Scolaris Content Display Scolaris Content Display

Methylphenidat für Kinder und Jugendliche mit Aufmerksamkeitsdefizit‐Hyperaktivitäts‐Syndrom (ADHS)

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Abikoff 2009 {published data only}

Concerta for organizational deficits in ADHD. The Brown University Child and Adolescent Behavior Letter2009; Vol. 25, issue 3:2. CENTRAL
Abikoff H, Nissley‐Tsiopinis J, Gallagher R, Zambenedetti M, Seyffert M, Boorady R, et al. Effects of MPH‐OROS on the organizational, time management, and planning behaviors of children with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2009;48(2):166‐75. CENTRAL

Ahmann 1993 {published data only}

Ahmann PA, Waltonen SJ, Olson KA, Theye FW, Van Erem AJ, LaPlant RJ. Placebo‐controlled evaluation of Ritalin side effects. Pediatrics 1993;91(6):1101‐6. CENTRAL

Armstrong 2012 {published data only}

Significant effect of OROS MPH ER on attention. Brown University Child & Adolescent Psychopharmacology Update2010; Vol. 12, issue 1:7‐8. CENTRAL
Armstrong RB, Damaraju CV, Ascher S, Schwarzman L, O'Neill J, Starr HL. Time course of treatment effect of OROS® methylphenidate in children with ADHD. Journal of Attention Disorders 2012;16(8):697‐705. CENTRAL
Wigal SB, Wigal T, Schuck S, Brams M, Williamson D, Armstrong RB, et al. Academic, behavioral, and cognitive effects of OROS® methylphenidate on older children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2011;21(2):121‐31. CENTRAL
Williamson D, Murray DW, Damaraju CV, Ascher S, Starr HL. Methylphenidate in children with ADHD with or without learning disability. Journal of Attention Disorders 2014;18(2):95‐104. [DOI: 10.1177/1087054712443411]CENTRAL

Arnold 2004 {published data only}

Arnold LE, Lindsay RL, Conners CK, Wigal SB, Levine AJ, Johnson DE, et al. A double‐blind, placebo‐controlled withdrawal trial of dexmethylphenidate hydrochloride in children with attention deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology2004; Vol. 14, issue 4:542‐54. CENTRAL

Ashare 2010 {published data only}

Ashare RL, Hawk LW, Shiels K, Rhodes JD, Pelham WE, Waxmonsky JG. Methylphenidate enhances prepulse inhibition during processing of task‐relevant stimuli in attention‐deficit/hyperactivity disorder. Psychophysiology 2010;47(5):838‐45. CENTRAL

Barkley 1989 {published data only}

Barkley RA, McMurray MB, Edelbrock CS, Robbins K. Side effects of methylphenidate in children with attention deficit hyperactivity disorder: a systemic, placebo‐controlled evaluation. Pediatrics 1990;86(2):184‐92. CENTRAL
Barkley RA, McMurray MB, Edelbrock CS, Robbins K. The response of aggressive and nonaggressive ADHD children to two doses of methylphenidate. Journal of the American Academy of Child and Adolescent Psychiatry 1989;28(6):873‐81. CENTRAL

Barkley 1991 {published data only}

Barkley RA, DuPaul GJ, McMurray MB. Attention deficit disorder with and without hyperactivity: clinical response to three dose levels of methylphenidate. Pediatrics 1991;87(4):519‐31. CENTRAL
DuPaul GJ, Barkley RA, McMurray MB. Response of children with ADHD to methylphenidate: interaction with internalizing symptoms. Journal of the American Academy of Child and Adolescent Psychiatry 1994;33(6):894‐903. CENTRAL

Barkley 2000 {published data only}

Barkley RA, Connor DF, Kwasnik D. Challenges to determining adolescent medication response in an outpatient clinical setting: comparing Adderall and methylphenidate for ADHD. Journal of Attention Disorders 2000;4(2):102‐13. CENTRAL

Bedard 2008 {published data only}

Bedard A‐C, Tannock R. Anxiety, methylphenidate response, and working memory in children with ADHD. Journal of Attention Disorders 2008;11(5):546‐57. CENTRAL

Ben 2002 {published data only}

Ben AL, Grizenko N, Mbekou V, Lageix P, Baron C, Schwartz G, et al. Dopamine transporter gene and behavioral response to methylphenidate (MPH) in children with attention deficit hyperactivity disorder (ADHD). American Journal of Medical Genetics. Abstracts of the Xth World Congress of Psychiatric Genetics; 2002 October 9‐13; Brussels, Belgium. 2002; Vol. 114 (7):724‐5. CENTRAL

Biederman 2003 {published data only}

Biederman J, Quinn D, Weiss M, Markabi S, Weidenman M, Edson K, et al. Efficacy and safety of Ritalin LA, a new, once daily, extended‐release dosage form of methylphenidate, in children with attention deficit hyperactivity disorder. Paediatric Drugs 2003;5(12):833‐41. CENTRAL

Bliznakova 2007 {published data only}

Bliznakova L, Gerstner S, Schmidt MH, Becker K. Methylphenidate double‐blind trial: indication and performing [Der methylphenidat‐doppelblindversuch ‐ indikation und durchfuhrung]. Klinische Pädiatrie 2007;219(1):9‐16. CENTRAL

Blum 2011 {published data only}

Blum NJ, Jawad AF, Clarke AT, Power TJ. Effect of osmotic‐release oral system methylphenidate on different domains of attention and executive functioning in children with attention‐deficit‐hyperactivity disorder. Developmental Medicine & Child Neurology 2011;53(9):843‐9. CENTRAL

Borcherding 1990 {published data only}

Borcherding BG, Keysor CS, Rapoport JL, Elia J, Amass J. Motor/vocal tics and compulsive behaviors on stimulant drugs: is there a common vulnerability?. Psychiatry Research 1990;33(1):83‐94. CENTRAL

Brams 2008 {published data only}

Brams M, Muniz R, Childress A, Giblin J, Mao A, Turnbow J, et al. A randomized, double‐blind, crossover study of once‐daily dexmethylphenidate in children with attention‐deficit hyperactivity disorder: rapid onset of effect. CNS Drugs 2008;22(8):693‐704. CENTRAL

Brams 2012 {published data only}

Dexmethylphenidate may be effective later in the day. The Brown University Child & Adolescent Psychopharmacology Update2012; Vol. 14, issue 11:8. CENTRAL
Brams M, Turnbow J, Pestreich L. Erratum: a randomized, double‐blind study of 30 versus 20 mg dexmethylphenidate extended‐release in children with attention‐deficit/ hyperactivity disorder: late‐day symptom control. Journal of Clinical Psychopharmacology 2012;32(6):766. Erratum for: Journal of Clinical Psychopharmacology 2012;32(5):637‐44. CENTRAL
Brams M, Turnbow J, Pestreich L, Giblin J, Childress A, McCague K, et al. A randomized, double‐blind study of 30 versus 20 mg dexmethylphenidate extended‐release in children with attention‐deficit/hyperactivity disorder: late‐day symptom control. Journal of Clinical Psychopharmacology. M. Brams, Menninger Department of Psychiatry, Baylor College of Medicine, 500 Westcott St, Houston, TX 77007, United States. E‐mail: [email protected]: Lippincott Williams and Wilkins (530 Walnut Street,P O Box 327, Philadelphia PA 19106‐3621, United States), 2012; Vol. 32, issue 5:637‐44. CENTRAL
Muniz R, Pestreich L, McCague K, Padilla A, Brams M, Childress A. Extended‐release dexmethylphenidate 30 mg improves late‐day attention deficit hyperactivity disorder (ADHD) symptom control in children with ADHD: a randomized, double‐blind crossover study. Journal of Child and Adolescent Psychopharmacology. Abstracts of the 50th Annual National Institute of Mental Health (NIMH) New Clinical Drug Evaluation Unit (NCDEU) Meeting; 2010 June 14–17; Boca Raton, Florida 2010;20(6):534‐5. CENTRAL
Padilla A, Pestreich L, McCague K, Muniz R. Late‐day attention deficit hyperactivity disorder (ADHD) symptom control improvement with extended‐release dexmethylphenidate in children with ADHD of all ethnicities: a sub‐analysis. Journal of Child and Adolescent Psychopharmacology. Abstracts of the 50th Annual National Institute of Mental Health (NIMH) New Clinical Drug Evaluation Unit (NCDEU) Meeting; 2010 June 14–17; Boca Raton, Florida 2010;20(6):534. CENTRAL
Silva RR, Brams M, McCague K, Pestreich L, Muniz R. Extended‐release dexmethylphenidate 30 mg/d versus 20 mg/d: duration of attention, behavior, and performance benefits in children with attention‐deficit/hyperactivity disorder. Clinical Neuropharmacology 2013;36(4):117‐21. CENTRAL

Brown 1984a {published data only}

Brown RT, Wynne ME, Slimmer LW. Attention deficit disorder and the effect of methylphenidate on attention, behavioral, and cardiovascular functioning. Journal of Clinical Psychiatry 1984;45(11):473‐6. CENTRAL

Brown 1985 {published data only}

Brown RT, Wynne ME, Medenis R. Methylphenidate and cognitive therapy: a comparison of treatment approaches with hyperactive boys. Journal of Abnormal Child Psychology 1985;13(1):69‐87. CENTRAL

Brown 1988 {published data only}

Brown RT, Sexson SB. A controlled trial of methylphenidate in black adolescents. Attentional, behavioral, and physiological effects. Clinical Pediatrics 1988;27(2):74‐81. CENTRAL
Brown RT, Sexson SB. Effects of methylphenidate on cardiovascular responses in attention deficit hyperactivity disordered adolescents. Journal of Adolescent Health Care 1989;10(3):179‐83. CENTRAL

Brown 1991 {published data only}

Brown RT, Jaffe SL, Silverstein J, Magee H. Methylphenidate and hospitalized adolescents with conduct disorder: dose effects on classroom behavior, academic performance, and impulsivity. Journal of Youth and Adolescence 1991;20(5):501‐18. CENTRAL

Buitelaar 1995 {published data only}

Buitelaar JK, Swaab‐Barneveld H, Van der Gaag RJ. Prediction of clinical response to methylphenidate in children with ADHD. X World Congress of Psychiatry; 1996 August 23‐26; Madrid, Spain. Madrid: World Psychiatric Association, 1996. CENTRAL
Buitelaar JK, Van der Gaag RJ, Swaab‐Barneveld H, Kuiper M. Pindolol and methylphenidate in children with attention‐deficit hyperactivity disorder. Clinical efficacy and side‐effects. Journal of Child Psychology and Psychiatry 1996;37(5):587‐95. CENTRAL
Buitelaar JK, Van der Gaag RJ, Swaab‐Barneveld H, Kuiper M. Prediction of clinical response to methylphenidate in children with attention‐deficit hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1995;34(8):1025‐32. CENTRAL

Bukstein 1998 {published data only}

Bukstein OG, Kolko DJ. Effects of methylphenidate on aggressive urban children with attention deficit hyperactivity disorder. Journal of Clinical Child Psychology 1998;27(3):340‐51. CENTRAL

Butter 1983 {published data only}

Butter HJ, Lapierre Y, Firestone P, Blank A. A comparative study of the efficacy of ACTH4‐9 analog, methylphenidate, and placebo on attention deficit disorder with hyperkinesis. Journal of Clinical Psychopharmacology 1983;3(4):226‐30. CENTRAL
Butter HJ, Lapierre Y, Firestone P, Blank A. Efficacy of ACTH 4‐9 analog, methylphenidate, and placebo on attention deficit disorder with hyperkinesis. Progress in Neuro‐Psychopharmacology & Biological Psychiatry 1984;8(4‐6):661‐4. CENTRAL

Carlson 1995 {published data only}

Carlson GA, Rapport MD, Kelly KL, Pataki CS. Methylphenidate and desipramine in hospitalized children with comorbid behavior and mood disorders: separate and combined effects on behavior and mood. Journal of Child and Adolescent Psychopharmacology 1995;5(3):191‐204. CENTRAL
Pataki CS, Carlson GA, Kelly KL, Rapport MD, Biancaniello TM. Side effects of methylphenidate and desipramine alone and in combination in children. Journal of the American Academy of Child and Adolescent Psychiatry 1993;32(5):1065‐72. CENTRAL

Carlson 2007 {published data only}

Carlson GA, Dunn D, Kelsey D, Ruff D, Ball S, Ahrbecker L, et al. A pilot study for augmenting atomoxetine with methylphenidate: safety of concomitant therapy in children with attention‐deficit/hyperactivity disorder. Child and Adolescent Psychiatry and Mental Health 2007;1(1):10. CENTRAL

Castellanos 1997 {published data only}

Castellanos FX. Stimulants and tic disorders: from dogma to data. Archives of General Psychiatry1999; Vol. 56, issue 4:337‐8. CENTRAL
Castellanos FX, Giedd JN, Elia J, Marsh WL, Ritchie GF, Hamburger SD, et al. Controlled stimulant treatment of ADHD and comorbid Tourette's syndrome: effects of stimulant and dose. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(5):589‐96. CENTRAL

Chacko 2005 {published data only}

Chacko A, Pelham WE, Gnagy EM, Greiner A, Vallano G, Bukstein O, et al. Stimulant medication effects in a summer treatment program among young children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2005;44(3):249‐57. CENTRAL

Childress 2009 {published data only}

Dose related safety and efficacy of dexmethylphenidate ER. The Brown University Child & Adolescent Psychopharmacology Update2009; Vol. 11, issue 11:7‐8. CENTRAL
Childress A, Muniz R, Miller J, Arnold V, Harper L, Gerstner O, et al. Fixed‐dose titration study of dexmethylphenidate extended release in children with ADHD: effects on teacher‐rated scales. Annals of Neurology 2007;62(Suppl 11):S125. CENTRAL
Childress AC, Spencer T, Lopez F, Gerstner O, Thulasiraman A, Muniz R, et al. Efficacy and safety of dexmethylphenidate extended‐release capsules administered once daily to children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2009;19(4):351‐61. CENTRAL
Greenbaum M, Muniz R, Brams M, Boellner S, Gerstner O, Borello M, et al. Cardiovascular safety of dexmethylphenidate extended release in children with ADHD. Annals of Neurology 2007;62(Suppl 11):S146. CENTRAL
Lopez F, Muniz R, Joyce JM, Franklin ER, Gerstner O, Borello M, et al. Fixed‐dose titration study of dexmethylphenidate extended release in children with ADHD: effects on parent‐rated scales. Annals of Neurology 2007;62(Suppl 11):S122. CENTRAL

Chronis 2003 {published data only}

Chronis A, Pelham WE, Gnagy E, Roberts J, Aronoff H. The impact of late‐afternoon stimulant dosing for children with ADHD on parent and parent‐child domains. Journal of Clinical Child and Adolescent Psychology 2003;32(1):118‐26. CENTRAL
Pelham WE, Gnagy EM, Chronis AM, Burrows‐MacLean L, Fabiano GA, Onyango AN, et al. A comparison of morning‐only and morning/late afternoon Adderall to morning‐only, twice‐daily, and three times‐daily methylphenidate in children with attention‐deficit/hyperactivity disorder. Pediatrics 1999;104(6):1300‐11. CENTRAL

Coghill 2007 {published data only}

Coghill DR, Rhodes SM, Matthews K. Chronic effects of the psychostimulant drug methylphenidate on neuropsychological functioning in drug‐naive boys with hyperkinetic disorder (ADHD). Journal of Psychopharmacology. Summer Meeting of the British Association for Psychopharmacology; 2003 July 20‐23; Cambridge, England. London: Sage Publications Ltd, 2003; Vol. 17 (3):A74. [WOS: 000185623500293]CENTRAL
Coghill DR, Rhodes SM, Matthews K. The neuropsychological effects of chronic methylphenidate on drug‐naive boys with attention‐deficit/hyperactivity disorder. Biological Psychiatry 2007;62(9):954‐62. CENTRAL

Coghill 2013 {published data only}

Banaschewski T, Soutullo C, Lecendreux M, Johnson M, Zuddas A, Hodgkins P, et al. Health‐related quality of life and functional outcomes from a randomized, controlled study of lisdexamfetamine dimesylate in children and adolescents with attention deficit hyperactivity disorder. CNS Drugs 2013;27(10):829‐40. CENTRAL
Banaschewski T, Soutullo C, Lecendreux M, Johnson M, Zuddas A, Hodgkins P, et al. The child health and illness profile as a measure of health‐related quality of life in stimulant‐treated children and adolescents with ADHD. European Child & Adolescent Psychiatry. Abstracts of the 15th International Congress of ESCAP ‐ European Society for Child and Adolescent Psychiatry; 2013 July 6‐10; Dublin, Ireland 2013;22(2 Suppl):S125‐6. CENTRAL
Coghill D, Banaschewski T, Lecendreux M, Soutollu C, Johnson M, Zuddas A, et al. The first European studies of lisdexamfetamine dimesylate in children and adolescents with attention‐deficit/hyperactivity disorder. European Child & Adolescent Psychiatry. Abstracts of the 15th International Congress of ESCAP ‐ European Society for Child and Adolescent Psychiatry; 2013 July 6‐10, Dublin, Ireland 2013;22(2 Suppl):S125. CENTRAL
Coghill D, Banaschewski T, Lecendreux M, Soutullo C, Johnson M, Zuddas A, et al. European, randomized, phase 3 study of lisdexamfetamine dimesylate in children and adolescents with attention‐deficit/hyperactivity disorder. European Neuropsychopharmacology 2013;23(10):1208‐18. CENTRAL
Coghill D, Banaschewski T, Lecendreux M, Soutullo C, Johnson M, Zuddas A, et al. Post hoc comparison of the efficacy of lisdexamfetamine dimesylate and osmotic‐release oral system methylphenidate in children and adolescents with ADHD. European Psychiatry. Abstracts of the 21th European Congress of Psychiatry 2013;28(Suppl 1):Article 957. CENTRAL
Coghill DR, Banaschewski T, Lecendreux M, Soutullo C, Zuddas A, Adeyi B, et al. Post hoc analyses of the impact of previous medication on the efficacy of lisdexamfetamine dimesylate in the treatment of attention‐deficit/hyperactivity disorder in a randomized, controlled trial. Neuropsychiatric Disease and Treatment 2014;10:2039‐47. CENTRAL
Coghill DR, Banaschewski T, Lecendreux M, Zuddas A, Dittmann RW, Otero IH, et al. Efficacy of lisdexamfetamine dimesylate throughout the day in children and adolescents with attention‐deficit/hyperactivity disorder: results from a randomized, controlled trial. European Child & Adolescent Psychiatry 2014;23(2):61‐8. CENTRAL
Doddamani L, Hodgkins P, Adeyi B, Squires LA, Civil R, Coghill DR. Functional impairment in children and adolescents with attention deficit hyperactivity disorder: results from short‐ and long‐term studies of lisdexamfetamine dimesylate. Australian and New Zealand Journal of Psychiatry. Proceedings of the Royal Australian and New Zealand College of Psychiatrists, RANZCP Annual Congress; 2014 May 11‐15; Perth, Australia. Informa Healthcare, 2014; Vol. 48:115. [EMBASE: 71565453]CENTRAL
Gasior M, Coghill D, Soutullo C, Lyne A, Johnson M. Poster 7: efficacy and safety of lisdexamfetamine dimesylate in children and adolescents with ADHD: a phase 3, randomized, double‐blind, multicenter, parallel‐group, placebo‐ and active‐controlled, dose‐optimized study in Europe. Acta Neuropsychiatrica. Abstracts of the Scandinavian College of Neuropsychopharmacology 53rd Annual Meeting; 2012 April 25‐27; Copenhagen, Denmark 2012;24(Suppl s1):24. [DOI: 10.1111/j.1601‐5215.2012.00665.x]CENTRAL
Hodgkins P, Coghill DR, Soutullo CA, Bloomfield R, Gasior M, Johnson M. Poster 14: effect of lisdexamfetamine dimesylate on functional impairment in children and adolescents with attention‐deficit/hyperactivity disorder. Acta Neuropsychiatrica. Abstracts of the Scandinavian College of Neuropsychopharmacology (SCNP) 53rd Annual Meeting; 2012 April 25‐27; Copenhagen, Denmark 2012;24(Suppl s1):27‐8. [DOI: 10.1111/j.1601‐5215.2012.00665.x]CENTRAL
Hodgkins P, Setyawan J, Banaschewski T, Soutullo C, Lecendreux M, Johnson M, et al. Health utility scores in children and adolescents with attention‐deficit/hyperactivity disorder: response to stimulant treatment. European Child & Adolescent Psychiatry. Abstracts of the 15th International Congress of ESCAP ‐ European Society for Child and Adolescent Psychiatry; 2013 July 6‐10; Dublin, Ireland 2013;22(Suppl 2):S127. CENTRAL
Zuddas A, Banaschewski T, Lecendreux M, Soutullo C, Johnson M, Anderson C, et al. Clinical efficacy of lisdexamfetamine dimesylate in children and adolescents with ADHD: a post‐hoc analysis. European Neuropsychopharmacology. Papers of the 25th European College of Neuropsychopharmacology Congress; 2012 October 13‐17; Vienna, Austria 2012;22(Suppl 2):S431. CENTRAL

Connor 2000 {published data only}

Connor DF, Barkley RA, Davis HT. A pilot study of methylphenidate, clonidine, or the combination in ADHD comorbid with aggressive oppositional defiant or conduct disorder. Clinical Pediatrics 2000;39(1):15‐25. CENTRAL

Cook 1993 {published data only}

Cook JR. The Effects of Methylphenidate on Resource Allocation in the Mental Processing of ADD Children [PhD thesis]. University of North Dakota, 1989. CENTRAL
Cook JR, Mausbach T, Burd L, Gascon GG, Slotnick HB, Patterson B, et al. A preliminary study of the relationship between central auditory processing disorder and attention deficit disorder. Journal of Psychiatry & Neuroscience 1993;18(3):130‐7. CENTRAL

Corkum 2008 {published data only}

Corkum P, Panton R, Ironside S, Macpherson M, Williams T. Acute impact of immediate release methylphenidate administered three times a day on sleep in children with attention‐deficit/hyperactivity disorder. Journal of Pediatric Psychology 2008;33(4):368‐79. CENTRAL
Ironside S, Davidson F, Corkum P. Circadian motor activity affected by stimulant medication in children with attention‐deficit/hyperactivity disorder. Journal of Sleep Research 2010;19(4):546‐51. CENTRAL

Cox 2006 {published data only}

Cox DJ, Merkel RL, Moore M, Thorndike F, Muller C, Kovatchev B. Relative benefits of stimulant therapy with OROS methylphenidate versus mixed amphetamine salts extended release in improving the driving performance of adolescent drivers with attention‐deficit/hyperactivity disorder. Pediatrics 2006;118(3):e704‐10. CENTRAL
Cox DJ, Moore M, Burket R, Merkel RL, Mikami AY, Kovatchev B. Rebound effects with long‐acting amphetamine or methylphenidate stimulant medication preparations among adolescent male drivers with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2008;18(1):1‐10. CENTRAL
Mikami AY, Cox DJ, Davis MT, Wilson HK, Merkel RL, Burket R. Sex differences in effectiveness of extended‐release stimulant medication among adolescents with attention‐deficit/hyperactivity disorder. Journal of Clinical Psychology in Medical Settings 2009;16(3):233‐42. CENTRAL
Wilson HK, Cox DJ, Merkel RL, Moore M, Coghill D. Effect of extended release stimulant‐based medications on neuropsychological functioning among adolescents with attention‐deficit/hyperactivity disorder. Archives of Clinical Neuropsychology 2006;21(8):797‐807. CENTRAL

Döpfner 2004 {published data only}

Döpfner M, Gerber WD, Banaschewski T, Breuer D, Freisleder FJ, Gerber‐von Müller G, et al. Comparative efficacy of once‐a‐day extended‐release methylphenidate, two‐times‐daily immediate‐release methylphenidate, and placebo in a laboratory school setting. European Child & Adolescent Psychiatry 2004;13(1 Suppl):i93‐101. CENTRAL
Döpfner M, Schröder S, Schmidt J, Lehmkuhl G. Duration of action of a single dose of methylphenidate Retard compared to twice immediate‐release methylphenidate in children and adolescents with ADHD [Wirkdauer einer einmaligen Gabe von Methylphenidat‐Retard im Vergleich zu zweimaliger Gabe von schnell freisetzendem Methylphenidat bei Kindern und Jugendlichen mit ADHS]. Klinikum der Universität zu Köln, Klinik und Poliklinik für Psychiatrie und Psychotherapie des Kindes und Jugendalters2003. CENTRAL
Gerber WD, Gerber‐von Müller G, Andrasik F, Niederberger U, Siniatchkin M, Kowalski JT, et al. The impact of a multimodal summer camp training on neuropsychological functioning in children and adolescents with ADHD: an exploratory study. Child Neuropsychology 2012;18(3):242‐55. CENTRAL
Gerber‐von Müller G, Petermann U, Petermann F, Niederberger U, Stephani U, Siniatchkin M, et al. ADHD summer camp: development and evaluation of a multimodal intervention program [Das ADHD‐summer‐camp‐entwicklung und evaluation eines multimodalen programms]. Kindheit und Entwicklung 2009;18(3):162‐72. CENTRAL
Lehmkuhl G. Double‐blind, non‐inferiority trial investigating the duration of action of Medikinet‐retard vs. immediate‐release methylphenidate vs. placebo across the day in children with attention deficit hyperactivity disorder (ADHD). Integrated final report. Phase III. Universitätsklinikum Essen. Project number Medikinet‐retard (R) Trial 6520‐0073‐012005. CENTRAL
Uebel H, Albrecht B, Kirov R, Heise A, Döpfner M, Freisleder FJ, et al. What can actigraphy add to the concept of labschool design in clinical trials?. Current Pharmaceutical Design 2010;16(22):2434‐42. CENTRAL

Douglas 1986 {published data only}

Douglas VI, Barr RG, O'Neill ME, Britton BG. Short term effects of methylphenidate on the cognitive, learning and academic performance of children with attention deficit disorder in the laboratory and the classroom. Journal of Child Psychology and Psychiatry, and Allied Disciplines 1986;27(2):191‐211. CENTRAL

Douglas 1995 {published data only}

Douglas VI, Barr RG, Desilets J, Sherman E. Do high doses of stimulants impair flexible thinking in attention‐deficit hyperactivity disorder?. Journal of the American Academy of Child and Adolescent Psychiatry 1995;34(7):877‐85. CENTRAL

DuPaul 1996 {published data only}

DuPaul GJ, Anastopoulos AD, Kwasnik D, Barkley RA, McMurray MB, DuPaul GJ. Methylphenidate effects on children with attention deficit hyperactivity disorder: self‐report of symptoms, side‐effects, and self‐esteem. Journal of Attention Disorders 1996;1(1):3‐15. CENTRAL

Duric 2012 {published data only}

Duric NS, Assmus J, Gundersen D, Elgen IB. Neurofeedback for the treatment of children and adolescents with ADHD: a randomized and controlled clinical trial using parental reports. BMC Psychiatry2012; Vol. 12:107. CENTRAL

Epstein 2011 {published data only}

Epstein JN, Brinkman WB, Froehlich T, Langberg JM, Narad ME, Antonini TN, et al. Effects of stimulant medication, incentives, and event rate on reaction time variability in children with ADHD. Neuropsychopharmacology 2011;36(5):1060‐72. CENTRAL

Fabiano 2007 {published data only}

Fabiano GA, Pelham WE, Gnagy EM, Burrows‐MacLean L, Coles EK, Chacko A, et al. The single and combined effects of multiple intensities of behavior modification and methylphenidate for children with attention deficit hyperactivity disorder in a classroom setting. School Psychology Review2007; Vol. 36, issue 2:195‐216. CENTRAL
Pelham WE, Burrows‐Maclean L, Gnagy EM, Fabiano GA, Coles EK, Wymbs BT, et al. A dose‐ranging study of behavioral and pharmacological treatment in social settings for children with ADHD. Journal of Abnormal Child Psychology 2014;42(6):1019‐31. CENTRAL

Findling 2006 {published data only}

Findling RL, Quinn D, Hatch SJ, Cameron SJ, DeCory HH, McDowell M. Comparison of the clinical efficacy of twice‐daily Ritalin and once‐daily Equasym XL with placebo in children with attention deficit/hyperactivity disorder. European Child & Adolescent Psychiatry 2006;15(8):450‐9. CENTRAL

Findling 2007 {published data only}

Findling RL, Short EJ, McNamara NK, Demeter CA, Stansbrey RJ, Gracious BL, et al. Methylphenidate in the treatment of children and adolescents with bipolar disorder and attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(11):1445‐53. CENTRAL

Findling 2008 {published data only}

Faraone SV, Glatt SJ, Bukstein OG, Lopez FA, Arnold LE, Findling RL. Effects of once‐daily oral and transdermal methylphenidate on sleep behavior of children with ADHD. Journal of Attention Disorders 2009;12(4):308‐15. CENTRAL
Findling RL, Bukstein OG, Melmed RD, Lopez FA, Sallee FR, Arnold LE, et al. "A randomized, double‐blind, placebo‐controlled, parallel‐group study of methylphenidate transdermal system in pediatric patients with attention‐deficit/hyperactivity disorder": correction. Journal of Clinical Psychiatry 2008;69(2):329. CENTRAL
Findling RL, Bukstein OG, Melmed RD, López FA, Sallee FR, Arnold LE, et al. A randomized, double‐blind, placebo‐controlled, parallel‐group study of methylphenidate transdermal system in pediatric patients with attention‐deficit/hyperactivity disorder. Journal of Clinical Psychiatry 2008;69(1):149‐59. Erratum in: Journal of Clinical Psychiatry. 2008;69(2):329. CENTRAL
Swanson JM. Transdermal methylphenidate more effective than placebo for treating ADHD. Evidence‐Based Mental Health 2008;11(4):118. CENTRAL

Findling 2010 {published data only}

Findling RL, Katic A, Rubin R, Moon E, Civil R, Li Y. A 6‐month, open‐label, extension study of the tolerability and effectiveness of the methylphenidate transdermal system in adolescents diagnosed with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2010;20(5):365‐75. CENTRAL
Findling RL, Turnbow J, Burnside J, Melmed R, Civil R, Li Y. A randomized, double‐blind, multicenter, parallel‐group, placebo‐controlled, dose‐optimization study of the methylphenidate transdermal system for the treatment of ADHD in adolescents. CNS Spectrums 2010;15(7):419‐30. CENTRAL
Keating GM. Methylphenidate transdermal system in attention‐deficit hyperactivity disorder in adolescents: profile report. Drugs in R&D 2012;12(3):171‐3. CENTRAL

Fine 1993 {published data only}

Fine S, Johnston C. Drug and placebo side effects in methylphenidate‐placebo trial for attention deficit hyperactivity disorder. Child Psychiatry and Human Development 1993;24(1):25‐30. CENTRAL
Johnston C, Fine S. Methods of evaluating methylphenidate in children with attention deficit hyperactivity disorder: acceptability, satisfaction and compliance. Journal of Pediatric Psychology 1993;18(6):717‐30. CENTRAL

Firestone 1981 {published data only}

Firestone P, Kelly MJ, Goodman JT, Davey J. Differential effects of parent training and stimulant medication with hyperactives: a progress report. Journal of the American Academy of Child and Adolescent Psychiatry 1981;20(1):135‐47. CENTRAL

Fitzpatrick 1992a {published data only}

Fitzpatrick PA, Klorman R, Brumaghim JT, Borgstedt AD. Effects of sustained‐release and standard preparations of methylphenidate on attention deficit disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1992;31(2):226‐34. CENTRAL
Fitzpatrick PS. Effects of Sustained‐Release and Standard Preparations of Methylphenidate on Attention Deficit Hyperactivity Disorder: Clinical Outcome, Performance, and Cognitive Event‐Related Potentials [PhD thesis]. New York, USA: University of Rochester, 1990. CENTRAL

Flapper 2008 {published data only}

Flapper BCT, Houwen S, Schoemaker MM. Fine motor skills and effects of methylphenidate in children with attention‐deficit‐hyperactivity disorder and developmental coordination disorder. Developmental Medicine & Child Neurology 2006;48(3):165‐9. CENTRAL
Flapper BCT, Schoemaker MM. Effects of methylphenidate on quality of life in children with both developmental coordination disorder and ADHD. Developmental Medicine & Child Neurology 2008;50(4):294‐9. CENTRAL

Forness 1992 {published data only}

Forness SR, Cantwell DP, Swanson JM, Hanna GL, Youpa D. Differential effects of stimulant medication on reading performance of boys with hyperactivity with and without conduct disorder. Journal of Learning Disabilities 1991;24(5):304‐10. CENTRAL
Forness SR, Swanson JM, Cantwell D, Guthrie D, Sena R. Response to stimulant medication across six measures of school‐related performance in children with ADHD and disruptive behavior. Behavioral Disorders 1992;18(1):42‐53. CENTRAL

Froehlich 2011 {published data only}

Froehlich TE, Epstein JN, Nick TG, Melguizo‐Castro MS, Stein MA, Brinkman WB, et al. Pharmacogenetic predictors of methylphenidate dose‐response in attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2011;50(11):1129‐39.e2. CENTRAL

Gadow 1990 {published data only}

Gadow KD, Nolan EE, Paolicelli LM, Sprafkin J. A procedure for assessing the effects of methylphenidate on hyperactive children in public school settings. Journal of Clinical Child Psychology 1991;20(3):268‐76. CENTRAL
Gadow KD, Nolan EE, Sverd J, Sprafkin J, Paolicelli L. Methylphenidate in aggressive‐hyperactive boys: I. Effects on peer aggression in public school settings. Journal of the American Academy of Child and Adolescent Psychiatry 1990;29(5):710‐8. CENTRAL
Gadow KD, Paolicelli LM, Nolan EE, Schwartz J, Sprafkin J, Sverd J. Methylphenidate in aggressive hyperactive boys: II. Indirect effects of medication treatment on peer behavior. Journal of Child and Adolescent Psychopharmacology 1992;2(1):49‐61. CENTRAL

Gadow 1995 {published data only}

Gadow D, Nolan E, Sprafkin J, Sverd J. School observations of children with attention‐deficit hyperactivity disorder and comorbid tic disorder: effects of methylphenidate treatment. Journal of Developmental and Behavioral Pediatrics 1995;16(3):167–76. CENTRAL
Gadow D, Sverd J, Sprafkin J, Nolan E, Ezor N. Efficacy of methylphenidate for attention‐deficit hyperactivity disorder in children with tic disorder. Annual Progress in Child Psychiatry and Child Development. New York: Brunner/Mazel Publishers, 1996:494‐522. CENTRAL
Gadow KD, Nolan EE, Sverd J. Methylphenidate in hyperactive boys with comorbid tic disorder: II. Short‐term behavioral effects in school settings. Journal of the American Academy of Child and Adolescent Psychiatry 1992;31(3):462‐71. CENTRAL
Gadow KD, Nolan EE, Sverd J, Sprafkin J, Schwartz J. Anxiety and depression symptoms and response to methylphenidate in children with attention‐deficit hyperactivity disorder and tic disorder. Journal of Clinical Psychopharmacology 2002;22(3):267‐74. CENTRAL
Gadow KD, Sverd J, Sprafkin J, Nolan EE, Grossman S. Long‐term methylphenidate therapy in children with comorbid attention‐deficit hyperactivity disorder and chronic multiple tic disorder. Archives of General Psychiatry 1999;56(4):330‐6. CENTRAL
Nolan EE, Gadow KD. Children with ADHD and tic disorder and their classmates: behavioral normalization with methylphenidate. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(5):597‐604. CENTRAL
Nolan EE, Gadow KD. Relation between ratings and observations of stimulant drug response in hyperactive children. Journal of Clinical Child Psychology 1994;23(1):78‐90. CENTRAL
Sprafkin J, Gadow KD. Double‐blind versus open evaluations of stimulant drug response in children with attention‐deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1996;6(4):215‐28. CENTRAL
Sverd J, Gadow KD, Nolan EE, Sprafkin J, Ezor SN. Methylphenidate in hyperactive boys with comorbid tic disorder: I. Clinic evaluations. Advances in Neurology 1992;58:271‐81. CENTRAL

Gadow 2007 {published data only}

Gadow KD, Nolan E, Sprafkin J, Sverd J. School observations of children with attention‐deficit hyperactivity disorder and comorbid tic disorder: effects of methylphenidate treatment. Journal of Developmental and Behavioral Pediatrics 1995;16(3):167‐76. CENTRAL
Gadow KD, Nolan EE, Sverd J, Sprafkin J, Schneider J. Methylphenidate in children with oppositional defiant disorder and both comorbid chronic multiple tic disorder and ADHD. Journal of Child Neurology 2008;23(9):981‐90. CENTRAL
Gadow KD, Sverd J, Nolan EE, Sprafkin J, Schneider J. Immediate‐release methylphenidate for ADHD in children with comorbid chronic multiple tic disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(7):840‐8. CENTRAL
Gadow KD, Sverd J, Sprafkin J, Nolan EE, Ezor SN. Efficacy of methylphenidate for attention‐deficit hyperactivity disorder in children with tic disorder. Archives of General Psychiatry 1995;52(6):444‐55. CENTRAL

Gadow 2011 {published data only}

Gadow KD, Nolan EE. Methylphenidate and comorbid anxiety disorder in children with both chronic multiple tic disorder and ADHD. Journal of Attention Disorders 2011;15(3):246‐56. CENTRAL

Garfinkel 1983 {published data only}

Garfinkel BD, Wender PH, Sloman L, O'Neill I. Tricyclic antidepressant and methylphenidate treatment of attention deficit disorder in children. Journal of the American Academy of Child and Adolescent Psychiatry 1983;22(4):343‐8. CENTRAL

Gonzalez‐Heydrich 2010 {published data only}

Gonzalez‐Heydrich J. OROS methylphenidate for attention‐deficit/hyperactivity disorder plus epilepsy. P and T 2006;31(12):725‐6. [EMBASE: 2007065321]CENTRAL
Gonzalez‐Heydrich J, Whitney J, Hsin O, Mrakotsky C, MacMillan C, Torres A, et al. Tolerability of OROS‐MPH 18 and 36 mg in paediatric epilepsy plus attention deficit/hyperactivity disorder (ADHD). Epilepsia. Proceedings of the 26th International Epilepsy Congress; 2005 August 28th ‐ September 1st; Paris, France 2005;46(Suppl s6):179. CENTRAL
Gonzalez‐Heydrich J, Whitney J, Waber D, Forbes P, Hsin O, Faraone SV, et al. Adaptive phase I study of OROS methylphenidate treatment of attention deficit hyperactivity disorder with epilepsy. Epilepsy & Behavior 2010;18(3):229‐37. CENTRAL
NCT00323947. Methylphenidate for treating attention deficit hyperactivity disorder in children with both ADHD and epilepsy. www.clinicaltrials.gov (accessed 1 May 2013). CENTRAL

Gorman 2006 {published data only}

Chang HTT. Effects of methylphenidate on performance and private speech of children with attention‐deficit/hyperactivity disorder during the Tower of Hanoi task. Dissertation Abstracts International: Section B: The Sciences and Engineering 2001;62(1‐B):540. CENTRAL
Gorman EB, Klorman R, Thatcher JE, Borgstedt AD. Effects of methylphenidate on subtypes of attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(7):808‐16. CENTRAL
Kopecky H, Chang HT, Klorman R, Thatcher JE, Borgstedt AD. Performance and private speech of children with attention‐deficit/hyperactivity disorder while taking the Tower of Hanoi test: effects of depth of search, diagnostic subtype, and methylphenidate. Journal of Abnormal Child Psychology 2005;33(5):625‐38. CENTRAL

Green 2011 {published data only}

Green T, Weinberger R, Diamond A, Berant M, Hirschfeld L, Frisch A, et al. The effect of methylphenidate on prefrontal cognitive functioning, inattention, and hyperactivity in velocardiofacial syndrome. Journal of Child and Adolescent Psychopharmacology 2011;21(6):589‐95. CENTRAL
Green T, Weinberger R, Weizman A, Kotler M, Gothelf D. Effect of methylphenidate on neurocognitive functioning in velocardiofacial syndrome: a randomized placebo‐controlled trial. Biological Psychiatry 2009;65(Suppl 8):147. CENTRAL

Greenhill 2002 {published data only}

Greenhill LL, Findling RL, Swanson JM, ADHD Study Group. A double‐blind, placebo‐controlled study of modified‐release methylphenidate in children with attention‐deficit/hyperactivity disorder. Pediatrics 2002;109(3):e39. CENTRAL

Greenhill 2006 {published data only}

Greenhill LL, Muniz R, Ball RR, Levine A, Pestreich L, Jiang H. Efficacy and safety of dexmethylphenidate extended‐release capsules in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(7):817‐23. CENTRAL

Grizenko 2012 {published data only}

Fortier ME, Sengupta SM, Grizenko N, Choudhry Z, Thakur G, Joober R. Genetic evidence for the association of the hypothalamic‐pituitary‐adrenal (HPA) axis with ADHD and methylphenidate treatment response. Neuromolecular Medicine 2013;15(1):122‐32. CENTRAL
Grizenko N, Bhat M, Schwartz G, Ter‐Stepanian M, Joober R. Efficacy of methylphenidate in children with attention‐deficit hyperactivity disorder and learning disabilities: a randomized crossover trial. Journal of Psychiatry & Neuroscience 2006;31(1):46‐51. CENTRAL
Grizenko N, Cai E, Jolicoeur C, Ter‐Stepanian M, Joober R. Effects of methylphenidate on acute math performance in children with attention‐deficit hyperactivity disorder. Canadian Journal of Psychiatry 2013;58(11):632‐9. CENTRAL
Grizenko N, Kovacina B, Amor LB, Schwartz G, Ter‐Stepanian M, Joober R, et al. Relationship between response to methylphenidate treatment in children with ADHD and psychopathology in their families. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(1):47‐53. CENTRAL
Grizenko N, Lachance M, Collard V, Lageix P, Baron C, Ben Amor L, et al. Sensitivity of tests to assess improvement in ADHD symptomatology. Canadian Child and Adolescent Psychiatry Review 2004;13(2):36‐9. CENTRAL
Grizenko N, Pereira RMR, Joober R. Sensitivity of scales to evaluate change in symptomatology with psychostimulants in different ADHD subtypes. Journal of the Canadian Academy of Child and Adolescent Psychiatry 2013;22(2):153‐8. CENTRAL
Grizenko N, Qi Zhang DD, Polotskaia A, Joober R. Efficacy of methylphenidate in ADHD children across the normal and the gifted intellectual spectrum. Journal of the Canadian Academy of Child and Adolescent Psychiatry 2012;21(4):282‐8. CENTRAL
Lee J, Grizenko N, Bhat V, Sengupta S, Polotskaia A, Joober R. Relation between therapeutic response and side effects induced by methylphenidate as observed by parents and teachers of children with ADHD. BMC Psychiatry 2011;11:70. CENTRAL
Ter‐Stepanian M, Grizenko N, Zappitelli M, Joober R. Clinical response to methylphenidate in children diagnosed with attention‐deficit hyperactivity disorder and comorbid psychiatric disorders. Canadian Journal of Psychiatry 2010;55(5):305‐12. CENTRAL
Thakur GA, Grizenko N, Sengupta SM, Schmitz N, Joober R. The 5‐HTTLPR polymorphism of the serotonin transporter gene and short term behavioral response to methylphenidate in children with ADHD. BMC Psychiatry 2010;10:50. CENTRAL
Thakur GA, Sengupta SM, Grizenko N, Choudhry Z, Joober R. Comprehensive phenotype/genotype analyses of the norepinephrine transporter gene (SLC6A2) in ADHD: relation to maternal smoking during pregnancy. PloS One 2012;7(11):e49616. CENTRAL

Gruber 2007 {published data only}

Gruber R, Grizenko N, Schwartz G, Bellingham J, Guzman R, Joober R. Performance on the continuous performance test in children with ADHD is associated with sleep efficiency. Sleep 2007;30(8):1003‐9. CENTRAL
Gruber R, Grizenko N, Schwartz G, Benamor L, Ter‐Stepanian M, Joober R. The associations between sleep and attention in children with attention deficit hyperactivity disorder while on placebo and while on methylphenidate. Sleep. Proceedings of the 19th Annual Meeting of the Associated Professional Sleep Societies; 2005 June 18‐23; Denver, CO. Westchester: American Academy of Sleep Medicine, 2005; Vol. 28:A93‐4. [WOS: 000228906100277]CENTRAL

Hale 2011 {published data only}

Hale JB, Reddy LA, Semrud‐Clikeman M, Hain LA, Whitaker J, Morley J, et al. Executive impairment determines ADHD medication response: implications for academic achievement. Journal of Learning Disabilities 2011;44(2):196‐212. CENTRAL
Kubas HA, Backenson EM, Wilcox G, Piercy JC, Hale JB. The effects of methylphenidate on cognitive function in children with attention‐deficit/hyperactivity disorder. Postgraduate Medicine 2012;124(5):33‐48. CENTRAL

Heriot 2008 {published data only}

Heriot SA, Evans IM, Foster TM. Critical influences affecting response to various treatments in young children with ADHD: a case series. Child: Care, Health and Development 2008;34(1):121‐33. CENTRAL

Hicks 1985 {published data only}

Gualtieri CT, Hicks RE, Mayo JP, Schroeder SR. The persistence of stimulant effects in chronically treated children: further evidence of an inverse relationship between drug effects and placebo levels of response. Psychopharmacology 1984;83(1):44‐7. CENTRAL
Hicks RE, Gualtieri CT, Mayo JP, Schroeder SR, Lipton MA. Methylphenidate and homeostasis: drug effects on the cognitive performance of hyperactive children. In: Bloomingdale Lewis M editor(s). Attention Deficit Disorder: Identification, Course and Treatment Rationale. New York, USA: SP Medical & Scientific Books, 1985:131‐41. CENTRAL

Hoeppner 1997 {published data only}

Hoeppner JA, Hale JB, Bradley AM, Byrnes M, Coury DL, Lennie L, et al. A clinical protocol for determining methylphenidate dosage levels in ADHD. Journal of Attention Disorders 1997;2(1):19‐30. CENTRAL

Horn 1991 {published data only}

Horn WF, Ialongo NS, Pascoe JM, Greenberg G, Packard T, Lopez M, et al. Additive effects of psychostimulants, parent training, and self‐control therapy with ADHD children. Journal of the American Academy of Child and Adolescent Psychiatry 1991;30(2):233‐40. CENTRAL
Ialongo NS, Horn WF, Pascoe JM, Greenberg G, Packard T, Lopez M, et al. The effects of a multimodal intervention with attention‐deficit hyperactivity disorder children: a 9‐month follow‐up. Journal of the American Academy of Child and Adolescent Psychiatry 1993;32(1):182‐9. CENTRAL

Ialongo 1994 {published data only}

Ialongo NS, Lopez M, Horn WF, Pascoe JM, Greenberg G. Effects of psychostimulant medication on self‐perceptions of competence, control, and mood in children with attention deficit hyperactivity disorder. Journal of Clinical Child Psychology 1994;23(2):161‐73. CENTRAL

Jacobi‐Polishook 2009 {published data only}

Jacobi‐Polishook T, Shorer Z, Melzer I. The effect of methylphenidate on postural stability under single and dual task conditions in children with attention deficit hyperactivity disorder ‐ a double blind randomized control trial. Journal of the Neurological Sciences 2009;280(1‐2):15‐21. CENTRAL

Jensen 1999 (MTA) {published data only}

Abikoff H. Tailored psychosocial treatments for ADHD: the search for a good fit. Journal of Clinical Child Psychology2001; Vol. 30, issue 1:122‐5. CENTRAL
Arnold LE, Abikoff HB, Cantwell DP, Conners CK, Elliott G, Greenhill LL, et al. National Institute of Mental Health Collaborative Multimodal Treatment Study of Children with ADHD (the MTA). Design challenges and choices. Archives of General Psychiatry 1997;54(9):865‐70. CENTRAL
Arnold LE, Abikoff HB, Cantwell DP, Conners CK, Elliott GR, Greenhill LL, et al. NIMH Collaborative Multimodal Treatment Study of Children with ADHD (MTA): design, methodology, and protocol evolution. Journal of Attention Disorders 1997;2(3):141‐58. CENTRAL
Arnold LE, Chuang S, Davies M, Abikoff HB, Conners CK, Elliott GR, et al. Nine months of multicomponent behavioral treatment for ADHD and effectiveness of MTA fading procedures. Journal of Abnormal Child Psychology 2004;32(1):39‐51. CENTRAL
Arnold LE, Elliott M, Sachs L, Bird H, Kraemer HC, Wells KC, et al. Effects of ethnicity on treatment attendance, stimulant response/dose, and 14‐month outcome in ADHD. Journal of Consulting and Clinical Psychology 2003;71(4):713‐27. CENTRAL
Carey WB. What the multimodal treatment study of children with attention deficit/hyperactivity disorder did and did not say about the use of methylphenidate for attention deficits. Pediatrics 2000;105(4):863‐4. CENTRAL
Conners CK, Epstein JN, March JS, Angold A, Wells KC, Klaric J, et al. Multimodal treatment of ADHD in the MTA: an alternative outcome analysis. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):159‐67. CENTRAL
Galanter CA, Carlson GA, Jensen PS, Greenhill LL, Davies M, Li W, et al. Response to methylphenidate in children with attention deficit hyperactivity disorder and manic symptoms in the multimodal treatment study of children with attention deficit hyperactivity disorder titration trial. Journal of Child and Adolescent Psychopharmacology 2003;13(2):123‐36. CENTRAL
Greene RW, Ablon JS. What does the MTA study tell us about effective psychosocial treatment for ADHD?. Journal of Clinical Child Psychology 2001;30(1):114‐21. CENTRAL
Greenhill LL, Abikoff HB, Arnold LE, Cantwell DP, Conners CK, Elliott G, et al. Medication treatment strategies in the MTA Study: relevance to clinicians and researchers. Journal of the American Academy of Child and Adolescent Psychiatry 1996;35(10):1304‐13. CENTRAL
Greenhill LL, Swanson JM, Vitiello B, Davies M, Clevenger W, Wu M, et al. Impairment and deportment responses to different methylphenidate doses in children with ADHD: the MTA titration trial. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):180‐7. CENTRAL
Hansen L, Thomsen PH. How to treat ADHD/DAMP? Is there a conclusive answer? A critical survey of the MTA trial. Ugeskrift for Laeger 2005;167(48):4555‐9. CENTRAL
Hinshaw SP. Moderators and mediators of treatment response for children with attention‐deficit/hyperactivity disorder: the multimodal treatment study of children with attention‐deficit/hyperactivity disorder. Archives of General Psychiatry 1999;56(12):1088‐96. CENTRAL
Hinshaw SP, March JS, Abikoff H, Arnold LE, Cantwell DP, Conners CK, et al. Comprehensive assessment of childhood attention‐deficit hyperactivity disorder in the context of a multisite, multimodal clinical trial. Journal of Attention Disorders 1997;1(4):217‐34. CENTRAL
Jensen PS, Arnold LE, Richters JE, Severe JB, Vereen D, Vitiello B, et al. A 14‐month randomized clinical trial of treatment strategies for attention‐deficit/hyperactivity disorder. Archives of General Psychiatry 1999;56(12):1073‐86. CENTRAL
Jensen PS, Arnold LE, Severe JB, Vitiello B, Hoagwood K. National Institute of Mental Health multimodal treatment study of ADHD follow‐up: changes in effectiveness and growth after the end of treatment. Pediatrics 2004;113(4):762‐9. CENTRAL
Jensen PS, Arnold LE, Swanson JM, Vitiello B, Abikoff HB, Greenhill LL, et al. 3‐Year follow‐up of the NIMH MTA study. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(8):989‐1002. CENTRAL
Jensen PS, Hinshaw SP, Kraemer HC, Lenora N, Newcorn JH, Abikoff HB, et al. ADHD comorbidity findings from the MTA study: comparing comorbid subgroups. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):147‐58. CENTRAL
Jensen PS, Hinshaw SP, Swanson JM, Greenhill LL, Conners CK, Arnold LE, et al. Findings from the NIMH Multimodal Treatment Study of ADHD (MTA): implications and applications for primary care providers. Journal of Developmental and Behavioral Pediatrics 2001;22(1):60‐73. CENTRAL
MTA Cooperative Group. National Institute of Mental Health Multimodal Treatment Study of ADHD follow‐up: 24‐month outcomes of treatment strategies for attention‐deficit/hyperactivity disorder. Pediatrics 2004;113(4):754‐61. CENTRAL
March JS, Swanson JM, Arnold LE, Hoza B, Conners CK, Hinshaw SP, et al. Anxiety as a predictor and outcome variable in the multimodal treatment study of children with ADHD (MTA). Journal of Abnormal Child Psychology 2000;28(6):527‐41. CENTRAL
Molina BSG, Flory K, Hinshaw SP, Greiner AR, Arnold LE, Swanson JM, et al. Delinquent behavior and emerging substance use in the MTA at 36 months: prevalence, course, and treatment effects. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(8):1028‐40. CENTRAL
Molina BSG, Hinshaw SP, Arnold LE, Swanson JM, Pelham WE, Hechtman L, et al. Adolescent substance use in the multimodal treatment study of attention‐deficit/hyperactivity disorder (ADHD) (MTA) as a function of childhood ADHD, random assignment to childhood treatments, and subsequent medication. Journal of the American Academy of Child and Adolescent Psychiatry 2013;52(3):250‐63. CENTRAL
Molina BSG, Hinshaw SP, Swanson JM, Arnold LE, Vitiello B, Jensen PS, et al. The MTA at 8 years: prospective follow‐up of children treated for combined‐type ADHD in a multisite study. Journal of the American Academy of Child and Adolescent Psychiatry 2009;48(5):484‐500. CENTRAL
Nieweg EH. Does ADHD medication stop working after 2‐3 years? On the surprising, but little known follow‐up of the MTA study [Is ADHD‐medicatie na 2‐3 jaar uitgewerkt? Over de verrassende, maar weinig bekende follow‐up van het MTA‐onderzoek]. Tijdschrift voor Psychiatrie 2010;52(4):245‐54. CENTRAL
Owens EB, Hinshaw SP, Kraemer HC, Arnold LE, Abikoff HB, Cantwell DP, et al. Which treatment for whom for ADHD? Moderators of treatment response in the MTA. Journal of Consulting and Clinical Psychology 2003;71(3):540‐52. CENTRAL
Pelham WE. The NIMH multimodal treatment study for attention‐deficit hyperactivity disorder: just say yes to drugs alone?. Canadian Journal of Psychiatry 1999;44(10):981‐90. CENTRAL
Pelham WE, Gnagy EM, Greiner AR, Hoza B, Hinshaw SP, Swanson JM, et al. Behavioral versus behavioral and pharmacological treatment in ADHD children attending a summer treatment program. Journal of Abnormal Child Psychology 2000;28(6):507‐25. CENTRAL
Richters JE, Arnold LE, Jensen PS, Abikoff H, Conners CK, Greenhill LL, et al. NIMH collaborative multisite multimodal treatment study of children with ADHD: I. Background and rationale. Journal of the American Academy of Child and Adolescent Psychiatry 1995;34(8):987‐1000. CENTRAL
Swanson J, Arnold LE, Kraemer H, Hechtman L, Molina B, Hinshaw S, et al. Evidence, interpretation, and qualification from multiple reports of long‐term outcomes in the Multimodal Treatment Study of Children with ADHD (MTA): part I: executive summary. Journal of Attention Disorders 2008;12(1):4‐14. CENTRAL
Swanson J, Arnold LE, Kraemer H, Hechtman L, Molina B, Hinshaw S, et al. Evidence, interpretation, and qualification from multiple reports of long‐term outcomes in the Multimodal Treatment Study of Children with ADHD (MTA): part II: supporting details. Journal of Attention Disorders 2008;12(1):15‐43. CENTRAL
Swanson JM, Elliott GR, Greenhill LL, Wigal T, Arnold LE, Vitiello B, et al. Effects of stimulant medication on growth rates across 3 years in the MTA follow‐up. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(8):1015‐27. CENTRAL
Swanson JM, Hinshaw SP, Arnold LE, Gibbons RD, Marcus S, Hur K, et al. Secondary evaluations of MTA 36‐month outcomes: propensity score and growth mixture model analyses. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(8):1003‐14. CENTRAL
Swanson JM, Kraemer HC, Hinshaw SP, Arnold LE, Conners CK, Abikoff HB, et al. Clinical relevance of the primary findings of the MTA: success rates based on severity of ADHD and ODD symptoms at the end of treatment. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):168‐79. CENTRAL
Vitiello B, Elliott GR, Swanson JM, Arnold LE, Hechtman L, Abikoff H, et al. Blood pressure and heart rate over 10 years in the Multimodal Treatment Study of Children with ADHD. American Journal of Psychiatry2012; Vol. 169, issue 2:167‐77. CENTRAL
Vitiello B, Severe JB, Greenhill LL, Arnold LE, Abikoff HB, Bukstein OG, et al. Methylphenidate dosage for children with ADHD over time under controlled conditions: lessons from the MTA. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):188‐96. CENTRAL

