Scolaris Content Display Scolaris Content Display

Sulfato de magnesio inhalado en el tratamiento del asma aguda

Contraer todo Desplegar todo

Referencias

Abreu‐Gonzalez 2002 {published data only}

Abreu‐Gonzalez J, Rodríguez‐Díaz CY. Magnesium and bronchodilator effect of beta‐adrenergic. American Journal of Respiratory and Critical Care Medicine 2002;165:A185. CENTRAL

Aggarwal 2006 {published data only}

Aggarwal P, Dwivedi S, Handa R. Nebulized magnesium sulfate and salbutamol combination compared to salbutamol alone in the treatment of acute bronchial asthma: a randomized study. Annals of Emergency Medicine 2004;44(4 Suppl):S38. CENTRAL
Aggarwal P, Sharad S, Handa R, Dwiwedi SN, Irshad M. Comparison of nebulised magnesium sulphate and salbutamol combined with salbutamol alone in the treatment of acute bronchial asthma: a randomised study. Emergency Medicine Journal 2006;23(5):358‐62. CENTRAL

Ahmed 2013 {published data only}

Ahmed S, Sutradhar S, Miah A, Bari M, Hasan M, Alam M, et al. Comparison of salbutamol with normal saline and salbutamol with magnesium sulphate in the treatment of severe acute asthma. Mymensingh Medical Journal : MMJ 2013;22(1):1‐7. CENTRAL

Alansari 2015 {published data only}

Alansari K, Ahmed W, Davidson B, Alamri M, Zakaria I. Nebulized magnesium for moderate and severe pediatric asthma: a randomized trial. American Journal of Respiratory and Critical Care Medicine 2015;191:A2622. CENTRAL
Alansari K, Ahmed W, Davidson B, Alamri M, Zakaria I, Alrifaai M. Nebulized magnesium for moderate and severe pediatric asthma: a randomized trial. Pediatric Pulmonology 2015;50(12):1191‐9. CENTRAL
Rasheed H, Al‐Ansari K. Inhaled magnesium for moderate and severe paediatric asthma. Archives of Disease in Childhood 2014;99(Suppl 2):A101‐2. CENTRAL

Ashtekar 2008 {published data only}

Ashtekar CS, Powell C, Hood K, Doull I. Magnesium nebuliser trial (magnet): a randomised double‐blind placebo controlled pilot study in severe acute asthma. Archives of Disease in Childhood 2008;93:A100‐6. CENTRAL

Badawy 2014 {published data only}

Badawy M, Hasannen I. The value of magnesium sulfate nebulization in treatment of acute bronchial asthma during pregnancy. Egyptian Journal of Chest Diseases and Tuberculosis 2014;63(2):285‐9. CENTRAL
Badawy M, Hasannen I. The value of magnesium sulfate nebulization in treatment of acute bronchial asthma during pregnancy. European Respiratory Journal 2012;40(Suppl 56):1789. CENTRAL

Bessmertny 2002 {published data only}

Bessmertny O, DiGregorio RV, Cohen H, Becker E, Looney D, Golden J, et al. A randomized clinical trial of nebulized magnesium sulfate in addition to albuterol in the treatment of acute mild‐to‐moderate asthma exacerbations in adults. Annals of Emergency Medicine 2002;39(6):585‐91. CENTRAL

Dadhich 2005 {published data only}

Dadhich P, Tailor M, Gupta ML, Gupta R. Magnesium sulphate nebulisation in acute severe asthma [Abstract]. Indian Journal of Allergy, Asthma and Immunology 2005;19:117. CENTRAL
Dadhich P, Vats M, Lokendra D, Gupta RC, Gupta ML, Gupta N. Magnesium sulphate nebulization in acute severe asthma. Chest 2003;124(4):107S. CENTRAL

Drobina 2006 {published data only}

Drobina BJ, Kostic MA, Roos JA. Nebulized magnesium has no benefit in the treatment of acute asthma in the emergency department. Academic Emergency Medicine2006; Vol. 13, issue 5 Suppl. 1:S26. CENTRAL

Gallegos‐Solórzano 2010 {published data only}

Gallegos‐Solórzano MC, Pérez‐Padilla R, Hernández‐Zenteno RJ. Usefulness of inhaled magnesium sulfate in the coadjuvant management of severe asthma crisis in an emergency department. Pulmonary Pharmacology and Therapeutics 2010;23(5):432‐7. CENTRAL

Gaur 2008 {published data only}

Gaur SN, Singh A, Kumar R. Evaluating role of inhaled magnesium sulphate as an adjunct to salbutamol and ipratropium in severe acute asthma [Abstract]. Chest 2008;134(4_MeetingAbstracts):91003. [DOI: 10.1378/chest.134.4_MeetingAbstracts.p91003]CENTRAL

Goodacre 2013 {published data only}

Goodacre S. The 3Mg Study: A randomised trial of intravenous or nebulised magnesium sulphate versus placebo for acute severe asthma. ISCRTN Register2007. CENTRAL
Goodacre S, Bradburn M, Cohen J, Gray A, Benger J, Coats T. Prediction of unsuccessful treatment in patients with severe acute asthma. Emergency Medicine Journal 2014;31(e1):e40‐5. [DOI: 10.1136/emermed‐2013‐203046]CENTRAL
Goodacre S, Cohen J, Bradburn M, Gray A, Benger J, Coats T, 3Mg Research Team. Intravenous or nebulised magnesium sulphate versus standard therapy for severe acute asthma (3Mg trial): a double‐blind, randomised controlled trial. Lancet: Respiratory Medicine 2013;1(4):293‐300. [DOI: 10.1016/S2213‐2600(13)70070‐5]CENTRAL
Goodacre S, Cohen J, Bradburn M, Stevens J, Gray A, Benger J, et al. The 3Mg trial: a randomised controlled trial of intravenous or nebulised magnesium sulphate versus placebo in adults with acute severe asthma. Health Technology Assessment 2014;18(22):1‐168. [DOI: 10.3310/hta18220]CENTRAL
ISRCTN04417063. Magnesium sulphate for treatment of severe acute asthma. isrctn.com/ISRCTN04417063 (first received 26 February 2007). [DOI: 10.1186/ISRCTN04417063]CENTRAL

Hossein 2016 {published data only}

Hossein S, Pegah A, Davood F, Said A, Babak M, Mani M, et al. The effect of nebulized magnesium sulfate in the treatment of moderate to severe asthma attacks: a randomized clinical trial. American Journal of Emergency Medicine 2016;34(5):883‐6. CENTRAL

Hughes 2003 {published and unpublished data}

Hughes RJ, Goldkorn AL, Masoli M, Weatherall M, Brugess C, Beasley CRW. The use of nebulised isotonic magnesium sulphate as an adjuvant to salbutamol in the treatment of asthma in adults. Thorax 2002;57:iii10. CENTRAL
Hughes RJ, Goldkorn AL, Masoli M, Weatherall M, Burgess C, Beasley CR. Use of isotonic nebulized magnesium as an adjuvant to salbutamol in treatment of severe asthma in adults: randomised placebo‐controlled trial. Lancet 2003;361(9375):2114‐7. CENTRAL
Hughes RJ, Goldkorn AL, Masoli M, Weatherall M, Burgess C, Beasley R. The use of isotonic nebulised magnesium as an adjuvant to salbutamol in the treatment of severe asthma. American Journal of Respiratory and Critical Care Medicine2002; Vol. 165:A186. CENTRAL

Khashabi 2008 {published data only}

Khashabi J, Asadolahi S, Karamiyar M, Salari Lak S. Comparison of magnesium sulfate to normal saline as a vehicle for nebulized salbutamol in children with acute asthma: a clinical trial [Abstract]. European Respiratory Society 18th Annual Congress; 2008 Oct 3‐7; Berlin. 2008:[4597]. CENTRAL

Kokturk 2005 {published data only}

Kokturk N, Turktas H, Kara P, Mullaoglu S, Yilmaz F, Karamercan A. A randomized clinical trial of magnesium sulphate as a vehicle for nebulized salbutamol in the treatment of moderate to severe asthma attacks. Pulmonary Pharmacology and Therapeutics 2005;18(6):416‐21. CENTRAL
Turktas H, Kokturk N, Kara P, Mullaoglu SB, Yilmaz F, Karamercan A. A randomized clinical trial of magnesium sulphate as a vehicle for nebulized salbutamol in the treatment of moderate to severe asthma attacks [Abstract]. European Respiratory Journal2004; Vol. 24:343s. CENTRAL

Mahajan 2004 {published and unpublished data}

Mahajan P, Haritos D, Rosenberg N, Thomas R. Comparison of nebulized magnesium plus albuterol to nebulized albuterol plus saline in children with mild to moderate asthma. Journal of Emergency Medicine 2004;27(1):21‐5. CENTRAL

Mangat 1998 {published data only}

Mangat HS, D'Souza GA, Jacob MS. Nebulized magnesium sulphate versus nebulized salbutamol in acute bronchial asthma: a clinical trial. European Respiratory Journal 1998;12(2):341‐4. CENTRAL

Meral 1996 {published data only}

Meral A, Coker M, Tanac R. Inhalation therapy with magnesium sulfate and salbutamol in bronchial asthma. Turkish Journal of Pediatrics 1996;38(2):169‐75. CENTRAL

Mohammedzadeh 2014 {published data only}

Mohammedzadeh I, Mohammadi M, Khodabakhsh E. Effect of salbutamol and magnesium sulfate nebulizer compared with salbutamol and normal saline nebulizer in the treatment of acute asthma attack. Journal of Babol University of Medical Sciences 2014;16(3):7‐12. CENTRAL

Nannini 2000 {published data only}

Nannini LJ, Pendino JC, Corna RA, Mannarino S, Quispe R. Magnesium sulfate as a vehicle for nebulized salbutamol in acute asthma. American Journal of Medicine 2000;108(3):193‐7. CENTRAL

Neki 2006 {published data only}

Neki NS. A comparative clinical efficacy and safety profile of inhaled magnesium sulphate and salbutamol nebulisations in severe asthma [Abstract]. Indian Journal of Allergy, Asthma and Immunology 2006;20:131. CENTRAL

Powell 2013 {published data only}

Kolamunnage‐Dona R, Powell C, Williamson P. Modelling variable dropout in randomised controlled trials with longitudinal outcomes: application to the MAGNETIC study. Trials 2016;17(1):222. [DOI: 10.1186/s13063‐016‐1342‐0]CENTRAL
Petrou S, Boland A, Khan K, Powell C, Kolamunnage‐Dona R, Lowe J, et al. Economic evaluation of nebulized magnesium sulphate in acute severe asthma in children. International Journal of Technology Assessment in Health Care 2014;30(4):354‐60. [DOI: 10.1017/S0266462314000440]CENTRAL
Powell C. MAGNETIC; a randomised, double blind, placebo controlled study of nebulised magnesium sulphate in acute asthma in children [Abstract]. European Respiratory Journal. 2011; Vol. 38, issue Suppl 55:1406. []CENTRAL
Powell C, Kolamunnage‐Dona R, Lowe J, Boland A, Petrou S, Doull I, et al. MAGNEsium Trial In Children (MAGNETIC): a randomised, placebo‐controlled trial and economic evaluation of nebulised magnesium sulphate in acute severe asthma in children. Health Technology Assessment 2013;17(45):1‐216. [DOI: 10.3310/hta17450]CENTRAL
Powell C, Kolamunnage‐Dona R, Lowe J, Boland A, Petrou S, Doull I, et al. MAGNEsium trial in children (MAGNETIC): a randomised, placebo controlled trial and economic evaluation of nebulised magnesium sulphate in acute severe asthma in children. Health Technology Assessment 2013;17(45):1‐216. []CENTRAL
Powell C, Kolamunnage‐Dona R, Lowe J, Boland A, Petrou S, Doull I, et al. Magnesium sulphate in acute severe asthma in children (MAGNETIC): a randomised, placebo‐controlled trial. Lancet Respiratory Medicine 2013;1(4):301‐8. CENTRAL
Powell CVE. A randomised double, blind , placebo controlled study of nebulised magnesium sulphate in acute asthma in children –The MAGNETIC study. Archives of Disease in Children 2012;97:A2‐3. [DOI: 10.1136/archdischild‐2012‐301885.6]CENTRAL

Sarhan 2016 {published data only}

Sarhan H, El‐Garhy O, Ali M, Youssef N. The efficacy of nebulized magnesium sulfate alone and in combination with salbutamol in acute asthma. Drug Design, Development and Therapy 2016;10:1927‐33. [DOI: 10.2147/DDDT.S103147]CENTRAL

Turker 2017 {published data only}

Turker S, Dogru M, Yildiz F, Yilmaz SB. The effect of nebulised magnesium sulphate in the management of childhood moderate asthma exacerbations as adjuvant treatment. Allergologia et immunopathologia 2017;45(2):115‐20. CENTRAL

Balter 1989 {published data only}

Balter MS, Rebuck AS. Treatment of the recalcitrant asthmatic. Annals of Allergy 1989;63:297‐300. CENTRAL

Bede 2003 {published data only}

Bede O, Suranyi A, Pinter K, Szlavik M, Gyurkovits K. Urinary magnesium excretion in asthmatic children receiving magnesium supplementation: a randomized, placebo‐controlled, double‐blind study. Magnesium Research 2003;16:262‐70. CENTRAL

Bede 2004 {published data only}

Bede O, Suranyi A, Pinter K, Szlavik M, Gyurkovits K. Efficacy of long‐lasting magnesium supplementation in asthmatic children [Abstract]. European Respiratory Journal 2004;24:212s. CENTRAL

Bede 2008 {published data only}

Bede O, Nagy D, Suranyi A, Horvath I, Szlavik M, Gyurkovits K. Effects of magnesium supplementation on the glutathione redox system in atopic asthmatic children. Inflammation Research 2008;57:279‐86. CENTRAL

Bernstein 1995 {published data only}

Bernstein WK, Khasgir T, Khastgir A, Hernandex E, Miller J, Schonfeld SA, et al. Lack of effectiveness of magnesium in chronic stable asthma. Archives of Internal Medicine 1995;155:271‐6. CENTRAL

Cairns 1996 {published data only}

Cairns CB, Kraft M. Magnesium attenuates the neutrophil respiratory burst in adult asthmatic patients. Academic Emergency Medicine 1996;3(12):1093‐7. CENTRAL

Castillo Rueda 1991 {published data only}

del Castillo Rueda A, Recarte Garcia‐Andrade C, Torres Segovia FJ. Magnesium, the 4th drug in asthma treatment?. Revista Clinica Espanola 1991;189(5):250. CENTRAL

Chande 1992 {published data only}

Chande VT, Skoner DP. A trial of nebulized magnesium sulfate to reverse bronchospasm in asthmatic patients. Annals of Emergency Medicine 1992;21(9):1111‐5. CENTRAL

Corbridge 1995 {published data only}

Corbridge TC, Hall JB. The assessment and management of adults with status‐asthmaticus. American Journal of Respiratory and Critical Care Medicine 1995;151(5):1296‐316. CENTRAL

DiGregorio 1999 {published data only}

DiGregorio RV, Bessmertny O, Pringle G, Cohen H. Effect of nebulized magnesium sulfate on serum magnesium levels in asthmatic patients. ASHP Midyear Clinical Meeting1999; Vol. 34:[P‐383E]. CENTRAL

Emelyanov 1990 {published data only}

Emelyanov AV, Fedoseev GB, Emanuel VL, Sinitsina TM, Krunchak LV. Effect of magnesium‐sulfate aerosol on external respiration in bronchial‐asthma patients. Klinicheskaya Meditsina 1990;68(11):31‐4. CENTRAL

Emelyanov 1996 {published data only}

Emelyanova AV, Goncharova VA, Sinitsina TM. Magnesium sulfate in management of bronchial asthma. Klinicheskaya Meditsina (Mosk) 1996;74(8):55‐8. CENTRAL

Emelyanov 1997 {published data only}

Emelyanov AV, Goncharova VA, Sinitsyna TM. Magnesium sulphate aerosol efficacy in bronchial asthma. Terapevticheskii Arkhiv 1997;69(3):35‐9. CENTRAL

Fathi 2014 {published data only}

Fathi N, Hosseini SA, Tavakkol H, Khodadady A, Tabesh H. Effect of oral magnesium citrate supplement on lung function and magnesium level in patients with asthma. [Persian]. Journal of Mazandaran University of Medical Sciences 2014;24(111):43‐51. CENTRAL

Fedoseev 1991 {published data only}

Fedoseev GB, Emelyanov AV, Malakauskas KK, Goncharova V, Sinitsina TM, Didur MD, et al. Therapeutic potentialities of magnesium‐sulfate in bronchial‐asthma. Terapevticheskii Arkhiv 1991;63(12):27‐9. CENTRAL

Gandia 2012 {published data only}

Gandia F, Guenard H, Sriha B, Tabka Z, Rouatbi S. Inhaled magnesium sulphate in the treatment of bronchial hyperresponsiveness. Magnesium Research 2012;25(4):168‐76. CENTRAL

Gurkan 1999 {published data only}

Gurkan F, Hospolat K, Bosnak M, Dikici B, Derman O, Ece A. Intravenous magnesium sulphate in the management of moderate to severe acute asthmatic children nonresponding to conventional therapy. European Journal of Emergency Medicine 1999;6(3):201‐5. CENTRAL

Harari 1998 {published data only}

Harari M, Barzillai R, Shani J. Magnesium in the management of asthma: critical review of acute and chronic treatments, and Deutsches Medizinisches Zentrum's (DMZ's) clinical experience at the Dead Sea. Journal of Asthma 1998;35(7):525‐36. CENTRAL

Hardin 2001 {published data only}

Hardin KA, Kallas HJ, McDonald RJ. Pharmacologic management of the hospitalized pediatric asthma patient. Clinical Reviews in Allergy and Immunology 2001;20(3):293‐326. CENTRAL

