Scolaris Content Display Scolaris Content Display

Smanjenje doze inhalacijskih kortikosteroida u terapiji astme kod odraslih osoba

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Godard 2008 {published data only}

Godard P, Attali V. Comparison of different treatment strategies in stepping down combination treatment withdrawing the LABA versus reducing the ICS dose [Abstract]. Proceedings of the American Thoracic Society. 2006:A213. CENTRAL
Godard P, Greillier P, Pigearias B, Nachbaur G, Desfougeres J‐L, Attali V. Maintaining asthma control in persistent asthma: comparison of three strategies in a 6‐month double‐blind randomised study. Respiratory Medicine 2008;102(8):1124‐31. CENTRAL

Gunn 1997 {published data only}

Gunn SD, Choudhury M, Au K, Burnett RJ, Black HL, Turbitt ML. Inhaled steroid treatment reduction in stable asthmatics: a comparison of beclomethasone/budesonide metered dose inhaler with half dose treatment using budesonide Turbohaler, given once or twice daily. British Journal of Clinical Research 1997;8:67‐80. CENTRAL

Hawkins 2003 {published data only}

Hawkins G, McMahon AD, Twaddle S, Wood SF, Ford I, Thompson NC. Stepping down inhaled corticosteroids in asthma: a randomised controlled trial. Thorax 2002;57(Suppl III):iii11. CENTRAL
Hawkins G, McMahon AD, Twaddle S, Wood SF, Ford I, Thomson NC, et al. Stepping down inhaled corticosteroids in asthma: randomised controlled trial. BMJ 2003;326(7399):1115‐8. CENTRAL
See S, Rubin S. Tapering inhaled steroids effective for chronic asthma. Journal of Family Practice 2003;52(10):748‐51. CENTRAL

Juniper 1991 {published data only}

Juniper EF, Kline PA, Vanzieleghem MA, Hargreave FE. Reduction of budesonide after a year of increased use: a randomised controlled trial to evaluate whether improvements in airway responsiveness and clinical asthma are maintained. Journal of Allergy and Clinical Immunology 1991;87(2):483‐9. CENTRAL

Knox 2007 {published data only}

Knox A, Langan J, Martinot JB, Gruss C, Höfner D. Comparison of a step‐down dose of once‐daily ciclesonide with a continued dose of twice‐daily fluticasone propionate in maintaining control of asthma. Current Medical Research Opinion 2007;23(10):2387‐94. CENTRAL

Magnussen 2000 {published data only}

Magnussen H, Magnussen H, Comparative Inhaled Steroid Investigation Group CISIG. Equivalent asthma control after dose reduction with HFA‐134a beclomethasone solution aerosol. Respiratory Medicine 2000;94(6):549‐55. CENTRAL

References to studies excluded from this review

Aalbers 2004 {published data only}

Aalbers R. Fixed or adjustable maintenance‐dose budesonide/formoterol compared with fixed maintenance‐dose salmeterol/fluticasone propionate in asthma patients aged [greater‐than or equal to]16 years: post hoc analysis of a randomized, double‐blind open‐label extension, parallel‐group study. Clinical Drug Investigation 2010;30(7):439‐51. CENTRAL
Aalbers R, Backer V, Kava TT, Omenaas ER, Sandström T, Jorup C, et al. Adjustable maintenance dosing with budesonide/formoterol compared with fixed‐dose salmeterol/fluticasone in moderate to severe asthma. Current Medical Research Opinion 2004;20(2):225‐40. CENTRAL
Aalbers R, Harris A, Naya I. Adjustable dosing with budesonide/formoterol achieves sustained guideline 'well‐controlled asthma' following step down in treatment [Abstract]. European Respiratory Journal. 2005; Vol. 26:Abstract No. 431. CENTRAL

Adachi 2001 {published data only}

Adachi M, Kohno Y, Minoguchi K, Yasurou K, Kochno Y. Step‐down and step‐up therapy in moderate persistent asthma. International Archives of Allergy and Immunology 2001;124(1‐3):414‐6. CENTRAL

ALA 2007 {published data only}

The American Lung Association Asthma Clinical Research Centers. Randomized comparison of strategies for reducing treatment in mild persistent asthma. New England Journal of Medicine 2007;356(20):2027‐39. CENTRAL

Anonymous 1979 {published data only}

Anonymous. Double‐blind trial comparing two dosage schedules of beclomethasone dipropionate aerosol with a placebo in chronic bronchial asthma. Second report of the Brompton Hospital/Medical Research Council Collaborative Trial. British Journal of Diseases of the Chest 1979;73(2):121‐32. CENTRAL

Baba 1999 {published data only}

Baba K, Hattori T, Sakakibara A, Kobayashi T, Takagi K. The usefulness of pranlukast or seratrodast for step‐down of inhaled corticosteroid therapy in adult chronic asthma. American Journal of Respiratory and Critical Care Medicine 1999;159(3; Part 2 of 2):A626. CENTRAL

Baba 2000 {published data only}

Baba K, Hattori T, Koishikawa I, Yoshida K, Kobayashi T, Takagi K. Serum eosinophil cationic protein for predicting the prognosis of a step‐down in inhaled corticosteroid therapy in adult chronic asthmatics. Journal of Asthma 2000;37(5):399‐408. CENTRAL

Bateman 2005 {published data only}

Bateman E, Atienza T, Mihaescu T, Duggan M, Jacques L, Goldfrad C. Asthma control is maintained if treatment with fluticasone propionate/salmeterol (FSC; advair/seretide) is stepped down [Abstract]. American Thoracic Society 2005 International Conference. 2005:C12. CENTRAL
Bateman ED, Jacques L, Goldfrad C, Atienza T, Mihaescu T, Duggan M. Asthma control can be maintained when fluticasone propionate/salmeterol in a single inhaler is stepped down. Journal of Allergy and Clinical Immunology 2006;117(3):563‐70. CENTRAL

Belda 2006 {published data only}

Belda J, Parameswaran K, Lemiere C, Kamada D, O'Byrne PM, Hargreave FE. Predictors of loss of asthma control induced by corticosteroid withdrawal. Canadian Respiratory Journal 2006;13(3):129‐33. CENTRAL

Boulet 1990 {published data only}

Boulet LP, Cartier A, Cockcroft DW, Gruber JM, Laberge F, MacDonald GF, et al. Tolerance to reduction of oral steroid dosage in severely asthmatic patients receiving nedocromil sodium. Respiratory Medicine 1990;84(4):317‐23. CENTRAL

Brambilla 1994 {published data only}

Brambilla C, Godard P, Lacronique J, Allaert FA, Duroux P, Blaive B, et al. A 3‐month comparative dose‐reduction study with inhaled beclomethasone dipropionate and budesonide in the management of moderate to severe adult asthma. Drug Investigation 1994;8(1):49‐56. CENTRAL

Britton 1997 {published data only}

Britton MG, Bone MF, Boyd G, Catterall JR, Ward MJ, Richards K. Comparison of a lower fixed dose of inhaled fluticasone propionate FP with a high dose step‐down regimen of FP in the prevention of re‐exacerbations after an acute severe attack of asthma requiring oral corticosteroid therapy. Thorax 1997;52(Suppl 6):A1/S3. CENTRAL

Bruggenjurgen 2005 {published data only}

Bruggenjurgen B, Selim D, Kardos P, Richter K, Vogelmeier C, Roll S, et al. Economic assessment of adjustable maintenance treatment with budesonide/formoterol in a single inhaler versus fixed treatment in asthma. Pharmacoeconomics 2005;23(7):723‐31. CENTRAL

Busse 2003 {published data only}

Busse W, Koenig SM, Oppenheimer J, Sahn SA, Yancey SW, Reilly D, et al. Steroid‐sparing effects of fluticasone propionate 100 microg and salmeterol 50 microg administered twice daily in a single product in patients previously controlled with fluticasone propionate 250 microg administered twice daily. Journal of Allergy and Clinical Immunology 2003;111(2):57‐65. CENTRAL

Campbell 1998 {published data only}

Campbell LM, Gooding TN, Aitchison WR, Smith N, Powell JA. Initial loading (400 micrograms twice daily) versus static (400 micrograms nocte) dose budesonide for asthma management. PLAN Research Group. International Journal of Clinical Practice 1998;52(6):361‐8, 370. CENTRAL

Casale 2003 {published data only}

Casale TB, Nelson HS, Kemp J, Parasuraman B, Uryniak T, Liljas B. Budesonide Turbuhaler delivered once daily improves health‐related quality of life and maintains improvements with a stepped‐down dose in adults with mild to moderate asthma. Annals of Allergy, Asthma & Immunology 2003;90(3):323‐30. CENTRAL

Chanez 2001 {published data only}

Chanez P, Karlstrom R, Godard P. High or standard initial dose of budesonide to control mild‐to‐moderate asthma?. European Respiratory Journal 2001;17(5):856‐62. CENTRAL