Johnston 1988 {published data only}

Johnston C, Pelham WE, Hoza J, Sturges J. Psychostimulant rebound in attention deficit disordered boys. Journal of the American Academy of Child and Adolescent Psychiatry 1988;27(6):806‐10. CENTRAL
Pelham WE, Sturges J, Hoza J, Schmidt C, Bijlsma JJ, Milich R, et al. Sustained release and standard methylphenidate effects on cognitive and social behavior in children with attention deficit disorder. Pediatrics 1987;80(4):491‐501. CENTRAL
Pelham WE, Hoza J. Behavioral assessment of psychostimulant effects on ADD children in a summer day treatment program. In: Prinz R editor(s). Advances in Behavioral Assessment of Children and Families. Vol. 3, Greenwich, CT: JAI Press, 1987:3‐34. CENTRAL
Pelham WE, Milich R, Walker JL. Effects of continuous and partial reinforcement and methylphenidate on learning in children with attention deficit disorder. Journal of Abnormal Psychology 1986;95(4):319‐25. CENTRAL

Kaplan 1990 {published data only}

Kaplan SL, Busner J, Kupietz S, Wassermann E, Segal B. Effects of methylphenidate on adolescents with aggressive conduct disorder and ADDH: a preliminary report. Journal of the American Academy of Child and Adolescent Psychiatry 1990;29(5):719‐23. CENTRAL

Kelly 1989 {published data only}

Kelly PC, Cohen ML, Walker WO, Caskey OL, Atkinson AW. Self‐esteem in children medically managed for attention deficit disorder. Pediatrics 1989;83(2):211‐7. CENTRAL

Kent 1995 {published data only}

Kent JD, Blader JC, Koplewicz HS, Abikoff H, Foley CA. Effects of late‐afternoon methylphenidate administration on behavior and sleep in attention‐deficit hyperactivity disorder. Pediatrics 1995;96(2):320‐5. CENTRAL

Kent 1999 {published data only}

Kent MA, Camfield CS, Camfield PR. Double‐blind methylphenidate trials: practical, useful, and highly endorsed by families. Archives of Pediatrics & Adolescent Medicine 1999;153(12):1292‐6. CENTRAL

Klorman 1990 {published data only}

Klorman R, Brumaghim JT, Fitzpatrick PA, Borgstedt AD. Clinical effects of a controlled trial of methylphenidate on adolescents with attention deficit disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1990;29(5):702‐9. CENTRAL
Klorman R, Brumaghim JT, Fitzpatrick PA, Borgstedt AD. Methylphenidate reduces abnormalities of stimulus classification in adolescents with attention deficit disorder. Journal of Abnormal Psychology 1992;101(1):130‐8. CENTRAL
Klorman R, Brumaghim JT, Fitzpatrick PA, Borgstedt AD. Methylphenidate speeds evaluation processes of attention deficit disorder adolescents during a continuous performance test. Journal of Abnormal Child Psychology 1991;19(3):263‐83. CENTRAL

Kolko 1999 {published data only}

Kolko DJ, Bukstein OG, Barron J. Methylphenidate and behavior modification in children with ADHD and comorbid ODD or CD: main and incremental effects across settings. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(5):578‐86. CENTRAL

Kollins 2006 (PATS) {published data only}

At a glance... study design sought to balance rigor, subject safety. The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 12:4‐5. CENTRAL
MPH‐related reductions in growth rates. The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 12:6. CENTRAL
PATS shows mixed effect of medication on functional outcomes. The Brown University Child & Adolescent Psychopharmacology Update2008; Vol. 10, issue 2:6. CENTRAL
PATS: efficacy of MPH in ADHD preschoolers. The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 12:5‐6. CENTRAL
PATS: safety and tolerability of MPH in ADHD preschoolers. The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 12:2‐4. CENTRAL
What are the effects of methylphenidate treatment in preschoolers with ADHD? Results from the Preschool ADHD Treatment Study (PATS). The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 12:1‐2. CENTRAL
Young ADHD patients may improve with low‐dose meds. Psychiatric Annals2006; Vol. 36, issue 12:826. CENTRAL
Abikoff HB, Vitiello B, Riddle MA, Cunningham C, Greenhill LL, Swanson JM, et al. Methylphenidate effects on functional outcomes in the Preschoolers with Attention‐Deficit/Hyperactivity Disorder Treatment Study (PATS). Journal of Child and Adolescent Psychopharmacology 2007;17(5):581‐92. CENTRAL
Ghuman JK, Riddle MA, Vitiello B, Greenhill LL, Chuang SZ, Wigal SB, et al. Comorbidity moderates response to methylphenidate in the Preschoolers with Attention‐Deficit/Hyperactivity Disorder Treatment Study (PATS). Journal of Child and Adolescent Psychopharmacology 2007;17(5):563‐80. CENTRAL
Greenhill LL. Preschool ADHD treatment study (PATS): science and controversy. Economics of Neuroscience 2001;3(5):49‐53. [EMBASE: 2001251865]CENTRAL
Greenhill LL, Kollins S, Abikoff H, McCracken J, Riddle M, Swanson J, et al. Efficacy and safety of immediate‐release methylphenidate treatment for preschoolers with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1284‐93. CENTRAL
Greenhill LL, Kollins S, Abikoff H, McCracken J, Riddle M, Swanson J, et al. Erratum: "Efficacy and safety of immediate‐release MPH treatment for preschoolers with ADHD". Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(1):141. Erratum for: Journal of the American Academy of Child and Adolescent Psychiatry 2006;45:1284‐93. CENTRAL
Kollins S, Greenhill L, Swanson J, Wigal S, Abikoff H, McCracken J, et al. Rationale, design, and methods of the Preschool ADHD Treatment Study (PATS). Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1275‐83. CENTRAL
Kollins SH, Greenhill L. Evidence base for the use of stimulant medication in preschool children with ADHD. Infants and Young Children 2006;19(2):132‐41. CENTRAL
March JS. The preschool ADHD Treatment Study (PATS) as the culmination of twenty years of clinical trials in pediatric psychopharmacology. Journal of the American Academy of Child and Adolescent Psychiatry 2011;50(5):427‐30. CENTRAL
McGough J, McCracken J, Swanson J, Riddle M, Kollins S, Greenhill L, et al. Pharmacogenetics of methylphenidate response in preschoolers with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1314‐22. CENTRAL
Reiff MI. Journal article reviews: attention‐deficit/hyperactivity disorder. Journal of Developmental and Behavioral Pediatrics2007; Vol. 28, issue 1:71‐2. CENTRAL
Riddle MA. New findings from the Preschoolers with Attention‐Deficit/Hyperactivity Disorder Treatment Study (PATS). Journal of Child and Adolescent Psychopharmacology 2007;17(5):543‐6. CENTRAL
Riddle MA, Yershova K, Lazzaretto D, Paykina N, Yenokyan G, Greenhill L, et al. The Preschool Attention‐Deficit/Hyperactivity Disorder Treatment Study (PATS) 6‐year follow‐up. Journal of the American Academy of Child and Adolescent Psychiatry 2013;52(3):264‐78.e2. CENTRAL
Swanson J, Greenhill L, Wigal T, Kollins S, Stehli A, Davies M, et al. Stimulant‐related reductions of growth rates in the PATS. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1304‐13. CENTRAL
Vitiello B, Abikoff HB, Chuang SZ, Kollins SH, McCracken JT, Riddle MA, et al. Effectiveness of methylphenidate in the 10‐month continuation phase of the Preschoolers with ADHD Treatment Study (PATS). Journal of Child and Adolescent Psychopharmacology 2007;17(5):593‐603. CENTRAL
Wagner KD. Methylphenidate treatment of ADHD in preschoolers. Psychiatric Times 2007;24(3):47. CENTRAL
Wigal T, Greenhill L, Chuang S, McGough J, Vitiello B, Skrobala A, et al. Safety and tolerability of methylphenidate in preschool children with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1294‐303. CENTRAL

Konrad 2004 {published data only}

Konrad K, Günther T, Hanisch C, Herpertz‐Dahlmann B. Differential effects of methylphenidate on attentional functions in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(2):191‐8. CENTRAL

Konrad 2005 {published data only}

Konrad K, Günther T, Heinzel‐Gutenbrunner M, Herpertz‐Dahlmann B. Clinical evaluation of subjective and objective changes in motor activity and attention in children with attention‐deficit/hyperactivity disorder in a double‐blind methylphenidate trial. Journal of Child and Adolescent Psychopharmacology 2005;15(2):180‐90. CENTRAL

Leddy 2009 {published data only}

Leddy JJ, Waxmonsky JG, Salis RJ, Paluch RA, Gnagy EM, Mahaney P, et al. Dopamine‐related genotypes and the dose‐response effect of methylphenidate on eating in attention‐deficit/hyperactivity disorder youths. Journal of Child and Adolescent Psychopharmacology 2009;19(2):127‐36. CENTRAL

Lehmkuhl 2002 {published data only}

Döpfner M, Banaschewski T, Schmidt J, Uebel H, Schmeck K, Gerber WD, et al. Long‐acting methylphenidate preparation in children with ADHD ‐ a multicenter study [Langzeitwirksames methylphenidat bei kindern mit aufmerksamkeitsdefizit‐hyperaktivitätsstörungen: eine multizentrische studie]. Nervenheilkunde: Zeitschrift fur interdisziplinaere Fortbildung 2003;22(2):85‐92. CENTRAL
Lehmkuhl G. Placebo‐controlled, double‐blind multicenter trial on the efficacy of sustained‐release methylphenidate in children suffering from attention deficit hyperactivity disorder (ADHD), Phase III. Integrated Final Report. Institut für medizinische Informatik, Biometrie und Epidemiologie, Universitätsklinikum Essen, Trial no 6520‐9979‐02, 1‐92002. CENTRAL
Sinzig J, Döpfner M, Lehmkuhl G, Uebel H, Schmeck K, Poustka F, et al. Long‐acting methylphenidate has an effect on aggressive behavior in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2007;17(4):421‐32. CENTRAL
Sinzig JK, Döpfner M, Plück J, Banaschewski T, Stephani U, Lehmkuhl G, et al. Does a morning dose of methylphenidate retard reduce hyperkinetic symptoms in the afternoon? [Lassen sich hyperkinetische auffälligkeiten am nachmittag durch eine morgengabe von methylphenidat retard vermindern?]. Zeitschrift für Kinder‐ und Jugendpsychiatrie und Psychotherapie 2004;32(4):225‐33. CENTRAL

Lijffijt 2006 {published data only}

Lijffijt M, Kenemans JL, Ter Wal A, Quik EH, Kemner C, Westenberg H, et al. Dose‐related effect of methylphenidate on stopping and changing in children with attention‐deficit/hyperactivity disorder. European Psychiatry 2006;21(8):544‐7. CENTRAL

Lin 2014 {published data only}

Lin DY, Kratochvil CJ, Xu W, Jin L, D'Souza DN, Kielbasa W, et al. A randomized trial of edivoxetine in pediatric patients with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2014;24(4):190‐200. CENTRAL

Lopez 2003 {published data only}

Lopez F, Silva R, Pestreich L, Muniz R. Comparative efficacy of two once daily methylphenidate formulations (Ritalin® LA™1 and Concerta®) and placebo in children with attention deficit hyperactivity disorder across the school day. Paediatric Drugs 2003;5(8):545‐55. CENTRAL
Lopez F, Silva R, Pestreich L, Muniz R. Erratum for: Comparative efficacy of two once daily methylphenidate formulations (Ritalin LA1 and Concerta) and placebo in children with attention deficit hyperactivity disorder across the school day. Pediatric Drugs 2003;5(12):832. Erratum for Pediatric Drugs 2003;5:545. [EMBASE: 2004010793]CENTRAL
Lopez FA, Silva RR, Pestreich L, Lee J, Muniz R. Comparative school‐day efficacy of Ritalin LA, Concerta, and placebo in children with attention deficit hyperactivity disorder. Annals of Neurology. Proceedings of the 32nd Annual Meeting of the Child Neurology Society; 2003 October 1‐4; Miami Beach, Florida. New York: John Wiley & Sons, 2003; Vol. 54 (Suppl 7):S143. [WOS: 000185260300393]CENTRAL

Lufi 1997 {published data only}

Lufi D, Parish‐Plass J, Gai E. The effect of methylphenidate on the cognitive and personality functioning of ADHD children. Israel Journal of Psychiatry and Related Sciences 1997;34(3):200‐9. CENTRAL

Lufi 2007 {published data only}

Lufi D, Gai E. The effect of methylphenidate and placebo on eye‐hand coordination functioning and handwriting of children with attention deficit hyperactivity disorder. Neurocase 2007;13(5):334‐41. CENTRAL

Manos 1999 {published data only}

Faraone SV, Short EJ, Biederman J, Findling RL, Roe C, Manos MJ. Efficacy of Adderall and methylphenidate in attention deficit hyperactivity disorder: a drug‐placebo and drug‐drug response curve analysis of a naturalistic study. International Journal of Neuropsychopharmacology 2002;5(2):121‐9. CENTRAL
Findling RL, Short EJ, Manos MJ. Developmental aspects of psychostimulant treatment in children and adolescents with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(12):1441‐7. CENTRAL
Findling RL, Short EJ, Manos MJ. Short‐term cardiovascular effects of methylphenidate and Adderall. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(5):525‐9. CENTRAL
Manos MJ, Short EJ, Findling RL. Differential effectiveness of methylphenidate and Adderall in school‐age youths with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(7):813‐9. CENTRAL

Martins 2004 {published data only}

Martins S, Tramontina S, Polanczyk G, Eizirik M, Swanson JM, Rohde LA. Weekend holidays during methylphenidate use in ADHD children: a randomized clinical trial. Journal of Child and Adolescent Psychopharmacology 2004;14(2):195‐206. CENTRAL

McBride 1988a {published data only}

McBride MC. An individual double‐blind crossover trial for assessing methylphenidate response in children with attention deficit disorder. Journal of Pediatrics 1988;113(1 Pt 1):137‐45. CENTRAL

McGough 2006 {published data only}

McGough JJ, Wigal SB, Abikoff H, Turnbow JM, Posner K, Moon E. A randomized, double‐blind, placebo‐controlled, laboratory classroom assessment of methylphenidate transdermal system in children with ADHD. Journal of Attention Disorders 2006;9(3):476‐85. CENTRAL
Wigal S, Turnbow J, Abikoff H, McGough J, Cohen J. Parent rated effects of transdermal methylphenidate in children with ADHD. International Journal of Neuropsychopharmacology. Abstracts from the XXVI Collegium Internationale Neuro‐Psychopharmacologicum; 2008 July 13‐17; Munich. 2008; Vol. 11 Suppl 1:232. CENTRAL

McInnes 2007 {published data only}

McInnes A, Bedard A‐C, Hogg‐Johnson S, Tannock R. Preliminary evidence of beneficial effects of methylphenidate on listening comprehension in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2007;17(1):35‐49. CENTRAL

Moshe 2012 {published data only}

Moshe K, Karni A, Tirosh E. Anxiety and methylphenidate in attention deficit hyperactivity disorder: a double‐blind placebo‐drug trial. Attention Deficit and Hyperactivity Disorders 2012;4(3):153‐8. CENTRAL

Muniz 2008 {published data only}

Muniz R, Brams M, Mao A, McCague K, Pestreich L, Silva R. Efficacy and safety of extended‐release dexmethylphenidate compared with d,l‐methylphenidate and placebo in the treatment of children with attention‐deficit/hyperactivity disorder: a 12‐hour laboratory classroom study. Journal of Child and Adolescent Psychopharmacology 2008;18(3):248‐56. CENTRAL
Silva R, Muniz R, McCague K, Childress A, Brams M, Mao A. Treatment of children with attention‐deficit/hyperactivity disorder: results of a randomized, multicenter, double‐blind, crossover study of extended‐release dexmethylphenidate and D,L‐methylphenidate and placebo in a laboratory classroom setting. Psychopharmacology Bulletin 2008;41(1):19‐33. CENTRAL

Murray 2011 {published data only}

Murray DW, Childress A, Giblin J, Williamson D, Armstrong R, Starr HL. Effects of OROS methylphenidate on academic, behavioral, and cognitive tasks in children 9 to 12 years of age with attention‐deficit/hyperactivity disorder. Clinical Pediatrics 2011;50(4):308‐20. CENTRAL

Musten 1997 {published data only}

Firestone P, Musten LM, Pisterman S, Mercer J, Bennett S. Short‐term side effects of stimulant medication are increased in preschool children with attention‐deficit/hyperactivity disorder: a double‐blind placebo‐controlled study. Journal of Child and Adolescent Psychopharmacology 1998;8(1):13‐25. CENTRAL
Musten LM. Efficacy of stimulant medication treatment of attention deficit hyperactivity disorder in preschool‐aged children. Dissertation Abstracts International: Section B: The Sciences and Engineering1998; Vol. 59, issue 3‐B:1374. CENTRAL
Musten LM, Firestone P, Pisterman S, Bennett S, Mercer J. Effects of methylphenidate on preschool children with ADHD: cognitive and behavioral functions. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(10):1407‐15. CENTRAL

Newcorn 2008 {published data only}

Newcorn JH, Kratochvil CJ, Allen AJ, Casat CD, Ruff DD, Moore RJ, et al. Atomoxetine and osmotically released methylphenidate for the treatment of attention deficit hyperactivity disorder: acute comparison and differential response. American Journal of Psychiatry 2008;165(6):721‐30. CENTRAL
Newcorn JH, Kratochvil CJ, Allen AJ, et al. Osmotically released methylphenidate compared to atomoxetine for ADHD. The Brown University Child & Adolescent Psychopharmacology Update2008; Vol. 10, issue 8:1. CENTRAL
Toplak ME. Osmotically released methylphenidate is more effective than atomoxetine in children and adolescents with ADHD. Evidence‐Based Mental Health 2009;12(1):19. CENTRAL

Nikles 2006 {published data only}

Nikles CJ, Mitchell GK, Del Mar CB, Clavarino A, McNairn N. An n‐of‐1 trial service in clinical practice: testing the effectiveness of stimulants for attention‐deficit/hyperactivity disorder. Pediatrics 2006;117(6):2040‐6. CENTRAL
Nikles CJ, Mitchell GK, Del Mar CB, McNairn N, Clavarino A. Long‐term changes in management following n‐of‐1 trials of stimulants in attention‐deficit/hyperactivity disorder. European Journal of Clinical Pharmacology 2007;63(11):985‐9. CENTRAL

Oesterheld 1998 {published data only}

Oesterheld JR, Kofoed L, Tervo R, Fogas B, Wilson A, Fiechtner H. Effectiveness of methylphenidate in Native American children with fetal alcohol syndrome and attention deficit/hyperactivity disorder: a controlled pilot study. Journal of Child and Adolescent Psychopharmacology 1998;8(1):39‐48. CENTRAL

Overtoom 2003 {published data only}

Overtoom CCE, Verbaten MN, Kemner C, Kenemans JL, Van Engeland H, Buitelaar JK, et al. Effects of methylphenidate, desipramine, and L‐dopa on attention and inhibition in children with attention deficit hyperactivity disorder. Behavioural Brain Research 2003;145(1‐2):7‐15. CENTRAL

Palumbo 2008 {published data only}

Cannon M, Pelham WH, Sallee FR, Palumbo DR, Bukstein O, Daviss WB. Effects of clonidine and methylphenidate on family quality of life in attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2009;19(5):511‐7. CENTRAL
Daviss WB, Patel NC, Robb AS, McDermott MP, Bukstein OG, Pelham WE, et al. Clonidine for attention‐deficit/hyperactivity disorder: II. ECG changes and adverse events analysis. Journal of the American Academy of Child and Adolescent Psychiatry 2008;47(2):189‐98. CENTRAL
Palumbo DR, Sallee FR, Pelham WE, Bukstein OG, Daviss WB, McDermott MP. Clonidine for attention‐deficit/hyperactivity disorder: I. Efficacy and tolerability outcomes. Journal of the American Academy of Child and Adolescent Psychiatry 2008;47(2):180‐8. CENTRAL

Pearson 2013 {published data only}

Methylphenidate dosing improved behavior in children with ASD. The Brown University Child & Adolescent Psychopharmacology Update2013; Vol. 15, issue 8:4‐5. CENTRAL
Pearson DA, Santos CW, Aman MG, Arnold LE, Casat CD, Mansour R, et al. Effects of extended release methylphenidate treatment on ratings of attention‐deficit/hyperactivity disorder (ADHD) and associated behavior in children with autism spectrum disorders and ADHD symptoms. Journal of Child and Adolescent Psychopharmacology 2013;23(5):337‐51. CENTRAL

Pelham 1989 {published data only}

Pelham WE, Walker JL, Sturges J, Hoza J. Comparative effects of methylphenidate on ADD girls and ADD boys. Journal of the American Academy of Child and Adolescent Psychiatry 1989;28(5):773‐6. CENTRAL

Pelham 1990a {published data only}

Pelham Jr WE, Greenslade KE, Vodde‐Hamilton M, Murphy DA, Greenstein JJ, Gnagy EM, et al. Relative efficacy of long‐acting stimulants on children with attention deficit‐hyperactivity disorder: a comparison of standard methylphenidate, sustained‐release methylphenidate, sustained‐release dextroamphetamine, and pemoline. Pediatrics 1990;86(2):226‐37. CENTRAL

Pelham 1993a {published data only}

Pelham J, Carlson C, Sams SE, Vallano G, Dixon MJ, Hoza B, et al. Separate and combined effects of methylphenidate and behavior modification on boys with attention deficit‐hyperactivity disorder in the classroom. Journal of Consulting and Clinical Psychology 1993;61(3):506‐15. CENTRAL

Pelham 1999 {published data only}

Pelham WE, Aronoff HR, Midlam JK, Shapiro CJ, Gnagy EM, Chronis AM, et al. A comparison of Ritalin and Adderall: efficacy and time‐course in children with attention‐deficit/hyperactivity disorder. Pediatrics 1999;103(4):e43. CENTRAL

Pelham 2001a {published data only}

Pelham WE, Gnagy EM, Burrows‐Maclean L, Williams A, Fabiano GA, Morrisey SM, et al. Once‐a‐day Concerta methylphenidate versus three‐times‐daily methylphenidate in laboratory and natural settings. Pediatrics 2001;107(6):E105. CENTRAL

Pelham 2002 {published data only}

Pelham WE, Hoza B, Pillow DR, Gnagy EM, Kipp HL, Greiner AR, et al. Effects of methylphenidate and expectancy on children with ADHD: behavior, academic performance, and attributions in a summer treatment program and regular classroom settings. Journal of Consulting and Clinical Psychology 2002;70(2):320‐35. CENTRAL

Pelham 2005 {published data only}

Chacko A, Pelham WE, Gnagy EM, Greiner A, Vallano G, Bukstein O, et al. Stimulant medication effects in a summer treatment program among young children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2005;44(3):249‐57. CENTRAL
Pelham WE, Burrows‐MacLean L, Gnagy EM, Fabiano GA, Coles EK, Tresco KE, et al. Transdermal methylphenidate, behavioral, and combined treatment for children with ADHD. Experimental and Clinical Psychopharmacology 2005;13(2):111‐26. CENTRAL
Pelham WE, Manos MJ, Ezzell CE, Tresco KE, Gnagy EM, Hoffman MT, et al. A dose‐ranging study of a methylphenidate transdermal system in children with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2005;44(6):522‐9. CENTRAL

Pelham 2011 {published data only}

Pelham WE, Waxmonsky JG, Schentag J, Ballow CH, Panahon CJ, Gnagy EM, et al. Efficacy of a methylphenidate transdermal system versus t.i.d. methylphenidate in a laboratory setting. Journal of Attention Disorders 2011;15(1):28‐35. CENTRAL

Pelham 2014 {published data only}

Pelham WE, Burrows‐MacLean L, Gnagy EM, Fabiano GA, Coles EK, Wymbs BT, et al. A dose‐ranging study of behavioral and pharmacological treatment in social settings for children with ADHD. Journal of Abnormal and Child Psychology 2014;42(6):1019‐31. CENTRAL

Perez‐Alvarez 2009 {published data only}

Perez‐Alvarez F, Serra‐Amaya C, Timoneda‐Gallart CA. Cognitive versus behavioral ADHD phenotype: what is it all about?. Neuropediatrics 2009;40(1):32‐8. CENTRAL

Pliszka 1990 {published data only}

Pliszka SR. Effect of anxiety on cognition, behavior, and stimulant response in ADHD. In: Chess S, Hertzig ME editor(s). Annual Progress in Child Psychiatry and Child Development. London: Routledge, 1990:454‐66. CENTRAL
Pliszka SR. Effect of anxiety on cognition, behavior, and stimulant response in ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 1989;28(6):882‐7. CENTRAL

Pliszka 2000 {published data only}

Pliszka SR, Browne RG, Olvera RL, Wynne SK. A double‐blind, placebo‐controlled study of Adderall and methylphenidate in the treatment of attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2000;39(5):619‐26. CENTRAL

Pliszka 2007 {published data only}

Pliszka SR, Liotti M, Bailey BY, Perez R, Glahn D, Semrud‐Clikeman M. Electrophysiological effects of stimulant treatment on inhibitory control in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2007;17(3):356‐66. CENTRAL

Quinn 2004 {published data only}

Quinn D, Wigal S, Swanson J, Hirsch S, Ottolini Y, Dariani M, et al. Comparative pharmacodynamics and plasma concentrations of d‐threo‐methylphenidate hydrochloride after single doses of d‐threo‐methylphenidate hydrochloride and d,l‐threo‐methylphenidate hydrochloride in a double‐blind, placebo‐controlled, crossover laboratory school study in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(11):1422‐9. CENTRAL

Ramtvedt 2013 {published data only}

Ramtvedt BE, Aabech HS, Sundet K. Minimizing adverse events while maintaining clinical improvement in a pediatric attention‐deficit/hyperactivity disorder crossover trial with dextroamphetamine and methylphenidate. Journal of Child and Adolescent Psychopharmacology 2014;24(3):130‐9. CENTRAL
Ramtvedt BE, Røinås E, Aabech HS, Sundet KS. Clinical gains from including both dextroamphetamine and methylphenidate in stimulant trials. Journal of Child and Adolescent Psychopharmacology 2013;23(9):597‐604. CENTRAL
Ramtvedt BE, Sandvik L, Sundet K. Correspondence between children's and adults' ratings of stimulant‐induced changes in ADHD behaviours in a crossover trial with medication‐naive children. European Journal of Developmental Psychology 2014;11(6):687‐700. CENTRAL
Ramtvedt BE, Sundet K. Relationships between computer‐based testing and behavioral ratings in the assessment of attention and activity in a pediatric ADHD stimulant crossover trial. Clinical Neuropsychologist 2014;28(7):1146‐61. CENTRAL

Rapport 1985 {published data only}

Rapport MD, DuPaul GJ. Methylphenidate: rate‐dependent effects on hyperactivity. Psychopharmacology Bulletin 1986;22(1):223‐8. CENTRAL
Rapport MD, DuPaul GJ, Stoner G, Birmingham BK, Masse G. Attention deficit disorder with hyperactivity: differential effects of methylphenidate on impulsivity. Pediatrics 1985;76(6):938‐43. CENTRAL
Rapport MD, DuPaul GJ, Stoner G, Jones TJ. Comparing classroom and clinic measures of attention deficit disorder: differential, idiosyncratic, and dose‐response effects of methylphenidate. Journal of Consulting and Clinical Psychology 1986;54(3):334‐41. CENTRAL
Rapport MD, Stoner G, DuPaul GJ, Birmingham BK, Tucker S. Methylphenidate in hyperactive children: differential effects of dose on academic, learning, and social behavior. Journal of Abnormal Child Psychology 1985;13(2):227‐43. CENTRAL

Rapport 1987 {published data only}

Denney CB, Rapport MD. Predicting methylphenidate response in children with ADHD: theoretical, empirical, and conceptual models. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(4):393‐401. CENTRAL
DuPaul GJ, Rapport MD. Does methylphenidate normalize the classroom performance of children with attention deficit disorder?. Journal of the American Academy of Child and Adolescent Psychiatry 1993;32(1):190‐8. CENTRAL
DuPaul GJ, Rapport MD, Vyse SA. ADHD and methylphenidate responders: effects on behavior controlled by complex reinforcement schedules. International Clinical Psychopharmacology 1988;3(4):349‐61. CENTRAL
Kelly KL, Rapport MD, DuPaul GJ. Attention deficit disorder and methylphenidate: a multi‐step analysis of dose‐response effects on children's cardiovascular functioning. International Clinical Psychopharmacology 1988;3(2):167‐81. CENTRAL
Rapport MD, Denney C. Titrating methylphenidate in children with attention‐deficit/hyperactivity disorder: is body mass predictive of clinical response?. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(4):523‐30. CENTRAL
Rapport MD, Denney C, DuPaul GJ, Gardner MJ. Attention deficit disorder and methylphenidate: normalization rates, clinical effectiveness, and response prediction in 76 children. Journal of the American Academy of Child and Adolescent Psychiatry 1994;33(6):882‐93. CENTRAL
Rapport MD, DuPaul GJ, Kelly KL. Attention deficit hyperactivity disorder and methylphenidate: the relationship between gross body weight and drug response in children. Psychopharmacology Bulletin 1989;25(2):285‐90. CENTRAL
Rapport MD, Jones JT, DuPaul GJ, Kelly KL, Gardner MJ, Tucker SB, et al. Attention deficit disorder and methylphenidate: group and single‐subject analyses of dose effects on attention in clinic and classroom settings. Journal of Clinical Child Psychology 1987;16(4):329‐38. CENTRAL
Rapport MD, Quinn SO, DuPaul GJ, Quinn EP, Kelly KL. Attention deficit disorder with hyperactivity and methylphenidate: the effects of dose and mastery level on children's learning performance. Journal of Abnormal Child Psychology 1989;17(6):669‐89. CENTRAL
Rapport MD, Randall R, Moffitt C. Attention‐deficit/hyperactivity disorder and methylphenidate: a dose‐response analysis and parent‐child comparison of somatic complaints. Journal of Attention Disorders 2002;6(1):15‐24. CENTRAL
Rapport MD, Stoner G, DuPaul GJ, Kelly KL, Tucker SB, Schoeler T. Attention deficit disorder and methylphenidate: a multilevel analysis of dose‐response effects on children's impulsivity across settings. Journal of the American Academy of Child and Adolescent Psychiatry 1988;27(1):60‐9. CENTRAL
Vyse SA, Rapport MD. The effects of methylphenidate on learning in children with ADDH: the stimulus equivalence paradigm. Journal of Consulting and Clinical Psychology 1989;57(3):425‐35. CENTRAL

Rapport 2008 {published data only}

Rapport MD, Kofler MJ, Coiro MM, Raiker JS, Sarver DE, Alderson RM. Unexpected effects of methylphenidate in attention‐deficit/hyperactivity disorder reflect decreases in core/secondary symptoms and physical complaints common to all children. Journal of Child and Adolescent Psychopharmacology 2008;18(3):237‐47. CENTRAL

Riggs 2011 {published data only}

Riggs PD, Winhusen T, Davies RD, Leimberger JD, Mikulich‐Gilbertson S, Klein C, et al. Randomized controlled trial of osmotic‐release methylphenidate with cognitive‐behavioral therapy in adolescents with attention‐deficit/hyperactivity disorder and substance use disorders. Journal of the American Academy of Child and Adolescent Psychiatry 2011;50(9):903‐14. CENTRAL
Tamm L, Adinoff B, Nakonezny PA, Winhusen T, Riggs P. Attention‐deficit/hyperactivity disorder subtypes in adolescents with comorbid substance‐use disorder. American Journal of Drug and Alcohol Abuse 2012;38(1):93‐100. CENTRAL
Tamm L, Trello‐Rishel K, Riggs P, Nakonezny PA, Acosta M, Bailey G, et al. Predictors of treatment response in adolescents with comorbid substance use disorder and attention‐deficit/hyperactivity disorder. Journal of Substance Abuse Treatment 2013;44(2):224‐30. CENTRAL
Warden D, Riggs PD, Min SJ, Mikulich‐Gilbertson SK, Tamm L, Trello‐Rishel K, et al. Major depression and treatment response in adolescents with ADHD and substance use disorder. Drug and Alcohol Dependence 2012;120(1‐3):214‐9. CENTRAL
Winhusen TM, Lewis DF, Riggs PD, Davies RD, Adler LA, Sonne S, et al. Subjective effects, misuse, and adverse effects of osmotic‐release methylphenidate treatment in adolescent substance abusers with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2011;21(5):455‐63. CENTRAL

Rubinsten 2008 {published data only}

Rubinsten O, Bedard A‐C, Tannock R. Methylphenidate has differential effects on numerical abilities in ADHD children with and without co‐morbid mathematical difficulties. Open Psychology Journal 2008;1:11‐7. CENTRAL

Samuels 2006 {published data only}

Samuels JA, Franco K, Wan F, Sorof JM. Effect of stimulants on 24‐h ambulatory blood pressure in children with ADHD: a double‐blind, randomized, cross‐over trial. Pediatric Nephrology 2006;21(1):92‐5. CENTRAL

Schachar 1997a {published data only}

Charach A, Figueroa M, Chen S, Ickowicz A, Schachar R. Stimulant treatment over 5 years: effects on growth. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(4):415‐21. CENTRAL
Charach A, Ickowicz A, Schachar R. Stimulant treatment over five years: adherence, effectiveness, and adverse effects. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(5):559‐67. CENTRAL
Diamond IR, Tannock R, Schachar RJ. Response to methylphenidate in children with ADHD and comorbid anxiety. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(4):402‐9. CENTRAL
Law SF, Schachar RJ. Do typical clinical doses of methylphenidate cause tics in children treated for attention‐deficit hyperactivity disorder?. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(8):944‐51. CENTRAL
Schachar RJ, Tannock R, Cunningham C, Corkum PV. Behavioral, situational, and temporal effects of treatment of ADHD with methylphenidate. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(6):754‐63. CENTRAL

Schachar 2008 {published data only}

Schachar R, Ickowicz A, Crosbie J, Donnelly GAE, Reiz JL, Miceli PC, et al. Cognitive and behavioral effects of multilayer‐release methylphenidate in the treatment of children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2008;18(1):11‐24. CENTRAL

Schulz 2010 {published data only}

Schulz E, Fleischhaker C, Hennighausen K, Heiser P, Oehler K‐U, Linder M, et al. A double‐blind, randomized, placebo/active controlled crossover evaluation of the efficacy and safety of Ritalin® LA in children with attention‐deficit/hyperactivity disorder in a laboratory classroom setting. Journal of Child and Adolescent Psychopharmacology 2010;20(5):377‐85. CENTRAL

Schwartz 2004 {published data only}

Gruber R, Grizenko N, Schwartz G, Ben Amor L, Gauthier J, De Guzman R, et al. Sleep and COMT polymorphism in ADHD children: preliminary actigraphic data. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(8):982‐9. CENTRAL
Schwartz G, Amor LB, Grizenko N, Lageix P, Baron C, Boivin DB, et al. Actigraphic monitoring during sleep of children with ADHD on methylphenidate and placebo. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(10):1276‐82. CENTRAL

Sharp 1999 {published data only}

Borcherding BG, Keysor CS, Cooper TB, Rapoport JL. Differential effects of methylphenidate and dextroamphetamine on the motor activity level of hyperactive children. Neuropsychopharmacology 1989;2(4):255‐63. CENTRAL
Castellanos FX, Elia J, Kruesi MJ, Marsh WL, Gulotta CS, Potter WZ, et al. Cerebrospinal fluid homovanillic acid predicts behavioral response to stimulants in 45 boys with attention deficit/hyperactivity disorder. Neuropsychopharmacology 1996;14(2):125‐37. CENTRAL
Elia J, Borcherding BG, Rapoport JL, Keysor CS. Methylphenidate and dextroamphetamine treatments of hyperactivity: are there true nonresponders?. Psychiatry Research 1991;36(2):141‐55. CENTRAL
Elia J, Welsh PA, Gullotta CS, Rapoport JL. Classroom academic performance: improvement with both methylphenidate and dextroamphetamine in ADHD boys. Journal of Child Psychology and Psychiatry, and Allied Disciplines 1993;34(5):785‐804. CENTRAL
Schmidt ME, Kruesi MJ, Elia J, Borcherding BG, Elin RJ, Hosseini JM, et al. Effect of dextroamphetamine and methylphenidate on calcium and magnesium concentration in hyperactive boys. Psychiatry Research 1994;54(2):199‐210. CENTRAL
Sharp WS, Walter JM, Marsh WL, Ritchie GF, Hamburger SD, Castellanos FX. ADHD in girls: clinical comparability of a research sample. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(1):40‐7. CENTRAL

Shiels 2009 {published data only}

Shiels K, Hawk LW, Reynolds B, Mazzullo RJ, Rhodes JD, Pelham WE, et al. Effects of methylphenidate on discounting of delayed rewards in attention deficit/hyperactivity disorder. Experimental and Clinical Psychopharmacology 2009;17(5):291‐301. CENTRAL
Spencer SV, Hawk LW, Richards JB, Shiels K, Pelham WE, Waxmonsky JG. Stimulant treatment reduces lapses in attention among children with ADHD: the effects of methylphenidate on intra‐individual response time distributions. Journal of Abnormal Child Psychology 2009;37(6):805‐16. CENTRAL

Silva 2005a {published data only}

Silva R, Muniz R, Pestreich LK, Brams M, Childress A, Lopez FA. Efficacy of two long‐acting methylphenidate formulations in children with attention‐deficit/hyperactivity disorder in a laboratory classroom setting. Journal of Child and Adolescent Psychopharmacology 2005;15(4):637‐54. CENTRAL

Silva 2006 {published data only}

Silva RR, Muniz R, Pestreich L, Childress A, Brams M, Lopez FA, et al. Efficacy and duration of effect of extended‐release dexmethylphenidate versus placebo in schoolchildren with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2006;16(3):239‐51. CENTRAL

Silva 2008 {published data only}

Silva RR, Muniz R, Pestreich L, Brams M, Mao AR, Childress A, et al. Dexmethylphenidate extended‐release capsules in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2008;47(2):199‐208. CENTRAL
Silva RR, Muniz R, Pestreich L, Lopez FA, Childress A, Wang J. Once‐daily dexmethylphenidate: a placebo‐controlled crossover study in children with attention‐deficit/hyperactivity disorder. Annals of Neurology 2005;58(Suppl S9):S109. CENTRAL

Smith 1998 {published data only}

Evans SW, Pelham WE, Smith BH, Bukstein O, Gnagy EM, Greiner AR, et al. Dose‐response effects of methylphenidate on ecologically valid measures of academic performance and classroom behavior in adolescents with ADHD. Experimental and Clinical Psychopharmacology 2001;9(2):163‐75. CENTRAL
Smith BH. Reliability, validity and unique contributions of self‐reports by adolescents being treated for attention‐deficit hyperactivity disorder. Dissertation Abstracts International: Section B: The Sciences and Engineering. US: U Arizona, 1997; Vol. 58, issue 6‐B:3328. CENTRAL
Smith BH, Pelham WE, Evans S, Gnagy E, Molina B, Bukstein O, et al. Dosage effects of methylphenidate on the social behavior of adolescents diagnosed with attention‐deficit hyperactivity disorder. Experimental and Clinical Psychopharmacology 1998;6(2):187‐204. CENTRAL
Smith BH, Pelham WE, Gnagy E, Yudell RS. Equivalent effects of stimulant treatment for attention‐deficit hyperactivity disorder during childhood and adolescence. Journal of the American Academy of Child and Adolescent Psychiatry 1998;37(3):314‐21. CENTRAL

Smith 2004 {published data only}

Smith R, Larsen D, Derby KM, McLaughlin TF, Weber KP, Brown K, et al. A comparison of teacher checklists used over 15 days and a one‐day antecedent analysis to conduct a medication trial. Psychology in the Schools 2004;41(2):235‐40. CENTRAL

Smithee 1998 {published data only}

Smithee JA, Klorman R, Brumaghim JT, Borgstedt AD. Methylphenidate does not modify the impact of response frequency or stimulus sequence on performance and event‐related potentials of children with attention deficit hyperactivity disorder. Journal of Abnormal Child Psychology 1998;26(4):233‐45. CENTRAL

Solanto 2009 {published data only}

Solanto M, Newcorn J, Vail L, Gilbert S, Ivanov I, Lara R. Stimulant drug response in the predominantly inattentive and combined subtypes of attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2009;19(6):663‐71. CENTRAL

Stein 1996 {published data only}

Stein MA, Blondis TA, Schnitzler ER, O'Brien T, Fishkin J, Blackwell B, et al. Methylphenidate dosing: twice daily versus three times daily. Pediatrics 1996;98(4 Pt 1):748‐56. CENTRAL

Stein 2003 {published data only}

Stein MA, Sarampote C, Seymour K. Insomnia and tiredness in ADHD youth: relationship with methylphenidate dose, age, and weight. Pediatric Research. Annual Meeting of the Pediatric Academic Societies; 2004 May 4; San Francisco, CA. Baltimore: International Pediatric Research Foundation, 2004; Vol. 55 (4):74A. [WOS: 000220591100437]CENTRAL
Stein MA, Sarampote CS, Waldman ID, Robb AS, Conlon C, Pearl PL, et al. A dose‐response study of OROS methylphenidate in children with attention‐deficit/hyperactivity disorder. Pediatrics 2003;112(5):e404. CENTRAL
Stein MA, Seymour KE, Black DO, Sarampote CS, Robb A, Conlon C, et al. Effects and side effects of Concerta methylphenidate (MPH) in children with ADHD and comorbid internalizing symptoms. Pediatric Research. Annual Meeting of the Pediatric Academic Society; 2003 May 3‐6; Seattle, Washington. Baltimore: International Pediatric Research Foundation, 2003; Vol. 53 (4):555A. [WOS: 000181897903139]CENTRAL
Stein MA, Waldman ID, Sarampote CS, Seymour KE, Robb AS, Conlon C, et al. Dopamine transporter genotype and methylphenidate dose response in children with ADHD. Neuropsychopharmacology 2005;30(7):1374‐82. CENTRAL

Stein 2011 {published data only}

Stein MA, Waldman ID, Charney E, Aryal S, Sable C, Gruber R, et al. Dose effects and comparative effectiveness of extended release dexmethylphenidate and mixed amphetamine salts. Journal of Child and Adolescent Psychopharmacology2011; Vol. 21, issue 6:581‐8. CENTRAL
Wiebe S, Gruber R, Charney E, Aryal S, Waldman I, Newcorn J, et al. Sleep and emotional reactivity to extended release dexmethylphenidate versus mixed amphetamine salts: a double‐blind, placebo controlled study. European Child & Adolescent Psychiatry. Proceedings of the Eunethydis 1st International ADHD Conference: From Data to Best Clinical Practice; 2010 May 26‐28; Amsterdam, Netherlands 2010;19(Suppl 1):S82. CENTRAL

Stoner 1994 {published data only}

Stoner G, Carey SP, Ikeda MJ, Shinn MR. The utility of curriculum‐based measurement for evaluating the effects of methylphenidate on academic performance. Journal of Applied Behavior Analysis 1994;27(1):101‐13. CENTRAL

Sumner 2010 {published data only}

Sumner CR, Haynes VS, Teicher MH, Newcorn JH. Does placebo response differ between objective and subjective measures in children with attention‐deficit/hyperactivity disorder?. Postgraduate Medicine 2010;122(5):52‐61. CENTRAL

Sunohara 1999 {published data only}

Sunohara GA. Methylphenidate effects on focused and selective attention processing in children with ADHD. Dissertation Abstracts International: Section B: The Sciences and Engineering1998; Vol. 59, issue 6‐B:3113. CENTRAL
Sunohara GA, Malone MA, Rovet J, Humphries T, Roberts W, Taylor MJ. Effect of methylphenidate on attention in children with attention deficit hyperactivity disorder (ADHD): ERP evidence. Neuropsychopharmacology 1999;21(2):218‐28. CENTRAL

Swanson 1998 {published data only}

Swanson J, Wigal S, Greenhill L, Browne R, Waslick B, Lerner M, et al. Objective and subjective measures of the pharmacodynamic effects of Adderall in the treatment of children with ADHD in a controlled laboratory classroom setting. Psychopharmacology Bulletin 1998;34(1):55‐60. CENTRAL
Swanson JM, Wigal S, Greenhill LL, Browne R, Waslik B, Lerner M, et al. Analog classroom assessment of Adderall® in children with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 1998;37(5):519‐26. CENTRAL

Swanson 1999 {published data only}

Swanson J, Gupta S, Guinta D, Flynn D, Agler D, Lerner M, et al. Acute tolerance to methylphenidate in the treatment of attention deficit hyperactivity disorder in children. Clinical Pharmacology and Therapeutics 1999;66(3):295‐305. CENTRAL
Wigal SB, Gupta S, Guinta D, Swanson JM. Reliability and validity of the SKAMP rating scale in a laboratory school setting. Psychopharmacology Bulletin 1998;34(1):47‐53. CENTRAL

Swanson 2002a {published data only}

Swanson J, Gupta S, Lam A, Shoulson I, Lerner M, Modi N, et al. Development of a new once‐a‐day formulation of methylphenidate for the treatment of attention‐deficit/hyperactivity disorder: proof‐of‐concept and proof‐of‐product studies. Archives of General Psychiatry 2003;60(2):204‐11. CENTRAL
Swanson J, Sadeh A, Lerner MA, Wigal SB. Comparison of the impact of OROS methylphenidate HCI with methylphenidate tid and placebo on the sleep of children with ADHD. Journal of Developmental and Behavioral Pediatrics 2000;21(5):387‐8. CENTRAL
Swanson JM, Gupta S, Williams L, Agler D, Lerner M, Wigal S. Efficacy of a new pattern of delivery of methylphenidate for the treatment of ADHD: effects on activity level in the classroom and on the playground. Journal of the American Academy of Child and Adolescent Psychiatry 2002;41(11):1306‐14. CENTRAL
Swanson JM, Wigal SB, Lemer MA. Comparison of the efficacy and safety of OROS methylphenidate HCI with methylphenidate tid and placebo in children with ADHD. Pediatric Research 2000;47(4):34A. [WOS: 000086155300200]CENTRAL
Wigal S, Lerner M, Swanson J. Once‐daily methylphenidate formulation: impact on academic productivity and activity levels of children with attention‐deficit/hyperactivity disorder. European Neuropsychopharmacology 2002;12(Suppl 3):416. CENTRAL
Wigal S, Swanson JM, Lerner M. Comparison of duration of effect of OROS MPH with MPH tid in ADHD children. Annual Meeting of the American Psychiatric Association; 2001 May 5‐10. New Orleans: American Psychiatric Association, 2001. CENTRAL

Swanson 2002b {published data only}

Swanson J, Gupta S, Lam A, Shoulson I, Lerner M, Modi N, et al. Development of a new once‐a‐day formulation of methylphenidate for the treatment of attention‐deficit/hyperactivity disorder: proof‐of‐concept and proof‐of‐product studies. Archives of General Psychiatry 2003;60(2):204‐11. CENTRAL
Swanson J, Sadeh A, Lerner MA, Wigal SB. Comparison of the impact of OROS methylphenidate HCI with methylphenidate tid and placebo on the sleep of children with ADHD. Journal of Developmental and Behavioral Pediatrics 2000;21(5):387‐8. [WOS: 000089857900028]CENTRAL
Swanson J, Wigal S, Lerner M. Treatment with a controlled‐release formulation of methylphenidate for attention‐deficit/hyperactivity disorder: onset and duration of effect. European Neuropsychopharmacology 2002;12(Suppl 3):414. CENTRAL
Swanson JM, Gupta S, Williams L, Agler D, Lerner M, Wigal S. Efficacy of a new pattern of delivery of methylphenidate for the treatment of ADHD: effects on activity level in the classroom and on the playground. Journal of the American Academy of Child and Adolescent Psychiatry 2002;41(11):1306‐14. CENTRAL
Swanson JM, Wigal SB, Lemer MA. Comparison of the efficacy and safety of OROS methylphenidate HCI with methylphenidate tid and placebo in children with ADHD. Pediatric Research 2000;47(Suppl):34A. CENTRAL
Wigal S, Lerner M, Swanson J. Once‐daily methylphenidate formulation: impact on academic productivity and activity levels of children with attention‐deficit/hyperactivity disorder. European Neuropsychopharmacology 2002;12(Suppl 3):416. CENTRAL

Swanson 2004b {published data only}

Sonuga‐Barke EJ, Coghill D, Markowitz JS, Swanson JM, Vandenberghe M, Hatch SJ. Sex differences in the response of children with ADHD to once‐daily formulations of methylphenidate. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(6):701‐10. CENTRAL
Sonuga‐Barke EJ, Van Lier P, Swanson JM, Coghill D, Wigal S, Vandenberghe M, et al. Heterogeneity in the pharmacodynamics of two long‐acting methylphenidate formulations for children with attention deficit/hyperactivity disorder. A growth mixture modelling analysis. European Child & Adolescent Psychiatry 2008;17(4):245‐54. CENTRAL
Sonuga‐Barke EJS, Coghill D, Wigal T, DeBacker M, Swanson J. Adverse reactions to methylphenidate treatment for attention‐deficit/hyperactivity disorder: structure and associations with clinical characteristics and symptom control. Journal of Child and Adolescent Psychopharmacology 2009;19(6):683‐90. CENTRAL
Sonuga‐Barke EJS, Swanson JM, Coghill D, DeCory HH, Hatch SJ. Efficacy of two once‐daily methylphenidate formulations compared across dose levels at different times of the day: preliminary indications from a secondary analysis of the COMACS study data. BMC Psychiatry 2004;4:28. CENTRAL
Sonuge‐Barke EJS, Swanson J, Hatch S, Van Lier P, Vandenberghe M. Heterogeneity in ADHD children's response to two long‐acting methylphenidate formulations. Journal of Neural Transmission. Abstracts of the 39th International Danube Symposium for Neurological Sciences and Continuing Education and 1st International Congress on ADHD, from Childhood to Adult Disease 2007;114(7):LXXXIX. CENTRAL
Swanson JM, Wigal SB, Wigal T, Sonuga‐Barke E, Greenhill LL, Biederman J, et al. A comparison of once‐daily extended‐release methylphenidate formulations in children with attention‐deficit/hyperactivity disorder in the laboratory school (the Comacs Study). Pediatrics 2004;113(3 Pt 1):e206‐16. CENTRAL

Symons 2007 {published data only}

Symons FJ, Tervo RC, Kim O, Hoch J. The effects of methylphenidate on the classroom behavior of elementary school‐age children with cerebral palsy: a preliminary observational analysis. Journal of Child Neurology 2007;22(1):89‐94. CENTRAL

Szobot 2004 {published data only}

Szobot CM, Ketzer C, Parente MA, Biederman J, Rohde LA. The acute effect of methylphenidate in Brazilian male children and adolescents with ADHD: a randomized clinical trial. Journal of Attention Disorders 2004;8(2):37‐43. CENTRAL

Szobot 2008 {published data only}

Szobot CM, Rohde LA, Katz B, Ruaro P, Schaefer T, Walcher M, et al. A randomized crossover clinical study showing that methylphenidate‐SODAS improves attention‐deficit/hyperactivity disorder symptoms in adolescents with substance use disorder. Brazilian Journal of Medical and Biological Research 2008;41(3):250‐7. CENTRAL

Tannock 1989 {published data only}

Tannock R, Schachar RJ, Carr RP, Chajczyk D, Logan GD. Effects of methylphenidate on inhibitory control in hyperactive children. Journal of Abnormal Child Psychology 1989;17(5):473‐91. Erratum in: Journal of Abnormal Child Psychology 1990;18(1):119. CENTRAL
Tannock R, Schachar RJ, Carr RP, Logan GD. Dose‐response effects of methylphenidate on academic performance and overt behavior in hyperactive children. Pediatrics 1989;84(4):648‐57. CENTRAL

Tannock 1992 {published data only}

Tannock R, Schachar R. Methylphenidate and cognitive perseveration in hyperactive children. Journal of Child Psychology and Psychiatry and Allied Disciplines 1992;33(7):1217‐28. CENTRAL

Tannock 1993 {published data only}

Tannock R, Schachar RJ, Logan GD. Does methylphenidate induce overfocusing in hyperactive children?. Journal of Clinical Child Psychology 1993;22(1):28‐41. CENTRAL

