration of the study; cytochrome P450 3A4 (CYP3A4) inhibitors: participants who have received a potent CYP3A4 inhibitor within 4 weeks of visit 1 (e.g. clarithromycin, atazanavir, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir; ritonavir; saquinavir; telithromycin, troleandomycin, voriconazole, mibefradil, cyclosporine)

  • Compliance: Participant will not be eligible if he/she or his/her parent or legal guardian has any infirmity, disability, disease or geographical location that seems likely (in the opinion of the investigator) to impair compliance with any aspect of this study protocol, including visit schedule and completion of eDiaries and a paper medical conditions diary

  • Tobacco use: current smoker or smoking history of 10 pack‐years (e.g. 20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products within the past 3 months (i.e. cigarettes, cigars or pipe tobacco)

  • Affiliation with investigator's site: Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or an employee of the participating investigator

Other exclusion criteria at visit 2 and visit 5

  • Evidence of clinically significant abnormal laboratory tests during visit 1 that are still abnormal upon repeat analysis and are not believed to be due to disease(s) present. Each investigator will use his/her own discretion in determining the clinical significance of the abnormality

  • Changes in asthma medication (excluding salbutamol inhalation aerosol provided at visit 1)

  • Occurrence of a culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear during run‐in and open‐label treatment periods that led to a change in asthma management or, in the opinion of the investigator, is expected to affect participant's asthma status or ability of the participant to participate in the study

  • Any asthma exacerbation requiring systemic corticosteroids or injection or that resulted in overnight hospitalisation requiring additional treatment for asthma. Clinical visual evidence of oral candidiasis at visit 2 and visit 5

  • Positive urine pregnancy test for all females of childbearing potential at visit 2 and visit 5

  • Participants for whom the investigator decides that it is impossible for participant to do "switch (visit 2)" and "step‐down (visit 5)"

Interventions

  • Arm 1: FF/VI 100/25 mcg participants will receive FF/VI 100/25 mcg once‐daily via DPI for 8 weeks in the open‐label treatment period

  • Arm 2: FF 100 mcg participants will receive FP matching placebo twice‐daily (morning and evening) and FF 100 mcg once‐daily in the evening, via DPI for 12 weeks in the double‐blind treatment period

  • Arm 3: FP 250 mcg participants will receive FP 250 mcg twice‐daily (morning and evening) and FF matching placebo once‐daily in the evening, via DPI for 12 weeks in the double‐blind treatment period

  • Arm 4: FP 100 mcg participants will receive FP 100 mcg twice‐daily (morning and evening) and FF matching placebo once‐daily in the evening, via DPI for 12 weeks in the double‐blind treatment period

Outcomes

Primary outcome measures

  • Time to withdrawal due to poorly controlled (requires step‐up) asthma during period 2

  • Proportion of participants with well‐controlled asthma at the end of period 2

Secondary outcome measures

  • Mean change from baseline in clinic visit trough FEV1 at the end of period 2

  • Mean change from baseline in daily AM and PM PEF averaged during period 2

  • Mean change from baseline in percentage of symptom‐free 24‐hour periods during period 2

  • Mean change from baseline in percentage of rescue‐free 24‐hour periods during period 2

  • Mean change from baseline in ACT score during period 2

  • Proportion of participants with ACT score ≥ 20 at the end of period 2

Starting date

March 2014

Contact information

GlaxoSmithKline Research and Development Limited; [email protected]

Notes

NCT02301975

Trial name or title

An efficacy and safety study of fluticasone furoate/vilanterol 100/25 microgram (mcg) inhalation powder, fluticasone propionate/salmeterol 250/50 mcg inhalation powder and fluticasone propionate 250 mcg inhalation powder in adults and adolescents with persistent asthma

Methods

Randomised double‐blind double‐dummy parallel‐group multi‐centre non‐inferiority study

Participants

Inclusion criteria

  • Participants must give their signed and dated written informed consent to participate before commencing any study‐related activities

  • Participants must be outpatients ≥ 12 years of age at visit 1 who have had a diagnosis of asthma, as defined by the National Institutes of Health, for ≥ 12 weeks before visit 1 (Note: Countries with local restrictions prohibiting enrolment of adolescents will enrol only participants ≥ 18 years of age)

  • Participants may be male or an eligible female. An eligible female is defined as having non‐childbearing potential or having childbearing potential and a negative urine pregnancy test at screening and agrees to use an acceptable method of birth control consistently and correctly

  • Participants must have a best pre‐bronchodilator FEV1 ≥ 80% of predicted normal value

  • Participants are eligible if they have received mid‐dose ICS plus LABA (equivalent to FP/SAL 250/50 twice‐daily or an equivalent combination via separate inhalers) for at least the 12 weeks immediately preceding visit 1

  • All participants must be able to replace their current SABA treatment with albuterol/salbutamol aerosol inhaler at visit 1 for use, as needed, for the duration of the study. Participants must be able to withhold albuterol/salbutamol for ≥ 6 hours before study visits

  • If in the opinion of the investigator the participant's asthma is well controlled

Exclusion criteria

  • History of life‐threatening asthma, defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 5 years

  • Culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that is not resolved within 4 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, was expected to affect the participant's asthma status or the ability of the participant to participate in the study

  • Any asthma exacerbation requiring OCS within 12 weeks of visit 1 or resulting in an overnight hospitalisation requiring additional treatment for asthma within 6 months before visit 1

  • Participant must not have current evidence of atelectasis, bronchopulmonary dysplasia, chronic bronchitis, chronic obstructive pulmonary disease, pneumonia, pneumothorax, interstitial lung disease or any evidence of concurrent respiratory disease other than asthma

  • Participant must not have any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of results if the condition/disease was exacerbated during the study

  • Participant must not have used any investigational drug within 30 days before visit 1 or within 5 half‐lives (t½) of the prior investigational study, whichever is longer of the 2

  • Any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or to any intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to the constituents of RELVAR ELLIPTA inhaler, SERETIDE ACCUHALER/DISKUS inhaler or FP 250

  • History of severe milk protein allergy

  • Administration of prescription or non‐prescription medication that would significantly affect the course of asthma, or interact with study drug

  • Participant must not be using or require the use of immunosuppressive medications during the study

  • Participant will not be eligible if he/she or his/her parent or legal guardian has an infirmity, disability, disease or geographical location that seems likely (in the opinion of the investigator) to impair compliance with any aspect of this study protocol, including visit schedule and completion of daily diaries

  • Current tobacco smoker or smoking history of 10 pack‐years (20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products or inhaled marijuana within the past 3 months (e.g. cigarettes, cigars, electronic cigarettes, pipe tobacco)

  • Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or is an employee of the participating investigator

Interventions

  • Experimental 1: FF/VI 100/25 mcg by inhalation once‐daily (PM) via ELLIPTA plus placebo by inhalation twice‐daily (AM and PM) via ACCUHALER/DISKUS for 24 weeks. Interventions: drug: FF/VI 100/25 mcg via ELLIPTA inhaler; drug: placebo inhalation powder via ACCUHALER/DISKUS inhaler

  • Experimental 2: FP/SAL 250/50 mcg by inhalation twice‐daily (AM and PM) via ACCUHALER/DISKUS plus placebo by inhalation once‐daily (PM) via ELLIPTA for 24 weeks. Interventions: drug: placebo inhalation powders via ELLIPTA inhaler; drug: FP/SAL 250/50 mcg via ACCUHALER/DISKUS inhaler

  • Experimental 3: FP 250 mcg by inhalation twice‐daily (AM and PM) via ACCUHALER/DISKUS plus placebo by inhalation once‐daily (PM) via ELLIPTA for 24 weeks

Outcomes

Primary outcome measures

  • Change from baseline in clinic visit PM FEV1 (pre‐bronchodilator and pre‐dose) at the end of the 24‐week treatment period

Secondary outcome measures

  • Change from baseline in percentage of rescue‐free 24‐hour periods during 24‐week treatment period

  • Change from baseline in percentage of symptom‐free 24‐hour periods during 24‐week treatment period

  • Change from baseline in AM PEF averaged over 24‐week treatment period

  • Percentage of participants controlled at the end of the 24‐week treatment period

  • Change from baseline in PM PEF averaged over 24‐week treatment period

Starting date

February 2015

Contact information

GlaxoSmithKline Research and Development Limited; [email protected]

Notes

NCT02301975 2015

Trial name or title

An efficacy and safety study of fluticasone furoate/vilanterol 100/25 microgram (mcg) inhalation powder, FP/SAL 250/50 mcg inhalation powder and fluticasone propionate 250 mcg inhalation powder in adults and adolescents with persistent asthma

Methods

This study is a randomised double‐blind double‐dummy parallel‐group multi‐centre non‐inferiority study

Participants

Inclusion criteria

  • Participants must give their signed and dated written informed consent to participate before commencing any study‐related activities

  • Participants must be outpatients ≥ 12 years of age at visit 1 who have had a diagnosis of asthma, as defined by the National Institutes of Health, for ≥ 12 weeks before visit 1 (Note: Countries with local restrictions prohibiting enrolment of adolescents will enrol only participants ≥ 18 years of age)

  • Participants may be male or an eligible female. An eligible female is defined as having non‐childbearing potential or having childbearing potential and a negative urine pregnancy test at screening and agrees to use an acceptable method of birth control consistently and correctly

  • Participants must have FEV1 ≥ 80% of predicted normal value

  • Participants are eligible if they have received mid‐dose ICS plus LABA (equivalent to FP/SAL 250/50 twice‐daily or an equivalent combination via separate inhalers) for at least the 12 weeks immediately preceding visit 1

  • All participants must be able to replace their current SABA treatment with albuterol/salbutamol aerosol inhaler at visit 1 for use, as needed, for the duration of the study. Participants must be able to withhold albuterol/salbutamol for ≥ 6 hours before study visits

  • If in the opinion of the investigator, the participant's asthma is well controlled

Exclusion criteria

  • History of life‐threatening asthma, defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 5 years

  • Culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that is not resolved within 4 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, was expected to affect the participant's asthma status or the participant's ability to participate in the study

  • Any asthma exacerbation requiring oral corticosteroids within 12 weeks of visit 1 or resulting in an overnight hospitalisation requiring additional treatment for asthma within 6 months before visit 1

  • Participant must not have current evidence of atelectasis, bronchopulmonary dysplasia, chronic bronchitis, chronic obstructive pulmonary disease, pneumonia, pneumothorax, interstitial lung disease, or any evidence of concurrent respiratory disease other than asthma

  • Participant must not have any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of results if the condition/disease was exacerbated during the study

  • Participant must not have used any investigational drug within 30 days before visit 1 or within 5 half‐lives (t½) of the prior investigational study, whichever is longer of the 2

  • Any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or any intranasal, inhaled or systemic corticosteroid therapy. Known or suspected sensitivity to the constituents of RELVAR ELLIPTA inhaler, SERETIDE ACCUHALER/DISKUS inhaler or FP 250

  • History of severe milk protein allergy

  • Administration of prescription or non‐prescription medication that would significantly affect the course of asthma, or interact with study drug

  • Participant must not be using or require the use of immunosuppressive medications during the study

  • Participant will not be eligible if he/she or his/her parent or legal guardian has an infirmity, disability, disease or geographical location that seems likely (in the opinion of the investigator) to impair compliance with any aspect of this study protocol, including visit schedule and completion of daily diaries

  • Current tobacco smoker or has a smoking history of 10 pack‐years (20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products or inhaled marijuana within the past 3 months (e.g. cigarettes, cigars, electronic cigarettes, pipe tobacco)

  • Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or an employee of the participating investigator

Interventions

The study will enrol adult and adolescent asthmatic participants who are currently receiving mid‐dose inhaled corticosteroids (ICSs) plus a long‐acting beta2‐agonist (LABA) (equivalent to FP/SAL 250/50 microgram (mcg) twice‐daily (BD)), via a fixed‐dose combination product or through separate inhalers. The study consists of a LABA washout period of 5 days and a run‐in period of 4 weeks, followed by a treatment period of 24 weeks and a follow‐up contact period of 1 week. The total duration of the study is 30 weeks. Approximately 1461 participants will be randomised to 1 of the following 3 treatments (487 per treatment): FF/VI 100/25 mcg once‐daily (OD) in the evening (PM) via ELLIPTA inhaler plus placebo BD via ACCUHALER/DISKUS; FP/SAL 250/50 mcg BD via ACCUHALER/DISKUS inhaler plus placebo OD (PM) via ELLIPTA inhaler; FP 250 mcg BD via ACCUHALER/DISKUS inhaler plus placebo OD (PM) via ELLIPTA inhaler. In addition, all participants will be supplied with albuterol/salbutamol inhalation aerosol for use as needed to treat acute asthma symptoms