Harmanci 1996 {published data only}

Harmanci E, Ekici M, Erginel S, Ozdemir N, Ozkan G. Comparison of effects of nebulized to intravenous magnesium sulfate on bronchial hyperreactivity and expiratory flow rate in asthmatic patients. European Respiratory Journal 1996;9 Suppl 23:34s. CENTRAL

Hill 1995 {published data only}

Hill J, Britton J. Dose‐response relationship and time‐course of the effect of inhaled magnesium sulphate on airflow in normal and asthmatic subjects. British Journal of Clinical Pharmacology 1995;40:539‐44. CENTRAL

Hill 1997a {published data only}

Hill J, Lewis S, Britton J. Studies of the effects of inhaled magnesium on airway reactivity to histamine and adenosine monophosphate in asthmatic subjects. Clinical and Experimental Allergy 1997;27(5):546‐51. CENTRAL

Hill 1997b {published data only}

Hill J, Lewis S, Britton J. Studies of the effects of inhaled magnesium on airway reactivity to histamine and adenosine monophosphate in asthmatic subjects. Clinical and Experimental Allergy1997; Vol. 27:546‐51. CENTRAL

Irazuzta 2014 {published data only}

Irazuzta J, Pavlovich V, Paredes F. High dose magnesium sulfate infusions in children with status asthmaticus in the emergency department, efficacy study. Pediatric Critical Care Medicine 2014;15(4):99. CENTRAL

Irazuzta 2016 {published data only}

Irazuzta JE, Paredes F, Pavlicich V, Dominguez SL. High‐dose magnesium sulfate infusion for severe asthma in the emergency department: efficacy study. Pediatric Critical Care Medicine 2016;17(2):e29‐e33. CENTRAL

Kenyon 2001 {published data only}

Kenyon N, Albertson TE. Status asthmaticus: From the emergency department to the intensive care unit. Clinical Reviews in Allergy and Immunology 2001;20(3):271‐92. CENTRAL

Kreutzer 2001 {published data only}

Kreutzer ML, Louie S. Pharmacologic treatment of the adult hospitalized asthma patient. Clinical Reviews in Allergy and Immunology 2001;20(3):357‐83. CENTRAL

Manzke 1990 {published data only}

Manzke H, Thiemeier M, Elster P, Lemke J. Magnesium sulfate as adjuvant in beta‐2‐sympathicomimetic inhalation therapy of bronchial asthma. Pneumologie 1990;44(10):1190‐2. CENTRAL

McFadden 1995 {published data only}

McFadden ER. Asthma. Lancet 1995;345(8959):1215‐20. CENTRAL

Nannini 1997 {published data only}

Nannini Jr LJ, Hofer D. Effect of inhaled magnesium sulfate on sodium metabisulfite‐induced bronchoconstriction in asthma. Chest 1997;111:858‐61. CENTRAL

Nunez‐Torres 1995 {published data only}

Nunez‐Torres C, Abreu Glez J, Glez Martin I, Martinez Riera A, Glez Reimers E, Santolaria FF. The magnesium sulphate does not have bronchodilator effect in the acute bronchial asthma when it is used by inhaled way [abstract]. European Respiratory Society Annual Congress; 1995 Oct 9‐13; Barcelona1995; Vol. 8:98S. CENTRAL

Pelton 1998 {published data only}

Pelton R. Nutrients can reduce asthma severity. American Druggist 1998;215(5):34, 36. CENTRAL

Pelton 1999 {published data only}

Pelton R. Don't forget magnesium. American Druggist 1999;December:48‐9. CENTRAL

Petrov 2014 {published data only}

Petrov VI, Shishimorov IN, Perminov AA, Nefedov IV. [Influence of magnesium deficiency correction on the effectiveness of bronchial asthma pharmacotherapy in children] [Russian]. Eksperimental'naia i klinicheskaia farmakologiia 2014;77(8):23‐7. CENTRAL

Puente‐Maestu 1999 {published data only}

Puente‐Maestu L, Abad YM. Treatment of asthmatic crisis. Revista Clinica Espanola 1999;199(7):473‐7. CENTRAL

Qureshi 1999 {published data only}

Qureshi F. Management of children with acute asthma in the emergency department. Pediatric Emergency Care 1999;15(3):206‐14. CENTRAL

Rodger 2003 {published data only}

Rodger KA, McLachlan J. The effects of oral magnesium in asthma [abstract]. American Thoracic Society 99th International Conference; 2003 May 16‐21; Seattle. 2003:C104 Poster D42. CENTRAL

Rodrigo 2000 {published data only}

Rodrigo G, Rodrigo C, Burschtin O. Efficacy of magnesium sulfate in acute adult asthma: a meta‐analysis of randomized trials. American Journal of Emergency Medicine 2000;18(2):216‐21. CENTRAL

Rolla 1987a {published data only}

Rolla G, Bucca C, Bugiani M, Arossa W, Spinaci S. Reduction of histamine‐induced bronchoconstriction by magnesium in asthmatic subjects. Allergy 1987;42:186‐8. CENTRAL

Rolla 1987b {published data only}

Rolla G, Bucca C, Arossa W, Bugiani M. Magnesium attenuates methacholine‐induced bronchoconstriction in asthmatics. Magnesium 1987;6(4):201‐4. CENTRAL

Rolla 1988a {published data only}

Rolla G, Bucca C, Caria E. Dose‐related effect of inhaled magnesium‐sulfate on histamine bronchial challenge in asthmatics. Drugs under Experimental and Clinical Research 1988;14(9):609‐12. CENTRAL

Rolla 1988b {published data only}

Rolla G, Bucca C. Magnesium, beta‐agonists and asthma. Lancet 1988;April 30:989. CENTRAL

Scarfone 1998 {published data only}

Scarfone RJ, Loiselle JM, Joffe MD, Mull C, Stiller S, Thompson, et al. Magnesium sulfate in the emergency department treatment of acute asthma in children. Pediatrics 1998;102:711. CENTRAL

Scarfone 2000 {published data only}

Scarfone RJ, Lioselle JM, Joffe MD, Mull CC, Stiller S, Thompson K, et al. A randomized trial of magnesium in the emergency department treatment of children with asthma. Annals of Emergency Medicine 2000;36(6):572‐8. CENTRAL

Shishimorov 2015 {published data only}

Shishimorov I, Petrov V, Magnitskaya O, Perminov A, Nefedov I. Magnesium deficiency correction for improvement of children bronchial asthma treatment results. European Respiratory Journal 2015;46(0):PA1286. CENTRAL

Singh 2008a {published data only}

Singh AK, Gaur S, Kumar R. A randomized controlled trial of intravenous magnesium sulphate as an adjunct to standard therapy in acute severe asthma. Iranian Journal of Allergy, Asthma and Immunology2008; Vol. 7:221‐9. CENTRAL

Singh 2008b {published data only}

Singh AK, Gaur SN, Kumar R. Comparison of efficacy of intravenous and inhaled magnesium sulphate as an adjunct to standard therapy in acute severe asthma [Abstract]. European Respiratory Society 18th Annual Congress; 2008 Oct 3‐7; Berlin. 2008:[P3620]. CENTRAL

Singhi 2014 {published data only}

Singhi S, Grover S, Bansal A, Chopra K. Randomised comparison of intravenous magnesium sulphate, terbutaline and aminophylline for children with acute severe asthma. Acta Paediatrica 2014;103(12):1301‐6. CENTRAL

Sinitsina 1991 {published data only}

Sinitsina TM, Shchemelinina TI, Didur MD, Evsyukova EV, Emelyanov AV, Nazarova VA. The follow‐up of bronchial hyperreactivity in risk group subjects and bronchial‐asthma patients ‐ approaches to its correction. Terapevticheskii Arkhiv 1991;63(8):21‐5. CENTRAL

Skobeloff 1982 {published data only}

Skobeloff EM. An ion for the lungs. Academic Emergency Medicine 1982;3(12):1082‐4. CENTRAL

Sun 2014 {published data only}

Sun YX, Gong CH, Liu S, Yuan XP, Yin LJ, Yan L. Effect of inhaled MgSO4 on FEV1 and PEF in children with asthma induced by acetylcholine: a randomized controlled clinical trial of 330 cases. Journal of Tropical Pediatrics 2014;60(2):1268‐73. CENTRAL

Talukdar 2005 {published data only}

Talukdar T, Singhal P, Jain A, Kumar R, Gaur SN. Inhaled magnesium sulfate in the treatment of severe asthma. Indian Journal of Allergy Asthma and Immunology2005; Vol. 19:29‐35. CENTRAL

Teeter 1999 {published data only}

Teeter JG. Bronchodilator therapy in status asthmaticus. Chest 1999;115(4):911‐2. CENTRAL

Telia 2005 {published data only}

Telia A, Tutashvili M, Donguzashvili S, Pirtskhalava N. Effect of magnesium and furosemide on bronchial asthma. Georgian Medical News 2005, (128):55‐9. CENTRAL

Tereshchenko 2006 {published data only}

Tereshchenko S, Turgenkova N, Gorbacheva N, Procoptzeva N, Goncharov B, Vlasova M. Administration of isotonic nebulised magnesium sulphate as an adjuvant to ipratropium bromide in treatment of viral wheezing in children [Abstract]. European Respiratory Journal 2006;28:265s [P1537]. CENTRAL

Tetikkurt 1992 {published data only}

Tetikkurt C, Kocyigit E, Disci R. Bronchodilating effect of inhaled and intravenous magnesium‐sulfate (compared with aeresol terbutaline). Magnesium Bulletin 1992;14(2):49‐52. CENTRAL

Tetikkurt 1993 {published data only}

Tetikkurt C, Kocyigit E. The bronchodilating effect of inhaled magnesium. Magnesium Bulletin 1993;15(1):1‐2. CENTRAL

Torres 2012 {published data only}

Torres S, Sticco N, Bosch JJ, Lolster T, Siaba A, Rivarola M, et al. Effectiveness of magnesium sulfate as initial treatment of acute severe asthma in children, conducted in a tertiary‐level university hospital: a randomized, controlled trial. Archivos Argentinos de Pediatria 2012;110(4):291‐6. CENTRAL

Watanatham 2015 {published data only}

Watanatham S, Pongsamart G, Vangveeravong M, Daengsuwan T. Comparison efficacy and safety of inhaled magnesium sulfate to intravenous magnesium sulfate in childhood severe asthma exacerbation. Journal of Allergy and Clinical Immunology 2015;135(2):AB241. CENTRAL

Wijetunge 2002 {unpublished data only}

Wijietunge DB. A trial of nebulised magnesium sulphate versus placebo in addition to conventional bronchodilator treatment in acute asthma of moderate severity. webarchive.nationalarchives.gov.uk/20130506081956/http://www.hta.ac.uk/protocols/200600010002.pdf (accessed prior to 31 July 2017). CENTRAL

Wongwaree 2017 {published data only}

Wongwaree S. Comparison efficacy of randomized nebulized magnesium sulfate and ipratropium bromide/fenoterol in children with severe asthma exacerbation. Journal of allergy and clinical immunology 2017;139(2 Suppl 1):AB94. CENTRAL

Xu 2002 {published data only}

Xu CQ, Yang J, Meng XK. Clinical study of salbutamol combined with magnesium sulfate by nebulization in the treatment of paroxysmal asthma. Chinese Journal of Clinical Pharmacology and Therapeutics 2002;7:446‐8. CENTRAL

Yemelyanov 1997 {published data only}

Yemelyanov A, Fedoseev G, Gonocharova V, Sinitcina T, Lintsov A. The effects of magnesium sulfate aerosol on pulmonary function tests in asthmatic patients [abstract]. European Respiratory Journal 1997;10:472S. CENTRAL

Zandsteeg 2009 {published data only}

Zandsteeg AM, Hirmann P, Pasma HR, Yska JP, ten Brinke A. Effect of MgSO4 on fev1 in stable severe asthma patients with chronic airflow limitation. Magnesium Research 2009;22:256‐61. CENTRAL

Zhu 2003 {published data only}

Zhu BF, Tao YJ. 1,6‐Fructose diphosphate and magnesium sulfate therapy for 34 patients with severe asthma. Chinese Journal of Medical Writing 2003;10:888‐90. CENTRAL

Abd 1997 {published data only}

Abd El Kader F. Ventilatory, cardiovascular and metabolic responses to salbutamol, ipratropium bromide and magnesium sulfate in bronchial asthma: comparative study. Alexandria Medical Journal 1997;39(1):43‐64. CENTRAL

Bustamante 2000 {published data only}

Bustamante APL. Inhaled magnesium sulfate as adjunct therapy for moderate to severe asthma exacerbations, a randomized control clinical trial. Philippine Journal of Pediatrics 2000;49:237‐9. CENTRAL

ISRCTN61336225 {published data only}

Dawood M, Mahmoud LMZ. Magnesium sulfate in the treatment of acute asthma. ISRCTN registry2016. [http://www.isrctn.com/ISRCTN61336225]CENTRAL

Motamed 2015 {published data only}

IRCT2015111015446N8. Comparison of clinical and Spirometric response between nebulized salbutamol _ magnesium sulfate and nebulized salbutamol alone in acute asthma attack. en.search.irct.ir/view/26734 (first received 27 November 2015). CENTRAL

Saucedo 2015 {published data only}

NCT02584738. Nebulized magnesium sulfate as an adjunct to standard therapy in asthma exacerbation. clinicaltrials.gov/ct2/show/NCT02584738 (first received 14 October 2015). CENTRAL

Schuh 2016a {published data only}

Schuh S, Sweeney J, Freedman S, Coates A, Johnson D, Thompson G, et al. Magnesium nebulization utilization in management of pediatric asthma (MagNUM PA) trial: study protocol for a randomized controlled trial. Trials 2016;17:261. CENTRAL

Aburuz 2005

Aburuz S, McElnay J, Gamble J, Millership J, Heaney L. Relationship between lung function and asthma symptoms in patients with difficult to control asthma. Journal of Asthma 2005;42(10):859‐64.

Ashutosh 2000

Ashutosh K. Nitric oxide and asthma: a review. Current Opinion in Pulmonary Medicine 2000;6(1):21‐5.

Bain 2014

Bain E, Pierides KL, Clifton VL, Hodyl NA, Stark MJ, Crowther CA, et al. Interventions for managing asthma in pregnancy. Cochrane Database of Systematic Reviews 2014, Issue 10. [DOI: 10.1002/14651858.CD010660.pub2]

Blitz 2005

Blitz M, Blitz S, Hughes R, Diner B, Beasley R, Knopp J, et al. Aerosolized magnesium sulfate for acute asthma: a systematic review. Chest 2005;128(1):337‐44.

Bois 1962

Bois P. Effect of magnesium deficiency on mast cells and urinary histamine in rats. British Journal of Experimental Pathology 1963;44:151‐5.

BTS/SIGN 2016

SIGN 2016. Sign 153. British guideline on the management of asthma. A national clinical guideline. September 2016. brit‐thoracic.org.uk/document‐library/clinical‐information/asthma/btssign‐asthma‐guideline‐2016/ (accessed prior to 31 July 2017). [ISBN 9781909103474]

Carranza Rosenzweig 2004

Carranza Rosenzweig JR, Edwards L, Lincourt W, Dorinsky P, ZuWallack RL. The relationship between health‐related quality of life, lung function and daily symptoms in patients with persistent asthma. Respiratory Medicine 2004;98(12):1157‐65.

Cates 2004

Cates CCJ, Bara A, Crilly JA, Rowe BH. Holding chambers versus nebulisers for beta‐agonist treatment of acute asthma. Cochrane Database of Systematic Reviews 2004, Issue 1. [DOI: 10.1002/14651858.CD000052]

Del Castillo 1954

Del Castillo J, Engbaek L. The nature of the neuromuscular block produced by magnesium. Journal of Physiology 1954;124:370‐84.

GINA 2017

Global Initiative for Asthma (GINA). Global Strategy for Asthma Management and Prevention 2017. ginasthma.org (accessed prior to 31 July 2017).

Gourgoulianis 2001

Gourgoulianis KI, Chatziparasidis G, Chatziefthimiou A, Molyvdas PA. Magnesium as a relaxing factor of airway smooth muscles. Journal of Aerosol Medicine 2001;14(3):301‐7.

GRADEpro GDT [Computer program]

GRADE Working Group, McMaster University. GRADEpro GDT. Version accessed 8 May 2017. Hamilton (ON): GRADE Working Group, McMaster University, 2014.

Griffiths 2016

Griffiths B, Kew KM. Intravenous magnesium sulfate for treating children with acute asthma in the emergency department. Cochrane Database of Systematic Reviews 2016, Issue 4. [DOI: 10.1002/14651858.CD011050.pub2]

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. Available from handbook.cochrane.org.

Kew 2014

Kew K, Kirtchuk L, Michell C. Intravenous magnesium sulfate for treating adults with acute asthma in the emergency department. Cochrane Database of Systematic Reviews 2014, Issue 5. [DOI: 10.1002/14651858.CD010909.pub2]

Ling 2016

Ling ZG, Wu YB, Kong JL, Tang ZM, Liu W, Chen YQ. Lack of efficacy of nebulized magnesium sulfate in treating adult asthma: A meta‐analysis of randomized controlled trials. Pulmonary Pharmacology and Therapeutics 2016;41:40‐7.

Mohammed 2007

Mohammed S, Goodacre S. Intravenous and nebulised magnesium sulphate for acute asthma: systematic review and meta‐analysis. Emergency Medicine Journal 2007;24(12):823‐30.

Nadler 1987

Nadler J, Goodson S, Rude R. Evidence that prostacyclin mediates the vascular action of magnesium in humans. Hypertension 1987;9(4):379‐83.