Tannock 1995a {published data only}

Tannock R, Schachar R, Logan G. Methylphenidate and cognitive flexibility: dissociated dose effects in hyperactive children. Journal of Abnormal Child Psychology 1995;23(2):235‐66. CENTRAL

Tannock 1995b {published data only}

Tannock R, Fine J, Heintz T, Schachar RJ. A linguistic approach detects stimulant effects in two children with attention‐deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1995;5(3):177‐89. CENTRAL
Tannock R, Ickowicz A, Schachar R. Differential effects of methylphenidate on working memory in ADHD children with and without comorbid anxiety. Journal of the American Academy of Child and Adolescent Psychiatry 1995;34(7):886‐96. CENTRAL

Taylor 1987 {published data only}

Taylor E, Schachar R, Thorley G, Wieselberg HM, Everitt B, Rutter M. Which boys respond to stimulant medication? A controlled trial of methylphenidate in boys with disruptive behaviour. Psychological Medicine 1987;17(1):121‐43. CENTRAL

Taylor 1993 {published data only}

Taylor MJ, Voros JG, Logan WJ, Malone MA. Changes in event‐related potentials with stimulant medication in children with attention deficit hyperactivity disorder. Biological Psychology 1993;36(3):139‐56. CENTRAL

Tervo 2002 {published data only}

Tervo RC, Azuma S, Fogas B, Fiechtner H. Children with ADHD and motor dysfunction compared with children with ADHD only. Developmental Medicine & Child Neurology 2002;44(6):383‐90. CENTRAL

Tirosh 1993a {published data only}

Tirosh E, Elhasid R, Kamah SC, Cohen A. Predictive value of placebo methylphenidate. Pediatric Neurology 1993;9(2):131‐3. CENTRAL

Tirosh 1993b {published data only}

Tirosh E, Sadeh A, Munvez R, Lavie P. Effects of methylphenidate on sleep in children with attention‐deficient hyperactivity disorder. An activity monitor study. American Journal of Diseases of Children 1993;147(12):1313‐5. CENTRAL

Tourette's Syndrome Study Group 2002 {published data only}

Deputy SR. Treatment of ADHD in children with tics: a randomized controlled trial. Clinical Pediatrics 2002;41(9):736. CENTRAL
Kurlan R, Goldberg J. Clonidine and methylphenidate were effective for attention deficit hyperactivity disorder in children with comorbid tics. Evidence‐Based Medicine 2002;7(5):157. CENTRAL
Kurlan R, Tourette Syndrome Study Group. Treatment of attention‐deficit‐hyperactivity disorder in children with Tourette's syndrome (TACT Trial). Annals of Neurology. Abstracts from the American Neurological Association's 125th Annual Meeting; 2000 October 15‐18; Boston, Massachusetts 2000;48(6):953. CENTRAL
Tourette's Syndrome Study Group. Treatment of ADHD in children with tics: a randomized controlled trial. Neurology 2002;58(4):527‐36. CENTRAL

Tucker 2009 {published data only}

Tucker JD, Suter W, Petibone DM, Thomas RA, Bailey NL, Zhou Y, et al. Cytogenetic assessment of methylphenidate treatment in pediatric patients treated for attention deficit hyperactivity disorder. Mutation Research 2009;677(1‐2):53‐8. CENTRAL
Zhou Y, Muni R, Tucke JD, Kumar V. Extended‐release methylphenidate exposure and the frequency of cytogenetic abnormalities in children with attention‐deficit‐hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology. Proceedings of the 49th Annual National Institute of Mental Health (NIMH) New Clinical Drug Evaluation Unit (NCDEU) Meeting; 2009 June 29‐ July 2; Hollywood, Florida 2009;19(6):785. CENTRAL

Ullmann 1985 {published data only}

Ullmann RK, Sleator EK. Attention deficit disorder children with or without hyperactivity: which behaviors are helped by stimulants?. Clinical Pediatrics 1985;24(10):547‐51. [DOI: 10.1177/000992288502401001]CENTRAL

Ullmann 1986 {published data only}

Ullmann RK, Sleator EK. Responders, nonresponders, and placebo responders among children with attention deficit disorder: importance of a blinded placebo evaluation. Clinical Pediatrics 1986;25(12):594‐9. CENTRAL

Urman 1995 {published data only}

Urman R, Ickowicz A, Fulford P, Tannock R. An exaggerated cardiovascular response to methylphenidate in ADHD children with anxiety. Journal of Child and Adolescent Psychopharmacology 1995;5(1):29‐37. CENTRAL

Van der Meere 1999a {published data only}

Gunning WB. A Controlled Trial of Clonidine in Hyperkinetic Children. Erasmus Universiteit Rotterdam, 1992. CENTRAL
Van der Meere J, Gunning B, Stemerdink N. The effect of methylphenidate and clonidine on response inhibition and state regulation in children with ADHD. Journal of Child Psychology and Psychiatry and Allied Disciplines 1999;40(2):291‐8. CENTRAL

Wallace 1994 {published data only}

Wallace AE, Kofoed LL. Statistical analysis of single case studies in the clinical setting: the example of methylphenidate trials in children with attention‐deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1994;4(3):141‐50. CENTRAL

Wallander 1987 {published data only}

Wallander JL, Schroeder SR, Michelli JA, Gualtieri CT. Classroom social interactions of attention deficit disorder with hyperactivity children as a function of stimulant medication. Journal of Pediatric Psychology 1987;12(1):61‐76. CENTRAL

Waxmonsky 2008 {published data only}

Waxmonsky J, Pelham WE, Gnagy E, Cummings MR, O'Connor B, Majumdar A, et al. The efficacy and tolerability of methylphenidate and behavior modification in children with attention‐deficit/hyperactivity disorder and severe mood dysregulation. Journal of Child and Adolescent Psychopharmacology 2008;18(6):573‐88. CENTRAL

Whalen 1990 {published data only}

Whalen CK, Henker B, Granger DA. Ratings of medication effects in hyperactive children: viable or vulnerable?. Behavioral Assessment 1989;11(2):179‐99. CENTRAL
Whalen CK, Henker B, Granger DA. Social judgment processes in hyperactive boys: effects of methylphenidate and comparisons with normal peers. Journal of Abnormal Child Psychology 1990;18(3):297‐316. CENTRAL
Whalen CK, Henker B, Hinshaw SP, Granger DA. Externalizing behavior disorders, situational generality, and the type A behavior pattern. Child Development 1989;60(6):1453‐62. CENTRAL

Wigal 2003 {published data only}

Wigal SB, Sanchez DY, DeCory HH, D'Imperio JM, Swanson JM. Selection of the optimal dose ratio for a controlled‐delivery formulation of methylphenidate. Journal of Applied Research 2003;3(1):46‐63. CENTRAL

Wigal 2004 {published data only}

Wigal S, Swanson JM, Feifel D, Sangal RB, Elia J, Casat CD, et al. A double‐blind, placebo‐controlled trial of dexmethylphenidate hydrochloride and d,l‐threo‐methylphenidate hydrochloride in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(11):1406‐14. CENTRAL

Wigal 2013 {published data only}

Liquid version of methylphenidate shows efficacy in school trial. The Brown University Child & Adolescent Psychopharmacology Update2013; Vol. 15, issue 3:1‐3. CENTRAL
Wigal SB, Childress AC, Belden HW, Berry SA. NWP06, an extended‐release oral suspension of methylphenidate, improved attention‐deficit/hyperactivity disorder symptoms compared with placebo in a laboratory classroom study. Journal of Child and Adolescent Psychopharmacology 2013;23(1):3‐10. CENTRAL

Wigal 2014 {published data only}

Wigal SB, Greenhill LL, Nordbrock E, Connor DF, Kollins SH, Adjei A, et al. A randomized placebo‐controlled double‐blind study evaluating the time course of response to methylphenidate hydrochloride extended‐release capsules in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2014;24(10):562‐9. CENTRAL

Wilens 2006b {published data only}

Oral system methylphenidate for teen ADHD. The Brown University Child & Adolescent Psychopharmacology Update2006; Vol. 8, issue 3:4‐5. CENTRAL
Biederman J. P.6.053 Effectiveness and safety of the once‐daily OROS formulation of methylphenidate in adolescents with attention‐deficit/hyperactivity disorder. European Neuropsychopharmacology 2003;13(Suppl 4):S448. CENTRAL
Greenhill LL. Safety and efficacy of OROS MPH in adolescents with ADHD. Proceedings of the 156th Annual Meeting of the American Psychiatric Association; 2003 May 17‐22; San Francisco, CA. San Francisco, 2003:16. CENTRAL
McGough JJ, McBurnett K, Bukstein O, Wilens TE, Greenhill L, Lerner M, et al. Once‐daily OROS methylphenidate is safe and well tolerated in adolescents with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2006;16(3):351‐6. CENTRAL
Newcorn JH, Stein MA, Cooper KM. Dose‐response characteristics in adolescents with attention‐deficit/hyperactivity disorder treated with OROS methylphenidate in a 4‐week, open‐label, dose‐titration study. Journal of Child and Adolescent Psychopharmacology 2010;20(3):187‐96. CENTRAL
Wilens TE, McBurnett K, Bukstein O, McGough J, Greenhill L, Lerner M, et al. Multisite controlled study of OROS methylphenidate in the treatment of adolescents with attention‐deficit/hyperactivity disorder. Archives of Pediatrics & Adolescent Medicine 2006;160(1):82‐90. CENTRAL

Wilens 2008 {published data only}

López FA, Landgraf JM, Wilens TE. Quality of life and parent satisfaction with the methylphenidate transdermal system. European Neuropsychopharmacology. Papers of the 21st ECNP Congress; 2008 August 30 ‐ September 3; Barcelona, Spain. 2008; Vol. Suppl 4:S562. CENTRAL
López FA, Wilens TE, Wigal SB, Turnbow JM. Effects of variable wear times on transdermal methylphenidate in attention‐deficit/hyperactivity disorder. European Neuropsychopharmacology. Papers of the 21st ECNP Congress; 2008 August 30 ‐ September 3; Barcelona, Spain. 2008; Vol. 18:S561‐2. CENTRAL
Manos M, Frazier TW, Landgraf JM, Weiss M, Hodgkins P. HRQL and medication satisfaction in children with ADHD treated with the methylphenidate transdermal system. Current Medical Research and Opinion 2009;25(12):3001‐10. CENTRAL
Wilens TE, Boellner SW, López FA, Turnbow JM, Wigal SB, Childress AC, et al. Varying the wear time of the methylphenidate transdermal system in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2008;47(6):700‐8. CENTRAL

Wilens 2010 {published data only}

Wilens T, Hammerness P, Utzinger L, Georgiopoulos A, Doyle R, Brodziak K, et al. Before‐school ADHD symptoms and functioning in youth treated with the Methylphenidate Transdermal Patch (MTS). Journal of Child and Adolescent Psychopharmacology. Abstracts of the 49th Annual National Institute of Mental Health (NIMH) New Clinical Drug Evaluation Unit (NCDEU) Meeting; 2009; June 29 ‐ July 2; Hollywood, Florida. 2009; Vol. 19, issue 6:785‐6. CENTRAL
Wilens TE, Hammerness P, Martelon MK, Brodziak K, Utzinger L, Wong P. A controlled trial of the methylphenidate transdermal system on before‐school functioning in children with attention‐deficit/hyperactivity disorder. Journal of Clinical Psychiatry 2010;71(5):548‐56. CENTRAL

Wilkison 1995 {published data only}

Wilkison PC, Kircher JC, McMahon WM, Sloane HN. Effects of methylphenidate on reward strength in boys with attention‐deficit hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1995;34(7):897‐901. CENTRAL

Wodrich 1998 {published data only}

Wodrich DL, Kush JC. The effect of methylphenidate on teachers' behavioral ratings in specific school situations. Psychology in the Schools 1998;35(1):81‐8. CENTRAL

Wolraich 2001 {published data only}

Greenhill LL. Efficacy and safety of once‐daily methylphenidate HCl, standard methylphenidate and placebo in children with ADHD. Proceedings of the 153rd Annual Meeting of the American Psychiatric Association; 2000 May 13‐18; Chicago, Illinois. Chicago, 2000:NR. 667. CENTRAL
Greenhill LL. Evaluation of the efficacy and safety of Concerta (Methylphenidate HCI) extended‐release tablets, Ritalin, and placebo in children with ADHD. Neurology 2000;54(7):A420‐1. [WOS: 000086557801120]CENTRAL
Swanson J, Greenhill L, Pelham W, Wilens T, Wolraich M, Abikoff H, et al. Initiating Concerta (TM) (OROS methylphenidate HCl) qd in children with attention‐deficit hyperactivity disorder. Journal of Clinical Research 2000;3:59‐76. CENTRAL
Wolraich ML. [Efficacy and safety of OROS(R) methylphenidate HCl (MPH) extended‐release tablets (CONCERTA (TM)), conventional MPH, and placebo in children with ADHD]. International Journal of Neuropsychopharmacology. Proceedings of the XXIIst CINP Congress; 2000 July 9‐13; Brussels, Belgium. 2000; Vol. 3 Suppl 1:S329. CENTRAL
Wolraich ML. Efficacy and safety of OROS methylphenidate HCI (MPH) extended‐release tablets (Concerta TM), conventional MPH, and placebo in children with ADHD. International Journal of Neuropsychopharmacology. XXIIst CINP Congress 2000;3(Suppl S1):329. CENTRAL
Wolraich ML. Evaluation of efficacy and safety of OROS methylphenidate HCI (MPH) extended‐release tablets, methylphenidate tid, and placebo in children with ADHD. Pediatric Research 2000;47(4):36A. [WOS: 000086155300208]CENTRAL
Wolraich ML, Greenhill LL, Pelham W, Swanson J, Wilens T, Palumbo D, et al. Randomized, controlled trial of OROS methylphenidate once a day in children with attention‐deficit/hyperactivity disorder. Pediatrics 2001;108(4):883‐92. CENTRAL

Zeiner 1999 {published data only}

Zeiner P. Body growth and cardiovascular function after extended treatment (1.75 years) with methylphenidate in boys with attention‐deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1995;5(2):129‐38. CENTRAL
Zeiner P. Do the beneficial effects of extended methylphenidate treatment in boys with attention‐deficit hyperactivity disorder dissipate rapidly during placebo treatment?. Nordic Journal of Psychiatry 1999;53(1):55‐60. CENTRAL
Zeiner P, Bryhn G, Bjercke C, Truyen K, Strand G. Response to methylphenidate in boys with attention‐deficit hyperactivity disorder. Acta Paediatrica 1999;88(3):298‐303. CENTRAL

Zeni 2009 {published data only}

Zeni CP, Tramontina S, Ketzer CR, Pheula GF, Rohde LA. Methylphenidate combined with aripiprazole in children and adolescents with bipolar disorder and attention‐deficit/hyperactivity disorder: a randomized crossover trial. Journal of Child and Adolescent Psychopharmacology 2009;19(5):553‐61. CENTRAL

An 2013 {published data only}

An L, Cao X‐H, Cao Q‐J, Sun L, Yang L, Zou Q‐H, et al. Methylphenidate normalizes resting‐state brain dysfunction in boys with attention deficit hyperactivity disorder. Neuropsychopharmacology 2013;38(7):1287‐95. CENTRAL

Anderson 2002 {published data only}

Anderson CM, Polcari A, Lowen SB, Renshaw PF, Teicher MH. Effects of methylphenidate on functional magnetic resonance relaxometry of the cerebellar vermis in boys with ADHD. American Journal of Psychiatry 2002;159(8):1322‐8. CENTRAL

Barkley 1988a {published data only}

Barkley RA, Fischer M, Newby RF, Breen MJ. Development of a multimethod clinical protocol for assessing stimulant drug response in children with attention deficit disorder. Journal of Clinical Child Psychology 1988;17(1):14‐24. CENTRAL

Barkley 1997 {published data only}

Barkley RA, Koplowitz S, Anderson T, McMurray MB. Sense of time in children with ADHD: effects of duration, distraction, and stimulant medication. Journal of the International Neuropsychological Society 1997;3(4):359‐69. CENTRAL

Bart 2013 {published data only}

Bart O, Daniel L, Dan O, Bar‐Haim Y. Influence of methylphenidate on motor performance and attention in children with developmental coordination disorder and attention deficit hyperactive disorder. Research in Developmental Disabilities 2013;34(6):1922‐7. [PUBMED: 23584172]CENTRAL
Bart O, Podoly T, Bar‐Haim Y. A preliminary study on the effect of methylphenidate on motor performance in children with comorbid DCD and ADHD. Research in Developmental Disabilities 2010;31(6):1443‐7. [PUBMED: 20650602]CENTRAL

Bedard 2002 {published data only}

Bedard A‐C, Ickowicz A, Tannock R. Methylphenidate improves Stroop naming speed, but not response interference, in children with attention deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2002;12(4):301‐9. CENTRAL

Bedard 2003 {published data only}

Bedard A‐C, Ickowicz A, Logan GD, Hogg‐Johnson S, Schachar R, Tannock R. Selective inhibition in children with attention‐deficit hyperactivity disorder off and on stimulant medication. Journal of Abnormal Child Psychology 2003;31(3):315‐27. CENTRAL

Bedard 2004 {published data only}

Bedard A‐C, Martinussen R, Ickowicz A, Tannock R. Methylphenidate improves visual‐spatial memory in children with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(3):260‐8. CENTRAL

Bedard 2007 {published data only}

Bedard A‐C, Jain U, Hogg Johnson S, Tannock R. Effects of methylphenidate on working memory components: influence of measurement. Journal of Child Psychology and Psychiatry 2007;48(9):872‐80. CENTRAL

Beery 1994 {published data only}

Beery SH. Behavioral disinhibition, anxiety, and response to methylphenidate and behavior management in children with attention deficit hyperactivity disorder. Dissertation Abstracts International: Section B: The Sciences and Engineering 1994;56(2‐B):1096. CENTRAL

Ben‐Pazi 2006 {published data only}

Ben‐Pazi H, Shalev RS, Gross‐Tsur V, Bergman H. Age and medication effects on rhythmic responses in ADHD: possible oscillatory mechanisms?. Neuropsychologia 2006;44(3):412‐6. CENTRAL

Bental 2008 {published data only}

Bental B, Tirosh E. The effects of methylphenidate on word decoding accuracy in boys with attention‐deficit/hyperactivity disorder. Journal of Clinical Psychopharmacology 2008;28(1):89‐92. CENTRAL

Brown 1984b {published data only}

Brown RT, Wynne ME. Sustained attention in boys with attention deficit disorder and the effect of methylphenidate. Pediatric Nursing 1984;10(1):35‐9. CENTRAL

Buhrmester 1992 {published data only}

Buhrmester D, Whalen CK, Henker B, MacDonald V, Hinshaw SP. Prosocial behavior in hyperactive boys: effects of stimulant medication and comparison with normal boys. Journal of Abnormal Child Psychology 1992;20(1):103‐21. CENTRAL

Campbell 1996 {published data only}

Campbell L, Malone MA, Kershner JR, Roberts W, Humphries T, Logan WJ. Methylphenidate slows right hemisphere processing in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1996;6(4):229‐39. CENTRAL

Carlson 1991 {published data only}

Carlson CL, Pelham WE, Swanson JM, Wagner JL. A divided attention analysis of the effects of methylphenidate on the arithmetic performance of children with attention‐deficit hyperactivity disorder. Journal of Child Psychology and Psychiatry and Allied Disciplines 1991;32(3):463‐71. CENTRAL

Carlson 1992 {published data only}

Carlson CL, Pelham WE, Milich R, Dixon J. Single and combined effects of methylphenidate and behavior therapy on the classroom performance of children with attention‐deficit hyperactivity disorder. Journal of Abnormal Child Psychology 1992;20(2):213‐32. CENTRAL

Cox 2004b {published data only}

Cox DJ, Humphrey JW, Merkel RL, Penberthy JK, Kovatchev B. Controlled‐release methylphenidate improves attention during on‐road driving by adolescents with attention‐deficit/hyperactivity disorder. Journal of the American Board of Family Medicine 2004;17(4):235‐9. [DOI: 10.3122/jabfm.17.4.235]CENTRAL
Cox DJ, Merkel RL, Penberthy JK, Kovatchev B, Hankin CS. Impact of methylphenidate delivery profiles on driving performance of adolescents with attention‐deficit/hyperactivity disorder: a pilot study. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(3):269‐75. CENTRAL

Dawson 1998 {published data only}

Dawson NL. Effects of methylphenidate on neuropsychological functions myelomeningocele. Dissertation Abstracts International: Section B: The Sciences and Engineering. US: Illinois Inst of Technology, 1998, issue 12‐B:6805. CENTRAL

De Sonneville 1991 {published data only}

De Sonneville LM, Njiokiktjien C, Hilhorst RC. Methylphenidate‐induced changes in ADDH information processors. Journal of Child Psychology and Psychiatry and Allied Disciplines 1991;32(2):285‐95. CENTRAL

DeVito 2008 {published data only}

DeVito EE, Blackwell AD, Clark L, Kent L, Dezsery AM, Turner DC, et al. Methylphenidate improves response inhibition but not reflection‐impulsivity in children with attention deficit hyperactivity disorder (ADHD). Psychopharmacology 2009;202(1‐3):531‐9. [PUBMED: 18818905]CENTRAL
DeVito EE, Blackwell AD, Kent L, Ersche KD, Clark L, Salmond CH, et al. The effects of methylphenidate on decision making in attention‐deficit/hyperactivity disorder. Biological Psychiatry 2008;64(7):636‐9. [DOI: 10.1016/j.biopsych.2008.04.017]CENTRAL
DeVito EE, Sahakian BJ. Response to comments on 'Methylphenidate improves response inhibition but not reflection impulsivity in children with attention deficit hyperactivity disorder (ADHD)'. Psychopharmacology 2009;203(1):187. [DOI: 10.1007/s00213‐008‐1383‐5]CENTRAL

Evans 1986 {published data only}

Evans RW, Gualtieri CT, Amara I. Methylphenidate and memory: dissociated effects in hyperactive children. Psychopharmacology 1986;90(2):211‐6. CENTRAL

Fox 2014 {published data only}

Fox O, Adi‐Japha E, Karni A. The effect of a skipped dose (placebo) of methylphenidate on the learning and retention of a motor skill in adolescents with attention deficit hyperactivity disorder. European Neuropsychopharmacology 2014;24(3):391‐6. CENTRAL

Francis 2001 {published data only}

Francis S, Fine J, Tannock R. Methylphenidate selectively improves story retelling in children with attention deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2001;11(3):217‐28. CENTRAL

Gan 1982 {published data only}

Gan J, Cantwell DP. Dosage effects of methylphenidate on paired associate learning: positive/negative placebo responders. Journal of the American Academy of Child Psychiatry 1982;21(3):237‐42. CENTRAL

Granger 1996 {published data only}

Granger DA, Whalen CK, Henker B, Cantwell C. ADHD boys' behavior during structured classroom social activities: effects of social demands, teacher proximity, and methylphenidate. Journal of Attention Disorders 1996;1(1):16‐30. CENTRAL

Grizenko 2010 {published data only}

Grizenko N, Paci M, Joober R. Is the inattentive subtype of ADHD different from the combined/hyperactive subtype?. Journal of Attention Disorders 2010;13(6):649‐57. CENTRAL

Günther 2010 {published data only}

Günther T, Herpertz‐Dahlmann B, Konrad K. Sex differences in attentional performance and their modulation by methylphenidate in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2010;20(3):179‐86. CENTRAL

Halliday 1983 {published data only}

Halliday R, Callaway E, Naylor H. Visual evoked potential changes induced by methylphenidate in hyperactive children: dose/response effects. Electroencephalography and Clinical Neurophysiology 1983;55(3):258‐67. CENTRAL

Hanisch 2004 {published data only}

Hanisch C, Konrad K, Günther T, Herpertz‐Dahlmann B. Age‐dependent neuropsychological deficits and effects of methylphenidate in children with attention‐deficit/hyperactivity disorder: a comparison of pre‐ and grade‐school children. Journal of Neural Transmission 2004;111(7):865‐81. CENTRAL

Hazel‐Fernandez 2006 {published data only}

Hazel‐Fernandez LA, Klorman R, Wallace JM, Cook S. Methylphenidate improves aspects of executive function in African American children with ADHD. Journal of Attention Disorders 2006;9(4):582‐9. CENTRAL

Hinshaw 1989 {published data only}

Hinshaw SP, Buhrmester D, Heller T. Anger control in response to verbal provocation: effects of stimulant medication for boys with ADHD. Journal of Abnormal Child Psychology 1989;17(4):393‐407. [PUBMED: 2677081]CENTRAL
Hinshaw SP, Henker B, Whalen CK, Erhardt D, Dunnington RE. Aggressive, prosocial, and nonsocial behavior in hyperactive boys: dose effects of methylphenidate in naturalistic settings. Journal of Consulting and Clinical Psychology 1989;57(5):636‐43. [DOI: 10.1037/0022‐006X.57.5.636]CENTRAL

Hinshaw 1993 {published data only}

Hinshaw SP, Heller T, McHale JP. Covert antisocial behavior in boys with attention‐deficit hyperactivity disorder: external validation and effects of methylphenidate. Journal of Consulting and Clinical Psychology 1992;60(2):274‐81. CENTRAL

Humphries 1979 {published data only}

Humphries T, Swanson J, Kinsbourne M, Yiu L. Stimulant effects on persistence of motor performance of hyperactive children. Journal of Pediatric Psychology 1979;4(1):55‐66. CENTRAL

King 2009a {published data only}

King S, Waschbusch DA, Pelham WE, Frankland BW, Corkum PV, Jacques S. Subtypes of aggression in children with attention deficit hyperactivity disorder: medication effects and comparison with typical children. Journal of Clinical Child and Adolescent Psychology 2009;38(5):619‐29. CENTRAL

King 2009b {published data only}

King S, Waschbusch DA, Pelham WE, Frankland BW, Andrade BF, Jacques S, et al. Social information processing in elementary‐school aged children with ADHD: medication effects and comparisons with typical children. Journal of Abnormal Child Psychology 2009;37(4):579‐89. CENTRAL

Lange 2007 {published data only}

Lange KW, Tucha LT, Walitza S, Sontag TA, Stasik D, Linder M, et al. Effects of methylphenidate on multiple components of attention in children with attention deficit hyperactivity disorder. Journal of Neural Transmission. Abstracts of the 39th International Danube Symposium for Neurological Sciences and Continuing Education and 1st International Congress on ADHD, from Childhood to Adult Disease; 2007 June 2‐5; Wurzburg, Germany. 2007; Vol. 114 (7):LXXV. CENTRAL

Leitner 2007b {published data only}

Leitner Y, Barak R, Giladi N, Peretz C, Eshel R, Gruendlinger L, et al. Gait in attention deficit hyperactivity disorder: effects of methylphenidate and dual tasking. Journal of Neurology 2007;254(10):1330‐8. [PUBMED: 17401735]CENTRAL
Leitner Y, Doniger GM, Barak R, Simon ES, Hausdorff JM. A novel multidomain computerized cognitive assessment for attention‐deficit hyperactivity disorder: evidence for widespread and circumscribed cognitive deficits. Journal of Child Neurology 2007;22(3):264‐76. [PUBMED: 17621495 ]CENTRAL

Malone 1988 {published data only}

Malone MA, Kershner JR, Siegel L. The effects of methylphenidate on levels of processing and laterality in children with attention deficit disorder. Journal of Abnormal Child Psychology 1988;16(4):379‐95. CENTRAL

Malone 1993 {published data only}

Malone MA, Swanson JM. Effects of methylphenidate on impulsive responding in children with attention‐deficit hyperactivity disorder. Journal of Child Neurology 1993;8(2):157‐63. CENTRAL

Malone 1994 {published data only}

Malone MA, Couitis J, Kershner JR, Logan WJ. Right hemisphere dysfunction and methylphenidate effects in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 1994;4(4):245‐53. CENTRAL

Martin 2007 {published data only}

Martin CA, Guenthner G, Bingcang C, Rayens MK, Kelly TH. Measurement of the subjective effects of methylphenidate in 11‐ to 15‐year‐old children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2007;17(1):63‐73. CENTRAL

Mehta 2004 {published data only}

Mehta MA, Goodyer IM, Sahakian BJ. Methylphenidate improves working memory and set‐shifting in AD/HD: relationships to baseline memory capacity. Journal of Child Psychology and Psychiatry and Alllied Disciplines 2004;45(2):293‐305. CENTRAL

Milich 1989 {published data only}

Milich R, Licht BG, Murphy DA, Pelham WE. Attention‐deficit hyperactivity disordered boys' evaluations of and attributions for task performance on medication versus placebo. Journal of Abnormal Psychology 1989;98(3):280‐4. CENTRAL

Milich 1991 {published data only}

Milich R, Carlson CL, Pelham WE, Licht BG. Effects of methylphenidate on the persistence of ADHD boys following failure experiences. Journal of Abnormal Child Psychology 1991;19(5):519‐36. CENTRAL

Novak 1995 {published data only}

Novak GP, Solanto M, Abikoff H. Spatial orienting and focused attention in attention deficit hyperactivity disorder. Psychophysiology 1995;32(6):546‐59. CENTRAL

O'Toole 1997 {published data only}

O'Toole K, Abramowitz A, Morris R, Dulcan M. Effects of methylphenidate on attention and nonverbal learning in children with attention‐deficit hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(4):531‐8. CENTRAL

Peeke 1984 {published data only}

Peeke S, Halliday R, Callaway E, Prael R, Reus V. Effects of two doses of methylphenidate on verbal information processing in hyperactive children. Journal of Clinical Psychopharmacology 1984;4(2):82‐8. CENTRAL

Pelham 1985 {published data only}

Pelham WE, Bender ME, Caddell J, Booth S, Moorer SH. Methylphenidate and children with attention deficit disorder. Dose effects on classroom academic and social behavior. Archives of General Psychiatry 1985;42(10):948‐52. CENTRAL

Pelham 1990b {published data only}

Pelham WE, McBurnett K, Harper GW, Milich R, Murphy DA, Clinton J, et al. Methylphenidate and baseball playing in ADHD children: who's on first?. Journal of Consulting and Clinical Psychology 1990;58(1):130‐3. [PUBMED: 2319047]CENTRAL

Pelham 1992 {published data only}

Pelham WE, Murphy DA, Vannatta K, Milich R. Methylphenidate and attributions in boys with attention‐deficit hyperactivity disorder. Annual Progress in Child Psychiatry & Child Development. American Psychological Association, 1993:242‐65. CENTRAL
Pelham WE, Murphy DA, Vannatta K, Milich R, Licht BG, Gnagy EM, et al. Methylphenidate and attributions in boys with attention‐deficit hyperactivity disorder. Journal of Consulting and Clinical Psychology 1992;60(2):282‐92. [PsycINFO: 1994‐34504‐001]CENTRAL

Pelham 1997 {published data only}

Pelham WE, Hoza B, Kipp HL, Gnagy EM, Trane ST. Effects of methylphenidate and expectancy on ADHD children's performance, self‐evaluations, persistence, and attributions on a cognitive task. Experimental and Clinical Psychopharmacology 1997;5(1):3‐13. CENTRAL

Pelham 2001b {published data only}

Pelham WE, Waschbusch DA, Hoza B, Pillow DR, Gnagy EM. Effects of methylphenidate and expectancy on performance, self‐evaluations, persistence, and attributions on a social task in boys with ADHD. Experimental and Clinical Psychopharmacology 2001;9(4):425‐37. CENTRAL

Rapport 1995 {published data only}

Rapport MD, Loo S, Denney C. The paired associate learning task: is it an externally valid instrument for assessing methylphenidate response in children with attention deficit disorder?. Journal of Psychopathology and Behavioral Assessment 1995;17(2):125‐44. CENTRAL

Richardson 1988 {published data only}

Kupietz SS, Winsberg BG, Sverd J. Learning ability and methylphenidate (Ritalin(®)) plasma concentration in hyperkinetic children: a preliminary investigation. Journal of the American Academy of Child and Adolescent Psychiatry 1982;21(1):27‐30. CENTRAL
Richardson E, Kupietz SS, Winsberg BG, Maitinsky S, Mendell N. Effects of methylphenidate dosage in hyperactive reading‐disabled children: II. Reading achievement. Journal of the American Academy of Child and Adolescent Psychiatry 1988;27(1):78‐87. CENTRAL

Rubia 2003 {published data only}

Rubia K, Noorloos J, Smith A, Gunning B, Sergeant J. Motor timing deficits in community and clinical boys with hyperactive behavior: the effect of methylphenidate on motor timing. Journal of Abnormal Child Psychology 2003;31(3):301‐13. CENTRAL

Sangal 2006 {published data only}

Sangal RB, Sangal JM. Attention‐deficit/hyperactivity disorder: use of cognitive evoked potential (P300) to predict treatment response. Clinical Neurophysiology 2006;117(9):1996‐2006. CENTRAL

Sengupta 2008 {published data only}

Sengupta S, Grizenko N, Schmitz N, Schwartz G, Bellingham J, Polotskaia A, et al. COMT Val108/158Met polymorphism and the modulation of task‐oriented behavior in children with ADHD. Neuropsychopharmacology 2008;33(13):3069‐77. CENTRAL

Silk 2012 {published data only}

Silk T. Resting‐state functional connectivity anomalies in ADHD and responses to methylphenidate medication. Brain Connectivity2012; Vol. 2, issue 4:A128. CENTRAL

Smith 2013 {published data only}

Smith A, Cubillo A, Barrett N, Giampietro V, Simmons A, Brammer M, et al. Neurofunctional effects of methylphenidate and atomoxetine in boys with attention‐deficit/hyperactivity disorder during time discrimination. Biological Psychiatry 2013;74(8):615‐22. CENTRAL

Solanto 1986 {published data only}

Solanto MV. Behavioral effects of low‐dose methylphenidate in childhood attention deficit disorder: implications for a mechanism of stimulant drug action. Journal of the American Academy of Child and Adolescent Psychiatry 1986;25(1):96‐101. CENTRAL

Solanto 1997 {published data only}

Solanto MV, Wender EH, Bartell SS. Effects of methylphenidate and behavioral contingencies on sustained attention in attention‐deficit hyperactivity disorder: a test of the reward dysfunction hypothesis. Journal of Child and Adolescent Psychopharmacology 1997;7(2):123‐36. CENTRAL

Srinivas 1992 {published data only}

Srinivas NR, Hubbard JW, Quinn D, Midha KK. Enantioselective pharmacokinetics and pharmacodynamics of dl‐threo‐methylphenidate in children with attention deficit hyperactivity disorder. Clinical Pharmacology and Therapeutics 1992;52(5):561‐8. CENTRAL

Strand 2012 {published data only}

Strand MT, Hawk LW, Bubnik M, Shiels K, Pelham WE, Waxmonsky JG. Improving working memory in children with attention‐deficit/hyperactivity disorder: the separate and combined effects of incentives and stimulant medication. Journal of Abnormal Child Psychology2012; Vol. 40, issue 7:1193‐207. CENTRAL

Stray 2009 {published data only}

Stray LL, Stray T, Iversen S, Ruud A, Ellertsen B. Methylphenidate improves motor functions in children diagnosed with hyperkinetic disorder. Behavioral and Brain Functions 2009;5:21. CENTRAL

Swanson 1993 {published data only}

Swanson J, McBurnett K, Wigal T, Pfiffner LJ. Effect of stimulant medication on children with attention deficit disorder: a "review of reviews". Exceptional Children 1993;60(2):154‐61. CENTRAL

Szobot 2003 {published data only}

Szobot CM, Ketzer C, Cunha RD, Parente MA, Langleben DD, Acton PD, et al. The acute effect of methylphenidate on cerebral blood flow in boys with attention‐deficit/hyperactivity disorder. European Journal of Nuclear Medicine and Molecular Imaging 2003;30(3):423‐6. CENTRAL

Tannock 2000 {published data only}

Tannock R, Martinussen R, Frijters J. Naming speed performance and stimulant effects indicate effortful, semantic processing deficits in attention‐deficit/hyperactivity disorder. Journal of Abnormal Child Psychology 2000;28(3):237‐52. CENTRAL

Teicher 2003 {published data only}

Teicher MH, Polcari A, Anderson CM, Andersen SL, Lowen SB, Navalta CP. Rate dependency revisited: understanding the effects of methylphenidate in children with attention deficit hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2003;13(1):41‐51. CENTRAL

Teicher 2006 {published data only}

Teicher MH, Polcari A, Foley M, Valente E, McGreenery CE, Chang WW, et al. Methylphenidate blood levels and therapeutic response in children with attention‐deficit hyperactivity disorder: I. Effects of different dosing regimens. Journal of Child and Adolescent Psychopharmacology 2006;16(4):416‐31. CENTRAL

Teicher 2007 {published data only}

Teicher MH, Polcari A, McGreenery CE. Objective measures of activity and attention accurately identify methylphenidate doses associated with optimal clinical response in children with ADHD. Biological Psychiatry 2007;61(8 Suppl):66S. CENTRAL
Teicher MH, Polcari A, McGreenery CE. Utility of objective measures of activity and attention in the assessment of therapeutic response to stimulants in children with attention‐deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology 2008;18(3):265‐70. CENTRAL

Tillery 2000 {published data only}

Tillery KL. A double‐blind study of the Central Auditory Processing and Auditory Continuous Test performances of children with attention deficit hyperactivity disorder and central auditory processing disorder under Ritalin and placebo conditions. Dissertation Abstracts International: Section A: Humanities and Social Sciences1998; Vol. 58, issue 7‐A:2536. CENTRAL
Tillery KL, Katz J, Keller WD. Effects of methylphenidate (Ritalin) on auditory performance in children with attention and auditory processing disorders. Journal of Speech, Language, and Hearing Research 2000;43(4):893‐901. [PUBMED: 11386476]CENTRAL

Trommer 1991 {published data only}

Trommer BL, Hoeppner JA, Zecker SG. The go‐no go test in attention deficit disorder is sensitive to methylphenidate. Journal of Child Neurology 1991;6(1 Suppl):S128‐31. CENTRAL

Tsang 2012 {published data only}

Tsang TW, Kohn MR, Clarke SD, Williams LM. Cognition and emotion in child and adolescent ADHD. Biological Psychiatry. Proceedings of the 67th Society of Biological Psychiatry Annual Meeting; 2012 May 2‐4. 2012; Vol. 71 (8 Suppl):74S. CENTRAL

Tucha 2006 {published data only}

Tucha O, Prell S, Mecklinger L, Bormann‐Kischkel C, Kübber S, Linder M, et al. Effects of methylphenidate on multiple components of attention in children with attention deficit hyperactivity disorder. Psychopharmacology 2006;185(3):315‐26. CENTRAL

Verbaten 1994 {published data only}

Verbaten MN, Overtoom CC, Koelega HS, Swaab‐Barneveld H, Van der Gaag RJ, Buitelaar J, et al. Methylphenidate influences on both early and late ERP waves of ADHD children in a continuous performance test. Journal of Abnormal Child Psychology 1994;22(5):561‐78. CENTRAL

Waschbusch 2007 {published data only}

Waschbusch DA, Craig R, Pelham WE, King S. Self‐handicapping prior to academic‐oriented tasks in children with attention deficit/hyperactivity disorder (ADHD): medication effects and comparisons with controls. Journal of Abnormal Child Psychology 2007;35(2):275‐86. CENTRAL

Whalen 1987 {published data only}

Whalen CK, Henker B, Swanson JM, Granger D, Kliewer W, Spencer J. Natural social behaviors in hyperactive children: dose effects of methylphenidate. Journal of Consulting and Clinical Psychology 1987;55(2):187‐93. CENTRAL

Drtilkova 1997 {published data only}

Drtilkova I, Misurec J, Balastikova B. Methylphenidate, amphetaminil and mesocarb in children with hyperkinetic disorder: predicting value changes the pharmo‐EEG profile of psychostimulatory substances [Metylfenidat, amfetaminil a mesocarb u deti s hyperkinetickou poruchou: prediktivni hodnota zmen farmakologickeho EEG profilu psychostimulacnich latek]. Ceska a Slovenska Psychiatrie 1997;93(Suppl 3):44‐53. CENTRAL

Short 2004 {published data only}

Short EJ, Manos MJ, Findling RL, Schubel EA. A prospective study of stimulant response in preschool children: insights from ROC analyses. Journal of the American Academy of Child and Adolescent Psychiatry 2004;43(3):251‐9. CENTRAL

Bottelier 2014 {published data only}

Bottelier MA, Schouw MLJ, Klomp A, Tamminga HGH, Schrantee AGM, Bouziane C, et al. The effects of psychotropic drugs on developing brain (ePOD) study: methods and design. BMC Psychiatry 2014;14(1):48. CENTRAL

Gendron 2012 {published data only}

Gendron M, Rusak B, Rajda M, Corkum PV. Assessing the impact of methylphenidate on sleep in children with ADHD using polysomnography and actigraphy. Sleep 2012;35:A374. CENTRAL

NCT00483106 {unpublished data only}

NCT00483106. Clinical and pharmacological study of attention deficit with hyperactivity disorder (ADHD). https://clinicaltrials.gov/ (accessed 10 March 2014). CENTRAL

NCT02039908 {unpublished data only}

NCT02039908. Examining tolerance to CNS stimulants in ADHD. https://clinicaltrials.gov/ (accessed 4 April 2015). CENTRAL

Nurmi 2013 {published data only}

Nurmi EL, Mallya A, Mallya KS, Hellemann GS, McGough J, Loo SK, et al. Pharmacogenetics of growth effects complicating ADHD treatment. Neuropsychopharmacology. Proceedings of the 52nd Annual Meeting of the American College of Neuropsychopharmacology; 2013 December 8‐12; Hollywood, USA. 2013 (S2); Vol. 38:S492‐3. CENTRAL
Nurmi EL, Mallya KS, McGough J, Loo SK, Bilder RM, Whelan F, et al. Pharmacogenetic moderators of methylphenidate and guanfacine response in children and adolescents with ADHD. Neuropsychopharmacology. Proceedings of the 51st Annual Meeting of the American College of Neuropsychopharmacology; 2012 December 2‐6; Hollywood; USA. 2012; Vol. 38 (S1):S219. CENTRAL

Aasberg 1978

Aasberg M, Montgomery SA, Perris C, Schalling D, Sedvall G. A comprehensive psychopathological rating scale. Acta Psychiatrica Scandinavica. Supplementum 1978;57(S271):5‐27.

Abikoff 1980

Abikoff H, Gittelman R, Klein DF. Classroom observation code for hyperactive children: a replication of validity. Journal of Consulting and Clinical Psychology 1980;48(5):555‐65.

Achenbach 1986

Achenbach TM, Edelbrock CS. Manual for the Teacher's Report Form and Teacher Version of the Child Behavior Profile. Burlington, VT: University of Vermont, Department of Psychiatry, 1986.

Achenbach 1991

Achenbach TM. Integrative Guide to the 1991 CBCL/4‐18, YSR, and TRF Profiles. Burlington, VT: University of Vermont, Department of Psychiatry, 1991.

Achenbach 1991a

Achenbach TM. Manual for the Child Behavior Checklist/4‐18 and 1991 Profile. Burlington, VT: University of Vermont, Department of Pyshciatry, 1991.

Achenbach 1991b

Achenbach TM. Manual for the Teacher Report Form. Burlington, VT: University of Vermont, Department of Psychiatry, 1991.

Achenbach 2001

Achenbach TM, Rescorla LA. Manual for the ASEBA School‐Age Forms and Profiles. Burlington, VT: ASEBA, 2001.

AIM‐C 2013

AIM‐C 2015. ADHD Impact module ‐ child. bit.ly/1fgWlKo (accessed 6 July 2015).

Aman 1996

Aman MG, Tassé MJ, Rojahn J, Hammer D. The Nisonger CBRF: a child behavior rating form for children with developmental disabilities. Research in Developmental Disabilities 1996;17(1):41‐57.

American Academy of Pediatrics 2011

Subcommittee on Attention‐Deficit/Hyperactivity Activity Disorder, Steering Committee on Quality Improvement and Management. ADHD: clinical practice guideline for the diagnosis, evaluation, and treatment of attention‐deficit/hyperactivity disorder in children and adolescents. Pediatrics 2011;128(5):1007‐22.

Andrews 2013a

Andrews J, Guyatt G, Oxman AD, Alderson P, Dahm P, Falck‐Ytter Y, et al. GRADE guidelines: 14. Going from evidence to recommendations: the significance and presentation of recommendations. Journal of Clinical Epidemiology 2013;66(7):719–25.

Andrews 2013b

Andrews JC, Schünemann HJ, Oxman AD, Pottie K, Meerpohl JJ, Coello PA, et al. GRADE guidelines: 15. Going from evidence to recommendation ‐ determinants of a recommendation’s direction and strength. Journal of Clinical Epidemiology 2013;66(7):726–35.

Angold 1995

Angold A, Prendergast M, Cox A, Harrington R, Simonoff E, Rutter M. The Child and Adolescent Psychiatric Assessment (CAPA). Psychological Medicine 1995;25(4):739‐53.

APA 1980

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM‐III). 3rd Edition. Washington, DC: American Psychiatric Association, 1980.

APA 1987

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM III‐R). 3rd Edition. Washington, DC: American Psychiatric Association, 1987.

APA 1994

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM‐IV). 4th Edition. Washington, DC: American Psychiatric Association, 1994.

APA 2000

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM‐IV‐TR) Text revision. 4th Edition. Washington, DC: American Psychiatric Publishing, 2000.

APA 2013

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM‐5). 5th Edition. Washington, DC: American Psyciatric Association, 2013.

Balshem 2011

Balshem H, Helfand M, Schünemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401–6.

Barkley 1977a

Barkley RA. A review of stimulant drug research with hyperactive children. Journal of Child Psychology and Psychiatry 1977;18(2):137‐65.

Barkley 1981

Barkley RA. The use of psychopharmacology to study reciprocal influences in parent‐child interaction. Journal of Abnormal Child Psychology 1981;9(3):303‐10.

Barkley 1987

Barkley RA, Edelbrock CS. In: Prinz R editor(s). Assessing Situational Variation in Children's Behavior Problems: The Home and School Situations Questionnaires. Greenwich, CT: JAI Press, 1987.

Barkley 1988b

Barkley RA. Child behavior rating scales and checklists. In: Rutter M, Tuma AH, Lann I editor(s). Assessment and Diagnosis in Child Psychopathology. New York: Guilford Press, 1988:113‐55.

Barkley 1989a

Barkley RA. Hyperactive girls and boys: stimulant drug effects on mother‐child interactions. Journal of Child Psychology and Psychiatry 1989;30(3):379‐90.

Barkley 1990

Barkley RA, McMurray MB, Edelbrock CS, Robbins K. Side effects of methylphenidate in children with attention deficit hyperactivity disorder: a systemic, placebo‐controlled evaluation. Pediatrics 1990;86(2):184‐92.

Barkley 1991a

Barkley RA. Attention‐Deficit Hyperactivity Disorder: A Clinical Workbook. New York: Guilford Press, 1991.

Barkley 1991b

Barkley RA, DuPaul GJ, McMurray MB. Attention deficit disorder with and without hyperactivity: clinical response to three dose levels of methylphenidate. Pediatrics 1991;87(4):519‐31.

Barkley 1998

Barkley RA, Murphy KR, Bauermeister JJ. Attention‐Deficit Hyperactivity Disorder: A Clinical Workbook. 2nd Edition. New York: Guilford Press, 1998.

Becker 1993

Becker MP, Balagtas CC. Marginal modeling of binary cross‐over data. Biometrics 1993;49(4):997‐1009. [DOI: 10.2307/2532242]

Berkey 1996

Berkey CS, Mosteller F, Lau J, Antman E. Uncertainty of the time of first significance in random effects cumulative meta‐analysis. Controlled Clinical Trials 1996;17(5):357‐71.

Bero 2013

Bero L. Why the Cochrane risk of bias tool should include funding source as a standard item [editorial]. Cochrane Database of Systematic Reviews2013; Vol. 12. [DOI: 10.1002/14651858.ED000075]

Biederman 2003b

Biederman J. Pharmacotherapy for attention‐deficit/hyperactivity disorder (ADHD) decreases the risk for substance abuse: findings from a longitudinal follow‐up of youths with and without ADHD. Journal of Clinical Psychiatry 2003;64(Suppl 11):3‐8.

Bloch 2009

Bloch MH, Panza KE, Landeros‐Weisenberger A, Leckman JF. Meta‐analysis: treatment of attention‐deficit/hyperactivity disorder in children with comorbid tic disorders. Journal of the American Academy of Child and Adolescent Psychiatry 2009;48(9):884‐93.

Block 1998

Block SL. Attention‐deficit disorder: a paradigm for psychotropic medication intervention in pediatrics. Pediatric Clinics of North America 1998;45(5):1053‐83.

Brok 2008

Brok J, Thorlund K, Gluud C, Wetterslev J. Trial sequential analysis reveals insufficient information size and potentially false positive results in many meta‐analysis. Journal of Clinical Epidemiology 2008;61(8):763‐9.

Brok 2009

Brok J, Thorlund K, Wetterslev J, Gluud C. Apparently conclusive meta‐analysis may be inconclusive ‐ trial sequential analysis adjustment of random error risk due to repetitive testing of accumulating data in apparently conclusive neonatal meta‐analysis. International Journal of Epidemiology 2009;38(1):287‐98.

Brunetti 2013

Brunetti M, Shemilt I, Pregno S, Vale L, Oxman AD, Lord J, et al. GRADE guidelines: 10. Considering resource use and rating the quality of economic evidence. Journal of Clinical Epidemiology 2013;66(2):140–50.

Bruni 1996

Bruni O, Ottaviano S, Guidetti V, Romoli M, Innocenzi M, Cortesi F, et al. The Sleep Disturbance Scale for Children (SDSC). Construction and validation of an instrument to evaluate sleep disturbances in childhood and adolescence. Journal of Sleep Research 1996;5(4):251‐61.

Bussing 2008

Bussing R, Fernandez M, Harwood M, Hou W, Garvan CW, Eyberg SM, et al. Parent and teacher SNAP‐IV ratings of attention deficit hyperactivity disorder symptoms: psychometric properties and normative ratings from a school district sample. Assessment 2008;15(3):317‐28.

CADDRA 2011

Canadian Attention Deficit Hyperactivity Disorder Resource Alliance. Canadian ADHD Practice Guidelines. 3rd Edition. Toronto: CADDRA, 2011.

Castellanos 2006

Castellanos FX, Sonuga‐Barke EJ, Milham MP, Tannock R. Characterizing cognition in ADHD: beyond executive dysfunction. Trends in Cognitive Sciences 2006;10(3):117‐23.

Caye 2013

Caye A, Machado JD, Rohde LA. Evaluating parental disagreement in ADHD diagnosis: can we rely on a single report from home?. Journal of Attention Disorders 2013;XX(X):1‐6. [DOI: 10.1177/1087054713504134]

Chambers 1985

Chambers WJ, Puig‐Antich J, Hirsch M, Paez P, Ambrosini PJ, Tabrizi MA, et al. The assessment of affective disorders in children and adolescents by semistructured interview. Test‐retest reliability of the schedule for affective disorders and schizophrenia for school‐age children, present episode version. Archives of General Psychiatry 1985;42(7):696‐702.

Chan 2013

Chan A‐W, Tetzlaff JM, Altman DG, Laupacis A, Gøtzsche PC, Krleža‐Jerić K, et al. SPIRIT 2013 statement: defining standard protocol items for clinical trials. Annals of Internal Medicine2013; Vol. 158, issue 3:200‐7.

Charach 2011

Charach A, Yeung E, Climans T, Lillie E. Childhood attention‐deficit/hyperactivity disorder and future substance use disorders: comparative meta‐analyses. Journal of the American Academy of Child and Adolescent Psychiatry 2011;50(1):9‐21.

Charach 2013

Charach A, Carson P, Fox S, Ali MU, Beckett J, Lim CG. Interventions for preschool children at high risk for ADHD: a comparative effectiveness review. Pediatrics 2013;131(5):e1584‐604.

Chen 2006

Chen W, Taylor E. Parental account of children's symptoms (PACS), ADHD phenotypes and its applications to molecular genetic studies. In: Oades RD editor(s). Attention‐Deficit/Hyperactivity Disorders and the Hyperkinetic Syndrome: Current Ideas and Ways Forward. Hauppage, NY: Nova Science Publishing, 2006:3‐20.

Cherland 1999

Cherland E, Fritzpark R. Psychotic side effects of psychostimulants: a 5‐year review. Canadian Journal of Psychiatry 1999;44(8):811‐3.

Chervin 2000

Chervin RD, Hedger K, Dillon JE, Pituch KJ. Pediatric Sleep Questionnaire (PSQ): validity and reliability of scales for sleep‐disordered breathing, snoring, sleepiness, and behavioral problems. Sleep Medicine 2000;1(1):21‐32.

Clark 1993

Clark LA. Manual for the Schedule for Nonadaptive and Adaptive Personality (SNAP): Manual for Administration, Scoring, and Interpretation. Minneapolis: University of Minnesota Press, 1993.

Clark 1996

Clark LA. Schedule for Nonadaptive and Adaptive Personality. Manual for Administration, Scoring, and Interpretation. Minneapolis: University of Minnesota Press, 1996.

Conners 1985

Conners CK, Barkley RA. Rating scales and checklists for child psychopharmacology. Psychopharmacology Bulletin 1985;21(4):809‐43.

Conners 1995

Conners CK. The Conners' Parent Rating Scales. Toronto: Multi‐Health Systems, 1995.

Conners 1997a

Conners CK. Conners' Rating Scales. Revised. North Tonowanda: Multi‐Health Systems, 1997.

Conners 1997b

Conners CK, Wells KC, Parker JD, Sitarenios G, Diamond JM, Powell JW. A new self‐report scale for assessment of adolescent psychopathology: factor structure, reliability, validity, and diagnostic sensitivity. Journal of Abnormal Child Psychology 1997;25(6):487‐97. [DOI: 10.1023/A:1022637815797]

Conners 1998a

Conners CK, Sitarenios G, Parker JDA, Epstein JN. Revision and restandardization of the Conners Teacher Rating Scale (CTRS‐R): factor structure, reliability, and criterion validity. Journal of Abnormal Child Psychology 1998;26(4):279‐91.

Conners 1998b

Conners CK, Sitarenios G, Parker JD, Epstein JN. The revised Conners' Parent Rating Scale (CPRS‐R): factor structure, reliability, and criterion validity. Journal of Abnormal Child Psychology 1998;26(4):257‐68.

Conners 2001

Conners CK. Conners Rating Scales, Revised. North Tonawanda: Multi‐Health Systems, 2001.

Conners 2008

Conners CK. Conners 3rd Edition. Toronto: Multi‐Health Systems Inc, 2008.