Outcomes

Primary outcome measures

  • Change from baseline in clinic visit evening (PM) forced expiratory volume in 1 second (FEV1) (pre‐bronchodilator and pre‐dose) at the end of the 24‐week treatment period (time frame: baseline and week 24) (designated as safety issue: no) FEV1 is a measure of lung function and is defined as the maximal amount of air that can be forcefully exhaled in 1 second. Baseline will be the pre‐dose value obtained at the visit 3 clinic visit. Change from baseline will be calculated as the week 24 value minus the baseline value

Secondary outcome measures

  • Change from baseline in percentage of rescue‐free 24‐hour periods during 24‐week treatment period (time frame: baseline and weeks 1 to 24) (designated as safety issue: no). Participants will record the number of inhalations of rescue medication used during the day and night in a daily electronic diary (eDiary). A 24‐hour period in which a participant's responses to both morning and evening assessments indicated no rescue medication use will be considered to be a rescue‐free 24‐hour period. Baseline value will be derived from the last 7 days of the daily eDiary before randomisation of participant. Change from baseline will be calculated as the value during the 24‐week treatment period minus the baseline value

  • Change from baseline in percentage of symptom‐free 24‐hour periods during 24‐week treatment period (time frame: baseline and weeks 1 to 24) (designated as safety issue: no). Asthma symptoms will be recorded in a daily eDairy by participants every day morning and evening. A 24‐hour period in which a participant's responses to both morning and evening assessments indicated no symptoms will be considered to be a symptom‐free 24‐hour period. Baseline value will be derived from the last 7 days before randomisation of the participant. Change from baseline will be calculated as the value during the 24‐week treatment period minus the baseline value

  • Change from baseline in morning (AM) peak expiratory flow (PEF) averaged over 24‐week treatment period (time frame: baseline and weeks 1 to 24) (designated as safety issue: no). PEF is defined as maximum airflow during a forced expiration beginning with the lungs fully inflated. PEF will be measured by participants using a hand‐held electronic peak flow meter each morning before the dose of study medication and any rescue albuterol/salbutamol inhalation aerosol use. The best of 3 measurements will be recorded. Change from baseline (defined from the last 7 days before randomisation of the participant) will be calculated as the value of the averaged daily AM PEF over the 24‐week treatment period minus the baseline value

  • Percentage of participants controlled at the end of the 24‐week treatment period (time frame: week 24) (designated as safety issue: no). Percentage of participants controlled will be defined using an Asthma Control Test (ACT) score ≥ 20 at the end of the 24‐week treatment period. The ACT is a 5‐item questionnaire developed as a measure of a participant's asthma control that can be quickly and easily completed in clinical practice. The questionnaire will be self completed by participants

  • Change from baseline in PM PEF averaged over 24‐week treatment period (time frame: baseline and weeks 1 to 24) (designated as safety issue: no). PEF is defined as maximum airflow during a forced expiration beginning with the lungs fully inflated. PEF will be measured by participants using a hand‐held electronic peak flow meter each evening before the dose of study medication and any rescue albuterol/salbutamol inhalation aerosol use. Change from baseline (defined as the last 7 days before randomisation of participants) will be calculated as the value of the averaged daily PM PEF over the 24‐week treatment period minus the baseline value

Starting date

March 2015

Contact information

US GSK Clinical Trials Call Center

Notes

NCT02446418 2015

Trial name or title

A study to compare the efficacy of fluticasone furoate/vilanterol inhalation powder with usual inhaled corticosteroids (ICS)/long‐acting beta‐agonists (LABA) in persistent asthma

Methods

Multi‐centre open‐label randomised parallel‐group study

Participants

Inclusion criteria

  • Informed consent: capable of giving signed informed consent, which includes compliance with requirements and restrictions listed in the consent form and in this protocol

  • Gender and age: male or female participants ≥ 18 and ≤ 75 years of age at screening visit

Female participant is eligible to participate if she is not pregnant (as confirmed by a negative urine human chorionic gonadotrophin (hCG) test), is not lactating and at least one of the following conditions applies:

  • Non‐reproductive potential defined as pre‐menopausal females with 1 of the following: documented tubal ligation; documented hysteroscopic tubal occlusion procedure with follow‐up confirmation of bilateral tubal occlusion; hysterectomy; documented bilateral oophorectomy. Postmenopausal defined as 12 months of spontaneous amenorrhoea (in questionable cases, a blood sample with simultaneous follicle‐stimulating hormone (FSH) and oestradiol levels consistent with menopause (refer to laboratory reference ranges for confirmatory levels)); females on hormone replacement therapy (HRT) and whose menopausal status is in doubt will be required to use one of the highly effective contraception methods if they wish to continue their HRT during the study. Otherwise, they must discontinue HRT to allow confirmation of post‐menopausal status before study enrolment

  • Reproductive potential and agrees to follow 1 of the options listed below in the GSK Modified List of Highly Effective Methods for Avoiding Pregnancy in Females of Reproductive Potential (FRP) requirements from 30 days before the first dose of study medication and until week 24

GSK Modified List of Highly Effective Methods for Avoiding Pregnancy in FRP

This list does not apply to FRP with same sex partners, when this is their preferred and usual lifestyle, or for participants who are and will continue to be abstinent from penile‐vaginal intercourse on a long‐term and persistent basis: contraceptive subdermal implant that meets standard operating procedure (SOP) effectiveness criteria, including a < 1% rate of failure per year, as stated in the product label; intrauterine device or intrauterine system that meets SOP effectiveness criteria, including a < 1% rate of failure per year, as stated in the product label; oral contraceptive, either combined or progestogen alone; injectable progestogen; contraceptive vaginal ring; percutaneous contraceptive patches; male partner sterilisation with documentation of azoospermia before entry of female participant into the study, and this male is the sole partner for that participant; male condom combined with a vaginal spermicide (foam, gel, film, cream or suppository). These allowed methods of contraception are effective only when used consistently, correctly and in accordance with the product label. The investigator is responsible for ensuring that participants understand how to properly use these methods of contraception.

  • Types of participants: participants with documented physician's diagnosis of asthma ≥ 1 year, unsatisfactorily controlled asthma (ACT < 20 at screening and randomisation visit) treated by ICS alone and intended to be treated by ICS/LABA maintenance therapy; participant will be eligible for inclusion in this study only if affiliated with or a beneficiary of a Social Security category

  • Current asthma therapy: All participants must be prescribed maintenance therapy and receiving ICS alone without LABA for ≥ 4 weeks before randomisation visit; other background asthma medication such as anti‐leukotrienes or theophylline is permitted as an alternative to ICS alone, if initiated ≥ 4 weeks before screening visit

  • Participant questionnaires: Participants must be able to complete the questionnaires themselves

Exclusion criteria:

  • History of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the last 6 months before screening and randomisation visit

  • Participants having a severe and unstable asthma, with ACT score < 15 at screening and randomisation visit, history of repeated severe exacerbations (3/y) and/or a severe exacerbation in the previous 6 weeks before screening and randomisation visit

  • Chronic obstructive pulmonary disease (COPD): respiratory disease: A participant must not have current evidence or diagnosis of chronic obstructive pulmonary disease at screening visit

  • Current or former cigarette smokers with a history of cigarette smoking ≥ 10 pack‐years at screening (number of pack‐years = (number of cigarettes per day/20) × number of years smoked (e.g. 20 cigarettes per day for 10 years, 10 cigarettes per day for 20 years))

  • Other diseases/abnormalities: participants with historical or current evidence of uncontrolled or clinically significant disease at screening and randomisation visit. Significant is defined as any disease that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would affect the efficacy or safety analysis if the disease/condition was exacerbated during the study

  • Participants with a history of adverse reaction, including immediate or delayed hypersensitivity to any intranasal, inhaled or systemic corticosteroid and LABA therapy and to components of the inhalation powder (e.g. lactose, magnesium stearate) at screening and randomisation visit. In addition, participants with a history of severe milk protein allergy that, in the opinion of the Investigator, contraindicates the participant's participation

  • Investigational medications: A participant must not have used any investigational drug within 30 days before randomisation visit or within 5 half‐lives (t½) of the prior investigational study (whichever is longer of the 2) (if unsure, discuss with the medical monitor before screening)

  • Long‐term user of systemic corticosteroids: participant who, in the opinion of the Investigator, is considered to be a long‐term user of systemic corticosteroids for respiratory or other indications (if unsure, discuss with the medical monitor before screening) at screening visit

  • Participants treated by the monoclonal antibody omalizumab at screening visit. Treatment with omalizumab is not allowed during the study

  • Participants involved in other clinical trials at screening visit

  • Affiliation with investigator site: is an investigator, sub‐investigator, study co‐ordinator or employee of a participating investigator or study site, or immediate family member of the aforementioned who is involved in this study

  • Participants who plan to move away during the study from the geographical area where the study is being conducted

Interventions

To evaluate the efficacy and safety of FF/VI compared with 2 usual ICS/LABA fixed combinations (FP/SAL or budesonide/formoterol (BUD/F)) in participants with persistent asthma, in "close to real life" settings. FF/VI will be administered once‐daily (QD) via ELLIPTA dry powder inhaler (DPI), and FP/SAL or BUD/F will be administered twice‐daily (BID) via DISKUS and TURBUHALER DPI, respectively. ELLIPTA is a new powder inhaler designed to be easy to use. The total duration of individual participation will be approximately 6 months (24 weeks)

Outcomes

Primary outcome measures

  • Change from baseline in Asthma Control Test (ACT) total score at week 12 (time frame: baseline and week 12) (designated as safety issue: no). ACT is a validated self administered questionnaire utilising 5 questions to assess asthma control during the past 4 weeks on a 5‐point categorical scale (1 to 5). The total score is calculated as the sum of the scores from all 5 questions. By answering all 5 questions, a participant with asthma can obtain a score that may range between 5 and 25, with higher scores indicating better control. Change from baseline was calculated as total ACT score at week 12 minus total ACT score at baseline

Secondary outcome measures

  • Change from baseline in ACT score at week 24 (time frame: baseline and week 24) (designated as safety issue: no). ACT is a validated self administered questionnaire utilising 5 questions to assess asthma control during the past 4 weeks on a 5‐point categorical scale (1 to 5). The total score is calculated as the sum of the scores from all 5 questions. By answering all 5 questions, a participant with asthma can obtain a score that may range between 5 and 25, with higher scores indicating better control. Change from baseline was calculated as total ACT score at week 24 minus total ACT score at baseline

  • Percentage of participants making at least 1 Type A error (likely to be critical) and overall errors at week 12 and at week 24 independent of use at week 12 (time frame: week 12 and week 24) (designated as safety issue: no). Participants will be asked to read the instruction leaflet of the assigned device and will be instructed by the investigator on the proper use of inhalers. Then, participant will be asked to self administer dose of assigned study drug. Any errors (critical or non‐critical) made by the participant will be recorded by the healthcare professional (HCP). A critical error is defined as an error that may impact the ability of the drug to reach the lung and hence impact efficacy. Overall errors includes non‐critical errors

Starting date

July 2015

Contact information

US GSK Clinical Trials Call Center

Notes

NCT02730351 2016

Trial name or title

Crossover study comparing fluticasone furoate (FF)/vilanterol (VI) once‐daily versus fluticasone propionate (FP) twice‐daily in participants with asthma and exercise‐induced bronchoconstriction (EIB)

Methods

Multi‐centre randomised double‐blind double‐dummy cross‐over study with two 2‐week treatment periods separated by a 2‐week washout period

Participants

Participants with asthma and exercise‐induced bronchoconstriction (EIB) between 12 and 50 years of age

Inclusion criteria

Participants eligible for enrolment in the study must meet the following criteria

  • Informed consent: Participants must give their signed and dated written informed consent to participate before commencing any study‐related activities

  • Age range: 12 to 50 years of age, inclusive, at visit 1 (screening)

  • Diagnosis: diagnosis of asthma, as defined by the National Institutes of Health for ≥ 12 weeks before visit 1

  • Asthma severity: Participants must have a pre‐bronchodilator FEV1 ≥ 70% of predicted normal value. Predicted values will be based upon Global Lung Function Initiative equations for spirometry reference values

  • Evidence of EIB: Participants must answer "Yes" to at least 2 of the following 3 questions reflecting on the previous 12 months: Are you short of breath during exercise or other physical exertion? Do you wheeze after exercise or other physical exertion? Do you cough after exercise or other physical exertion?