NRAD 2014

Royal College of Physicians. Why asthma still kills: the national review of asthma deaths (NRAD) confidential enquiry report. rcplondon.ac.uk/projects/outputs/why‐asthma‐still‐kills. [ISBN 978‐1‐86016‐531‐3]

Powell 2012

Powell C, Dwan K, Milan S, Beasley R, Hughes R, Knopp‐Sihota J, et al. Inhaled magnesium sulfate in the treatment of acute asthma. Cochrane Database of Systematic Reviews 2012, Issue 12. [DOI: 10.1002/14651858.CD003898.pub5]

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Shan 2013

Shan Z, Rong Y, Yang W, Wang D, Yao P, Xie J, et al. Intravenous and nebulized magnesium sulfate for treating acute asthma in adults and children: A systematic review and meta‐analysis. Respiratory Medicine 2013;107(3):321‐30. [DOI: 10.1016/j.rmed.2012.12.001]

Song 2012

Song WJ, Chang YS. Magnesium sulfate for acute asthma in adults: a systematic literature review. Asia Pacific Allergy 2012;2(1):76‐85.

Su 2017

Su Z, Li R, Gai Z. Intravenous and nebulized magnesium sulfate for treating acute asthma in children: a systematic review and meta‐analysis. journals.lww.com/pec‐online/Abstract/publishahead/Intravenous_and_Nebulized_Magnesium_Sulfate_for.98881.aspx 2016 Oct 4 [Epub ahead of print].

Turner 2017

Turner DL, Ford WR, Kidd EJ, Broadley KJ, Powell C. Effects of nebulised magnesium sulphate on inflammation and function of the guinea‐pig airway. European Journal of Pharmacology 2017;801:79‐85.

Blitz 2004

Blitz M, Blitz S, Beasely R, Diner BM, Hughes R, Knopp JA, et al. Inhaled magnesium sulfate in the treatment of acute asthma. Cochrane Database of Systematic Reviews 2004, Issue 3. [DOI: 10.1002/14651858.CD003898.pub2]

Blitz 2005a

Blitz M, Blitz S, Beasely R, Diner BM, Hughes R, Knopp JA, et al. Inhaled magnesium sulfate in the treatment of acute asthma. Cochrane Database of Systematic Reviews 2005, Issue 3. [DOI: 10.1002/14651858.CD003898.pub3]

Blitz 2005b

Blitz M, Blitz S, Beasely R, Diner B, Hughes R, Knopp JA, et al. Inhaled magnesium sulfate in the treatment of acute asthma. Cochrane Database of Systematic Reviews 2005, Issue 4. [DOI: 10.1002/14651858.CD003898.pub4]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abreu‐Gonzalez 2002

Methods

Randomised, controlled, double blind study, 2 groups.

1 centre in Tenerife.

Participants

24 patients (Intervention 13, Control 11), adults, acute asthma, moderate obstruction.

Interventions

Intervention: 2 mL of MgSO₄ (isotonic) dose and 400 mcg of salbutamol (delivery probably by MDI).

Control: 2 mL of a physiological serum of an inhaled form, 400 mcg of salbutamol (delivery probably by MDI).

Nebuliser: no details.

Outcomes

FEV1 and PEF at 0, 15, 30 45 minutes.

Notes

Funding: Gobierno Autonomo Canarias.

Abstract only.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details but stated as "randomised".

Allocation concealment (selection bias)

Unclear risk

No details.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double blind.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No details.

Selective reporting (reporting bias)

Unclear risk

Abstract only and not all time points reported.

Aggarwal 2006

Methods

Double blind, randomised controlled trial, parallel.

1 emergency department in India.

Participants

Inclusion criteria: participants aged 13 to 60, BTS definition acute asthma (PEF and clinical features).

Exclusion criteria: first episode of wheeze, chronic bronchitis or emphysema, heart failure, angina, renal failure, temperature > 38 ºC,  ET tube required, no consent, pregnancy, failure to do peak flow.

Intervention: 50 randomised.

Mean age (years): 46.26 (13.96). 

Men:women: 27:23.

Acute severe: 29.

Acute life threatening: 21.

Smokers: 9.

Baseline PEF: 118.6 (41.3).

Duration of attack; days (SD) 4.16 (1.69).

Control: 50 randomised.

Mean age (years): 41.00 (16.66).

Men: women: 33:17.

Acute severe: 30.

Severe life threatening: 20.

Smokers: 5.

Baseline PEF: 111.6 (43.3).

Duration of attack; days (SD) 4.28 (1.99).

Interventions

Intervention: MgSO₄ (1 mL of 500 mg/mL MgSO₄) and salbutamol (1 mL of salbutamol) 8 mL distilled water – 295 mOsmol/kg ×3 in an hour.

Control: salbutamol 1 mL, 1.5 mL distilled water, 7.5 mL normal saline – 287 mOsmol/kg ×3 in an hour.

Treatment over 1 h; 3 nebulisers 20 minutes apart.  Follow‐up for 20 minutes. 

Ultrasonic nebuliser.

Outcomes

PEF, heart rate, systolic pressure, diastolic pressure, time in ED, blood gases (O₂ and CO₂— 0 and 120 minutes), magnesium levels (0 and 120 minutes).

Time points 0, 15, 60, 75, 120 minutes.

Notes

Funding: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number tables.

Allocation concealment (selection bias)

Low risk

Separate envelopes to ensure concealment until inclusion (where they were kept and whether tamper proof — not mentioned).

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The 2 researchers were blinded to the treatments so measurements (normal clinical outcomes) remained blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

50 participants both sides at beginning and 50 participants both sides completed the study with full outcome data.

Selective reporting (reporting bias)

Low risk

Follow‐up data and longer‐term outcome data not collected. No apparent indication of selective reporting.

Ahmed 2013

Methods

Randomised open controlled trial.

1 hospital in Bangladesh.

Participants

Inclusion criteria: severe acute asthma.

Exclusion criteria: none stated.

120 randomised.

Intervention: 60 randomised.

Control: 60 randomised.

Interventions

Intervention: salbutamol with MgSO₄.

Control: salbutamol with normal saline.

Outcomes

PEF, respiratory rate, pulse rate, systolic, diastolic blood pressure, adverse effects.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details but states randomized.

Allocation concealment (selection bias)

Unclear risk

No details given.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label trial.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label trial.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not clear how many participants completed the trial or if any were excluded from analysis.

Selective reporting (reporting bias)

Unclear risk

Conference abstract. no prospective trial registration identified, no outcome measures pre‐specified.

Alansari 2015

Methods

Double‐blind, randomised controlled trial.

1 Paediatric emergency centre, Qatar.

Participants

Inclusion criteria: moderate/severe asthma exacerbation, age 2‐14 years, previous diagnosis of asthma.

Exclusion criteria: prematurity, critical illness needing ICU admission for IV bronchodilator, NIV or invasive ventilation, transfer to other institution, history of hypersensitivity to MgSO₄, history of neuromuscular/cardiac/renal disease, underlying structural lung disease, received systemic steroid/theophylline/ipratropium in prior 72 h, consolidation on chest XR, received IV MgSO₄ before randomisation, prior participation in the study, haemodynamic instability.

Number randomised: 400.

Intervention: 208 randomised.

Mean age (years): 5.6 (3.1).

Male:female: 133:75.

Moderate:severe: 168:40.

Mean baseline asthma severity score: 7.6 (1.3).

Control: 192 randomised.

Mean age (years): 5.8 (3.1).

Male:female: 115:77.

Moderate:severe: 163:29.

Mean baseline asthma severity score: 7.5 (1.3).

Interventions

Intervention: 800 mg MgSO₄ (15 mL).

Control: 15 mL 0.9% NaCl.

Medication divided into 3 doses over 1 h.

Jet nebuliser.

Outcomes

Time to medical readiness for discharge, mean asthma severity score (4, 8, 12, 24, 36, 48 h), mean asthma severity score at discharge, need for revisit or readmission (2 weeks).

Adverse events: chest tightness and facial rash (1; intervention group). Excessive cough (1; control group). ICU admission (1; control group).

Notes

Funding: Hamad Medical Corporation; Number: 12095/12

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation without blocks.

Allocation concealment (selection bias)

Low risk

Randomisation list provided to pharmacy resulted in preparation of identical‐appearing sealed numbered vials.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All study personnel were blinded to treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All study personnel were blinded to treatment. The paper was not explicit re. outcome assessors ‒ they were assumed to also have been blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Over 90% completed the trial in both arms. Balanced number were excluded from each arm, with reasons given.

Selective reporting (reporting bias)

Low risk

Prospective trial registration. All listed outcomes are reported.

Ashtekar 2008

Methods

Parallel

1 Children’s Assessment Unit, 1 hospital (UHW).

Participants

Inclusion criteria: age range 2 to 16 years, acute severe asthma.

Exclusion criteria: chronic lung disease, congenital heart disease, unable to understand English.

17 randomised (8 boys).

Intervention: 7 completed.

Control: 10 completed.

Interventions

Intervention: 2.5 mL isotonic MgSO₄ (3 occasions at 20‐minute intervals), salbutamol and ipratropium bromide.

Control: 2.5 mL isotonic saline (3 occasions at 20‐minute intervals), salbutamol and ipratropium bromide.

3 dosages over 1 h: follow‐up for 240 minutes.

Outcomes

Asthma severity scores (ASS), the sum of wheeze, accessory muscle use and heart rate, were computed on 6 occasions over 4 h. The primary endpoint was the area under the curve of the ASS at the 6 time points for each child.

Notes

Funding: local R and D pilot funding.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation by pharmacy at source ‒ in ED as sequential vials (code in pharmacy).

Allocation concealment (selection bias)

Low risk

As above – absolute concealment.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial described as double blind: as above.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trial described as double blind: as above.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All data collected for the 17 patients.

Selective reporting (reporting bias)

Unclear risk

Abstract only. Outcomes partially reported.

Badawy 2014

Methods

Randomised controlled trial.

Outpatient department and Emergency department from 1 hospital, Egypt.

Participants

Inclusion criteria: pregnancy, acute exacerbation of asthma partially or not completely controlled on routine acute asthma therapy.

Exclusion criteria: congestive heart failure, history of angina, renal problems, history suggestive of pulmonary oedema, very severe asthma (altered consciousness, respiratory acidosis, needing intubation, arrest), any associated medical illness e.g. diabetes/hypertension, fever > 38°C, inability to perform PEF.

Number randomised: 60.

All participants female.

Intervention: 30 randomised.

Mean age (years): 25.7 (3.8).

Control: 30 randomised.

Mean age (years): 25.9 (4.0).

Interventions

Intervention: 500 mg (1 mL) MgSO₄ with 1 mL salbutamol solution and 8 mL 0.9% NaCl.

Control: 1 mL salbutamol solution with 9 mL 0.9% NaCl.

Treatments given over 8 minutes; max 3 sets of nebulisation 20 minutes apart.

Outcomes

PEF, FEV1, FVC, FEV1/FVC ratio, FEF 25‐75%, arterial blood pCO2, pO2 and pH, oxygen saturations, serum potassium. Recorded at end of therapy ‒ assumed to be 2 h from baseline.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Participants were randomly classified into groups comparable in socio‐demographic criteria, but no indication is given of random sequence generation. Baseline clinical characteristics are given, and there is no indication that the groups were balanced with regard to clinical criteria.

Allocation concealment (selection bias)

Unclear risk

Participants were randomized into 2 groups through sealed opaque envelopes, but no indication is given whether participants or research personnel were aware of group allocation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Participants were randomized into 2 groups through sealed opaque envelopes, but no mention of procedures to blind personnel.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No mention of procedures to blind personnel.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

On further correspondence, appropriate exclusion criteria were applied but no indication given whether excluded participants were balanced across groups, and no dropout data were given.

Selective reporting (reporting bias)

High risk

No prospective trial registration identified. Primary outcome on which this trial was powered is not stated. On further correspondence, adverse event data were given but no clinical baseline characteristics are given.

Bessmertny 2002

Methods

Design: parallel randomised controlled trial.
Method of randomisation: computer‐generated random numbers.
Concealment of allocation: yes.
Blinding: double‐blinded, placebo‐controlled.
Withdrawals/dropouts: 6 (4 unable to complete spirometry, 2 inappropriate randomisation).

Participants

Location: 1 university hospital in Brooklyn, NY.
Participants: 74 patients, presenting to the emergency department with acute asthma exacerbation, PEF between 40% and 80% predicted.
Exclusions: smoking history > 10 pack years, known hypersensitivity to albuterol or MgSO₄, known chronic obstructive pulmonary disease, known history of renal impairment, known history of cardiac dysrhythmias, congestive heart failure or angina, fever more than 38 °C, receipt of theophylline or anti‐cholinergic within 2 h of arrival to ED.

Interventions

Treatment: albuterol 2.5 mg/3 mL nebule followed by 384 mg isotonic MgSO₄ every 20 min × 3.
Control: albuterol 2.5 mg/3 mL nebule followed by normal saline every 20 min × 3.

Outcomes

Measured FEV1 every 20 minutes for 2 h.
Adverse events: no serious adverse events noted.

Notes

Funding: supported by an unrestricted educational grant from Astra Pharmaceutical Company; no Astra Pharmaceutical Company products were used in the study. Mouthpieces for the spirometer were supplied at no charge from Mallinkrodt Nellcor Puritan Bennett. Circulaire nebulizers were supplied by Westmed Inc. at a reduced rate.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An assigned third party randomised participants by means of a computer‐generated random table (1:1 randomisation) to either the treatment or control group.

Allocation concealment (selection bias)

Low risk

An assigned third party randomised participants by means of a computer‐generated random table (1:1 randomisation) to either the treatment or control group.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded, placebo‐controlled. A log of the identification number and specific treatment of each participant was kept and remained closed to the investigators until the completion of the study.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blinded, placebo‐controlled. A log of the identification number and specific treatment of each participant was kept and remained closed to the investigators until the completion of the study. Outcomes were assessed every 20 minutes for 2 h.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 3 in each group.

Albuterol plus normal saline solution (3 unable to complete spirometry); and albuterol plus magnesium (2 inappropriate randomisation, 1 unable to perform spirometry).

Selective reporting (reporting bias)

High risk

Mean values only given for FEV1, no SDs and the text reports that there were no statistically significant differences in FEV1 between the groups. The text also states "The analysis of continuous safety variables (BP, pulse rate, respiratory rate, oxygen saturation, and serum magnesium concentrations) did not demonstrate any clinically or statistically significant differences between the 2 groups at any point during the study."

Dadhich 2005

Methods

Random allocation into 3 groups parallel study.

Participants

Location: 1 emergency department teaching hospital in India.

Acute severe asthma , PEF < 50%.

Group A = 24

Group B = 26

Group C = 21

Interventions

Group A: salbutamol; Group B; salbutamol and MgSO₄; Group C MgSO₄ alone; no details on dose or frequency.

Outcomes

FEV1, FVC, FEV1/FVC, PEF,

"Vital parameters"

Notes

2 abstracts only (the same).

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly allocated.

Allocation concealment (selection bias)

Unclear risk

No details.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No details.

Selective reporting (reporting bias)

Unclear risk

Abstract only and no data reported except there was a significant improvement in groups B and C compared to group A.

Drobina 2006

Methods

Parallel.

Participants

A total of 110 participants.

Interventions

Intervention: received the control treatment with the addition

of 150 mg of MgSO₄ (0.3 mL of 50% MgSO₄ heptahydrate) to each nebulised dose of medication.

Control: received nebulised treatments of albuterol sulfate 0.5% (5 mg/mL) combined with 0.5 mg of ipratropium bromide 0.02% inhalation solution (Atrovent).

Outcomes

Vital signs and peak flow measurements were also assessed at the end of each treatment (a maximum of 3 treatments) and just prior to discharge.

A 24‐hour follow‐up call was made to each participant, during which peak flow measurements were again obtained.

Notes

Abstract only.

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised but no detail.

Allocation concealment (selection bias)

Unclear risk

Described as randomised but no detail.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double blind but no detail.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Described as double blind but no detail.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Very limited information ‒ impossible to judge.

Selective reporting (reporting bias)

Unclear risk

Abstract only. Vital signs are mentioned as being recorded but are not reported.

Gallegos‐Solórzano 2010

Methods

RCT, parallel

Participants

Inclusion criteria: adults, >18 years in the emergency dept with asthmatic crisis, FEV1 < 60% predicted.

Exclusion criteria: smokers, those with ambulatory use of systemic steroids, with associated co‐morbidities (neuropathy, nephropathy, heart disease, liver disease), fever at admission, use of dietary supplements with MgSO₄, irreversible airway obstruction (persistent abnormal spirometry), near‐fatal asthma, requirement of endotracheal intubation at admission, anatomic abnormalities of the bronchial tree (bronchiectasis, tuberculosis), history of pulmonary or thoracic surgery, hypersensitivity to MgSO₄, and pregnancy or breastfeeding.

Location: National Institute of Respiratory Diseases, a tertiary care teaching hospital and national referral centre in Mexico City.

Date of study: June 2008 to March 2009.

Intervention: 60 randomised, 30 completed.

Mean age (years): 34.3 (12.4).

Men:women: 9:21.

Control: 52 randomised, 30 completed.

Mean age (years): 40.3 (11.6).

Men:women: 9:21.

Interventions

Each nebulisation lasted 20 mins.

Intervention: standard nebulisation but diluted with 3 mL (333 mg) of 10% isotonic MgSO₄ (Magnefusin PISA, Guadalajara, Mexico; 1 g/10 mL). Also received 125 mg of IV methylprednisolone.

Control: 1 IV dose of 125 mg methylprednisolone and nebulisation with 7.5 mg of albuterol and 1.5 mg of ipratropium bromide in 3 divided doses. Standard nebulisation diluted in 3 mL of isotonic saline solution (SS) as placebo.

Outcomes

FEV1 post‐BD (absolute in litres and as percentage of predicted), clinical improvement, oxygen saturation, admission to the ED, admission to the asthma ward, hospital readmissions.

At 30‐min post‐nebulisation, patients were clinically and functionally re‐evaluated. Also evaluated at 30 days.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised.