Connor 2002

Connor DF. Preschool attention deficit hyperactivity disorder: a review of prevalence, diagnosis, neurobiology, and stimulant treatment. Journal of Developmental and Behavioral Pediatrics 2002;23(1 Suppl):S1‐S9.

Corkum 2007

Corkum P, Andreou P, Schachar R, Tannock R, Cunningham C. The Telephone Interview Probe: a novel measure of treatment response in children with attention deficit hyperactivity disorder. Educational and Psychological Measurement 2007;67(1):169‐85.

Cox 2004a

Cox DJ, Merkel RL, Penberthy JK, Kovatchev B, Hankin CS. Impact of methylphenidate delivery profiles on driving performance of adolescents with attention‐deficit/hyperactivity disorder: a pilot study. Journal of the American Acadademy of Child and Adolescent Psychiatry 2004;43(3):269‐75.

CTU 2011

Copenhagen Trial Unit. TSA ‐ trial sequential analysis. www.ctu.dk/tsa (accessed 4 March 2015).

Curtin 2002

Curtin F, Elbourne D, Altman DG. Meta‐analysis combining parallel and cross‐over clinical trials. III: The issue of carry‐over. Statistics in Medicine 2002;21(15):2161‐73.

DuPaul 1991a

DuPaul GJ. Parent and teacher ratings of ADHD symptoms: psychometric properties in a community‐based sample. Journal of Clinical Child Psychology 1991;20(3):245‐53.

DuPaul 1992

DuPaul GJ, Barkley RA. Situational variability of attention problems: psychometric properties of the Revised Home and School Situations Questionnaires. Journal of Clinical Child Psychology 1992;21(2):178‐88.

Döpfner 2000

Döpfner M, Lehmkuhl G. Diagnostik‐System für Psychische Störungen im Kindes‐ und Jugendalter nach ICD‐10 und DSM‐IV (DISYPS‐KJ). Bern: Huber, 2000.

Döpfner 2008

Döpfner M, Görtz‐Dorten A, Lehmkuhl G. Diagnostik‐System für Psychische Störungen im Kindes‐ und Jugendalter nach ICD‐10 und DSM‐IV. DISYPS‐II. Bern: Huber, 2008.

Efstratopoulou 2013

Efstratopoulou M, Simons J, Janssen R. Concordance among physical educators’, teachers’, and parents’ perceptions of attention problems in children. Journal of Attention Disorders 2013;17(5):437‐43.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JP, Curtin F, Worthington HV, Vail A. Meta‐analyses involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140‐9. [PUBMED: 11914310]

Engert 2008

Engert V, Pruessner JC. Dopaminergic and noradrenergic contributions to functionality in ADHD: the role of methylphenidate. Current Neuropharmacology 2008;6(4):322‐8.

Epstein 2014

Epstein T, Patsopoulos NA, Weiser M. Immediate‐release methylphenidate for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database of Systematic Reviews 2014, Issue 9. [DOI: 10.1002/14651858.CD005041.pub2]

Ercan 2001

Ercan ES, Amado S, Somer O, Çıkoğlu S. Development of a test battery for attention deficit hyperactivity and disruptive behavior disorders [Dikkat eksikligi hiperaktivite bozuklugu ve yikici davranis bozukluklari icin bir test bataryasi gelistirme cabasi]. Turkish Journal of Child and Adolescent Mental Health 2001;8(3):132‐44.

Fabiano 2006

Fabiano GA, Pelham WE, Waschbusch DA, Gnagy EM, Lahey BB, Chronis AM, et al. A practical measure of impairment: psychometric properties of the impairment rating scale in samples of children with attention deficit hyperactivity disorder and two school‐based samples. Journal of Clinical Child and Adolescent Psychology 2006;35(3):369‐85.

Faraone 2002

Faraone SV, Biederman J, Roe C. Comparative efficacy of Adderall and methylphenidate in attention‐deficit/hyperactivity disorder: a meta‐analysis. Journal of Clinical Psychopharmacology 2002;22(5):468‐73.

Faraone 2006

Faraone SV, Biederman J, Spencer TJ, Aleardi M. Comparing the efficacy of medications for ADHD using meta‐analysis. Medscape General Medicine 2006;8(4):4.

Faraone 2009

Faraone SV. Using meta‐analysis to compare the efficacy of medications for attention‐deficit/hyperactivity disorder in youths. P & T: A Peer‐Reviewed Journal for Formulary Management 2009;34(12):678‐94.

Faraone 2010

Faraone SV, Glatt SJ. A comparison of the efficacy of medications for adult attention‐deficit/hyperactivity disorder using meta‐analysis of effect sizes. Journal of Clinical Psychiatry 2010;71(6):754‐63.

Fischer 1998

Fischer M, Newby RF. Use of the restricted academic task in ADHD dose‐response relationships. Journal of Learning Disabilities 1998;31(6):608‐12.

Fish 1985

Fish B. Children's Psychiatric Rating Scale (scoring). Psychopharmacology Bulletin 1985;21(4):1764‐985.

Fitzpatrick 1992b

Fitzpatrick PA, Klorman R, Brumaghim JT, Borgstedt AD. Effects of sustained‐release and standard preparations of methylphenidate on attention deficit disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1992;31(2):226‐34.

Gadow 1994

Gadow KD, Sprafkin J. Child Symptom Inventories Manual. Stony Brook, NY: Checkmate Plus, 1994.

Gadow 1996

Gadow KD, Sprafkin J, Nolan EE. ADHD School Observation Code. New York: Checkmate Plus, 1996.

Gale 1986

Gale J, Pfefferbaum B, Suhr MA, Overall JE. The Brief Psychiatric Rating Scale for Children: a reliability study. Journal of Clinical Child Psychology 1986;15(4):341‐5.

Gilmore 2001

Gilmore A, Milne R. Methylphenidate in children with hyperactivity: review and cost‐utility analysis. Pharmacoepidemology and Drug Safety 2001;10(2):85‐94.

Gittleman‐Klein 1976

Gittleman‐Klein R, Klein DF. Methylphenidate effects in learning disabilities: psychometric changes. Archives of General Psychiatry 1976;33(6):655‐64.

Gluud 2015b

The Nordic Trial Alliance Working Group 6 on Transparency and Registration. Report on transparency and registration in clinical research in the Nordic countries. bit.ly/1S2H1Np (accessed 30 April 2015).

Gould 2009

Gould M S, Walsh B T, Munfakh J L, Kleinman M, Duan N H, Olfson M, et al. Sudden Death and Use of Stimulant Medications in Youths. American Journal of Psychiatry 2009;166(9):992‐1001.

Goyette 1978

Goyette CH, Conners CK, Ulrich RF. Normative data on revised Conners Parent and Teacher Rating Scales. Journal of Abnormal Child Psychology 1978;6(2):221‐36.

GRADEpro 2014 [Computer program]

McMaster Unviersity. GRADEpro. McMaster Unviersity, 2014.

Greenhill 2006b

Greenhill L, Kollins S, Abikoff H, McCracken J, Riddle M, Swanson J, et al. Efficacy and safety of immediate release methylphenidate treatment for preschoolers with ADHD. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(11):1284‐93.

Guy 1976

Guy W. ECDEU Assessment Manual for Psychopharmacology ‐ Revised (DHHS Publ No ADM 91‐338). Rockville, MD, US: Department of Health and Human Services, 1976:218‐22.

Guy 1976a

Guy W. ECDEU Assessment Manual for Psychopharmacology. Rockville, MD: US Department of Health, Education, and Welfare Public Health Service Alcohol, Drug Abuse, and Mental Health Administration, 1976.

Guyatt 2011a

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction ‐ GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383–94.

Guyatt 2011b

Guyatt GH, Oxman AD, Kunz R, Atkins D, Brozek J, Vist G, et al. GRADE guidelines: 2. Framing the question and deciding on important outcomes. Journal of Clinical Epidemiology 2011;64(4):395–400.

Guyatt 2011c

Guyatt GH, Oxman AD, Vist G, Kunz R, Brozek J, Alonso‐Coello P, et al. GRADE guidelines: 4. Rating the quality of evidence ‐ study limitations (risk of bias). Journal of Clinical Epidemiology 2011;64(4):407–15.

Guyatt 2011d

Guyatt GH, Oxman AD, Montori V, Vist G, Kunz R, Brozek J, et al. GRADE guidelines: 5. Rating the quality of evidence ‐ publication bias. Journal of Clinical Epidemiology 2011;64(12):1277–82.

Guyatt 2011e

Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence ‐ imprecision. Journal of Clinical Epidemiology 2011;64(12):1283–93.

Guyatt 2011f

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 7. Rating the quality of evidence ‐ inconsistency. Journal of Clinical Epidemiology 2011;64(12):1294–302.

Guyatt 2011g

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 8. Rating the quality of evidence ‐ indirectness. Journal of Clinical Epidemiology 2011;64(12):1303–10.

Guyatt 2011h

Guyatt GH, Oxman AD, Sultan S, Glasziou P, Akl EA, Alonso‐Coello P, et al. GRADE guidelines: 9. Rating up the quality of evidence. Journal of Clinical Epidemiology 2011;64(12):1311–6.

Guyatt 2013a

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso‐Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66(2):151–7.

Guyatt 2013b

Guyatt GH, Oxman AD, Santesso N, Helfand M, Vist G, Kunz R, et al. GRADE guidelines: 12. Preparing summary of findings tables ‐ binary outcomes. Journal of Clinical Epidemiology 2013;66(2):158–72.

Guyatt 2013c

Guyatt GH, Thorlund K, Oxman AD, Walter SD, Patrick D, Furukawa TA, et al. GRADE guidelines: 13. Preparing summary of findings tables and evidence profiles ‐ continuous outcomes. Journal of Clinical Epidemiology 2013;66(2):173–83.

Gøtzsche 2015

Gøtzsche P. Comments on the editorial by Sterne: Why the Cochrane risk of bias tool should include funding source as a standard item [editorial]. Comment by Peter Gøtzsche 9 January 2014. Cochrane Database of Systematic Reviews 10 March 2015.

Hanwella 2011

Hanwella R, Senanayake Mi, De Silva V. Comparative efficacy and acceptability of methylphenidate and atomoxetine in treatment of attention deficit hyperactivity disorder in children and adolescents: a meta‐analysis. BMC Psychiatry 2011;11:176.

Hartman 2007

Hartman CA, Rhee SH, Willcutt EG, Pennington, BF. Modeling rater disagreement for ADHD: are parents or teachers biased?. Journal of Abnormal Child Psychology 2007;35(4):536‐42. [PUBMED: 17333362]

Heal 2006

Heal DJ, Pierce DM. Methylphenidate and its isomers: their role in the treatment of attention‐deficit hyperactivity disorder using a transdermal delivery system. CNS Drugs 2006;20(9):713‐38.

Higgins 2011

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

Holden 2013

Holden SE, Jenkins‐Jones S, Poole CD, Morgan CL, Coghill D, Currie CJ. The prevalence and incidence, resource use and financial costs of treating people with attention deficit/hyperactivity disorder (ADHD) in the United Kingdom (1998 to 2010). Child and Adolescent Psychiatry and Mental Health 2013;11(7):34.

Hollis 1999

Hollis S, Campbell F. What is meant by intention to treat analysis? Survey of published randomised controlled trials. BMJ 1999;319(7211):670‐4.

Holmskov 2014 [pers comm]

Holmskov M. Data requested [personal communication]. Email to: Dr Leddy 11 June 2014.

Humphrey 1982

Humphrey LL. Children's and teachers' perspectives on children's self‐control: the development of two rating scales. Journal of Consulting and Clinical Psychology 1982;50(5):624‐33.

ICH 1996

ICH Expert Working Group. International conference on harmonisation of technical requirements for registration of pharmaceuticals for human use. ICH harmonised tripartite guideline. Guideline for good clinical practice E6(R1). http://bit.ly/1B0jeJg (accessed 4 March 2014).

Iversen 2006

Iversen L. Neurotransmitter transporters and their impact on the development of psychopharmacology. British Journal of Pharmacology 2006;147 Suppl 1:S82‐8.

Jakobsen 2013

Jakobsen JC, Gluud C. The necessity of randomised clinical trials. British Journal of Medicine and Medical Research 2013;3(4):1453‐68.

Jakobsen 2014

Jakobsen JC, Wetterslev J, Winkel P, Lange T, Gluud C. Thresholds for statistical and clinical significance in systematic reviews with meta‐analytic methods. BMC Medical Research Methodology 2014;14:120. [DOI: 10.1186/1471‐2288‐14‐120]

Jensen 2001

Jensen PS, Hinshaw SP, Kraemer HC, Lenora N, Newcorn JH, Abikoff HB, et al. ADHD comorbidity findings from the MTA study: comparing comorbid subgroups. Journal of the American Academy of Child and Adolescent Psychiatry 2001;40(2):147‐58.

Kadesjö 2001

Kadesjö B, Gillberg C. The comorbidity of ADHD in the general population of Swedish school‐age children. Journal of Child Psychology and Psychiatry and Allied Disciplines 2001;42(4):487‐92.

Kadesjö 2002

Kadesjö B. ADHD in Children and Adults [ADHD hos barn och vuxna]. Stockholm: Socialstyrelsen, 2002.

Kambeitz 2014

Kambeitz J, Romanos M, Ettinger U. Meta‐analysis of the association between dopamine transporter genotype and response to methylphenidate treatment in ADHD. Pharmacogenomics Journal 2014;14(1):77‐84.

Kanjwal 2012

Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Grubb BP. Use of methylphenidate in the treatment of patients suffering from refractory postural tachycardia syndrome. American Journal of Therapeutics 2012;19(1):2‐6.

Kelsey 2004

Kelsey DK, Sumner CR, Casat CD, Coury DL, Quintana H, Saylor KE, et al. Once‐daily atomoxetine treatment for children with attention‐deficit/hyperactivity disorder, including an assessment of evening and morning behavior: a double‐blind, placebo‐controlled trial. Pediatrics 2004;114(1):e1‐8.

Kendall 1979

Kendall PC, Wilcox LE. Self‐control in children: development of a rating scale. Journal of Consulting and Clinical Psychology 1979;47(6):1020‐9.

Kidd 2000

Kidd PM. Attention deficit/hyperactivity disorder (ADHD) in children: rationale for its integrative management. Alternative Medicine Review 2000;5(5):402‐28.

Kieling 2010

Kieling C, Kieling RR, Rohde LA, Frick PJ, Moffitt T, Nigg JT, et al. The age at onset of attention deficit hyperactivity disorder. American Journal of Psychiatry 2010;167(1):14‐6.

Kimko 1999

Kimko HC, Cross JT, Abernethy DR. Pharmacokinetics and clinical effectiveness of methylphenidate. Clinical Pharmacokinetics 1999;37(6):457–70.

King 2006

King S, Griffin S, Hodges Z, Weatherly H, Asseburg C, Richardson G, et al. A systematic review and economic model of the effectiveness and cost‐effectiveness of methylphenidate, dexamfetamine and atomoxetine for the treatment of attention deficit hyperactivity disorder in children and adolescents. Health Technology Assessment. NIHR Health Technology Assessment Programme, 2006; Vol. 10, issue 23:iii‐iv, xiii‐146.

Kjaergard 2001

Kjaergard LL, Villumsen J, Gluud C. Reported methodologic quality and discrepancies between large and small randomized trials in meta‐analyses. Annals of Internal Medicine 2001;135(11):982‐9.

Krogh 2013a [pers comm]

Krogh HB. Requested data from the trial [personal communication]. Email to: Dr Brams 23 September 2013.

Krogh 2013b [pers comm]

Krogh HB. Requestion data on inclusion criteria [personal communication]. Email to: Dr Cox 8 October 2013.

Krogh 2013c [pers comm]

Krogh HB. Data received from author [personal communication]. Email to: Dr Wolraich 2 July 2013.

Krogh 2014a [pers comm]

Krogh HB. Randomisation procedure and analyses used in our trial [personal communication]. Email to: Russel Barkley 15 February 2014.

Krogh 2014b [pers comm]

Krogh BH. Data requested [personal communication]. Email to: Dr JohnsonJune 2014.

Krogh 2014c [pers comm]

Krogh HB. Data regarding randomisation procedure. Email to: Dr Klorman 21 January 2014.

Lachar 1986

Lachar D, Klein R, Boersma D. Personality Inventory for Children: approaches to actuarial interpretation in clinic and school settings. In: Knoff H editor(s). The Assessment of Child and Adolescent Personality. New York: Guilford Press, 1986:273‐308.

Ladd 1996

Ladd GW, Profilet SM. The Child Behavior Scale: a teacher‐report measure of young children's aggressive, withdrawn, and prosocial behaviors. Developmental Psychology 1996;32(6):1008‐24.

Lahey 2006

Lahey BB, Pelham WE, Chronis A, Massetti G, Kipp H, Ehrhardt A, et al. Predictive validity of ICD‐10 hyperkinetic disorder relative to DSM‐IV attention‐deficit/hyperactivity disorder among younger children. Journal of Child Psychology and Psychiatry and Allied Disciplines 2006;47(5):472‐9.

Landgraf 1998

Landgraf JM, Maunsell E, Speechley KN, Bullinger M, Campbell S, Abetz L, et al. Canadian‐French, German and UK versions of the Child Health Questionnaire: methodology and preliminary item scaling results. Quality of Life Research 1998;7(5):433‐45.

Landgraf 2002

Landgraf JM, Rich M, Rappaport L. Measuring quality of life in children with attention‐deficit/hyperactivity disorder and their families: development and evaluation of a new tool. Archives of Pediatrics and Adolescent Medicine 2002;156(4):384‐91.

Lau 1995

Lau J, Schmid CH, Chalmers TC. Cumulative meta‐analysis of clinical trials builds evidence for exemplary medical care. Journal of Clinical Epidemiology 1995;48(1):45‐57.

Lavigne 2012

Lavigne JV, Dulcan MK, LeBailly SA, Binns HJ. Can parent reports serve as a proxy for teacher ratings in medication management of attention‐deficit hyperactivity disorder?. Journal of Developmental & Behavioral Pediatrics 2012;33(4):336‐42.

Leckman 1988

Leckman JF, Towbin K, Ort SI. Clinical assessment of tic disorder severity. In: Cohen DJ, Bruun RD, Leckman JF editor(s). Tourette's Syndrome and Tic Disorders: Clinical Understanding and Treatment. Oxford: John Wiley & Sons, 1988:55‐78.

Leckman 1989

Leckman JF, Riddle MA, Hardin MT, Ort SI, Swartz KL, Stevenson J, et al. The Yale Global Tic Severity Scale: initial testing of a clinician‐rated scale of tic severity. Journal of the American Academy of Child and Adolescent Psychiatry 1989;28(4):566‐73.

Lee 2008

Lee SI, Schachar RJ, Chen SX, Ornstein TJ, Charach A, Barr C, et al. Predictive validity of DSM‐IV and ICD‐10 criteria for ADHD and hyperkinetic disorder. Journal of Child Psychology and Psychiatry and Allied Disciplines 2008;49(1):70‐8.

Liberati 2009

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Götzsche PC, Ioannidis JPA, et al. Research methods and reporting: the PRISMA statement for reporting systematic reviews and meta‐analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ 2009;339:b2700.

Light 2015

Light DW, Lexchin J. The FDA´s new clothes. BMJ2015; Vol. 651:h4897. [DOI: 10.1136/bmj.h4897]

Loney 1982

Loney J, Milich R. Hyperactivity, inattention, and aggression in clinical practice. In: Wolraich M, Routh DK editor(s). Advances in Developmental and Behavioral Pediatrics. Vol. 3, Greenwich: JAI Press, 1982:113‐47.

Loney 1984

Loney J. A short parent scale for subgrouping childhood hyperactivity, aggression. Annual Meeting of the American Psychological Association; 1984 August 24‐28; Toronto. Toronto, 1984.

Lundh 2012

Lundh A, Sismondo S, Lexchin J, Busuioc OA, Bero L. Industry sponsorship and research outcome. Cochrane Database of Systematic Reviews 2012, Issue 12. [DOI: 10.1002/14651858.MR000033.pub2]

Magnusson 2014a [pers comm]

Magnusson FL. Received information on randomisation and allocation concealment procedure [personal communication]. Email to: Dr Klorman 17 April 2014.

Magnusson 2014b [pers comm]

Magnusson FM. Data received from author [personal communication]. Email to: Dr Rapport 8 May 2014.

Magnusson 2014c [pers comm]

Magnusson F. Data received from author [personal communication]. Email to: Dr KircherMay 2014.

Maia 2014

Maia CR, Cortese S, Caye A, Deakin TK, Polanczyk GV, Polanczyk CA, et al. Long‐term efficacy of methylphenidate immediate‐release for the treatment of childhood ADHD: a systematic review and meta‐analysis. Journal of Attention Disorders 2010 December 14 [Epub ahead of print]. [DOI: 10.1177/1087054714559643]

Marsee 2007

Marsee MA, Frick PJ. Exploring the cognitive and emotional correlates to proactive and reactive aggression in a sample of detained girls. Journal of Abnormal Child Psychology 2007;35(6):969‐81.

McGough 2006a

McGough JJ, Wigal SB, Abikoff H, Turnbow JM, Posner K, Moon E. A randomized, double‐blind, placebo‐controlled, laboratory classroom assessment of methylphenidate transdermal system in children with ADHD. Journal of Attention Disorders 2006;9(3):476‐85.

Mendelsohn 1978

Mendelsohn M, Erdwins C. The Disruptive Behavior Scale: an objective assessment of unmanageable social behavior in adolescents. Journal of Clinical Psychology 1978;34(2):426‐8.

Milich 1980

Milich R, Roberts MA, Loney J, Caputo J. Differentiating practice effects and statistical regression on the Conners Hyperkinesis Index. Journal of Abnormal Child Psychology 1980;8(4):549‐52.

Miller 1997

Miller LS, Koplewicz HS, Klein RG. Teacher ratings of hyperactivity, inattention, and conduct problems in preschoolers. Journal of Abnormal Child Psychology 1997;25(2):113‐9.

Moffit 2007

Moffit TE, Melchior M. Why does the worldwide prevalence of childhood attention deficit hyperactivity disorder matter?. American Journal of Psychiatry 2007;146(6):856‐8.

Moher 1998

Moher D, Pham B, Jones A, Cook DJ, Jadad A, Moher M, et al. Does quality of reports of randomised trials affect estimates of intervention efficacy reported in meta‐analysis?. Lancet 1998;352(9128):609‐13.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group. Preferred reporting items for systematic reviews and meta‐analyses: the PRISMA statement. Open Medicine 2009;3(3):123‐30.

Moher 2010

Moher D, Hopewell S, Schulz KF, Montori V, Gøtzsche PC, Devereaux J, et al. Research methods and reporting: CONSORT 2010 explanation and elaboration: updated guidelines for reporting parallel group randomised trials. BMJ Clinical Research Edition 2010;340:c869. [DOI: http://dx.doi.org/10.1136/bmj.c869]

Moher 2015

Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Preferred reporting items for systematic review and meta‐analysis protocols (PRISMA‐P) 2015 statement. Systematic Reviews 2015;4:1. [DOI: 10.1186/2046‐4053‐4‐1]

Moncrieff 2004

Moncrieff J, Wessely S, Hardy R. Active placebos versus antidepressants for depression. Cochrane Database of Systematic Reviews 2004, Issue 1. [DOI: 10.1002/14651858.CD003012.pub2]

MTA 1999

MTA Cooperation Group. A 14 month randomised clinical trial of treatment strategies for attention‐deficit/hyperactivity disorder. The MTA Cooperative Group. Multimodal treatment study of children with ADHD. Archives of General Psychiatry 1999;56(12):1073‐86.

Murray 2007

Murray DW, Kollins SH, Hardy KK, Abikoff HB, Swanson JM, Cunningham C, et al. Parent versus teacher ratings of attention‐deficit/hyperactivity disorder symptoms in the Preschoolers with Attention‐Deficit/Hyperactivity Disorder Treatment Study (PATS). Journal of Child and Adolescent Psychopharmacology 2007;17(5):605‐19.

Murray 2009

Murray DW, Bussing R, Fernandez M, Hou W, Garvan CW, Swanson JM, et al. Psychometric properties of teacher SKAMP ratings from a community sample. Assessment 2009;16(2):193‐208.

Mustafa 2013

Mustafa RA, Santesso N, Brozek J, Akl EA, Walter SD, Norman G, et al. The GRADE approach is reproducible in assessing the quality of evidence of quantitative evidence syntheses. Journal of Clinical Epidemiology 2013;66(7):736‐42.

NICE 2009

National Collaborating Centre for Mental Health commissioned by the National Institute for Health and Clinical Excellence. Attention Deficit Hyperactivity Disorder. Diagnosis and Management of ADHD in Children, Young People and Adults. National Clinical Practice Guideline Number 72. Leicester and London: The British Psychological Society and The Royal College of Psychiatrists, 2009.

Nigg 2005

Nigg JT, Casey BJ. An integrative theory of attention‐deficit/hyperactivity disorder based on the cognitive and affective neurosciences. Development and Psychopathology 2005;17(3):785‐806.

Nilausen 2014 [pers comm]

Nilausen TD. Requested data [personal communication]. Email to: Dr Kolko 13 January 2014.

Owens 2000

Owens JA, Spirito A, McGuinn M. The Children's Sleep Habits Questionnaire (CSHQ): psychometric properties of a survey instrument for school‐aged children. Sleep 2000;23(8):1043‐51.

Pasini 2007

Pasini A, Paloscia C, Alessandrelli R, Porfirio MC, Curatolo P. Attention and executive functions profile in drug naive ADHD subtypes. Brain & Development 2007;29(7):400‐8.

Pelham 1993b

Pelham WE. Pharmacotherapy for children with attention‐deficit hyperactivity disorder. School Psychology Review 1993;22(2):199‐227.

Pelham 2005a

Pelham WE, Burrows‐MacLean L, Gnagy EM, Fabiano GA, Coles EK, Tresco KE, et al. Transdermal methylphenidate, behavioral, and combined treatment for children with ADHD. Experimental and Clinical Psychopharmacology 2005;13(2):111‐26.

Pfefferbaum‐Levine 1983

Pfefferbaum‐Levine B, Overall JE. Diagnostic factor structure of the Children's Psychiatric Rating Scale (C.P.R.S.). Journal of Clinical Child Psychology 1983;12(2):167‐73.

Pliszka 1998

Pliszka S. The use of psychostimulants in the paediatric patient. Pediatric Clinics of North America 1998;45(5):1085‐98.

Pliszka 2007a

Pliszka S, AACAP Work Group on Quality Issues. Practice parameter for the assessment and treatment of children and adolescents with attention‐deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry 2007;46(7):894‐921.

Polanczyk 2007

Polanczyk G, Rohde LA. Epidemiology of attention‐deficit/hyperactivity disorder across the lifespan. Current Opinion in Psychiatry 2007;20(4):386‐92.

Polanczyk 2010

Polanczyk G, Laranjeira R, Zalesk IM, Pinsky I, Caetano R, Rohde LA. ADHD in a representative sample of the Brazilian population: estimated prevalence and comparative adequacy of criteria between adolescents and adults according to the response theory. International Journal of Methods in Psychiatric Research 2010;19(3):177‐84. [DOI: 10.1002/mpr.319]

Polanczyk 2014

Polanczyk GV, Willcutt EG, Salum GA, Kieling C, Rohde LA. ADHD prevalence estimates across three decades: an updated systematic review and meta‐regression analysis. International Journal of Epidemiology 2014;43(2):434‐42. [DOI: 10.1093/ije/dyt261]

Polderman 2007

Polderman TJ, Derks EM, Hudziak JJ, Verhulst FC, Posthuma D, Boomsma DI. Across the continuum of attention skills: a twin study of the SWAN ADHD rating scale. Journal of Child Psychology and Psychiatry 2007;48(11):1080‐7.

Poznanski 1983

Poznanski EO, Cook SC, Carroll BJ, Corzo H. Use of the Children's Depression Rating Scale in an inpatient psychiatric population. Journal of Clinical Psychiatry 1983;44(6):200‐3.

Požgain 2014

Požgain I, Požgain Z, Degmečić D. Placebo and nocebo effect: a mini review. Psychiatria Danubina 2014;26(2):100‐7.

Punja 2013

Punja S, Zorzela L, Hartling L, Urichuk L, Vohra S. Long‐acting versus short‐acting methylphenidate for paediatric ADHD: a systematic review and meta‐analysis of comparative efficacy. BMJ Open 2013;3(3):e002312.

Raman 2015

Raman SR, Marshall SW, Gaynes BN, Haynes K, Naftel AJ, Stürmer T. An observational study of pharmacological treatment in primary care of children with ADHD in the United Kingdom. Psychiatric Services 2015 March 1 [Epub ahead of print].

Ramstad 2013a [pers comm]

Ramstad E. Data requested from author [personal communication]. Email to: Dr Ialongo 1 August 2013.

Ramstad 2013b [pers comm]

Ramstad E. Request on data from the Rhode Island CLC study [personal communication]. Email to: Dr DuPaul 5 September 2013.

Ramstad 2013c [pers comm]

Ramstad E. Data requestioned [personal communication]. Email to: Dr Rapport 22 July 2013.

Ramstad 2013d [pers comm]

Ramstad E. Data requested from author [personal communication]. Email to: Dr Tirosh 1 August 2013.

Ramstad 2014 [pers comm]

Ramstad E. Data received from authors [personal communication]. Email to: Dr VitielloJuly 2014.

Reichow 2013

Reichow B, Volkmar FR, Bloch MH. Systematic review and meta‐analysis of pharmacological treatment of the symptoms of attention‐deficit/hyperactivity disorder in children with pervasive developmental disorders. Journal of Autism and Developmental Disorders 2013;43(10):2435‐41.

Rentz 2005

Rentz AM, Matza LS, Secnik K, Swensen A, Revicki DA. Psychometric validation of the Child Health Questionnaire (CHQ) in a sample of children and adolescents with attention‐deficit/hyperactivity disorder. Quality of Life Research 2005;14(3):719‐34.

Review Manager 2014 [Computer program]

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

Riley 2004

Riley AW, Forrest CB, Starfield B, Rebok GW, Robertson JA, Green BF. The Parent Report Form of the CHIP‐Child Edition: reliability and validity. Medical Care 2004;42(3):210‐20.

Routh 1978

Routh D. Hyperactivity. In: Magrab P editor(s). Psychological Management of Paediatric Problems. Baltimore: University Park Press, 1978.

Rowe 1997

Rowe KS, Rowe KJ. Norms for parental ratings on Conners' Abbreviated Parent‐Teacher Questionnaire: implications for the design of behavioral rating inventories and analyses of data derived from them. Journal of Abnormal Child Psychology 1997;25(6):425‐51.

Savović 2012a

Savović J, Jones H, Altman D, Harris R, Jüni P, Pildal J, et al. Influence of reported study design characteristics on intervention effect estimates from randomised controlled trials: combined analysis of meta‐epidemiological studies. Health Technology Assessment 2012;16(35):1‐82.

Savović 2012b

Savović J, Jones HE, Altman DG, Harris RJ, Jüni P, Pildal J, et al. Influence of reported study design characteristics on intervention effect estimates from randomized, controlled trials. Annals of Internal Medicine 2012;157(6):429–38.

Scahill 2000

Scahill L, Schwab‐Stone M. Epidemiology of ADHD in school‐age children. Child and Adolescent Psychiatric Clinics of North America 2000;9(3):541‐55.

Schachar 1997b

Schachar RJ, Tannock R, Cunningham C, Corkum PV. Behavioral, situational, and temporal effects of treatment of ADHD with methylphenidate. Journal of the American Academy of Child and Adolescent Psychiatry 1997;36(6):754‐63.

Schachar 2007

Schachar R, Chen S, Crosbie J, Goos L, Ickowicz A, Charach A. Comparison of the predictive validity of hyperkinetic disorder and attention deficit hyperactivity disorder. Journal of the Canadian Academy of Child and Adolescent Psychiatry 2007;16(2):90‐100.

Schachter 2001

Schachter HM, Pham B, King J, Langford S, Moher D. How efficacious and safe is short‐acting methylphenidate for the treatment of attention‐deficit disorder in children and adolescents? A meta‐analysis. Canadian Medical Association Journal 2001;165(11):1475‐88.

Schmidt 2009

Schmidt S, Petermann F. Developmental psychopathology: attention deficit hyperactivity disorder (ADHD). BMC Psychiatry 2009;9:58.

Schroll 2015

Schroll J, Bero L. Editorial: regulatory agencies hold the key to improving Cochrane Reviews of drugs. bit.ly/1D6XyYc (accessed 6 July 2015).

Schulz 1995

Schulz KF, Chalmers I, Hayes RJ, Altman DG. Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA 1995;273(5):408‐12.

Schulz 2012

Schulz KP, Fan J, Bédard AC, Clerkin SM, Ivanov I, Tang CY, et al. Common and unique therapeutic mechanisms of stimulant and nonstimulant treatments for attention‐deficit/hyperactivity disorder. Archives of General Psychiatry 2012;69(9):952‐61.

Sergeant 2003

Sergeant JA, Geurts H, Huijbregts S, Scheres A, Oosterlaan J. The top and the bottom of ADHD: a neuropsychological perspective. Neuroscience and Biobehavioural Reviews 2003;27(7):583‐92.

Shaffer 1983

Shaffer D, Gould MS, Brasic J, Ambrosini P, Fisher P, Bird H, et al. A Children's Global Assessment Scale (CGAS). Archives of General Psychiatry 1983;40(11):1228‐31.

Shaw 2012

Shaw M, Hodgkins P, Caci H, Young S, Kahle J, Woods AG, et al. A systematic review and analysis of long‐term outcomes in attention deficit hyperactivity disorder: effects of treatment and non‐treatment. BMC Medicine Sep 2012;10:99.

SIGN 2009

Scottish Intercollegiate Guidelines Network. Management of attention deficit and hyperkinetic disorders in children and young people. http://www.sign.ac.uk/guidelines/fulltext/112/ (accessed 30 April 2015).

Silva 2005b

Silva RR, Alpert M, Pouget E, Silva V, Trosper S, Reyes K, et al. A rating scale for disruptive behavior disorders, based on the DSM‐IV item pool. Psychiatric Quarterly 2005;76(4):327‐39. [PUBMED: 16217627]

Solanto 1998

Solanto MV. Neuropsychopharmacological mechanisms of stimulant drug action in attention‐deficit hyperactivity disorder: a review and integration. Behavioural Brain Research 1998;94(1):127‐52.

Sonuga‐Barke 2007

Sonuga‐Barke EJS, Coghill D, Markowitz JS, Swanson JM, Vandenberghe M, Hatch SJ. Sex differences in response of children with ADHD to once‐daily formulations of methylphenidate. Journal of American Academy of Child & Adolescent Psychiatry 2007;46(6):701‐10.

Sprafkin 1986

Sprafkin J, Grayson P, Gadow K, Nolan EE, Paolicelli LM. Code for Observing Social Activity (COSA). Stony Brook: State University of New York, Department of Psychiatry and Behavioral Science, 1986.

Steinhausen 1993

Steinhausen H‐C. LF ‐ Teacher Questionnaire after Rutter, Tizard and Whitmore ‐ German edited version [Lehrerfragebogen nach Rutter, Tizard und Whitmore ‐ Deutsche bearbeitete Fassung]. Psychische Störungen bei Kindern und Jugendlichen. Lehrbuch der Kinder‐ und Jugendpsychiatrie. München: Urban Und Schwarzenberg, 1993.

Sterne 2013

Sterne J. Why the Cochrane risk of bias tool should not include funding source as a standard item [editorial]. Cochrane Database of Systematic Reviews2013; Vol. 12. [DOI: 10.1002/14651858.ED000076]

Stevenson 1989

Stevenson RD, Wolraich ML. Stimulant medication therapy in the treatment of children with attention deficit hyperactivity disorder. Pediatric Clinics of North America 1989;36(5):1183‐97.

Storebø 2015b [pers comm]

Storebø OJ, Gluud C. Criticism to "Immediate‐release methylphenidate for attention deficit hyperactivity disorder (ADHD) in adults" [personal communication]. Email to: T Epstein via Wiley Online Feedback form 10 May 2015.

Storebø in press

Storebø OJ, Pedersen N, Krogh H, Ramstad E, Nilausen TD, Skoog M, et al. Methylphenidate for attention deficit hyperactivity disorder (ADHD) in children and adolescents ‐ assessment of harms in observational studies. Cochrane Database of Systematic Reviews in press.

Swanson 1992

Swanson JM. School‐based Assessments and Interventions for ADD Students. Irvine, CA: KC Publishing, 1992.

Swanson 2004a

Swanson JM, Wigal SB, Wigal T, Sonuga‐Barke E, Greenhill LL, Biederman J, et al. A comparison of once‐daily extended‐release methylphenidate formulations in children with attention‐deficit/hyperactivity disorder in the laboratory school (the COMACS Study). Pediatrics 2004;113(3):e206‐16.

Swanson 2006

Swanson JM, Schuck S, Mann M, Carlson C, Hartman K, Sergeant JA, et al. Categorical and dimensional definitions and evaluations of symptoms of ADHD: the SNAP and SWAN Rating Scales. http://bit.ly/1Kj9akF (accessed 4 March 2015).

Swanson 2009

Swanson J. The MTA follow‐up into adolescence: implications for personalized treatment. Oral presentation at The International Society for Research in Child and Adolescent Psychopathology (ISRCAP); 2009 June 19; Seattle. 2009.

Thorlund 2009

Thorlund K, Devereaux PJ, Wetterslev J, Guyatt G, Ioannidis JP, Thabane L, et al. Can trial sequential monitoring boundaries reduce spurious inferences from meta‐analysis?. International Journal of Epidemiology 2009;38(1):276‐86.

Timimi 2004

Timimi S, Taylor E. ADHD is best understood as a cultural construct. British Journal of Psychiatry 2004;184(1):8‐9.

Todd 2008

Todd RD, Huang H, Henderson CA. Poor utility of the age of onset criterion for DSM‐IV attention deficit/hyperactivity disorder: recommendations for DSM‐V and ICD‐11. Journal of Child Psychology and Psychiatry and Allied Disciplines 2008;49(9):942‐9.

Torrance 1982

Torrance GW, Boyle MH, Horwood SP. Application of multi‐attribute utility theory to measure social preferences for health states. Operations Research 1982;30(6):1043‐69.

Tripp 1999

Tripp G, Luk SL, Schaughency EA, Singh R. DSM‐IV and ICD‐10: a comparison of the correlates of ADHD and hyperkinetic disorder. Journal of the American Academy of Child and Adolescent Psychiatry 1999;38(2):156‐64.

Turgay 1994

Turgay A. Disruptive Behavior Disorders: Child and Adolescent Screening and Rating Scales for Children, Adolescents, Parents and Teachers. West Blomfield, Michigan: Integrative Therapy Institute Publication, 1994.

Ullmann 1984

Ullmann RK, Sleator EK, Sprague RL. A new rating scale for diagnosis and monitoring of ADD children. Psychopharmacology Bulletin 1984;20(1):160‐4.

US FDA 2011

US Food, Drug Administration. Communication about an ongoing safety review of stimulant medications used in children with attention‐deficit/hyperactivity disorder (ADHD). http://1.usa.gov/1M5AYoq (accessed 6 May 2011).

Van der Meere 1999b

Van der Meere J, Gunning B, Stemerdink N. The effect of methylphenidate and clonidine on response inhibition and state regulation in children with ADHD. Journal of Child Psychology and Psychiatry and Allied Disciplines 1999;40(2):291‐8.

Van der Oord 2008

Van der Oord S, Prins PJM, Oosterlaan J, Emmelkamp PMG. Efficacy of methylphenidate, psychosocial treatments and their combination in school‐aged children with ADHD: a meta‐analysis. Clinical Psychology Review 2008;28(5):783‐800.

Visser 2014

Visser SN, Danielson ML, Bitsko RH, Holbrook JR, Kogan MD, Ghandour RM, et al. Trends in the parent‐report of health care provider‐diagnosed and medicated attention‐deficit/hyperactivity disorder: United States, 2003‐2011. Journal of the American Academy of Child and Adolescent Psychiatry 2014;53(1):34‐46.e2.

Vitiello 2008

Vitiello B. Understanding the risk of using medications for attention deficit hyperactivity disorder with respect to physical growth and cardiovascular function. Child and Adolescent Psychiatry Clinics of North America 2008;17(2):459‐74.

Wahler 1976

Wahler RG, House AE, Stambaugh EE. Ecological Assessment of Child Problem Behavior: A Clinical Package for Home, School, and Institutional Settings. Oxford: Pergamon, 1976.

Ward 1993

Ward MF, Wender PH, Reimherr FW. The Wender Utah Rating Scale: an aid in the retrospective diagnosis of childhood attention deficit hyperactivity disorder. American Journal of Psychiatry 1993;150(6):885‐90.

Wechsler 1945

Wechsler D. A standardized memory scale for clinical use. Journal of Psychology: Interdisciplinary and Applied 1945;19(1):87‐95.

Wetterslev 2008

Wetterslev J, Thorlund K, Brok J, Gluud C. Trial sequential analysis may establish when firm evidence is reached in cumulative meta‐analysis. Journal of Clinical Epidemiology 2008;61(1):64‐75.

Wetterslev 2009

Wetterslev J, Thorlund K, Brok J, Gluud C. Estimating required information size by quantifying diversity in random‐effects model meta‐analyses. BMC Medical Research Methodology 2009;9:86.

WHO 1992

World Health Organization. The ICD‐10 Classification of Mental and Behavioural Disorders. Clinical Descriptions and Diagnostic Guidelines. Geneva: WHO, 1992.

Wigal 1998

Wigal SB, Gupta S, Guinta D, Swanson JM. Reliability and validity of the SKAMP rating scale in a laboratory school setting. Psychopharmacology Bulletin 1998;34(1):47‐53.

Wilens 2006a

Wilens TE, Gignac M, Swezey A, Monuteaux MC, Biederman J. Characteristics of adolescents and young adults with ADHD who divert or misuse their prescribed medications. Journal of the American Academy of Child and Adolescent Psychiatry 2006;45(4):408‐14.

Wilkinson 2006

Wilkinson GS, Robertson GJ. Wide Range Achievement Test 4. Lutz, FL: Psychological Assessment Resources, 2006.

Willcut 2012

Willcutt EG. The prevalence of DSM‐IV attention‐deficit/hyperactivity disorder: a meta‐analytic review. Neurotherapeutics 2012;9(3):490‐9.

Wolraich 2003

Wolraich ML, Lambert W, Doffing MA, Bickman L, Simmons T, Worley K. Psychometric properties of the Vanderbilt ADHD Diagnostic Parent Rating Scale in a referred population. Journal of Pediatric Psychology 2003;28(8):559‐67.

Wood 2008

Wood L, Egger M, Gluud LL, Schulz KF, Juni P, Altman DG, et al. Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta‐epidemiological study. BMJ 2008;336(7644):601–5.

Woodcock 2001

Woodcock RW, Mcgrew KS, Mather N. Woodcock‐Johnson III Tests of Achievement. Itasca, IL: Riverside Publishers, 2001.

Young 1978

Young RC, Biggs JT, Ziegler VE, Meyer DA. A rating scale for mania: reliability, validity and sensitivity. British Journal of Psychiatry 1978;133(11):429‐35.

Zhang 2005

Zhang S, Faries DE, Vowles M, Michelson D. ADHD Rating Scale IV: psychometric properties from a multinational study as clinician‐administered instrument. International Journal of Methods in Psychiatric Research 2005;14(4):186‐201.

Zito 2000

Zito J, Safer DJ, DosReis S, Gardner JF, Boles M, Lynch FT. Trends in the prescribing of psychotropic medications to preschoolers. JAMA 2000;283(8):1025‐30.

Storebø 2012

Storebø OJ, Rosendal S, Skoog M, Groth C, Bille T, Buch Rasmussen K, et al. Methylphenidate for attention deficit hyperactivity disorder (ADHD) in children and adolescents. Cochrane Database of Systematic Reviews 2012, Issue 5. [DOI: 10.1002/14651858.CD009885]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abikoff 2009

Methods

Eight‐week double‐blind, randomised, placebo‐controlled, cross‐over trial with 2 interventions

  1. OROS methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participant included: 19 (15 boys, 4 girls). Participants randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 19

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV (combined (42%), hyperactive‐impulsive (0%), inattentive (58%))

Age: mean 10.05 years (SD 1.62, range 8 to 13)

IQ: mean 107.1 (SD 14.3)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: ODD (26.3%), anxiety disorder (10.5%), dysthymic disorder (5.3%), conduct disorder (5.3%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV criteria for ADHD (combined or inattentive type)

  2. Meeting dimensional criteria for ADHD symptom severity on Conners' Teacher Rating Scale ‐ Revised, long form, defined as a score ≥ 1.5 SD above age and sex norms

  3. Impaired Organisational, Time Management and Planning treatment (OTMP), defined by a mean total score ≥ 1 SD below the norm on the Children’s Organizational Skills Scale ‐ Parent, and the Children’s Organizational Skills Scale ‐ Teacher

  4. Score ≥ 80 on the Wechsler Abbreviated Scale of Intelligence

Exclusion criteria

  1. Diagnosis of autism, major depression, substance abuse, obsessive‐compulsive disorder, post‐traumatic stress disorder, panic disorder, tic disorders, significant suicidality

  2. Lifetime history of psychosis or mania

  3. Learning disability according to a school individualised educational plan

  4. Taking other CNS medications

Interventions

Participants were randomly assigned to 1 of 2 possible orders of OROS methylphenidate and placebo

Mean methylphenidate dosage: 48.3 mg (range 18 ± 54 mg); weight‐based final OROS methylphenidate dose was 1.3 mg/kg

Administration schedule: not stated

Duration of each medication condition: 4 weeks: 2 weeks titration and 2 weeks optimal dose

Washout before study initiation: 2 days between interventions

Titration period: 2 weeks after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Nolan and Pelham Scale, Fourth Edition, parent‐rated at baseline, after weeks 1 and 2 (during titration) and at end of treatment (4 weeks)

  2. Swanson, Nolan and Pelham Scale, Fourth Edition, teacher‐rated at baseline, after weeks 1 and 2 (during titration) and at end of treatment (4 weeks)

Notes

Sample calculation: no

Ethics approval: Study protocol was reviewed and approved by the University’s institutional review board

Comment from study authors

  1. The 4 weeks of OROS methylphenidate treatment was relatively brief, and post‐treatment measures were obtained after children had been taking their optimal titration dose for 2 weeks

Key conclusion of study authors

  1. OROS methylphenidate reduced children’s OTMP deficits, and these improvements were associated with improvement in ADHD symptoms. Some children remained impaired in OTMP even after effective stimulant treatment for ADHD symptoms. These youngsters may require other treatments that target OTMP deficits

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: December 2013. No supplemental information provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Post‐treatment scores for all 19 study children were obtained from parents. Because 1 child’s treatment was delayed and ran beyond the end of the school year, teacher data on 18 youngsters were analysed

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No protocol published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Investigator‐initiated trial funded by a grant from Ortho‐McNeil Janssen Scientific Affairs to Dr. Abikoff

Conflicts of interest: Drs. Abikoff and Gallagher have a contract with Multi‐Health Systems to further develop the Children’s Organizational Skills Scale (COSS) used in this study. Dr. Abikoff has served on the ADHD Advisory Board of Shire Pharmaceuticals and of Novartis Pharmaceuticals. Dr. Boorady has served on the ADHD Advisory Board and Speakers’ Bureau of Shire Pharmaceuticals. Other study authors report no conflicts of interest

Ahmann 1993

Methods

Three‐week double‐blind, placebo‐controlled, cross‐over trial, in which participants were randomly assigned to

  1. Methylphenidate (low and high doses; Ritalin)

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 234. Participants were randomly assigned to low‐dose methylphenidate (0.3 mg/kg), high‐dose methylphenidate (0.5 mg/kg) or placebo

Number of participants followed up: 206

Number of withdrawals: not stated, but it is described in the text that 4 children experienced severe side effects while taking Ritalin and could not complete the protocol

Regarding the 206 participants

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: 5 to 15 years

IQ: > 70

Sex: 161 boys, 45 girls

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Informed consent from parent

  2. DSM‐III‐R diagnosis of ADHD

In addition, ≥ 3 of the following criteria had to be met

  1. Attention score on the ADD‐H Comprehensive Teacher Rating Scale: at or below 25th percentile

  2. Hyperactivity score on the ADD‐H Comprehensive Teacher Rating Scale: at or below 25th percentile

  3. Inattention/Passivity score on Conners' Teacher Rating Scale (28 items): ≥ 2 SD above the mean

  4. Hyperactivity Index on Conners' Teacher Rating Scale (28 items): ≥ 2 SD above the mean

  5. Hyperactivity Index on Conners' Parent Rating Scale (48 items): ≥ 2 SD above the mean

Children were divided into responders and non‐responders based on the following criteria

  1. Parent reported 1 SD improvement on the Hyperactivity Index of the Conners' Parent Rating Scale (48 items) or gave a positive narrative comment, and

  2. Teacher reported ≥ 2 of the following

    1. 10 percentile improvement in Attention Score on the ADD‐H Comprehensive Teacher Rating Scale

    2. 10 percentile improvement in Hyperactivity Score on the ADD‐H Comprehensive Teacher Rating Scale

    3. 1 SD improvement on the Inattention/Passivity Scale of Conners' Teacher Rating Scale (28 items)

    4. 1 SD improvement on the Hyperactivity Index of Conners' Parent Rating Scale (48 items)

    5. Positive narrative comment

Exclusion criterion

  1. Children with a history of seizures, mental retardation, Tourette’s syndrome or other significant neurological history were not eligible for the study

Interventions

Participants were randomly assigned to different orders of low‐dose Ritalin (0.3 mg/kg), high‐dose Ritalin (0.5 mg/kg) or placebo

Administration schedule: 3 times a day

Duration of each medication condition: 7 consecutive days

Washout before study initiation: not stated

Titration period: not stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADD‐H Comprehensive Teacher Rating Scale (24 items): rated each week

  2. Conners' Teacher Rating Scale (28 items): rated each week

  3. Conners' Parent Rating Scale (48 items): rated each week

Non‐serious adverse events

  1. Barkley Side Effects Questionnaire: parent‐rated, weekly

Notes

Sample calculation:no

Ethics approval: yes; protocol was approved by the Institutional Review Board at the Marshfield Medical Center

Key conclusion of study authors

  1. The Barkley Side Effects Questionnaire proved to be clinically effective in tracking Ritalin side effects and should be incorporated into the routine evaluation and monitoring of ADHD patients for whom stimulants are prescribed

Comment from review authors

  1. Study divided participants into 2 groups: responders and non‐responders. 147 children were determined to be Ritalin responders. Both responders and non‐responders (n = 206) were included in the analysis of side effects

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: not able to find study authors’ contact information; therefore not able to obtain supplemental information regarding study design and data on ADD‐H Comprehensive Teacher Rating Scale, Conners' Teacher Rating Scale, Conners' Parent Rating Scale

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned, but not described how and/or by whom

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind trial, identical appearing pills

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind trial, identical appearing pills

Incomplete outcome data (attrition bias)
All outcomes

Low risk

206 had sufficient data for analyses

Selection bias: Participants were divided into responders and non‐responders. However, data from this article pertain to side effects only and include both groups

Selective reporting (reporting bias)

Unclear risk

Not able to obtain protocol or other information

Vested interest bias

Low risk

Study was funded by Marshfield Clinic grants

Armstrong 2012

Methods

Two identical, concurrently conducted, phase 4, double‐blind, randomised, cross‐over, analogue classroom trials with 2 interventions

  1. OROS methylphenidate

  2. Placebo

Phases

  1. Screening/washout phase: up to 28 days

  2. Titration: up to 6 weeks

  3. Double‐blind assessment period, with the following subperiods

    1. Open‐label OROS methylphenidate

    2. School day 1: OROS methylphenidate or placebo

    3. Open‐label OROS methylphenidate: ≥ 7 days

    4. School day 2: OROS methylphenidate or placebo

Participants

Number of participants screened: not stated

Number of participants included: 78 (55 boys, 23 girls)

Number of participants followed up: 71

Number of withdrawals before randomisation: 7

Number of withdrawals after randomisation: 0

Diagnosis of ADHD: DSM‐IV‐TR (combined (81%), hyperactive‐impulsive (0%), inattentive (19%))

Age: mean 10.1 years (range 9 to 12 years)

IQ: > 80

Methylphenidate naive: not stated

Ethnicity: Caucasian (58%), African American (28%), other (14%)

Country: USA

Setting: out‐patient clinic

Comorbidity: anxiety (0%), depressive disorders (0%), learning disability (32%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. 9 to 12 years of age

  2. DSM‐IV‐TR diagnosis of ADHD

  3. Baseline score on ADHD Rating Scale, Fourth Edition, in the 90th percentile or greater relative to the general population of children of the same age and sex

  4. Participants receiving medication for ADHD at the time of study enrolment exhibited an inadequate response to their then‐current stimulant dose and completed a washout equivalent to 5 half‐lives of the given medication before completing baseline assessments

  5. Attendance at regular school

  6. Ability to read and understand English

  7. To be eligible for the double‐blind, randomised assessment period, participants had to reach their individualised dose of OROS methylphenidate, defined as

    1. Score on the ADHD Rating Scale, Fourth Editiion (as scored by parent or guardian): ≤ 75th percentile for age and sex

    2. Score on the ADHD Rating Scale, Fourth Editiion (as scored by parent or guardian): between 75th and 85th percentiles for age and sex after (1) dose decrease for tolerability (1 dose decrease by 18 mg to a minimum of 18 mg/d was allowed) or (2) having reached dosage of 54 mg/d

Exclusion criteria

  1. History or current diagnosis of epilepsy, severe anxiety or conduct or psychotic disorders

  2. Pervasive developmental, eating, obsessive‐compulsive, sleep, major depressive, bipolar or chronic tic disorder, substance use disorder

  3. Personal or family history of Tourette's syndrome

  4. Known cardiac abnormalities: clinically significant abnormalities in ECG results; family history of sudden death or ventricular arrhythmia

  5. Inability to take or tolerate OROS methylphenidate

  6. Allergies to MPH or other ingredients of OROS methylphenidate

  7. Known gastrointestinal narrowing or significant gastrointestinal problems

  8. Glaucoma

  9. Use of medication with CNS effects (excluding bronchodilators)

  10. Clinically significant laboratory and ECG abnormalities and blood pressure in the 95th percentile or greater for age, sex and height

  11. Study participants were prohibited from using any caffeine‐containing products on study visit days or laboratory assessment days, and were limited to 1 (12‐ounce) caffeinated beverage a day during study participation

  12. Estimated full‐scale IQ 80, as measured by the Wechsler Abbreviated Scale of Intelligence

  13. Scores of ≥ 2 SD less than mean scores for age on the Gray Oral Reading Test (GORT), the Comprehensive Test of Phonological Processing (CTOPP), or the Wechsler Individual Achievement Test ‐ Second Edition (WIAT‐II AB13))

  14. Weight < 3rd percentile for age

  15. History of hospitalisation for treatment of a mood, anxiety or psychotic disorder

  16. History of failed response to methylphenidate

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of OROS methylphenidate and placebo

Mean OROS methylphenidate daily dosage: 40.5 mg

Administration schedule: once daily (morning)

Average duration of OROS methylphenidate treatment: 40 days

Duration of placebo intervention: 1 day

Washout before study initiation: up to 28 days

Medication‐free period between interventions: no

Titration period: before randomisation, up to 6 weeks

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Atkins, M‐Flynn and Pelham Scale, observer‐rated, 4 hours post dose (on the 2 laboratory days)

Serious adverse events

  1. Serious adverse effects assessed on the 2 laboratory days and during the open‐label period

Non‐serious adverse events

  1. Adverse effects, vital signs and body weight, assessed on the 2 laboratory days

Notes

Sample calculation: yes

Ethics approval: yes

Key conclusions of study authors

  1. Wigal 2011 (Armstrong 2012): OROS methylphenidate dose to reduce core symptoms of ADHD to within the normal range also improved performance on a variety of academic tasks in school‐aged children compared with placebo. Reported adverse effects were consistent with those of prior studies

  2. Armstrong 2012 (Armstrong 2012): Robust treatment effect occurred with OROS methylphenidate; onset was at 1 hour post treatment and persisted for ≥ 12.5 hours after dosing

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; a history of failed response to methylphenidate was an exclusion criterion. Only children demonstrating the required decrease in ADHD symptoms with MPH‐OROS within the labelled dosing range were included in the randomised phase of the study. Children who may have required a dose > 54 mg to achieve full therapeutic effect may also have been excluded

Any withdrawals due to adverse events: yes (n = 2)

Email correspondence with study authors: June 2013 to June 2014. Obtained supplemental efficacy data (Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale) and safety data. Awaiting data through the Yale Open Data Access Project

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Wigal 2011; computer‐generated randomisation schedule

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Wigal 2011; blinding of investigators and participants maintained throughout the study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Wigal 2011; blinding of investigators and participants maintained throughout the study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information on incomplete outcome data

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

All outcomes reported according to protocol

Vested interest bias

High risk

Supported by Ortho‐McNeil‐Janssen Scientific Affairs, LLC. Phase IV study

Conflicts of interest: Several study authors had affiliations with pharmaceutical companies producing methylphenidate

Arnold 2004

Methods

Seven‐centre US study consisting of a 6‐week, open‐label, dose‐titration phase (Part A) and a 2‐week, double‐blind, randomised, parallel‐group, placebo‐controlled withdrawal study (Part B) with 2 arms

  1. Dexmethylphenidate

  2. Placebo

Participants

Number of patients screened: 116

Regarding Part A

  1. Number of participants included: 89 (72 boys, 17 girls)

  2. Number of participants followed up: 76

  3. Number of withdrawals: 13

  4. DSM‐IV diagnosis of ADHD (combined 80%)

  5. Age range: 6 to 16 years

  6. IQ: not stated

  7. Methylphenidate naive: 71.9%

  8. Ethnicity: not stated

  9. Country: USA

  10. Comorbidity: not stated

  11. Comedication: not stated

  12. Sociodemographics: not stated

Regarding Part B

  1. Number of participants included: 75 (61 boys, 14 girls)

  2. Number randomly assigned: methylphenidate 35, placebo 40

  3. Number followed up in each arm: methylphenidate 34, placebo 39

  4. Number of withdrawals in each arm: methylphenidate 1, placebo 1

  5. DSM‐IV diagnosis of ADHD (combined (80%), hyperactive‐impulsive (0%), inattentive (20%))

  6. Age range: 6 to 16 years

  7. IQ: > 70 (mean not stated)

  8. MPH‐naive (methylphenidate 82.9%, placebo 62.5%)

  9. Ethnicity: Caucasian (methylphenidate 80%, placebo 75%), African American (methylphenidate 14.3%, placebo 12.5%), Hispanic (methylphenidate 5.7%, placebo 12.5%)

  10. Country: USA

  11. Setting: out‐patient clinic and hospital

  12. Comorbidity: not stated

  13. Comedication: antihistamines, non‐steroidal anti‐inflammatory agents, multi‐vitamins, nasal decongestants or other analgesics or antipyretics (methylphenidate 34.3%, placebo 40.0%)

  14. Sociodemographics: not stated

No significant differences in baseline demographics were noted between the 2 groups. Thus, slightly more treatment‐naive participants were receiving d‐MPH than placebo

Inclusion criteria

  1. 6 to 17 years of age

  2. Enrolled in school

  3. DSM‐IV diagnosis of ADHD, any subtype

  4. Within 30% of normal body weight

  5. Able to participate for the full 8 weeks

Exclusion criteria

  1. History or evidence of cardiovascular, renal, respiratory (other than asthma/allergy), endocrine or immune system disease

  2. History of substance abuse

  3. Hypersensitivity to d,l‐MPH or other stimulants

  4. Treatment with any investigational drug within 30 days of screening

  5. Other significant CNS disorders

  6. Treatment with antidepressants, neuroleptics/antipsychotics, mood stabilisers, anticonvulsants, beta‐blockers, alpha‐2‐agonists, other stimulants, thyroid medications, long‐term oral steroids or sedatives/hypnotics

  7. Concurrent treatment with other psychoactive drug

Interventions

Part A

Dexmethylphenidate dosage: 2.5 to 10 mg, twice daily depending on individual participants' prior medication experience. Children who had received d,l‐MPH began with half their total daily d,l‐MPH dose administered as dexmethylphenidate but not more than 20 mg/d; those who had not previously received d,l‐MPH started d‐MPH at 2.5 mg twice daily

Duration of intervention: 6 weeks

Treatment compliance: not stated

Part B

Participants were randomly assigned to dexmethylphenidate or placebo

Mean methylphenidate dosage: 68.6% of dexmethylphenidate continuers and 79.5% of placebo participants were receiving 20 mg at end of Part B, mean (SD) not stated

Administration schedule: 10 mg twice daily. Time points 7 AM to 8 AM and 11:30 AM to 12 PM

Duration of intervention: 2 weeks

Titration period: 6 weeks, initiated before randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms (Part B)

  1. Swanson, Nolan and Pelham Scale, teacher‐rated at baseline and at end of treatment

  2. Swanson, Nolan and Pelham Scale, parent‐rated at baseline and at end of treatment, 3 and 6 hours post dose

Non‐serious adverse events (Parts A and B)

  1. Monitoring of adverse events and changes from baseline in vital signs (pulse and blood pressure), physical examination and clinical laboratory parameters throughout the study

Notes

Comments from study authors

  1. Limitations

    1. Study design: Treatment effects in such trials may be larger than those seen in unselected populations because, in the randomised withdrawal phase, responders were pre‐selected from the open‐label titration phase to the drug phase.