  • Concurrent antiasthma therapy: Participants must be taking low‐ to moderate‐dose inhaled steroids for 12 weeks before visit 1 to participate, with no change in dose for the 4 weeks before visit 1

  • Gender: Participants may be male or an eligible female. A female is eligible to enter and participate in the study if she is of non‐childbearing potential (i.e. physiologically incapable of becoming pregnant, including any female who is post‐menopausal or surgically sterile). Surgically sterile females are defined as those with a documented hysterectomy and/or bilateral oophorectomy or tubal ligation. Post‐menopausal females are defined as amenorrhoeic for longer than 1 year, with an appropriate clinical profile (e.g. age appropriate, > 45 years), in the absence of hormone replacement therapy

OR

  • Childbearing potential: has a negative pregnancy test at screening, and agrees to acceptable contraceptive methods approved in local country, when used consistently and correctly (i.e. in accordance with the approved product label and instructions of the physician for the duration of the study ‐ screening to follow‐up contact)

  • Albuterol/salbutamol use: All participants must be able to replace their current short‐acting beta2‐agonist with albuterol/salbutamol, to be used only on an as‐needed basis for the duration of the study. Each participant must be judged capable of withholding albuterol/salbutamol for ≥ 6 hours before performing spirometric evaluations

  • Physical capacity: Each participant must be physically able to perform exercise challenges on a treadmill when bronchodilators have been withheld

Exclusion criteria

Participants are not eligible for enrolment in the study if they meet the following criteria

  • Intermittent asthma, seasonal asthma or exercise‐induced bronchoconstriction only: Participants with only intermittent or seasonal asthma or only exercise‐induced asthma are excluded from participation in this study

  • History of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 10 years

  • Asthma exacerbation: any asthma exacerbation requiring oral corticosteroids within 12 weeks of visit 1, or that resulted in an overnight hospitalisation requiring additional treatment for asthma within 6 months before visit 1

  • Symptomatic allergic rhinitis: Participants with symptomatic allergic rhinitis at visit 1 may be treated for up to 4 weeks with intranasal corticosteroids, followed by a repeat screening visit to determine eligibility before entry into the study. Participants who continue to be symptomatic after up to 4 weeks of treatment will be excluded

  • 12‐Lead electrocardiogram (ECG): A participant is not eligible if he/she has an abnormal, clinically significant ECG as determined by investigator at the screening visit.

  • Pregnancy: women who are pregnant or lactating or are planning on becoming pregnant during the study

  • Respiratory infection: culture‐documented or suspected bacterial or viral infection of the upper or lower respiratory tract, sinus or middle ear that is not resolved within 4 weeks of visit 1 and led to a change in asthma management or, in the opinion of the investigator, is expected to affect participant's asthma status or ability of participant to participate in the study

  • Concurrent respiratory disease: A participant must not have current evidence of atelectasis, bronchopulmonary dysplasia, chronic bronchitis, chronic obstructive pulmonary disease (COPD) (current or past diagnosis including asthma/COPD overlap), pneumonia, pneumothorax, interstitial lung disease or any evidence of concurrent respiratory disease other than asthma

  • Other concurrent diseases/abnormalities: A participant must not have any clinically significant, uncontrolled condition or disease state that, in the opinion of the investigator, would put the safety of the patient at risk through study participation or would confound interpretation of efficacy results if the condition/disease was exacerbated during the study

  • Investigational medications: A participant must not have used any investigational drug within 30 days before visit 1 or within 5 half‐lives (t1/2) of the prior investigational study, whichever is longer of the 2 periods

  • Allergies: drug allergy: any adverse reaction including immediate or delayed hypersensitivity to any beta2‐agonist or sympathomimetic drug, or to any intranasal, inhaled or systemic corticosteroid therapy, or excipients used with FF/VI 100/25 or FP 250 (i.e. drug, lactose or magnesium stearate); milk protein allergy: history of severe milk protein allergyl latex allergy: history of allergy or sensitivity to latex that, in the opinion of the investigator, contraindicates participation of the patient in the study

  • Concomitant medication: administration of prescription or non‐prescription medication that would significantly affect the course of asthma, or interact with study drug

  • Immunosuppressive medication: A participant must not be using or require the use of immunosuppressive medications during the study

  • Compliance: A participant will not be eligible if he/she or his/her parent or legal guardian has an infirmity, disability, disease or geographical location that seems likely (in the opinion of the investigator) to impair compliance with any aspect of this study protocol

  • Tobacco/Marijuana use: current tobacco smoker or has a smoking history of ≥ 10 pack‐years (20 cigarettes/d for 10 years). Participant may not have used inhaled tobacco products or inhaled marijuana within the past 3 months (e.g. cigarettes, cigars, electronic cigarettes, pipe tobacco)

  • Affiliation with investigator's site: Participant will not be eligible for this study if he/she is an immediate family member of the participating investigator, subinvestigator or study co‐ordinator, or an employee of the participating investigator

Interventions

This study is designed to compare fluticasone furoate (FF)/vilanterol (VI) once‐daily vs fluticasone propionate (FP) twice‐daily in participants with asthma and exercise‐induced bronchoconstriction (EIB)

Outcomes

Primary outcome measures

  • Maximal percent decrease from pre‐exercise forced expiratory volume in 1 second (FEV1) following exercise challenge at 12 hours post evening dose at the end of 2‐week treatment period

  • Maximal percent decrease will be defined as percent change from pre‐exercise FEV1 to minimum FEV1 collected within 1 hour following exercise challenge at 12 hours post dose

Secondary outcome measures

  • Maximal percent decrease from pre‐ exercise FEV1 following exercise challenge at 23 hours post evening dose at the end of 2‐week treatment period

  • Maximal percent decrease will be defined as percent change from pre‐exercise FEV1 to minimum FEV1 collected within 1 hour following exercise challenge at 23 hours post dose. Pre‐exercise FEV1 will be defined as FEV1 value collected before the exercise challenge test at 23 hours post dose. Serial spirometry will be performed at time points 5, 10, 15, 30, 45 and 60 minutes post exercise challenges

  • Proportion of participants with a 30‐minute post‐challenge FEV1 that was no more than 5% lower than their pre‐exercise FEV1 following the exercise challenge at 12 hours and 23 hours post evening dose at the end of 2‐week treatment period

  • Serial spirometry will be performed at time points 5, 10, 15, 30, 45 and 60 minutes post exercise challenges

  • Weighted mean for percent decrease from pre‐exercise FEV1 following exercise challenge at 12 hours and 23 hours post evening dose at the end of 2‐week treatment period

  • Serial spirometry will be performed at time points 5, 10, 15, 30, 45 and 60 minutes post exercise challenges

Starting date

March 2016

Contact information

GlaxoSmithKline

Notes

NCT02753712 2016

Trial name or title

A study to evaluate the effect of fluticasone/formoterol breath actuated inhaler (BAI) or Relvar Ellipta DPI on ventilation heterogeneity in asthma

Methods

A randomised assessor‐blinded parallel‐group trial

Participants

Inclusion criteria for participants on Seretide Accuhaler 250/50 µg at screening

  • Male and female participants ≥ 18 years old

  • Adequate contraception

  • Documented clinical history of asthma for ≥ 6 months before screening visit

  • Using Seretide Accuhaler at a stable dose of 250/50 μg twice‐daily at screening for ≥ 8 weeks

  • Uncontrolled asthma as defined by Asthma Control Questionnaire (ACQ‐6) score ≥ 1.0

  • R5‐R20 ≥ 0.10 kPa/L/s as measured on impulse oscillometry during screening visit

  • Historical evidence (within 24 months) of eosinophilic airways disease evidenced by sputum eosinophil count ≥ 3% and/or FeNO 35 ppb

Inclusion criteria for participants on equivalent /higher dose or other ICS‐LABAs or higher dose of Seretide at screening

  • Male and female participants ≥ 18 years old

  • Adequate contraception

  • Documented clinical history of asthma for ≥ 6 months before screening visit

  • R5‐R20 ≥ 0.07 kPa/L/s as measured on impulse oscillometry during screening visit 5. Historical evidence (within past 24 months) of eosinophilic airways disease, evidenced by sputum eosinophil count ≥ 3% and/or FeNo ≥ 35 ppb

Exclusion criteria for all participants

  • Any severe chronic respiratory disease other than asthma

  • Participant has a smoking history ≥ 10 "pack‐years" (i.e. ≥ 1 pack of 20 cigarettes/d for 10 years or 10 packs/d for 1 year, etc)

  • Current smoking history within 12 months before screening visit

  • Near fatal or life‐threatening (including intubation) asthma within the past year

  • Known history of systemic (injectable or oral) corticosteroid medication within 1 month of visit 1

  • Evidence of clinically unstable disease as determined by medical history or physical examination that, in the investigator's opinion, precludes entry into the study. 'Clinically unstable' is defined as any disease that, in the opinion of the Investigator, would put the patient at risk through study participation, or would affect the outcome of the study

  • In the investigator's opinion, a clinically significant upper or lower respiratory infection within 4 weeks before visit 1

  • Current evidence or known history of alcohol and/or substance abuse within 12 months before screening visit

  • Participant has taken β‐blocking agents, tricyclic antidepressants, monoamine oxidase inhibitors, astemizole, quinidine type antiarrhythmics or potent CYP 3A4 inhibitors such as ketoconazole within 1 week before screening visit

  • Current use of bronchodilators/anti‐inflammatory agents other than those specified in the protocol

  • Known or suspected sensitivity to study drug or excipients

  • Participation in a clinical drug study within 30 days of screening visit

  • Current participation in a clinical study

Exclusion criteria for participant or participants undergoing OR‐MRI and HD‐CT

  • Contraindication for MRI scanning (as assessed by local MRI safety questionnaire), which includes but is not limited to presence of non‐MRI compatible artificial heart valves, hydrocephalus shunts, intracranial aneurysm clips, joint replacements or metal implants, pacemakers or other cardiac rhythm management devices, claustrophobia, history of metal in the eye, presence of shrapnel from a war injury, callipers or braces, dentures, dental plates or hearing aids that include metal and cannot be removed, history of epilepsy or blackouts, ear implants, piercings that cannot be removed, intrauterine contraceptive device or coil

  • Inability to stay in the supine position for the duration of the scanning procedure

  • Obesity (body weight > 140 kg)

Interventions

Comparison between fluticasone/formoterol BAI vs fluticasone/vilanterol DPI (Relvar Ellipta DPI)

Outcomes

Primary outcome measures

  • Measurement of peripheral airway resistance (R5‐R20)

  • Measurement of peripheral airway resistance (R5‐R20)

Secondary outcome measures

  • Ventilation heterogeneity (using functional respiratory imaging)

  • Distal airway volume and resistance (using impulse oscillometry)

  • Evaluation of asthma control (using ACQ‐6)

  • Evaluation of health status (using AQLQ

Starting date

April 2016

Contact information

Mundipharma Research Limited

Notes

New 2014 (NCT01551758)

Trial name or title

A randomised effectiveness study comparing fluticasone furoate (FF, GW685698)/vilanterol (VI, GW642444) with standard treatment in chronic obstructive pulmonary disease (COPD)

Methods

This is a phase III multi‐centre randomised open‐label study

Participants

Inclusion criteria

Participants eligible for enrolment in the study must meet all of the following criteria

  • Types of participants: participants with documented GP diagnosis of COPD, and currently receiving maintenance therapy

  • Informed consent: Participants must be able to provide informed consent, have their consent signed and dated. Participants must be able to complete the electronic participant questionnaires or allow a proxy to do so on their behalf

  • Gender and age: male or female participants ≥ 40 years of age at visit 1. A female is eligible to enter and participate in the study if she is of non‐childbearing potential (i.e. physiologically incapable of becoming pregnant, including any female who is post‐menopausal or surgically sterile). Surgically sterile females are defined as those with a documented hysterectomy and/or bilateral oophorectomy or tubal ligation. Post‐menopausal females are defined as amenorrhoeic for longer than 1 year with an appropriate clinical profile (e.g. age appropriate, history of vasomotor symptoms). However, in questionable cases, a blood sample with FSH > 40 MIU/mL and oestradiol < 40 pg/mL (< 140 pmol/L) is confirmatory. Or with childbearing potential has a negative urine pregnancy test at visit 2, and agrees to 1 of the highly effective and acceptable contraceptive methods used consistently and correctly (i.e. in accordance with the approved product label and instructions of the physician for the duration of the study ‐ visit 2 to the end of the study)

  • Participants with exacerbation history

  • Current COPD maintenance therapy

Exclusion criteria

Participants meeting any of the following criteria must not be enrolled in the study

  • Participants with any life‐threatening condition (e.g. low probability (in the opinion of the GP/Investigator) of 12‐month survival due to severity of COPD or co‐morbid condition) at point of entry into the study

  • Other diseases/abnormalities: participants with historical or current evidence of uncontrolled or clinically significant disease. Significant is defined as any disease that, in the opinion of the GP/ Investigator, would put the safety of the patient at risk through participation or would affect the efficacy or safety analysis if the disease/condition was exacerbated during the study

  • Participants with unstable COPD, defined as the occurrence of the following in the 2 weeks before visit 2: acute worsening of COPD that is managed by the participant with corticosteroids or antibiotics or that requires treatment prescribed by a physician