Allocation concealment (selection bias)

Low risk

After randomisation, diluents were prepared by a physician outside the study who was not responsible for the participants’ care and only had control of the pre‐filled syringes.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind.

Both diluents are odourless, tasteless and colourless to the eye and did not differ when transparency was measured.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The physician responsible for the participants’ care along with the nurse and respiratory therapist were blinded to the type of treatment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reasons given for dropouts in both groups in the CONSORT diagram.  It seems as though there are a high percentage of dropouts but the majority are post‐randomisation exclusions based on exclusion criteria.

Selective reporting (reporting bias)

Low risk

All outcomes stated in the Methods section are reported.  Best judgement with no access to trial protocol.

Gaur 2008

Methods

Parallel RCT.

Participants

Age: 18 to 60 years.

Location: emergency department of a tertiary referral centre in India.

Acute asthma and FEV1 < 30% predicted.

Intervention: 30.

Control: 30.

Interventions

Intervention: nebulised similarly using isotonic MgSO₄ (3 mL of 3.2 g%) as a vehicle ‒ unsure if this is “Nebulized salbutamol and ipratropium”.

Control: nebulised salbutamol and ipratropium using isotonic saline as a vehicle thrice at 20‐min intervals.

Outcomes

FEV% predicted at 120 minutes, pooled discharge rate proportion of groups attaining PEF > 60% predicted and relief in dyspnoea at 30, 60, 90, 120 min).

Notes

Abstract only.

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised.

Allocation concealment (selection bias)

Unclear risk

Not stated.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Single blind – no further details.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Single blind – no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated.

Selective reporting (reporting bias)

High risk

1 outcome partially reported and not significant. Abstract only.

Goodacre 2013

Methods

Double blind, randomised controlled trial.

34 emergency departments, UK.

Participants

Inclusion criteria: severe (BTS/SIGN quantified) asthma attack, age ≥16 years.

Exclusion criteria: life‐threatening features, contraindication to MgSO₄, participant unable to give verbal/written consent, previous participation in the study; criteria amended to exclude those who had received MgSO₄ in the past 24 h.

1109 randomised.

Intervention 1 (nebulised MgSO₄): 339 randomised.

Mean age (years): 36.5 (14.8).

Men:women: 107:232.

Smokers: 98.

Mean predicted PEF (L/min): 430 (118.8).

Intervention 2 (intravenous MgSO₄): 406 randomised.

Mean age (years): 35.6 (13.1).

Men:women: 130:279.

Smokers: 138.

Mean predicted PEF (L/min): 431.8 (116.9).

Control: 364 randomised.

Mean age (years): 36.4 (14.1).

Men:women: 112:252.

Smokers: 127.

Mean predicted PEF (L/min): 435.0 (110.8).

Interventions

Intervention 1: 100 mL 0.9% NaCl IV and 2 mmol MgSO₄ in 7.5 mL 0.9% NaCl nebulised.

Intervention 2: 8 mmol MgSO₄ in 100 mL 0.9% NaCl IV and 7.5 mL 0.9% NaCl nebulised.

Control: 100 mL 0.9% NaCl IV and 7.5 mL 0.9% NaCl nebulised.

IV infusion given once over 20 mins, nebulisers given 3 times, each over 20 minutes.

Outcomes

Admission (4 h, 7 days); change in participant's assessment of breathlessness via visual analogue scale, change in PEF, heart rate, respiratory rate, BP, oxygen saturations (1, 2 h); adverse events (2 h); mortality, length of hospital stay, admission to HDU or ICU.

Adverse events: treatment group 41 adverse events; control group 36 adverse events.

Notes

Funding: UK National Institute for Health Research Health Technology Assessment Programme.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Simple and blocked randomisation sequences used to allocate participants to numbered treatment packs.

Allocation concealment (selection bias)

Low risk

Allocated treatment pack numbers were only revealed after participant details recorded and the participant irreversibly entered into the trial.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, hospital staff, and research staff were masked to allocated treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants, hospital staff, and research staff were masked to allocated treatment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Over 95% of randomised participants in each arm were included in primary analysis. All participants clearly accounted for in flow diagram. There was an inevitable 'drop off' in participants available at each time point for many of the secondary outcomes; it is unclear what impact this may have had on the results.

Selective reporting (reporting bias)

Low risk

Prospective trial registration identified. All primary and secondary outcomes listed in the trial register were reported.

Hossein 2016

Methods

Double blind, randomised controlled trial.

2 emergency departments, Iran.

Participants

Inclusion criteria: moderate/severe asthma exacerbation defined by PEFR < 40% to 69% predicted or limiting speech/normal activity, age > 16 years.

Exclusion criteria: need for immediate intubation, significant impairment of heart function, kidney or liver disease, fever > 38.3 °C, chronic lung disease, pregnancy, lactation, pneumonia.

50 randomised.

Intervention: 25 randomised.

Mean age (years): 52.4 (16.9).

Men:women: 11:14.

Acute moderate: 3.

Acute severe: 22.

Mean predicted PEF (%): 15.1 (4.7).

Control: 25 randomised.

Mean age (years): 53.9 (16.2).

Men:women: 14:11.

Acute moderate: 3.

Acute severe: 21

Mean predicted PEF (%): 14.7 (6.4).

Interventions

Intervention: 3 mL MgSO₄ solution (260 mmol/L) nebulised.

Control: 3 mL 0.9% NaCl nebulised.

Nebulised medication given every 20 to 60 minutes.

Outcomes

Predicted PEFR (%), oxygen saturations, respiratory rate, dyspnoea severity index (20, 60 minutes); need for admission, serious side‐effect rate (60 minutes).

Adverse effects: no "serious side‐effects" reported.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation software used.

Allocation concealment (selection bias)

Unclear risk

Data not given.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both patients and investigators were blinded to the content of identical treatment vials.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Both patients and investigators were blinded to the content of identical treatment vials.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The report does not state that all 25 randomised participants in each arm completed the trial, but as the trial finished at 60 mins is it likely that they did.

Selective reporting (reporting bias)

Unclear risk

Trial registered while recruiting. Pre‐specified primary and secondary outcomes are reported; note omission of PEFR/dyspnoea scale reporting at 40 mins and no data given to support report of "no treatment‐related complications". Clear mistakes in reporting of vital signs.

Hughes 2003

Methods

Design: parallel randomised controlled trial.
Method of randomisation: unknown.
Concealment of allocation: yes.
Blinding: double‐blinded, placebo‐controlled.
Withdrawals/dropouts: 6 (4 COPD, 2 pneumonia).

Participants

Location: 2 university hospitals in New Zealand.
Participants: 52 patients, presenting to the emergency department with acute asthma exacerbation, FEV1 < 50% predicted.
Exclusions: known irreversible lung disease, pneumonia, pregnancy, significant renal/cardiac impairment, hypotension (sBP < 100 mmHg), required intubation.

Interventions

Standard of care: salbutamol 2.5 mg nebulised ×1 or more, hydrocortisone 100 mg IV at presentation.
Treatment: salbutamol 2.5 mg nebule with 2.5 mL isotonic MgSO₄ (250 mmol/L) every 30 min ×3.
Control: salbutamol 2.5 mg nebule with 2.5 mL normal saline every 30 min ×3.
Participants were unable to distinguish solutions.

Outcomes

Measured at baseline and after each treatment (every 30 min ×3): FEV1, % predicted FEV1, BP, heart rate, O₂ saturation.
Requirement for admission at 90 minutes.
Adverse events: no serious adverse events noted.

Notes

Funding: the study was funded by a research grant from the University of Otago. The study sponsor had no role in study design, data collection, data analysis, data interpretation, or writing of the report

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to their treatment groups in accordance with the allocation sequence determined by the hospital pharmacy.

Allocation concealment (selection bias)

Unclear risk

No information.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded, placebo‐controlled. Participants and investigators were unaware of treatment allocation through provision by the hospital pharmacy of pre‐prepared identical unmarked syringes containing the study drug.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blinded, placebo‐controlled. Participants and investigators were unaware of treatment allocation through provision by the hospital pharmacy of pre‐prepared identical unmarked syringes containing the study drug. Outcomes assessed every 30 minutes.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

6 in total.

MgSO₄ (1 COPD, 1 pneumonia).

Saline (3 COPD, 1 pneumonia).

Selective reporting (reporting bias)

High risk

The primary outcome, FEV1, was fully reported but other outcomes were not. "The change in blood pressure and heart rate did not differ between the two groups. No clinically significant adverse events were reported."

Khashabi 2008

Methods

Parallel RCT.

Participants

Location: authors based in Iran.

Participants: 40 asthmatic children in total between 2 groups.

Mean age: 3.55 years.

Interventions

Intervention: nebulised salbutamol, as a vehicle isotonic MgSO₄ mixed with salbutamol.

Control: nebulised salbutamol, as a vehicle 2.5 mL of normal saline.

Outcomes

Days of hospital stay, hours of need for oxygen, respiratory distress.

Measured 1 h before and 1 h after the second course of treatment.

Notes

Abstract only.

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly enrolled.

Allocation concealment (selection bias)

Unclear risk

Not stated.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double blind.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated.

Selective reporting (reporting bias)

Low risk

Outcomes stated as measured, reported. Abstract only.

Kokturk 2005

Methods

Parallel RCT

Participants

Inclusion criteria: moderate to severe asthma attacks, 18 to 60 years.

Exclusion criteria: patients with febrile disease, diabetes, congestive heart failure, atherosclerotic heart disease, intractable hypertension, chronic obstructive lung disease, renal and hepatic failure and arrhythmia were excluded from the study. Pregnant and breast‐feeding women, patients who had already taken theophylline, antihistaminics, and systemic steroids in the previous 24 h, who had acute or chronic respiratory failure, who had been on long‐term oxygen therapy, and a history of allergy to salbutamol and MgSO₄ have been excluded as well.

Location: emergency department, Turkey.

Intervention: 14.

Mean age: 46.43 (years) (3.31) range 18 to 3.

Men:women: 4:10.

Control: 12.

Mean age: 37.83 (years) (9.26) range 20 to 52.

Men:women: 3:9.

Interventions

Every 20 mins for first hour and every hour for the rest of 4 h.

Intervention: isotonic MgSO₄ (2.5mL) + salbutamol (2.5 mL).

Control: salbutamol (2.5 mL) + saline (2.5 mL).

Outcomes

PEF, clinical scores, discharge rates, admission rates.

20th, 60th, 120th, 180th, 240th minute (180 and 240 not compared as most patients completed study in 2 h).

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised ‒ details of sequence generation not included in trial report.

Allocation concealment (selection bias)

Unclear risk

Information not available in trial report.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Single blind – no further details.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Single blind – no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Information provided in trial report on discharges from both groups up to 240 minutes.

Selective reporting (reporting bias)

Low risk

No apparent indication of selective reporting.

Mahajan 2004

Methods

Design: parallel randomised controlled trial.
Method of randomisation: table of random numbers.
Concealment of allocation: not stated.
Blinding: double‐blinded, placebo‐controlled.
Withdrawals/dropouts: none described.

Participants

Location: 1 paediatric emergency department in Detroit, Michigan.
Participants: 62 patients age 5 to 17, presenting to the emergency department with acute asthma exacerbation, FEV1 between 45% and 75% predicted.
Exclusions: Fever (> 39 °C), chronic disease (bronchopulmonary dysplasia, cystic fibrosis), known allergy to albuterol or magnesium, received any of steroids, theophylline or ipratropium bromide in the prior 3 days.

Interventions

Treatment: albuterol 2.5 mg nebule with 2.5 mL isotonic MgSO₄ (6.3% solution); 1 dose.
Control: albuterol 2.5 mg nebule with 2.5 mL normal saline; 1 dose.
Both groups received corticosteroids (2 mg/kg) after inhaled treatment.

Outcomes

Lung function (FEV1 and % predicted FEV1) at baseline, then at 10 and 20 minutes after treatment.
Also report vital signs and hospital admission rates.
State that none of the patients showed any side effects.

Notes

Funding: this work was funded by an unrestricted grant from the Division of Pediatric Emergency Medicine, Children’s Hospital of Michigan, Detroit, Michigan

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A table of random numbers was used to provide randomisation and this was performed by a senior research pharmacist at the institution.

Allocation concealment (selection bias)

Low risk

A table of random numbers was used to provide randomisation and this was performed by a senior research pharmacist at the institution.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded, placebo controlled. The study medications were provided in identical syringes and both the pharmacy and the investigator were blinded to their contents.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blinded, placebo controlled. The study medications were provided in identical syringes and both the pharmacy and the investigator were blinded to their contents. Outcomes assessed at 10 and 20 minutes after treatment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

None described.

Selective reporting (reporting bias)

Low risk

All outcomes stated in the methods section are reported.

Mangat 1998

Methods

Design: parallel randomised controlled trial.
Method of randomisation: unknown.
Concealment of allocation: yes.
Blinding: double‐blind, placebo‐controlled.
Withdrawals/dropouts: 0.

Participants

Location: emergency department, St John's Medical College Hospital, India.
Screened: 63.
Participants: 33, 12 to 60 years of age, known or newly diagnosed asthmatics with PEF < 300 L/min.
Exclusions: patient enrolled at prior presentation, febrile, lower respiratory tract infection, history or evidence of cardiac/renal/hepatic dysfunction, pregnancy, requirement for ventilatory care, oral/parenteral bronchodilators within previous 6 h, steroids within previous 12 h.

Interventions

Standard of care: hydrocortisone 100 mg IV.
Treatment: MgSO₄ 3 mL (3.2% solution = 95 mg) nebulised every 20 min ×4.
Control: salbutamol 3 mL (2.5 mg) nebulised every 20 min ×4.

Outcomes

Clinical score: Fischl Index, clinical examination.

Pulmonary function: PEF.

Vitals: respiratory rate, heart rate, BP, pulsus paradoxus.

Admission rates, vital signs.

Adverse events/side effects:

  • treatment: 1 case mild transient hypotension with spontaneous resolution.

  • control group: 1 case mild transient hypotension with spontaneous resolution, 1 case palpitations, 2 cases fine tremors in hand.

Notes

Funding: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised.

Allocation concealment (selection bias)

Unclear risk

Information not available.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled. Outcomes assessed at 20 minute intervals.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

None described.

Selective reporting (reporting bias)

Unclear risk

Pulsus paradoxus and BP are mentioned but not reported, but pulsus paradoxus is included as part of the Fischl index.

Meral 1996

Methods

Design: randomised controlled trial.
Method of randomisation: unknown.
Concealment of allocation: unknown.
Blinding: unknown.
Withdrawals/dropouts: 0.

Participants

Location: Department of Paediatric Asthma of Ege University Hospital, Turkey.
Participants: 40 randomly selected and divided into 2 groups of 20. Mean ages 10.6 and 11 years of age. Previously diagnosed as asthmatic using ATS definitions; PEF decreased by ≥ 25%.
Exclusions: medication within 12 h of study, cardiac/renal dysfunction.

Interventions

Treatment: MgSO₄ 2 mL (280 mmol/L, 258 mOsm, pH 6.7).
Control: salbutamol 2.5 mg in 2.5 mL.
Administration: nebulised, inhaled over 10 to 15 minutes.

Outcomes

Evaluations at: 5, 15, 30, 60, 180,240 and 360 minutes.
Clinical score: Davis‐Leffert‐Dabbous respiratory distress score pulmonary function: PEF.
Adverse reactions/side effects: none observed.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Patients were randomly selected for the study and divided into 2 groups.

Allocation concealment (selection bias)

Unclear risk

Information not available.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

None described.

Selective reporting (reporting bias)

High risk

No statistical differences were found between the groups for respiratory rate, heart rate and BP. It is also unclear as to the time point reported as although 5 minutes was prespecified, there were also several other time points specified and only the maximum values were presented.

Mohammedzadeh 2014

Methods

Randomised controlled trial.

1 hospital, Iran.

Participants

Inclusion criteria: moderate to severe asthma (GINA‐defined) with acute attack.

Exclusion criteria: corticosteroid therapy, steroid/theophylline/ipratropium in past 72 h, chronic lung disease e.g. bronchopulmonary dysplasia/CF, allergy to MgSO₄ or salbutamol, not co‐operative.

80 randomised.

Intervention 1 (nebulised MgSO₄): 40 randomised.

Mean age (years): 9 (2.2).

Male:female: 10:30.

Control: 40 randomised.

Mean age (years): 8.5 (2.4)

Male:female: 17:23.

Interventions

Intervention: 3 mL 7.5% MgSO₄, 0.15 mg/kg salbutamol.

Control: 3 mL normal saline, 0.15 mg/kg salbutamol.

3 doses at 20 minute intervals.

Outcomes

Pulmonary index, PEFR, adjusted PEFR at 30, 60 and 90 minutes

Notes

Funding: Babol University of Medical Sciences ‒ Research and Technology Institute.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as "divided into two groups randomly" but no details given.

Allocation concealment (selection bias)

Unclear risk

Data not given.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as "double‐blind" in prospective trial registration but no details given.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as "double‐blind" in prospective trial registration but no details given.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participants lost to follow‐up.

Selective reporting (reporting bias)

Low risk

Prospectively registered; planned outcomes were fully reported.

Nannini 2000

Methods

Design: randomised controlled trial.
Method of randomisation: unknown.
Concealment of allocation: yes.
Blinding: double‐blind, placebo‐controlled.
Solutions were pre‐packaged in identical appearing vials.
Withdrawals/dropouts: 3 participants were enrolled more than once, only the initial visit was used in the analysis.

Participants

Location: emergency departments in 4 Argentinian hospitals.
Participants: 35 patients at least 18 years of age presenting to the emergency department with an acute asthma exacerbation who were able to have PEF measured were enrolled.
(% predicted PEF: 38 + 18 in treatment group, 38 + 12 in control group).
Exclusions: current smokers of ≥ 5 pack years, concurrent medical illness, pregnant, breast feeding, oral or parenteral steroids within the previous 7 days.