Chiu 2011 {published data only}

Chiu K‐C, Hsu J‐Y, Lin M‐S, Liu W‐T, Wang C‐H, Kuo H‐P. Comparison of the efficacy of cicleosonide with budesonide in mild to moderate asthma patients after step‐down therapy [Abstract]. European Respiratory Society Annual Congress. 2011; Vol. 38:721s [P3965]. CENTRAL
Chiu KC, Chou YL, Hsu JY, Lin MS, Lin CH, Chou PC, et al. Comparison of the efficacy of ciclesonide with that of budesonide in mild to moderate asthma patients after step‐down therapy: a randomised parallel‐group study. NPJ Primary Care Respiratory Medicine 2014;24:14010. CENTRAL

Chung 2002 {published data only}

Chung KF, Holgate S, O'Brien J, Fox H, Thirlwell J. Inhaled corticosteroid dose reducing effect of amalizumab in patients with controlled, severe asthma according to usage of inhaled long acting beta agonists [Abstract]. Journal of Allergy, Asthma and Immunolgy 2002;109(Suppl 1):Abstract No: 726. CENTRAL

Davies 1977 {published data only}

Davies G, Thomas P, Broder I, Mintz S, Silverman F, Leznoff A, et al. Steroid‐dependent asthma treated with inhaled beclomethasone dipropionate. A long‐term study. Annals of Internal Medicine 1977;86(5):549‐53. CENTRAL

Dorinsky 2003 {published data only}

Dorinsky P, Stauffer J, Waitkus‐Edwards K, Yancey S, Prillaman BA, Sutton L. "Stepping down" from fluticasone propionate/salmeterol 100/50mcg diskus results in loss of asthma control [Abstract]. European Respiratory Journal 2004;24(Suppl 48):309s. CENTRAL
Dorinsky P, Yancey S, Reilly D, Stauffer J, Edwards L, Sutton L. Control of airway inflammation is maintained in asthma patients following a reduction in ICS dose with fluticasone propionate/salmeterol (FSC) compared with higher dose fluticasone propionate (FP) alone [Abstract]. European Respiratory Journal. 2004; Vol. 24:308s. CENTRAL
Dorinsky PM, Stauffer J, Waitkus‐Edwards K, Yancey S, Prillaman BA, Sutton L. "Stepping down" from fluticasone propionate/salmeterol 100/50mcg Diskus(R) results in loss of asthma control: lack of effect of ethnic origin [Abstract]. Chest 2004;126(4 Suppl):758S‐a. CENTRAL
Dorinsky PM, Yancey SW, Reilly D, Edwards L. The effectiveness of fluticasone propionate/salmeterol 100/50mcg Diskus(R) (FSC) as an inhaled corticosteroid‐sparing agent: effect of baseline asthma severity [Abstract]. Chest. 2003; Vol. 124:87S‐b, 88. CENTRAL
Dorinsky PM, Yancey SW, Waitkus‐Edwards K, Edwards L, Dorinsky PM, Yancey SW, et al. Clinical markers of worsening asthma with the fluticasone propionate/salmeterol 100/50 mcg Diskus(R) (FSC) vs fluticasone propionate (FP) 250 mcg alone in patients requiring FP 250 mcg BID for asthma stability [Abstract]. Chest 2003;124(4):88S. CENTRAL
Oppenheimer J, Stauffer J, Waitkus‐Edwards K, Yancey S, Prillaman B, Sutton L, et al. 'Stepping down' from fluticasone propionate/salmeterol 100/50mcg diskus results in loss of asthma control [Abstract]. American Thoracic Society 100th International Conference. 2004:A37 Poster J94. CENTRAL

Fardon 2005 {published data only}

Fardon TC, Haggart K, Lee DKC, Lipworth BJ. Stepping down inhaled corticosteroids in severe asthma with long acting bronchodilators utilising effort dependent and independent measures of pulmonary function [Abstract]. American Thoracic Society 2005 International Conference. 2005:[B35] [Poster: G3]. CENTRAL

Fardon 2007 {published data only}

Fardon T, Haggart K, Lee DKC, Lipworth BJ. A proof of concept study to evaluate stepping down the dose of fluticasone in combination with salmeterol and tiotropium in severe persistent asthma. Respiratory Medicine 2007;101(6):1218‐28. CENTRAL

FitzGerald 2003 {published data only}

FitzGerald JM, Sears MR, Boulet LP, Becker AB, McIvor AR, Ernst P, et al. Adjustable maintenance dosing with budesonide/formoterol reduces asthma exacerbations compared with traditional fixed dosing: a five‐month multicentre Canadian study. Canadian Respiratory Journal 2003;10(8):427‐34. CENTRAL
FitzGerland JM, Sears MR, Boulet LP, Becker AB, Boulet L‐P, Becker AB. Erratum: Adjustable maintenance dosing with budesonide/formoterol reduces asthma exacerbations compared with traditional fixed dosing: a five month multicentre Canadian study [Canadian Respiratory Journal (2003) vol. 10 (8) (427‐434)]. Canadian Respiratory Journal 2004;11(1):20. CENTRAL

FitzGerald 2005 {published data only}

FitzGerald JM, Boulet LP, Follows RMA. The CONCEPT trial: a 1‐year, multicenter, randomized, double‐blind, double‐dummy comparison of a stable dosing regimen of salmeterol/fluticasone propionate with an adjustable maintenance dosing regimen of formoterol/ budesonide in adults with persistent asthma. Clinical Therapeutics 2005;27(4):393‐406. CENTRAL
Price DB, Williams AE, Yoxall S. Salmeterol/fluticasone stable‐dose treatment compared with formoterol/budesonide adjustable maintenance dosing: impact on health‐related quality of life. Respiratory Research 2007;8:46. CENTRAL

FLIQ96 2005 {published data only}

FLIQ96 GlaxoSmithKline. Effect of fourteen weeks treatment with fluticasone propionate step‐down dosing from 1000mcg/day in comparison with a fixed dose of 200mcg/day followed by an open phase with placebo administered for 8 weeks in subjects with mild asthma who exacerbate during treatment with beta‐2 agonist (prn). GlaxoSmithKline Clinical Study Register2005. CENTRAL

Fowler 2002 {published data only}

Fowler S, Currie GP, Lipworth BJ. Step down therapy for asthma with inhaled steroids alone or in combination with a long acting beta2 agonist [Abstract]. Journal of Allergy, Asthma and Immunolgy 2002;109(Suppl 1):Abstract No: 745. CENTRAL

Haggart 2004 {published data only}

Haggart K, Fardon TC, Lee DKC, Lipworth BJ. Stepping down inhaled steroids in severe asthma with long acting bronchodilators: utilising effort dependent and independent measures of pulmonary function [Abstract]. Thorax 2004;59(Suppl II):ii72. CENTRAL

Hamada 2008 {published data only}

Hamada K, Yasuba H, Tanimura K, Hiramatu M, Kobayashi Y, Kita H. How can we stop ICS? Risk control therapy by as needed inhaled fluticasone after stepping down [Abstract]. Journal of Allergy and Clinical Immunology. 2008; Vol. 121:S219 [844]. CENTRAL

Kardos 2001 {published data only}

Kardos P, Brüggenjürgen B, Martin A, Meyer‐Sabellek W, Richter K, Vogelmeier C, et al. Treatment of bronchial asthma using a new adjustable combination treatment plan: Asthma Control Plan (ATACO) [German]. Pneumologie 2001;55(5):253‐7. CENTRAL

Kawagishi 2000 {published data only}

Kawagishi Y, Oosaki R, Kashii T, Kawasaki A, Fujishita T, Arai N, et al. Long‐term prognosis of asthmatic patients treated with low‐dose beclomethasone dipropionate. Arerugi 2000;49(5):391‐6. CENTRAL

Keonig 2004 {published data only}

Keonig S, Waitkus‐Edwards K, Yancey S, Prillman B, Dorinsky P, Prillaman B. Loss of asthma control when patients receiving fluticasone propionate/salmeterol 100/50 µg Diskus are "stepped‐down" to fluticasone propionate, salmeterol or montelukast alone [Abstract]. Journal of Allergy and Clinical Immunology. 2004; Vol. 113:S94. CENTRAL

Massanari 2008 {published data only}

Massanari M, Jimenez P, Kianifard F, Maykut R, Zeldin R. The omalizumab associated decrease in peripheral blood eosinophils in moderate severe IgE mediated asthma is sustained following inhaled steroid dose reduction [Abstract]. American Thoracic Society International Conference. 2008:A105. CENTRAL

McKinlay 2011 {published data only}

McKinlay L, Williamson PA, Short PM, Fardon TC, Lipworth BJ. Proof of concept study to evaluate step‐down therapy with inhaled corticosteroid alone or additive therapy on surrogate inflammatory markers in asthma. British Journal of Clinical Pharmacology 2011;71(1):128‐31. CENTRAL