    2. Another possible limitation is the duration of discontinuation (2 weeks)

Key conclusion of study authors

  1. Dexmethylphenidate is safe, tolerable and effective, with a 6‐hour duration of effect suggested by significant differences from placebo at 6 hours on double‐blind discontinuation

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: October 2013. Supplemental information regarding additional information was received. However, study authors advised us to contact the sponsoring drug company for additional information. This process has been difficult, and no further communication was attempted to request additional information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was central, irrespective of whether the drug was pre‐packaged and pre‐randomised, or if it was bottled and labelled by an unblinded dispenser who had no contact with participants and kept the other staff blind

Allocation concealment (selection bias)

Low risk

Randomisation was central. "In all industry studies I have been involved with, either the drug was pre‐packaged and pre‐randomized or it was bottled and labeled by an unblinded dispenser who had no contact with patients and kept the other staff blind"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. In Part B, participants/guardians and medical personnel were blinded to the drug. Also, d‐methylphenidate was available in tablets, each identical in appearance to a matching placebo. Study drug (or placebo) was dispensed in bottles containing a weekly supply, labelled for use at “Home” and “School”, with the strength designated “A,” “B” or “C”

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind. In Part B, participants/guardians and medical personnel were blinded to the drug

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT sample was used in analysis of efficacy parameters: Participants who received dexmethylphenidate and had a Part B baseline efficacy evaluation and ≥ 1 post‐baseline assessment

Selective reporting (reporting bias)

Low risk

No protocol published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Study was supported by the Celgene Corporation

Conflicts of interest: Drs. Arnold, Wigal and Bohan received research funding from Celgene for the study reported. Dr. Wigal and Dr. West are on the Advisory Panel and Speakers' Bureau for Novartis. Dr. Arnold and Dr. Bohan are on the Speakers' Bureau for Novartis. Dr. Zeldis is Chief Medical Officer and Vice President of Medical Affairs at the Celgene Corporation

Ashare 2010

Methods

Three‐day, double‐blind, randomised, placebo‐controlled medication assessment (cross‐over)

Participants

Number of patients screened: not stated

Number of participants included: 50. Participants were randomly assigned to methylphenidate (low dose, high dose) or placebo

Number of participants followed up: 36 (28 boys, 8 girls)

Number of withdrawals: 14

Diagnosis of ADHD: DSM‐IV (combined (61%), hyperactive‐impulsive (8%), inattentive (31%))

Age: mean 10.5 years (range 9 to 12)

IQ: mean 102 (SD 13)

Methylphenidate naive: 7 participants

Ethnicity: Caucasian (80%), African American (17%), mixed race (3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (47%), conduct disorder (17%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD

  2. 9 to 12 years of age

Exclusion criteria

  1. Full‐scale IQ < 80

  2. History of seizures, neurological disorders and other medical problems

  3. Contraindicating psychostimulant treatment

  4. Current use of non‐ADHD psychotropic medications

  5. History or concurrent diagnosis of pervasive developmental disorder or psychosis, and sensory problems that would make it difficult to complete the task

Interventions

Participants were randomly assigned to 2 doses of long‐acting methylphenidate (OROS methylphenidate; Concerta) once a day and placebo

  1. One (low dose) provided equivalent effects to t.i.d. immediate‐release methylphenidate at 0.3 mg/kg dose, producing a total daily dose of 0.9 mg/kg

  2. The other (high dose) was equivalent to t.i.d. immediate‐release methylphenidate 0.6 mg/kg dose, producing a total daily dose of 1.8 mg/kg

Methylphenidate dosage: low dose: mean 40 mg (SD 9.2); high dose: mean 76.5 mg (SD 13.2)

Administration schedule: once daily, morning, 90 minutes before trial

Duration of trial: 3 days

Washout before study initiation: 24 hours before participation. No washout between interventions. To promote reasonable tolerability of the medication in participants who were stimulant naive or were previously prescribed only very low doses (0.4 mg/kg/d), 13 children were order‐restricted, so they received the 0.3 mg/kg t.i.d.‐equivalent dose before receiving the 0.6 mg/kg t.i.d.‐equivalent dose. Among those who were not order‐restricted, 6 possible drug orders were counterbalanced across participants

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Adverse events were rated daily by camp counsellors and parents using the Pittsburgh Side Effects Rating Scale

  2. Blood pressure was rated daily during time of peak medication effects

  3. Pulse rate was rated daily during time of peak medication effects

Notes

Sample calculation: no information

Ethics approval: yes

Comment from study authors

  1. No child had marked elevation of either cardiovascular parameter

Key conclusion of study authors

  1. Findings of the study fit with the clinical literature on ADHD and the hypothesis that methylphenidate enhances interference control for important environmental stimuli

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: wrote to study authors in July 2014 to ask for data on side effects, pulse and blood pressure; have received no response as yet

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information about randomisation

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind, no further information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing data

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

Supported by grants from the National Institute of Mental Health (NIMH) and from the National Institute on Drug Abuse (NIDA)

Barkley 1989

Methods

Triple‐blind, randomised, cross‐over trial with 3 interventions

  1. Placebo

  2. Methylphenidate low dose (0.30 mg/kg)

  3. Methylphenidate high dose (0.5 mg/kg)

Participants

Number of participants screened: not stated

Number of participants included: 83 (71 boys, 12 girls). Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 80

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐III‐R (subtype distribution not stated)

Age: mean 8.2 years (range 5 to 13)

IQ: mean 105.1

Methylphenidate naive: 85%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: mothers, married (n = 48), divorced (n = 13), unmarried or widowed (n = 13)

Inclusion criteria

  1. 6 to 13 years of age

  2. ADHD according to DSM‐III‐R

  3. Complaints from teacher, parents or both, of significant inattention, overactivity and impulsivity

  4. Appearance of symptoms before 7 years of age

  5. Symptoms for 12 months

  6. Above the 93rd percentile of the hyperactivity scale on parent or teacher report forms of the Child Behavior Checklist (CBCL)

  7. Simple language IQ score > 80 on the Peabody Picture Vocabulary Test ‐ Revised

Exclusion criteria

  1. Gross sensory or motor deficits

  2. Tic disorders, Tourette's syndrome or an immediate family history of such

  3. Seizures

  4. Gross brain damage

  5. Autism

  6. Thought disturbance or schizoid, schizotypal or frank psychotic features

  7. Significant cardiac problems or high blood pressure

  8. Excessive levels of anxiety or fear

  9. Levels of depression that exceed or equal the problems of ADHD

  10. Most participants (n = 74) were subdivided into children placed 2 SD above the normal mean on the Aggressive scale of the parent form of the Child Behavior Checklist (T score > 70) and those who did not have ≥ 2 SD above the normal mean to form aggressive (n = 37) and non‐aggressive subgroups (n = 37). Groups did not differ in age, years of education, maternal age or maternal years of education

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of 0.3 mg/kg methylphenidate, 0.5 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: morning and noon

Duration of each medication condition: 7 to 10 days

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: unused capsules returned to the clinic each week for adherence check

No family was discontinued from the study because of non‐compliance with the drug regimen, defined as more than 1 day of failure to take medication, or 2 missed capsules per week

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale ‐ Revised: parent‐rated at the end of each drug condition

  2. Conners' Teacher Rating Scale ‐ Revised: teacher‐rated at the end of each drug condition

  3. Child Attention Profile: teacher‐rated at the end of each drug condition

General behaviour

  1. Home Situations Questionnaire: parent‐rated at the end of each drug condition (7 to 10 days)

  2. School Situations Questionnaire: teacher‐rated at the end of each drug condition (7 to 10 days)

Adverse effects

  1. Barkley Side Effects Rating Scale: parent‐ and teacher‐rated at the end of each drug condition (7 to 10 days)

Notes

Sample calculation: not stated

Ethics approval: Study was approved by the Human Subjects Committee at the medical centre

Comments from study authors

  1. Limitations

    1. Rates of side effects were based on a large sample that was screened before admission to the drug trial

    2. Low doses of medication to detect side effects

    3. Use of rating scale rather than direct behavioural observation

Key conclusions of study authors

  1. In their drug responding, aggressive and non‐aggressive participants were quite similar. The few exceptions involved measures of conduct, on which aggressive participants were initially rated as more extreme and subsequently showed a greater degree of improvement from medication than non‐aggressive participants

  2. With this dose range, stimulants result in few/mild side effects; systematic monitoring of side effects suggested before/during clinical trials of stimulants

Comment from review authors

  1. The Barkley Side Effects Rating Scale was labelled generically as a “behaviour questionnaire” to disguise its intended use as a monitoring tool for potential side effects. The purpose was to prevent prejudice on the part of respondents, who might potentially distort their ratings if they knew these could be side effects of the medication

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes; 3. One child discontinued because of nervous facial tics, headache and dizziness; a second as the result of excessive thinking and disjointed thinking (during high‐dose methylphenidate); and a third because of headache, dizziness and increased hyperactivity

Email correspondence with study authors: July 2013. We obtained additional information regarding funding and ethics approval. Unfortunately, it was not possible to receive from the study authors supplemental data on ADHD symptoms and general behaviour

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A completely counterbalanced design was used, with participants randomly assigned in relatively equal numbers to 1 of 6 possible drug conditions

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both medication and placebo were crushed and placed within orange opaque gelatin capsules to disguise distinctive differences in flavour between medication and placebo and dose differences across conditions. Children and their parents and teachers, as well as the research assistant evaluating the children, were blinded to medication conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Children and their parents and teachers, as well as the research assistant evaluating the children, were blinded to medication conditions

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information on incomplete outcome data

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no information

Selective reporting (reporting bias)

Low risk

No selective reporting

Vested interest bias

Low risk

Study was internally funded by the medical school

Barkley 1991

Methods

Triple‐blind, placebo‐controlled, cross‐over design with participants randomly assigned to the following conditions

  1. Three doses of methylphenidate (5 mg, 10 mg and 15 mg b.i.d.)

  2. Placebo

Each intervention period lasted 1 week

Participants

Number of participants screened: not stated

Number of participants included: 40. Participants were randomly assigned to 1 of 6 possible drug condition orders (only 6 drug orders, so the highest dose was never given unless preceded by the moderate dose)

Number of participants followed up: 40 (36 boys, 4 girls)

Number of withdrawals: 0

Diagnosis of ADD: DSM‐III‐R (+H: 58%, ‐H: 42%)

Age: mean 8.6 years (range 6 to 12)

IQ: 103.5

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: borderline and low internalising symptoms. No others stated

Comedication: not stated

Sociodemographics: 54.8 on Hollingheads 2 Factor Index

Participants were divided into different categories

  1. Based on type of ADD

    1. ADD with hyperactivity (n = 2)

    2. ADD without hyperactivity (n = 17)

  2. Based on severity of internalising symptoms by maternal ratings on the Internalising scale of the Child Behavior Checklist (CBCL)

    1. 'High internalising group' (> 70) (n = 12)

    2. 'Borderline internalising group' (65 to 70) (n = 17)

    3. 'Low internalising group' (< 65) (n = 11)

Inclusion criteria

  1. Diagnosis of ADD(H) according to DSM‐III‐R

  2. IQ estimate of 80 or higher on a standardised IQ test given within the past year or on the Wechsler Intelligence Scale for Children ‐ Revised, given at study screening

  3. Was the biological child of both current parents or had been adopted by them shortly after birth (within the first year)

  4. No evidence of deafness, blindness, severe language delay, cerebral palsy, epilepsy, autism or psychosis, as established through medical history, parental interview and child play diagnostic interview

Additional criteria for children with combined ADHD

  1. Teacher complaints of short attention span, impulsivity and overactivity as revealed by parent reports

  2. Duration of 6 months for these problems

  3. Age of onset of these problems before 7 years

  4. Score > 93rd percentile on the Inattention and Overactivity scales of the Child Attention Problems Rating Scale

  5. No history of treatment with stimulant drugs, or, if such a history, physician consent to stop taking medication for 48 hours before evaluation in the study

Additional criteria for children with ADD‐H

  1. Same criteria as for children with combined ADHD, with the exception of the fourth criterion. Instead, score > 93rd percentile on the Inattention scale of the Child Attention Problems Rating Scale, but a score < 84th percentile on the Overactivity scale of the Child Attention Problems Rating Scale

Differences regarding the 2 ADHD groups

  1. They differed significantly on child’s IQ score, with the ADD without hyperactivity group scoring significantly lower than the ADD with hyperactivity group. Child IQ was correlated with all of the dependent measures (Measures that we used in this review were not affected by differences in IQ)

Exclusion criteria

  1. History of tics or Tourette's syndrome, given the controversy over whether stimulants may create or exacerbate these conditions

  2. Those with a history of cardiac surgery, high blood pressure or cerebral vascular accident, given the known cardiac pressor effects of stimulants

  3. Those with a history of adverse reactions to a stimulant

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate (5 mg, 10 mg and 15 mg) and placebo

Administration schedule: b.i.d., morning and noon

Duration of each medication condition: 1 week

Washout before study initiation: no

Titration period: none, but highest dose was never given unless preceded by moderate dose

Compliance: Children were permitted to miss 1 day of medication over 7 days and still remain in the study. No families were removed from this study because of non‐compliance as defined in this way

Outcomes

ADHD symptoms

  1. Parent‐rated

    1. Attention Deficit Hyperactivity Disorder Rating Scale: rated at the end of each drug condition order

    2. Home Situations Questionnaire: number of problem settings and mean severity of problems, rated at the end of each drug condition order

  2. Teacher‐rated (teachers completed questionnaires at the conclusion of each medication condition)

    1. Self Control Rating Scale

    2. Child Attention Problems

    3. School Situations Questionnaire: number of problem settings and mean severity of problems

Non‐serious adverse events

  1. Home Side Effects Rating Scale: parent‐rated at the end of each drug condition order

  2. School Side Effects Rating Scale: parent‐rated at the end of each drug condition order

Notes

Sample calculation: no

Ethics approval: Study was approved by the Institutional Review Board at the medical centre

Key conclusion of study authors

  1. This study indicates that ADHD, inattentive type, and ADHD, combined type, do not show dramatic differences in their manner of responding to methylphenidate across 3 dose levels (5 mg, 10 mg and 15 mg), with both groups displaying generally positive drug responses. However, more children with ADHD, inattentive type, had minimal or no response or did best on the low dose of medication, whereas the vast majority of children with ADHD, combined type, showed a positive response, primarily to moderate to high doses of methylphenidate

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; excluded those with history of adverse reactions, but included both naive and prior users of antipsychotics

Email correspondence with study author: 18 January 2013. Dr. Barkley informed us that data from the study on side effects, for example, are no longer available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to 1 of 6 possible drug conditions

Allocation concealment (selection bias)

Low risk

Hospital pharmacy prepared placebo (lactose powder) and methylphenidate by crushing and placing them into 6 orange opaque gelatin capsules

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Triple‐blind design

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Triple‐blind design

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No incomplete outcome data

Selection bias: no

Selective reporting (reporting bias)

Unclear risk

Protocol not available

Vested interest bias

Low risk

Research was supported by the National Institute of Mental Health (NIMH)

Conflicts of interest: no information

Barkley 2000

Methods

Double‐blind, placebo‐controlled, within‐participant, cross‐over trial with 3 interventions

  1. Methylphenidate: 5 mg, 10 mg, twice daily

  2. Amphetamine and dextroamphetamine mixed salts (Adderall): 5 mg, 10 mg, twice daily

  3. Placebo

Phases: 5, but high doses of each stimulant always followed lower dose of the same stimulant

Participants

Number of participants screened: 46

Number of participants included: 38. Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 35 (30 boys, 5 girls)

Number of withdrawals: 2. One was a post hoc exclusion

Diagnosis of ADHD: DSM‐IV (subtype not described)

Age: mean 14 years (range 12 to 17)

IQ: mean 103.9 (range 80 to 141)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. Adolescents, 12 to 17 years of age, with a DSM‐IV diagnosis of ADHD

Exclusion criteria

  1. History of motor or vocal tics, Tourette’s syndrome, cardiac surgery, high blood pressure, cerebral vascular accident, hyperthyroidism or pregnancy or lactation

  2. Adverse reactions to stimulant medications

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of 5 mg methylphenidate followed by 10 mg methylphenidate, 5 mg Adderall followed by 10 mg Adderall and placebo

  1. 5 mg, 10 mg methylphenidate; placebo; 5 mg, 10 mg Adderall

  2. 5 mg, 10 mg Adderall; placebo; 5 mg, 10 mg methylphenidate

  3. Placebo; 5 mg, 10 mg methylphenidate; 5 mg, 10 mg Adderall

  4. 5 mg, 10 mg Adderall; 5 mg, 10 mg methylphenidate; placebo

Mean methylphenidate dosage: low dose 10 mg/d; high dose 20 mg/d

Administration schedule: AM and midday

Duration of each medication condition: 1 week

Washout before study initiation: none

Medication‐free period between interventions: none

Titration period: none, although 5 mg dose was given before 10 mg, initiated after randomisation

Treatment compliance: Parents were required to return all unused capsules, but nothing further was said about this

Outcomes

ADHD symptoms

  1. Parent‐ and teacher‐rated ADHD/ODD Rating Scales: completed over previous treatment week

Non‐serious adverse events

  1. Barkley Side Effects Rating Scale: completed by adolescent, parent and teacher at the end of each treatment week

Notes

Sample calculation: not described

Ethics approval: yes

Comments from study authors

  1. Teens are more independent of their parents than are younger children, spending more time outside parental supervision. This raises serious questions about the sensitivity of parental reports to drug and dose response

  2. Limitations: Most noteworthy was the poor co‐operation of teachers. As a consequence, the statistical power of the study to detect drug effects on these measures, often the most sensitive to stimulant drug effects, was greatly reduced

Key conclusion of study authors

  1. In conclusion, the present study suggested that both Adderall and methylphenidate may have been clinically effective in the management of teens with ADHD when non‐blinded, global clinical judgements of improvement, which were based on multiple sources of information, were used. Even so, these positive drug responses could not be documented at the group level of statistical analysis by using more specific and systematic ratings by parents and teachers. Clinicians undertaking stimulant trials in such contexts need to be aware of the many challenges to the internal validity of these procedures that are likely to occur in drug trials with teens

Comment from review authors

  1. To exclude participants with low IQ was a post hoc decision

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; excluded patients who had a history of adverse events to stimulants

Email correspondence with study authors: January 2014. We received additional information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Email correspondence with study author: “Randomization was done by me as best as I can recall” (Krogh 2014a [pers comm])

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Opaque gelatin capsules were prepared by the pharmacist

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Completer analysis reported 11/46 teens LTFU, 15 parents LTFU, 33 English teachers and 31 Maths teachers LTFU

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Email correspondence with study author: "All planned analyses were done and all measures we collected as treatment endpoints were analyzed" (Krogh 2014a [pers comm])

Vested interest bias

Low risk

University of Massachusetts Medical School

Bedard 2008

Methods

Four‐day randomised, double‐blind, placebo‐controlled, cross‐over trial with 2 interventions in 2 groups

Interventions

  1. Methylphenidate

  2. Placebo

Groups

  1. ADHD with anxiety

  2. ADHD without anxiety

Participants

Number of participants screened: not stated

Number of participants included: 130. Participants were randomly assigned to 1 of 11 possible drug condition orders

Number of participants followed up: 130 (110 boys, 20 girls)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (63%), hyperactive‐impulsive (30%), inattentive (6%))

Age: mean 9 years (SD 1.46, range 6 to 12)

IQ: mean 104.11

Methylphenidate naive: 70%

Ethnicity: Caucasian (90%)

Country: Canada

Setting: out‐patient clinic

Comorbidity: specific learning disorder (34%), conduct disorder (24%), oppositional defiant disorder (26%), generalised anxiety disorder (17%), separation anxiety disorder (11%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. DSM‐IV diagnosis via clinical diagnostic assessment: confirmed by Parental Interview for Child Symptoms (PICS) and Teacher Telephone Interview (TTI)

Exclusion criteria

  1. IQ < 80

  2. Evidence of neurological dysfunction, poor physical health or uncorrected sensory impairments

  3. History of psychosis based on physician enquiry

  4. Primary language spoken at home not English

Interventions

Participants were randomly assigned to 1 of 11 possible drug condition orders of methylphenidate and placebo. Children weighing < 25 kg received 5 mg, 10 mg and 15 mg of methylphenidate; children weighing ≥ 25 kg received 10 mg, 15 mg and 20 mg of methylphenidate

Mean methylphenidate dosage: 0.28 mg/kg, 0.45 mg/kg, 0.61 mg/kg

Administration schedule: not stated

Duration of each medication condition: 1 day

Washout before study initiation: none

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Iowa‐Conners’ Rating Scale. Scale was completed by the examiner at the end of each session

Notes

Sample calculation: no

Ethics approval: approved by the institutional ethics review board

Comment from study authors

  1. We cannot predict that similar results would hold with longer‐term treatment or with extended‐release preparations

Key conclusions of study authors

  1. Findings provide insight into potential mechanisms underlying individual differences in treatment response in ADHD, which may facilitate more targeted treatments

  2. Results from the present study demonstrate that methylphenidate produces moderate but beneficial effects on selected aspects of working memory that are known to be impaired in ADHD. Furthermore, comorbid anxiety may be a predictor of working memory treatment response

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: December 2013. Not able to get supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Master randomisation tables were prepared by the research support pharmacist at the hospital by using simple randomisation with restrictions (high dose not to be given on the first possible drug day nor immediately following placebo; no directly ascending or descending dose order). Therefore, a balanced block 22 design was used

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Examiner, psychiatrist, participant and participant’s family were not informed about participant’s randomisation order or daily medication status until completion. Placebo and active medication were prepared by the hospital pharmacist and were powdered and packaged in an opaque capsule to prevent identification of contents by colour, taste or volume

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trained clinicians, blinded to other aspects of the participant’s assessment, conducted interviews independently

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Low risk

Funding and operating grant from the Canadian Institute of Health Research and funding from the Canada Research Chairs Programme

Conflicts of interest: none

Ben 2002

Methods

Two‐week, double‐blind, placebo‐controlled, cross‐over trial with 1 intervention

  1. Methylphenidate 0.5 mg/kg/d

  2. Placebo

The purpose of the study was to investigate the relationship between SLC6A3 3’ end variable number tandem repeat (VNTR) polymorphism and behavioural/therapeutic response to methylphenidate

Phases: not stated

Participants

Number of participants screened: not stated

Number of participants included: 42 (all boys). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.2 ± 1.8 years (range not stated)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: Canada

Setting: not stated

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. Not stated

Exclusion criterion

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate (0.5 mg/kg/d) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: not stated

Time points: not stated

Duration of each medication condition: not stated

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: not stated. No information

Treatment compliance: not stated

Outcomes

General behaviour

  1. Conners’ Continuous Performance Test Overall Index: before and 45 minutes after administration of placebo or methylphenidate

  2. Restricted Academic Situation Scale: before and 45 minutes after administration of placebo or methylphenidate

  3. Conners’ Global Index Scale: parent and teacher reports, time points not stated

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusion of study authors

  1. Although preliminary, these results suggest that the SLC6A3 3’ end variable number tandem repeat may modulate therapeutic response to methylphenidate as evaluated by teachers

Comments from review authors

  1. Abstract only. Abstract includes no raw information on clinical effectiveness; includes mostly details of the relationship gene/methylphenidate effect

  2. No information on adverse events was provided in this abstract

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated

Any withdrawals due to adverse events: not stated

Email correspondence with study authors: October 2013 and April 2014. We wrote to the study authors twice to request supplementary information on data. No reply was received. Therefore we have no useable data from this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

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

Inadequate information contained in the abstract

Selection bias: not stated

Selective reporting (reporting bias)

Unclear risk

Protocol not available

Vested interest bias

Unclear risk

Not stated

Biederman 2003

Methods

Fifteen‐site, multi‐centre, double‐blind, randomised, 2‐week parallel trial with 2 arms

  1. Extended‐release methylphenidate (Ritalin LA)

  2. Placebo

Phases: 3. Pre‐randomisation (4 weeks titration plus 1 week washout), randomisation, double‐blind treatment and open‐label extension

Participants

Number of participants screened: unknown

Number of participants included: 164

Number of participants titrated: 161 (122 boys, 39 girls)

Number of participants randomly assigned: methylphenidate 66, placebo 71

Number of participants followed up: methylphenidate 63, placebo 71

Number of withdrawals: methylphenidate 3, placebo 0
Diagnosis of ADHD: DSM‐IV (combined (75.8%), hyperactive‐impulsive (1.2%), inattentive (18.6 %))

Age: mean 8.81 years (range 6 to 14)

IQ: not stated

Methylphenidate naive: 94 (58.4%)

Ethnicity: Caucasian (85.7%), African American (3.7%), Asian (1.2%), other (9.3%)

Country: USA and Canada

Setting: out‐patient clinic (naturalistic school setting)

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated. No significant difference in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Boys and girls 6 to 14 years of age who met DSM‐IV criteria for ADHD and were receiving treatment with methylphenidate, or de novo patients

  2. Meeting ADHD criteria in the structured diagnostic interview (National Institute of Mental Health Diagnostic Interview Schedule for Children, Fourth Edition)

  3. Attending school in a classroom setting with the same teacher, who, for the duration of the study, would perform weekly assessments

  4. Functioning, in the opinion of the investigator, at age‐appropriate levels academically

  5. Female patients of childbearing age needed to have a negative pregnancy test and if sexually active had to be using adequate and reliable contraception for the duration of the study

  6. Ongoing behavioural therapies for ADHD were permitted to continue, but participants were not to initiate behavioural therapy during the trial

  7. Informed consent

Exclusion criteria

  1. Patients with somatic or psychiatric disorders that could contraindicate treatment or confound efficacy or safety assessments, or those who required treatment with drugs other than methylphenidate

  2. Known hypersensitivity to the study drug

  3. Likelihood of non‐compliance

  4. History of substance abuse

  5. Living with a person with a substance abuse disorder

  6. Pregnancy

  7. Use of other investigational drugs during the study period

Interventions

Participants were randomly assigned to extended‐release methylphenidate or placebo

Mean methylphenidate dosage: not stated

Administration schedule: once daily in the morning

Duration of intervention: 2 weeks (mean: methylphenidate 13.91, placebo 13.96)

Titration period: 1 week before randomisation
Treatment compliance: 130 completed treatment (methylphenidate 61, placebo 69)

Outcomes

ADHD symptoms

  1. Primary efficacy outcome measure

    1. Conners’ ADHD/DSM‐IV Scale, Teachers: Total subscale, school day weekly assessments

  2. Secondary efficacy measure

    1. Conners’ ADHD/DSM‐IV Scale, Teachers: Inattentive subscale

    2. Conners’ ADHD/DSM‐IV Scale, Teachers: Hyperactive‐Impulsive subscale

    3. Conners’ ADHD/DSM‐IV Scale, Parents: Total subscale, weekends

    4. Conners’ ADHD/DSM‐IV Scale, Parents: Inattentive subscale

    5. Conners’ ADHD/DSM‐IV Scale, Parents: Hyperactive‐Impulsive subscale

Non‐serious adverse events

  1. Monitoring of adverse events

  2. Routine laboratory tests (haematology, blood chemistry, urine)

  3. Vital signs (sitting blood pressure and pulse)

  4. Height, weight and performance of physical examinations and drug screening

Notes

Sample calculation: yes. A total of 128 participants (n = 64 per treatment group) were required for analysis of the primary efficacy variable, based on an effect size of 0.5 with a power of 80% and a 2‐tailed α‐level of 0.05

Ethics approval: yes. An institutional review board approved this study at each participating site

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Any withdrawals due to adverse events: yes

Comment from study authors

  1. Limitations: First, participants in this study were primarily male Caucasians with ADHD. Second, the short study duration does not predict long‐term efficacy or safety. Third, selection of responders during the titration phase may have affected the outcomes of this study. In clinical practice, response rate may be lower than that observed among the current sample

Key conclusion of study authors

  1. Results demonstrate that extended‐release methylphenidate (Ritalin LA) administered once daily for up to 2 weeks achieved outcomes statistically superior to placebo in children with ADHD

Comments from review authors

  1. Mean methylphenidate dosage not stated, only range (10 mg/d to 40 mg/d)

  2. IQ not stated

Email correspondence with study authors. April 2014. Emailed study authors for additional information/data but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed by Novartis Drug Supply Management, which used a validated system that automates random assignment of treatment groups to randomisation numbers

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not clear whether investigator was blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not clear whether investigator was blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

LOCF analysis of ITT population. ITT population included all participants who received the double‐blind study drug, and from whom ≥ 1 Conners’ ADHD/DSM‐IV Scale, Teachers, was obtained

Selective reporting (reporting bias)

Low risk

No protocol was published. Outcomes of interest have been reported

Vested interest bias

High risk

Funding was received from Novartis

Bliznakova 2007

Methods

Eleven‐day N‐of‐1 randomised, double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 1 boy

Number included: 1. The participant was randomly assigned to methylphenidate and placebo across the 11 days

Number of participants followed up: 1

Number of withdrawals: 1

Diagnosis of ADHD: ICD‐10 (predominantly hyperactive type)

Age: 15 years

IQ: not stated

Methylphenidate naive: no

Ethnicity: not stated

Country: Germany

Setting: not stated

Comorbidity: not stated

Comedication: not stated

Sociodemographics: 2 parents

Interventions

The participant was randomly assigned to methylphenidate and placebo across 11 days

Mean methylphenidate dosage: not stated

Administration schedule: not stated

Duration of each medication condition: The condition was changed daily, but placebo was given for 6 days and methylphenidate for 5 days

Washout before study initiation: not relevant

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: 100% according to Table 6

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale: rated daily

  2. Parent/Teacher Rating Scale: rated daily

Non‐serious adverse events

  1. Assessment of stomachaches (yes/no) daily

Notes

Sample calculation: yes

Ethics approval: not stated

Key conclusion of study authors

  1. Double‐blind study showed significant symptom reduction under the medication condition, which was also noted by the participant himself. Furthermore, towards the end of the study, the somatic complaints were gone

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: December 2013. Emailed study author to request information about missing data but received no response. Not possible to use data from this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: not stated

Blum 2011

Methods

This double‐blind, placebo‐controlled, cross‐over trial with the child's clinically most effective dose as identified by a systematic open‐label titration procedure investigated whether components of attention and executive functioning improve when children with ADHD are treated with OROS methylphenidate

Two‐week, cross‐over trial with 2 interventions

  1. OROS methylphenidate

  2. Placebo

Participants

Number of participants screened: 41

Number of participants included: 34. Participants were assigned to OROS methylphenidate and placebo in random order

Number of participants followed up: 30 (24 boys, 6 girls)

Number of withdrawals: 4

Diagnosis of ADHD: DSM‐IV TR (combined (100%))

Age: mean 8 years 6 months (range 6 years 5 months to 12 years 6 months)

IQ: mean 97.8 (range 77 to 132)

Methylphenidate naive: number not stated

Ethnicity: Caucasian (80%), African American (13.3%), other (6.7%)

Country: USA

Setting: out‐patient clinic

Comorbidity type: oppositional defiant disorder (40%), specific learning difficulty (33.3%), anxiety (6.67%), dysthymia (3.3%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children 6 to 12 years of age, in first grade or higher

  2. DSM‐IV‐TR diagnosis of ADHD combined type

  3. Parent and teacher ratings on the ADHD Rating Scale, Fourth Edition, of at least the 85th centile for hyperactivity/impulsivity and inattention, or both (if the child was not taking medication at enrolment)

  4. IQ > 75 on the Wechsler Abbreviated Scale of Intelligence

Exclusion criteria

  1. Children with a past or current diagnosis of a chronic tic disorder, a pervasive developmental disorder, cerebral palsy, bipolar disorder, major depression, head injury requiring hospitalisation, psychotic disorder, glaucoma, cardiovascular disease, epilepsy, obsessive‐compulsive disorder serious enough to warrant separate treatment or suicidal or homicidal behaviour or ideation

  2. History of side effects with methylphenidate requiring discontinuation of the medication

  3. Children were also excluded if they were known to be unable to swallow a tablet

  4. Use within 14 days of a monoamine oxidase inhibitor

  5. Long‐term treatment with coumarin, clonidine or tricyclic antidepressants

Interventions

Participants were randomly assigned to OROS methylphenidate and placebo

Methylphenidate dosage: 9 children treated with 18 mg, 13 with 36 mg and eight with 54 mg of OROS methylphenidate

Administration schedule: not stated

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: 2‐ to 3‐week open‐label, multi‐dose‐titration protocol to determine the child’s optimal dose as recommended by practice guidelines

Treatment compliance: 30 children completed the study; however, compliance regarding study medication is not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition, both parent‐ and teacher‐rated: rated at the end of each week of the different medication trial

Non‐serious adverse events

  1. Stimulant Drug Side Effects Rating Scale, parent‐rated: rated at the end of each week of the different medication trial. These data are not reported in the article

Notes

Sample calculation: not stated

Ethics approval: yes; approved by the Committee for the Protection of Human Subjects at The Children’s Hospital of Philadelphia

Comment from study authors

  1. In conducting analyses for the cross‐over study, our team examined the effects of crossing participants from the first treatment condition to the second treatment condition (i.e. the carry‐over effect).(.....) Because no statistically significant sequence effects were noted, data from both periods were combined, and final analyses were reduced to paired comparisons

Key conclusion of study authors

  1. When OROS methylphenidate was used to treat children with ADHD at the clinically most effective dose, general improvement was noted on tasks requiring response inhibition; response to treatment in other domains was variable or was not demonstrated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: January 2014: Emailed study authors twice to request additional information but received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned and counterbalanced across participants

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Referred to as double‐blind but no information provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Referred to as double‐blind but no information provided

Incomplete outcome data (attrition bias)
All outcomes

High risk

Completer analysis reported

Selective reporting (reporting bias)

Low risk

Clinical trial ID: NCT00530257

Vested interest bias

High risk

Study was supported by an investigator‐initiated grant from Ortho McNeil Janssen Scientific Affairs, the manufacturer of OROS methylphenidate (Concerta)

Borcherding 1990

Methods

Eleven‐week, double‐blind, placebo‐controlled, cross‐over study with 3 interventions

  1. Methylphenidate

  2. Dextroamphetamine

  3. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 46 (all boys). Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: 45

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 8.6 years (range 6 to 12)

IQ: mean 106.1 (range > 80)

Methylphenidate naive: 13 have not received past stimulant treatment

Ethnicity: Caucasian (72%), African American (22%), Asian/Hispanic (6%)

Country: USA

Setting: out‐patient clinic

Comorbidity: medically healthy

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Medically healthy

  2. Full‐scale IQ score > 80 on the Wechsler Intelligence Scale

  3. Score 2 SD or above age norms on Factor 4 (Hyperactivity) of Conners' Teacher Rating Scale

Exclusion criteria

  1. Medical or neurological disease, including chronic motor tics or Tourette’s syndrome

  2. Other primary Axis I psychiatric disorders

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of methylphenidate, dextroamphetamine and placebo

Mean methylphenidate dosage: 1.3 mg/kg

Administration schedule: twice daily, 9:00 AM and 1:00 PM

Duration of each medication condition: 3 weeks

Washout before study initiation: 2 weeks

Medication‐free period between interventions: none

Titration period: During the 3 weeks, low dose was given week 1, intermediate dose week 2 and high dose week 3

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Subject Treatment Emergent Symptom Scale completed weekly by the physician and the child’s parents; reflected both symptoms and observed effects

  2. Attention given to onset and duration of abnormal movements or obsessive‐compulsive behaviours. Collected from several sources

Notes

Sample calculation: no

Ethics approval: not stated

Comment from study authors

  1. Most movements and compulsive behaviours were seen only by staff sensitive to these possible effects

Key conclusions of study authors

  1. Dextroamphetamine tended to produce more compulsive behaviours, which were also more likely to resemble obsessive‐compulsive disorder, than did methylphenidate

  2. Abnormal movements and compulsive behaviours tended to co‐occur with methylphenidate only; no general Tourette's‐obsessive‐compulsive disorder diathesis was found for this population

  3. An important clinical point from these data is that abnormal movements and compulsive behaviour due to treatment with 1 stimulant should not necessarily be a cause for discontinuation of stimulant drug treatment; rather, the same stimulant at a different dose or a different stimulant should be tried

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Double‐blind random fashion

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Oral medication in identical capsules was administered at 9:00 AM and 1:00 PM

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants except 1 (who experienced adverse events) completed the study

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol; however, all outcomes stated in the Methods have been reported

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: not stated

Brams 2008

Methods

Randomised, double‐blind, cross‐over, multi‐centre study evaluating the efficacy of the following over an 8‐hour laboratory classroom day in children with ADHD

  1. Extended‐release dexmethylphenidate (20 mg/d)

  2. Placebo

Phases

  1. Baseline: day 0

  2. Period 1: days 1 to 7 (Sunday to Saturday), when Saturday is assessment day (8‐hour laboratory classroom day)

  3. Period 2: days 7 to 15

  4. Final visit: day 15

Participants

Number of participants screened: 92

Number of participants included: 86 (53 boys, 33 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 86

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (87.2%), hyperactive‐impulsive (0%), inattentive (12.8%))

Age: mean 9.5 years (range 6 to 12)

IQ: > 70

Methylphenidate naive: 0%

Ethnicity: Caucasian (48.8%), African American (24.4%), Asian (2.3%), Hispanic (23.3%)

Country: USA

Setting: multi‐centre, out‐patient clinic

Comorbidity: not stated

Comedication: no antidepressant or other antipsychotic medication

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV criteria for a diagnosis of ADHD of any type, as established by the Kiddie Schedule for Affective Disorders and Schizophrenia‐Present and Lifetime Versions

  2. 6 to 12 years of age

  3. Only children whose parents or legal guardians, or both, provided written informed consent before any study‐related procedures were performed were enrolled

  4. Females of child‐bearing potential were required to have a negative urine pregnancy test before enrolment and, if sexually active, to be using adequate and reliable contraception (e.g. double‐barrier method), which was documented in the medical record

Exclusion criteria

  1. Children or their parents/guardians were unable to understand or follow instructions as needed to participate in the study

  2. Children deemed by investigators to have below‐average cognitive capacity, or to be home‐schooled

  3. Previously diagnosed with Gilles de la Tourette's syndrome or a tic disorder (medication‐induced tics were not excluded)

  4. History of seizure disorder, or history of, or concurrent, significant medical or psychiatric illness or substance abuse disorder

  5. Taking an antidepressant or other antipsychotic medication; those who initiated psychotherapy within the 3 months preceding screening and those with a positive urine drug screen were deemed ineligible

  6. Poor response or known sensitivity to all methylphenidate or dexmethylphenidate formulations based on past medical history

  7. Taking other medications for ADHD

  8. Prospective participants taking or planning to take any other investigational drug within 30 days of the start of the study

  9. Previously participated in an analogue classroom study within 6 months before screening.

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of once‐daily, extended‐release dexmethylphenidate 20 mg (Focalin XR (Novartis Pharmaceuticals Corporation) and placebo

Mean methylphenidate dosage: 20 mg

Administration schedule: once daily in the morning

Duration of each medication condition: 7 days

Washout before study initiation: 1 week before the study

Titration period: Before study participation, all participants were stabilised on a total daily dose or nearest equivalent dose of methylphenidate 40 mg to 60 mg or dexmethylphenidate 20 mg to 30 mg for ≥ 2 weeks before screening

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Primary efficacy outcome

    1. Change on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: combined score from pre‐dose to the 0.5‐hour post‐dose time point during the 8‐hour classroom day. Rated by observer

    2. Change in Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: combined scores from pre‐dose to 1, 2, 4, 6 and 8 hours post dose. Rated on classroom day by observers

    3. Change from pre‐dose on Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: Attention and Deportment scores at all time points (0.5, 1, 2, 4, 6 and 8 hours post dose)

    4. Conners’ ADHD/DSM‐IV Scales, Parent: completed by parent/legal guardian on classroom day

Non‐serious adverse events

  1. Vital signs: recorded at each visit (days 0, 7, 14)

  2. Spontaneously reported adverse events, including serious adverse events, at the end of each treatment period

  3. Heart rate and blood pressure measured at pre‐dose, 4 and 8 hours post dose at days 7 and 14

  4. Weight at screening and at day 15

  5. ECG at screening and at day 15

Notes

Sample calculation: yes

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; children with poor response or known sensitivity to methylphenidate or dexmethylphenidate were excluded

Comments from study authors

  1. Several limitations with regard to the design of this study should be considered in interpretation of these data

  2. Participant population was required to have had previous exposure to methylphenidate or dexmethylphenidate. It is likely that this patient population already demonstrated a therapeutic response to methylphenidate or dexmethylphenidate, as well as tolerance to these drugs

  3. Only a fixed dosage of 20 mg/d was evaluated, making it difficult to compare results observed in the current study with those of other doses currently available, or when treatment is optimised

Key conclusions of study authors

  1. Compared with placebo, once‐daily, extended‐release dexmethylphenidate 20 mg provided rapid and significant improvement at 0.5 hours post dose in attention, deportment and academic performance, which was sustained for 8 hours post dose

  2. Overall, once‐daily extended‐release dexmethylphenidate 20 mg was well tolerated

  3. In an analysis of parental assessment of diary responses, children appeared better organised, and morning preparation for school was smoother and less frustrating, with once‐daily extended‐release dexmethylphenidate compared with placebo

Email correspondence with study authors: September 2013. We received an email from Dr. Brams, in which we were told that Novartis had control and ownership of study data. Consequently, we had to contact the Public Affairs Department at Novartis to request the information (e.g. protocols) (Krogh 2013a [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation list was generated by the study sponsor, who used an automated random assignment of treatment sequences to randomisation numbers in the specified ratio

Allocation concealment (selection bias)

Low risk

All study medications and packaging were identical in appearance for blinding purposes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, parents, study centre personnel and those who assessed outcomes were blinded to study treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants, parents, study centre personnel and those who assessed outcomes were blinded to study treatment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The safety population consisted of all participants who took ≥ 1 dose of study medication. The efficacy population included all randomly assigned participants who provided valid efficacy measurements for both treatment periods

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

High risk

Sponsored by Novartis Pharmaceuticals Corporation

Conflicts of interest: First study author has been a speaker, consultant and advisory board member for Novartis and Shire

Brams 2012

Methods

Randomised, double‐blind, 3‐period × 3‐treatment cross‐over study in a 12‐hour laboratory classroom setting with 3 interventions

  1. 20 mg extended‐release dexmethylphenidate

  2. 30 mg extended‐release dexmethylphenidate

  3. Placebo

Each period lasted 7 days

Participants

Number of participants screened: not stated

Number of participants included: 165. Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 157

Number of withdrawals: 8

Diagnosis of ADHD: DSM‐IV (combined or predominantly hyperactive‐impulsive subtype)

Age: mean 9.6 years (range 9.3 to 10.0)

IQ: above normal

Sex: 57% boys, 43% girls

Methylphenidate naive: 0%

Ethnicity: Caucasian (38.2%), African American (31.5%), Hispanic (22.4%), other (7.9%)

Country: USA

Setting: out‐patient clinic (laboratory classroom)

Comorbidity: no significant medical illness

Comedication: no information

Sociodemographics: no information

Inclusion criteria

  1. Males and females 6 to 12 years of age

  2. Meeting DSM‐IV criteria for primary diagnosis of ADHD combined subtype or predominantly hyperactive‐impulsive subtype

  3. Female of childbearing potential required to have a negative urine pregnancy test before enrolment and, if sexually active, to use adequate and reliable contraception

  4. Stabilised on a total daily dose or nearest equivalent dose of 40 mg to 60 mg of methylphenidate or 20 mg to 30 mg dexmethylphenidate (36 mg and 54 mg of extended‐release methylphenidate and 10 mg to 20 mg of transdermal methylphenidate were allowed) for ≥ 2 weeks before screening

Exclusion criteria

  1. Children or their parents/guardians were unable to understand or follow instructions necessary to responsibly participate in the study

  2. Children were deemed by the investigator to have below average cognitive ability

  3. Home‐schooled

  4. Previously diagnosed with Gilles de la Tourette's disorder or similar tic disorder (medication‐induced tics were not excluded)

  5. History of a seizure disorder

  6. History of or concurrent long QT syndrome or QTc > 450 milliseconds at screening, or any clinically significant ECG abnormality

  7. Significant medical or psychiatric illness or substance abuse disorder

  8. Children were taking an antidepressant or other antipsychotic medications

  9. Children initiated psychotherapy within the 3 months before screening

  10. Positive urine drug screen

  11. Children with a poor prior response, or known sensitivity, to all methylphenidate or D‐MPH products, based on medical history

  12. Children currently taking non‐methylphenidate‐based medications for ADHD

  13. Those taking or planning to take any other investigational drug within 30 days of study start

  14. Children who had previously participated in an analogue classroom study within 6 months before screening

  15. Alanine aminotransferase, aspartate aminotransferase, gamma glutamyl transferase or serum creatinine > 2× the upper limit of normal at screening

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of 20 mg extended‐release dexmethylphenidate, 30 mg extended‐release dexmethylphenidate and placebo

Administration schedule: once daily, morning

Duration of each medication condition: 7 days

Washout before study initiation: 1 week

Medication‐free period between interventions: no

Titration period: none (fixed doses)

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale (‐combined, ‐attention and ‐deportment): performed by independent blinded raters throughout the 12‐hour testing period

Serious adverse events

  1. Spontaneously reported serious adverse events were recorded weekly

One participant experienced 2 serious adverse events (peritonsillar abscess and oral bullae) while receiving 20 mg extended‐release dexmethylphenidate and was hospitalised for 6 days for the peritonsillar abscess. Serious adverse events were considered not related to study drug. Participant discontinued the study for missed study drug during hospitalisation

Non‐serious adverse events

  1. Vital signs were assessed, and spontaneously reported adverse effects were recorded weekly

  2. Heart rate and blood pressure were measured after weeks 1 and 2

  3. Weight was measured and ECG tests were conducted at screening and at the final visit

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; children with a poor prior response, or known sensitivity, to all methylphenidate or dexmethylphenidate products based on medical history were excluded

Comments from study authors (limitations)

  1. Limited exposure to both doses of extended‐release dexmethylphenidate for each participant to 1 week

  2. Potential for carry‐over effects between study periods due to cross‐over design

  3. Children with the inattentive subtype of ADHD were excluded from this analysis

  4. Efficacy data presented as change from pre‐dose scores, rather than as effect sizes or response rates

  5. Pharmacokinetic and pharmacodynamic data were not collected and analysed

  6. Results reported for school‐aged children may not be relevant to other ADHD patient populations

  7. Higher percentage of females recruited

Key conclusions of study authors

  1. Significantly greater improvement in ADHD symptoms was noted with 30 mg extended‐release dexmethylphenidate compared with 20 mg extended‐release dexmethylphenidate at hours 10 through 12

  2. Tolerability was comparable between doses. 30‐mg dose of extended‐release dexmethylphenidate may provide further benefit to patients who do not maintain optimal symptom control later in the day with 20‐mg extended‐release dexmethylphenidate

  3. ADHD symptoms significantly improved with 30 mg extended‐release dexmethylphenidate compared with 20 mg extended‐release dexmethylphenidate at hours 10 to 12 in all ethnic parameters, with a statistically significant difference in the Caucasian subgroup

  4. 30 mg extended‐release dexmethylphenidate may provide further benefit to patients of all ethnic backgrounds who do not obtain optimal late‐day symptom control with 20 mg extended‐release dexmethylphenidate

Email correspondence with study authors: September 2013. Not possible to contact study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to 1 of 6 treatment sequences. All participants were given the lowest available number from the randomisation numbers provided at each site. A randomisation list was produced by using a validated system that automated the random assignment of treatment sequences to randomisation numbers in the specified ratio

Allocation concealment (selection bias)

Low risk

Randomisation data were kept strictly confidential until the time of unblinding

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All study medications and packaging were identical in appearance for blinding purposes. Participants, parents, study centre personnel and those who assessed outcomes were blinded to study treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All study medications and packaging were identical in appearance for blinding purposes. Participants, parents, study centre personnel and those who assessed outcomes were blinded to study treatment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Eight drop‐outs from the methylphenidate group. The ITT population included all randomly assigned participants who took ≥ 1 dose of study medication and had ≥ 1 post‐dose efficacy measurement. The safety population consisted of all participants who took ≥ 1 dose of study medication

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

High risk

Funding by Novartis Pharmaceuticals Corporation with the following involvement reported: design and conduct of the study; collection, management, analysis and interpretation of data; and preparation, review and approval of the manuscript. All study authors are employees or consultants or have received research grants from pharmaceutical companies

Conflicts of interest: All study authors are employees or consultants or have received research grants from pharmaceutical companies

Brown 1984a

Methods

Four‐week cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 2 weeks of placebo and 2 weeks of methylphenidate treatment with sequence according to randomisation

Participants

Number of participants screened: not stated

Number of participants included: 11 (all boys). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 11

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III

Age: mean 10 years, 5 months (range 9 years 1 month to 12 years 1 month)

IQ: > 80

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. DSM‐III diagnosis of ADD

Exclusion criteria

  1. Known neurological or sensory impairment

  2. IQ > 80

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.3 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: b.i.d.