Interventions

Standard of care: all patients received supplemental oxygen. If patient condition worsened patient may receive salbutamol 2.5 mg nebulised at discretion of physician.
Treatment: 0.5 mL salbutamol (2.5 mg) diluted in 3 mL isotonic MgSO₄ (286 mOsm, 7.5% = 225 mg).
Control: 0.5 mL salbutamol (2.5 mg) diluted in 3 mL normal saline.
Administration: jet nebulised using oxygen at 10 L/min via mouthpiece until dry.

Outcomes

Measurements made at baseline, 10 minutes after treatment and 20 minutes after treatment.
Pulmonary functions: primary endpoint : % increase in peak flow = ((change/baseline) × 100).
Other: peak flow (best of 3 attempts).
Vital signs: respiratory rate, pulse rate, BP.
Duration of emergency room care.
No adverse events reported in either the experimental or control group.

Notes

Funding: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised.

Allocation concealment (selection bias)

Unclear risk

Information not available.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled.

Incomplete outcome data (attrition bias)
All outcomes

High risk

3 patients were enrolled more than once, only the initial visit was used in the analysis but treatment group not stated.

Selective reporting (reporting bias)

High risk

There were no significant differences between the groups in changes in BP, heart rate, or respiratory rate at either 10 minutes or 20 minutes.

Neki 2006

Methods

Parallel.

Participants

Inclusion criteria: patients in age group of 15 to 60 years with severe bronchial asthma, as judged by Fischl index having PEF < 300 L/min or FEV in 1st second less than 40% of the predicted value were included in the study.

Exclusion criteria: all patients who had received oral inhaler or parenteral bronchodilators in the past 6 h or steroid in the previous 12 h were excluded from the study.

Adults and children with severe asthma (15 to 60 years) ‒ 40 participants.

30 female and 10 male but unclear how divided between groups.

Intervention: 20 completed.

Control: 20 completed.

Interventions

Intervention: given 4 doses of nebulised solution of "3.2G%" MgSO₄, 20 minutes apart.

Control: received 4 doses of nebulised salbutamol (each dose of 3 mL containing 25 mg), 20 minutes apart.

Outcomes

PEF (L/min),

respiratory rate, Fischl index and SaO₂.

Notes

Abstract only.

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Details of random sequence generation not included in trial report. There is no reference to randomisation in trial report and trial not reported as randomised – seeking clarification from author.

Allocation concealment (selection bias)

Unclear risk

Details of allocation concealment not included in trial report. There is no reference to randomisation in trial report and trial not reported as randomised – seeking clarification from author.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

If the trial was not blinded, there is a strong likelihood that outcome assessment was not blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information.

Selective reporting (reporting bias)

Unclear risk

Abstract only. No apparent indication of selective reporting.

Powell 2013

Methods

Double blind, randomised controlled trial.

30 emergency departments or children's assessment units, UK.

Participants

Inclusion criteria: severe (BTS/SIGN quantified) asthma exacerbation after conventional treatment, age 2 to 16 years.

Exclusion criteria: coexisting respiratory disease, severe renal disease, severe liver disease, known pregnancy, known previous reaction to magnesium, inability to give informed consent, previous randomisation into the trial, life‐threatening symptoms, current or previous (in the 3 months preceding screening) involvement with a trial of a medicinal product.

508 randomised.

Intervention: 252 randomised.

Median age (years): 4 (3 to 7).

Male:female: 143:109.

Control: 256 randomised.

Median age (years): 4 (3 to 7).

Male:female: 150:106.

Interventions

Intervention: 2.5 mL MgSO₄ (250 mmol/L) nebulised.

Control: 2.5 mL isotonic saline nebulised.

3 doses given at roughly 20 minute intervals.

Outcomes

Mean Yung asthma severity score, treatment step‐down (60 minutes); length of stay, need for additional intravenous bronchodilator, admission to PICU/HDU or intubation, adverse events (until discharge).

Adverse events: treatment group 47, control group 59

Notes

Funding: National Institute of Health Research Health Technology Assessment Programme.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A computer‐generated blocked randomisation sequence stratified by centre was generated by an independent statistician who had no further involvement in the study.

Allocation concealment (selection bias)

Low risk

Treatment packs were identical in appearance and numbered sequentially for each centre.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants (patients, clinicians, research team, and statisticians) were masked to the treatment allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The statistical analyses were completed with masked data, with treatment groups revealed only after final analyses had been completed.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Over 90% randomised participants in each arm were included in the adjusted primary analysis. All participants who withdrew or were excluded are clearly accounted for in the flow diagram.

Selective reporting (reporting bias)

Low risk

Prospectively registered trial. All listed outcomes are reported.

Sarhan 2016

Methods

Double blind, randomised controlled trial.

Chest and emergency departments at 1 hospital, Egypt.

Participants

Inclusion criteria: diagnosis of asthma.

Exclusion criteria: fever, lower respiratory tract infection, cardiac/renal/hepatic dysfunction, needed NIV/intubation, near‐fatal asthma, pregnancy, lactation, failed to use PEF meter, inhaled/oral/intravenous bronchodilator use within past 6 h or steroid use within past 12 h.

30 randomised.

Intervention 1 (magnesium): 10 randomised.

Mean age (years): 33.5 (17.8).

Men:women: 4:6.

Mean % of predicted PEF at presentation: 33.9 (9.8).

Intervention 2 (salbutamol and placebo) : 10 randomised.

Mean age (years): 48.6 (9.9).

Men:women: 3:7.

Mean % of predicted PEF at presentation: 36.4 (10.5).

Intervention 3 (salbutamol and magnesium): 10 randomised.

Mean age (years): 51.3 (15.8).

Men:women: 7:3.

Mean % of predicted PEF at presentation: 34.1 (9.4).

Interventions

Intervention 1: 3 mL MgSO₄ (3.3% solution) nebulised.

Intervention 2: 0.5 mL salbutamol (0.5% solution) in 2.5 mL isotonic saline nebulised.

Intervention 3: 0.5 mL salbutamol (0.5% solution) in 2.5 mL MgSO₄ (4% solution) nebulised.

4 doses given at 20 minute intervals.

Ultrasonic nebuliser.

Outcomes

PEF improvement, respiratory rate, heart rate, blood pressure, oxygen saturations, improvement in Fischl index of clinical severity, adverse event rate (all at "final" time point, assumed to be 2 h).

Adverse events: no events "severe enough to warrant withdrawal" reported.

Notes

Funding: not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Reports patients were randomised into 3 groups but no details given.

Allocation concealment (selection bias)

Unclear risk

Data not given.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as "double blind" but no details given about who was blinded or how.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as "double blind" but no details given about who was blinded or how.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The report does not specify how many randomised participants completed the trial.

Selective reporting (reporting bias)

Unclear risk

No prospective trial registration identified. Primary and secondary outcomes not defined. No power calculation reported.

Turker 2017

Methods

Double blind, randomised controlled trial.

1 emergency department, Turkey

Participants

Inclusion criteria: children aged 3 to 15 years with asthma admitted to the emergency department due to a moderate asthma exacerbation.

Exclusion criteria: any associated chronic diseases such as cystic fibrosi and bronchiectasis.

100 randomised.

Intervention: 50 randomised.

Mean age, months (SD): 76.06 (27.33).

Male:female: 25:25.

Median (IQR) modified pulmonary index score at presentation 8 (7‐8).

Control: 50 randomised.

Mean age, months (SD): 74.96 (33.65).

Male:female: 29/21.

Median (IQR) modified pulmonary index score at presentation 7 (7 to 9).

Interventions

Intervention: nebulised salbutamol (0.15 mg/kg) + 1 mL magnesium sulfate (15%) + 1.5 mL isotonic saline.

Control: nebulised salbutamol (0.15 mg/kg) + 1.5 mL isotonic saline.

3 doses given at 20 min intervals.

Outcomes

Primary outcome: Modified Pulmonary Index Score (MPIS); secondary outcomes: hospitalisation rates, symptoms of magnesium imbalance such as nausea, vomiting, abdominal pain, chest pain, headache, fatigue, hypotension and fever.

Notes

Funding: "this research did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors".

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Patients were assigned consecutively to the control or intervention group based on a stratified randomisation procedure" but no further detail about how the randomisation sequence was generated.

Allocation concealment (selection bias)

Unclear risk

Data not given.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as "double‐blind" but no details of who was blinded and the blinding procedure.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as "double‐blind" but no details of who was blinded and the blinding procedure.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"All patients enrolled in the study completed it".

Selective reporting (reporting bias)

High risk

No trial registration or prospective protocol identified. Adverse events reported as: "no side effect caused by magnesium was observed in any of the patients in the study". Modified pulmonary index score reported numerically at 120 minutes only; other time points presented graphically with no measure of variance

ASS: Asthma Severity Score (ASS)
ATS: American Thoracic Society
BP: blood pressure
BTS: British Thoracic Society
COPD: Chronic obstructive pulmonary disease
ED: emergency department
FEV1: Forced expiratory volume in 1 second
FVC: Forced vital capacity
h: hour(s)
IV: intravenous
MDI: metered dose inhaler
MgSO₄: magnesium sulfate
PEF: Peak Expiratory Flow Rate
R&D: research and development
sBP: systolic blood pressure
SD: standard deviation
SIGN: Scottish Intercollegiate Guidelines Network

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Balter 1989

Review

Bede 2003

Oral supplementation in chronic asthma

Bede 2004

Oral supplementation in chronic asthma

Bede 2008

Oral supplementation in chronic asthma

Bernstein 1995

Study does not assess people with acute asthma

Cairns 1996

Study does not assess people with acute asthma

Castillo Rueda 1991

Letter to the Editor

Chande 1992

Study of stable asthma and methacholine challenge tests

Corbridge 1995

Review

DiGregorio 1999

Not a randomised controlled study

Emelyanov 1997

Study not a randomised trial and in mild‐to‐moderate persistent asthma rather than acute asthma

Emelyanov 1990

Not a randomised controlled trial

Emelyanov 1996

Exercise induced bronchospasm and challenge test. Not a randomised controlled trial.

Fathi 2014

Oral supplementation in chronic asthma

Fedoseev 1991

Study does not assess people with acute asthma and is not a randomised controlled trial

Gandia 2012

Study of stable asthma and methacholine challenge tests

Gurkan 1999

Randomised controlled trial of intravenous MgSO4

Harari 1998

Review

Hardin 2001

Review

Harmanci 1996

Stable asthma histamine‐induced bronchospasm adults

Hill 1995

Study does not assess people with acute asthma. Dose response study in 20 normal individuals and 19 with chronic asthma.

Hill 1997a

Study does not assess people with acute asthma. Stable asthma histamine challenge tests.

Hill 1997b

Stable adult asthma with histamine challenges

Irazuzta 2014

Randomised controlled trial of intravenous MgSO₄

Irazuzta 2016

Randomised controlled trial of intravenous MgSO₄

Kenyon 2001

Review

Kreutzer 2001

Review

Manzke 1990

Paediatric exercise‐induced bronchospasm. Not a randomised controlled trial.

McFadden 1995

Review

Nannini 1997

Study does not assess people with acute asthma

Nunez‐Torres 1995

Not a randomised controlled trial

Pelton 1998

Study does not assess people with acute asthma

Pelton 1999

Review

Petrov 2014

Oral supplementation in uncontrolled and partly controlled atopic asthma

Puente‐Maestu 1999

Review

Qureshi 1999

Review

Rodger 2003

Oral supplementation on people with unstable asthma

Rodrigo 2000

Systematic review, includes intravenous MgSO₄

Rolla 1987a

Study does not assess people with acute asthma

Rolla 1987b

Study does not assess people with acute asthma

Rolla 1988a

Study does not assess people with acute asthma

Rolla 1988b

Letter to the editor

Scarfone 1998

Randomised controlled trial of intravenous MgSO₄

Scarfone 2000

Intravenous MgSO₄

Shishimorov 2015

Oral supplementation in children with uncontrolled asthma

Singh 2008a

Intravenous MgSO₄

Singh 2008b

Comparison between inhaled versus intravenous MgSO₄

Singhi 2014

Randomised controlled trial of intravenous MgSO₄

Sinitsina 1991

Not a randomised controlled trial

Skobeloff 1982

Editorial

Sun 2014

Study of stable asthma and methacholine challenge tests

Talukdar 2005

Not a randomised controlled trial

Teeter 1999

Review

Telia 2005

Study does not assess people with acute asthma

Tereshchenko 2006

Looking at ipratropium bromide mixed with either MgSO₄ or saline for bronchiolitis (up to age 11.5 months)

Tetikkurt 1992

Study does not assess people with acute asthma

Tetikkurt 1993

Study does not assess people with acute asthma

Torres 2012

Randomised controlled trial of intravenous MgSO₄

Watanatham 2015

Randomised controlled trial of intravenous versus nebulised MgSO₄

Wijetunge 2002

No response to attempts made to contact first author from 2002 to 2012. First author sadly died in 2014.

Wongwaree 2017

Randomised controlled trial of nebulized magnesium sulfate versus ipratropium bromide/fenoterol in children with severe asthma exacerbation

Xu 2002

Not a randomised controlled trial

Yemelyanov 1997

Study does not assess people with acute asthma

Zandsteeg 2009

Study does not assess people with acute asthma (stable chronic asthma) and is not a randomised controlled trial

Zhu 2003

Intravenous MgSO₄ and not a randomised controlled trial

MgSO₄: magnesium sulfate

Characteristics of studies awaiting assessment [ordered by study ID]

Abd 1997

Methods

"Ventilatory, cardiovascular and metabolic responses to salbutamol, ipratropium bromide and magnesium sulfate in bronchial asthma: comparative study"

Participants

No details

Interventions

No details

Outcomes

No details

Notes

Full‐text unobtainable

Bustamante 2000

Methods

"Inhaled magnesium sulfate as adjunct therapy for moderate to severe asthma exacerbations, a randomized control clinical trial"

Participants

No details

Interventions

No details

Outcomes

No details

Notes

Full‐text unobtainable

ISRCTN61336225

Methods

Prospective double‐blind placebo controlled trial

Participants

Children diagnosed as asthmatic according to The Global Initiative for Asthma (GINA) guidelines, aged 5 to 14 years old, capable of measuring PEFR, presenting with moderate to severe acute exacerbation according to paediatric asthma severity score and PEFR

Interventions

Group A: participants receive inhaled salbutamol solution (0.15 mL/kg) plus isotonic magnesium sulfate (2 mL) in a nebulizer chamber;

Group B: participants receive inhaled salbutamol solution (0.15 mL/kg), diluted with placebo (normal saline 2 mL) in a nebulizer chamber.

Outcomes

1. Asthma severity measured using the Pediatric Asthma Severity Score (PASS) at baseline, 20, 40 and 60 minutes post‐nebulisation

2. Oxygen saturation measured using pulse oximetry at baseline, 20, 40 and 60 minutes post‐nebulisation

3. Lung function assessed through measuring peak expiratory flow rate (PEFR) at baseline, 20, 40 and 60 minutes post‐nebulisation

Notes

Trial stated as complete February 2016 but no associated publication identified. Contact person emailed on 7 September 2017 to enquire about status of results/publication. No response received at time of review publication.

Characteristics of ongoing studies [ordered by study ID]

Motamed 2015

Trial name or title

Comparison of clinical and spirometric response between nebulized salbutamol, MgSO₄ and nebulized salbutamol alone in acute asthma attack

Methods

Randomized, double‐blind controlled trial

1 hospital, Iran

Participants

Inclusion criteria: having a history of asthma, a minimum 18 years and maximum 65 years

Exclusion criteria: COPD; kidney disease; CHF; pneumonitis; underlying respiratory disease

146 randomised

Interventions

Intervention: nebulized MgSO₄ 1/5 mL (20 g / 100 mL) with salbutamol 2/5 mL

Control: normal saline with nebulized salbutamol 2/5 mL

Outcomes

Clinical state, FEV1, PEFR

Starting date

22 March 2014

Contact information

[email protected]

Notes

Saucedo 2015

Trial name or title

Nebulized Magnesium Sulfate as an Adjunct to Standard Therapy in Asthma Exacerbation

Methods

Randomized, double‐blind controlled trial

1 paediatric emergency department, Mexico

Participants

Inclusion criteria: clinical history of asthma, clinical diagnosis of moderate or severe asthma exacerbations, age 2 to 15 years

Exclusion criteria: coexistence of lung disease, severe kidney or liver disease, pregnancy, previous reaction to magnesium, no parental consent, prior inclusion in this study, presence of life‐threatening co‐morbidities, need for advanced airway management, life‐threatening symptoms.

Estimated enrolment: 152

Interventions

Intervention: nebulized salbutamol 2.5 mg (2 to 5 years) or 5 mg (≥ 6 years) and ipratropium bromide 250 mcg mixed with 2.5 mL of isotonic MgSO₄ (150 mg) per dose every 20 minutes during the first hour, continued with nebulized standard treatment every hour for 4 h, plus IV methylprednisolone or PO prednisolone 2 mg/kg/day for each treatment.

Control: nebulized salbutamol 2.5 mg (2‐5 years) or 5 mg (≥ 6 years) and ipratropium bromide 250 mcg mixed with 2.5 mL of isotonic saline per dose every 20 minutes during the first hour, continued with nebulized standard treatment every hour for 4 h, plus IV methylprednisolone or PO prednisolone 2 mg/kg/day for each treatment.

Outcomes

Primary outcome measure: change from Baseline Preschool Respiratory Assessment Measure (PRAM) at 20, 40, 60, 120, 180 and 240 minutes after beginning treatment.

Secondary outcome measures: rate of hospitalisation at 4 h, change from baseline heart rate, respiratory rate and blood pressure at 20, 40, 60, 120, 180 and 240 minutes after beginning treatment.