Mikloweit 2000 {published data only}

Mikloweit P, Zachgo W, Hader S, Reiss Plagemann C, Grimm A, Drews G. Lung function before and 12 weeks after switching to a reduced dose of HFA‐beclomethasone dipropionate. American Journal of Respiratory and Critical Care Medicine 2000;161(3 Suppl):A186. CENTRAL

Obase 2013 {published data only}

Obase Y, Ikeda M, Kurose K, Abe M, Shimizu H, Ohue Y, et al. Step‐down of budesonide/formoterol in early stages of asthma treatment leads to insufficient anti‐inflammatory effect. Journal of Asthma 2013;50(7):718‐21. CENTRAL

Paggiaro 2011 {published data only}

Paggiaro P, Nicolini G, Crimi N, Fabbri L, Olivieri D, Rossi A, et al. Six months step down treatment from high dose ICS/LABA combination therapy in asthma [Abstract]. American Journal of Respiratory and Critical Care Medicine. 2011; Vol. 183:A1278. CENTRAL
Paggiaro P, Nicolini G, Crimi N, Fabbri LM, Olivieri D, Rossi A, et al. Asthma control and lung function after step down from high dose ICS/LABA combination therapy [Abstract]. European Respiratory Society Annual Congress. 2011; Vol. 38:722s [P3967]. CENTRAL

Reddel 2007 {published data only}

Reddel HK, Peyters MJ, Wark PA, Sand IB, Jenkins CR. Comparison of the efficacy of seretide and flixotide when down‐titrating the inhaled corticosteroid dose [Abstract]. Respirology. (Carlton, Vic.), 2007; Vol. 12:A40. CENTRAL

Rumbak 1998 {published data only}

Rumbak M, Self T, Kelso T, Eberle L, Abou‐Shala N, Learned S, et al. Moderate to high dose inhaled corticosteroids in adult asthmatics: does salmeterol facilitate step down therapy? [Abstract]. European Respiratory Journal. 1998; Vol. 12:19s, P157. CENTRAL

Schmier 2003 {published data only}

Schmier J, Leidy NK, Gower R. Reduction in oral corticosteroid use with mometasone furoate dry powder inhaler improves health‐related quality of life in patients with severe persistent asthma. Journal of Asthma 2003;40(4):383‐93. CENTRAL

Shamsul 2007 {published data only}

Shamsul AI, Hadzri HM, Noradina AT, Fauzi MA, Hamid AJ, Rosalina AM, et al. Step‐down approach in chronic stable asthma; a comparison of reducing dose inhaled formoterol/budesonide with maintaining inhaled budesonide [Abstract]. Respirology. (Carlton, Vic.), 2007; Vol. 12:A141. CENTRAL

Ställberg 2003 {published data only}

Ställberg B, Olsson P, Jörgensen LA, Lindarck N, Ekström T. Budesonide/formoterol adjustable maintenance dosing reduces asthma exacerbations versus fixed dosing. International Journal of Clinical Practice 2003;57(8):656‐61. CENTRAL

Additional references

Adams 2005

Adams NP, Bestall JB, Malouf R, Lasserson TJ, Jones PW. Beclomethasone versus placebo for chronic asthma. Cochrane Database of Systematic Reviews 2005, Issue 1. [DOI: 10.1002/14651858.CD002738]

Adams 2008

Adams N, Bestall J, Lasserson TJ, Jones PW, Cates CJ. Fluticasone versus placebo for chronic asthma in adults and children. Cochrane Database of Systematic Reviews 2008, Issue 4. [DOI: 10.1002/14651858.CD003135.pub4]

Ahmad 2015

Ahmad S. Stopping long‐acting beta2‐agonists (LABA) for adults with asthma well controlled by LABA and inhaled corticosteroids. Cochrane Database of Systematic Reviews 2015, Issue 6. [DOI: 10.1002/14651858.CD011306.pub2]

Bateman 2004

Bateman ED, Boushey HA, Bousquet J, Busse WW, Clark TJ, Pauwels RA, et al. Can guideline‐defined asthma control be achieved? The Gaining Optimal Asthma ControL study. American Journal of Respiratory and Critical Care Medicine 2004;170:836‐44.

Bateman 2008

Bateman ED, Hurd SS, Barnes PJ, Bousquet J, Drazen JM, FitzGerald M, et al. Global strategy for asthma management and prevention: GINA executive summary. European Respiratory Journal 2008;31(1):143‐78.

Colice 2006

Colice GL. New developments in inhaled corticosteroids. Allergy and Asthma Proceedings 2006;27(5):332‐40.

Custovic 2012

Custovic A, Simpson A. The role of inhalant allergens in allergic airways disease. Journal of Investigational Allergology and Clinical Immunology 2012;22(6):393‐401.

Ducharme 2010

Ducharme FM, Ni Chroinin M, Greenstone I, Lasserson, TJ. Addition of long‐acting beta2‐agonists to inhaled steroids versus higher dose inhaled steroids in adults and children with persistent asthma. Cochrane Database of Systematic Reviews 2010, Issue 4. [DOI: 10.1002/14651858.CD005533.pub2]

Fisseler‐Eckhoff 2011

Fisseler‐Eckhoff A, Bartsch H, Zinsky R, Schirren J. Environmental isocyanate‐induced asthma: morphologic and pathogenetic aspects of an increasing occupational disease. International Journal of Environmental Research and Public Health 2011;8(12):3672‐87.

GINA 2016

Global Initiative for Asthma (GINA). From the Global Strategy for Asthma Management and Prevention, 2016. www.ginasthma.org (accessed 19 October 2016).

Gionfriddo 2015

Gionfriddo MR, Hagan JB, Hagan CR, Volcheck GW, Castaneda‐Guarderas A, Rank MA. Stepping down inhaled corticosteroids from scheduled to as needed in stable asthma: systematic review and meta‐analysis. Allergy and Asthma Proceedings 2015;36(4):262‐7.

Global Asthma Network 2014

Global Asthma Network. The Global Asthma Report 2014. http://www.globalasthmareport.org/resources/Global_Asthma_Report_2014.pdf (accessed 16 April 2015).

Hagan 2014

Hagan JB, Samant SA, Volcheck GW, Li JT, Hagan CR, Erwin PJ, et al. The risk of asthma exacerbation after reducing inhaled corticosteroids: a systematic review and meta‐analysis of randomized controlled trials. Allergy 2014;69(4):510‐6.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. www.cochrane‐handbook.org.

Jaakkola 2008

Jaakkola JJ, Knight TL. The role of exposure to phthalates from polyvinyl chloride products in the development of asthma and allergies: a systematic review and meta‐analysis. Environmental Health Perspectives 2008;116(7):845‐53.

Juniper 1990

Juniper EF, Kline PA, Vanzieleghemt MA, Ramsdale EH, O'Byrnett PM, Hargreave FE. Long‐term effects of budesonide on airway responsiveness and clinical asthma severity in inhaled steroid‐dependent asthmatics. European Respiratory Journal 1990;3:1122‐7.

Juniper 1993

Juniper EF, Guyatt GH, Ferrie PJ, Griffith LE. Measuring quality of life in asthma. American Review of Respiratory Disease 1993;147:832‐8.

Kew 2013

Kew KM, Karner C, Mindus SM, Ferrara G. Combination formoterol and budesonide as maintenance and reliever therapy versus combination inhaler maintenance for chronic asthma in adults and children. Cochrane Database of Systematic Reviews 2013, Issue 12. [DOI: 10.1002/14651858.CD009019.pub2]

Kew 2015

Kew KM, Beggs S, Ahmad S. Stopping long‐acting beta2‐agonists (LABA) for children with asthma well controlled on LABA and inhaled corticosteroids. Cochrane Database of Systematic Reviews 2015, Issue 5. [DOI: 10.1002/14651858.CD011316.pub2]

Lipworth 1992

Lipworth BJ, McDevitt DG. Inhaled 2‐adrenoceptor agonists in asthma: help or hindrance?. British Journal of Clinical Pharmacology 1992;33:129‐38.

Lipworth 1999

Lipworth BJ. Systemic adverse effects of inhaled corticosteroid therapy: a systematic review and meta‐analysis. Archives of Internal Medicine 1999;159(9):941‐55.

NHS 2013

National Health Service. Respiratory National Charts NHS Business Services Authority. October 2013. http://www.nhsbsa.nhs.uk/PrescriptionServices/Documents/PPDPrescribingAnalysisCharts/Resp_Oct_2013_National.pdf (accessed 14 April 2015).

Partridge 2006

Partridge MR, van der Molen T, Myrseth SE, Busse WW. Attitudes and actions of asthma patients on regular maintenance therapy: the INSPIRE study. BMC Pulmonary Medicine 2006;6(13):1‐9.

Pearce 1999

Pearce N, Pekkanen J, Beasley R. How much asthma is really attributable to atopy?. Thorax 1999;54(3):268‐72.

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.

Rimmington 1997

Rimmington LD, Aronoffsky L, Mowatt A, Wharburton E, Ryland I, Pearson MG. Use of a simple patient focused asthma morbidity score. European Respiratory Journal 1997;11:1289.