Duration of each medication condition: 2 weeks

Washout before study initiation: not stated

Medication‐free period between interventions: time of day the pills were taken not stated

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale: rated at the end of each school week

  2. Conners' Teacher Rating Scale: rated at the end of each school week

Non‐serious adverse events

  1. Cardiovascular measures: heart rate, systolic and diastolic blood pressure. Heart rate was recorded after the child had rested for 5 minutes by placing a stethoscope over the precordium and measuring the rate for 1 minute. Blood pressure was obtained with a sphygmomanometer with the child seated after he had rested 5 minutes. Tested at the end of each 2‐week drug period

  2. “No deleterious side effects”

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Of additional importance in the present study was the finding that methylphenidate had no deleterious side effects and was well tolerated by all children participating in the research project

  2. Table 2 shows that individual variations in heart rate and blood pressure associated with methylphenidate trials are quite large

Key conclusions of study authors

  1. Results demonstrated significant improvement in sustained attention and impulse control, as well as in ratings of social behaviour, by both teachers and parents

  2. Cardiovascular functioning did not significantly increase as a function of methylphenidate

Email correspondence with study authors: November 2013. We received additional information regarding ethics approval, sample calculation, etc., from study authors. However, it was not possible to receive all requested data, as the study author no longer possessed raw data from the study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The sequence of the 2 medication conditions was randomly assigned, but no information was provided on methods

Allocation concealment (selection bias)

Low risk

Triple blinding; dosage was administered twice daily in the form of opaque capsules packaged by hospital pharmacists to conceal the contents

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Child and parent, teacher and the physician were blinded to the child’s medication condition

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Physician was blinded to the child’s medication concealment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data provided on all 11 participants

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

Unclear risk

Funded by National institute of Mental Health and National institutes of Health. Placebo and methylphenidate were supplied by CIBA‐GEIGY Corporation, Summit, New Jersey

Conflicts of interest: not stated

Brown 1985

Methods

Twelve‐week, randomised, parallel trial with 4 arms

  1. Cognitive training

  2. Methylphenidate combined with cognitive training

  3. No treatment (not randomly assigned)

  4. Cognitive training programme: individual, twice‐weekly, 1‐hour sessions for a total of 24 sessions spanning a 3‐month period

Participants

Number of participants included: methylphenidate + cognitive training 10, cognitive training 10

Number of participants followed up: methylphenidate + cognitive training 10, cognitive training 10

Number of withdrawals: methylphenidate + cognitive training 0, cognitive training 0

Diagnosis of ADHD: DSM‐III (types not stated)

Age: mean 11.36 years (range 6.4 to 11.9)

IQ: 101.92 (range 91 to 136)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: not stated

Comorbidity: not stated

Comedication: no. No child was receiving any psychopharmacological treatment

Sociodemographics: not stated. No significant differences in baseline demographics between the 2 groups

Inclusion criteria

  1. Demonstrating ADHD symptoms in serious and persistent form, agreed by parents and teachers

  2. Symptoms present for ≥ 12 months (parents to verify)

  3. Meeting criteria for ADD including hyperactivity

  4. According to DSM‐III

  5. Reading deficit of ≥ 2 grade levels

Exclusion criteria

  1. Symptoms seem to stem from stress at home or from inconsistent child management

  2. No major diseases or obvious physical defects (gross neurological, sensory, motor impairment or psychosis)

Interventions

Participants were randomly assigned to methylphenidate + cognitive training or to cognitive training only

Mean methylphenidate dosage: 0.3 mg/kg (range 5 mg/d to 15 mg/d)

Administration schedule: twice daily (morning and lunch)

Duration of intervention: 12 weeks + 3 months (only with medication)

Titration period: none

Treatment compliance: not stated

Cognitive training programme: individual, twice‐weekly, 1‐hour sessions for a total of 24 sessions spanning a 3‐month period

Outcomes

ADHD symptoms

  1. Conners’ Parent Rating Scale and Conners’ Abbreviated Teacher Rating Scale: baseline, 12 weeks, 3 months

  2. Teacher Rating of Attention: baseline, 12 weeks, 3 months

  3. Teacher Rating of Impulsivity: baseline, 12 weeks, 3 months

Notes

Sample calculation: no

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Children in the 2 medication treatment conditions demonstrated improvement in attentional deployment and in behavioural ratings

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label methylphenidate

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Not blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants followed up

Selective reporting (reporting bias)

Low risk

No protocol/design published. All pre‐specified outcomes of interest have been reported

Vested interest bias

Low risk

Funding: research supported by US Public Health Services Grant from the National Institute of Mental Health (NIMH), and by the Biomedical Research Award from the National Institutes of Health (NIH). Methylphenidate provided by CIBA‐GEIGY Corporation, Summit, New Jersey

Conflicts of interest: not stated

Brown 1988

Methods

Eight‐week, double‐blind, randomised, cross‐over trial with 4 interventions

  1. Placebo

  2. 0.15 mg/kg methylphenidate

  3. 0.30 mg/kg methylphenidate

  4. 0.5 mg/kg methylphenidate

Participants

Number of participants screened: not stated

Number of participants included: 11 (all boys). Participants were randomly assigned to possible drug condition orders

Number of participants followed up: 11

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 13 years, 7 months (range 12 years and 10 months to 14 years and 10 months)

IQ: full‐scale mean 92.91 (range 86 to 104)

Methylphenidate naive: not stated, but none of the participants had been treated with stimulants during the year preceding the study

Ethnicity: African American (100%)

Country: USA

Setting: out‐patient clinic

Comorbidity: conduct disorder, socialised aggressive (45%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. Sexual rating of ≥ 3 according to Tanner’s classification of stages of development, to ensure post‐pubertal status

  2. ADD according to DSM‐III

  3. Score of ≥ 15 on the Abbreviated Conners' Teacher Rating Scale

Exclusion criterion

  1. Mental retardation or gross neurological disorders

Interventions

Interventions (mean dosage)

  1. Placebo

  2. Methylphenidate low (0.15 mg/kg)

  3. Methylphenidate medium (0.30 mg/kg)

  4. Methylphenidate high (0.5 mg/kg)

Mean methylphenidate dosage: methylphenidate low (4.38 mg), medium (12.55 mg), high (21.28 mg)

Administration schedule: twice daily; morning and noon

Duration of each medication condition: 2 weeks

Washout before study initiation: none (but no stimulant treatment for the past year)

Titration period: none

Treatment compliance: Compliance was determined to be satisfactory

Outcomes

ADHD symptoms

At the end of each 2‐week trial, parents and teachers completed the following rating scales

  1. Conners' Parent Rating Scale ‐ Revised

  2. Abbreviated Conners' Parent Hyperactivity Index

  3. Abbreviated Conners' Teacher Hyperactivity Index

  4. ADD/H Comprehensive Teacher Rating Scale

Non‐serious adverse events

  1. Side Effects Rating Scale (includes questions about sleep disturbances, dysphoria, decreased appetite, physiological complaints such as headaches and generalised anxiety), parent‐rated (assessing the preceding week)

  2. Cardiovascular measures (heart rate, resting systolic and diastolic measures for blood pressure) assessed ≥ 1 hour after administration of methylphenidate or placebo. Apical pulse rates taken for 1 full minute

  3. Weight (during every clinic visit)

Notes

Sample calculation: no

Ethics approval: yes; Institutional Review Board (IRB) at Emory University

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Significant drug effects were found for most measures. In general, higher doses resulted in the most beneficial response in behavioural, academic and laboratory measures of attention and impulsivity. However, a significant linear increase occurred in diastolic blood pressure. Results suggest that methylphenidate is an effective adjunct to the treatment of ADD in adolescents

Comment from study authors

  1. We do not know whether our findings can be generalised in non‐black populations

Email correspondence with study authors: October 2013. We received from study authors additional information about ethics approval, planned outcomes and participants followed up. Unfortunately, it was not possible for study authors to provide other data that we needed because the study was conducted many years ago, and study authors no longer had the data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Drug order was randomly assigned across participants; no further description

Allocation concealment (selection bias)

Low risk

All medication was prepared in identical capsules by hospital pharmacists. Medication was dispensed in dated envelopes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All medication was prepared in identical capsules by hospital pharmacists. Medication was dispensed in dated envelopes. Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No withdrawals

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Researchers administered all measures that were proposed and reported these data in the published report

Vested interest bias

Low risk

Biomedical Research Support Grant Program, Division of Research Resources, National Institutes of Health and Emory University Research

Conflicts of interest: none reported

Brown 1991

Methods

Double‐blind, randomised, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: methylphenidate 10 mg, methylphenidate 15 mg, methylphenidate 20 mg, placebo

Participants

Study consisted of 22 participants, but only 7 had ADD. As outcomes were reported separately for these 7 participants, we were able to include the study

Number of participants screened: 25

Number of participants included: 22 (all boys). Participants were randomly assigned to 1 of 4 possible drug condition orders (in counterbalanced order)

Number of participants followed up: 22

Number of withdrawals: 0

Diagnosis: DSM‐III (conduct disorder, with 7 of 22 also diagnosed with ADD)

Age: mean:15.8 years (range 12.9 to18.9)

IQ: 96.22 (SD 15.12, range 80 to 123)

Methylphenidate naive: not stated

Ethnicity: Caucasian (100%)

Country: USA

Setting: hospital/out‐patient clinic

Comorbidity: conduct disorder (100%)

Comedication: no

Sociodemographics: middle and upper‐middle class

Inclusion criterion

  1. Hospitalised adolescents diagnosed with conduct disorder

Exclusion criterion

  1. Mental retardation, psychosis and organic brain disorder

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of methylphenidate (10 mg, 15 mg, 20 mg) and placebo

Mean methylphenidate dosage: 0.15 mg/kg, 0.22 mg/kg and 0.31 mg/kg

Administration schedule: twice daily, 8:00 AM and 12:00 PM

Duration of each medication condition: 4 days

Washout before study initiation: not stated

Titration period: 1 day before first phase

Treatment compliance: 100%

Outcomes

General behaviour

  1. Conners' Teacher Rating Scale (conduct factor): teacher‐rated daily

Non‐serious adverse events

  1. Side Effects Rating Scale (including 17 known adverse events for methylphenidate): observer‐rated daily

  2. Cadiovascular measures were recorded 90 minutes after 12:00 PM administration

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; initially participants received a 1‐day open trial of methylphenidate. Three participants were excluded because of intolerability

Comments from study authors

  1. ... in our study, mean milligram per kilogram doses were lower than in previously published reports … doses may simply have been too small to induce any real change in behaviour

  2. Another limitation that may have influenced the results is timing of the measurements of behaviour. As behavioural ratings were made by teachers at the end of the day, it is possible that medication effects (particularly for lower doses) had dissipated by the time the ratings were made

Key conclusions of study authors

  1. In summary, results are of theoretical importance and with additional research may suggest the potential efficacy of stimulants for treating adolescents with conduct disorders in the absence of ADD

  2. The present data may be interpreted to suggest that ADD may be managed with stimulant medication when it presents comorbidly with conduct disorder

Comment from review authors

  1. Data on the conduct disorder + ADD group were reported separately in the study ‐ that is why we could use the data

Email correspondence with study authors: unable to locate contact details for study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

All participants received each of the 4 doses in 1 of 24 possible randomly assigned sequences

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Methylphenidate and placebo were packaged in coloured gelatin capsules by the hospital pharmacist to avoid detection of dose, visually or by taste

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Under double‐blinded conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants followed up

Selective reporting (reporting bias)

Unclear risk

No protocol published. Not all pre‐specified outcomes of interest have been reported (e.g. scores on Conners' Teacher Rating Scale)

Vested interest bias

Unclear risk

Conflicts of interest: not stated

Buitelaar 1995

Methods

Randomised, cross‐over trial with 3 interventions

  1. Pindolol

  2. Methylphenidate

  3. Placebo

Phases

  1. Phase 1: 4‐week treatment block

  2. Phase 2: 2‐week drug‐free interval

  3. Phase 3: 4‐week treatment block

First 32 participants were randomly assigned to interventions 1 to 3 in the first treatment block, and to intervention 1 or 2 in the second treatment block. Next 20 participants were randomly assigned to intervention 2 or 3 in the first treatment block, and to intervention 2 or 3 in the second treatment block

Participants

Number of participants screened: not stated

Number of participants included: 52 (46 boys, 6 girls); however, because of an incomplete block design, only 46 were treated with methylphenidate and 31 were treated with placebo in first or second treatment block

Number of participants followed up: 52

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 9.3 years (range 6 to 13)

IQ: mean 94.2

Methylphenidate naive: 100%

Ethnicity: not stated

Country: the Netherlands

Setting: out‐patient clinic

Comorbidity: conduct disorder (38%); depressive disorder, dysthymia or major depressive disorder (15%); anxiety disorder, overanxious disorder or avoidant disorder (42%); psychomotor epilepsy (2%)

Comedication: antiepileptic medication (carbamazepine) at a fixed dosage (2%)

Sociodemographics: 20% were from families of high socioeconomic status, 50% of middle socioeconomic status and 30% of low socioeconomic status (on the Hollingshead Index). No significant difference in baseline characteristics were noted between groups of children treated with methylphenidate, pindolol or placebo

Inclusion criteria

  1. ADHD according to DSM‐III‐R criteria

  2. Scores in the clinical range on both the Child Behavior Checklist and Conners' Teacher Rating Scale, Hyperactivity factors

  3. Deficits in attention performance on a reaction time task or a continuous performance task in neuropsychological testing

  4. No previous treatment with psychotropic medication

  5. Clinical indication for drug treatment

Exclusion criteria

  1. Diagnosis of tic disorder or pervasive developmental disorder

  2. Family history of tic disorder

  3. Contraindications to treatment with blockers such as cardiac disease, in particular, conduction abnormalities and bradycardia, hypotension, obstructive pulmonary disease and insulin‐dependent diabetes

Interventions

Participants were randomly assigned to possible drug condition orders of 40 mg pindolol, 20 mg methylphenidate and placebo

Fixed dosage: 10 mg methylphenidate, twice daily (approximately 0.6 mg/kg/d)

Administration schedule: morning and noon

Duration of each medication condition: 4 weeks

Washout before study initiation: no (medication naive)

Medication‐free period between interventions: 2 weeks

Titration period: yes. After randomisation, during the first 3 days of a treatment period, participants received 1 morning dose (10 mg methylphenidate, 20 mg pindolol or placebo). After completion of endpoint assessment, medication was tapered off (3 days with 1 morning dose)

Treatment compliance: good to very good in 96% of children. Two children had poor compliance under methylphenidate treatment as the result of side effects

Outcomes

ADHD symptoms

  1. 10‐Item Abbreviated Conners' Rating Scale: rated by parents, teachers and a psychologist

  2. 93‐Item Conners' Parents Rating Scale

  3. 39‐Item Conners' Teachers Rating Scale

Parents and teachers completed ratings at baseline, at week 2 and at endpoint of each treatment period. The psychologist completed ratings at baseline and at endpoint of each treatment period. Furthermore, ACRS was rated 30 minutes after drug administration

Non‐serious adverse events

  1. Adverse effects checklist (encompassing 20 possible side effects, modified from the Stimulant Drug Side Effects Rating Scale, rated by parents after 2 and 4 weeks of treatment

  2. Treatment‐emergent adverse effects were further assessed systematically at endpoint by research psychiatrist

  3. Pulse and blood pressures were recorded at each clinical visit

Notes

Sample calculation: yes; for comparison of pindolol with methylphenidate (50 participants)

Ethics approval: yes; approved by the Committee for Research on Human Subjects of Utrecht University Hospital

Comments from study authors

  1. Interim analysis of side effects indicated that pindolol was associated with significantly more intense adverse effects when compared with placebo and methylphenidate. Consequently, pindolol was dropped from the study design, and the next 20 participants were included in a randomised methylphenidate‐placebo cross‐over design

  2. Study was limited by use of fixed dosage, with exclusive focus on behavioural symptoms ‐ not on improvement in neuropsychological measures of information processing, and with an incomplete and unbalanced block design

Key conclusions of study authors

  1. Beta‐blocker pindolol appeared to be modestly effective in the treatment of behavioural symptoms of children with ADHD. Data suggest some utility for pindolol in treating hyperactivity and conduct problems in children with ADHD, but safety concerns about troubling side effects clearly limit use of pindolol in ADHD

  2. Only strong levels of response could be predicted by baseline characteristics. Severity of disorder based on clinical judgement and improvement after a single dose of methylphenidate are found to be important contributors to response prediction

Comments from review authors

  1. Study designed to compare usage of pindolol and methylphenidate

  2. Few participants

  3. Study design changed during the study because of adverse effects. Phase 2 changed intervention from pindolol versus methylphenidate to methylphenidate versus placebo

  4. Phase 1: Given the relatively small sample sizes, effects of single treatments (i.e. methylphenidate vs placebo, and pindolol vs placebo) were not robust enough to reach conventional levels of statistical significance

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no; only medication‐naive participants

Any withdrawals due to adverse events: no; however, in 4 participants, dosages of methylphenidate had to be adjusted in the first 2 weeks of the study because of increased agitation, restlessness and insomnia. Two participants remained on 5 mg of methylphenidate for 4 weeks, whereas dosage for the other 2 participants could be gradually increased to 10 mg methylphenidate in the last 2 weeks

Email correspondence with study authors: January to March 2014. Requested but did not receive from study authors supplemental efficacy and safety data and information regarding randomisation, allocation concealment and blinding

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned, not further described

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, not further described. Methylphenidate and placebo were administered in identical‐looking tablets

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Two participants from the methylphenidate group had bad compliance but were included in the analyses, as an ITT analysis was planned

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no, but design of study changed during the course of the study because of adverse events

Selective reporting (reporting bias)

Unclear risk

Protocol not identified

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: no affiliations with pharmaceutical companies stated

Bukstein 1998

Methods

Cross‐over trial with 3 interventions

  1. Methylphenidate 0.3 mg MPH/kg

  2. Methylphenidate 0.6 mg MPH/kg

  3. Placebo

Phases: Study included 2 phases: a baseline phase and the medication trial itself. The baseline phase occurred during the first 2 weeks (9 days) of the programme, when the children were medication‐free. Each medication condition was administered for 7 days of the programme during the 21‐day trial

Participants

Number of participants screened: not stated

Number of participants included: 18. Participants were randomly assigned to 1 of 3 possible drug condition orders

Number of participants followed up: 18 (14 boys, 4 girls)

Number of withdrawals: 0

DSM‐III‐R criteria for ADHD and oppositional defiant disorder or conduct disorder

Age: mean 9.4 years (range 6.1 to 12.2)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: Caucasian (17%), African American (83%)

Country: USA

Setting: out‐patient clinic (summer school at clinic)

Comorbidity: oppositional defiant disorder (56%) and conduct disorder (44%)

Comedication: no

Sociodemographics: Participants were predominantly from lower socioeconomic classes, with an average Hollingshead Index of Social Status of 3.83 (SD 1.65, range 1 to 5). Thirteen (72%) of the participants' families were receiving public assistance. Only 4 of the children lived with both biological parents; 12 (67%) lived with their biological mother only. Inner city environment characterised by higher than average rates of poverty and community violence. No significant differences in baseline demographics between groups

Inclusion criterion

  1. DSM‐III criteria for ADHD and oppositional defiant disorder or conduct disorder, while attending the Summer Treatment and Enrichment Program (STEP)

Exclusion criterion

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate (0.3 mg/kg or 0.6 mg/kg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 8:30 AM, 11:45 AM and 3:00 PM

Duration of each medication condition: 7 days

Washout before study initiation: 9 days

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: poor compliance with the weekend medication condition; most families missed ≥ 1 dose each weekend of the trial. Poor compliance with the 3‐dose regimen was so widespread that trialists omitted from the trial all data on weekend doses

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale: rated by staff daily

  2. IOWA Conners' Parent Rating Scale: rated by parents daily

  3. Conners' Teacher Rating Scale: rated daily

Non‐serious adverse event

  1. Side Effects Rating Scale: adaptation of the Barkley Side Effects Rating Scale, rated daily by staff and also rated by parents

Notes

Key conclusions of study authors

  1. Staff ratings of behaviour of children in the programme and in an academic classroom showed that children displayed significant improvement in ADHD symptoms and aggressive behaviour with low‐ and high‐dose methylphenidate conditions

  2. At home, parents and guardians reported few significant differences in behaviour ratings between placebo and methylphenidate

  3. In both settings, methylphenidate was well tolerated, with few side effects found during active drug conditions

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Schedule for each condition was randomly assigned across the 5 weekdays to minimise programme effects; the only qualifying condition was that approximately one‐half of the 7 days of each medication condition would occur during each half of the 21‐day trial

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Nurse and other Summer Treatment and Enrichment Program (STEP) staff, children and parents were blinded to dosages and schedules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Each child's daily data were collected and entered by trained research associates, who were unaware of medication status

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All included in the analyses

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

None

Butter 1983

Methods

One‐week, double‐blind, parallel trial with 3 arms

  1. Adrenocorticotropic hormone

  2. Methylphenidate (10 mg, 15 mg or 20 mg, weight adjusted)

  3. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 30 (all boys)

Number of participants randomly assigned: methylphenidate 10, placebo 10

Number of participants followed up: methylphenidate 10, placebo 10

Diagnosis of ADHD: DSM‐III

Age: mean in years (range 6 to 12)

IQ: above 85

Methylphenidate naive: not stated

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated. No information about significant differences in baseline demographics between groups

Inclusion criteria

  1. Clinical ADHD diagnosis of attention deficit disorder with hyperkinesis (DSM‐III)

  2. Hyperkinesis rating required a score of 15 or higher on Conners' Short Form Rating Scale, and hyperkinetic behaviour had to be apparent throughout most of the day

  3. Untreated behaviour had to be a cause of severe difficulty both at home and at school

Exclusion criterion

  1. Wechsler Intelligence Scale for Children (WISC) score < 85 or abnormal perceptual functioning

Interventions

Participants were randomly assigned to adrenocorticotropic hormone, methylphenidate or placebo

Mean methylphenidate dosage: 0.5 mg/kg

Administration schedule: once daily, 7.30 AM

Duration of intervention: 1 week. One week drug‐free followed by 1 week of placebo treatment. After placebo washout, randomly assigned to adrenocorticotropic hormone, methylphenidate or placebo

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Short Form Rating Scale: completed by parents and clinicians

  2. Conners' Rating Scale Teachers: rated during placebo and active drug phases

Non‐serious adverse event

  1. EEG, haematology, liver and kidney function test, blood pressure and pulse, blood chemistry, urinalysis before and after treatment

EEG, haematology, blood chemistry and urinalysis were within normal limits before treatment and remained so after treatment

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Children treated with methylphenidate show significantly greater vasomotor reactivity, behavioural improvement and learning receptivity than children treated with adrenocorticotropic hormone and placebo

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

After placebo washout, treatment was assigned in a double‐blind and random manner

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Neurologist assessing EEG blinded. Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Low risk

Funded by the Scientific Development Group, Organon International B.V., Oss, the Netherlands

Conflicts of interest: none

Carlson 1995

Methods

Double‐blind, placebo‐controlled, repeat‐measures (across drug and dosage), cross‐over trial with 6 interventions

  1. Methylphenidate at 10 mg twice daily

  2. Methylphenidate at 15 mg twice daily

  3. Methylphenidate at 20 mg twice daily

  4. Desipramine alone

  5. Desipramine + methylphenidate

  6. Placebo

Phases: Each child received placebo, desipramine, each of the 3 doses of methylphenidate (10 mg, 15 mg, 20 mg) and combined desipramine and methylphenidate (at the same 3 doses)

Participants

Number of participants screened: not stated

Number of participants included: 16 (all boys). Participants were randomly assigned to different drug condition orders and placebo

Number of participants followed up: 16

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (62.5%), inattentive (12.5%), “in partial remission” (25%))

Age: mean: not reported (range 7.9 to 12.10 years)

IQ: mean not stated (range overall 81 to 121; range verbal 74 to 113, range performance 73 to 126)

Methylphenidate naive: 4 (25%)

Ethnicity: Caucasian (87.5%), African American (12.5%)

Country: USA

Setting: in‐patient ward

Comorbidity: yes; major depressive disorder (68.75%), dysthymic disorder (31.25%). “All had ODD, CD or both”

Comedication: yes

Sociodemographics: not stated

Inclusion criteria

  1. Initial team diagnosis of ADHD, non‐bipolar major depressive disorder, dysthymic disorder or a combination thereof

  2. ≥ 7 years old

  3. Wechsler Intelligence Scale for Children, Revised, full‐scale IQ > 80

Exclusion criteria

  1. Medical contraindication to medications being investigated

  2. Family history of bipolar disorder in first‐ or second‐degree relatives

  3. Abnormal baseline laboratory values or ECG

Interventions

Participants were randomly assigned to different possible drug condition orders of methylphenidate (10 mg, 15 mg and 20 mg), twice daily at 7:30 AM and 11:30 AM × 6 days, then 1‐day washout; the titrated therapeutic level (125 ng/mL to 225 ng/mL) of desipramine twice daily at 3:30 PM and 7: 30 PM × 3 weeks minimum before final measures taken; and placebo

Mean methylphenidate dosage: not stated

Duration of each medication condition: 1 week

Washout before study initiation: 14 days

Medication‐free period between intervention: 1 day

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADD/H Comprehensive Teacher Rating Scale (ACTerRS): teacher‐rated

General behaviour

  1. Humphrey's Teacher Self‐Control Rating Scale

  2. Inpatient Global Rating Scale

Non‐serious adverse events

  1. Subjective Treatment Emergent Symptom Scale: side effect ratings, collected weekly by nurse

  2. Somatic factor of the Inpatient Global Rating Scale: collected weekly by nurse

  3. Childrens’ Depression Rating Scale ‐ Revised: Appetite and Sleep disturbance items collected weekly by research psychologist

  4. Cardiovascular side effects (daily morning blood pressure and pulse rates + weekly ECG)

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusion of study authors

  1. “With regard to differential efficacy, the major findings of this study were that a combination of MMI and MPH (at 20 mg) was somewhat more effective than DMI or MPH alone for improving hyperactive, inattentive and oppositional defiant, and 'aggressive' behaviours across both school and unit settings. Efficacy was also demonstrated for MPH and DMI alone in school, but less so on the unit”

Comment from review authors

  1. Small sample of 16 participants entering the trial seems heterogeneous and opportunistic rather than clearly defined, with a mixture of diagnoses including mood, ADHD and oppositional defiant disorder/conduct disorder. This makes it difficult to believe that the results are reliable

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: maybe; hence exclusion criterion number 1: medical contraindication to medications being investigated

Email correspondence with study authors. Emailed first study author to ask for outcome data in mean and SD format. Study authors replied in July 2014 to say that they were unable to help us

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Not stated

Allocation concealment (selection bias)

High risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications were packaged in identical grey capsules (size 00) that were administered 4 times per day

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

For this protocol, all raters (including teachers, nurses, psychologist and physicians, except for the attending child psychiatrist, who controlled the desipramine dosage but did not rate the children), children and parents were blinded to all medication conditions

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Unclear risk

Not stated

Carlson 2007

Methods

Six‐week, multi‐centre, double‐blind, parallel, randomised controlled trial (RCT) with 2 arms

  1. Atomoxetine + methylphenidate

  2. Atomoxetine + placebo

RCT was preceded by a 4‐week, open‐label, atomoxetine and placebo phase, during which participants who had adequate response were removed

Participants

Number of participants screened: not stated

Number of participants included: phase 1 (4‐week open‐label atomoxetine and placebo phase) 25 (1 boy, 20 girls); phase 2 (6‐week, double‐blind RCT) 17

Number of participants randomly assigned: methylphenidate 9, placebo 8

Number of participants followed up: methylphenidate 8, placebo 7

Number of withdrawals: methylphenidate 1, placebo 1

Diagnosis of ADHD: DSM‐IV (combined (79% in phase 1))

Age: phase 1 mean 9.6 years (range 6 to 12)

IQ: > 70

Stimulant‐naive: 4%

Ethnicity: Caucasian (83%), other (17%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (50% in phase 1)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria, including assessment details

  1. 6 to 12 years of age

  2. DSM‐IV diagnosis of ADHD, any type

  3. Rating on ADHD Rating Scale, Fourth Edition, Parent Version ‐ Investigator Administered and Scored Version of ≥ 1.5 SD above age and sex norms

  4. Severity rating of at least moderate on the Clinical Global Impressions Severity Scale

  5. History (preceding 12 months) of insufficient response to an adequate stimulant trial, which was defined as gradual titration of stimulant medication for ≥ 2 weeks at specified doses for each medication. Inadequate response was determined by the child's prescribing physician, who also documented his or her opinion that a change in treatment was needed

  6. Patients must be of normal intelligence, as assessed by the investigator (i.e. without a general impairment of intelligence, and likely, in the investigator's judgement, to achieve a score ≥ 70 on an IQ test) (Administration of a formal IQ test is not an entry requirement for this study. Specific learning disabilities are not considered general impairments of intelligence)

  7. Patients must be able to swallow capsules

Exclusion criteria

  1. Weighed < 22 kg or > 60 kg at study entry

  2. Had any other Axis I diagnosis, including pervasive developmental disorder, mood or anxiety disorder (Presence of comorbid oppositional defiant disorder was not an exclusion criterion)

  3. Bipolar disorder, autism

  4. Any medical conditions that would contraindicate the use of atomoxetine or extended‐release methylphenidate

  5. History of any seizure disorder and/or Rolandic seizures (other than febrile seizures) or prior ECG abnormalities in the absence of seizures, or history of taking (or are currently taking) anticonvulsants for seizure control

  6. History of severe allergies to > 1 class of medication or multiple adverse drug reactions, including hypersensitivity to methylphenidate

  7. History of intolerance or non‐response to atomoxetine

  8. Used any concomitant psychotropic or excluded medications

  9. Ingestion of any excluded medications 5 days before baseline ratings and randomisation

Interventions

Participants were randomly assigned to extended‐release OROS methylphenidate or placebo

Cointervention: atomoxetine

Methylphenidate mean dosage: 1.02 mg/kg

Administration schedule: once daily

Duration of intervention: 6 weeks

Titration period for methylphenidate: after randomisation to target dose of 1.08 mg/kg/d (max 1.2 mg/kg/d)

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition, Parent Version ‐ Investigator Administered and scores: parent‐rated

  2. Conners' Parent Rating Scale Revised, Short Form: parent‐rated at baseline and at weeks 4, 5, 6 and 10

General behaviour

  1. Weekly Parent Ratings of Evening and Morning Behaviour: parent‐rated at baseline and at weeks 4, 5, 6 and 10

Non‐serious adverse events

  1. Vital signs, at each visit

  2. Weight, at each visit

  3. Spontaneous adverse event reports, at each visit

Notes

Sample calculation: Included sample (25 participants) is too small in relation to the sample calculation (85 participants)

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no, but exclusion of atomoxetine/placebo responders before study phase 2

Comment from study authors

  1. The present findings can be applied only to children with an inadequate stimulant response. Given the small study sample, particularly in the combination treatment groups, the findings must be considered preliminary

Key conclusion of study authors

  1. Methylphenidate appears to be safely combined with atomoxetine, but conclusions are limited by small sample

Comment from review authors

  1. For the review, we used data from study phase 2, that is, the RCT with 2 arms (intervention: methylphenidate + atomoxetine; control: placebo + atomoxetine)

Email correspondence with study authors. June to November 2013. We received from study authors and sponsoring pharmaceutical company, supplemental information regarding IQ and blinding procedures, as well as data on weight, treatment‐emergent adverse events and ECG

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned via an interactive voice response system to receive extended‐release methylphenidate or placebo

Allocation concealment (selection bias)

Low risk

Investigators and participants were blinded to the precise visit at which randomisation to blinded placebo or Concerta occurred, as this visit is not identified in the investigator’s copy of the protocol, and as use of blinded study drug begins at visit 2 and continues up to visit 8

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

Low risk

Efficacy outcome measures were conducted on the ITT sample by using an LOCF method

Selection bias: no

Selective reporting (reporting bias)

Low risk

Protocol identified

Vested interest bias

High risk

Funding: Research was funded by Eli Lilly and Company, Indianapolis, Indiana

Conflicts of interest: Dr. Carlson has received research support or has consulted with the following companies: Abbott Laboratories, Cephalon, Eli Lilly and Company, Janssen, McNeil, Otsuka and Shire Pharmaceuticals. Dr. Dunn has received research support or has served on Speakers' Bureaus of the following companies: AstraZeneca, Eli Lilly and Company, NIH, Otsuka and Pfizer Pharmaceuticals. Drs. Kelsey, Ruff, Ball and Allen and Ms. Ahrbecker are employees and/or shareholders of Eli Lilly and Company

Castellanos 1997

Methods

Nine‐week, double‐blind, cross‐over trial with 3 interventions for 3 weeks each

  1. Methylphenidate

  2. Dextroamphetamine

  3. Placebo

Study was followed up by an open clinical follow‐up of 22 months

Participants

Number of participants screened: 64

Number of participants included: 22 (all boys). Participants were randomly assigned to different possible drug condition orders

Number of participants followed up: 20

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐III‐R

Age: mean 9.4 years (range 6 to 13)

IQ: mean 98.8

Methylphenidate naive: 0

Ethnicity: Caucasian (80%), African American (10%), Asian (5%), Hispanic (5%)

Country: USA

Setting: out‐patient clinic

Comorbidity: Tourette’s disorder (95%), chronic motor tics (5%), conduct disorder (5%), oppositional defiant disorder (30%), reading disorder (5%), overanxious disorder (5%), obsessive‐compulsive disorder (10%), enuresis (20%)

Comedication: 4 received haloperidol

Sociodemographics: no information

Inclusion criteria

  1. DSM‐III criteria for Tourette’s disorder, with tics confirmed by a knowledgeable clinician ≥ 1 year before referral

  2. Symptoms of ADHD present in ≥ 2 settings

  3. Conners' hyperactivity factor scores from home teacher ≥ 2 SD > age norms

Exclusion criteria

  1. Full‐scale IQ < 75 (Wechsler Intelligence Scale for Children ‐ Revised)

  2. Evidence of medical or neurological disease

  3. Any other Axis I psychiatric disorder except obsessive‐compulsive disorder, conduct or oppositional disorder, overanxious disorder and specific developmental disorders

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of methylphenidate and placebo. Methylphenidate was increased weekly. For body weight > 30 kg, weekly methylphenidate doses were 15 mg/dose, 25 mg/dose and 45 mg/dose b.i.d. (for weight ≤30 kg, 12.5: 25 mg/dose and 45 mg/dose, b.i.d.)

Mean methylphenidate dosage: main cohort 1.20 mg/kg, second cohort 0.69 mg/kg, third cohort 1.22 mg/kg

Administration schedule: twice a day: breakfast and lunch

Duration of each medication condition: 3 weeks. First cohort underwent weekly increases in stimulant doses described as low, medium and high. Second cohort underwent increase described as low, medium and medium, and the third cohort as low, high and high

Washout before study initiation: minimum 4 weeks

Medication‐free period between interventions: 20 hours

Titration period: during the first 3 weeks of intervention

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Conners’ Teacher Rating Scale: completed by day programme teachers, weekly ratings

Non‐serious adverse events

  1. Adverse effects

  2. Tic severity: Unified Rating Scale (Tourette Syndrome Association): variety, frequency, intensity, complexity and interference of motor and vocal tics based on observations by programme staff and derived by consensus at weekly meetings

Notes

Sample calculation: no

Ethics approval: approved by the National Institute of Mental Health (NIMH) Institutional Review Board

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Limitations

    1. Small sample size

    2. Drug dosage was not randomly assigned

    3. Allowed 4 participants to continue on a constant dose of haloperidol

    4. Exploration of different dose schedules

    5. As methylphenidate undergoes much more rapid and complete metabolism than dextroamphetamine, we would expect blood level curves of the 2 stimulants to differ markedly

Key conclusion of study authors

  1. Substantial minority of comorbid participants had consistent worsening of tics while taking stimulants, although most experienced improvement in ADHD symptoms with acceptable effects on tics

Comment from review authors

  1. Study consists of a main study of 12 boys followed by 2 smaller cohorts of, respectively, 6 and 4 boys

Email correspondence with study authors: January 2014. Corresponding author was not able to supply us with further information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were randomly assigned

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Capsule formulation prepared by a pharmacy

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Two drop‐outs; their data are not included in subsequent analyses. One of these was dropped from the study because of acute exacerbation of tics, and 1 because of excessively disruptive behaviour

Selective reporting (reporting bias)

High risk

Yes; they seem to have reported only hyperactivity scores from Conners’ Teacher Rating Scale

Vested interest bias

Low risk

None declared

Conflicts of interest: not stated

Chacko 2005

Methods

Six‐week within‐participant, randomised, placebo‐controlled, double‐blind, daily cross‐over trial with 3 interventions

  1. Methylphenidate at 0.3 mg/kg given at 7:45 AM and 11:45 AM

  2. Methylphenidate at 0.6 mg/kg given at 7:45 AM and 11:45 AM

  3. Placebo given at 7:45 AM and 11:45 AM

Phases: daily cross‐over between medication conditions. Two weeks of baseline and adjustment period before 6‐week medication trial. Data from this paper pertain to 5‐ and 6‐year‐old children attending a summer treatment programme between 1987 and 1997

Participants

Number of participants screened: not stated

Number of participants included: 36 (32 boys, 4 girls). Participants were randomly assigned to 1 of 3 possible groups

Number of participants followed up: 36

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (n = 35, subtype not stated); DSM‐IV (n = 1, combined type)

Age: mean 6.13 years (range 5 to 6)

IQ: mean 102 (SD 15.50)

Methylphenidate naive: not reported

Ethnicity: Caucasian (86%), other (14%)

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: oppositional defiant disorder (50%), conduct disorder (27.7%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosis of ADHD according to DSM

  2. Between 5 and 6 years of age

  3. Parental consent

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned, daily, to 1 of 3 possible interventions: 0.3 mg/kg methylphenidate, 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: b.i.d. 7:45 AM and 11:45 AM from Monday through Thursday

Duration of each medication condition: daily shifted

Washout before study initiation: not stated

Medication‐free period between interventions: maximum 18‐hour gap between doses (11:45 AM to 7:45 AM the following day) and no medication Thursday to Sunday

Titration period: Before the 6‐week trial began, participants underwent a 2‐week baseline and adjustment period

Treatment compliance: not stated

Outcomes

Serious adverse events

  1. None reported

Non‐serious adverse events

  1. "Classroom teachers, counselors, and parents completed daily ratings of the presence and severity of common stimulant side effects"

Side effects are reported as occurring: "none" (i.e. symptom was assessed and was found absent), "mild" (i.e. symptom was present but was not sufficient to cause concern among child, peers or adults), "moderate" (i.e. symptom caused impairment of functioning or social embarrassment to the degree that benefits of medication must be considerable to justify risks of continuing medication) or "severe" (i.e. symptom caused significant impairment of functioning or social embarrassment to the degree that the child should not continue to receive medication). Rating of "moderate" or "severe" signifies clinically significant side effects. Side effects are based on staff/parent subjective report on severity of the side effect. Children were considered to have significant side effects in a particular area if clinically significant side effects were reported within that area for ≥ 50% of observations in a given condition

Notes

Sample calculation: no information

Ethics approval: no information

Comments from study authors

  1. Study limitations. Most participants were white males, limiting the generalisability of findings to females and ethnic minorities

  2. Furthermore, the sample size did not allow statistical analyses of ADHD subtype or comorbidity

Key conclusion of study authors

  1. Stimulant medication is an effective treatment for young children diagnosed with ADHD; however, multiple domains of functioning must be assessed for the most effective dose for young children with ADHD to be determined

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: unclear; 2 weeks of adjustment period before the medication condition ‐ no information on how many were screened and how many participated during baseline weeks

Comments from review authors

  1. Participants in this study were attending a summer treatment programme between 1987 and 1997

  2. Other articles from summer treatment programme

    1. Pelham and Hoza, 1987

    2. Pelham and Smith, 2000

    3. Pelham and Hoza, 1996

    4. Pelham et al., 1990

    5. Pelham et al., 1993, 1999, 2002

Email correspondence with study authors: December 2013. We emailed study authors to request additional information about study sample and side effects. Study authors replied to say that the data are no longer available. Data from this study could not be used in meta‐analyses

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Active medication and placebo were disguised in opaque capsules and were dispensed in daily pill reminders. Each condition occurred one or two times per week, with the order of the conditions randomized on a daily basis. Data were averaged across days within conditions for each child"

Allocation concealment (selection bias)

Unclear risk

“Each condition occurred one or two times per week, with the order of the conditions randomized on a daily basis”

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

“Active medication and placebo were disguised in opaque capsules and were dispensed in daily pill reminders”

Double‐blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Side effects were measured for 30 participants

Selective reporting (reporting bias)

High risk

Unclear how the population sample was selected from the group of attendees at these programmes over a 10‐year period

Vested interest bias

High risk

During the conduct of this research, Dr. Pelham was supported by grants from the National Institute of Mental Health (NIMH) (MH48157, MH47390, MH45576, MH50467, MH53554, MH62946), NIAAA (AA06267, AA11873), National Institute on Drug Abuse (NIDA) (DA05605, DA12414), National Institute of Neurological Disorders and Stroke (NINDS) (NS39087), National Institute for Environmental Studies (NIES) (ES05015) and National Institute of Child Health and Human Development (NICHHD) (HD42080)

Conflicts of interest: Several study authors have affiliations with medical companies

Childress 2009

Methods

Five‐week, multi‐centre, multiple‐setting, phase III, double‐blind, randomised, placebo‐controlled, parallel trial with 4 arms

  1. Placebo

  2. Extended‐release dexmethylphenidate hydrochloride 10 mg

  3. Extended‐release dexmethylphenidate hydrochloride 20 mg

  4. Extended‐release dexmethylphenidate hydrochloride 30 mg

One to 3 weeks titration and 2 to 4 weeks maintenance period dependent on the allocated active drug group

Participants

Number of participants screened: 332

Number of participants included: 253 (163 boys, 90 girls)

Number of participants randomly assigned: extended‐release dexmethylphenidate hydrochloride 188 (10 mg, n = 66; 20 mg, n = 62; 30 mg, n = 60), placebo 65

Number of participants followed up: extended‐release dexmethylphenidate hydrochloride 161 (10 mg, n = 56; 20 mg, n = 54; 30 mg, n = 51), placebo 57

Number of withdrawals: extended‐release dexmethylphenidate hydrochloride 27 (10 mg, n = 10; 20 mg, n = 8; 30 mg, n = 9), placebo 8

Diagnosis of ADHD: DSM‐IV (combined (73.9%), hyperactive‐impulsive (2.8%), inattentive (21.7%), missing data (1.6 %))

Age: mean 8.7 years (SD 1.84, range not reported)

IQ: not stated

Methylphenidate naive: 69.2%

Ethnicity: Caucasian (57.7%), African American (28.9%), Asian (0.8%), other (12.6%)

Country: 34 centres in the United States

Setting: out‐patient clinic

Comorbidity: respiratory, thoracic and mediastinal disorders (methylphenidate 26.9%, placebo 38.1%), immune system disorders (methylphenidate 22.5%, placebo 14.3%), nervous system disorders (methylphenidate 18.7%, placebo 19.0%), surgical and medical procedures (methylphenidate 15.4%, placebo 11.0%), infections and infestations (methylphenidate 14.3%, placebo 17.5%) and skin and subcutaneous tissue disorders (methylphenidate 11.0%, placebo 9.5%)

Comedication: ≥ 1 concomitant medication or non‐drug therapy after start of study (methylphenidate 39.0%, placebo 44.4%). The most common concomitant medications were analgesics, antihistamines and allergy medications

Sociodemographics: not stated. Treatment groups were well balanced in relation to participant background characteristics, and baseline demographics were comparable among treatment groups

Inclusion criteria

  1. 6 to 12 years of age

  2. DSM‐IV‐TR diagnosis of ADHD

  3. Patients attending school had to have the same teacher (English or Math) for the entire duration of the study, who was willing and able to spend sufficient time with the patient to make valid weekly assessments

  4. Drug naive or not treated with any methylphenidate‐related medication during the month before the study

  5. Patients receiving psychological or behavioural therapies before the screening visit were considered eligible to participate, provided that therapy had been ongoing for ≥ 3 months with the same therapist

  6. Patients had to have academic competence appropriate to their age and the following subscale total scores on Conners’ ADHD/DSM‐IV Scales ‐ Teacher Version: For boys, baseline scores on Conners’ ADHD/DSM‐IV Scales ‐ Teacher Version, Total subscale were required to be 27 for those 6 to 8 years old, 24 for those 9 to 11 years old and 19 for those 12 years old. For girls, respective baseline cutoff scores on Conners' ADHD/DSM‐IV Scales ‐ Teacher Version, for the same age groups were 16, 13 and 12

Exclusion criteria

  1. Home‐schooled children

  2. Any medical condition that interfered with study assessments or that was not stable for ≥ 3 months before screening

  3. Clinically significant abnormalities detected during screening

  4. Family history of long‐QT syndrome, current diagnosis or history of cardiac abnormalities, seizures, psychiatric disorders such as schizophrenia, schizoaffective disorder, severe obsessive‐compulsive disorder, conduct disorder, autism, chronic tic disorder, Tourette's disorder or any mood or anxiety disorder

  5. Antidepressants, antipsychotics, herbal preparations with psychotropic effects, amphetamine‐based medications, benzodiazepines, barbiturates, sedatives or hypnotics, monoamine oxidase inhibitors and atomoxetine had to be stopped 1 to 4 weeks before randomisation according to their half‐lives. All concomitant medications that could interfere with absorption, metabolism and distribution of study drug were excluded from the start of screening until the end of all evaluations. Over‐the‐counter analgesics, short‐term antibiotic treatment for minor infections and any medication needed to treat adverse events were allowed

  6. Additionally, patients who were judged by the investigator as likely to be non‐compliant with study procedures, including those with a suspected history of substance abuse and those living with a person diagnosed with a substance abuse disorder, or whose parent or guardian was unable or unwilling to complete Conners’ ADHD/DSM‐IV Scales ‐ Parent Version

  7. Pregnancy or lactation

  8. Positive drug screen

Interventions

Participants were randomly assigned to extended‐release dexmethylphenidate hydrochloride or placebo

Fixed methylphenidate dosage: 10 mg, 20 mg or 30 mg

Administration schedule: once daily, morning

Duration of intervention: 5 weeks

Washout before study initiation: up to 28 days (duration dependent on the half‐life of any previous psychotropic medication)

Titration period: 1 to 3 weeks, initiated after randomisation

Treatment compliance: 86% completed

Outcomes

ADHD symptoms

  1. Conners' ADHD/DSM‐IV ‐ Teacher Version: rated by teacher at baseline and each week

  2. Conners' ADHD/DSM‐IV ‐ Parent Version (including 12‐item ADHD Index and 18‐item DSM‐IV Subscale total): rated by parent or guardian at baseline and each week

Non‐serious adverse events

  1. Regular monitoring and recording of adverse events, serious adverse events, vital signs, body weight, ECG, physical examination, haematology parameters, blood chemistry and urinalysis. While vital signs were recorded at every visit, physical, laboratory and ECG evaluations were completed during the screening visit and the final study visit. Weight was recorded at baseline and at the final visit. Notable value for weight loss was defined as a decrease from baseline weight at study end of 7%

Notes

Sample calculation: yes; 252 participants (63 per treatment group)

Ethics approval: Institutional review boards or ethical review committees at each centre approved the study

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Limitations: short duration of study, forced‐dose titration, not powered to assess differences in treatment effects within extended‐release dexmethylphenidate hydrochloride groups

Key conclusions of study authors

  1. All 3 doses of extended‐release dexmethylphenidate hydrochloride (10 mg, 20 mg or 30 mg a day) were significantly more effective than placebo in improving ADHD symptoms, as confirmed by teacher, parent and clinician

  2. Additionally, extended‐release dexmethylphenidate hydrochloride was well tolerated and demonstrated a consistent safety profile. Mean changes from baseline in vital signs in extended‐release dexmethylphenidate hydrochloride groups were small, clinically irrelevant, unrelated to dose and similar to placebo

  3. Despite the forced‐titration design of the study, the cardiovascular safety profile of extended‐release dexmethylphenidate hydrochloride (10 mg/d, 20 mg/d and 30 mg/d) is similar to placebo in children with ADHD

Comment from review authors

  1. Additionally, participants judged by the investigator as likely to be non‐compliant with study procedures were excluded

Email correspondence with study authors in December 2013. We have contacted study authors twice in an attempt to obtain supplemental information regarding blinding, allocation concealment and weight loss. We have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed by using a validated system that automated the assignment of treatment arms to randomisation numbers in the specified ratio

Allocation concealment (selection bias)

Low risk

Allocation concealed

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, but no detailed description

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind, but no detailed description

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT population. LOCF

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

High risk

This study was funded by Novartis Pharmaceuticals Corporation. Novartis Pharma has been helping with development of the manuscript

Conflicts of interest: Several study authors have received research support from, are speakers for, are consultants of, are on the Advisory Board, have served on the Speakers' Bureaus of or are employees of several pharmaceutical companies

Chronis 2003

Methods

Eight‐week within‐participant, placebo‐controlled, randomised, cross‐over trial (Summer Treatment Camp (STP) with 3 interventions)

  1. Methylphenidate

  2. Adderall

  3. Placebo

Phases: 2‐week baseline assessment followed by 6‐week medication trial

Participants

Number of participants screened: 48

Number of participants included: 21 (19 boys, 2 girls). Participants were randomly assigned to 1 of 7 drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated
Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 10.26 years (±1.9, range 6 to 12)

IQ: mean 109.9 (±18.8)

Methylphenidate naive: 14 children had received methylphenidate before start of the study

Ethnicity: White (100%)

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: learning problems (42.9%), oppositional defiant disorder (66.7%), conduct disorder (23.8%)

Comedication: not stated

Sociodemographics (median family income = $35,000/y (< $15,000/y to > $100,000/y); 66.7% of parents married

Inclusion criterion

  1. DSM‐IV diagnosis of ADHD

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to 1 of 7 possible drug condition orders of immediate‐release methylphenidate, Adderall and placebo

  1. Placebo at 7:30 AM, 11: 30 AM and 3:30 PM

  2. 0.3 mg/kg methylphenidate at 7:30 AM, 11:30 AM and 3:30 PM

  3. 0.3 mg/kg methylphenidate at 7:30 AM and 11:30 AM with 0.15 mg/kg at 3:30 PM

  4. 0.3 mg/kg methylphenidate at 7:30 AM only

  5. 0.3 mg/kg Adderall at 7:30 AM and at 3:30 PM

  6. 0.3 mg/kg Adderall at 7:30 AM with 0.15 mg/kg received at 3:30 PM

  7. 0.3 mg/kg Adderall at 7:30 AM only

Mean methylphenidate dosage: not stated

Administration schedule: methylphenidate once, twice or 3 times daily according to the randomisation procedure

Duration of each medication condition: All participants received medication each Monday through Friday throughout a period of 6 weeks for a 24‐day clinical assessment period. Assessment period was divided into three 8‐day segments. Within each segment, placebo occurred twice and each other condition occurred once, with the order of conditions randomly assigned on a daily basis

Washout before study initiation: not stated

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale: rated by counsellors at the end of each day, by teachers after the classroom period each day and by parents in the evening

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale: rated by counsellors and teachers daily, rated by parents each evening with the addition of an item assessing difficulty falling asleep

Notes

Sample calculation: no

Ethics approval: yes

Any withdrawals due to adverse events: none during the trial

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Additional (late‐afternoon) stimulant dose has beneficial effects on parent‐child interactions

  2. At the end of the Summer Treatment Programme (STP) for the 21 children, it was determined that 5 children did not show a sufficient positive response

Key conclusions of study authors

  1. Single morning dose of Adderall had effects for an entire school day

  2. Single dose of Adderall was equivalent to immediate‐release methylphenidate twice/day

Comments from review authors

  1. No mean methylphenidate dose

  2. Behavioural intervention was also implemented during the entire 8‐week study period

  3. As intervention sequences were switched on a daily basis according to randomisation, we did not ask about first period data from this study

Email correspondence with study authors: July 2014. Emailed study authors twice to request additional information. Study author was not able to provide us with additional data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

“Randomized by day”, no description of how randomisation took place

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

To ensure blinding, placebo capsules were given at 11:30 AM in the Adderall conditions and for applicable doses in the other conditions. Active medication and placebo were disguised in opaque gelation capsules by a local pharmacy and were dispensed in daily pill reminders by the study doctor. Furthermore, children were informed that they would be receiving 2 different kinds of medication to see how well they worked, and that some days they would receive inactive pills, but that they, their counsellors or teachers and their parents would not know what kind of pill they would get each day

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Children were informed that they would be receiving 2 different kinds of medication to see how well they worked, and that some days they would receive inactive pills, but that they, their counsellors or teachers and their parents would not know what kind of pill they would get each day

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No mention of drop‐out

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

All outcomes reported

Vested interest bias

High risk

Supported by a grant from Shire‐Richwood Pharmaceuticals, Incorporated ‐ manufacturer of Adderall ‐ and from the National Institute of Mental Health (NIMH)

Coghill 2007

Methods

Twelve‐week randomised, placebo‐controlled, double‐blind cross‐over trial with 3 interventions

  1. Methylphenidate 0.3 mg/kg

  2. Methylphenidate 0.6 mg/kg

  3. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 75 (all boys). Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (combined type); ICD‐10 (hyperkinetic disorder)

Age: mean not reported (range 7 to 15 years)