  • Long‐term user of oral corticosteroids: participants who, in the opinion of the GP/Investigator, are considered to be long‐term users of oral corticosteroids for respiratory or other indications (if unsure, discuss with the medical monitor before screening)

  • Drug/food allergy: participants with a history of hypersensitivity to any of the study medications (e.g. beta‐agonists, corticosteroids) or components of the inhalation powder (e.g. lactose, magnesium stearate). In addition, participants with a history of severe milk protein allergy that, in the opinion of the GP/investigator, contraindicates the patient's participation

  • Investigational medications: A participant must not have used any investigational drug treatment within 30 days before visit 2 or within 5 half‐lives (t½) of the prior investigational study (whichever is the longer of the 2)

  • Participants who plan to move away during the study period from the geographical area where the study is being conducted and/or participants who have not consented to inclusio of their medical records in the electronic medical records database that is operational in the Salford area

Interventions

This study is designed to compare the effectiveness and safety of FF/VI. Inhalation powder (100 mcg FF, GW685698)/25 mcg VI, GW642444)) delivered once‐daily via a novel dry powder inhaler (NDPI) compared with existing COPD maintenance therapy over 12 months in participants diagnosed with COPD. Participants who meet eligibility criteria are randomised and will enter a 12‐month treatment period

Outcomes

Primary outcome measures

  • Mean annual rate of moderate and severe exacerbations (time frame: 12 months) (designated as safety issue: no). A moderate exacerbation is defined by the participant receiving an exacerbation‐related prescription of oral corticosteroids and/or antibiotic (with NHS contact) not requiring hospitalisation. A severe exacerbation is defined as an exacerbation‐related hospitalisation

Secondary outcome measures

  • Variety of healthcare utilisation endpoints (time frame: 12 months). (designated as safety issue: no). Healthcare utilisation

  • Serious adverse events and non‐serious adverse drug reactions (time frame: 12 months). (designated as safety issue: no). Frequency and types of serious adverse events and non‐serious adverse drug reactions

  • Patient‐reported outcomes (time frame: 12 months). (designated as safety issue: no). Patient‐reported outcomes

Starting date

January 2012

Contact information

GlaxoSmithKline

Notes

Woodcock 2014 (NCT01706198)

Trial name or title

An effectiveness study comparing fluticasone furoate (FF, GW685698)/vilanterol (VI, GW642444) with standard treatment in asthma

Methods

Multi‐centre randomised open‐label study

Participants

Inclusion criteria

Participants eligible for enrolment in the study must meet all of the following criteria

  • Informed consent: Participants must be able to provide informed consent, have their consent signed and dated

  • Types of participants: participants with documented GP diagnosis of asthma as their primary respiratory disease

  • Current antiasthma therapy: All participants must be prescribed maintenance therapy and must be receiving ICS with or without LABA (fixed combination or via separate inhalers) and for at least 4 weeks before visit 2. Other background asthma medications such as anti‐leukotrienes are permitted

  • All participants receiving ICS monotherapy or ICS/LABA combination (this can be a fixed‐dose combination or an ICS alone or LABA alone in separate inhalers) must have had symptoms in the past week before visit 2. Symptoms are defined by daytime symptoms more than twice per week, use of a short‐acting beta2‐agonist bronchodilator more than twice per week, any limitation of activities or any nocturnal symptoms/awakening (Symptoms are based on participant's recall and are consistent with the GINA and in principle with BTS/SIGN guidelines)

  • Participant questionnaires: Participants must be able to complete the electronic participant questionnaires as well as those questionnaires completed by phone or provide a proxy (e.g. partner/relative/friend who can do so on their behalf)

  • Gender and age: male or female participants ≥ 18 years of age at visit 1. A female is eligible to enter and participate in the study if she is of:non‐childbearing potential (i.e. physiologically incapable of becoming pregnant, including any female who is post‐menopausal or surgically sterile). Surgically sterile females are defined as those with a documented hysterectomy and/or bilateral oophorectomy or tubal ligation. Post‐menopausal females are defined as amenorrhoeic for longer than 1 year with an appropriate clinical profile (e.g. age appropriate, history of vasomotor symptoms). However, in questionable cases, a blood sample with FSH > 40 MIU/mL and oestradiol < 40 pg/mL (< 147 pmol/L) is confirmatory

OR

Childbearing potential with a negative urine pregnancy test at visit 2, and agrees to one of the highly effective and acceptable contraceptive methods used consistently and correctly (i.e. in accordance with the approved product label and instructions of the physician for the duration of the study ‐ visit 2 to the end of the study)

Exclusion criteria

Participants meeting any of the following criteria must not be enrolled in the study

  • Recent history of life‐threatening asthma: defined for this protocol as an asthma episode that required intubation and/or was associated with hypercapnia, respiratory arrest or hypoxic seizures within the past 6 months

  • COPD: respiratory disease: Participant must not have current evidence or GP diagnosis of chronic obstructive pulmonary disease

  • Other diseases/abnormalities: participants with historical or current evidence of uncontrolled or clinically significant disease. Significant is defined as any disease that, in the opinion of the GP/investigator, would put the safety of the patient at risk through study participation or would affect the efficacy or safety analysis if the disease/condition was exacerbated during the study

  • Drug/food allergy: participants with a history of hypersensitivity to any of the study medications (e.g. beta2‐agonists, corticosteroid) or components of the inhalation powder (e.g. lactose, magnesium stearate). In addition, participants with a history of severe milk protein allergy that, in the opinion of the GP/ Investigator, contraindicates the patient's participation

  • Investigational medications: Participant must not have used any investigational drug within 30 days before visit 2 or within 5 half‐lives (t½) of the prior investigational study (whichever is longer of the 2) (if unsure, discuss with the medical monitor before screening)

  • Long‐term user of systemic corticosteroids: participant who, in the opinion of the GP/investigator, is considered to be a long‐term user of systemic corticosteroids for respiratory or other indications (if unsure, discuss with the medical monitor before screening)

  • Participants who are using LABA without an ICS as asthma maintenance therapy

  • Participants who plan to move away during the study period from the geographical area where the study is being conducted and/or participants who have not consented to inclusion of their medical records in the electronic medical records database that is operational in the Salford area

Interventions

This study is designed to compare the effectiveness and safety of FF/VI inhalation powder ((100 mcg FF), GW685698)/25 mcg VI, GW642444) or 200 mcg FF, GW685698)/25 mcg VI, GW642444)) delivered once‐daily via a novel dry powder inhaler (NDPI) compared with the existing asthma maintenance therapy over 12 months in participants diagnosed with asthma. Participants who meet the eligibility criteria are randomised and will enter a 12‐month treatment period

Outcomes

Primary outcome measures

  • Percentage of participants who have an ACT total score ≥ 20 at week 24 (6th month) assessment (time frame: week 24) (designated as safety issue: no]

  • Percentage of participants who have an ACT total score ≥ 20 at week 24 (6th month) assessment

Secondary outcome measures

  • Percentage of participants with asthma control (ACT total score ≥ 20) (time frame: weeks 12, 40 and 52) (designated as safety issue: no). Percentage of participants with asthma control (ACT total score ≥ 20)

  • Mean change from baseline in ACT total score (time frame: weeks 12, 24, 40 and 52) (designated as safety issue: no). Mean change from baseline in ACT total score

  • Percentage of participants who have an increase from baseline ≥ 3 in ACT total score (time frame: weeks 12, 24, 40 and 52) (designated as safety issue: no). Percentage of participants who have an increase from baseline ≥ 3 in ACT total score

  • Asthma‐related secondary care contacts (time frame: 12 months) (designated as safety issue: no). Asthma‐related secondary care contacts

  • Asthma‐related primary care contacts (time frame: 12 months) (designated as safety issue: no). Asthma‐related primary care contacts

  • All secondary care contacts (time frame: 12 months) (designated as safety issue: no). All secondary care contacts

  • All primary care contacts (time frame: 12 months) (designated as safety issue: no). All primary care contacts

  • Mean annual rate of severe asthma exacerbations (time frame: 12 months) (designated as safety issue: no). Mean annual rate of severe asthma exacerbations

  • Number of salbutamol inhalers (adjusted to equivalence of 200 actuations) collected by participants from study‐enrolled community pharmacies over the entire treatment period (time frame: 12 months) (designated as safety issue: no). Number of salbutamol inhalers (adjusted to equivalence of 200 actuations) collected by participants from study‐enrolled community pharmacies over the entire treatment period

  • Time to discontinuation or modification of initial therapy (time frame: 12 months) (designated as safety issue: no). Time to discontinuation or modification of initial therapy

  • Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) total score at week 52 (time frame: week 52) (designated as safety issue: no). Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) total score at week 52

  • Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) environmental stimuli domain score at week 52 (time frame: week 52) (designated as safety issue: no). Percentage of participants who have an increase from baseline ≥ 0.5 in AQLQ(S) environmental stimuli domain score at week 52

Starting date

November 2012

Contact information

US GSK Clinical Trials Call Center

Notes

ACT: Asthma Control Test

AE: adverse event

AM: morning

AQLQ: Asthma Quality of Life Questionnaire

BTS: British Thoracic Society

COPD: chronic obstructive pulmonary disease

DPI: dry powder inhaler

ECG: electrocardiogram

FEV1: forced expiratory volume in one second

FF: fluticasone furoate

FP: fluticasone propionate

FRP: female reproductive potential

FSH: follicle‐stimulating hormone

GINA: Global Initiative for Asthma

GP: general practitioner

HCP: healthcare practitioner

HRT: hormone replacement therapy

ICS: inhaled corticosteroid

LABA: long‐acting beta2‐agonist

LAMA: long‐acting muscarinic agonist

LTRA: leukotriene receptor antagonist

MAO: monoamine oxidase

NHANES: National Health and Nutrition Examination Survey

OCS: oral corticosteroid

PEF: peak expiratory flow

PM: afternoon

SABA: short‐acting beta2‐agonist

SAL: salmeterol

SIGN: Scottish Intercollegiate Guidelines Network

SOP: standard operating procedure

Data and analyses

Open in table viewer
Comparison 1. FF/VI 100/25 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

329

Mean Difference (Fixed, 95% CI)

0.3 [0.14, 0.46]

Analysis 1.1

Comparison 1 FF/VI 100/25 versus placebo, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

2 Exacerbations Show forest plot

2

161

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.2

Comparison 1 FF/VI 100/25 versus placebo, Outcome 2 Exacerbations.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 2 Exacerbations.

3 Serious adverse events Show forest plot

5

721

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.3

Comparison 1 FF/VI 100/25 versus placebo, Outcome 3 Serious adverse events.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 3 Serious adverse events.

4 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.17 [0.09, 0.26]

Analysis 1.4

Comparison 1 FF/VI 100/25 versus placebo, Outcome 4 FEV1 Litres.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 4 FEV1 Litres.

5 PEFR AM L/min (change from baseline at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.3 [26.59, 40.01]

Analysis 1.5

Comparison 1 FF/VI 100/25 versus placebo, Outcome 5 PEFR AM L/min (change from baseline at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 5 PEFR AM L/min (change from baseline at 12 wk).

6 PEFR PM L/min (change from baseline at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

28.2 [21.67, 34.73]

Analysis 1.6

Comparison 1 FF/VI 100/25 versus placebo, Outcome 6 PEFR PM L/min (change from baseline at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 6 PEFR PM L/min (change from baseline at 12 wk).

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

339

Mean Difference (Fixed, 95% CI)

1.9 [1.22, 2.58]

Analysis 1.7

Comparison 1 FF/VI 100/25 versus placebo, Outcome 7 Change in asthma symptoms (measured by ACT).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 7 Change in asthma symptoms (measured by ACT).

Open in table viewer
Comparison 2. FF/VI 100/25 versus same dose of FF

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.15 [‐0.00, 0.30]

Analysis 2.1

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

2 Exacerbations Show forest plot

2

2425

Odds Ratio (M‐H, Fixed, 95% CI)

1.38 [0.86, 2.22]

Analysis 2.2

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 2 Exacerbations.

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 2 Exacerbations.

3 Serious adverse events Show forest plot

5

1258

Odds Ratio (M‐H, Fixed, 95% CI)

1.61 [0.42, 6.17]

Analysis 2.3

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 3 Serious adverse events.

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 3 Serious adverse events.

4 Trough FEV1 (L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.08 [0.02, 0.14]

Analysis 2.4

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 4 Trough FEV1 (L).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 4 Trough FEV1 (L).

5 PEFR AM (change from baseline at 12 wk) Show forest plot

2

Mean Difference (Fixed, 95% CI)

20.29 [15.72, 24.85]

Analysis 2.5

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 5 PEFR AM (change from baseline at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 5 PEFR AM (change from baseline at 12 wk).