Rogers 2012

Rogers L, Reibman J. Stepping down asthma treatment: how and when. Current Opinion in Pulmonary Medicine 2012;18(1):70‐5.

SIGN/BTS 2016

Scottish Intercollegiate Guidelines Network/British Thoracic Society. British guideline on the management of asthma. A national clinical guideline. Edinburgh/London: SIGN/BTS, 2016. [ISBN 978 1 909103 47 4]

Tse 1984

Tse CS, Bernstein IL. Corticosteroid aerosols in the treatment of asthma. Pharmacotherapy 1984;4(6):334‐42.

WHO 2014

World Health Organization. Adolescents: health risks and solutions. Fact sheet N°345. Updated May 2014. http://www.who.int/mediacentre/factsheets/fs345/en/ (accessed on 10 June 2015).

Xepapadaki 2009

Xepapadaki P, Manios Y, Liarigkovinos T, Grammatikaki E, Douladiris N, Kortsalioudaki C, et al. Association of passive exposure of pregnant women to environmental tobacco smoke with asthma symptoms in children. Pediatric Allergy and Immunology 2009;20:423‐9.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Godard 2008

Methods

Study design: randomised controlled trial

Total duration of study: 24 weeks.

'Run‐in' period: 8 weeks. All participants received salmeterol/fluticasone propionate combination (SFC) at a dose of 50/250 μg twice daily.

Number of study centres and locations: 124 centres (no locations specified)

Study setting: not stated

Date of study: not stated

Participants

Enrolled (N): 603

Randomised (n): 475 (SFC 50/250, n = 159; SFC 50/100, n = 157; FP 250, n = 159)

Analysed (n): 464 (SFC 50/250, n = 154; SFC 50/100, n = 156; FP 250, n = 154)

Withdrawals (n): 63

Median age (range), years: SFC 50/250, 46.5 (18‐81); SFC 50/100, 43.0 (18‐75); FP 250 42.0 (18‐77)

Age range, years: 18‐81

Gender (% female): SFC 50/250, 48.1; SFC 50/100, 46.2; FP 250 51.3

Severity of condition: well controlled on step 2 or 3. Mean % predicted prebronchodilator FEV1 (SD) as follows: SFC 50/250, 87.8 (18.2); SFC 50/100, 91.2 (17.8); FP 250, 90.8 (17.2)

Diagnostic criteria: Asthma control was assessed using the GOAL definitions of 'well controlled' and 'total control'.

Baseline lung function (mean morning PEF (SD), L/min): SFC 50/250, 465.6 (113.2); SFC 50/100, 467.9 (111.2); FP 250, 463.7 (105.1)

Smoking history, % smokers or ex‐smokers: SFC 50/250, 24.7; SFC 50/100, 21.3; FP 250, 16.2

Inclusion criteria: aged ≥18 years; documented history of asthma (≥ 6 months) well controlled with current treatment (ICS at a dose of CFC beclomethasone dipropionate or equivalent and a long‐acting beta2‐agonist at recommended dose) at a stable dose for ≥ 4 weeks before initial clinic visit (V1); respiratory tract infection, with acute exacerbation requiring emergency department treatment/hospitalisation or use of oral/parenteral steroids, within 4 weeks of V1; any change in asthma maintenance treatment within 4 weeks

Exclusion criteria: smoking history ≥ 10 pack‐years; respiratory tract infection

Details of criteria for stepping down treatment: All participants received SFC 50/250 μg twice daily and were randomised to remain on SFC 50/250 or move to 1 of the 2 step‐down treatment arms if their asthma was assessed as 'well controlled' over the last 2 weeks of the run‐in period; asthma control was assessed according to GOAL definitions (see Bateman 2004).

Interventions

Intervention 1: SFC 50/100 μg twice daily

Intervention 2: FP 250 μg twice daily (not relevant to review)

Comparison: SFC 50/250 μg twice daily

Concomitant medications: Short‐acting bronchodilators (previously used as rescue medication) and antihistamines were permitted, provided they had been used for at least 4 weeks.

Excluded medications: All previous asthma medications were discontinued at entry into the run‐in period, except short‐acting bronchodilators (previously used as rescue medication) and antihistamines, provided they had been used for at least 4 weeks.

Outcomes

Primary outcomes: mean morning PEF over the first 12 weeks of randomised treatment

Secondary outcomes: mean morning PEF over the last 12 weeks of randomised treatment; daily symptoms; use of short‐acting bronchodilator as rescue medication; FEV1; asthma control based on GOAL definitions of total control and 'well‐controlled' (see Bateman 2004)

Notes

Funding for trial: not stated

Notable conflicts of interest of trial authors: Three of the trial authors had received sponsorship and had attended advisory boards for various pharmaceutical companies, including AstraZeneca, GlaxoSmithKline and Boehringer‐Ingelheim; 3 authors are employees of GlaxoSmithKline.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information provided

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study reported as double blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Study reported as double blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data provided for all randomised individuals. We note that study authors reported lung function results only for the per‐protocol population, whereas they reported all other outcomes for the intent‐to‐treat population.

Selective reporting (reporting bias)

High risk

Study authors reported the primary outcome for the per‐protocol data set on the basis that this is a non‐inferiority study. Furthermore, the primary outcome considers lung function only over the first 12 weeks of treatment; a secondary outcome assessed lung function in the full analysis set but considered only the second 12 weeks of treatment. All in all, findings were quite confusing and inconsistent. This trial was not reported as registered, and we cannot source a protocol.

Other bias

High risk

The protocol suggests that only participants whose condition was well controlled within the last 2 weeks of the run‐in period would go on to randomisation; however, it appears that a relatively high proportion of participants whose asthma was not controlled were included in the full analysis set. Results of this study are not well reported, and as the study does not appear to have been prospectively registered, and a protocol was not cited, it is difficult to ascertain whether selective outcome reporting occurred. Study sponsorship is not reported, although several authors worked for GSK. Key exclusion criteria of poor control according to ACQ were not defined or reported. A large proportion of poorly controlled randomised participants were not included in the primary outcome analysis (but were included in the secondary outcome analysis). Reporting was confusing.

Gunn 1997

Methods

Study design: randomised controlled trial, multi‐centre, open label

Total duration of study: 2‐week run‐in period; 12‐week treatment period

'Run‐in' period: 2‐week run in period, during which participants remained on their existing doses of ICS ('high‐dose' budesonide 400 μg twice daily, beclomethasone 400 μg twice daily or beclomethasone 500 μg twice daily via a pMDI with a spacer; or 'low‐dose' budesonide or beclomethasone 200 μg twice daily)

Number of study centres and locations: UK

Study setting: primary care

Withdrawals: 147/631 (23%) randomised participants withdrew during the treatment period

Date of study: not stated

Participants

N: 631 patients were randomised after a 2‐week run‐in period.

Mean age (range), years: budesonide OD: 44.1 (16.5‐80.2); budesonide BID: 45.7 (16.7‐77.0); no ICS dose change: 40.9 (16.2‐80.2)

Gender M/F, n: budesonide OD: 100/128; budesonide BID: 90/101; no ICS dose change: 100/112

Severity of condition: baseline mean morning PEFR (SD), L/min: controlled on step 2. Budesonide OD: 437.2 (106.5); budesonide BID: 447.4 (111.3); no ICS dose change: 445.8 (100.9)

Diagnostic criteria: mild, well controlled

Baseline lung function ‐ mean morning PEFR (SD), L/min: budesonide OD: 437.2 (106.5); budesonide BID: 447.4 (111.3); no ICS dose change: 445.8 (100.9)

Smoking history: not stated

Inclusion criteria: aged ≥ 16 years; documented diagnosis of asthma (currently stable); asthma considered by physician to be well controlled (as per BTS guidelines); receiving 200 μg twice daily (low dose) or 400/500 μg twice daily (high dose) budesonide or beclomethasone (via a pMDI ± spacer) for 6 months before entry; patients on the higher dose of steroid were required to have used a large volume spacer for a minimum of 4 weeks before entry

Exclusion criteria: pregnant, at risk of pregnancy, breast feeding, brittle asthma, night shift workers. Within 3 months: any increase in total daily inhaled steroid dose; exacerbation resulting in hospitalisation or requiring nebulisation, oral/injectable/rectal steroids, beta blockers, sodium cromoglycate, sodium nedocromil, any unlicensed medication or fluticasone propionate. Within 1 week before the study: Patients were not permitted to have taken theophylline (or derivatives), any long‐acting bronchodilators, ipratropium/oxitropium bromide or ketotifen.