IQ: > 80

Methylphenidate naive: 100%

Ethnicity: not stated

Country: United Kingdom

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (41.3%), conduct disorder (28%), depressive disorder (4%), generalised anxiety disorder (2.7%), separation anxiety disorder (4%), tic disorder (2.7%), social phobia (1.3%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Eligible boys (scoring 1.5 SD from the mean on both Conners’ Parent Rating Scale, Short Version, and Conners’ Teacher Rating Scale, Short Version) were interviewed by an experienced child and adolescent psychiatrist using the Kiddie‐Schedule for Affective Disorders and Schizophrenia Present and Lifetime

  2. Those meeting criteria for hyperkinetic disorder (HD F 90) (ICD‐10) and combined subtype (DSM‐IV) were invited to participate

Exclusion criteria

  1. History of neurological impairment/learning disability

  2. IQ < 80

  3. Chronic physical illness

  4. Sensory or motor impairment

  5. Current or previous exposure to stimulant medication

  6. Abuse of any illegal drugs

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders: 3 mg/kg/dose methylphenidate or 6 mg/kg/dose methylphenidate and placebo

Administration schedule: twice daily

Duration of each medication condition: 4 weeks

Washout before study initiation: no

Titration period: none

Treatment compliance: assessed by pill count and clinical enquiry but not further described

Outcomes

ADHD symptoms

  1. Conners' Global Index, Parent Version and Teacher Version (10 item): rated by parents and teachers, each after 4 weeks

Notes

Sample calculation: no

Ethics approval: yes

Key conclusions of study authors

  1. Chronic methylphenidate predominantly enhanced neuropsychological functioning on "recognition memory" component tasks with modest "executive" demands

  2. Neuropsychological measures offer only modest contributions to the prediction of clinical responses to methylphenidate in ADHD

Comment from study authors

  1. Doses (0.3 mg/kg and 0.6 mg/kg) were chosen to reflect low‐ and high‐dose regimens, respectively

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors in 2013. Emailed study authors twice with a request for additional information regarding protocol and number of participants on which analyses were based. Have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned by an independent clinical trials pharmacist (using a computer‐generated random number sequence with block design to ensure equal numbers in each treatment arm)

Allocation concealment (selection bias)

Low risk

Participants were randomly assigned by an independent clinical trials pharmacist

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Placebo‐controlled, double‐blind, no further information

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Clinical status was assessed by interview conducted by an experienced, blinded child and adolescent psychiatrist. Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

No description about how many participants were included in analyses

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Unclear risk

This work was supported by a local trust through a Tenovus Scotland initiative

Conflicts of interest: Some study authors have affiliations with different pharmaceutical companies

Coghill 2013

Methods

Seven‐week, multi‐centre, double‐blind, parallel, dose‐optimised trial with 3 arms

  1. Lisdexamfetamine dimesylate

  2. OROS methylphenidate

  3. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 221 (181 boys, 40 girls)

Number of participants randomly assigned: methylphenidate 112, placebo 111

Number of participants followed up: methylphenidate 74, placebo 42

Number of withdrawals: methylphenidate 38, placebo 68

Diagnosis of ADHD: DSM‐IV‐TR (combined (methylphenidate 86.4%, placebo 79.1%), hyperactive‐impulsive (methylphenidate 0.9%, placebo 6.4%), inattentive (methylphenidate 12.7%, placebo 14.5%)

Age: mean 10.9 years (range 6 to 17)

IQ: normal

Methylphenidate naive: methylphenidate 60 (54.1%), placebo 58 (52.7%)

Ethnicity: Caucasian (methylphenidate 96.4%, placebo 98.2%), African American (0%), Asian (0%), Hispanic/Latino (methylphenidate 1.8%, placebo 0%)

Countries: Germany, Sweden, Spain, Hungary, France, the UK, Italy, Belgium, Poland, the Netherlands

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (methylphenidate 9.0%, placebo 7.3%), concomitant psychiatric diagnosis (methylphenidate 26.1%, placebo 18.2%)

Comedication: not stated

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Any sex

  2. 6 to 17 years of age

  3. Meeting criteria for ADHD according to DSM‐IV‐TR

  4. Baseline ADHD moderate severity with ADHD‐RS‐IV score of 28 or higher

  5. Age‐appropriate intellectual functioning

  6. Blood pressure measurements within the 95th percentile for age, sex and height

  7. Ability to swallow a capsule

  8. Girls of childbearing potential had to have a negative urine pregnancy test at baseline and to comply with any contraceptive requirements of the protocol

Exclusion criteria

  1. Failure to respond to previous OROS methylphenidate therapy

  2. Presence of a conduct disorder (excluding oppositional defiant disorder)

  3. Pregnancy or lactation

  4. Weight below 22.7 kg; body mass index (BMI, kg/m2) > 97th percentile for age and sex

  5. Positive urine drug test (with the exception of patient’s current ADHD therapy)

  6. Clinically significant electrocardiogram or laboratory abnormalities

  7. Suspected substance abuse or dependence disorder (excluding nicotine) within the previous 6 months

  8. History of seizures

  9. Tics or Tourette’s disorder

  10. Known structural cardiac abnormality

  11. Any other condition that might increase vulnerability to the sympathomimetic effects of a stimulant drug

  12. Patients whose current ADHD medication provided effective control of symptoms with acceptable tolerability

  13. Patient currently considered a suicide risk, with previous suicide attempt or with history of, or currently demonstrating, active suicidal ideation

  14. Patient with glaucoma

  15. Patient with documented allergy, hypersensitivity or intolerance to amphetamine or methylphenidate

  16. Patient with documented allergy, hypersensitivity or intolerance to any excipients in test or reference products

  17. Patients with known family history of sudden cardiac death or ventricular arrhythmia

  18. Patients with pre‐existing severe gastrointestinal tract narrowing (pathological or iatrogenic)

  19. Patients unable to tolerate the study drug were withdrawn from the study

Interventions

Participants were randomly assigned to OROS methylphenidate or placebo

Mean methylphenidate dosage: 45.4 ± 12.7 mg/d (9.9% 18 mg, 19.8% 36 mg, 53.2% 54 mg)

Administration schedule: once a day, 7:00 AM

Duration of intervention: 7 weeks

Titration period: 4‐week stepwise dose‐optimisation period after randomisation. Three‐week dose‐maintenance period followed by 1‐week washout and safety follow‐up

Treatment compliance: 2 discontinued in placebo group because of non‐compliance, 3 in methylphenidate group

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition: total score at endpoint, investigator‐rated at baseline and weekly for 7 weeks. Endpoint was defined as the last on‐therapy, post‐randomisation treatment visit with a valid ADHD Rating Scale, Fourth Edition, total score

Quality of life

  1. Health Utilities Index‐2: parent‐rated at baseline and at weeks 4 and 7. Scoring ranges from 0.00 (dead) to 1.00 (perfect health). Higher scores represent better health status

  2. Child Health and Illness Profile, Child Edition: Parent Report Form: parent‐rated at baseline and at weeks 4 and 7. Questionnaire comprises 76 items classed into 5 domains and 12 associated subdomains. Most items relate to the past 4 weeks; the remainder are not associated with a specific time period. Parents use a 5‐point response format to assess each item. Achievement is considered the primary health‐related quality of life outcome

Non‐serious adverse events

  1. Safety assessments included treatment‐emergent adverse events, clinical laboratory evaluations, physical examinations, vital signs and ECGs, observer‐rated weekly for 8 weeks

Notes

Sample calculation: yes

Ethics approval: approved by an independent ethics committee/institutional review board and regulatory agency at each centre (as appropriate) before study initiation

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Comments from study authors

  1. Individuals with comorbid conditions, such as post‐traumatic stress disorder, bipolar affective disorder or severe anxiety disorder, were excluded from this study

  2. As a result of European regulations, the maximum dose of OROS methylphenidate administered in this study was 54 mg/d. However, it is possible that the smaller than usual placebo response did, at least in part, contribute to this large treatment effect. ADHD is currently less frequently diagnosed in Europe than in North America, with evidence of underrecognition and underdiagnosis (healthcareimprovementscotland.org). Consequently, it is likely that individuals who are diagnosed are at the more severe end of the spectrum and therefore would be less likely to show a response to placebo. Consistent with this suggestion, reassessment of data from the Multimodal Treatment of ADHD (MTA) study revealed that children who had been diagnosed with ADHD on the basis of DSM‐IV criteria, but also met the more restrictive International Classification of Diseases 10th Revision criteria for hyperkinetic disorder, showed a more robust response to medication compared with those who met DSM‐IV criteria alone

Key conclusions of study authors

  1. In this European 7‐week phase III study, lisdexamfetamine dimesylate was more effective than placebo in improving symptoms in children and adolescents with ADHD

  2. Compared with placebo, significant improvements in both ADHD core symptoms and global functioning were observed

  3. Lisdexamfetamine dimesylate was well tolerated, with treatment‐emergent adverse effects consistent with those reported in previous studies

  4. Robust efficacy outcomes were also observed for OROS methylphenidate, which was included in this study as a reference arm

Comment from review authors

  1. Very confused on the number of participants included in the different analyses

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Interactive voice/web response system was used to allocate a unique randomisation number to each participant. Randomisation was stratified by country and age group (6 to 12 or 13 to 17 years of age)

Allocation concealment (selection bias)

Low risk

Interactive voice/web response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study drugs were overencapsulated and appeared identical

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Masking: double‐blinded (participant, caregiver, investigator, outcomes assessor)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

LOCF approach was used when efficacy assessments were incomplete for a participant owing to early withdrawal from the study or for missing data. However, as the review authors cannot understand the relationship between the n of the full analysis set and the endpoint data, risk of bias was assessed as unclear

Selection bias (e.g. titration before randomisation → exclusion): none

Selective reporting (reporting bias)

Low risk

Outcomes according to protocol

Vested interest bias

High risk

Funded by Shire Development LLC

Conflicts of interest: C. Anderson, R. Civil, N. Higgins, A. Lyne and L. Squires are employees of Shire and own stock/stock options. Some study authors have received compensation for serving as consultants or speakers, or they or the institutions they work for have received research support or royalties from different companies or organisations

Connor 2000

Methods

Three‐month, randomised, blind, parallel‐group study with 3 arms

  1. Methylphenidate and clonidine

  2. Clonidine monotherapy

  3. Methylphenidate monotherapy

Participants

Number of participants screened: 24

Number of participants included: 24 (methylphenidate + clonidine 8 boys, 0 girls; placebo + clonidine 8 boys, 0 girls)

Number of participants randomly assigned: methylphenidate + clonidine 8, placebo + clonidine 8, placebo + methylphenidate 8

Number of participants followed up: methylphenidate + clonidine 8, placebo + clonidine 6

Number of withdrawals: methylphenidate + clonidine 0, placebo + clonidine 2

Diagnosis of ADHD: DSM‐III‐R (combined (100%))

Mean age: methylphenidate + clonidine 10.1 years, placebo + clonidine 9.3 years (range 6 to 16)

IQ: > 70

Methylphenidate naive: 54%

Ethnicity: Caucasian (11%), African American (1%), Asian (0%), Hispanic (0%), other (0%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder or conduct disorder (100%)

Comedication: not reported

Sociodemographics: No significant differences in baseline demographics were noted between groups

Inclusion criteria

  1. DSM‐III‐R for ADHD

  2. DSM‐III‐R for oppositional defiant disorder or conduct disorder, score of 1.5 SD above the mean for age and sex on the Parent Child Behavior Checklist, Attention problems scale (T score > 65) and a score on the Teacher Child Attention Problem Rating Scale of ≥ 93rd percentile

  3. Score 1.5 SD above the mean for age and sex on the Parent or Teacher Child Behavior Checklist, Delinquency or Aggression problems scale

  4. Normal findings from general physical examination by family physician within 6 months before study entry

Exclusion criterion

  1. Medical history that contraindicated use of stimulants or clonidine

Interventions

Participants were randomly assigned to methylphenidate + clonidine or placebo + clonidine

Methylphenidate dose: 35.0 (5.67) mg/d

Administration schedule: b.i.d.

Duration of intervention: 12 weeks

Titration period: 4 weeks after randomisation

Treatment compliance: “All subjects were acceptably compliant with the protocol”

Outcomes

General behaviour

  1. Disruptive Behavior Disorders Rating Scale: parent‐ and teacher‐rated monthly

  2. Home Situations Questionnaire: parent‐rated monthly

  3. School Situations Questionnaire: teacher‐rated monthly

Non‐serious adverse events

  1. Possible side effects of methylphenidate and clonidine: Combined Stimulant/Clonidine Side Effects Rating Scale, parent‐ and teacher‐rated monthly

  2. Pulse and blood pressure obtained monthly. ECG measured after first month. Height and weight obtained monthly

Notes

Intervention group: methylphenidate + clonidine; control group: placebo + clonidine

Ethics approval: yes

Sample calculation: no

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: June 2013. We obtained additional information from study authors, but it was not possible to receive supplemental data. We could not perform a meta‐analysis on any of the outcomes, as we did not have relevant data and transformation was not possible

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Eight participants were randomly assigned to each group. Three participants were previous methylphenidate treatment failures and refused randomisation to the methylphenidate‐alone study arm. These 3 participants were partially randomly assigned to methylphenidate and clonidine or to clonidine alone. All other children were fully randomly assigned

Allocation concealment (selection bias)

Low risk

After study completion, the medication blind was broken. All medication capsules and placebo capsules were prepared by the UMMS (University of Massachusetts Medical School) Pharmacy in identical capsules to disguise taste and smell. All participants in all study groups received an equal number of capsules per day

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All teachers, school nurses, parents, children and research assistants completing dependent measures were blinded to the child's treatment group for the study duration. Completion of electrocardiograms (ECGs) for only 2 clonidine treatment groups may have broken blinding for parent raters and for the child (but not for teacher raters nor for research assistants administering the GDS, who remained blinded as to whether the child had received an ECG). This is not relevant to us, as we are using data only on the 2 groups completing ECGs

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

As stated above

Incomplete outcome data (attrition bias)
All outcomes

Low risk

LOCF

Selection bias: no

Selective reporting (reporting bias)

Low risk

Reply from study author on our request for the protocol: protocol described in the study

Vested interest bias

Low risk

Supported by a UMMS (University of Massachusetts Medical School) Small Grants Project Award

Conflicts of interest: not stated

Cook 1993

Methods

Six‐week, double‐blind, single cross‐over, placebo‐controlled study conducted on a group of 15 participants with ADD and a comparison group of 10 age‐matched participants who did not have ADD

ADD group was randomly assigned to 1 of 2 experimental groups

  1. Methylphenidate

  2. Placebo

Participants

Regarding the ADD group

Number of participants screened: not stated

Number of participants included: 15. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 15

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (100% met DSM‐III criteria for ADD with hyperactivity)

Age: mean 104.5 months (range 6 to 10 years)

IQ: mean 110.5 (SD 7.15)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: central auditory processing disorder (80%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Male

  2. 6 to 10 years of age

  3. IQ > 85

  4. Clinical diagnosis of ADD from paediatrician and met DSM‐III criteria for ADD on both Parent and Teacher Versions of the Swanson, Nolan and Pelham Scale; scored ≥ 15 points on the Parent Version of the Abbreviated Conners' Rating Scale

Exclusion criteria

Participants were excluded if they had

  1. Seizures

  2. Cerebral palsy

  3. Learning disabilities

  4. Speech or language problems

  5. Vision or peripheral hearing problems

  6. Thought disorder

  7. Abnormal auditory brainstem evoked potentials

  8. Previous drug treatment for ADD

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 0.30 mg/kg (0.057)

Administration schedule: not stated

Duration of each medication condition: 3 weeks

Washout before study initiation: not relevant, 100% treatment naive

Medication‐free period between interventions: minimum of 48 hours between the 2 interventions

Titration period: Dose was titrated up to a maximum (methylphenidate tablets or placebo tablets) over the first 3 weeks of the experimental period

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Nolan and Pelham Scale: teacher‐ and parent‐rated, only items on the Inattention and Impulsivity subdomains, measured at the end of each treatment condition

  2. ADD/H Comprehensive Teacher Rating Scale: measured at the end of each treatment condition

  3. Abbreviated Conners' Rating Scale: parent‐rated, measured at the end of each treatment condition

Notes

Sample calculation: no information

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Limitations: The conclusion should not be generalised to results obtained with measures other than the 3 behaviour rating scales. The sample is small. Stimulant treatment periods were brief, and results may not have been maintained at the same level if the study had been conducted over a longer period

Comment from review authors

  1. Read relevant parts of Dr. Cook's Ph.D. dissertation to get additional information about the study

Key conclusion of study authors

  1. The implications of these results are 3‐fold. First, sustained attention is a critical feature of performance on tests of central auditory processing disorder, and current diagnostic criteria for central auditory processing disorder make clinical separation of the 2 disorders problematic. Second, stimulants appear to be a useful treatment for symptoms of both ADD and central auditory processing disorder. Third, tests of central auditory processing disorder may provide a useful measure of ADD symptoms and response to stimulants

Email correspondence with study authors: September 2013. Emailed study author requesting additional information about the study and data. Dr. Cook referred to his Ph.D. dissertation, which we managed to get

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were assigned to groups by a table of random numbers known only to the pharmacist

Allocation concealment (selection bias)

Low risk

Drugs were coded and administered by a pharmacist, and clinical titration of dosage was done by the participant’s paediatrician; neither practitioner was involved in data collection

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Physician, audiologist, teachers and parents involved in behaviour ratings and participants themselves were blinded to assigned treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Physician, audiologist, teachers and parents involved in behaviour ratings and participants themselves were blinded to assigned treatment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No incomplete outcome data

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

Low risk

Supported by the Medical Center Rehabilitation Hospital Foundation and the School of Medicine, University North Dakota; the Veterans Hospital; the Dakota Clinic; and The Neuropsychiatric Institute, Fargo, North Dakota

Conflicts of interest: not stated

Corkum 2008

Methods

Three‐week, blind medication trial with cross‐over design, in which children were randomly assigned to the following

  1. Two different doses of methylphenidate (low and moderate)

  2. Placebo

Participants

Number of participants recruited: 28

Number of participants included: 21 (15 boys, 6 girls)

Number of withdrawals: 7 children excluded from final data analyses

Diagnosis of ADHD: DSM‐IV (combined type (52%), hyperactive‐impulsive type (10%), inattentive type (38%))

Age: mean not reported (range 6 years 1 month to 12 years 1 month)

IQ: no intellectual disability

Methylphenidate naive: All participants were stimulant medication‐naive

Comorbidity: learning disabilities (29%), oppositional defiant disorder (10%), conduct disorder (0%), generalised anxiety disorder (0%), depression (0%)

Setting: out‐patient clinic

Country: Canada

Ethnicity: All participants were Caucasian

Sociodemographics: predominantly middle‐class families

Inclusion criteria

  1. Stimulant medication‐naive

  2. DSM‐IV criteria for 1 of the 3 ADHD subtypes

  3. Received a recommendation to initiate a trial of methylphenidate following assessment

  4. Parents/caregivers agreed to initiate a stimulant medication trial through the clinic paediatrician

Exclusion criteria

  1. IQ < 1 SD below the mean on the Wechsler Intelligence Scale for Children, Fourth Edition

  2. Known neurological, metabolic or seizure disorder

  3. Currently taking other psychotropic medications or medications for sleep disturbances

  4. Symptoms of an intrinsic sleep disorder (i.e. sleep apnoea, restless legs syndrome or periodic limb movements in sleep) or a sleep‐onset disorder based on parent report

  5. Reached criteria for another mental health disorder that was considered

  6. Primary to the ADHD diagnosis (e.g. autism)

Interventions

Participants were randomly assigned to 1 of 3 medication‐dosing schedules, including 1 week of baseline, placebo and low and moderate immediate‐release methylphenidate (ritalin) dose condition. Children weighing ≤ 25 kg received 5‐mg and 10‐mg doses; children weighing > 25 kg received 10‐mg and 15‐mg doses. Children received medication
t.i.d. (8:00 AM, 12:00 PM, 4:00 PM)

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Conners’ Parent and Teacher Rating Scale ‐ Revised, Short Form: rated weekly

Non‐serious adverse events

  1. Sleep Disturbances Scale for Children based on child’s sleep over previous week: rated by parents

  2. Actigraphy

  3. Sleep diary

Notes

Sample calculation: not stated

Ethics approval: yes

Key conclusion of study authors

  1. Based on findings from the current study, we would encourage physicians and parents to closely monitor children’s sleep when treating ADHD with stimulant medication, and to carefully weigh the benefits of improved behavioural functioning while taking medication against the potential negative consequences of sleep

Comment from review authors

  1. Paper includes data on adverse effects: sleep disturbance (actigraph data, sleep diary data, Sleep Disturbances Scale for Children data) related to methylphenidate treatment compared with placebo

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: August to October 2013. We received supplemental information regarding additional data from study authors, but we never received first period data from the cross‐over trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Baseline data from 1 week were collected, followed by a 3‐week medication trial with random assignment of children to 1 of 3 medication‐dosing schedules. The original, fully randomised schedule was modified after a pilot study, conducted before the current study, indicated that children receiving a moderate dose before the low dose of methylphenidate reported increased side effects and were more likely to stop taking the third dose (4:00 PM) of medication

Allocation concealment (selection bias)

Low risk

Pharmacy local to the ADHD clinic prepared both placebo and active medication, which were packaged into identical gelatin capsules

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Child, family, school personnel and study investigators were unaware of the randomisation schedule. This information was made available only to the paediatrician and the pharmacist

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Child, family, school personnel and study investigators were unaware of the randomisation schedule. This information was made available only to the paediatrician and the pharmacist

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

7 children excluded from final data analyses for the following reasons: actigraphic problems (i.e. data failure/loss of actigraph/refusal to wear actigraph) (n = 5), withdrawal of consent due to marital discord (n = 1) and decision to try alternative medication immediately before the start of the medication trial (n = 1)

Selective reporting (reporting bias)

Low risk

No subsequent correspondence with first study author regarding additional information on reporting

Vested interest bias

Low risk

Research was supported by a grant from the Izaak Walton Killam

IWK Health Centre in Halifax, Nova Scotia

Conflicts of interest: "none declared"

Cox 2006

Methods

Randomised, double‐blind, placebo‐controlled, cross‐over trial of adolescent drivers with ADHD who were assessed on a driving simulator after taking

  1. 72 mg of OROS methylphenidate

  2. 30 mg of extended‐release mixed amphetamine salts

  3. Placebo

Phases: 3 (2 relevant phases)

Participants

Number of participants screened: not stated

Number of participants included: 35 (19 boys, 16 girls). Participants were randomly assigned to 1 of 2 relevant (n = 3) possible drug condition orders

Number of participants followed up: 35

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (combined 21 (60%), hyperactive‐impulsive 2 (6%), inattentive 12 (34%))

Age: mean 17.8 years (range 16 to 19)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: agoraphobia (2.9%), conduct disorder and marijuana abuse (2.9%), obsessive‐compulsive disorder (2.9%), obsessive‐compulsive disorder and hypomania (2.9%), nicotine dependence (5.7%)

Comedication: 2 were taking no medication, 21 were taking methylphenidate and 12 were taking amphetamine formulations at the start of the study

Sociodemographics: not stated

Inclusion criteria

  1. Adolescent drivers

  2. 16 to 19 years of age

  3. ADHD according to DSM‐IV. To meet inclusion criteria for a diagnosis of ADHD, adolescents first needed to surpass clinical cutoffs for ADHD on a commonly used parent rating scale, the ADHD Rating Scale, Fourth Edition

  4. Psychiatrist confirmed ADHD diagnosis with the Standardized Interview for Adult ADHD (DSM‐IV)

  5. Positive history of stimulant responsiveness, as disclosed by adolescents and by parent reports; current licence to drive and reported daily driving activity

Exclusion criteria

  1. Adolescents were excluded when they had a history of tics or any adverse reaction to stimulant medication

  2. History of substance abuse disclosed by patient or parent

  3. Co‐existing medical condition or medication usage known to interfere with safe administration of stimulant medications

Interventions

Participants were randomly assigned to 1 of 2 relevant drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 72 mg/d

Administration schedule: 2 overlaid capsules/tablets in blister packs each day on awakening

Duration of each medication condition: 17 days

Washout before study initiation: not stated

Medication‐free period between interventions: 4 to 21 days between OROS methylphenidate and mixed amphetamine salts

Titration period: half dose (36 mg/d OROS methylphenidate) days 1 to 5, full dose (72 mg/d OROS methylphenidate) days 6 to 17 initiated after randomisation

Treatment compliance: not stated. Pill counts were completed at each study visit

Outcomes

Non‐serious adverse events

  1. Throughout the study, only 1 adverse event was reported ‐ urinary difficulty. This adverse event occurred during treatment with 36 mg of OROS methylphenidate and was resolved after 2 days without discontinuation of the medicine

  2. Self‐Reported Stimulant Drug Side Effects Rating Scale on days 5 and 10

Notes

Sample calculation: no

Ethics approval: yes

Comments from study authors

  1. Automobile accidents are the leading cause of death among adolescents, and collisions are 2 to 4 times more likely to occur among adolescents with ADHD

  2. Studies have demonstrated that stimulants improve driving performance

  3. Results provide evidence supporting most literature on children with short‐acting stimulants; longer‐acting stimulants appear equally effective for female and male post‐pubertal adolescents with ADHD

  4. Limitation of this study is that few participants with hyperactive subtype were included, limiting extrapolation of results to this subgroup

Key conclusions of study authors

  1. This study validates the use of stimulants to improve driving performance in adolescents with attention deficit hyperactivity disorder

  2. In this study, OROS methylphenidate promoted significantly improved driving performance compared with placebo and extended‐release mixed amphetamine salts

Comments from review authors

  1. Effects of methylphenidate were also measured, but not in comparison with placebo. Therefore, we could not use these data in the review

  2. Some participants are older than 18 years. However, as mean age is > 19 years, this study is still included but was tested by sensitivity analyses

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; excluded if any history of adverse events to stimulant medications

Email correspondence with study authors: February 2014. We received additional information from study authors. Data from the Self‐Reported Stimulant Drug Side Effects Rating Scale are no longer available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Using a random‐numbers table, each participant was assigned"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Treatments were provided in different forms: "Participants took 2 overlaid capsules/tablets in blister packs each day on awakening". Participants and research assistants were blinded to medication conditions

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"Participants were either tested on placebo or were not required to come in for testing". Participants and research assistants were blinded to medication condition

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Email correspondence with study author: no drop‐outs and all were followed‐up (Krogh 2013b [pers comm])

Selection bias (e.g. titration after randomisation → exclusion): none described

Selective reporting (reporting bias)

Low risk

Email correspondence with study author, who stated that all planned outcome measures and analyses are described in the papers (Krogh 2013b [pers comm])

Vested interest bias

High risk

Study was supported by funding from McNeil Pediatrics, a division of McNeil‐PPC Incorporated

Conflicts of interest: none described

Douglas 1986

Methods

Five‐day cross‐over trial with 2 interventions

  1. Methylphenidate (0.3 mg/kg capsules each dose, morning + afternoon)

  2. Placebo (100 mg lactose capsules)

Phases

  1. Screening session about a week before testing (given practice on all tasks in the test battery; appropriate level was established for each child on tasks graded for difficulty level)

  2. Five days of testing. Children received drug (D) or placebo (P) according to 1 of 4 possible orders: (1) PDDPP: (2) DPPDD; (3) DDPPD; (4) PPDDP. Testings were received each morning according to 4 test orders: (1) ABCDEF; (2) ACBEDF; (3) ADEBCF and (4) AEDCBF

Before the study was undertaken, 16 drug and test order combinations for the morning test battery were ordered randomly. As participants entered the study, they were assigned to these drug‐plus‐test order combinations until all 16 participants had been assigned. The order of the 2 tests in the afternoon battery (arithmetic and word discovery) was alternated over children, and each child received tests in the same order over 5 days of testing

Participants

Number of participants screened: not stated

Number of participants included: 16 (15 boys, 1 girl). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 16

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (subtypes not stated)

Age: mean 9.2 years (range 6 to 11.6)

IQ: mean103.19 (range 89 to 125)

Methylphenidate naive: 13 (81%)

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: Children’s families varied from I to V on the Hollingshead and Redlich Index (with most families falling within level III to IV (V being the poorest)

Inclusion criteria

  1. Met criteria for a DSM‐III diagnosis of ADHD (American Psychiatric Association, APA 1980)

  2. Had to receive ratings above 1.5 (on a 0 to 3 scale) on the Hyperactivity Index of Revised Conners' Parent and Teacher Rating Scales

Participants were referred to the Hyperactivity Project at Montreal Children’s Hospital by paediatricians or school personnel. Referral was based on presence of the following symptoms

  1. Inattentiveness

  2. Impulsivity

  3. Hyperactivity

  4. Restlessness

  5. Poor compliance and poor self control

Symptoms were of sufficient severity to prompt referring physicians to consider a trial on stimulant medication

Exclusion criteria

  1. Psychosis

  2. Serious visual, auditory or language deficits

  3. Diagnosed as brain damaged

  4. Restless behaviour attributable to emotional problems or a stressful home environment

  5. Appearance of symptoms before age five and evidence that symptoms were chronic and pervasive

Interventions

Participants were randomly assigned to 1 of 4 possible orders of drug (D) or placebo (P): (1) PDDPP; (2) DPPDD; (3) DDPPD; (4) PPDDP. On days when participants were assigned to active medication, they received a capsule containing the quantity of medication closest to a calculated dose of 0.3 mg/kg for each dose (morning and afternoon)

Mean methylphenidate dosage: not stated

Administration schedule: morning capsule administered approximately 1 hour after breakfast and 45 minutes before morning test battery was administered. Second capsule, identical to the morning capsule, administered before child left for lunch and returned to school. Time between morning and afternoon capsule was approximately 3½ hours

Duration of each medication condition: 1 day each; in all 2 or 3 days, depending on order of drugs assigned

Washout before study initiation: 24 hours before screening, 48 hours before first testing day

Titration period: none

Treatment compliance: capsule administered by examiner

Outcomes

ADHD symptoms

  1. Hyperactivity Index from Conners' Revised Teacher Rating Scale: examiner‐rated, at end of each morning of individual testing

  2. Hyperactivity Index from Conners' Revised Teacher Rating Scale: teacher‐rated, each afternoon

Score for the Index is based on the mean of item ratings on a 4‐point scale (0 to 3)

Notes

Sample calculation: no

Ethics approval: yes

Key conclusions of study authors

  1. Results indicate methylphenidate‐induced improvement in most measures

  2. Drug‐induced changes reflected increased output, accuracy and efficiency and improved learning acquisition. Evidence of increased effort and self correcting behaviours was found

  3. It is argued that review authors have underestimated the potential of stimulants to improve performance of ADD‐H children on academic, learning and cognitive tasks

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: none stated

Email correspondence with study authors: July 2014. Emailed study authors to ask for additional information. Received information on ethics approval, but first study author was not able to provide additional data or information on the study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Capsule was administered by examiner

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

In a few cases in which data were missing, scores from 1 or 2 days were used to compute means for drug and placebo conditions. No information on drop‐out rate

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

Low risk

Research was supported by Grant Number MA 6913, from the Medical Research Council of Canada

Conflicts of interest: no information

Douglas 1995

Methods

Eight‐day, double‐blind, cross‐over trial in which participants were randomly assigned to different doses of methylphenidate and placebo

  1. 0.3 mg/kg methylphenidate

  2. 0.6 mg/kg methylphenidate

  3. 0.9 mg/kg methylphenidate

  4. Placebo

Phases

  1. Assessment week 1: 4 different doses given on 4 different days

  2. Assessment week 2: same structure as assessment week 1

Participants

Number of participants screened: not stated

Number of participants included: 17 (16 boys, 1 girl). Participants were randomly assigned to 1 of 24 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R. Also had to meet DSM‐III criteria for ADD with hyperactivity

Age: mean 9 years 5 months (range 6 years 3 months to 11 years 9 months)

IQ: mean 104.3 (range 89.0 to 127)

Methylphenidate naive: 53%

Ethnicity: not stated

Country: Canada

Setting: treatment centre (hospital) or out‐patient clinic

Comorbidity: oppositional defiant disorder (71%), conduct disorder (18%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DMS‐III‐R criteria for ADHD

  2. DSM‐III criteria for attention deficit disorder with hyperactivity

  3. Ratings from both mothers and teachers at or above a criterion score of 1.5 on the Hyperactivity Index of the Revised Conners' Rating Scale

Exclusion criteria

  1. IQ < 85

  2. Serious visual, auditory or speech deficits

  3. Evidence of organic damage

  4. Evidence suggesting that symptoms could be attributed to emotional problems or a stressful home environment

Interventions

Participants were randomly assigned to 1 of 24 possible drug condition orders of methylphenidate (0.3 mg/kg, 0.6 mg/kg, 0.9 mg/kg) and placebo

Mean methylphenidate dosage: 0.3 mg/kg: 9.71; 0.6 mg/kg: 19.42; 0.9 mg/kg: 29.14

Administration schedule: once daily

Duration of each medication condition: 1 day; each child received each dosage twice during the study (i.e. during first and second assessment weeks)

Washout before study initiation: In the case of children currently receiving stimulants, medication was discontinued ≥ 48 hours before screening

Titration period: none

Treatment compliance: To ensure compliance, medications were administered at the laboratory

Outcomes

Non‐serious adverse events

  1. Not described as a measure for the trial, but the Barkley Side Effects Rating Scale is mentioned

Notes

Sample calculation: no information

Ethics approval: no information.

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Under acute dosage conditions as used in this study, methylphenidate doses up to 0.9 mg/kg had an increasingly positive effect on measures of mental flexibility and other cognitive processes

Email correspondence with study authors: October 2013. Emailed last study author twice to get supplemental information (protocol, ethics approval, data on side effects, etc.) but received no response. Therefore we included no data from this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Drug order was determined by consecutive assignment to a randomly ordered list of 24 possible combinations of 4 medication levels for each of the 2 testing weeks

Allocation concealment (selection bias)

Low risk

Drug order was determined by consecutive assignment to a randomly ordered list of 24 possible combinations of 4 medication levels for each of the 2 testing weeks. To maximise blindness of examiners, all participants received a different drug order for assessment weeks 1 and 2

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications containing active drug and placebo were prepared in identical opaque gelatin capsules and were administered in a double‐blind fashion

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

To maximise blindness of examiners, all participants received a different drug order during assessment weeks 1 and 2

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated whether any participants were LTFU

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Low risk

Funded by grants from the Medical Research Council of Canada and by William T. Grant Foundation Faculty Scholar Program

Conflicts of interest: none

DuPaul 1996

Methods

Four‐week, double‐blind, placebo‐controlled, cross‐over trial in which participants were randomly assigned to 3 doses of methylphenidate

  1. 0.16 mg/kg

  2. 0.29 mg/kg

  3. 0.42mg/kg

  4. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 24 (19 boys, 5 girls). Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 24

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 11.09 years (range 9 to 15)

IQ: > 70

Methylphenidate naive: not stated

Ethnicity: Caucasian (100%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (21%), conduct disorder (% not reported)

Comedication: not stated

Sociodemographics: Children were primarily from lower middle class and middle class families

Inclusion criteria

  1. Parent and/or teacher referral to an out‐patient ADHD clinic due to reported problems with inattention, impulsivity and/or overactivity

  2. Parent interview indicating that child met DSM‐III‐R (American Psychiatric Association, APA 1987) criteria for ADHD

  3. Independent diagnosis of ADHD by psychologist and paediatrician using DSM‐III‐R criteria for ADHD

  4. Parent or teacher ratings on the Attention problem scale of the Child Behavior Checklist (Achenbach 1991), resulting in a T score ≥ 65 (i.e. 1.5 SD above the mean)

  5. ≥ 9 years old and able to read self report questionnaires independently

Exclusion criteria

  1. Evidence of mental retardation, gross sensory or motor disabilities, seizure disorder, autism, psychosis, tic disorders or Tourette’s syndrome, or significant cardiac problems

  2. Currently receiving psychotropic medication

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of low‐ (0.16 mg/kg; SD 0.08), moderate‐ (0.29 mg/kg; SD 0.11 kg) and high‐dose (0.42 mg/kg; SD 0.14) methylphenidate and placebo

Administration schedule: b.i.d., morning, noon

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Titration period: none

Treatment compliance: No participant was removed from the investigation for non‐compliance (e.g. > 1 day of failure to administer medication as scheduled)

Outcomes

ADHD symptoms

  1. ADHD Rating Scale: teacher and parent ratings

Non‐serious adverse events

  1. Barkley Side Effects Rating Scale (17 items, 0 = absent, severity rated from 0 to 9): rated by participants at the end of each dosage condition

Notes

Sample calculation: no information

Ethics approval: Human Subjects Research Board at the University of Massachusetts Medical Center

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Conclusions based on present findings are limited by several factors

  2. Among others, sample size may have diminished power to detect methylphenidate effects on key variables, especially analyses of side effect ratings

  3. Also results are generalisable only to children with ADHD between 9 and 15 years of age

Email correspondence with study authors: January 2013 to August 2013. Emailed first study author regarding additional information. Not able to get all data requested, as study author no longer has these data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Children were randomly assigned to 1 of 6 possible orders of methylphenidate dosage

Allocation concealment (selection bias)

Low risk

Medication was prepared by the hospital pharmacy in increments of 5 mg and packaged within opaque gelatin capsules

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, their parents and teachers and the research assistant in charge of collecting data were blinded to the order of medication

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants, their parents and teachers and the research assistant in charge of collecting data were blinded to the order of medication

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Analyses on all dependent measures in the study are reported in the article

Vested interest bias

Low risk

No conflicts of interest

Duric 2012

Methods

Randomised, controlled trial, parallel with 3 arms

  1. Neurofeedback group

  2. Methylphenidate group

  3. Methylphenidate + neurofeedback group

Participants

Number of participants screened: 628

Number of participants included: 130

Number of participants randomly assigned: methylphenidate 44, control 42

Number of participants followed up: methylphenidate 30 (23 boys, 7 girls), control 30 (22 boys, 8 girls)

Number of withdrawals: methylphenidate 14, control 12

Diagnosis of ADHD: ICD‐10 (subtype not stated)

Age: methylphenidate 11.2 (SD 2.8), control 11.4 (SD 3.1)

IQ: mean 87

Methyphenidate‐naive: not stated

Ethnicity: not stated

Country: Norway

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. ICD‐10 criteria for diagnosis of ADHD

  2. 6 to 18 years of age

  3. IQ > 70

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to methylphenidate and neurofeedback (intervention group) or to neurofeedback (control group)

Methylphenidate dosage: 20 mg daily to 60 mg daily. No placebo pill

Administration schedule: twice per day

Duration of intervention: 11 to 13 weeks

Titration period: not stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Two core ADHD symptoms – attention and hyperactivity, Assessment of Disruptive Behavior Disorders Rating Scale for parents: rated at baseline and 1 week after neurofeedback had been completed (between weeks 11 and 13 after start of intervention)

Notes

Ethics approval: yes

Comments from study authors

  1. Factors that may result in ADHD symptom improvement: extraordinary amount of time spent with therapist during neurofeedback, better motivation for change in ADHD symptoms and cognitive‐behavioural training induced under neurofeedback

  2. For Attention rating, methylphenidate + neurofeedback and methylphenidate, no significant differences were noted between pre‐treatment and post‐treatment values

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: March to June 2013. Emailed first study author. All questions were answered

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Children with ADHD were randomly placed into 3 groups

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, as it is not a “pure” placebo group

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of parents

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

Low risk

No

Conflicts of interest: Study authors declare no potential conflicts of interests with regard to authorship or publication of this article

Döpfner 2004

Methods

Randomised, double‐dummy, double‐blind, cross‐over, multi‐centre trial with 3 interventions

  1. Immediate‐release methylphenidate 10 mg to 40 mg

  2. Extended‐release methylphenidate 10 mg to 40 mg

  3. Placebo

Phases: Trial was subdivided into 5 stages: pre‐screening, run‐in phase (duration: 1 workday), trial phases 1 and 2 (duration in each case: 4 workdays plus weekend) and trial phase 3 (duration: 4 workdays). Participants also received a behavioural therapy intervention and social skills training at school

Participants

Number of participants screened: not stated

Number of participants included: 82. Participants were randomly assigned to different orders of immediate‐release methylphenidate, extended‐release methylphenidate and placebo

Number of participants followed up: 79 (71 boys, 8 girls)

Number of withdrawals: 3

Regarding the group followed up

Diagnosis of ADHD: DSM‐IV or ICD‐10 (combined (92.4%), hyperactive‐impulsive (0%), inattentive (7.6%))

Age: mean 10 years (range 6 to 16)

IQ: 103 ± 10.4

Methylphenidate naive: 0%

Ethnicity: not stated

Country: Germany

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder/conduct disorder (44%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Meeting criteria for an ICD‐10 diagnosis of Hyperkinetic disorder (F90) or for a DSM‐IV diagnosis of ADHD

  2. Between 8 and 15 years of age

  3. Methylphenidate responders on the basis of clinical assessment and after careful titration

  4. All patients had to be treated with immediate‐release methylphenidate at least twice daily or once daily with a retard preparation

  5. During previous month, methylphenidate dosage had to be unchanged. Daily methylphenidate dosage was ≥ 10 mg

  6. IQ ≥ 85

  7. Body weight > 20 kg

  8. Written informed consent of parents and participants to join the trial

Exclusion criteria

  1. Patients attending schools for mentally handicapped, sensory handicapped or physically handicapped children

  2. Patients who, during the past 4 weeks, were treated with other medication because of ADHD, apart from methylphenidate

  3. Diagnosis of a severe developmental disorder or psychosis

  4. Previous convulsive disorder; EEG indicated susceptibility to convulsions

  5. Case history of pathological changes in liver function or liver disease

  6. Severe depressive disorder (Child Behavior Checklist, teacher‐rated, > 70 on the Anxiety‐depression scale) or a severe anxiety disorder according to clinical diagnosis

Interventions

Participants were randomly assigned

Mean methylphenidate dosage: 22 mg ± 6 mg. Dosage was identical in methylphenidate groups but did not exceed 1 mg/kg body weight. Thus, 9 (11%) participants received a daily dose of 10 mg, 54 (68%) received 20 mg, 14 (17%) received 30 mg and 2 (3 %) received 40 mg.

Administration schedule: 9:00 AM and 1:00 PM

Duration of each medication condition: 4 days, and for trial phase 1 + 2 (also weekends)

Washout before study initiation: none

Medication‐free period between interventions: none

Titration period: had to be oriented to the optimum individual dosage previously determined in clinical treatment trials initiated before randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: rated by clinic personnel/caregiver staff for each child at 9:00 AM, 11:00 AM, 12:30 PM, 3:00 PM and 4.15 PM

  2. Fremdbeurteilungsbogen für Hyperkinetische Störungen: staff/personnel‐rated, at 1:00 PM and 4:45 PM

Non‐serious adverse events

  1. Questionnaire on side effects (Side Effects Rating Scale)

Notes

Sample calculation: not stated

Ethics approval: approved by local university ethics committees

Comment from study authors

  1. Althogh the analogue classroom attempts to mimic many aspects of a regular school classroom, it represents a unique setting that may influence behaviour. Analogue assessments included only ADHD participants; no control or normal participants were available for comparison

Carry‐over effect: As no evidence for possible carry‐over effects was noted, no secondary analyses for carry‐over effects were performed

Key conclusions of study authors

  1. These data provide support for the benefit of this novel, once‐daily methylphenidate preparation for the treatment of ADHD

  2. On all measures analysed, both b.i.d. immediate‐release methylphenidate and extended‐release methylphenidate produced significant improvement relative to placebo. Moreover, extended‐release methylphenidate was not significantly different from b.i.d. immediate‐release methylphenidate, even longer than 7 hours after dosing

  3. Longer duration of action of Medikinet Retard has the potential to simplify psychostimulant treatment, thus reducing dose diversion and eliminating the need for in‐school administration

Comments from review authors

  1. For the article in German (Döpfner 2003 in Döpfner 2004), only 1 review author who knew German extracted the data. All other articles have been assessed for data by 2 review authors. We received Döpfner 2003 from HB Pharma

  2. Eighteen participants in the sample from Clinic for Neuropediatrics, University of Kiel, received response cost token‐based behaviour training (RCT) (Gerber 2012 in Döpfner 2004)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate: yes; included only methylphenidate responders

Email correspondence with study authors. We received some data from study authors in July 2013. We sent 2 additional emails to different study authors to request data in July 2014 but received no answer

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Order in which participants were allocated to respective treatment arms was randomly assigned

Allocation concealment (selection bias)

Unclear risk

Order in which participants were allocated to respective treatment arms was randomly assigned

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

To guarantee a double‐blind trial, the double‐dummy method was used (i.e. participants took the capsule once a day and the tablets twice daily). Only 1 of the 2 galenical forms contained the active substance; the other form contained placebo. In the placebo group, participants took both placebo capsules and placebo tablets

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

No protocol obtained

Vested interest bias

High risk

Study was conducted and sponsored by MEDICE Arzneimittel Pütter GmbH & Co. KG as part of the drug approval process for Medikinet‐RetardTM

Conflicts of interest: Some study authors have affiliations with medical companies

Epstein 2011

Methods

Cross‐over trial with 2 interventions

Three preliminary phases to determine optimal dose followed by 2‐phase trial of the following

  1. Optimal dose of methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: not clear

Number of participants followed up: 93 (numbers of boys and girls: not stated)

Number of withdrawals: not clear

Diagnosis of ADHD: DSM‐IV (combined (n = 45), hyperactive‐impulsive (n = 48), inattentive (n = 0))

Age: mean 8.11 years (range 7 to 11)

IQ: mean 105.58

Methylphenidate naive: 100%

Ethnicity: Caucasian (75%), African American (22%), other (3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 34), conduct disorder (n = 4), anxiety disorders (n = 31), mood disorders (n = 2)

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. Children 7 to 11 years of age who met the diagnostic criteria for ADHD, plus 6 non‐overlapping symptoms in a symptom domain on the Diagnostic Interview for Children ‐ Parent Report and Vanderbilt Teacher Rating Scale; both parents and teachers reported ≥ 4 symptoms in that domain

Exclusion criterion

  1. Children with an IQ below 80 (on the Wechsler Abbreviated Scale of Intelligence) or a score below 80 on the Reading or Numerical operations subtests of the Wechsler Individual Achievement Test or possible organic brain injury

Interventions

Participants were randomly assigned to an optimal dose of methylphenidate and placebo

Mean methylphenidate dosage: 1.13 mg/kg

Administration schedule: testing 1 to 4 hours after medication ingestion

Duration of each medication condition: 1 week

Washout before study initiation: no apparent washout

Titration period: Each of the 3 doses was trialled for 1 week before random assignment to identify an optimal dose

Treatment compliance: not reported

Outcomes

ADHD symptoms

  1. Vanderbilt ADHD Rating Scale: completed by parents and teachers at the end of 1 week

Non‐serious adverse events

  1. Referred to the Pittsburgh Side Effects Rating Scale: completed by parents and teachers at the end of each week, but data were not reported

Notes

Sample calculation: not stated

Ethics approval: not stated

Funding: National Institute of Health (NIH) and National Institute of Mental Health (NIMH)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Although we used a placebo‐controlled, double‐blind titration trial to determine optimal dosage, the highest dosage used in this trial was 54 mg for children ≧ 25 kg and 36 mg for children < 25 kg

  2. Also, a significant minority of children (24%) exited the titration trial, with the placebo dosage as their optimal dosage, which is comparable with the stimulant response rate in other studies. The fact that 19% of children in the optimal dose condition received the same stimulant dosage (i.e. placebo) as was received by the placebo control group may have affected the ability of this study to detect between‐group differences for some study outcomes (e.g. accuracy)

Key conclusion of study authors

  1. None regarding our outcomes of interest

Email correspondence with study authors: April 2014. We were able to obtain supplemental information regarding data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Double‐blind"; capsules were "identical" (p 2, p 1063)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Double‐blind", capsules were "identical" (p 2, p 1063)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not clear whether any participants were LTFU

Selective reporting (reporting bias)

Low risk

No selective reporting

Vested interest bias

Low risk

No evidence of vested interests

No evidence of conflicts of interest

Fabiano 2007

Methods

Randomised, double‐blind, within‐participant design, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 0.15 mg/kg, 0.30 mg/kg and 0.60 mg/kg. Medication was randomly assigned for each child and varied daily during a 9‐week summer treatment programme

Participants

Number of participants screened: not stated

Number of participants included: 48 (44 boys, 4 girls). Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 47

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (combined (not stated), hyperactive‐impulsive (not stated), inattentive (not stated))

Age: mean 9.35 years (SD 1.98, range 5 to 12)

IQ: 106.33 (SD 14.61)

Methylphenidate naive: not stated

Ethnicity: Caucasian (79%); African American (12.5%); Hispanic, Native American or mixed race (8.5%)

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, diagnostic criteria for ADHD

  2. Estimated full‐scale IQ ≥ 80

  3. No documented adverse response or non‐response to methylphenidate

  4. No medical condition that would contraindicate use of methylphenidate

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of immediate‐release methylphenidate (0.15 mg/kg, 0.30 mg/kg and 0.60 mg/kg) and placebo

Mean methylphenidate dosage: 5.03 mg (range 2.5 to 10), 10.8 mg (range 5 to 20) and 21 mg (range 12.5 to 30)

Administration schedule: 3 times daily (7:45 AM, 11:45 PM and 3:45 PM)

Duration of each medication condition: varied daily

Washout before study initiation: none

Titration period: none/duration initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale (inattention‐impulsivity‐overactivity): teacher‐ and observer‐rated, daily

General behaviour

  1. IOWA Conners' Rating Scale (oppositional defiance): teacher‐ and observer‐rated, daily

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale: teacher‐rated daily

  2. Clinically significant adverse events: observer‐rated daily

Notes

Sample calculation: no

Ethics approval: yes

Comments from study authors

  1. "The study was conducted in an analogue classroom setting" (as opposed to a community classroom)

  2. "The treatments used were of short duration"

  3. "Observers and raters were blind to medication condition but not to behaviour modification conditions"

  4. Ratings from the Pittsburgh Side Effects Rating Scale were averaged across days within drug condition (regardless of behaviour modification condition) for the 47 children. One participant's medication was discontinued because of parental concerns about side effects (mainly buccal‐lingual movements) after 2 days of treatment, and 1 child's afternoon dose was reduced on 0.60 mg/kg days because of parent‐reported anxiety and mood symptoms. No other children had side effects rated by the teacher at an average level of moderate or severe

Comments of review authors

  1. This study of placebo versus methylphenidate was conducted during different conditions of behaviour modification. We should consider whether to use only data from the non‐behaviour modification condition or data from all conditions

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: April 2014. We obtained supplemental information regarding risk of bias. No further information was received

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation and allocation concealment were completed with a random number generator by a researcher not involved in treatment

Allocation concealment (selection bias)

Low risk

Randomisation and allocation concealment were completed with a random number generator by a researcher not involved in treatment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Medication assessment procedure was double‐blinded" (p 201) "children, their parents, and all clinical staff members were blinded to medication condition" (p 202)

Medication was prepared in opaque capsules by a pharmacist not otherwise involved in the study. It was administered to children by research staff who were not involved in administration of behavioural treatment nor in daily activities

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Observers and raters were blinded to medication conditions

Incomplete outcome data (attrition bias)
All outcomes

High risk

One child's parents elected to stop medication for the child after 2 days because of their concerns about possible side effects of the medication. This child was not included in the analyses

Selective reporting (reporting bias)

Low risk

No protocol/design was published. All pre‐specified outcomes of interest have been reported

Vested interest bias

Low risk

This study was funded by the National Institute of Mental Health (NIMH) grant MH62946

Conflicts of interest: supported only by National Institutes

Findling 2006

Methods

Three‐week, randomised, double‐blind, parallel trial with 3 arms

  1. Immediate‐release methylphenidate (Ritalin)

  2. Modified‐release methylphenidate (Equasym, EqXL)

  3. Placebo

Participants

Number of participants screened: 346

Number of participants randomly assigned: 327

Number of participants included: 318; immediate‐release methylphenidate 133, modified‐release methylphenidate (EqXL) 139, placebo 46

Number of participants followed up: immediate‐release methylphenidate 120, modified‐release methylphenidate (EqXL) 120, placebo 39 (number in each arm is only per protocol population)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (combined (71%), hyperactive‐impulsive (6%), inattentive (23%))

Age: mean 9.5 years (range not reported)

IQ: above 80

Sex: 252 boys, 66 girls

Methylphenidate naive: 0%

Ethnicity: Caucasian (86%), African Caribbean (5.2%), Asian (0.3%), Hispanic (1.6%), other (6.9%)

Country: Australia, Canada, USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Male and female children

  2. 6 to 12 years of age

  3. Stable dose of methylphenidate 3 weeks before screening

  4. Diagnosed with ADHD on the basis of DSM‐IV criteria for any subtype and confirmed by administration of the Kiddie‐Sads‐Present and Lifetime Versions interview at screening

  5. Attending a school setting in which a single teacher could make morning and afternoon assessments of the child’s behaviour

Exclusion criteria

  1. Female who had experienced menarche

  2. Comorbid psychiatric disorder requiring medication

  3. History of seizure or tic disorder or family history of Tourette’s disorder

  4. IQ test score below 80, or functioning at a level of intelligence indicative of an IQ below 80

  5. Use of unapproved medication(s)

  6. Use of an investigational product within 30 days before study entry

  7. Concurrent chronic or acute illness, disability or medication that might confound the results of rating tests

  8. Diagnosed with hyperthyroidism, glaucoma or eating disorder

  9. Current substance abuse disorder or living with someone with a current substance abuse disorder

  10. Demonstrated lack of response to methylphenidate

Interventions

Participants were randomly assigned to immediate‐release methylphenidate, extended‐release methylphenidate or placebo

Mean methylphenidate dosage: not stated

Administration schedule: twice daily, morning and lunch

Duration of intervention: 3 weeks

Titration period: All participants were stable while taking methylphenidate medication before randomisation

Treatment compliance: not stated. Children with a previous total daily dose of 10 mg to 20 mg immediate‐release methylphenidate or 20 mg extended‐release methylphenidate were randomly assigned to receive 10 mg immediate‐release methylphenidate twice daily, 20 mg modified‐release methylphenidate (EqXL) once daily or placebo; children given a previous total daily dose of 25 mg to 40 mg immediate‐release methylphenidate of > 20 mg to < 40 mg extended‐release methylphenidate were randomly assigned to receive 20 mg immediate‐release methylphenidate twice daily, 40 mg modified‐release methylphenidate (EqXL) once daily or placebo; children given a previous total daily dose > 40 mg immediate‐release methylphenidate or < 40 mg extended‐release methylphenidate were randomly assigned to receive 20 mg immediate‐release methylphenidate twice daily, 60 mg modified‐release methylphenidate (EqXL) once daily or placebo