6 PEFR PM (change from baseline at 12 wk) Show forest plot

2

Mean Difference (Fixed, 95% CI)

18.52 [14.03, 23.01]

Analysis 2.6

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 6 PEFR PM (change from baseline at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 6 PEFR PM (change from baseline at 12 wk).

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.6 [‐0.04, 1.24]

Analysis 2.7

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Open in table viewer
Comparison 3. FF/VI 100/25 versus same dose VI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

1

53

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.1

Comparison 3 FF/VI 100/25 versus same dose VI, Outcome 1 Serious adverse events.

Comparison 3 FF/VI 100/25 versus same dose VI, Outcome 1 Serious adverse events.

Open in table viewer
Comparison 4. FF/VI 100/25 versus FP 500 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

1

301

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.10, 2.47]

Analysis 4.1

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 1 Exacerbations.

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 1 Exacerbations.

2 Serious adverse events Show forest plot

1

301

Odds Ratio (M‐H, Fixed, 95% CI)

0.20 [0.05, 0.80]

Analysis 4.2

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 2 Serious adverse events.

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 2 Serious adverse events.

Open in table viewer
Comparison 5. FF/VI 100/25 versus FPS 250/50 bd

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

677

Mean Difference (Fixed, 95% CI)

0.09 [‐0.03, 0.21]

Analysis 5.1

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 1 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 1 Change in quality of life (measured by AQLQ at 24 wk).

2 Exacerbations Show forest plot

1

806

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.05, 5.52]

Analysis 5.2

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 2 Exacerbations.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 2 Exacerbations.

3 Serious adverse events Show forest plot

1

806

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.21, 2.99]

Analysis 5.3

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 3 Serious adverse events.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 3 Serious adverse events.

4 FEV1 Show forest plot

1

Mean Difference (Fixed, 95% CI)

‐0.02 [‐0.07, 0.03]

Analysis 5.4

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 4 FEV1.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 4 FEV1.

5 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.24 [‐0.20, 0.68]

Analysis 5.5

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 5 Change in asthma symptoms (measured by ACT).

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 5 Change in asthma symptoms (measured by ACT).

Open in table viewer
Comparison 6. FF/VI 100/25 µg versus FF/VI 200/25 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

2

515

Odds Ratio (M‐H, Fixed, 95% CI)

2.02 [0.50, 8.19]

Analysis 6.1

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 1 Exacerbations.

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 1 Exacerbations.

2 Serious adverse events Show forest plot

2

515

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.18]

Analysis 6.2

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 2 Serious adverse events.

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 2 Serious adverse events.

Open in table viewer
Comparison 7. FF/VI 200/25 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

1

114

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 7.1

Comparison 7 FF/VI 200/25 versus placebo, Outcome 1 Exacerbations.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 1 Exacerbations.

2 Serious adverse events Show forest plot

1

114

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 7.2

Comparison 7 FF/VI 200/25 versus placebo, Outcome 2 Serious adverse events.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 2 Serious adverse events.

3 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.21 [0.13, 0.29]

Analysis 7.3

Comparison 7 FF/VI 200/25 versus placebo, Outcome 3 FEV1 Litres.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 3 FEV1 Litres.

4 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.9 [0.12, 1.68]

Analysis 7.4

Comparison 7 FF/VI 200/25 versus placebo, Outcome 4 Change in asthma symptoms (measured by ACT).

Comparison 7 FF/VI 200/25 versus placebo, Outcome 4 Change in asthma symptoms (measured by ACT).

Open in table viewer
Comparison 8. FF/VI 200/25 µg versus FP 500 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

2

606

Mean Difference (Fixed, 95% CI)

0.05 [‐0.08, 0.17]

Analysis 8.1

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

2 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.03 [‐0.15, 0.21]

Analysis 8.2

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

3 OLD***Health‐related quality of life Show forest plot

2

606

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.08, 0.17]

Analysis 8.3

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 3 OLD***Health‐related quality of life.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 3 OLD***Health‐related quality of life.

4 Exacerbations Show forest plot

2

611

Odds Ratio (M‐H, Fixed, 95% CI)

0.70 [0.22, 2.20]

Analysis 8.4

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 4 Exacerbations.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 4 Exacerbations.

5 Serious adverse events Show forest plot

3

1003

Odds Ratio (M‐H, Fixed, 95% CI)

0.61 [0.25, 1.49]

Analysis 8.5

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 5 Serious adverse events.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 5 Serious adverse events.

6 PEFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

28.6 [20.23, 36.97]

Analysis 8.6

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 6 PEFR.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 6 PEFR.

7 PEFR AM Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.0 [24.84, 41.16]

Analysis 8.7

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 7 PEFR AM.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 7 PEFR AM.

8 PEFR PM Show forest plot

1

Mean Difference (Fixed, 95% CI)

26.2 [18.04, 34.36]

Analysis 8.8

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 8 PEFR PM.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 8 PEFR PM.

9 % symptom‐free days Show forest plot

1

Mean Difference (Fixed, 95% CI)

4.8 [‐2.84, 12.44]

Analysis 8.9

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 9 % symptom‐free days.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 9 % symptom‐free days.

10 Change in asthma symptoms (measured by ACT) Show forest plot

1

332

Mean Difference (Fixed, 95% CI)

0.8 [0.01, 1.59]

Analysis 8.10

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 10 Change in asthma symptoms (measured by ACT).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 10 Change in asthma symptoms (measured by ACT).

Open in table viewer
Comparison 9. FF/VI 200/25 versus same dose of FF

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.08 [‐0.08, 0.24]

Analysis 9.1

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

2 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

307

Mean Difference (Fixed, 95% CI)

0.05 [‐0.14, 0.24]

Analysis 9.2

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

3 Serious adverse events Show forest plot

1

391

Odds Ratio (M‐H, Fixed, 95% CI)

6.06 [0.72, 50.84]

Analysis 9.3

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 3 Serious adverse events.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 3 Serious adverse events.

4 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.19 [0.10, 0.28]

Analysis 9.4

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 4 FEV1 Litres.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 4 FEV1 Litres.

5 PEFR AM Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.6 [25.41, 41.79]

Analysis 9.5

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 5 PEFR AM.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 5 PEFR AM.

6 PEFR PM Show forest plot

1

Mean Difference (Fixed, 95% CI)

30.7 [22.51, 38.89]

Analysis 9.6

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 6 PEFR PM.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 6 PEFR PM.

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

317

Mean Difference (Fixed, 95% CI)

0.3 [‐0.50, 1.10]

Analysis 9.7

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

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

Comparison 1 FF/VI 100/25 versus placebo, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).
Figuras y tablas -
Analysis 1.1

Comparison 1 FF/VI 100/25 versus placebo, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 2 Exacerbations.
Figuras y tablas -
Analysis 1.2

Comparison 1 FF/VI 100/25 versus placebo, Outcome 2 Exacerbations.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 1.3

Comparison 1 FF/VI 100/25 versus placebo, Outcome 3 Serious adverse events.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 4 FEV1 Litres.
Figuras y tablas -
Analysis 1.4

Comparison 1 FF/VI 100/25 versus placebo, Outcome 4 FEV1 Litres.

Comparison 1 FF/VI 100/25 versus placebo, Outcome 5 PEFR AM L/min (change from baseline at 12 wk).
Figuras y tablas -
Analysis 1.5

Comparison 1 FF/VI 100/25 versus placebo, Outcome 5 PEFR AM L/min (change from baseline at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 6 PEFR PM L/min (change from baseline at 12 wk).
Figuras y tablas -
Analysis 1.6

Comparison 1 FF/VI 100/25 versus placebo, Outcome 6 PEFR PM L/min (change from baseline at 12 wk).

Comparison 1 FF/VI 100/25 versus placebo, Outcome 7 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 1.7

Comparison 1 FF/VI 100/25 versus placebo, Outcome 7 Change in asthma symptoms (measured by ACT).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).
Figuras y tablas -
Analysis 2.1

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 2 Exacerbations.
Figuras y tablas -
Analysis 2.2

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 2 Exacerbations.

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 2.3

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 3 Serious adverse events.

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 4 Trough FEV1 (L).
Figuras y tablas -
Analysis 2.4

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 4 Trough FEV1 (L).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 5 PEFR AM (change from baseline at 12 wk).
Figuras y tablas -
Analysis 2.5

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 5 PEFR AM (change from baseline at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 6 PEFR PM (change from baseline at 12 wk).
Figuras y tablas -
Analysis 2.6

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 6 PEFR PM (change from baseline at 12 wk).

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 2.7

Comparison 2 FF/VI 100/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Comparison 3 FF/VI 100/25 versus same dose VI, Outcome 1 Serious adverse events.
Figuras y tablas -
Analysis 3.1

Comparison 3 FF/VI 100/25 versus same dose VI, Outcome 1 Serious adverse events.

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 1 Exacerbations.
Figuras y tablas -
Analysis 4.1

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 1 Exacerbations.

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 4.2

Comparison 4 FF/VI 100/25 versus FP 500 µg, Outcome 2 Serious adverse events.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 1 Change in quality of life (measured by AQLQ at 24 wk).
Figuras y tablas -
Analysis 5.1

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 1 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 2 Exacerbations.
Figuras y tablas -
Analysis 5.2

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 2 Exacerbations.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 5.3

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 3 Serious adverse events.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 4 FEV1.
Figuras y tablas -
Analysis 5.4

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 4 FEV1.

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 5 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 5.5

Comparison 5 FF/VI 100/25 versus FPS 250/50 bd, Outcome 5 Change in asthma symptoms (measured by ACT).

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 1 Exacerbations.
Figuras y tablas -
Analysis 6.1

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 1 Exacerbations.

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 6.2

Comparison 6 FF/VI 100/25 µg versus FF/VI 200/25 µg, Outcome 2 Serious adverse events.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 1 Exacerbations.
Figuras y tablas -
Analysis 7.1

Comparison 7 FF/VI 200/25 versus placebo, Outcome 1 Exacerbations.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 7.2

Comparison 7 FF/VI 200/25 versus placebo, Outcome 2 Serious adverse events.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 3 FEV1 Litres.
Figuras y tablas -
Analysis 7.3

Comparison 7 FF/VI 200/25 versus placebo, Outcome 3 FEV1 Litres.

Comparison 7 FF/VI 200/25 versus placebo, Outcome 4 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 7.4

Comparison 7 FF/VI 200/25 versus placebo, Outcome 4 Change in asthma symptoms (measured by ACT).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).
Figuras y tablas -
Analysis 8.1

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).
Figuras y tablas -
Analysis 8.2

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 3 OLD***Health‐related quality of life.
Figuras y tablas -
Analysis 8.3

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 3 OLD***Health‐related quality of life.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 4 Exacerbations.
Figuras y tablas -
Analysis 8.4

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 4 Exacerbations.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 5 Serious adverse events.
Figuras y tablas -
Analysis 8.5

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 5 Serious adverse events.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 6 PEFR.
Figuras y tablas -
Analysis 8.6

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 6 PEFR.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 7 PEFR AM.
Figuras y tablas -
Analysis 8.7

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 7 PEFR AM.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 8 PEFR PM.
Figuras y tablas -
Analysis 8.8

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 8 PEFR PM.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 9 % symptom‐free days.
Figuras y tablas -
Analysis 8.9

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 9 % symptom‐free days.

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 10 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 8.10

Comparison 8 FF/VI 200/25 µg versus FP 500 µg, Outcome 10 Change in asthma symptoms (measured by ACT).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).
Figuras y tablas -
Analysis 9.1

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 1 Change in quality of life (measured by AQLQ at 12 wk).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).
Figuras y tablas -
Analysis 9.2

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 2 Change in quality of life (measured by AQLQ at 24 wk).

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 9.3

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 3 Serious adverse events.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 4 FEV1 Litres.
Figuras y tablas -
Analysis 9.4

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 4 FEV1 Litres.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 5 PEFR AM.
Figuras y tablas -
Analysis 9.5

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 5 PEFR AM.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 6 PEFR PM.
Figuras y tablas -
Analysis 9.6

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 6 PEFR PM.

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).
Figuras y tablas -
Analysis 9.7

Comparison 9 FF/VI 200/25 versus same dose of FF, Outcome 7 Change in asthma symptoms (measured by ACT).