Details of criteria for stepping‐down treatment: Participants were eligible for randomisation if their diary cards showed that they had no nocturnal wakening due to asthma in the previous 7 nights, and if they fulfilled 3 of the following criteria:

asthma symptoms of no more than mild severity experienced on 3 or fewer days of the previous 7 days; using ≤ 1 puff per day of inhaled bronchodilator on a maximum of 5 of the last 7 days; circadian variation in PEFR < 20% in the previous 7 days; morning PEFR ≥ 80% or predicted or best (if this value was greater than predicted) on 5 of the 7 previous days

Interventions

Intervention: Participants on an initial high dose of ICS (budesonide 400 μg twice daily or beclomethasone 400 μg twice daily or beclomethasone 500 μg twice daily delivered via a pMDI and a spacer device) were randomised to receive budesonide 200 μg twice daily via a turbuhaler or 400 μg once daily (i.e. both groups represent a halving of the initial ICS dose). Participants on an initial low dose of ICS (budesonide or beclomethasone 200 μg twice daily) were randomised to receive budesonide 100 μg twice daily via a turbuhaler or 200 μg once daily (i.e. both groups represent a halving of the initial ICS dose).

Comparison: No change in initial dose of budesonide or beclomethasone.

Concomitant medications: Each patient was given terbutaline (Bricanyl) turbuhaler 500 μg prn for rescue mediation during the run‐in and throughout the study.

Excluded medications: See exclusion criteria.

Outcomes

Primary outcomes: morning PEFR recorded by diary cards (recorded at baseline, and at 4, 8 and 12 weeks)

Secondary outcomes: evening PEFR, proportion of symptom‐free days/nights, proportion of beta2‐agonist‐free days/nights, sleep disturbance (all recorded via diary cards) quality of life (Juniper Asthma Quality of Life Questionnaire (Juniper 1993); PEFR measured at clinic visits; asthma severity measured at clinic visits; asthma control)

Notes

Funding for trial: not stated; likely Astra Pharmaceuticals

Notable conflicts of interest of trial authors: not stated. One study author was an employee of Astra Pharmaceuticals.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information provided

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The study was open label.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The study was open label, and it does not appear that outcome assessors were blinded to treatment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Incomplete primary outcome data were reported, but the number of participants for whom data were missing was similar across OD/BD/pMDI groups.

Selective reporting (reporting bias)

High risk

No study protocol available. The data for high‐dose and low‐dose groups were pooled and were not presented individually. Study authors stated that separate data were not presented individually because no significant differences between the 2 dose groups were found for any of the analyses performed.

Other bias

Unclear risk

Unclear risk of bias: This is a complicated study, and some changes in inhaler device appear to have occurred at the same time as changes in dose. Participants entered the run‐in period on their existing dose of ICS ('high' or 'low') and were later randomised to remain on their existing dose, or step down to half the dose in 1 of 2 different formats (half the dose twice daily, or the same dose but only once daily). No data were reported for the run‐in period.

Unclear risk of bias: Funding for the study is not reported. The paper has industry authors, and the company manufactures products that seem to match the products reported upon. Funding for the study is not declared, but one study author is employed by Astra.

Hawkins 2003

Methods

Study design: randomised controlled trial, double‐blind, parallel group

Total duration of study: 1 year

'Run‐in' period: 1 month

Number of study centres and locations: general practices in Western and Central Scotland

Study setting: primary care (general practice)

Withdrawals: 24/130 participants in the stepdown group and 22/129 in the control group discontinued the intervention. Analyses were performed on all randomised participants.

Date of study: The study was performed between May 1999 and October 2001.

Participants

N: 259 participants were randomised.

Mean age (SD), years: step‐down 52.8, (14.5); control 55 (15.2)

Age range: 18‐86 years

Gender (M/F), n: step‐down, 54/76; control, 54/75

Severity of condition: controlled on high‐dose ICS (at least 1000 μg BDP) plus possibly other drugs (steps 2‐4)

Baseline lung function ‐ % predicted pre‐salbutamol FEV1 (SD), L/min: step‐down, 80.3 (19.2); control, 80.1 (18.6)

Smoking history ‐ current/former/never, n: step‐down, 16/44/70; control, 17/49/63

Inclusion criteria: aged ≥ 18 years; diagnosis of asthma ≥ 1 year; treated with ≥ 800 μg inhaled BDP (or budesonide or fluticasone propionate at equivalent dosage)

Exclusion criteria: required oral corticosteroids or attended general practice or hospital within 2 months; inability to use peak flow meter; treatment with immunosuppressive drugs; serious illness; alcohol, substance or drug misuse; pregnancy; participation in other research within the past 6 months

Details of criteria for stepping down treatment: stable asthma (i.e. good control) assessed at end of run‐in period and at 3, 6, 9 and 12 months. Good control was defined as an asthma morbidity score ≤ 2, no visits to general practice or hospital since previous visit and peak flow ≥ target flow on 8 of the previous 14 days; if peak flow data were missing, the first two criteria were used.

Interventions

Intervention: step‐down ‐ 50% reduction in ICS dose

Comparison: no change in ICS dose

Concomitant medications: Reliever inhalers were permitted. 36.9% of the step‐down group and 30.2% of the control group were receiving a concomitant LABA.

Excluded medications: immunosuppresive drugs

Outcomes

Primary outcomes: proportion of participants experiencing an asthma exacerbation, asthma control (short asthma morbidity score (Rimmington 1997); scores ranged from 0 (perfect control) to 8 (very poor control))

Secondary outcomes: adverse events, health‐related quality of life (EuroQoL and St George's Respiratory Questionnaire), annual corticosteroid dose

Notes

Funding for trial: NHS R&D Programme on Asthma Management

Notable conflicts of interest of trial authors: Study authors had received funding, and various pharmaceutical companies including GlaxoSmithKline provided the study inhalers.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Well‐described randomisation with computer‐generated randomisation stratified by centre

Allocation concealment (selection bias)

Low risk

Computer‐allocated randomisation sequence; randomisation code withheld from investigators until study completion

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and personnel were blinded to treatment allocation via use of identical inhaler packs.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Randomisation code was maintained blind until the end of the study.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Primary outcome data were reported for all participants as intention to treat. Some data for health status secondary outcome measures were missing (not explained), but the number of participants for whom data were missing was similar in both treatment groups. Lung function was not reported during or at the end of the treatment period.

Selective reporting (reporting bias)

High risk

Protocol was not available. It is not clear why study authors did not present lung function as, according to the Methods section, participants did monitor lung function for 2 weeks before each visit. Detailed adverse event data were not presented.

Other bias

Low risk

Study medication was provided by industry, but study was funded by NHS R&D programme on asthma development. No industry was involved in authorship of the paper.

Juniper 1991

Methods

Study design: randomised controlled trial, double‐blind, parallel group

Total duration of study: 3 months

'Run‐in' period: no run‐in (this is a follow‐up extension to a previous study)

Number of study centres and locations: Firestone Regional Chest and Allergy Clinic at St Joseph's Hospital and the McMaster University Medical Centre in Hamilton, Canada

Study setting: secondary care (asthma clinic)

Withdrawals: All 28 participants completed the study

Date of study: not reported

Participants

N: 28. A subgroup of 14 participants were relevant to this review.

Mean age: not reported. Mean age in parent study was ˜ 42 years (Juniper 1990).

Age range: not reported

Gender: not reported

Severity of condition: controlled on step 2 (mild to moderate: approximately half of participants were 'steroid dependent')

Baseline lung function: Individual participant data were reported. At entry to initial study, all participants had airway hyper‐responsiveness to methacholine (PC20 < 8.0 mg/mL) and symptomatic asthma.

Smoking history: not reported

Inclusion criteria and exclusion criteria: successful completion of previous study

Details of criteria for step‐down treatment: not reported

Interventions

Intervention: a halving of the budesonide dose in steroid‐dependent participants (n = 6)

Comparison: no change in budesonide dose among steroid‐dependent participants (n = 8)

Concomitant medications: Bronchodilator medication was permitted (long‐acting vs short‐acting not specified).

Excluded medications: not reported

Outcomes

Primary outcomes: airway responsiveness to methacholine (measured with a standardised tidal breathing protocol); clinical asthma severity (i.e. asthma control assessed via asthma severity questionnaire). The questionnaire comprised 6 items: awakened at night by symptoms; awakened in the morning by symptoms; limitation of normal daily activities; sputum; use of bronchodilator more than 4 times per day; FEV1 prebronchodilator < 70% predicted (One point was scored for each of the first 5 items that had been positive on ≥ 1 day during the previous week; 1 point was scored for reduced spirometry; therefore, the maximum asthma severity score (i.e. worst control) was 6).

Secondary outcomes: bronchodilator use; allergen exposure score; upper respiratory tract infection score

Notes

Funding for trial: Funding was not reported.

Notable conflicts of interest of trial authors: Conflicts of interest were not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information provided

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was reported as double‐blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study was reported as double‐blind.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All 28 randomised participants completed the study, and it appears that data were reported for all 28 participants.

Selective reporting (reporting bias)

High risk

Study authors state, "During analysis, it was found that all the outcomes in the two reduction groups were very similar, and also, the outcomes in the two groups in whom steroids were not reduced were very similar. Therefore, for simplicity, the data have been combined and are presented as two groups, reduced and maintained".