Starting date

September 2015

Contact information

[email protected]

Notes

Estimated study completion date: January 2018

Schuh 2016a

Trial name or title

Magnesium nebulization utilization in management of paediatric asthma (MagNUM PA) trial: study protocol for a randomized controlled trial

Methods

Randomized double‐blind controlled trial in 7 Canadian paediatric emergency departments

Participants

The trial will include 816 otherwise healthy children who are 2 to 17 years old, having had at least 1 previous wheezing episode, have received systemic corticosteroids, and have a Pediatric Respiratory Assessment Measure (PRAM) ≥ 5 points after 3 salbutamol and ipratropium treatments for a current acute asthma exacerbation.

Interventions

3 doses nebulized salbutamol with either 600 mg MgSO₄ or placebo 20 min apart.

Outcomes

Primary outcome: hospitalisation within 24 h of the start of the experimental therapy for persistent respiratory distress or supplemental oxygen.

Secondary outcomes include all‐cause hospitalisation within 24 h, PRAM, vital signs, number of bronchodilator treatments by 240 min, association between the difference in the primary outcome between the groups, age, gender, baseline PRAM, atopy, and “viral induced wheeze” phenotype.

Starting date

November 2014

Contact information

Suzanne Schuh: [email protected]
Division of Paediatric Emergency Medicine, The Hospital for Sick Children, Child Health Evaluative Sciences, SickKids Research Institute, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada

Notes

Estimated completed: December 2017

RCT: randomised controlled trial

Data and analyses

Open in table viewer
Comparison 1. MgSO4 + SABA + ipratropium versus SABA + ipratropium

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pulmonary function (% FEV1) Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

3.28 [1.06, 5.49]

Analysis 1.1

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 1 Pulmonary function (% FEV1).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 1 Pulmonary function (% FEV1).

1.1 90 minutes

1

60

Mean Difference (IV, Fixed, 95% CI)

8.57 [1.99, 15.15]

1.2 120 minutes

1

60

Mean Difference (IV, Fixed, 95% CI)

2.60 [0.25, 4.95]

2 Pulmonary function % predicted PEF Show forest plot

2

636

Mean Difference (IV, Fixed, 95% CI)

0.05 [‐2.33, 2.42]

Analysis 1.2

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 2 Pulmonary function % predicted PEF.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 2 Pulmonary function % predicted PEF.

3 Clinical severity scores (closest to 60 mins) Show forest plot

2

1130

Std. Mean Difference (IV, Fixed, 95% CI)

0.01 [‐0.11, 0.12]

Analysis 1.3

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 3 Clinical severity scores (closest to 60 mins).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 3 Clinical severity scores (closest to 60 mins).

3.1 Yung ASS at 60 minutes

1

472

Std. Mean Difference (IV, Fixed, 95% CI)

‐0.17 [‐0.35, 0.02]

3.2 Change in dyspnoea VAS at 60 minutes

1

658

Std. Mean Difference (IV, Fixed, 95% CI)

0.13 [‐0.02, 0.28]

4 Admission at first presentation Show forest plot

4

1308

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.91, 1.00]

Analysis 1.4

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 4 Admission at first presentation.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 4 Admission at first presentation.

4.1 Adults

3

800

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.87, 1.03]

4.2 Children

1

508

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.92, 1.01]

5 HDU/ITU admission Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.5

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 5 HDU/ITU admission.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 5 HDU/ITU admission.

5.1 Admission to HDU (adults)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Admission to ICU (adults)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Admission to PICU/HDU or intubation (children)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Readmission Show forest plot

2

750

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [0.84, 3.87]

Analysis 1.6

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 6 Readmission.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 6 Readmission.

7 Respiratory rate at 60 mins Show forest plot

2

723

Mean Difference (IV, Fixed, 95% CI)

0.70 [‐0.14, 1.53]

Analysis 1.7

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 7 Respiratory rate at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 7 Respiratory rate at 60 mins.

8 Heart rate at 60 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 8 Heart rate at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 8 Heart rate at 60 mins.

9 Systolic blood pressure at 60 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 9 Systolic blood pressure at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 9 Systolic blood pressure at 60 mins.

10 Diastolic blood pressure at 60 mins Show forest plot

1

674

Mean Difference (IV, Fixed, 95% CI)

2.40 [0.29, 4.51]

Analysis 1.10

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 10 Diastolic blood pressure at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 10 Diastolic blood pressure at 60 mins.

11 Serious adverse events (during admission) Show forest plot

2

557

Risk Difference (M‐H, Fixed, 95% CI)

‐0.03 [‐0.06, ‐0.00]

Analysis 1.11

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 11 Serious adverse events (during admission).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 11 Serious adverse events (during admission).

11.1 Adults

1

50

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.07, 0.07]

11.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.04 [‐0.06, ‐0.01]

12 Any adverse event (during admission) Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.01 [‐0.03, 0.05]

Analysis 1.12

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 12 Any adverse event (during admission).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 12 Any adverse event (during admission).

12.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.02 [‐0.02, 0.07]

12.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.02 [‐0.09, 0.05]

13 Serious adverse events (within 30 days) Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.13

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 13 Serious adverse events (within 30 days).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 13 Serious adverse events (within 30 days).

14 Any adverse event (within 30 days) Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.14

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 14 Any adverse event (within 30 days).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 14 Any adverse event (within 30 days).

15 Adverse event: hypotension Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.02 [‐0.01, 0.04]

Analysis 1.15

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 15 Adverse event: hypotension.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 15 Adverse event: hypotension.

15.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.03 [‐0.01, 0.07]

15.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.00 [‐0.02, 0.01]

16 Adverse event: flushing Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.00 [‐0.01, 0.01]

Analysis 1.16

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 16 Adverse event: flushing.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 16 Adverse event: flushing.

16.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.00 [‐0.01, 0.02]

16.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.00 [‐0.02, 0.01]

Open in table viewer
Comparison 2. MgSO4 + SABA versus SABA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pulmonary function % predicted FEV1 Show forest plot

4

208

Mean Difference (IV, Fixed, 95% CI)

3.34 [‐1.58, 8.26]

Analysis 2.1

Comparison 2 MgSO4 + SABA versus SABA, Outcome 1 Pulmonary function % predicted FEV1.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 1 Pulmonary function % predicted FEV1.

1.1 Adults

3

146

Mean Difference (IV, Fixed, 95% CI)

2.18 [‐3.30, 7.67]

1.2 Children

1

62

Mean Difference (IV, Fixed, 95% CI)

8.10 [‐3.03, 19.23]

2 % predicted FEV1: subgroup: severity Show forest plot

3

188

Mean Difference (IV, Fixed, 95% CI)

4.12 [‐1.81, 10.06]

Analysis 2.2

Comparison 2 MgSO4 + SABA versus SABA, Outcome 2 % predicted FEV1: subgroup: severity.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 2 % predicted FEV1: subgroup: severity.

2.1 Severe (FEV1 <50% predicted)

1

52

Mean Difference (IV, Fixed, 95% CI)

9.90 [0.05, 19.75]

2.2 Moderate

2

136

Mean Difference (IV, Fixed, 95% CI)

0.84 [‐6.59, 8.27]

3 Pulmonary function PEF L/min Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 2.3

Comparison 2 MgSO4 + SABA versus SABA, Outcome 3 Pulmonary function PEF L/min.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 3 Pulmonary function PEF L/min.

3.1 Adults

3

155

Mean Difference (IV, Fixed, 95% CI)

11.91 [‐4.12, 27.95]

3.2 Children

1

80

Mean Difference (IV, Fixed, 95% CI)

11.90 [‐6.86, 30.66]

4 Admission to hospital Show forest plot

6

375

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.52, 1.15]

Analysis 2.4

Comparison 2 MgSO4 + SABA versus SABA, Outcome 4 Admission to hospital.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 4 Admission to hospital.

4.1 Adults

4

213

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.45, 1.07]

4.2 Children

2

162

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.44, 2.98]

5 Heart rate at 120 mins Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 MgSO4 + SABA versus SABA, Outcome 5 Heart rate at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 5 Heart rate at 120 mins.

6 Respiratory rate at 120 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 MgSO4 + SABA versus SABA, Outcome 6 Respiratory rate at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 6 Respiratory rate at 120 mins.

7 Diastolic blood pressure at 120 mins Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

0.72 [‐1.35, 2.80]

Analysis 2.7

Comparison 2 MgSO4 + SABA versus SABA, Outcome 7 Diastolic blood pressure at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 7 Diastolic blood pressure at 120 mins.

8 Systolic blood pressure at 120 mins Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

0.89 [‐2.69, 4.48]

Analysis 2.8

Comparison 2 MgSO4 + SABA versus SABA, Outcome 8 Systolic blood pressure at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 8 Systolic blood pressure at 120 mins.

9 Serious adverse events Show forest plot

5

243

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.04, 0.04]

Analysis 2.9

Comparison 2 MgSO4 + SABA versus SABA, Outcome 9 Serious adverse events.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 9 Serious adverse events.

9.1 Adults

4

181

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.04, 0.04]

9.2 Children

1

62

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.06, 0.06]

10 Any adverse events Show forest plot

5

694

Risk Difference (M‐H, Fixed, 95% CI)

‐0.01 [‐0.05, 0.03]

Analysis 2.10

Comparison 2 MgSO4 + SABA versus SABA, Outcome 10 Any adverse events.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 10 Any adverse events.

10.1 Adults

4

329

Risk Difference (M‐H, Fixed, 95% CI)

‐0.02 [‐0.10, 0.06]

10.2 Children

1

365

Risk Difference (M‐H, Fixed, 95% CI)

‐0.01 [‐0.03, 0.01]

Open in table viewer
Comparison 3. MgSO4 versus SABA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical severity score Show forest plot

3

93

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.62, 0.36]

Analysis 3.1

Comparison 3 MgSO4 versus SABA, Outcome 1 Clinical severity score.

Comparison 3 MgSO4 versus SABA, Outcome 1 Clinical severity score.

1.1 Fischl index final score (120 mins)

1

33

Mean Difference (IV, Fixed, 95% CI)

‐0.33 [‐1.07, 0.41]

1.2 Fischl index score (time point unclear)

1

40

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐1.11, 0.71]

1.3 Change in Fischl index at 120 mins

1

20

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.67, 1.27]

2 Admission to hospital Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3 MgSO4 versus SABA, Outcome 2 Admission to hospital.

Comparison 3 MgSO4 versus SABA, Outcome 2 Admission to hospital.

3 Heart rate (120 mins) Show forest plot

1

20

Mean Difference (IV, Fixed, 95% CI)

21.20 [0.17, 42.23]

Analysis 3.3

Comparison 3 MgSO4 versus SABA, Outcome 3 Heart rate (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 3 Heart rate (120 mins).

4 Respiratory rate Show forest plot

2

60

Mean Difference (IV, Fixed, 95% CI)

‐2.40 [‐3.91, ‐0.89]

Analysis 3.4

Comparison 3 MgSO4 versus SABA, Outcome 4 Respiratory rate.

Comparison 3 MgSO4 versus SABA, Outcome 4 Respiratory rate.

5 Systolic pressure (120 mins) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.5

Comparison 3 MgSO4 versus SABA, Outcome 5 Systolic pressure (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 5 Systolic pressure (120 mins).

6 Diastolic pressure (120 mins) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.6

Comparison 3 MgSO4 versus SABA, Outcome 6 Diastolic pressure (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 6 Diastolic pressure (120 mins).

7 Serious adverse events Show forest plot

2

53

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.10, 0.10]

Analysis 3.7

Comparison 3 MgSO4 versus SABA, Outcome 7 Serious adverse events.

Comparison 3 MgSO4 versus SABA, Outcome 7 Serious adverse events.

8 Mild‐Moderate Side Effects Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.8

Comparison 3 MgSO4 versus SABA, Outcome 8 Mild‐Moderate Side Effects.

Comparison 3 MgSO4 versus SABA, Outcome 8 Mild‐Moderate Side Effects.

Study flow diagram: review update
Figuras y tablas -
Figure 1

Study flow diagram: review update

'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.

In the control group 82 people out of 100 had hospital admission , compared to 78 (95% CI 75 to 82) out of 100 for the active treatment group.
Figuras y tablas -
Figure 3

In the control group 82 people out of 100 had hospital admission , compared to 78 (95% CI 75 to 82) out of 100 for the active treatment group.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 1 Pulmonary function (% FEV1).
Figuras y tablas -
Analysis 1.1

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 1 Pulmonary function (% FEV1).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 2 Pulmonary function % predicted PEF.
Figuras y tablas -
Analysis 1.2

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 2 Pulmonary function % predicted PEF.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 3 Clinical severity scores (closest to 60 mins).
Figuras y tablas -
Analysis 1.3

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 3 Clinical severity scores (closest to 60 mins).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 4 Admission at first presentation.
Figuras y tablas -
Analysis 1.4

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 4 Admission at first presentation.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 5 HDU/ITU admission.
Figuras y tablas -
Analysis 1.5

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 5 HDU/ITU admission.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 6 Readmission.
Figuras y tablas -
Analysis 1.6

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 6 Readmission.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 7 Respiratory rate at 60 mins.
Figuras y tablas -
Analysis 1.7

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 7 Respiratory rate at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 8 Heart rate at 60 mins.
Figuras y tablas -
Analysis 1.8

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 8 Heart rate at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 9 Systolic blood pressure at 60 mins.
Figuras y tablas -
Analysis 1.9

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 9 Systolic blood pressure at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 10 Diastolic blood pressure at 60 mins.
Figuras y tablas -
Analysis 1.10

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 10 Diastolic blood pressure at 60 mins.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 11 Serious adverse events (during admission).
Figuras y tablas -
Analysis 1.11

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 11 Serious adverse events (during admission).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 12 Any adverse event (during admission).
Figuras y tablas -
Analysis 1.12

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 12 Any adverse event (during admission).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 13 Serious adverse events (within 30 days).
Figuras y tablas -
Analysis 1.13

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 13 Serious adverse events (within 30 days).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 14 Any adverse event (within 30 days).
Figuras y tablas -
Analysis 1.14

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 14 Any adverse event (within 30 days).

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 15 Adverse event: hypotension.
Figuras y tablas -
Analysis 1.15

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 15 Adverse event: hypotension.

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 16 Adverse event: flushing.
Figuras y tablas -
Analysis 1.16

Comparison 1 MgSO4 + SABA + ipratropium versus SABA + ipratropium, Outcome 16 Adverse event: flushing.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 1 Pulmonary function % predicted FEV1.
Figuras y tablas -
Analysis 2.1

Comparison 2 MgSO4 + SABA versus SABA, Outcome 1 Pulmonary function % predicted FEV1.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 2 % predicted FEV1: subgroup: severity.
Figuras y tablas -
Analysis 2.2

Comparison 2 MgSO4 + SABA versus SABA, Outcome 2 % predicted FEV1: subgroup: severity.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 3 Pulmonary function PEF L/min.
Figuras y tablas -
Analysis 2.3

Comparison 2 MgSO4 + SABA versus SABA, Outcome 3 Pulmonary function PEF L/min.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 4 Admission to hospital.
Figuras y tablas -
Analysis 2.4

Comparison 2 MgSO4 + SABA versus SABA, Outcome 4 Admission to hospital.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 5 Heart rate at 120 mins.
Figuras y tablas -
Analysis 2.5

Comparison 2 MgSO4 + SABA versus SABA, Outcome 5 Heart rate at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 6 Respiratory rate at 120 mins.
Figuras y tablas -
Analysis 2.6

Comparison 2 MgSO4 + SABA versus SABA, Outcome 6 Respiratory rate at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 7 Diastolic blood pressure at 120 mins.
Figuras y tablas -
Analysis 2.7

Comparison 2 MgSO4 + SABA versus SABA, Outcome 7 Diastolic blood pressure at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 8 Systolic blood pressure at 120 mins.
Figuras y tablas -
Analysis 2.8

Comparison 2 MgSO4 + SABA versus SABA, Outcome 8 Systolic blood pressure at 120 mins.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 9 Serious adverse events.
Figuras y tablas -
Analysis 2.9

Comparison 2 MgSO4 + SABA versus SABA, Outcome 9 Serious adverse events.

Comparison 2 MgSO4 + SABA versus SABA, Outcome 10 Any adverse events.
Figuras y tablas -
Analysis 2.10

Comparison 2 MgSO4 + SABA versus SABA, Outcome 10 Any adverse events.

Comparison 3 MgSO4 versus SABA, Outcome 1 Clinical severity score.
Figuras y tablas -
Analysis 3.1

Comparison 3 MgSO4 versus SABA, Outcome 1 Clinical severity score.

Comparison 3 MgSO4 versus SABA, Outcome 2 Admission to hospital.
Figuras y tablas -
Analysis 3.2

Comparison 3 MgSO4 versus SABA, Outcome 2 Admission to hospital.

Comparison 3 MgSO4 versus SABA, Outcome 3 Heart rate (120 mins).
Figuras y tablas -
Analysis 3.3

Comparison 3 MgSO4 versus SABA, Outcome 3 Heart rate (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 4 Respiratory rate.
Figuras y tablas -
Analysis 3.4

Comparison 3 MgSO4 versus SABA, Outcome 4 Respiratory rate.

Comparison 3 MgSO4 versus SABA, Outcome 5 Systolic pressure (120 mins).
Figuras y tablas -
Analysis 3.5

Comparison 3 MgSO4 versus SABA, Outcome 5 Systolic pressure (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 6 Diastolic pressure (120 mins).
Figuras y tablas -
Analysis 3.6

Comparison 3 MgSO4 versus SABA, Outcome 6 Diastolic pressure (120 mins).

Comparison 3 MgSO4 versus SABA, Outcome 7 Serious adverse events.
Figuras y tablas -
Analysis 3.7

Comparison 3 MgSO4 versus SABA, Outcome 7 Serious adverse events.

Comparison 3 MgSO4 versus SABA, Outcome 8 Mild‐Moderate Side Effects.
Figuras y tablas -
Analysis 3.8

Comparison 3 MgSO4 versus SABA, Outcome 8 Mild‐Moderate Side Effects.