Summary of findings for the main comparison. Vilanterol and fluticasone furoate compared with placebo for asthma

VI and FF compared with placebo for asthma

Patient or population: people with asthma

Settings: community

Intervention: VI and FF

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Placebo

VI and FF

Health‐related quality of life

0.61
(SE 0.061),
n = 149

0.91 (SE 0.055),

n = 180

MD 0.30, 95% CI 0.14 to 0.46

Bleecker 2012 (N = 609 participants, 515 completed study) compared VI/FF 100/25 mcg vs placebo in respect of health‐related quality of life and indicated a significant advantage for VI/FF 100/25 mcg

Moderatea

Asthma exacerbation

Not estimable

Only 2 studies (Allen 2013 and Kempsford 2012a) compared VI/FF 100/25 mcg vs placebo in respect of exacerbations; both studies reported no exacerbations in either treatment arm

Very lowb

Serious adverse events

Not estimable

Five trials (Allen 2013; Bleecker 2012; Kempsford 2012a; Oliver 2012; Oliver 2013) made this same comparison in relation to serious adverse events; all 5 reported no serious adverse events in VI/FF100/25 mcg or placebo arms

Very lowb

FEV1

0.196 L
(SE 0.0310),

n = 193

0.368 L

(SE 0.0304),

n = 200

MD 0.17 L, 95% CI 0.09 to 0.26

Significant difference in favour of VI/FF 100/25 mcg vs placebo with respect to mean change in trough FEV1 (pre‐bronchodilator and pre‐dose) from baseline to week 12 in 1 trial (Bleecker 2012) (N = 609 participants, 515 completed study) (MD 0.17 L, 95% CI 0.09 to 0.26), and a similar effect was found in a small cross‐over trial (Kempsford 2012a) over a 2‐week period in the morning (MD 0.377 L, 90% CI 0.293 to 0.462) and in the evening (MD 0.422 L, 90% CI 0.337 to 0.507)

Moderatec

Peak expiratory flow

‐0.4 L/min (SE 2.42),
n = 203

32.9 L/min

(SE 2.42),

n = 201

MD 33.30 L/min,

95% CI 26.59 to 40.01

Bleecker 2012 (N = 609 participants, 515 completed study) compared VI/FF 100/25 mcg vs placebo as mean change from baseline in daily morning (AM) PEF averaged over 12‐week treatment period; researchers noted a significant difference in favour of VI/FF 100/25 mcg (MD 33.30 L/min, 95% CI 26.59 to 40.01). The same trial showed a similar advantage in favour of VI/FF 100/25 mcg vs placebo in the evening over this period (28.20 L/min, 95% CI 21.67 to 34.73). A small cross‐over trial (Kempsford 2012a) produced a similar effect in favour of VI/FF 100/25 mcg vs placebo over a 2‐week period in the morning (MD 44.0 L/min, 90% CI 31.2 to 56.9) and in the evening (MD 69.0 L/min, 90% CI 55.9 to 82.1)

Moderatec

Asthma symptoms

14.6

(SE 2.15),

n = 202

32.5 (SE 2.14),

n = 201

MD 17.90, 95% CI 11.95 to 23.85

Only 1 trial (Bleecker 2012) (N = 609 participants, 515 completed study) made VI/FF vs placebo comparison with respect to asthma symptoms, indicating a clear advantage for VI/FF 100/25 mcg

Moderatea

Adverse events

Not estimable

Several trials reported a range of adverse events for which overall aggregation was not possible. These are tabulated in Table 8

Moderated

*The basis for the assumed risk (e.g. 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)
AM: morning; CI: confidence interval; FEV1: forced expiratory volume in one second; FF: fluticasone furoate; GRADE: Grades of Recommendation, Assessment, Development and Evaluation Working Group; MD: mean difference; OR: odds ratio; PEF: peak expiratory flow; PM: afternoon; RR: risk ratio; SE: standard error; VI: vilanterol

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

aPoint deducted to reflect that these data were derived from only one trial

bInvestigators reported no events in either arm of these trials

cPoint deducted to reflect that data contributing to the main result (MD 0.17 L, 95% CI 0.09 to 0.26) were obtained from only one trial

dPoint deducted, as we were unable to combine data on this outcome; results are presented in a separate table

Figuras y tablas -
Summary of findings for the main comparison. Vilanterol and fluticasone furoate compared with placebo for asthma
Table 8. Adverse events

Study

FF/VI

100/25 mcg

FF 100 mcg

FF 200 mcg

FF/VI

200/25 mcg

VI 25 mcg

FP/SAL

250/50 mcg twice‐daily

FP 500 mcg

Prednisolone

10 mg

Placebo

Allen 2013

6 weeks' duration. On‐treatment AEs

23/56 (41.00%)

21/56 (38.00%)

5/15 (33.00%)

16/58 (28.00%)

Bateman 2014

≥24 to 78 weeks' duration. On‐treatment AEs

636/1009 (63.00%)

652/1010 (65.00%)

Bernstein 2014

12 weeks' duration

54/346 (15.61%)

67/347 (19.31%)

52/346 (15.03%)

‐‐

Bleecker 2012

12 weeks' duration

29/201 (14.43%)

20/205 (9.76%)

22/203 (10.84%)

Busse 2013

52 weeks' duration. On‐treatment AEs

139/201 (69.15%)

134/202 (66.34%)

73/100 (73.00%)

Lee 2014

Cross‐over trial. 3 of 7 treatments (2 weeks) separated by 12 to 14‐day washout periods

43/172 (25%)

25/187 (13%)

Lin 2013

12 weeks' duration. Any AE

40/155 (26.00%)

41/154 (27.00%)

Kempsford 2012

Cross‐over trial. Each period lasted 14 days with a 14 to 21‐day washout period between periods

11/24 (45.83%) AM

12/25 (48.00%) PM

8/23 (34.78%)

NCT01134042

24 weeks' duration

66/194 (34.02%)

62/197 (31.47%)

73/195 (37.44%)

NCT01453023

Cross‐over trial. 11 weeks (for a single period)

4/25 (16.00%)

1/25 (4.00%)

Oliver 2012

Cross‐over trial. 28 days for each period

11/51 (21.57%)

18/51 (35.29%)

15/51 (29.41%)

Oliver 2013

Cross‐over trial. 21 days

20/27 (74.07%)

19/27 (70.37%)

22/26 (84.62%)

19/27 (70.37%)

Woodcock 2013

24 weeks' duration

110/403 (27.30%)

106/403 (26.30%)

Fractions shown in the table indicate the proportions of people who suffered one or more adverse events of any cause in each treatment arm

AE: adverse event; F: fluticasone furoate; FP: fluticasone propionate; SAL: salmeterol; VI: vilanterol

Figuras y tablas -
Table 8. Adverse events
Table 1. Summary of study characteristics

Study

Duration (weeks)

Severity at baseline

Inclusion criteria

Adverse events

Allen 2013

6

Reversibility > 12%

 

FEV1 > 50% of predicted

Adults

Comply with treatment

 

Clinical diagnosis of asthma for ≥ 12 weeks

Cortisol urinary excretion, serum AUC and trough

Bateman 2014

24 to 78

Reversibility > 12%

 

FEV1 > 50% to 90% of predicted

Adults

Using ICS

 

History of ≥ 1 exacerbation requiring hospitalisation or steroids in the past year

None

Bernstein 2014

12

Reversibility > 12%

FEV1 50% to 80% of predicted

ICS for > 12 weeks before study

 

> 12 years of age

Yes, not clear

Bleecker 2012

12

Pre‐bronchodilator FEV1 40% to 90% of predicted normal

Reversibility FEV1 ≥ 12%

ICS for 12 weeks before study

 

> 12 years of age

Details not stated

Busse 2013

52

Pre‐bronchodilator FEV1 40% to 90% of predicted normal

Reversibility FEV1 ≥ 12%

 

Adults

Clinical diagnosis of asthma

 

ICS at high dose

Details not stated

Hojo 2015

4

ACT suggesting poor control and FEV1 mean  70% (SD 11%)

Asthma ≥ 20 years of age

No, conference abstract only

Lee 2014

4

Pre‐bronchodilator FEV1 40% to 80% of predicted

Demonstrated reversibility by ≥ 12%

Need for regular controller therapy for minimum of 8 weeks

Stable dose of ICS for ≥ 4 weeks

≥ 18 years of age

Diagnosis of asthma for ≥ 6 months

 

No

Lin 2013

12

Reversibility of disease: demonstrated ≥ 12% and

FEV1 40% to 90%

 

ICS, with or without LABA, for ≥ 12 weeks

Clinical diagnosis of asthma for 12 weeks

 

Adults

No

Kempsford 2012

6 to 8

Pre‐bronchodilator FEV1 ≥ 60% of predicted.

 

18 and 70 years of age inclusive

Using an ICS, with or without a SABA, for ≥ 12 weeks before screening

Participants who are current non‐smokers, who have not used inhaled tobacco products in the 12‐month period preceding screening visit

Body weight ≥ 50 kg and BMI within the range 19.0 to 29.9 kg/m2

Yes, details not stated

NCT01134042

24

Pre‐bronchodilator FEV1 40% to 90% of predicted

Reversibility FEV1 ≥ 12%

 

Current asthma therapy that includes an ICS for ≥ 12 weeks before first visit

Adults

Cortisol, ECG, mouth swabs, various blood parameters

NCT01453023

14

Mild to moderate (GINA)

Stable asthma therapy (FP, total daily dose ≤ 400 mcg or equivalent) and SABA inhaler for ≥ 4 weeks before screening

5 to 12 years of age

Clinical diagnosis of asthma 6 months before

Controlled asthma (Childhood ACT > 19)

Not stated

Oliver 2012

8

Pre‐bronchodilator FEV1 > 70% of predicted at screening

 

Methacholine challenge PC20 < 8 mg/mL at screening

 

 

Adults

Stable asthma therapy (FP, total daily dose ≤ 400 mcg or equivalent) and SABA inhaler for ≥ 4 weeks before screening

BMI within the range 18.5 to 35.0 kg/m2

 

 

Not stated

Oliver 2013

3 with 3 weeks' washout

Pre‐bronchodilator FEV1 > 70% of predicted at screening

 

Methacholine challenge PC20 < 8 mg/mL at screening

 

Stable asthma therapy (FP, total daily dose ≤ 400 mcg or equivalent) and SABA inhaler for ≥ 4 weeks before screening

BMI within the range 18.5 to 35.0 kg/m2

Adults

Not stated

Woodcock 2013

24

Reversibility ≥ 12% and 200 mL within 10 to 40 minutes following 2 to 4 inhalations of albuterol

FEV1 40% to 85% predicted normal

Currently using ICS therapy

Clinical diagnosis of asthma

Adults  

Not stated

ACT: Asthma Control Test

AUC: area under the curve

BMI: body mass index

ECG: electrocardiogram

FEV1: forced expiratory volume in one second

FP: fluticasone propionate

GINA: Global Initiative for Asthma

ICS: inhaled corticosteroid

LABA: long‐acting beta2‐agonist

PC20: provocative concentration of methacholine estimated to result in a 20% reduction in FEV1

SABA: short‐acting beta2‐agonist

Figuras y tablas -
Table 1. Summary of study characteristics
Table 2. Health‐related quality of life

Study

score (change from baseline)

FF/VI 100/25 mcg

Mean (SE), N

FF 100 mcg

Mean (SE), N

FF 200 mcg

Mean (SE), N

FF/VI 200/25 mcg

Mean (SE), N

FP/SAL 250/50 mcg twice‐daily

FP 500 mcg

Placebo

MD (95% CI)

Bleecker 2012

AQLQ change from baseline at 12 weeks

0.91 (0.055),

n = 180

0.76 (0.055),

n = 184

0.61 (0.061),

n = 149

0.15 (0.00 to 0.30), 0.30 (0.14 to 0.46), 0.15 (‐0.01 to 0.31)

Lin 2013

AQLQ change from baseline at 12 weeks

0.80 (0.069),

n = 140

0.69 (0.074),

n = 123

0.12 (‐0.08 to 0.32)

NCT01134042

AQLQ change from baseline at 12 weeks

0.66 (0.061),

n = 154

0.74 (0.056),

n = 180

0.74 (0.059),

n = 163

‐0.08 (‐0.24 to 0.08), ‐0.08 (‐0.25 to 0.09), 0.00 (‐0.16 to 0.16)

NCT01134042

AQLQ change from baseline at 24 weeks

0.88 (0.071),

n = 140

0.93 (0.065),

n = 167

0.90 (0.068),

n = 156

‐0.05 (‐0.24 to 0.14), ‐0.02 (‐0.21 to 0.17), 0.03 (‐0.15 to 0.21)

Woodcock 2013

AQLQ change from baseline at 168 days

0.46 (0.043),

n = 342

0.37 (0.043), n = 335

0.09 (‐0.03 to 0.21)

Woodcock 2013

EQ‐5D change from baseline at 168 days

5.5 (0.60),

n = 343

4.1 (0.60), n = 349

1.4 (‐0.3 to 3.0)

AQLQ: asthma quality of life questionnaire; CI: confidence interval; EQ‐5D: EuroQuality of Life‐5D questionnaire; FF: fluticasone furoate; FP: fluticasone propionate; MD: mean difference; SAL: salmeterol; SE: standard error; VI: vilanterol

Figuras y tablas -
Table 2. Health‐related quality of life
Table 3. Asthma exacerbation