No protocol was available; no prespecified analysis plan was prepared. Group data were combined as described above.

Other bias

Low risk

None identified

Knox 2007

Methods

Study design: randomised controlled, double‐blind, double‐dummy, parallel group

Total duration of study: 14 weeks

'Run‐in' period: 2 weeks

Number of study centres and locations: 16 centres (8 each in UK and Belgium)

Study setting: not stated

Withdrawals: 5 participants (CIC 160 μg, n = 4; FP 250 μg, n = 1)

Date of study: October 2004 to July 2005

Participants

N: 111 randomised

Mean age, years: CIC 160 μg OD: 43; FP 250 μg BID: 46

Age range, years: 18‐75

Gender M/F, n: CIC 160 μg OD: 28/30; FP 250 μg BID: 30/23

Severity of condition: controlled on step 2

Baseline lung function ‐ mean (SD) FEV1, L: CIC 160 μg OD: 3.272 (0.869); FP 250 μg BID: 3.146 (0.823)

Smoking history ‐ non‐smoker/ex‐smoker/current smoker, n: CIC 160 μg OD: 38/18/2; FP 250 μg BID: 34/18/1

Inclusion criteria: male and female patients aged 17–75 years; diagnosis of asthma as defined by American Thoracic Society guidelines for at least 6 months, but otherwise in good health; FEV1 ≥ 90% of predicted; maintained asthma control over previous 3 months using fluticasone propionate 250 μg twice daily, or equivalent, with short‐acting bronchodilator use as rescue medication only

Exclusion criteria: concomitant severe disease, such as a lower respiratory tract infection; chronic obstructive pulmonary disease or other relevant lung diseases; more than 1 emergency care visit or hospitalisation due to asthma exacerbations in the previous year; or clinically relevant abnormal laboratory values suggesting an
unknown disease. Other exclusion criteria were use of systemic glucocorticoids, long‐acting β2‐agonists,
oral β2‐agonists and sustained‐release xanthines within 3 months before study entry; pregnancy and breast‐feeding among
female patients; and ex‐smokers or current smokers with ≥ 10 pack‐years.

Details of criteria for step‐down treatment: Participants were randomised to step‐down (for eligibility, see inclusion and exclusion criteria).

Interventions

Intervention: ciclesonide 160 μg OD (i.e. ˜ 50% reduction according to GINA 2016)

Comparison: fluticasone propionate 250 μg BID (i.e. no change)

Concomitant medications: short‐acting bronchodilator used as rescue medication only

Excluded medications: See exclusion criteria.

Outcomes

Primary outcomes: efficacy ‐ percentage of days with asthma control (defined as days without asthma symptoms and without rescue medication use); asthma symptom‐free days; rescue medication‐free days; and nocturnal awakening‐free days. Safety ‐ adverse events

Secondary outcomes: efficacy ‐ FEV1; forced vital capacity (FVC); PEF from spirometry; PEF from participant diaries measured on a Mini‐Wright PEF meter; asthma symptom scores from participant diaries (sum scores based on a 9‐point scale, with 0 indicating no symptoms); use of rescue medication; number of participants with an asthma exacerbation; and time to onset of the first asthma exacerbation. Safety ‐ vital signs (blood pressure and pulse rate); standard laboratory tests (including haematology, blood chemistry and urinalysis); and number of participants with oral candidiasis

Notes

Funding for trial: This study was funded and sponsored by ALTANA Pharma AG, a member of the Nycomed Group.

Notable conflicts of interest of trial authors: Editorial assistance for preparation of the manuscript was provided by Nathan Price‐Lloyd, PhD, Medicus International, which was funded by ALTANA Pharma AG, a member of the Nycomed Group. Study authors reported no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information provided

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was reported as double‐blind, double‐dummy.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study was reported as double‐blind, double‐dummy.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analyses were performed for safety analyses and comprised all randomised participants. Some data for lung function analyses were missing, but only from 3 participants in the step‐down group.

Selective reporting (reporting bias)

Unclear risk

Protocol was not available; however, the range of outcomes seems fairly comprehensive.

Other bias

Low risk

None identified

Magnussen 2000

Methods

Study design: randomised controlled, double‐blind, double‐dummy, parallel group

Total duration of study: 14 weeks

'Run‐in' period: 4 weeks

Number of study centres and locations: 18 pulmonology practices

Study setting: pulmonology outpatient practices

Withdrawals: none reported

Date of study: November 1996 to October 1997

Participants

N: 150

Mean (SD) age, years: 400 μg/day BDP: 43 (15); 1000 μg/day BDP: 42 (15)

Age range: not reported

Gender ‐ M/F, n: 400 μg/day BDP: 22/50; 1000 μg/day BDP: 30/48

Severity of condition: step 2

Baseline lung function ‐ mean (SE) FEV1, L: 400 μg/day BDP: 2.77 (0.09); 1000 μg/day BDP: 2.85 (0.09)

Smoking history: not reported

Inclusion criteria and exclusion criteria, allowable range: age 18‐75 years; use of inhaled steroids for ≥ 3 months (BDP 1000 mg or BUD 800‐1000 mg); use of β2‐agonists on demand (≥ 1 puffs/d); reversible airflow obstruction assessed within the last 2 years; change in FEV ≥ 12%; change in PEF ≥ 20%; bronchial hyper‐responsiveness to inhaled histamine (PC20 FEV1 ≥ 4 mg/mL); baseline FEV1 ≥ 60% of predicted; variability of baseline FEV1 during run‐in period ≤ 15%

Details of criteria for step‐down treatment: Participants were randomised to step‐down (for eligibility, see inclusion and exclusion criteria).

Interventions

Intervention: hydrofluoroalkane beclomethasone 400 μg/day (i.e. < 50% dose reduction)

Comparison: chlorofluorocarbon beclomethasone 1000 μg/day.

Concomitant medications: not reported; likely that use of short‐acting bronchodilators as rescue medication was permitted

Excluded medications: none specified

Outcomes

Primary outcomes: Efficacy ‐ morning peak flow; Safety ‐ adverse events

Secondary outcomes: evening peak flow, FEV1, concentration of inhaled histamine causing a 20% decline in FEV1, frequency of β2‐agonist use, daily asthma symptom score (0 represents no symptoms; 5 represents severe symptoms); and sleep disturbance score. Safety ‐ oropharyngeal candidiasis; reported hoarseness; clinical laboratory tests (i.e. haematology, serum chemistry, urine analysis); and vital signs (i.e. sitting pulse rate, blood pressure, ECG)

Notes

Funding for trial: 3M Medica (Borken, Germany)

Notable conflicts of interest of trial authors: not reported; however, several study authors were employees of 3M Medica

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information provided

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was reported as double‐blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study was reported as double‐blind.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Outcome data appear complete. Data appear to be reported for all randomised participants.

Selective reporting (reporting bias)

High risk

Protocol was not available. Reporting of safety results appears to be fairly selective (SAEs not reported, details of individual AEs not reported).

Other bias

Low risk

None identified

Abbreviations: BDP, beclomethasone dipropionate; BID, twice daily; BTS, British Thoracic Society; CFC, chlorofluorocarbon; CIC, ciclesonide; ECG, electrocardiogram; FEV1, forced expiratory volume in 1 second; FP, fluticasone propionate; FVC, forced vital capacity; GOAL, Gaining Optimal Asthma Control study; ICS, inhaled corticosteroid; LABA, long‐acting beta agonist; NHS, National Health Service; OD, once daily; PC20, provocative concentration that produces a 20% reduction in FEV1 from baseline value; PEF, peak expiratory flow; PEFR, peak expiratory flow rate; pMDI, pressurised metered‐dose inhaler; QoL, quality of life; R&D, research and development; SD, standard deviation; SE, standard error; SFC, salmeterol formoterol combination; UK, United Kingdom.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Aalbers 2004

Wrong intervention

Adachi 2001

Wrong patient population

ALA 2007

Wrong intervention

Anonymous 1979

Wrong intervention

Baba 1999

Wrong study design

Baba 2000

Wrong intervention

Bateman 2005

Wrong comparator

Belda 2006

Wrong study design

Boulet 1990

Wrong route of administration

Brambilla 1994

Wrong comparator

Britton 1997

Wrong study design

Bruggenjurgen 2005

Wrong intervention

Busse 2003

Wrong study design

Campbell 1998

Wrong intervention

Casale 2003

Wrong intervention

Chanez 2001

Wrong intervention

Chiu 2011

Wrong intervention

Chung 2002

Wrong intervention

Davies 1977

Wrong intervention

Dorinsky 2003

Wrong intervention

Fardon 2005

Wrong intervention

Fardon 2007

Wrong intervention

FitzGerald 2003

Wrong intervention

FitzGerald 2005

Wrong intervention

FLIQ96 2005

Wrong study design

Fowler 2002

Wrong intervention

Haggart 2004

Wrong intervention

Hamada 2008

Wrong study design

Kardos 2001

Wrong intervention

Kawagishi 2000

Wrong study design

Keonig 2004

Wrong intervention

Massanari 2008

Wrong intervention

McKinlay 2011

Wrong intervention

Mikloweit 2000

Wrong study design

Obase 2013

Wrong intervention

Paggiaro 2011

Wrong comparator

Reddel 2007

Wrong intervention

Rumbak 1998

Wrong intervention

Schmier 2003

Wrong route of administration

Shamsul 2007

Wrong intervention

Ställberg 2003

Wrong intervention

Data and analyses

Open in table viewer
Comparison 1. ICS dose reduction versus no change in ICS dose (no concomitant LABA)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbation requiring OCS Show forest plot

2

261

Odds Ratio (M‐H, Random, 95% CI)

1.86 [0.16, 21.09]

Analysis 1.1

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 1 Exacerbation requiring OCS.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 1 Exacerbation requiring OCS.