Summary of findings for the main comparison. MgSO4 + SABA + ipratropium compared to SABA + ipratropium in the treatment of acute asthma

MgSO+ SABA + ipratropium compared to SABA + ipratropium in the treatment of acute asthma

Patient or population: adults and children with acute exacerbation of asthma
Setting: emergency department/inpatient
Intervention: MgSO₄ + SABA + ipratropium
Comparison: SABA + ipratropium

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with SABA + ipratropium

Risk with MgSO4 + SABA + ipratropium

Pulmonary function (% predicted FEV1)

(90 to 120 minutes)

The mean pulmonary function (% predicted FEV1) was 65%

% predicted FEV1 was 3.28% higher
(1.06 higher to 5.49 higher)

120
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2 3

Outcome measured at 90 mins in 1 study and 120 mins in the other.

1 study (Gaur 2008) has reported much smaller standard deviations and contributes almost 90% of analysis weight

Pulmonary function % predicted PEF

(60 minutes)

The mean pulmonary function % predicted PEF was 50.45%

% predicted PEF was 0.05 higher
(2.33 lower to 2.42 higher)

636
(2 RCTs)

⊕⊕⊕⊝
MODERATE 2 4 5

Both studies in adults

Mean control group % predicted PEF was 36% in 1 study and 64.9% in the other

Clinical severity scores

(60 minutes)

The mean dyspnoea VAS was 31.8; the mean Yung ASS was 4.95

SMD 0.01 higher
(0.11 lower to 0.12 higher)

1130
(2 RCTs)

⊕⊕⊝⊝
LOW 2 6

1 study reported Yung ASS and the other change in dyspnoea VAS

Admission at first presentation

819 per 1000

778 per 1000
(745 to 819)

RR 0.95
(0.91 to 1.00)

1308
(4 RCTs)

⊕⊕⊕⊝
MODERATE 7 8 9

Adults vs children test for subgroup difference: P = 0.72, I² = 0%

Readmission

(7 to 30 days)

26 per 1000

46 per 1000
(22 to 100)

RR 1.80
(0.84 to 3.87)

750
(2 RCTs)

⊕⊕⊝⊝
LOW 10

Outcome measured at 7 days in 1 study and 30 days in the other.

Serious adverse events (during admission)

43 per 1000

Not estimable. See comment.

557
(2 RCTs)

⊕⊕⊕⊝
MODERATE 11

Risk difference: −0.03 (95% CI −0.06 to 0.00)

Adults vs children test for subgroup difference: P = 0.39, I² = 0%

Goodacre 2013 also reported participants with 1 or more SAE within 30 days: 35/332 in the MgSO₄ group and 28/358 in the placebo group (RD: 0.03; 95% CI −0.02 to 0.07)

Any adverse event (during admission)

144 per 1000

Not estimable. See comment.

1197
(2 RCTs)

⊕⊕⊕⊕
HIGH

Risk Difference: 0.01 (95% CI −0.03 to 0.05)

Adults vs children test for subgroup difference: P = 0.34, I² = 0%

Goodacre 2013 also reported participants with 1 or more adverse event within 30 days: 52/332 in the MgSO₄ group and 36/358 in the placebo group (OR 1.66, 95% CI 1.05 to 2.62)

*The risk in the intervention group (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).

ASS: asthma severity score; CI: Confidence interval; RD: risk difference; RR: Risk ratio; OR: Odds ratio; VAS: visual analogue scale

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

1 One study contributing most of weight at unclear risk of bias in multiple domains (−1 study limitations)

2 I² > 50% (−1 inconsistency)

3 Studies equal size but one study contributes almost 90% of weight to analysis due to much smaller standard deviations. Result no longer significant if random‐effects model applied (−1 imprecision)

4 Although one study at unclear risk of bias in several domains, the larger study, which contributes vast majority of weight to analysis, if of high methodological quality (no downgrade)

5 Although confidence interval includes no difference, they are sufficiently tight to effectively rule out an important between‐group difference (no downgrade)

6 Confidence intervals include both harm and benefit of intervention (−1 imprecision)

7 Although two of the studies at unclear risk of bias in several domains the two large studies contributing nearly 95% of weight in analysis are both of high methodological quality (no downgrade)

8 Although the I² = 52%, the two large studies contributing to this analysis show consistent results (no downgrade)

9 Confidence intervals include no difference (−1 imprecision)

10 Confidence intervals include no difference and appreciable harm or benefit of the intervention (−2 imprecision)

11 Events rare and confidence intervals include no difference (−1 imprecision)

Figuras y tablas -
Summary of findings for the main comparison. MgSO4 + SABA + ipratropium compared to SABA + ipratropium in the treatment of acute asthma
Summary of findings 2. MgSO4 + SABA compared to SABA in the treatment of acute asthma

MgSO+ SABA compared to SABA in the treatment of acute asthma

Patient or population: adults and children with acute exacerbation of asthma
Setting: emergency department/inpatient
Intervention: MgSO₄ + SABA
Comparison: SABA

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with SABA

Risk with MgSO4 + SABA

Pulmonary function % predicted FEV1

(20 minutes to 2 to 3 h)

The mean pulmonary function % predicted FEV1 was 56.55%

% predicted FEV1 was 3.34% higher
(1.58 lower to 8.26 higher)

208
(4 RCTs)

⊕⊕⊝⊝
LOW 1 2

Adults vs children test for subgroup difference: P = 0.35, I² = 0%

Severe vs moderate asthma exacerbation test for subgroup difference: P = 0.15, I² = 51.8% (favouring a greater effect in the more severe subgroup)

Pulmonary function PEF L/min ‐ Adults

(20 minutes to 2 to 3 h)

The mean pulmonary function PEF was 233 L/min

PEF was 11.91 L/min higher
(4.12 lower to 27.95 higher)

155
(3 RCTs)

⊕⊕⊝⊝
LOW 1 2

Pulmonary function PEF L/min ‐ Children

(60 minutes)

The mean pulmonary function PEF was 143.5

PEF was 11.9 L/min higher
(6.86 lower to 30.66 higher)

80
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

Admission to hospital at initial presentation

202 per 1000

158 per 1000 (105 to 233)

RR 0.78, (0.52 to 1.15)

375
(6 RCTs)

⊕⊕⊝⊝
LOW 1 2

Adults vs children test for subgroup difference: P = 0.35, I² = 0%

Serious adverse events

(During ED/hospital admission)

Not estimable

Not estimable. See comment

243
(5 RCTs)

⊕⊕⊝⊝
LOW 1 4

Risk difference: 0.00 (95% CI −0.04 to 0.04)

No events reported

Any adverse events

(During ED/hospital admission)

107 per 1000

Not estimable. See comment

694
(5 RCTs)

⊕⊕⊝⊝
LOW 1 2

Risk difference: −0.01 (95% CI −0.05 to 0.03)

Adults vs children test for subgroup difference: P = 0.77, I² = 0%

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; ED: emergency department; FEV1: forced expiratory volume in 1 second; OR: Odds ratio; PEF: peak expiratory flow; RD: risk difference; RR: Risk ratio

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

1 Several studies were at unclear or high risk of bias in one or more domain (−1 study limitations)

2 Confidence intervals include both possible harm and benefit of the intervention (−1 imprecision)

3 Study at unclear risk of bias in several domains (−1 study limitations)

4 No events reported but less than 250 participants in total. Risk difference confidence intervals include a possible important harm or benefit of the intervention (−1 imprecision)

Figuras y tablas -
Summary of findings 2. MgSO4 + SABA compared to SABA in the treatment of acute asthma
Summary of findings 3. MgSO4 compared to SABA in the treatment of acute asthma

MgSOcompared to SABA in the treatment of acute asthma

Patient or population: adults and children with acute exacerbation of asthma
Setting: emergency department/inpatient
Intervention: MgSO₄
Comparison: SABA

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with SABA

Risk with MgSO4

Lung function

Reported narratively in text

Clinical severity score ‐ Fischl index

(120 minutes)

The Fischl index score was 2.1

Fischl index score 0.13 lower
(0.62 lower to 0.36 higher)

93
(3 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2 3

Time point 120 minutes in 2 studies and unclear in the third study

Wide range of control group scores (0.3, 0.76 and 4.81). Scale out of 7 with higher score indicating more severe symptoms. 4.81 reported in study with unclear time point.

Admission to hospital at initial presentation

118 per 1000

62 per 1000
(6 to 625)

RR 0.53
(0.05 to 5.31)

33
(1 RCT)

⊕⊝⊝⊝
VERY LOW 4 5

Serious adverse events

(During ED/hospital admission)

Not estimable

Not estimable. See comment

53
(2 RCTs)

⊕⊕⊝⊝
LOW 1 6

Risk difference: 0.00 (95% CI −0.10 to 0.10)

No events reported

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; RR: Risk ratio; OR: Odds ratio;

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

1 Several studies at unclear or high risk of bias in one or more domains (−1 study limitations)

2 Confidence intervals include both possible harm and benefit of the intervention (−1 imprecision)

3 Time‐point for measurement unclear in one study (−1 indirectness)

4 Study at unclear risk of bias in several domains (−1 study limitations)

5 One small study. Confidence intervals include appreciable harm or benefit of the intervention (−2 imprecision)

6 Two small studies. No events reported. Risk difference confidence intervals include appreciable harm or benefit of the intervention (−1 for imprecision)

Figuras y tablas -
Summary of findings 3. MgSO4 compared to SABA in the treatment of acute asthma
Table 1. Summary of Severity

Study

Severity of asthma exacerbation

Diagnosis based on

Population (adult/mixed/paediatric)

MgSO₄ and SABA and Ipratropium bromide versus SABA and Ipratropium

Ashtekar 2008

Severe

BTS definition clinical features

Paediatric (2 to 16)

Drobina 2006

Unclear

PEF and clinical signs

Adults

Gallegos‐Solórzano 2010

Moderate to severe

FEV1 < 60%

Adults >18

Gaur 2008

Severe

FEV1 < 30%

Adults (18 to 60)

Goodacre 2013

Severe

BTS definition

Adult (≥ 16)

Hossein 2016

Moderate to severe

PEF < 70% and clinical signs

Adult (> 16)

Powell 2013

Severe after conventional treatment

BTS definition

Paediatric (2 to 16)

MgSO4 and SABA versus SABA

Abreu‐Gonzalez 2002

Moderate

FEV1 and PEF at baseline

Adults

Aggarwal 2006

Severe and life threatening

BTS definition clinical features and PEF

Mixed (13 to 60)

Ahmed 2013

Severe

PEF

Not documented

Alansari 2015

Moderate to severe

Clinical score

Paediatric (2 to 14)

Badawy 2014

Unclear

N/A

Adult

Bessmertny 2002

Moderate to severe

PEF between  40% to 80%

Adults (18 to 65)

Dadhich 2005

Severe

PEF < 50%

Adults

Hughes 2003

Severe

FEV1 < 50%

Adults (16 to 65)

Khashabi 2008

Unclear

Clinically defined as respiratory distress

Paediatric (mean age 3.55 years)

Kokturk 2005

Moderate to severe

Clinical scores and PEF

Adults (18 to 60)

Mahajan 2004

Moderate to severe

FEV1 between 45% and 75%

Paediatric (5 to 17)

Mohammedzadeh 2014

Moderate to severe

GINA definition

Paediatric (5 to 14)

Nannini 2000

Severe

PEF < 50%

Adult (> 18)

Sarhan 2016

Unclear

PEF < 300L/min

Mixed (11 to 70)

Turker 2017

Moderate

Not described

Children (3 to 15)

MgSO₄ versus SABA

Dadhich 2005

Severe

PEF < 50%

Adults

Mangat 1998

Moderate to severe

PEF < 300 L/Min

Mixed (12 to 60)

Meral 1996

Moderate to severe

PEF < 75%

Paediatric

Neki 2006

Severe

FEV1 < 40% or PEF < 300 L/Min

Adult (15 to 60)

Sarhan 2016

Unclear

PEF < 300L/min

Mixed (11 to 70)

BTS: British Thoracic Society

GINA: Global Initiative for Asthma
FEV1: Forced expiratory volume in one second
PEF: Peak Expiratory Flow Rate

Figuras y tablas -
Table 1. Summary of Severity
Table 2. Summary of  Characteristics of the studies – where patients were recruited from, additional treatment, exclusion criteria and side effects.

Study

Presentation to which department?

Origin

Primary outcome(s)

Total n randomised

Side effects (patients in study)

Pharmaceutical exclusions

Other Interventions

MgSO₄ and SABA and Ipratropium bromide versus SABA and Ipratropium

Ashtekar 2008

Children’s Assessment Unit after GP referral

Cardiff, Wales

ASS (Yung)

17

1 tingling in fingers and 1 transient hypotension

None stated

All management followed the BTS/SIGN guidelines; all children received 2 mg/kg prednisolone

Drobina 2006

ED

USA

PEF, admissions

110

No comment on side effects in paper

Not stated

All subjects received 50 mg of oral prednisone at the onset of the treatment

Gallegos‐Solórzano 2010

ED

Mexico City, Mexico

% change FEV1,

O₂ post treatment, admission rates

112

Dry and bitter mouth (MgSO₄ group 1), dizziness (MgSO₄ 1; placebo 1)

Use of steroids prior to presentation

All participants received one IV dose of 125 mg methylprednisolone at admission and 1 mg/kg/day for 10 days prednisolone,on discharge. Other treatments were administered according to the treating physician

Gaur 2008

ED

Delhi, India

FEV1

60

None reported

None stated

All participants received IV hydrocortisone on arrival

Goodacre 2013

ED

UK

Admission within 7d, visual analogue scale for breathlessness at 2 h

703

AEs (41 MgSO₄/salbutamol; 36 placebo/salbutamol)

MgSO₄ in the past 24 h

All participants were managed according to BTS/SIGN guidelines (consisting of
oxygen, nebulised salbutamol (5 mg), nebulised ipratropium (500 μg), and oral prednisolone administered during recruitment, followed by up to 5 mg salbutamol added to each trial nebuliser. Other treatments were provided at the discretion of the clinician

Hossein 2016

ED

Tehran, Iran

PEFR improvement, admission rate

50

No serious side effects reported

None stated

All participants received 50 mg oral prednisolone

Powell 2013

ED and children's assessment units

UK

Yung asthma severity score

508

47 in MgSO₄ group and 59 in control group

None

Hospital‐defined conventional treatment

MgSO4 and SABA versus SABA

Abreu‐Gonzalez 2002

Tenerife Spain

FEV1, PEF

24

None reported

None stated

Not stated

Aggarwal 2006

ED

New Delhi India

PEF

100

Palpitations (MgSO₄/salbutamol 13; salbutamol/placebo 11) and tremors (7; 7).

None stated

Clinicians free to administer steroids, salbutamol, IV hydrocortisone if judged to be required

Ahmed 2013

Mymensingh, Bangladesh

PEF

120

None reported

None stated

Not stated

Alansari 2015

Paediatric emergency centre

Doha, Qatar

Time to readiness for discharge

400

Chest tightness and facial rash (MgSO₄/salbutamol 191), excessive cough (placebo/salbutamol 174)

None stated

All participants received methylprednisolone 1 mg/kg IV every 12h and additional nebulised albuterol at clinicians' discretion

Badawy 2014

Outpatient department and ED

Sohag, Egypt

Exacerbations post intervention, delivery outcome, post‐partum health status

60

None reported

None stated

All participants received 100 mg hydrocortisone IV, 500 mg aminophylline IV

Bessmertny 2002

ED

Brooklyn, USA

FEV1 (% pred)

74

No SAEs reported

No theophylline or anticholinergics 2 h prior to presentation

Intravenous hydrocortisone, 2 mg/kg
every 6 h, was administered to patients who failed to show an adequate improvement of pulmonary function after 3 initial doses of albuterol

Dadhich 2005

ED

Ajmer India

PEF

71

"Side effects were self limiting"

Not stated

Not stated

Hughes 2003

ED

Wellington New Zealand

FEV1

52

None reported

None

All participants received 100 mg hydrocortisone IV

Khashabi 2008

Urmia, Iran

Reduced mean duration of O₂ therapy in MgSO₄ group,

no change in Respiratory Distress Score)

40

No side effects

Not stated

Not stated

Kokturk 2005

ED

Gazi, Turkey

PEF difference

26

Transient hypotension (1 MgSO₄), palpitation (1 salbutamol)

None

All participants received 1 mg/kg prednisolone. Theophylline, anticholinergics and salbutamol given at clinicians discretion

Mahajan 2004

ED

Detroit, USA

% change in FEV1

62

No side effects

Steroids, ipratropium or theophylline in the last 3 days.