Study

FF/VI

100/25 mcg

FF 100 mcg

FF

200 mcg

FF/VI

200/25 mcg

FP/SAL

250/50 mcg twice‐daily

FP

500 mcg

Prednisolone

10 mg

Placebo

Allen 2013a

6 weeks' duration

0/56 (0.00%)

0/56 (0.00%)

0/15 (0.00%)

0/58 (0.00%)

Bateman 2014

≥ 24 to 78 weeks' duration

Time to first severe exacerbation (HR 0.80, 95% CI 0.64 to 0.99). Annualised rate of severe exacerbation 25% reduction (95% CI 5% to 40%)

154/1009 (15.26%)

186/1010 (18.42%)

Busse 2013

52 weeks' duration

3/201 (1.49%)

6/202 (2.97%)

3/100 (3.00%)

Lin 2013

12 weeks' duration

1/155 (0.65%)

3/154 (1.95%)

Kempsford 2012

Cross‐over trial. Each period lasted 14 days with a 14 to 21‐day washout period between periods

0/24 (0.00%) AM

0/25 (0.00%) PM

0/23 (0.00%)

Woodcock 2013b

24 weeks' duration

1/403 (0.25%)

2/403 (0.50%)

aOne participant in the FF/VI 100/25 mcg group experienced a severe asthma exacerbation concurrent with sinusitis and was withdrawn owing to lack of efficacy. The participant did not require hospitalisation, and the event, which was not classified as an AE, resolved following treatment with prednisone

bThe incidence of asthma exacerbations was low, and no difference was noted between groups (3% vs 2% on FP/SAL vs FF/VI, respectively (on‐treatment events)). Eight (2%) participants in the FF/VI group and seven (2%) in the FP/SAL group withdrew because of exacerbation. One patient in the FF/VI group and two in the FP/SAL group were hospitalised because of exacerbation

AM: morning; CI: confidence interval; FF: fluticasone furoate; FP: fluticasone propionate; HR: hazard ratio; PM: afternoon; SAL: salmeterol; VI: vilanterol

Figuras y tablas -
Table 3. Asthma exacerbation
Table 4. Serious adverse events

Study

FF/VI

100/25 mcg

FF 100 mcg

FF

200 mcg

FF/VI

200/25 mcg

VI 25 mcg

FP/SAL

250/50 mcg twice‐daily

FP 500 mcg

Prednisolone

10 mg

Placebo

Allen 2013

6 weeks' duration. Post‐treatment period SAEs

0/56 (0.00%)

0/56 (0.00%)

0/15 (0.00%)

0/58 (0.00%)

Bateman 2014

≥ 24 to 78 weeks' duration. On‐treatment SAEs

41/1009 (4.06%)

29/1010 (2.87%)

Bernstein 2014

12 weeks' duration

4/346 (1.16%)

3/347 (0.86%)

1/346 (0.29%)

Bleecker 2012

12 weeks' duration

0/201 (0.00%)

1/205 (0.49%)

0/203 (0.00%)

Busse 2013

52 weeks' duration. On‐treatment SAEs

3/201 (1.49%)

1/202 (0.50%)

7/100 (7.00%)

Lee 2014

Cross‐over trial. Three of 7 treatments (2 weeks) separated by 12 to 14‐day washout periods

1/172

(0.006%)

0/187

(0%)

Lin 2013

12 weeks' duration

1/155 (0.65%)

2/154 (1.30%)

Kempsford 2012

Cross‐over trial. Each period lasted 14 days with a 14 to 21‐day washout period

0/24 (0.00%) AM.

0/25 (0.00%) PM.

0/23 (0.00%)

NCT01134042

24 weeks' duration

1/194 (0.52%)

6/197 (3.05%)

2/195 (1.03%)

NCT01453023

Cross‐over trial. 11 weeks (for a single period)

0/25 (0.00%)

0/25 (0.00%)

Oliver 2012a

Cross‐over trial. 28 days for each period

0/51 (0.00%)

0/51 (0.00%)

0/51 (0.00%)

Oliver 2013

Cross‐over trial. 21 days

0/27 (0.00%)

0/27 (0.00%)

0/26 (0.00%)

0/27 (0.00%)

Woodcock 2013

24 weeks' duration

4/403 (0.99%)

5/403 (1.24%)

aThe main paper reports that 1 of the 52 withdrew during the study owing to an SAE, which occurred 4 days after the last dose in the FF 100 treatment period. This participant was provisionally diagnosed with moderate (grade 2) Still’s disease. Six weeks later, the participant was hospitalised. A diagnosis of histiocytic necrotising lymphadenitis (Kikuchi’s disease) was made on the basis of histology of an excised lymph node. Tapered prednisolone treatment, initiated at 60 mg per day, has been successful

FF: fluticasone furoate; FP: fluticasone propionate; SAE: serious adverse event; SAL: salmeterol; VI: vilanterol

Figuras y tablas -
Table 4. Serious adverse events
Table 5. Forced expiratory flow in one second (FEV1)

Study

measure

time point/

duration

FF/VI

100/25 mcg

Mean (SE), N, of MD (95% CI)

FF 100 mcg

Mean (SE), N

FF 200 mcg

Mean (SE), N

FF/VI 200/25 mcg

Mean (SE), N

VI 25 mcg

Mean (SE), N

FP/SAL250/50 mcg twice‐daily

Mean (SE), N

FP 500 mcg

Mean (SE), N

Placebo

Mean (SE), N

MD (95% CI, unless otherwise stated)

Bernstein 2014

Trough FEV1

At 0 to 12 weeks

Change in baseline trough FEV1 from baseline to week 12

0.441 L (0.022)

0.365 L (0.022)

0.457 L (0.022)

‐‐

Bleecker 2012

Trough FEV1

At 0 to 12 weeks

Mean change in trough FEV1 (pre‐bronchodilator and pre‐dose) from baseline to

week 12

0.368 L (0.0304),

n = 200

0.332 L (0.0302),

n = 203

0.196 L (0.0310),

n = 193

0.04 L (‐0.05 to 0.12) 0.17 L (0.09 to 0.26) 0.14 L (0.05 to 0.22)

Lee 2014

Trough FEV1 combining all treatment periods

At 0 to 2 weeks

3 of 7 treatments (2 weeks) separated by 12 to 14‐day washout periods

0.200 L,

n = 158

0.087 L,

n = 158

Lin 2013

12 weeks' duration

Adjusted treatment difference 0.108 L (0.040 to 0.176)

Kempsford 2012

Weighted mean FEV1 over the day

At day 14

Weighted mean FEV1, over 0 to 24 hours post dose at day 14

Cross‐over trial. Each period lasted 14 days with a 14 to 21‐day washout period

AM dose: 3.188 L
(3.112 to 3.265), n = 24

PM dose: 3.233 L
(3.159 to 3.306), n = 25

2.811 L
(2.729 to 2.893),

n = 20

AM vs placebo 0.377 L (90% CI 0.293 to 0.462)

PM vs placebo 0.422 L (90% CI 0.337 to 0.507)

AM vs PM ‐0.44 L

(90% CI ‐0.125 to 0.36)

(Kempsford 2012)

Day 14 pre‐treatment (trough) AM FEV1

At day 14

AM dose: 3.191 L
(3.087 to 3.295), n = 24

PM dose: 3.285 L
(3.187 to 3.383),

n = 25

2.788 L
(2.684 to 2.892),

n = 22

AM vs placebo 0.403 L (90% CI 0.272 to 0.533)

PM vs placebo 0.496 L (90% CI 0.369 to 0.624)

AM vs PM ‐0.094 L (90% CI ‐0.221 to 0.034)

(Kempsford 2012)

Day 14 pre‐treatment (trough) PM FEV1

At day 14

AM dose: 3.153 L
(3.049 to 3.258), n = 24

PM dose: 3.188 L
(3.088 to 3.288), n = 25

2.879 L
(2.775 to 2.982),

n = 23

AM vs placebo 0.275 L (90% CI 0.169 to 0.380)

PM vs placebo 0.309 L (90% CI 0.205 to 0.413)

AM vs PM ‐0.034

(90% CI ‐0.138 to 0.070)

NCT01134042

Change in baseline trough FEV1

At 24 weeks

Change from baseline in clinic visit trough (pre‐bronchodilator and pre‐dose) FEV1 at end of 24‐week treatment period

0.201 L (0.0303),

n = 186

0.394 L (0.0302),

n = 187

0.183 L (0.0300),

n = 190

‐0.19 L (‐0.28 to ‐0.11) 0.02 L (‐0.06 to 0.10) 0.21 L (0.13 to 0.29)

(NCT01134042)

Change from baseline in weighted mean serial FEV1 over 24 hours

At 24 weeks

Change from baseline in weighted mean serial FEV1 over 0 to 24 hours post dose at week 24

0.328 L (0.0493),

n = 83

0.464 L (0.0470),

n = 89

0.258 L (0.0483),

n = 86

‐0.14 L (‐0.27 to ‐0.00) 0.07 L (‐0.07 to 0.21) 0.21 L (0.07, 0.34)

Oliver 2012

23 hours post challenge

At day 29

Cross‐over trial ‐ 28 days for each period

Weighted mean change from baseline in FEV1 between 0 and 2 hours following 22 to 23‐hour post‐treatment allergen challenge at day 29 of each treatment period

‐0.227 L (0.0550),

n = 46

‐0.210 L (0.0549),

n = 49

‐0.372 L (0.0557),

n = 45

FF vs placebo 0.162 L (0.087 to 0.237)

FF/VI vs placebo 0.145 L (0.069 to 0.222)

FF/VI vs FF ‐0.017 L (‐0.091 to 0.057)

(Oliver 2012)

Decrease from baseline 23 hours post challenge

At day 29

Maximum % decrease from baseline FEV1 between 0 and 2 hours following 22 to 23‐hour post‐treatment allergen challenge at day 29 of each treatment period (time frame: baseline and at day 29 of each treatment period (up to study day 197))

‐13.206% (2.0491),

n = 46

‐14.040% (2.0435),

n = 49

‐24.991% (2.0736),

n = 45

FF vs placebo 10.951% (8.053 to 13.848)
FF/VI vs placebo 11.785%

(8.849 to 14.721)
FF/VI vs FF 0.834% (‐2.010 to 3.678)

(Oliver 2012)

Change from baseline FEV1

23 hours post challenge

Minimum FEV1 absolute change from baseline between 0 and 2 hours following 22 to 23‐hour post‐treatment allergen challenge at day 29 of each treatment period

‐0.478 L (0.0767),

n = 46

‐0.479 L (0.0765),

n = 49

‐0.809 L (0.0775),

n = 45

FF vs placebo 0.330 L (0.232 to 0.429)
FF/VI vs placebo 0.331 L (0.231 to 0.43)
FF/VI vs FF 0.001 L (‐0.096 to 0.097)

Oliver 2013

Change from baseline 4 to 10 hours post challenge

At day 21

Cross‐over trial ‐ 21 days

LAR: absolute change from baseline in minimum FEV1 between 4 and 10 hours following 1‐hour post‐treatment allergen challenge at day 21 of each treatment period

‐0.216 L
(‐0.343 to ‐0.088),

n = 26

‐0.188 L
(‐0.315 to ‐0.061),

n = 27

‐0.536 L
(‐0.676 to ‐0.396),

n = 22

‐0.731 L
(‐0.878 to ‐0.584),

n = 20

(Oliver 2013)

Change from baseline 4 to 10 hours post challenge

At day 21

LAR: absolute change from baseline in weighted mean FEV1 between 4 and 10 hours following 1‐hour post‐treatment allergen challenge at day 21 of each treatment period

0.018 L
(‐0.089 to 0.125), n = 26

0.018 L
(‐0.089 to 0.124),

n = 27

‐0.298 L
(‐0.415 to ‐0.181),

n = 22

‐0.466 L
(‐0.589 to ‐0.343),

n = 20

Woodcock 2013

Change from baseline trough FEV1

At day 168

24 weeks' duration

0.281 L (0.0191),

n = 397

0.300 L (0.0193),

n = 389

‐0.019 L (‐0.073 to 0.034)

AM: morning; CI: confidence interval; FEV1: forced expiratory volume in one second; FF: fluticasone furoate; FP: fluticasone propionate; h: hour; LAR: late asthmatic response; MD: mean difference; PM: afternoon; SAL: salmeterol; SE: standard error; VI: vilanterol

Figuras y tablas -
Table 5. Forced expiratory flow in one second (FEV1)
Table 6. Peak expiratory flow

Study

Duration (weeks)

Measure of PEF

FF/VI 100/25 mcg

Mean (SD, unless otherwise stated), N

FF 100 mcg

Mean (SD, unless otherwise stated), N

FF 200 mcg

Mean (SE), N

FF/VI 200/25 mcg

Mean (SE ), N

FP 500 mcg

Mean (SE), N

Placebo

Mean (SE, unless otherwise stated), N

MD (95% CI, unless otherwise stated)

Bernstein 2014

12

Change from baseline, AM

Change from baseline in AM PEF Averaged over 12‐week treatment period

44.3 L/min (2.25)

19.1 L/min (2.25)

47.7 L/min (2.25)

25.20 L/min (18.96 to 31.44), 100/25 vs 100 FF

12

Change from baseline, PM

Change from baseline in AM PEF Averaged over 12‐week treatment period

39.7 L/min (2.24)

15.5 L/min (2.24)

41.7 L/min (2.24)

24.20 L/min (17.99 to 30.41), 100/25 vs 100 FF

Bleecker 2012

12

Change from baseline, PM

Mean change from baseline in daily PM PEF averaged over 12‐week treatment period

26.4 L/min

(SE 2.35), n = 201

14.1 L/min

(SE 2.34), n = 204

‐1.8 L/min (2.36),

n = 202

12.30 L/min (5.80 to 18.80), 28.20 L/min (21.67 to 34.73), 15.90 L/min (9.39 to 22.41)

Hojo 2015

4

Change from baseline, AM

Only 1 (FF/VI) condition reported. Trial reported as conference abstract with limited information

Lee 2014

Baseline to day 15

Least squares mean change calculated from baseline to day 15

Least squares mean change in last 7 days, mean PEF

24.1 (2.46) AM

21.4 (2.58) PM

n = 172

‐2.9 (2.44) AM

‐5.2 (2.51) PM

n = 187

Lin 2013

12

12 weeks' duration.