2 Asthma control Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 2 Asthma control.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 2 Asthma control.

3 All‐cause SAEs Show forest plot

2

742

Odds Ratio (M‐H, Random, 95% CI)

1.24 [0.25, 6.25]

Analysis 1.3

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 3 All‐cause SAEs.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 3 All‐cause SAEs.

4 Steroid‐related AEs Show forest plot

2

261

Odds Ratio (M‐H, Random, 95% CI)

0.76 [0.16, 3.54]

Analysis 1.4

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 4 Steroid‐related AEs.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 4 Steroid‐related AEs.

5 Juniper AQLQ score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.5

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 5 Juniper AQLQ score (change from baseline).

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 5 Juniper AQLQ score (change from baseline).

6 Lung function, PEFR morning (L/min) Show forest plot

3

875

Mean Difference (IV, Random, 95% CI)

‐5.98 [‐19.47, 7.51]

Analysis 1.6

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 6 Lung function, PEFR morning (L/min).

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 6 Lung function, PEFR morning (L/min).

7 Lung function, FEV1 (L) Show forest plot

2

261

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.12, 0.08]

Analysis 1.7

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 7 Lung function, FEV1 (L).

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 7 Lung function, FEV1 (L).

Open in table viewer
Comparison 2. ICS dose reduction versus no change in ICS dose (concomitant LABA)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbation requiring OCS Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

1.31 [0.82, 2.08]

Analysis 2.1

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 1 Exacerbation requiring OCS.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 1 Exacerbation requiring OCS.

2 Asthma control (short asthma morbidity score), change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 2 Asthma control (short asthma morbidity score), change from baseline.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 2 Asthma control (short asthma morbidity score), change from baseline.

3 Asthma control (Asthma Severity Questionnaire) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 3 Asthma control (Asthma Severity Questionnaire).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 3 Asthma control (Asthma Severity Questionnaire).

4 All‐cause SAEs Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

0.60 [0.11, 3.33]

Analysis 2.4

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 4 All‐cause SAEs.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 4 All‐cause SAEs.

5 EuroQoL score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 5 EuroQoL score (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 5 EuroQoL score (change from baseline).

6 St. George's Respiratory Scale score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 6 St. George's Respiratory Scale score (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 6 St. George's Respiratory Scale score (change from baseline).

7 Lung function, PEFR morning (L/min) (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.7

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 7 Lung function, PEFR morning (L/min) (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 7 Lung function, PEFR morning (L/min) (change from baseline).

8 Lung function, reduction in FEV1 (% predicted, change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.8

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 8 Lung function, reduction in FEV1 (% predicted, change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 8 Lung function, reduction in FEV1 (% predicted, change from baseline).

9 Exacerbation requiring hospitalisation Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

4.06 [0.45, 36.86]

Analysis 2.9

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 9 Exacerbation requiring hospitalisation.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 9 Exacerbation requiring hospitalisation.

10 Exacerbation requiring ED visit Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.10

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 10 Exacerbation requiring ED visit.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 10 Exacerbation requiring ED visit.

11 Mortality Show forest plot

1

310

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 2.11

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 11 Mortality.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 11 Mortality.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.1 Exacerbation requiring OCS.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.1 Exacerbation requiring OCS.

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.1 Exacerbation requiring OCS.
Figuras y tablas -
Figure 5

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.1 Exacerbation requiring OCS.

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.3 All‐cause SAEs.
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.3 All‐cause SAEs.

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.4 All‐cause SAEs.
Figuras y tablas -
Figure 7

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.4 All‐cause SAEs.

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.4 Steroid‐related AEs.
Figuras y tablas -
Figure 8

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.4 Steroid‐related AEs.

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.7 Lung function, FEV1 (L).
Figuras y tablas -
Figure 9

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.7 Lung function, FEV1 (L).

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.6 Lung function, PEFR morning (L/min).
Figuras y tablas -
Figure 10

Forest plot of comparison: 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), outcome: 1.6 Lung function, PEFR morning (L/min).

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.9 Exacerbation requiring hospitalisation.
Figuras y tablas -
Figure 11

Forest plot of comparison: 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), outcome: 2.9 Exacerbation requiring hospitalisation.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 1 Exacerbation requiring OCS.
Figuras y tablas -
Analysis 1.1

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 1 Exacerbation requiring OCS.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 2 Asthma control.
Figuras y tablas -
Analysis 1.2

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 2 Asthma control.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 3 All‐cause SAEs.
Figuras y tablas -
Analysis 1.3

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 3 All‐cause SAEs.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 4 Steroid‐related AEs.
Figuras y tablas -
Analysis 1.4

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 4 Steroid‐related AEs.

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 5 Juniper AQLQ score (change from baseline).
Figuras y tablas -
Analysis 1.5

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 5 Juniper AQLQ score (change from baseline).

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 6 Lung function, PEFR morning (L/min).
Figuras y tablas -
Analysis 1.6

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 6 Lung function, PEFR morning (L/min).

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 7 Lung function, FEV1 (L).
Figuras y tablas -
Analysis 1.7

Comparison 1 ICS dose reduction versus no change in ICS dose (no concomitant LABA), Outcome 7 Lung function, FEV1 (L).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 1 Exacerbation requiring OCS.
Figuras y tablas -
Analysis 2.1

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 1 Exacerbation requiring OCS.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 2 Asthma control (short asthma morbidity score), change from baseline.
Figuras y tablas -
Analysis 2.2

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 2 Asthma control (short asthma morbidity score), change from baseline.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 3 Asthma control (Asthma Severity Questionnaire).
Figuras y tablas -
Analysis 2.3

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 3 Asthma control (Asthma Severity Questionnaire).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 4 All‐cause SAEs.
Figuras y tablas -
Analysis 2.4

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 4 All‐cause SAEs.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 5 EuroQoL score (change from baseline).
Figuras y tablas -
Analysis 2.5

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 5 EuroQoL score (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 6 St. George's Respiratory Scale score (change from baseline).
Figuras y tablas -
Analysis 2.6

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 6 St. George's Respiratory Scale score (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 7 Lung function, PEFR morning (L/min) (change from baseline).
Figuras y tablas -
Analysis 2.7

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 7 Lung function, PEFR morning (L/min) (change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 8 Lung function, reduction in FEV1 (% predicted, change from baseline).
Figuras y tablas -
Analysis 2.8

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 8 Lung function, reduction in FEV1 (% predicted, change from baseline).

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 9 Exacerbation requiring hospitalisation.
Figuras y tablas -
Analysis 2.9

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 9 Exacerbation requiring hospitalisation.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 10 Exacerbation requiring ED visit.
Figuras y tablas -
Analysis 2.10

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 10 Exacerbation requiring ED visit.

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 11 Mortality.
Figuras y tablas -
Analysis 2.11

Comparison 2 ICS dose reduction versus no change in ICS dose (concomitant LABA), Outcome 11 Mortality.

Summary of findings for the main comparison. ICS dose reduction compared with no change in ICS dose (no concomitant LABA) for adults with asthma

ICS dose reduction compared with no change in ICS dose (no concomitant LABA) for adults with asthma

Patient or population: adults with asthma
Setting: primary care and specialist centres
Intervention: ICS dose reduction
Comparison: no change in ICS dose (no concomitant LABA)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with no change in ICS dose (no concomitant LABA)

Risk with ICS dose reduction

Exacerbation requiring OCS
Follow‐up: range 10 weeks to 12 weeks

8 per 1000

14 per 1000
(1 to 140)

OR 1.86
(0.16 to 21.09)

261
(2 RCTs)

⊕⊝⊝⊝
Very lowa

No clear benefit or harm of stepping down the dose of ICS (very low‐quality evidence)

Asthma control
assessed by: Asthma Symptom Scale from: 0 (no symptoms) to 5 (severe symptoms)
Follow‐up: 10 weeks

Mean asthma control score in the no change in ICS dose group was 1.79.