All participants received 2 mg/kg of prednisone

Mohammedzadeh 2014

Babol, Iran

Pulmonary index, PEFR, adjusted PEFR

80

Corticosteroids; steroids, theophylline or ipratropium use within last 72 h

Not stated

Nannini 2000

ED

4 hospitals in Argentina

PEF, admissions

35

None reported

Oral or parenteral steroids in the last 7 days

No other medications were permitted during the study except
supplemental oxygen; if the patient’s condition worsened, a 2.5 mg dose of nebulized salbutamol was administered at the discretion of the treating physician

Sarhan 2016

Chest and ED

Minia, Egypt

Clinical improvement, PEFR

30

None severe enough to warrant withdrawal

Bronchodilators in last 6 h, steroids in last 12 h

Nebulised salbutamol, IV hydrocortisone, IV aminophylline at clinicians' discretion

Turker 2017

ED

Turkey

Modified pulmonary index score

100

"No side effect caused by magnesium was observed in any of the patients in the study"

Not stated

Nebulised salbutamol (0.15 mg/kg), methylprednisolone 1 mg/kg IV; Oxygen was given to patients with SaO2≤ 95%

MgSO₄ versus SABA

Dadhich 2005

ED

Ajmer India

PEF

71

"Side effects were self limiting"

Not stated

Not stated

Mangat 1998

ED

St John’s College, India

PEF, Fischl index score, admissions

33

Transient self limiting hypotension (1) palpitation (1) tremors (2) all in control group and only 1 transient hypotension in MgSO₄ group (33)

Oral parenteral bronchodilators (6 h) steroids (last 12 h)

All participants received 100 mg hydrocortisone IV

Meral 1996

Izmir, Turkey

% change in PEF

ASS (Davies Leffert, Dabbous score)

40

No side effects

Beta2‐agonists or theophylline in the last 12 h

No other medication given

Neki 2006

Amritsar Punjab

PEF, RR, Fischl index

40

Oral, inhaled or parenteral steroids in last 12 h

All participants received 100 mg hydrocortisone IV

Sarhan 2016

Chest and ED

Minia, Egypt

Clinical improvement, PEFR

30

None severe enough to warrant withdrawal

Bronchodilators in last 6 h, steroids in last 12 h

Nebulised salbutamol, IV hydrocortisone, IV aminophylline at clinicians' discretion

ASS: Asthma Severity Score; BP: blood pressure; ED: emergency department; FEV1: Forced expiratory volume in 1 second; h: hour(s)
HR: heart rate; IV: intravenous; MgSO₄: magnesium sulfate; PEF: Peak Expiratory Flow Rate; SAEs: serious adverse events

Figuras y tablas -
Table 2. Summary of  Characteristics of the studies – where patients were recruited from, additional treatment, exclusion criteria and side effects.
Table 3. Summary of Interventions

Study (N)

Magnesium sulfate

Control

Dose

N

Co‐interventions

Dose

N

Co‐interventions

MgSO₄ and SABA and Ipratropium bromide versus SABA and Ipratropium

Ashtekar 2008

2.5 mL isotonic MgSO₄ (151 mg /dose)

7

500 mcg Ipratropium bromide

2.5 mg salbutamol or 5 mg salbutamol (depending on age) 3 times per h

2.5 mL of isotonic saline)

10

Same as for MgSO₄ group

Drobina 2006

150 mg MgSO₄ (0.3 mL of 50% MgSO₄ heptahydrate)

60

Albuterol sulfate (0.5%) 5 mg/mL) and 0.5 mg ipratropium bromide (0.02% inhalation solution) (frequency*)

No placebo so volume will be less: i.e. blinding may be an issue)

50

Same as for MgSO₄ group

Gallegos‐Solórzano 2010

3 mL (333 mg) of 10% isotonic MgSO₄ (1 g/10 mL)

60 (30 withdrawals)

2.5 mg albuterol and 500 mcg ipratropium 3 doses per hour

3 mL isotonic saline

52 (22 withdrawals)

Same as for MgSO₄ group

Gaur 2008

3 mL (3.2 g%)

isotonic MgSO₄

30

Salbutamol and ipratropium (dose*, frequency*)

Saline

30

Same as for MgSO₄ group

Goodacre 2013

2 mmol MgSO₄

339 (7 withdrawal)

7.5 mL 0.9% NaCl nebulised, 3 doses; 100 mL 0.9% NaCl IV once, BTS/SIGN standard treatments plus others at clinicians' discretion

7.5 mL 0.9% saline nebulised, 3 doses, 100 mL 0.9% NaCl IV once

364 (7 withdrawal)

BTS/SIGN standard treatments plus others at clinicians' discretion

Hossein 2016

3 mL (260 mmol/L) MgSO4

25

2.5 mg salbutamol, 0.5 mg ipratropium nebulised every 20 to 60 minutes, 50 mg oral prednisolone (once*)

3 mL 0.9% NaCl

25

Same as for MgSO₄ group

Powell 2013

2.5 mL 250 mmol/L MgSO₄

252 (13 withdrawals)

3 doses every 20 min. Hospital‐defined conventional treatment

2.5 mL isotonic saline

256 (10 withdrawals)

Same as for MgSO₄ group

MgSO4 and SABA versus SABA

Abreu‐Gonzalez 2002

2 mL MgSO₄ (isotonic)

13

400 mcg salbutamol

(once*)

2 mL of a physiological serum of an inhaled form

11 patients

11

400 mcg salbutamol

Aggarwal 2006

1 mL of 500 mg/mL MgSO₄

50

1 mL salbutamol (dose*, 8 mL distilled water, (295 mOsml/kg) 3 times per h

ultrasonic nebuliser

7.5 mL normal saline

50

1 mL salbutamol (dose*),

1.5 mL distilled water

(287 mOsml/kg) 3 times per h

Ahmed 2013

MgSO₄ (dose* frequency*)

60

Not recorded

Normal saline (dose* frequency*)

60

Not recorded

Alansari 2015

800 mg (15 mL) MgSO₄

208 (17 withdrawals)

5 mg albuterol, divided into 3 doses over 1 h. Methylprednisolone 1 mg/kg IV every 12 h. 3 doses nebulized 1 mL albuterol (5 mg/mL), 250 mcg ipratropium, 2 mL normal saline before trial doses started

15 mL 0.9% NaCl

192 (18 withdrawals)

Same as for MgSO₄ group

Badawy 2014

500 mg (1mL) MgSO₄

30

1 mL salbutamol solution (dose*), 8 mL 0.9% NaCl, max 3 doses with 20 mins apart. 100 mg hydrocortisone IV, 500 mg aminophylline IV (once*)

1 mL 0.9% NaCl

30

Same as for MgSO₄ group

Bessmertny 2002

MgSO₄ (384 mg)

37 (3 withdrawals)

Followed by ( i.e. not mixed) albuterol 2.5 mg/mL 3 times per h

Normal saline (no volume documented)

37 (3 withdrawals)

Same as for MgSO₄ group

Dadhich 2005

MgSO₄

26

No doses in any group or co‐interventions described

Not stated

24

No doses in any group or co‐interventions described

Hughes 2003

2.5 mL isotonic MgSO₄ (250 mmol/L 151 mg)

28 patients

28

2.5 mg salbutamol 3 times per

30 minutes

2.5 mL normal saline

24

Same as for MgSO₄ group

Khashabi 2008

Isotonic MgSO₄

(dose*, frequency*)

*

Salbutamol (dose*)

2.5 mL normal saline (frequency*)

*

Same as for MgSO₄ group

Kokturk 2005

Isotonic MgSO₄ (2.5 mL)

14

Salbutamol (dose*) 3

times per h then 1 per h for 3 h

2.5 mL normal saline

12

Same as for MgSO₄ group

Mohammedzadeh 2014

3 mL 7.5% MgSO₄

40

0.15 mg/kg salbutamol 3 doses, every 20 min

3 mL normal saline

40

Same as for MgSO₄ group

Mahajan 2004

2.5 mL Isotonic (6.3%) MgSO₄ solution

31

Albuterol 2.5 mg 1 dose

2.5 mL normal saline

31

Same as for MgSO₄ group

Nannini 2000

3 mL isotonic MgSO₄

(286 mOsml, 7.5%, 225 mg)

19

0.5 mL 2.5 mg salbutamol

1 dose*

3 mL normal saline

16

Same as for MgSO₄ group

Sarhan 2016

2.5 mL MgSO4 (100 mg), 0.5 mL salbutamol (2.5 mg)

10

4 doses at 20 min intervals. If needed: additional nebulised salbutamol, IV hydrocortisone, IV aminophylline

2.5 mL isotonic saline

10

Same as for MgSO4 group

Turker 2017

1 mL magnesium sulfate (15%) + 1.5 mL isotonic saline

50

3 doses at 20 min intervals. Also nebulised salbutamol (0.15 mg/kg), methylprednisolone 1 mg/kg IV; Oxygen was given to patients with SaO2 ≤ 95%

1.5 mL isotonic saline

50

Same as for MgSO₄ group

MgSO₄ versus SABA

Dadhich 2005

MgSO₄

21

No doses in any group or co‐interventions described

Not stated

24

No doses in any group or co‐interventions described

Mangat 1998

3.2% solution MgSO₄ = 95 mg)

16

4 doses every 20 minutes

3 mL (2.5 mg) salbutamol

17

Four doses every 20 minutes

Meral 1996

2 mL MgSO₄ (280 mmol/L)

20

1* dose given over 10 to 15 minutes

Salbutamol 2.5 mg in 2.5 mL

20

1 dose* given over 10 to 15 minutes

Neki 2006

20 patients

3.2 G % MgSO₄

20

4 doses every 20 min

3 mL of 25 mg* salbutamol (likely decimal point missing)

20

Same as for MgSO4 group

Sarhan 2016

3 mL (100 mg) MgSO4

10

4 doses at 20 min intervals. If needed: additional nebulised salbutamol, IV hydrocortisone, IV aminophylline

0.5 mL salbutamol (2.5 mg)

10

Same as for MgSO₄ group

TOTAL: 2907 randomised to comparisons of interest. 130 withdrawn, 2777 completed

TOTAL: 1476 randomised, 70 withdrawn = 1406 completed intervention

TOTAL: 1431 randomised, 60 withdrawn = 1371 completed control

* denotes uncertainty

Figuras y tablas -
Table 3. Summary of Interventions
Table 4. Outcomes

Study ID (author, date of publication)

Review primary outcomes

Review secondary outcomes

FEV1

PEF

Clinical severity scores

Hospital admissions

Duration of symptoms

Vital signs

Adverse effects

MgSO₄ and SABA and Ipratropium bromide versus SABA and Ipratropium

Ashtekar 2008

N

N

Y

N

N

N

Y

Drobina 2006

N

P

N

N

N

N

P

Gallegos‐Solórzano 2010

Y

N

N

N

N

N

Y

Gaur 2008

Y

N

N

N

N

N

N

Goodacre 2013

N

Y

N

Y

N

Y

Y

Hossein 2016

N

Y

Y

Y

N

Y

N

Powell 2013

N

N

Y

P

N

N

Y

MgSO4 and SABA versus SABA

Abreu‐Gonzalez 2002

Y

Y

N

N

N

N

N

Aggarwal 2006

N

Y

N

Y

N

Y

Y

Ahmed 2013

N

P

N

N

N

N

N

Alansari 2015

N

N

Y

P

N

N

Y

Badawy 2014

Y

Y

N

N

N

Y

N

Bessmertny 2002

P

N

N

N

N

N

Y

Dadhich 2005

P

P

N

N

N

N

Y

Hughes 2003

Y

N

N

Y

N

N

Y

Khashabi 2008

N

N

N

N

N

N

N

Kokturk 2005

N

Y

P

Y

N

N

Y

Mahajan 2004

Y

N

N

Y

N

N

Y

Mohammedzadeh 2014

N

Y

Y

N

N

N

N

Nannini 2000

N

Y

N

Y

N

N

Y

Sarhan 2016

N

Y

Y

N

N

Y

N

Turker 2017

N

N

Y

Y

N

N

Y

MgSO₄ versus SABA

Dadhich 2005

P

P

N

N

N

N

Y

Mangat 1998

N

Y

N

Y

N

N

Y

Meral 1996

N

Y

N

N

N

N

Y

Neki 2006

N

Y

N

N

N

Y

N

Sarhan 2016

N

Y

Y

N

N

Y

N

N ‒ the study did not report the outcome but it is not clear whether the outcome was measured or not

Y ‒ full reporting

P ‒ partial reporting

Figuras y tablas -
Table 4. Outcomes
Comparison 1. MgSO4 + SABA + ipratropium versus SABA + ipratropium

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pulmonary function (% FEV1) Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

3.28 [1.06, 5.49]

1.1 90 minutes

1

60

Mean Difference (IV, Fixed, 95% CI)

8.57 [1.99, 15.15]

1.2 120 minutes

1

60

Mean Difference (IV, Fixed, 95% CI)

2.60 [0.25, 4.95]

2 Pulmonary function % predicted PEF Show forest plot

2

636

Mean Difference (IV, Fixed, 95% CI)

0.05 [‐2.33, 2.42]

3 Clinical severity scores (closest to 60 mins) Show forest plot

2

1130

Std. Mean Difference (IV, Fixed, 95% CI)

0.01 [‐0.11, 0.12]

3.1 Yung ASS at 60 minutes

1

472

Std. Mean Difference (IV, Fixed, 95% CI)

‐0.17 [‐0.35, 0.02]

3.2 Change in dyspnoea VAS at 60 minutes

1

658

Std. Mean Difference (IV, Fixed, 95% CI)

0.13 [‐0.02, 0.28]

4 Admission at first presentation Show forest plot

4

1308

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.91, 1.00]

4.1 Adults

3

800

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.87, 1.03]

4.2 Children

1

508

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.92, 1.01]

5 HDU/ITU admission Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.1 Admission to HDU (adults)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Admission to ICU (adults)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Admission to PICU/HDU or intubation (children)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Readmission Show forest plot

2

750

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [0.84, 3.87]

7 Respiratory rate at 60 mins Show forest plot

2

723

Mean Difference (IV, Fixed, 95% CI)

0.70 [‐0.14, 1.53]

8 Heart rate at 60 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

9 Systolic blood pressure at 60 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

10 Diastolic blood pressure at 60 mins Show forest plot

1

674

Mean Difference (IV, Fixed, 95% CI)

2.40 [0.29, 4.51]

11 Serious adverse events (during admission) Show forest plot

2

557

Risk Difference (M‐H, Fixed, 95% CI)

‐0.03 [‐0.06, ‐0.00]

11.1 Adults

1

50

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.07, 0.07]

11.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.04 [‐0.06, ‐0.01]

12 Any adverse event (during admission) Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.01 [‐0.03, 0.05]

12.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.02 [‐0.02, 0.07]

12.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.02 [‐0.09, 0.05]

13 Serious adverse events (within 30 days) Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

14 Any adverse event (within 30 days) Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

15 Adverse event: hypotension Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.02 [‐0.01, 0.04]

15.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.03 [‐0.01, 0.07]

15.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.00 [‐0.02, 0.01]

16 Adverse event: flushing Show forest plot

2

1197

Risk Difference (M‐H, Fixed, 95% CI)

0.00 [‐0.01, 0.01]

16.1 Adults

1

690

Risk Difference (M‐H, Fixed, 95% CI)

0.00 [‐0.01, 0.02]

16.2 Children

1

507

Risk Difference (M‐H, Fixed, 95% CI)

‐0.00 [‐0.02, 0.01]

Figuras y tablas -
Comparison 1. MgSO4 + SABA + ipratropium versus SABA + ipratropium
Comparison 2. MgSO4 + SABA versus SABA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pulmonary function % predicted FEV1 Show forest plot

4

208

Mean Difference (IV, Fixed, 95% CI)

3.34 [‐1.58, 8.26]

1.1 Adults

3

146

Mean Difference (IV, Fixed, 95% CI)

2.18 [‐3.30, 7.67]

1.2 Children

1

62

Mean Difference (IV, Fixed, 95% CI)

8.10 [‐3.03, 19.23]

2 % predicted FEV1: subgroup: severity Show forest plot

3

188

Mean Difference (IV, Fixed, 95% CI)

4.12 [‐1.81, 10.06]

2.1 Severe (FEV1 <50% predicted)

1

52

Mean Difference (IV, Fixed, 95% CI)

9.90 [0.05, 19.75]

2.2 Moderate

2

136

Mean Difference (IV, Fixed, 95% CI)

0.84 [‐6.59, 8.27]

3 Pulmonary function PEF L/min Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

3.1 Adults

3

155

Mean Difference (IV, Fixed, 95% CI)

11.91 [‐4.12, 27.95]

3.2 Children

1

80

Mean Difference (IV, Fixed, 95% CI)

11.90 [‐6.86, 30.66]

4 Admission to hospital Show forest plot

6

375

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.52, 1.15]

4.1 Adults

4

213

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.45, 1.07]

4.2 Children

2

162

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.44, 2.98]

5 Heart rate at 120 mins Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

6 Respiratory rate at 120 mins Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

7 Diastolic blood pressure at 120 mins Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

0.72 [‐1.35, 2.80]

8 Systolic blood pressure at 120 mins Show forest plot

2

120

Mean Difference (IV, Fixed, 95% CI)

0.89 [‐2.69, 4.48]

9 Serious adverse events Show forest plot

5

243

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.04, 0.04]

9.1 Adults

4

181

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.04, 0.04]

9.2 Children

1

62

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.06, 0.06]

10 Any adverse events Show forest plot

5

694

Risk Difference (M‐H, Fixed, 95% CI)

‐0.01 [‐0.05, 0.03]

10.1 Adults

4

329

Risk Difference (M‐H, Fixed, 95% CI)

‐0.02 [‐0.10, 0.06]

10.2 Children

1

365

Risk Difference (M‐H, Fixed, 95% CI)

‐0.01 [‐0.03, 0.01]

Figuras y tablas -
Comparison 2. MgSO4 + SABA versus SABA
Comparison 3. MgSO4 versus SABA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical severity score Show forest plot

3

93

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.62, 0.36]

1.1 Fischl index final score (120 mins)

1

33

Mean Difference (IV, Fixed, 95% CI)

‐0.33 [‐1.07, 0.41]

1.2 Fischl index score (time point unclear)

1

40

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐1.11, 0.71]

1.3 Change in Fischl index at 120 mins

1

20

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.67, 1.27]

2 Admission to hospital Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3 Heart rate (120 mins) Show forest plot

1

20

Mean Difference (IV, Fixed, 95% CI)

21.20 [0.17, 42.23]

4 Respiratory rate Show forest plot

2

60

Mean Difference (IV, Fixed, 95% CI)

‐2.40 [‐3.91, ‐0.89]

5 Systolic pressure (120 mins) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

6 Diastolic pressure (120 mins) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

7 Serious adverse events Show forest plot

2

53

Risk Difference (M‐H, Fixed, 95% CI)

0.0 [‐0.10, 0.10]

8 Mild‐Moderate Side Effects Show forest plot

1

Risk Difference (M‐H, Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 3. MgSO4 versus SABA