39.1 L/min (3.01), n = 155

10.5 L/min (3.03), n = 154

Adjusted treatment difference 28.5 L/min (20.1 to 36.9)

Kempsford 2012

12 days

Pre‐treatment

Pre‐treatment PEF at days 1 to 12

Cross‐over trial. Each period lasted 14 days with a 14 to 21‐day washout period

AM dose:

510.4 L/min
(95% CI 492.9 to 527.8), n = 24

PM dose:

535.3 L/min
(95%CI 518.1 to 552.5), n = 25

466.3 L/min
(95% CI 448.8 to 483.9), n = 24

AM vs placebo 44.0 L/min (90% CI 31.2 to 56.9)

PM vs placebo 69.0 L/min (90% CI 55.9 to 82.1)

AM vs PM ‐25.0 L/min (90% CI ‐37.9 to ‐12.0)

12 days

Pre‐treatment

Pre‐treatment PEF (PM) at days 1 to 12

AM dose:

517.6 L/min
(95% CI 503.0 to 532.2), n = 24

PM dose:

521.4 L/min
(95% CI 507.1 to 535.7), n = 26

453.2 L/min
(95% CI 438.5 to 467.9), n = 24

AM vs placebo 64.4 L/min (90% CI 52.9 to 76.0)

PM vs placebo 68.2 L/min (90% CI 56.5 to 79.8)

AM vs PM ‐3.7 L/min (90% CI ‐15.2 to 7.7)

NCT01134042

24

Change from baseline, AM

4 weeks

Mean change from baseline in daily trough (AM) PEF averaged over 24‐week treatment period

18.2 L/min (2.97),

n = 193

51.8L/min (2.94), n = 197

18.8L/min (2.95), n = 195

‐33.60 L/min (‐41.79 to, ‐25.41), ‐0.60 L/min (‐8.80 to 7.60), 33.00 L/min (24.84 to 41.16)

24

Change from baseline, PM

Mean change from baseline in daily trough (PM) PEF averaged over 24‐week treatment period

9.1 L/min (2.98),

n = 192

39.8 L/min (2.93), n = 197

13.6 L/min (2.96), n = 194

‐30.70 L/min (‐38.89 to ‐22.51), ‐4.50 L/min (‐12.73 to 3.73), 26.20 L/min (18.04 to 34.36)

AM: morning; CI: confidence interval; FF: fluticasone furoate; PEF: peak expiratory flow; PM: evening; SD: standard deviation; SE: standard error; VI: vilanterol

Figuras y tablas -
Table 6. Peak expiratory flow
Table 7. Asthma symptoms

Study

Measure

FF/VI 100/25 mcg

Mean (SE)

FF 100 mcg

Mean (SE)

FF 200 mcg

Mean (SE)

FF/VI 200/25 mcg

Mean (SE)

FP/SAL250/50 mcg twice‐daily

Mean (SE)

FP 500 mcg

Mean (SE)

Placebo

Mean (SE)

MD (95% CI)

Bateman 2014

≥ 24 to 78 weeks' duration

Responder analysis results: ORs for FF/VI vs FF at week 12 (1.49, 95% CI 1.20 to 1.84), week 36 (1.49, 95% CI 1.21 to 1.83) and at endpoint (1.50, 95% CI 1.23 to 1.82)

ACQ7 mean difference and responder analysis

NR

NR

Bernstein 2014

Change from baseline in percentage of symptom‐free 24‐hour periods during 12‐week treatment

Change from baseline % symptom‐free days

27.2 (1.74)

n = 345

19.4 (1.74)

n = 346

29.0 (1.74)

n = 346

‐‐

Bleecker 2012

Change from baseline in % of symptom‐free 24‐hour periods during 12‐week treatment period

Change from baseline % symptom‐free days

32.5 (2.14),

n = 201

20.4 (2.13),

n = 204

14.6 (2.15), n = 202

12.10 (6.18 to 18.02), 17.90 (11.95 to 23.85), 5.80 (‐0.13 to 11.73)

Hojo 2015

Trial reported as conference abstract with limited information

Change from baseline ACT score

Lee 2014

LS mean change in symptom‐free days during 2‐week treatment period

LS mean change in symptom‐free days (SE)

7.3 (1.67) n = 172

5.8 (1.64) n = 187

Lin 2013

% of symptom‐free 24‐hour periods, weeks 1 to 12

% symptom‐free days

25.4 (2.74),

n = 155

20.6 (2.77),

n = 152

4.9 (‐2.8 to 12.5)

NCT01134042

Change from baseline in ACT scores at week 12

Change from baseline ACT score

3.9 (0.29), n = 164

4.8 (0.27), n = 183

3.9 (0.28), n = 169

‐0.90 (‐1.68 to ‐0.12), 0.00 (‐0.79 to 0.79), 0.90 (0.14 to 1.66)

(NCT01134042)

Change from baseline in ACT scores at week 24

Change from baseline ACT score

5.2 (0.30), n = 147

5.5 (0.28), n = 170

4.7 (0.29), n = 162

‐0.30 (‐1.10 to 0.50), 0.50 (‐0.32 to 1.32), 0.80 (0.01 to 1.59)

Woodcock 2013

Change from baseline in ACT scores at day 168 and at

24 weeks

Change from baseline ACT score

2.3 (0.16), n = 354

2.0 (0.16),

n = 348

0.2 (‐0.2 to 0.7)

ACT: asthma control test; CI: confidence interval; FF: fluticasone furoate; FP: fluticasone propionate; LS: least squares; MD: mean difference; NR: not reported; OR: odds ratio; SAL: salmeterol; SE: standard error; VI: vilanterol

Figuras y tablas -
Table 7. Asthma symptoms
Comparison 1. FF/VI 100/25 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

329

Mean Difference (Fixed, 95% CI)

0.3 [0.14, 0.46]

2 Exacerbations Show forest plot

2

161

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

5

721

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.17 [0.09, 0.26]

5 PEFR AM L/min (change from baseline at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.3 [26.59, 40.01]

6 PEFR PM L/min (change from baseline at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

28.2 [21.67, 34.73]

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

339

Mean Difference (Fixed, 95% CI)

1.9 [1.22, 2.58]

Figuras y tablas -
Comparison 1. FF/VI 100/25 versus placebo
Comparison 2. FF/VI 100/25 versus same dose of FF

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.15 [‐0.00, 0.30]

2 Exacerbations Show forest plot

2

2425

Odds Ratio (M‐H, Fixed, 95% CI)

1.38 [0.86, 2.22]

3 Serious adverse events Show forest plot

5

1258

Odds Ratio (M‐H, Fixed, 95% CI)

1.61 [0.42, 6.17]

4 Trough FEV1 (L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.08 [0.02, 0.14]

5 PEFR AM (change from baseline at 12 wk) Show forest plot

2

Mean Difference (Fixed, 95% CI)

20.29 [15.72, 24.85]

6 PEFR PM (change from baseline at 12 wk) Show forest plot

2

Mean Difference (Fixed, 95% CI)

18.52 [14.03, 23.01]

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.6 [‐0.04, 1.24]

Figuras y tablas -
Comparison 2. FF/VI 100/25 versus same dose of FF
Comparison 3. FF/VI 100/25 versus same dose VI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

1

53

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 3. FF/VI 100/25 versus same dose VI
Comparison 4. FF/VI 100/25 versus FP 500 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

1

301

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.10, 2.47]

2 Serious adverse events Show forest plot

1

301

Odds Ratio (M‐H, Fixed, 95% CI)

0.20 [0.05, 0.80]

Figuras y tablas -
Comparison 4. FF/VI 100/25 versus FP 500 µg
Comparison 5. FF/VI 100/25 versus FPS 250/50 bd

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

677

Mean Difference (Fixed, 95% CI)

0.09 [‐0.03, 0.21]

2 Exacerbations Show forest plot

1

806

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.05, 5.52]

3 Serious adverse events Show forest plot

1

806

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.21, 2.99]

4 FEV1 Show forest plot

1

Mean Difference (Fixed, 95% CI)

‐0.02 [‐0.07, 0.03]

5 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.24 [‐0.20, 0.68]

Figuras y tablas -
Comparison 5. FF/VI 100/25 versus FPS 250/50 bd
Comparison 6. FF/VI 100/25 µg versus FF/VI 200/25 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

2

515

Odds Ratio (M‐H, Fixed, 95% CI)

2.02 [0.50, 8.19]

2 Serious adverse events Show forest plot

2

515

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.18]

Figuras y tablas -
Comparison 6. FF/VI 100/25 µg versus FF/VI 200/25 µg
Comparison 7. FF/VI 200/25 versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbations Show forest plot

1

114

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serious adverse events Show forest plot

1

114

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.21 [0.13, 0.29]

4 Change in asthma symptoms (measured by ACT) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.9 [0.12, 1.68]

Figuras y tablas -
Comparison 7. FF/VI 200/25 versus placebo
Comparison 8. FF/VI 200/25 µg versus FP 500 µg

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

2

606

Mean Difference (Fixed, 95% CI)

0.05 [‐0.08, 0.17]

2 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.03 [‐0.15, 0.21]

3 OLD***Health‐related quality of life Show forest plot

2

606

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.08, 0.17]

4 Exacerbations Show forest plot

2

611

Odds Ratio (M‐H, Fixed, 95% CI)

0.70 [0.22, 2.20]

5 Serious adverse events Show forest plot

3

1003

Odds Ratio (M‐H, Fixed, 95% CI)

0.61 [0.25, 1.49]

6 PEFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

28.6 [20.23, 36.97]

7 PEFR AM Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.0 [24.84, 41.16]

8 PEFR PM Show forest plot

1

Mean Difference (Fixed, 95% CI)

26.2 [18.04, 34.36]

9 % symptom‐free days Show forest plot

1

Mean Difference (Fixed, 95% CI)

4.8 [‐2.84, 12.44]

10 Change in asthma symptoms (measured by ACT) Show forest plot

1

332

Mean Difference (Fixed, 95% CI)

0.8 [0.01, 1.59]

Figuras y tablas -
Comparison 8. FF/VI 200/25 µg versus FP 500 µg
Comparison 9. FF/VI 200/25 versus same dose of FF

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in quality of life (measured by AQLQ at 12 wk) Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.08 [‐0.08, 0.24]

2 Change in quality of life (measured by AQLQ at 24 wk) Show forest plot

1

307

Mean Difference (Fixed, 95% CI)

0.05 [‐0.14, 0.24]

3 Serious adverse events Show forest plot

1

391

Odds Ratio (M‐H, Fixed, 95% CI)

6.06 [0.72, 50.84]

4 FEV1 Litres Show forest plot

1

Mean Difference (Fixed, 95% CI)

0.19 [0.10, 0.28]

5 PEFR AM Show forest plot

1

Mean Difference (Fixed, 95% CI)

33.6 [25.41, 41.79]

6 PEFR PM Show forest plot

1

Mean Difference (Fixed, 95% CI)

30.7 [22.51, 38.89]

7 Change in asthma symptoms (measured by ACT) Show forest plot

1

317

Mean Difference (Fixed, 95% CI)

0.3 [‐0.50, 1.10]

Figuras y tablas -
Comparison 9. FF/VI 200/25 versus same dose of FF