MD 0.22 lower
(1.05 lower to 0.61 higher)

150
(1 RCT)

⊕⊕⊝⊝
Lowb

No clear benefit or harm of stepping down the dose of ICS (low‐quality evidence)

All‐cause SAEs
Follow‐up: mean 12 weeks

8 per 1000

9 per 1000
(2 to 45)

OR 1.24
(0.25 to 6.25)

742
(2 RCTs)

⊕⊕⊝⊝
Lowc

No clear benefit or harm of stepping down the dose of ICS (low‐quality evidence)

Steroid‐related AEs
Follow‐up: range 10 weeks to 12 weeks

31 per 1000

23 per 1000
(5 to 100)

OR 0.76
(0.16 to 3.54)

261
(2 RCTs)

⊕⊝⊝⊝
Very lowd

No clear benefit or harm of stepping down the dose of ICS (very low‐quality evidence)

Health‐related quality of life (change from baseline)
assessed by: AQLQ
Follow‐up: 12 weeks

Mean change from baseline in health‐related quality of life for the no change in ICS dose group was 0.02.

MD 0.21 lower
(0.33 lower to 0.09 lower)

554
(1 RCT)

⊕⊝⊝⊝
Very lowe

No clear benefit or harm of stepping down the dose of ICS (very low‐quality evidence); MCID is 0.5 for AQLQ

Lung function, FEV1 (L)
assessed by: spirometry
Follow‐up: range 10 weeks to 12 weeks

Mean FEV1 in the no change in ICS dose group was 3.15 litres.

MD 0.02 litres lower
(0.12 lower to 0.08 higher)

261
(2 RCTs)

⊕⊕⊝⊝
Lowf

No clear benefit or harm of stepping down the dose of ICS (low‐quality evidence)

Exacerbations requiring hospitalisation ‐ not reported

Outcome not reported by included studies

*Risk in the intervention group (and its 95% confidence interval) is based on assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
aThe quality of the evidence was downgraded once for indirectness (included studies were performed at specialist centres) and twice for imprecision (no events reported by Magnussen 2000; confidence intervals include null effect and appreciable benefit or harm).

bThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for indirectness (single study representative of one setting and drug regimen).

cThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for imprecision (confidence intervals include null effect and appreciable benefit or harm).

dThe quality of the evidence was downgraded once for risk of bias (selective reporting), once for indirectness (representative of specialist centres) and once for imprecision (confidence intervals include null effect and appreciable benefit or harm).

eThe quality of the evidence was downgraded twice for risk of bias (selective reporting and lack of blinding (subjective outcome)) and once for indirectness (single study representative of one setting and drug regimen).

fThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for imprecision (confidence intervals include null effect and appreciable benefit or harm).
AE, adverse event; AQLQ, Asthma Quality of Life Questionnaire; CI, confidence interval; FEV1, forced expiratory volume in one second; GRADE, Grades of Recommendation, Assessment, Development and Evaluation; ICS, inhaled corticosteroid; LABA, long‐acting beta agonist; MCID, minimum clinically important difference; MD, mean difference; OCS, oral corticosteroid; OR, odds ratio; RCT, randomised controlled trial; RR, risk ratio; SAE, serious adverse event.

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to the estimate of effect.
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of effect but may be substantially different.
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of effect.
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

Figuras y tablas -
Summary of findings for the main comparison. ICS dose reduction compared with no change in ICS dose (no concomitant LABA) for adults with asthma
Summary of findings 2. ICS dose reduction compared with no change in ICS dose (concomitant LABA) for adults with asthma

ICS dose reduction compared with no change in ICS dose (concomitant LABA) for adults with asthma

Patient or population: adults with asthma
Setting: primary and secondary care
Intervention: ICS dose reduction
Comparison: no change in ICS dose (concomitant LABA)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with no change in ICS dose (concomitant LABA)

Risk with ICS dose reduction

Exacerbation requiring OCS
Follow‐up: range 4 months to 12 months

148 per 1000

186 per 1000
(125 to 266)

OR 1.31
(0.82 to 2.08)

569
(2 RCTs)

⊕⊕⊝⊝
Lowa

No clear benefit or harm of stepping down the dose of ICS with respect to exacerbations requiring OCS (low‐quality evidence)

Asthma control (short asthma morbidity score)
Follow‐up: 12 months

Mean asthma control score was 1.43.

MD 0.16 higher
(0.34 lower to 0.66 higher)

242
(1 RCT)

⊕⊕⊝⊝
Lowb

No clear benefit or harm of stepping down the dose of ICS with respect to asthma control (low‐quality evidence)

All‐cause SAEs
Follow‐up: range 4 months to 12 months

35 per 1000

22 per 1000
(4 to 109)

OR 0.60
(0.11 to 3.33)

569
(2 RCTs)

⊕⊕⊝⊝
Lowa

No clear benefit or harm of stepping down the dose of ICS with respect to all‐cause SAEs (low‐quality evidence)

Steroid‐related AEs ‐ not reported

St. George's Respiratory Scale score (change from baseline)
Follow‐up: 12 months

Score 0‐100. 100 = greatest impact of chest disease on life; MCID is 4 units.

Mean change from baseline in HRQoL score was 7.4.c

MD 0.13 higher
(2.8 lower to 3.06 higher)

229
(1 RCT)

⊕⊕⊝⊝
Lowb

No clear benefit or harm of stepping down the dose of ICS with respect to HRQoL (low‐quality evidence)

Exacerbation requiring hospitalisation
Follow‐up: range 4 months to 12 months

4 per 1000

14 per 1000
(2 to 116)

OR 4.06
(0.45 to 36.86)

569
(2 RCTs)

⊕⊕⊝⊝
Lowd

No clear benefit or harm of stepping down the dose of ICS with respect to exacerbations requiring hospitalisation (low‐quality evidence)

Lung function, reduction in FEV1 (% predicted, change from baseline)
Follow‐up: 3 months

Mean change from baseline in % predicted FEV1 was ‐0.75%.

MD 2.45 lower
(8.88 lower to 3.98 higher)

14
(1 RCT)

⊕⊝⊝⊝
Very lowe

No clear benefit or harm of stepping down the dose of ICS with respect to lung function (very low‐quality evidence)

*Risk in the intervention group (and its 95% confidence interval) is based on assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
aThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for imprecision (confidence intervals include null effect and appreciable benefit or harm).

bThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for indirectness (single study representative of one setting and drug regimen).

cNote that study authors reported the change to the lowest SGRQ score during follow‐up.

dThe quality of the evidence was downgraded once for risk of bias (selective reporting) and once for imprecision (confidence intervals include null effect and appreciable benefit or harm).

eThe quality of the evidence was downgraded once for risk of bias (selective reporting), once for indirectness (single study representative of one setting or drug regimen) and once for imprecision (wide CI).

AE, adverse event; CI, confidence interval; FEV1, forced expiratory volume in one second; GRADE, Grades of Recommendation, Assessment, Development and Evaluation; HRQoL, health‐related quality of life; ICS, inhaled corticosteroid; LABA, long‐acting beta agonist; MCID, minimum clinically important difference; MD, mean difference; OCS, oral corticosteroid; OR, odds ratio; RCT, randomised controlled trial; RR, risk ratio; SAE, serious adverse event.

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

Figuras y tablas -
Summary of findings 2. ICS dose reduction compared with no change in ICS dose (concomitant LABA) for adults with asthma
Comparison 1. ICS dose reduction versus no change in ICS dose (no concomitant LABA)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbation requiring OCS Show forest plot

2

261

Odds Ratio (M‐H, Random, 95% CI)

1.86 [0.16, 21.09]

2 Asthma control Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 All‐cause SAEs Show forest plot

2

742

Odds Ratio (M‐H, Random, 95% CI)

1.24 [0.25, 6.25]

4 Steroid‐related AEs Show forest plot

2

261

Odds Ratio (M‐H, Random, 95% CI)

0.76 [0.16, 3.54]

5 Juniper AQLQ score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

6 Lung function, PEFR morning (L/min) Show forest plot

3

875

Mean Difference (IV, Random, 95% CI)

‐5.98 [‐19.47, 7.51]

7 Lung function, FEV1 (L) Show forest plot

2

261

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.12, 0.08]

Figuras y tablas -
Comparison 1. ICS dose reduction versus no change in ICS dose (no concomitant LABA)
Comparison 2. ICS dose reduction versus no change in ICS dose (concomitant LABA)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exacerbation requiring OCS Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

1.31 [0.82, 2.08]

2 Asthma control (short asthma morbidity score), change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Asthma control (Asthma Severity Questionnaire) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 All‐cause SAEs Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

0.60 [0.11, 3.33]

5 EuroQoL score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

6 St. George's Respiratory Scale score (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

7 Lung function, PEFR morning (L/min) (change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

8 Lung function, reduction in FEV1 (% predicted, change from baseline) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

9 Exacerbation requiring hospitalisation Show forest plot

2

569

Odds Ratio (M‐H, Random, 95% CI)

4.06 [0.45, 36.86]

10 Exacerbation requiring ED visit Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

11 Mortality Show forest plot

1

310

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 2. ICS dose reduction versus no change in ICS dose (concomitant LABA)