Outcomes

ADHD symptoms

  1. Swanson, Nolan and Pelham, Fourth Edition: teacher‐rated at baseline and weekly

  2. Swanson, Nolan and Pelham, Fourth Edition: parent‐rated at baseline and weekly, at the end of the child’s day

  3. IOWA Conners' Scale (Inattention‐Impulsivity‐Overactivity): teacher and parent

General behaviour

  1. IOWA Conners' Rating Scale (oppositional defiance): teacher‐ and parent‐rated, at baseline and weekly (2 hours post dose and 2 to 4 hours post lunch)

Serious adverse events

  1. Only 1 serious adverse event (neutropenia) was reported during the study in EqXL treatment, but the investigator considered it unlikely to be related to the study medication

Non‐serious adverse events

  1. Barkley Side Effects Rating Scale: parent‐ and teacher‐rated at baseline and weekly

  2. Adverse events, laboratory parameters, vital signs, physical exam, observer‐rated, time point not specified

Notes

Sample calculation: yes

Ethics approval: yes; independent ethics committee at each clinical site before study initiation

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Key conclusion of study authors

  1. Modified‐release methylphenidate (EqXL) given once daily was non‐inferior to immediate‐release methylphenidate given twice daily. Both treatments were superior to placebo in reducing ADHD symptoms

Comment from review authors

  1. September 2013. Not possible to contact study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomised" but did not state how

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both EqXL (modified‐release methylphenidate) capsules and immediate‐release methylphenidate tablets were overencapsulated in hard gelatin capsules identical to the placebo capsule

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Stated LOCF but primary efficacy population was the per protocol (PP) population, defined as participants who received study treatment and had ≥ 1 efficacy measurement after the first dose

Selective reporting (reporting bias)

Low risk

No protocol/design was published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Funding for this study was provided by Celltech Americas Incorporated, currently part of UCB (Union Chimique Belge)

Conflicts of interest: Drs. Hatch and DeCory and Miss Cameron were employees of Celltech at the time of this study. Dr. Findling received research support, acted as a consultant and/or served on a Speakers' Bureau for Abbott, AstraZeneca, Bristol‐Myers Squibb, Celltech‐Medeva, Forest, GlaxoSmithKline, Johnson & Johnson, Lilly, New River, Novartis, Otsuka, Pfizer, Sanofi‐Synthelabo, Shire, Solvay and Wyeth. Dr. Quinn claims no competitive interests. Dr. McDowell has consulted for Janssen‐Cilag and Lilly

Findling 2007

Methods

Four‐week, double‐blind, randomised, placebo‐controlled, cross‐over trial with 4 interventions

Methylphenidate (at 3 different doses, in the morning and at midday)

  1. 5 mg

  2. 10 mg

  3. 15 mg

  4. Placebo

Phases

Participants were assigned to receive, at random, 1 of 6 possible dosing orders that included the following

  1. Placebo, 5 mg, 10 mg, 15 mg

  2. Placebo, 10 mg, 15 mg, 5 mg

  3. 5 mg, placebo, 10 mg, 15 mg

  4. 5 mg, 10 mg, 15 mg, placebo

  5. 10 mg, 15 mg, placebo, 5 mg

  6. 10 mg, 15 mg, 5 mg, placebo

Schedules were designed in such a way that participants did not receive the 15 mg dose before the 10 mg dose, so in the event that a participant experienced adverse events while taking a lower dose, the 15 mg dose was not administered

Participants

Number of participants screened: not stated

Number of participants included: 20. Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 16 (12 boys, 4 girls)

Number of withdrawals: 4

Demographic data regarding the 16 who completed the study

Diagnosis of ADHD: DSM‐IV (combined (94%), inattentive (6%))

Age: mean 10.43 years (range 5 to 17)

IQ: > 70

Methylphenidate naive: not stated

Ethnicity: White (75%), African American (6%), Hispanic (19%)

Country: USA

Setting: out‐patient clinic

Comorbidity: bipolar (100%), oppositional defiant disorder (50%), conduct disorder (25%), enuresis (12.5%), encopresis (12.5%)

Comedication: 100% divalproex sodium. Some received clonidine for sleep at night

Sociodemographics: not stated. No statistically significant differences in distribution based on sex, ethnicity, age group, rate/proportion of comorbid oppositional defiant disorder or comorbid conduct disorder were found between 6 six dosing order groups

Inclusion criteria

  1. 5 to 17 years of age

  2. Individuals meeting DSM‐IV criteria for a diagnosis of bipolar spectrum disorder and a comorbid diagnosis of ADHD were eligible for study participation

  3. Treated with fixed doses of mood stabilisers at the time of study enrolment for ≥ 5 days before receiving study medication

  4. Eligible if study physician’s clinical assessment indicated the need for a psychostimulant for treatment of "dysfunctional residual symptoms of ADHD"

Exclusion criteria

  1. Mental retardation

  2. Pervasive developmental disorder

  3. Inability to swallow pills

  4. History of alcohol or other substance abuse or dependence within 6 months before enrolment

  5. Active neurological or other medical condition suspected to be related to mood symptoms

  6. Pregnant females, those intending to become pregnant and sexually active female patients who were using an inadequate form of birth control were not permitted to participate

  7. Participation required a negative qualitative pregnancy test within 2 weeks of receiving the first dose of double‐blind treatment for female patients of childbearing potential

  8. Significant symptoms of mania (Young Mania Rating Scale score > 13) or depression (CDRS‐R > 40) during the week before enrolment and anticipated dosing changes for mood‐stabilising agents

  9. Individuals receiving a tricyclic antidepressant or antipsychotic agent and those with symptoms of psychosis or suicidal ideation

  10. Females nursing an infant and patients experiencing significant medical or neurological illness were not permitted to participate

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate (5 mg b.i.d., 10 mg b.i.d., 15 mg b.i.d.) and placebo

Mean methylphenidate dosage: not provided, as the trial compares methylphenidate at different doses

Administration schedule: morning and midday

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: between lunchtime dose and morning dose the following day

Titration period: none. Dosing schedules were designed in such a way that patients did not receive the 15 mg dose before the 10 mg dose, so in the event that a participant experienced adverse events while on a lower dose, the 15 mg dose was not administered

Treatment compliance: One of 20 screened participants was withdrawn from the study because of poor compliance. Among the 16 who participated, no compliance issues were reported

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition: rated weekly by parents

  2. Conners' Parent Rating Scale (48‐item): rated weekly

General behaviour

  1. Conners' Parent Rating Scale, Conduct problem subscale

Non‐serious adverse events

  1. Child Depression Rating Scale ‐ Revised: rated weekly

  2. Young Mania Rating Scale: rated weekly.

  3. Side Effects/Behavior Monitoring Scale: rated weekly at the study visit

  4. Resting blood pressure and pulse recorded each week

  5. Weight documented at baseline and at end of study

Notes

Sample calculation: yes

Ethics approval: yes

Comments from study authors (limitations)

  1. Small sample size

  2. Full consideration of dosing order effects was not possible because of the modest size of this study cohort

Key conclusion of study authors

  1. Euthymic youths with bipolar disorder and ADHD may benefit from short‐term concomitant treatment with methylphenidate

Comment from review authors

  1. All participants have a bipolar disorder

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes; 2

Email correspondence with study authors: November 2013. We wrote to the study author twice to request a copy of the protocol and information about sample size and allocation concealment but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A random numbers table was generated and was used by a pharmacist to assign dose orders to participants. Counterbalancing was applied in such a way that as each dose order was used, its number was eliminated from the next dose order assignment

Allocation concealment (selection bias)

Low risk

Placebo and methylphenidate in identical capsules. No participants were discontinued from this trial because of broken integrity of the blind

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

At the conclusion of the 4‐week study, study physician, participant and participant's guardian determined a "best dose week", taking into consideration behaviour ratings and reports of any adverse events. After all assessments had been completed, the study blind was broken to reveal the dose that had been prescribed during the previously identified 'best dose week'. No participants were discontinued from this trial because of broken integrity of the blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"After all of the assessments had been completed, the study blind was then broken to reveal the dose that had been prescribed during the previously identified 'best dose week'"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All data present

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Unclear risk

Stanley Medical Research Institute

Conflicts of interest: Some study authors have affiliations with pharmaceutical companies

Findling 2008

Methods

Seven‐week randomised, phase III, double‐blind, multi‐centre, parallel‐group, placebo‐controlled, naturalistic home and school trial with 3 arms

  1. Methylphenidate transdermal system patch + placebo capsule

  2. OROS methylphenidate capsule + placebo patch

  3. Placebo capsule + placebo patch

Five‐week titration phase, 2‐week maintenance phase

Participants

Number of participants screened: not stated

Number of participants included: 282 (187 boys, 95 girls)

Number of participants randomly assigned (and administered ≥ 1 dose of study medication): methylphenidate transdermal system 100, OROS methylphenidate 94, placebo 88

Number of participants followed up: methylphenidate transdermal system 71, OROS methylphenidate 66, placebo 32

Number of withdrawals: methylphenidate transdermal system 27, OROS methylphenidate 25, placebo 53

Diagnosis of ADHD: DSM‐IV‐TR (combined (80.5%), hyperactive‐impulsive (1.4%), inattentive (17.0%), unclassified (1.1%))

Age: mean 8.8 years (range 6 to 12). Methylphenidate transdermal system 8.9, OROS methylphenidate 8.8, placebo 8.5

IQ: ≥ 80

Methylphenidate naive: 86%

Ethnicity: Caucasian (77.3%), African American (14.5%), Asian (0.7%), Hispanic (not stated), other (7.4%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated. No significant differences in baseline demographics (age, sex, ethnicity, ADHD subtype, prior ADHD medication use) among the 3 groups

Inclusion criteria

  1. 6 to 12 years of age, inclusive

  2. ADHD, DSM‐IV‐TR

  3. Stimulant‐naive or known to be stimulant‐responsive

  4. IQ ≥ 80

  5. Total score of 26 on ADHD Rating Scale, Fourth Edition, while unmedicated

  6. Normal laboratory parameters and vital signs, including ECG

  7. Females of childbearing potential must have a negative serum beta human chorionic gonadotropin (HCG) pregnancy test at screening and a negative urine pregnancy test at baseline

Exclusion criteria

  1. Comorbid psychiatric diagnosis (except oppositional defiant disorder)

  2. History of seizures during the past 2 years

  3. Tic disorder

  4. Any concurrent illness or skin disorder that might compromise safety or study assessments

  5. Ingestion of clonidine, atomoxetine, antidepressants, antihypertensives, investigational medications, hepatic or cytochrome (p 450) enzyme‐altering agents, medications with central nervous system effects, sedatives, antipsychotics or anxiolytics within the 30 days before study entry

  6. Overweight (body mass index (BMI)‐for‐age > 90th percentile)

Interventions

Participants were randomly assigned to methylphenidate patch, OROS methylphenidate or placebo

Titration period: 5 weeks (after randomisation) of optimisation to 1 of 4 total daily dosage strength. OROS methylphenidate: 18 mg, 27 mg, 36 mg and 54 mg. Methylphenidate transdermal system and placebo transdermal system: 10 mg, 15 mg, 20 mg and 30 mg over a 9‐hour period in patches of 12.5 cm², 18.75 cm² , 25 cm² and 37.5 cm², respectively

Administration schedule: Treatments were administered at approximately 7:00 AM each morning; patches were applied to the hip area and were worn for approximately 9 hours daily, different hip each day

Mean patch wear time: 8.70 (0.51) to 9.46 (0.53) hours

Duration of intervention: 7 weeks

Washout before study initiation: up to 28 days if applicable

Treatment compliance: mean compliance 97% to 99% during both study phases

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition: clinician‐rated, weekly

  2. Conners' Teacher Rating Scale: teacher‐rated, twice weekly

  3. Conners' Parent Rating Scale: parent‐rated, twice weekly

Non‐serious adverse events

  1. Evaluations for safety were performed at the end of each week during both dose‐optimisation and dose‐maintenance phases. Furthermore, adverse events were spontaneously reported as coded via MedDRA (7.0) Adverse Event Dictionary

  2. Blood pressure, pulse, oral temperature, weight: weekly

  3. Laboratory parameters: week 7

  4. Sleep‐related behaviours rated by parents each week using Children`s Sleep Habits Questionnaire

  5. Skin reactions due to the patch investigated each week; Dermal Response Scale

  6. ECG: final dose‐optimisation visit and final study visit

Notes

Sample calculation: yes (258 participants)

Ethics approval: yes

Comments from study authors

  1. It is important to note, however, that effects reported are for baseline and endpoint reports of sleep problems; thus, these results may not generalise to the titration period. For example, sleep problems may have resulted in dosage adjustments and attenuation of sleep problems during titration

  2. Respondents did not enter the maintenance phase if spontaneously reported side effects could not be controlled by adjusting the dose. Thus, participants with extreme insomnia would not have entered the maintenance phase

  3. Rating scales for sleep problems may lack validity

Key conclusions of study authors

  1. Results of this study suggest that the methylphenidate transdermal system is an efficacious treatment option for children with ADHD

  2. Results of our analysis suggest that emergence or worsening of sleep problems in response to treatment of ADHD symptoms with OROS methylphenidate or methylphenidate transdermal system generally should not be a major concern to clinicians, children with ADHD or their parents after titration to an optimal dose, as described for this protocol. However, these suggestions should be considered in the light of other research that supports an effect of methylphenidate on insomnia and other sleep difficulties, especially when methylphenidate will be administered to children with pre‐existing sleep difficulties. These findings should not be generalised to children who have not been titrated to an optimal dose

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes. An inclusion criterion is that participants are stimulant‐naive or stimulant‐responsive

Any withdrawals due to adverse events: yes; 11

Email correspondence with study authors. June to September 2013. We attempted to obtain supplemental efficacy and safety data from study authors but without success

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers schedule

Allocation concealment (selection bias)

Low risk

Computer‐generated random numbers schedule

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, double‐dummy: Participants received both a patch and a capsule to be administered each day. Methylphenidate and placebo capsules were overencapsulated to blind the identity of the capsule’s content

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

ITT, LOCF

Selection bias: yes; non‐responders excluded during the study. Participants who did not reach an acceptable condition by the final dose‐optimisation visit (week 5) were withdrawn from the study

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

High risk

This study was funded by Shire Development Incorporated, Wayne, Pennsylvania

Conflicts of interest: Some study authors received research support, acted as consultants and/or served on a Speakers' Bureau for several pharmaceutical companies

Findling 2010

Methods

Phase IIIB, randomised, double‐blind, parallel‐group, placebo‐controlled, multi‐centre, dose‐optimisation study evaluating the efficacy and safety of the following in adolescents 13 to 17 years of age with ADHD

  1. Methylphenidate transdermal system (10‐, 15‐, 20‐ or 30‐mg/9‐hour patches)

  2. Placebo transdermal system

Study consisted of 4 experimental periods

  1. Screening and washout

  2. Dose optimisation (5 weekly visits)

  3. Dose maintenance (5 monthly visits)

  4. Seven‐day post‐treatment follow‐up

Follow‐up consisted of an open‐label extension study conducted to evaluate the safety and efficacy of the methylphenidate transdermal system (10‐, 15‐, 20‐ or 30‐mg/9‐hour patches) for participants who completed all required study visits; consisted of 3 experimental periods

Participants

By using 85% power to detect an effect size of 0.5 between active treatment and placebo at the significance level of 5%, it was estimated that 112 participants were needed for methylphenidate transdermal system groups and 56 for placebo transdermal system groups. Assuming a 20% drop‐out rate, ˜ 210 participants (methylphenidate transdermal system 140, placebo transdermal system 70) were required for the study

For the double‐blind, randomised, controlled trial

Number of participants screened: not stated

Number of participants included: 217 (162 boys, 55 girls)

Number of participants randomly assigned: methylphenidate 145, placebo 72

Number of participants followed up (ITT population): methylphenidate 143, placebo 72

Number of withdrawals: methylphenidate 2, placebo 0

Number that completed 7‐week dose‐optimisation/dose‐maintenance phase: methylphenidate 95, placebo 72

Diagnosis of ADHD: DSM‐IV (types not stated)

Age: mean 14.6 years (SD 1.3, range 13 to 17)

IQ: ≥ 80

Methylphenidate naive: 122 (56%)

Ethnicity: Caucasian (77%), African American (18%), Asian (0.5%), other (4.5%)

Country: USA

Setting: out‐patient clinic

Comorbidity: 0%

Comedication: not stated

Sociodemographics: not stated. No significant difference in baseline demographics were noted between the 2 groups

For the open‐label extension (regarding safety measures)

Number of participants included: 163 (previously taking methylphenidate 110, placebo 53)

Number of participants followed up: 162 (121 boys, 41 girls)

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (types not stated)

Age: 14.5 years (SD 1.24, range 13 to 17)

IQ: ≥ 80

Methylphenidate naive: 122 (56%)

Ethnicity: Caucasian (78%), African American (17%), Asian (0.6%), other (4.4%)

Country: USA

Comorbidity: 0 %

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Male or female adolescents

  2. 13 to 17 years of age

  3. Primary diagnosis of ADHD according to DSM‐IV

  4. IQ score > 80

  5. Total score ≥ 26 on the ADHD Rating Scale, Fourth Edition, at baseline

  6. Participants were required to have ECG results within normal range or variants that were not clinically significant, as judged by investigators in conjunction with the central laboratory

  7. Blood pressure measurements within the 95th percentile for age, sex and height

  8. No current or past skin disease or other skin problems, including sensitive skin or signs of skin irritation

  9. Females must have a negative urine pregnancy test at entry and must agree to use acceptable contraceptives throughout the study period and for 30 days the last dose of IP

Participant and parent of legally authorised representative (LAR) are able, willing and likely to fully comply with study procedures and restrictions

  1. Regarding the 6‐month open‐label study: Participants must have completed all required study visits or a 5‐week dose‐optimisation period without achieving an acceptable condition (i.e. ≧ 25% decrease from baseline in a participant's ADHD Rating Scale, Fourth Edition, score with minimal side effects)

Exclusion criteria

  1. Conduct disorder or comorbid psychiatric illness (such as clinically significant obsessive‐compulsive disorder, depressive or anxiety disorder; post‐traumatic stress disorder; psychosis; bipolar illness; or pervasive developmental disorder); history of structural cardiac abnormality, cardiomyopathy, cardiac rhythm abnormalities or other serious cardiac problems; suicidal ideation; alcohol or other substance abuse (except caffeine or nicotine) within the past 6 months

  2. Seizures during the previous 2 years and a history of being non‐responsive to psychostimulant treatment; use of clonidine, atomoxetine, antidepressants, sedatives, antipsychotics, anxiolytics, P450 enzyme‐altering agents or other investigational medications within 30 days before screening

  3. Female participant who is pregnant or lactating

Regarding the 6‐month open‐label study

  1. Participants were not eligible to participate in the extension study if they were discontinued from the antecedent study because of a protocol violation (including non‐compliance) or had experienced an adverse event for which continued treatment would be medically contraindicated, or a serious adverse event

  2. Participants with considerable general medical illness (except mild, stable asthma) or an unstable medical condition, disability or other condition the investigator believed might interfere with or prevent completion of the study

Interventions

Participants were randomly assigned to methylphenidate transdermal (patches) or placebo

Mean methylphenidate dosage at week 7: 10 mg (4.2%), 15 mg (16.7%), 20 mg (24.0%), 30 mg (55.2%); median exposure time: 48 days (range 4 to 57)

Administration schedule: single patch in the morning, once daily for 9 hours

Duration of intervention: 7 weeks

Titration period: 5 weeks, initiated after randomisation

Treatment compliance: 124 fulfilled the protocol. However, it is not stated in the article how compliance regarding the medication had to be assessed to fulfil the protocol

Mean methylphenidate dosage at month 6: 10 mg (5.6%), 15 mg (7.9%), 20 mg (32.6%), 30 mg (53.9%); median exposure time: 168 days (range 3 to 200)

Administration schedule: single patch in the morning, once daily for 9 hours

Duration of intervention: 6 months

Titration period: 5 weeks of the 6 months

Treatment compliance: not stated. 88 fulfilled the protocol. However, it is not stated in the article how compliance regarding the medication had to be assessed to fulfil the protocol

Outcomes

Regarding the 7‐week parallel study

ADHD symptoms

  1. ADHD Rating Scale, Clinicians: rated baseline and weekly for 7 weeks (each study visit)

  2. Conners' Parent Rating Scale ‐ Revised, Parents: rated baseline and weekly for 7 weeks (each study visit)

Serious adverse events

  1. None

Non‐serious adverse events

  1. Adverse events monitored at each study visit. All AEs were coded using the MedDRA, Version 7.0

  2. Height measurements were performed at screening and at the final double‐blind study visit

  3. Vital signs (systolic and diastolic blood pressure, pulse) and weight, at screening, at baseline and at each study visit

The investigator determined the clinical significance of any physical examination, height or vital sign measurement that was outside the normal range. Clinically significant deviations from measurements recorded at screening were reported as AEs. A 12‐lead ECG evaluation was obtained at screening, at baseline, at week 5 and at the final double‐blind study visit.Dermal skin reaction: Dermal Scale (DRS) was used to evaluate observed skin findings: range 0 to 7, where 0 shows no evidence of irritation

Regarding 6‐month open‐label study

  1. Dose optimisation (5 weekly visits); dose maintenance (5 monthly visits)

    1. AEs were monitored at each study visit and were assessed by an open‐ended inquiry along with specific dermatological questions asked by an investigator or a qualified evaluator. AEs were considered treatment‐emergent if they began or worsened on or after application of the first patch, and occurred before or at the same time as application of the patch. AEs coded and defined using the MedDRA, Version 7.03, at 7‐day post‐treatment follow‐up

    2. Height: measured at month 6 visit by the investigator

    3. Weight: recorded at all visits (5) by the investigator

    4. Vital signs (systolic blood pressure, diastolic blood pressure, pulse): measured at all study visits by the investigator. 12‐Lead ECG performed at entry, week 4, month 3 and month 6 by the investigator

    5. Blood and urine samples collected at entry, week 4, month 4 and month 6

    6. Dermal skin reaction: measured by DRS at each study visit.

    7. Sleep: measured by non‐validated Post‐Sleep Questionnaire. Measured at 6‐month visit

Changes noted between evaluation data at study entry and data obtained at schedule visits deemed to be clinically significant by the investigator were considered an adverse event

Notes

Sample calculation: yes

Ethics approval: yes

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Given the short duration of this study, results do not characterise the long‐term effects of treatment with methylphenidate transdermal system

  2. It is important to note that participants who failed to respond to psychostimulants in the past and those with conduct disorder and other psychiatric comorbidity were excluded from the study

  3. Regarding the 6‐month study: no clinically significant findings between laboratory evaluation parameters obtained post entry relative to screening values obtained at the antecedent study

  4. Limitations: This study used an open‐label design; thus tolerability and effectiveness assessments are susceptible to observer bias. Another way in which results of long‐term, open‐label continuation studies may be biased is that participants who enrol in a study after participating in an antecedent trial may represent that subset of patients who have had improvement in ADHD symptoms and/or who did not experience substantial adverse events in the antecedent study

Key conclusions of study authors

  1. Methylphenidate transdermal system therapy was generally well tolerated and resulted in significantly greater improvement in ADHD symptoms among adolescents when compared with the placebo transdermal system

  2. Reported adverse events included those typically observed for oral methylphenidate, with the exception of generally mild application site erythema associated with transdermal delivery

  3. Regarding the 6‐month open‐label study: Methylphenidate transdermal system was generally well tolerated, and adverse events were generally typical of those associated with oral methylphenidate, with the exception of application site reactions associated with transdermal delivery of methylphenidate

Comments from review authors

  1. As already stated, people who earlier had failed to respond to psychostimulants were not included in the study. Therefore, results can be generalised only to responders

  2. Risk of bias table done only for the 7‐week parallel‐group study

  3. Regarding the 6‐month open‐label study: Participants who had not reached an acceptable response by the end of week 5 were withdrawn from the study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were allocated the next sequential randomisation number and received treatment that corresponded with that randomised number as given by an interactive voice response system

Allocation concealment (selection bias)

Low risk

Randomisation schedule was produced by computer software that incorporated a standard procedure for generating random numbers

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, but no information on methylphenidate and placebo blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

Efficacy analyses were performed on the ITT population, defined as all randomly assigned participants who received ≥ 1 dose of study medicine and ≥ 1 post‐baseline primary efficacy assessment. Safety population was defined as all randomly assigned participants who received ≥ 1 dose of study medicine.

Selection bias (e.g. titration after randomisation → exclusion): yes. No further dose titration was permitted for any participant after week 5, and participants who had not reached an acceptable response by the end of week 5 were withdrawn from the study

Selective reporting (reporting bias)

Low risk

Protocol registered 12 July 2007. First participant consent was obtained 29 August 2007 for the open‐label study

Vested interest bias

High risk

This study was funded by Shire Development Incorporated, which was involved in study design, conduct and data analysis. The open‐label study was industry‐sponsored

Conflicts of interest: Dr. Findling has acted as consultant to, has served on Speakers' Bureaus of and/or has received research support from Abbott, Addrenex, AstraZeneca, Biovail, Bristol‐Myers Squibb, Eli Lilly, Forest Pharmaceuticals, GlaxoSmithKline, KemPharm, Johnson & Johnson, Lundbeck, Neuropharm, Novartis, Noven, Organon, Otsuka, Pfizer, sanofi‐aventis, Sepracor, Shire, Solvay, Supernus, Validus and Wyeth. Dr. Turnbow receives or has received research support, acted as a consultant and/or served on Speakers' Bureaus for Eli Lilly, Novartis US, sanofi‐aventis, Shire and UCB (Union Chimique Belge). Dr. Burnside has acted as consultant to, has served on Speakers’ Bureaus of and/or has received research support from Eli Lilly, Johnson & Johnson, Shire and Wyeth. Dr. Melmed has acted as consultant to, has served on Speakers' Bureaus of and/or has received research support from Bristol‐Myers, Eli Lilly, McNeil, Novartis and Shire. Drs. Civil and Li are full‐time employees of Shire Development Incorporated

Fine 1993

Methods

Three‐week double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate b.i.d. (2 doses: 0.3 mg/kg and 0.6 mg/kg)

  2. Placebo b.i.d.

Phases: medical trial or typical clinical procedure, concluded with recommendation for treatment, follow‐up (6 weeks and 3 months)

Participants

Number of participants screened: 24

Number of participants randomly assigned: 12 "typical clinical procedure", 12 "medical trial"

Number of participants included: 12 (sex not stated). Participants in the medical trial group were randomly assigned to different possible drug condition orders

Number of participants followed up: 12

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 101.58 months (range 6 to 10 years)

IQ: not stated

Methylphenidate naive: 12

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics (socioeconomic status calculated using Blishen Index; lower score indicates higher SES): medical trial: mean 3.42 (SD 0.90); typical clinical procedure: mean 3.50 (SD 1.88)

Inclusion criterion

  1. ADHD diagnosis. Parent interview with psychiatrist, several parent (mother AND father) and teacher ratings. Included if any positive

Exclusion criteria

  1. Physical or intellectual handicap

  2. Tic disorders

Interventions

Participants in the medical trial group were randomly assigned to different possible drug condition orders of 0.3 mg/kg and 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: b.i.d.

Duration of each medication condition: "the different interventions were randomly assigned across days"

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: not stated

Treatment compliance: missed pills mean 1.92 (SD 1.44). Furthermore, compliance was assessed by urine test. By 6‐week follow‐up, parent‐reported compliance was 83%; by 3 months, it was 73%

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Rating Scale: rated on weekdays by teachers and on weekends by parents

  2. Swanson, Nolan and Pelham: rated daily, but not stated by whom

Non‐serious adverse events

  1. Side effects questionnaire: rated weekly by parents ("Across the 12 children, ratings were available from an average of 2.58 placebo days, 1.33 low‐dose MPH days and 1.75 high‐dose MPH days")

Notes

Sample calculation: not stated

Ethics approval: not stated

Comments from study authors

  1. Small sample size

  2. Rely on parent ratings to assess side effects

Key conclusions of study authors

  1. Fine 1993: "Our analyses revealed that several side‐effects appeared equally often on placebo as on active medication and the parents’ reports of side effects are significantly related to reports of ADHD symptomatology" (p 28)

  2. Johnston 1993 (in Fine 1993): "In summary, this study finds mixed support for the prediction that medication trials would enhance acceptability, satisfaction, and compliance associated with MPH" (p 728)

Comments from review authors

  1. This trial aims to examine differences in attitudes of parents whose children participate in a medical trial or are subjected to "typical clinical procedure"

  2. Only the medical trial group represents a cross‐over design

  3. Fine 1993 deals only with the medical trial group, and Johnston 1993 (in Fine 1993) deals with both

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: June 2014. Emailed study authors for supplemental information and received the following response: "I did look at the questions, but unfortunately, given how long ago the study was conducted, I do not still have the data necessary to answer" (Krogh 2014b [pers comm]

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

24 children were matched in pairs on sex and age and then were randomly assigned to the medication trial or to the typical clinical procedure. Higher and lower doses of methylphenidate and placebo were randomly assigned across days by the hospital pharmacy

Allocation concealment (selection bias)

Low risk

Higher and lower doses of methylphenidate and placebo were randomly assigned across days by the hospital pharmacy. Active medication and placebo were packaged in capsule form to disguise taste and visual differences, and were dispensed in envelopes for daily use

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, teacher, child and resident were blinded to the daily medication status of the child. "Active medication and placebo were packaged in capsule form to disguise taste and visual differences, and were dispensed in envelopes for daily use". All evaluations were conducted by psychiatric residents blinded to the hypotheses of the study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parents, teacher, child and resident were blinded to the daily medication status of the child. All evaluations were conducted by psychiatric residents blinded to the hypotheses of the study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Across 12 children, ratings were available for an average of 2.58 placebo days, 1.33 low‐dose methylphenidate days and 1.75 high‐dose methylphenidate days. Ratings for each child were averaged across each of the 3 treatment conditions. Data for all participants were available for measures of missed pills and appointments. One participant was missing on measures of parent‐reported compliance at 6‐week follow‐up, and 2 participants at 3‐month follow‐up. Three participants lacked data for the number of missing envelopes. Four participants did not provide urine for analysis, and 4 were missing physician reports of compliance (6‐week and 3‐month follow‐ups). As the result of intervening school holidays, teacher reports of compliance are available for only 16 children (8 in each condition) at each follow‐up.
Methylphenidate non‐responders were not excluded. Did not state the method used to account for missing outcome data

Selective reporting (reporting bias)

Unclear risk

All analyses mentioned are described, but no protocol was identified

Vested interest bias

High risk

Funding from CIBA‐GEIGY Canada

Firestone 1981

Methods

Three‐month, randomised, double‐blind, placebo‐controlled, parallel study, wherein participants were randomly assigned to 3 arms

  1. Parent training + placebo (control group)

  2. Parent training + methylphenidate (intervention group)

  3. Methylphenidate

Participants

Number of  participants screened: 91

Number of participants included: not stated (includes boys and girls)

Number of participants randomly assigned: not stated

Number of participants followed up: intervention 18, control 13

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III

Age: mean 7.32 years (range 5 to 9)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: not stated

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

IQ: 116

Sociodemographics: all children living at home with ≥ 1 parent. No significant differences in age and IQ between treatment groups. No data on remaining parameters

Inclusion criteria

  1. 5 to 9 years of age

  2. Fit DSM‐III criteria for attention deficit disorder with hyperactivity, showing overactivity, short attention span, impulsivity, aggressiveness and oppositional behaviour, both at home and in school, since before 4 years of age

  3. Hyperactivity Index of Conners' Behavior Rating Scale for teachers ≥ 15

  4. IQ > 85

Exclusion criteria

  1. Brain damage

  2. Epilepsy

  3. Psychosis

Interventions

Participants were randomly assigned to methylphenidate + parent training (intervention group) or to placebo + parent training (control group). First 3 to 4 weeks: Methylphenidate was titrated (after randomisation), starting with 5 mg b.i.d. (morning, noon), 7 days a week. After the titration period: Methylphenidate was given only on school days

Average methylphenidate dosage: 22 mg/d (range 10 mg/d to 30 mg/d)

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale 2, Conners' Parent Rating Scale subscale

  2. Hyperactivity Index: rated by mothers post treatment

Notes

Ethics approval: no information

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusions of study authors

  1. All groups showed improvement at home and in school; only with methylphenidate administration were gains in measures of attention and impulse control also seen

  2. Results also revealed greater improvement in academic achievement and in classroom behaviour in medication groups as compared with placebo groups

  3. No evidence showed significant benefit from the addition of parent training to administration of medication

Comment from review authors

  1. First study author has retired; we were not able to get data from him (e.g. protocol, randomisation method)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Children were randomly assigned; no description of how

Allocation concealment (selection bias)

Unclear risk

No data

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, teachers, therapists and those testing the children were unaware of medication conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parents, teachers, therapists and those testing the children were unaware of medication conditions

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No data

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): none

Selective reporting (reporting bias)

Unclear risk

No data

Vested interest bias

Low risk

Ministry of Health

Conflicts of interest: not stated

Fitzpatrick 1992a

Methods

Cross‐over trial with 4 interventions

  1. Sustained‐release methylphenidate

  2. Standard methylphenidate

  3. Combination

  4. Placebo

Phases: 4 pharmaceutical conditions, each lasting 2 weeks (including weekends), with different dosage schedules based on weight of child and type of intervention

Participants

Number of participants screened: not stated

Number of participants included: 19 (17 boys, 2 girls). Participants were randomly assigned to 1 of 24 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADD: DSM‐III during Diagnostic Instrument for Childhood and Adolescence (ADD with hyperactivity 16/19) (ADD without hyperactivity 3/19)

Age: mean 8.71 years (SD 1.33, range 6.9 to 11.5)

IQ: 114.11 (SD 13.34)

Methylphenidate naive: 18

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional disorder (n = 12), oppositional + conduct disorder (n = 1), enuresis (n = 2), encopresis (n = 2), phobia (n = 1), overanxious (n = 1), adjustment disorder (n = 1)

Comedication: not stated

Sociodemographics: middle class (Hollingshead 4‐factor mean 38.11, SD 13.18)

Inclusion criteria

  1. Not explicitly stated

Exclusion criteria

  1. Not explicitly stated

Interventions

Participants were randomly assigned to 1 of 24 (3 methylphenidate and 1 placebo) possible drug condition orders of sustained‐release methylphenidate (SR), standard methylphenidate (SA), methylphenidate combination and placebo

Mean methylphenidate dosage: SRSA‐MPH 17.1 mg (0.56 mg/kg), SASR‐MPH 20 mg (0.67 mg/kg), combination 11.8 mg SA MPH + 20 mg SR MPH (0.38 mg/kg SA, 0.67 mg/kg SR)

Administration schedule: sustained‐release methylphenidate, mornings (8:00 AM) daily, SA MPH morning (8:00 AM) and noon daily

Duration of each medication condition: 2 weeks

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: not stated

Treatment compliance: parents phoned weekly to encourage compliance. School nurse contacted at the beginning of each individual's participation to promote co‐operation and to check on compliance

Outcomes

ADHD symptoms

  1. Conners' Hyperactivity Index: parent‐ and teacher‐rated, weekly (although results were averaged over treatment phase)

  2. IOWA Inattention/Overactivity and Aggression/Non‐compliance Scales: parent‐ and teacher‐rated, weekly

  3. Hyperactivity, Attention and Aggression subscales of Loney’s Time on Task Scale (TOTS): every 2 weeks

General behaviour

  1. Selected items of Child Psychiatric Scale: observer, end of each laboratory session (i.e. after 14 days of treatment)

Non‐serious adverse events

  1. Interviewed: 12 side effect symptoms: drawn from Subject’s Treatment Emergent Symptom Scale, parents, end of each treatment phase

  2. Body weight: start of each laboratory session

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusions of study authors

  1. Methylphenidate conditions were superior to placebo and were comparable with one another

  2. Findings suggest comparable effectiveness for sustained and standard preparations of methylphenidate

  3. Improved behaviour and improved information processing under stimulant conditions

Comment from review authors

  1. This study was not actually randomised; therefore we cannot use data on ADHD symptoms and general behaviour ‐ only data on adverse events

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not clear

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Double‐blind trial consisting of 4 pharmacological conditions, each lasting 2 weeks and ordered according to a Latin square (i.e. not randomly assigned)

Allocation concealment (selection bias)

High risk

Not randomised

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. Blindness was maintained by administering placebo tablets consisting of the vehicle for the SA and SR preparations, respectively, as appropriate for each condition

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Data for 6 participants on Paired Associate Learning Test excluded, as they could not read well, but this was not 1 of our outcomes

Selection bias (e.g. titration after randomisation): no, but because of emergent side effects, reductions of 2.5 mg SA MPH per dose were performed blindly for 1 participant in the combined condition. Similarly, blinded (but sham) adjustments of SA placebo were made for 2 participants in the placebo phase and for 1 in the SR condition

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

Research was supported by National Institute of Mental Health (NIMH) grant MH38118

Conflicts of interest: not stated

Flapper 2008

Methods

Four‐week, double‐blind, randomised, placebo‐controlled, cross‐over study of methylphenidate and placebo with weekly switches of 4 dosage levels; capsules of

  1. 0.5 mg/kg

  2. 0.75 mg/kg

  3. 1 mg/kg

  4. Placebo

Methylphenidate‐sensitive children continued in an open‐label study for 4 weeks

Participants

Number of participants screened: 80

Number of participants included: 30 (22 boys, 8 girls). Participants were randomly assigned to 1 of the possible drug orders of low‐dose, moderate‐dose and high‐dose methylphenidate and placebo

Number of participants followed up: 30

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (37%), inattentive (57%), hyperactive‐impulsive (7%))

Age: mean 105.2 months (SD 25.1, range 7 to 12 years)

IQ: > 70

Methylphenidate naive: 100%

Ethnicity: not stated

Country: the Netherlands

Setting: out‐patient clinic

Comorbidity: developmental co‐ordination disorder (100%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD, DSM‐IV

  2. Developmental co‐ordination disorder, DSM‐IV

  3. ADHD symptoms had to be severe for ≥ 6 items on the DSM‐IV ADHD Rating Scale, Parent Version, investigator administered and scored

  4. Total score on the Movement Assessment Battery for Children below the fifth centile

Exclusion criteria

  1. Comorbid disorders (including pervasive developmental disorder)

  2. Not medication‐naive

  3. IQ score below normal range (< 70), as assessed by the Wechsler Intelligence Scale for Children – Revised (Dutch Edition)

Interventions

Participants were randomly assigned to possible drug orders of the 3 daily doses of methylphenidate (0.5 mg/kg, 0.75 mg/kg, 1 mg/kg) and placebo

Mean optimal dosage: 0.66 mg/kg/d (SD 0.22)

Administration schedule: b.i.d.

Duration of each medication condition: 1 week

Washout before study initiation: none. All were medication naive

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. 18‐Item ADHD Rating Scale, Fourth Edition: rated weekly (each Friday) by parents and teachers

Notes

Sample calculation: yes

Ethics approval: Procedures were performed in accordance with the ethical standards of the University Medical Centre, University of Groningen

Comment from study authors

  1. One aim of the study was to investigate the effectiveness of methylphenidate in improving the fine motor performance of children with ADHD and developmental co‐ordination disorder (DCD). To prevent confounding by fluctuations in ADHD symptoms, these had to be reduced by selecting methylphenidate‐sensitive children

Key conclusions of study authors

  1. Children with ADHD/DCD and their parents rated overall quality of life as poorer than for healthy controls, manifested in domains of motor and autonomic functioning, as well as cognitive and psychosocial functioning

  2. In our study, significant improvements in health‐related quality of life were noted after treatment with methylphenidate, as was improvement in symptoms of ADHD and motor functioning

  3. Fine motor performance in children with ADHD‐DCD was poorer before use of methylphenidate than afterwards

  4. Impairment in manual dexterity and poor quality of handwriting and drawing improved after methylphenidate use, but performance remained poorer than in the control group

Comment from review authors

  1. Only data from the 4‐week cross‐over trial are useful for the review; therefore all data extracted were derived from the cross‐over period

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: August 2013 to January 2014. We obtained supplemental information regarding participant demographics and efficacy data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random order by pharmacist

Allocation concealment (selection bias)

Low risk

Medication codes were broken at time 2 (endpoint)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, teachers, children and paediatrician were kept blinded to the child’s drug condition and dosage level

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parents, teachers, children and paediatrician were kept blinded to the child’s drug condition and dosage level

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant withdrew; no ITT was needed. Seven children whose ADHD symptoms were not sensitive enough to methylphenidate (effect < 25%) were excluded from the 4‐week open‐label period. However, as we are using in this review only data from the preceding cross‐over period, exclusion of these children does not cause high risk of bias

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No reporting bias

Vested interest bias

Low risk

For this study, no funding was available. This double‐blind placebo‐controlled (DBPC) trial of methylphenidate was performed as a clinical treatment programme as best clinical practice to determine the effects of methylphenidate and optimal dose compared with placebo

Conflicts of interest: no affiliations with pharmaceutical companies or similar stated

Forness 1992

Methods

Double‐blind, placebo‐controlled, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 4

Participants

Number of participants screened: 82

Number of participants included: 71 (all boys). Participants were randomly assigned to different possible drug condition orders of placebo and 3 doses of methylphenidate

Number of participants followed up: Conners' Teacher (n = 47), Conners' Parent (n = 69)

Number of withdrawals: Conners' Teacher 33.8%, Conners' Parent 12.6%

Diagnosis of ADHD: DSM‐III‐R (hyperactive‐impulsive (100%))

Age: mean 9.3 years (range 7 to 11)

IQ: mean 106.2 (range 67 to 137)

Methylphenidate naive: 100%

Ethnicity: ethnic minority (11.3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: conduct disorder (30/71)

Comedication: no; had no psychotropics for a month before

Sociodemographics: Alll participants were middle to upper class

Inclusion criteria

  1. Boys

  2. 7 to 11 years of age

  3. Recruited from referrals to 2 clinics: University of California, Irvine Child Development Center for Children with ADD, and University of California, Los Angeles Child Psychiatry Outpatient Department for children with psychiatric disorders

  4. Meet cutoff for inattention/overactivity

Exclusion criterion

  1. IQ below 85

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.3, 0.6 and 1.0 mg/kg MPH (unless dosages would exceed 20 mg, so the low dose would be 0.15 mg/kg) 3 times a day and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 3 times a day – 7:30 AM, 11:30 AM, 3:30 PM

Duration of each medication condition: 7 days

Washout before study initiation: not stated

Titration period: nil

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Rating Scale: teachers and parents

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. At the very least , teachers may be cautious about optimistic claims of positive response to methylphenidate

Email correspondence with study authors. Emailed study authors to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Reported as double‐blind but not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Reported as double‐blind but not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion): no, but high loss to follow‐up for teachers' scores on Conners' Teacher Rating Scale

Washout period between medication conditions: unclear

Selective reporting (reporting bias)

Unclear risk

Not clear

Vested interest bias

Low risk

Study supported by National Institute of Mental Health (NIMH) grant MH38686

Conflicts of interest: no affiliations described

Froehlich 2011

Methods

Randomised, double‐blind, placebo‐controlled, cross‐over trial of multiple methylphenidate doses in stimulant‐naive school‐aged children

Phases: 3 active dosage weeks and 1 week of placebo

Participants

Number of participants screened: 162

Number of participants included: 105. Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 89 (65 boys, 24 girls)

Number of withdrawals: 16

Diagnosis of ADHD: DSM‐IV (combined (48%), hyperactive‐impulsive (0%), inattentive (52%))

Age: mean 8.13 years (range 7 to 11)

IQ: mean: 105.34 ± 12.65

Methylphenidate naive: 100%

Ethnicity: Caucasian (79%), African American (18%), Hispanic (2%), other (1%)

Country: USA.

Setting: out‐patient clinic

Comorbidity: anxiety disorder (17%), mood disorder (2%), disruptive behaviour disorder (36%)

Comedication: not stated

Inclusion criteria

  1. ADHD according to DSM‐IV criteria for onset age, pervasiveness and impairment, inattentive or combined type

  2. Stimulant‐naive

  3. 7 to 11 years of age

  4. 6 non‐overlapping symptoms in a symptom domain (as per Diagnostic Interview Schedule for Children, and Vanderbilt ADHD Teacher Rating Scale), and both parent and teacher reported ≥ 4 symptoms in that domain

Exclusion criteria

  1. Hyperactive‐impulsive‐type participants

  2. IQ ≦ 80

  3. Mania/hypomania

  4. Comorbid oppositional defiant disorder, conduct disorder, depression and anxiety, if they were determined to be the primary cause of ADHD symptoms, or required different treatment

  5. Medical history suggesting significant brain injury

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of OROS methylphenidate (18 mg, 27 mg, 36 mg for children ≤ 25 kg; 18 mg, 36 mg or 54 mg for children > 25 kg; sample mean maximum dose 1.57 mg/kg/d) and placebo

Administration schedule: once daily

Duration of each medication condition: 1 week

Washout before study initiation: drug naive

Medication‐free period between interventions: not stated

Titration period: none initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Parents and teachers: Vanderbilt ADHD Rating Scales: completed at baseline and each week of the trial

Notes

Sample calculation: yes

Ethics approval: yes; Cincinnati Children’s Hospital Institutional Review Board

Comments from study authors

  1. Additional limitations include restricted duration of follow‐up and heterogeneity of our sample due to recruitment from a variety of sources. Further, our sample had more inattentive (52%) than combined subtype (48%) participants

  2. Although consistent with subtype distribution in many epidemiological samples, this differs from clinic settings, where combined type is most common

Key conclusions of study authors

  1. Results of this double‐blind, placebo‐controlled, ADHD pharmacogenetic trial of psychostimulant‐naive school‐aged children suggest that dopamine active transporter (DAT) and dopamine receptor (DR) D4 variations may be associated with unique methylphenidate dose‐response curves

  2. Children lacking the DAT 10‐repeat allele and those with the DRD4 4‐repeat allele had a more robust methylphenidate response compared with those of alternate genotypes, consistent with improved response for the ADHD susceptibility low‐risk alleles

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study author: October/November 2013. Contacted study author to obtain mean and SD for ADHD symptoms. Never received additional data. Therefore no further email correspondence regarding allocation concealment, randomisation, etc.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned to different drug orders. No description about how

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Study medication consisted of identical capsules filled with an inert white powder (placebo) or prescribed dose of Concerta overencapsulated to preserve double‐blind

Double‐blind, but not specified who was blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Study medication consisted of identical capsules filled with an inert white powder (placebo) or prescribed dose of Concerta overencapsulated to preserve double‐blind

Double‐blind, but not specified who was blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data provided on 89 participants who completed the study

Selective reporting (reporting bias)

Unclear risk

Not able to find protocol or trial identifier

Vested interest bias

Low risk

National Institute of Mental Health (NIMH) and Cincinnati Children’s Hospital Center for Education and Research Therapeutics Award

Conflicts of interest: Dr. Epstein receives funding from Eli Lilly and Co. Dr. Stein has received research support from Eli Lilly and Co., McNeil Pharmaceuticals, Novartis and Shire. He has served on a Speakers' Bureau for Novartis and has served as consultant to Novartis, Shire and Shinogi Pharmaceuticals

Gadow 1990

Methods

Six‐week double‐blind, cross‐over trial in which participants received the following in random order

  1. Low dose of methylphenidate

  2. Moderate dose of methylphenidate

  3. Placebo

Furthermore, a single case report from the study is described

Participants

Number of participants screened: not stated

Number of participants included: 11 (11 boys, 0 girls). Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: between 9 and 10, depending on ratings

Number of withdrawals: 1 to 2

Diagnosis of ADHD: DSM‐III (type not stated)

Age: mean not reported (range 5.9 to 11.9 years)

Case report: 10 years of age

IQ: > 75

Stimulant‐naive: 6

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: disruptive behaviour disorder

Comedication: not reported

Sociodemographics: Children represented a full range of socioeconomic backgrounds

Inclusion criteria   

  1. Boys

  2. 5 to 12 years of age

  3. ADHD diagnosis according to DSM‐III

  4. Scored above research cutoff (> 7) on the Aggression scale of IOWA Conners' Teacher Rating Scale OR were considered aggressive by their classroom teacher and were above cutoff on ≥ 1 other conduct problem scale

  5. Scored ≥ 15 on the Abbreviated Teacher Rating Scale

  6. Above cutoff on the Oppositional DIsorder or Conduct Disorder Index of the Parent or Teacher Version of the Stony Brook Child Psychiatric Checklist, Version 3

Exclusion criteria

  1. IQ < 70

  2. Psychosis, pervasive developmental disorder, dangerous to self or others

  3. Seizure disorder, major organic brain dysfunction, medical illness, contraindication to medication treatment

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of low‐dose (0.3 mg/kg) and moderate‐dose (0.6 mg/kg) (upper limit 25 mg) methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: b.i.d., morning, noon, 3.5 hours apart, 7 days a week

Duration of each medication condition: 2 weeks

Washout before study initiation: yes

Titration period: When moderate dose was not preceded by low‐dose condition, the child was gradually built up to the moderate dose. Data from these titration days were excluded from the analyses

Treatment compliance: pill count, no further information

Outcomes

ADHD symptoms

  1. Conners' Abbreviated Teacher Rating Scale

  2. Conners' Abbreviated Parent Rating Scale

Non‐serious adverse events

  1. Stimulant Side Effects Checklist: rated systematically

Notes

Sample calculation: no information

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Sample size was small, which decreased the probability of detecting clinically significant group differences between treatment conditions

Key conclusion of study authors

  1. Results of this study indicate that methylphenidate‐induced improvements in the classroom behaviour of aggressive hyperactive boys are associated with concomitant changes in the demeanor of classmates sitting in close proximity to the drug‐treated child

Email correspondence with study authors: July 2013. Emailed first study author twice to get additional information (funding, ethics approval, etc.) and data from the study. Study authors not able to provide us with additional data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Dose schedules were assigned on a random basis

Allocation concealment (selection bias)

Low risk

Medication and placebo pills were identical and were dispensed to parents and school nurses in dated, sealed envelopes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, teachers, observers, treating physicians and children were blinded to dose and order

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parents, teachers, observers, treating physicians and children were blinded to dose and order

Incomplete outcome data (attrition bias)
All outcomes

Low risk

For classroom analyses, data on 10 boys were analysed, and for lunch room analyses, data on 9 boys were analysed

Selection bias (e.g. titration before randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Email sent to first study author. No answer; therefore not able to get information

Vested interest bias

Unclear risk

Email sent to first study author in July 2013. No answer

Ciba Pharmaceutical Company supplied methylphenidate placebo

Conflicts of interest: not stated

Gadow 1995

Methods

Randomised, placebo‐controlled, double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate in 2 or 3 dosages

  2. Placebo

Phases

  1. Washout if medications before trial

  2. Eight‐week randomised, controlled trial (RCT) with 2 weeks on each arm

  3. Open‐label follow‐up at 24 months

Participants

Number of participants screened: not stated

Number of participants included: 34 (31 boys, 3 girls). Participants were randomly assigned to different orders of the 3 dosages

Number of participants followed up: RCT 34, MED (minimal effective dose, after RCT) 27; 12‐month follow‐up 30; 18‐month follow up 26; 24‐month follow‐up 26

Number of withdrawals: RCT: 0

Diagnosis of ADHD: DSM‐III‐R (subtypes not stated)

Age: mean 8 years and 10 months (range 6.1 years to 11.9 years)

IQ: mean 105.9 (SD 13.7, range no information)

Methylphenidate naive: 24 (71%)

Ethnicity: Caucasian (85%), African American (3%), Asian (3%), Hispanic (9%), other (0%)

Country: USA

Setting: out‐patient clinic

Comorbidity: Data from only 21 children from Gadow et al. (1995): tics (100%); anxiety or depressive disorder, or both (8/21; 38%); obsessive‐compulsive disorder (3/2; 14%); most of the children also had oppositional defiant disorder or conduct disorder and academic problems

Comedication: not during RCT. Four children were treated with an anti‐tic medication in combination with methylphenidate at some time during the course of follow‐up (neuroleptic 3, clonidine 1)

Sociodemographics: no information

Inclusion criteria

  1. Meet DSM III‐R diagnostic criteria for ADHD and chronic motor tic disorder or Tourette's disorder

  2. ADHD had to be a primary reason for seeking clinical services

  3. In general, had to be above the cutoff on 2 or 3 parent‐ and teacher‐rated hyperactivity and/or ADHD behaviour rating scales

  4. Written signed statement from parents consenting to their child’s participation

Exclusion criteria

  1. Dangerous to self or others

  2. Tics: the major clinical management concern

  3. Psychosis

  4. IQ < 70

  5. Seizure disorder

  6. Major organic brain dysfunction

  7. Major medical illness

  8. Contraindications to medication (other than tics)

  9. Pervasive developmental disorder

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of doses: 0.1 mg/kg (mean 4.4 mg), 0.3 mg/kg (mean 9.0 mg) and 0.5 mg/kg (mean 14.0 mg) of methylphenidate and placebo

Upper dosage limit was 20 mg. When the 0.5‐mg/kg dose was not preceded by a low‐dose condition, the child was gradually built up to the moderate dose. Build‐up days occasionally fell on scheduled school observation days. Observers were unaware of these days and observations were conducted as usual, but these data were excluded from the analyses

Administration schedule: b.i.d. (or t.i.d.) at morning and noon, approximately 3.5 hours apart, 7 days a week

Duration of each medication condition: 2 weeks

Washout before study initiation: methylphenidate 1 week, antipsychotic 3 weeks, clonidine 2 weeks

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: Parents and nurses were asked to return unused medication envelopes, which allowed researchers to assess compliance. No further information was provided in the paper

Regarding 24‐month follow‐up: Total daily dose of methylphenidate, MED (minimal effective dose ‐ after RCT) mean 16.5 mg (range 5 mg to 40 mg); second visit mean 28.5 mg (range 15 mg to 60 mg); third visit mean 29.2 mg (range 10 mg to 90 mg); and fourth visit mean 34.5 mg (range 15 mg to 92 mg)

Outcomes

During 8‐week RCT

ADHD symptoms

  1. Parents

    1. Conners' Abbreviated Parent Rating Scale: rated Saturday and Sunday each week

    2. Mothers’ Objective Method for Subgrouping: rated by parents Saturday and Sunday each week

  2. Teachers

    1. Conners' Abbreviated Teacher Rating Scale, 10‐item: rated 2 days per week for each intervention period

    2. Iowa Conners' Teacher Rating Scale: rated 2 days per week for each intervention period

General behaviour

  1. Peer Conflict Scale: parent‐ and teacher‐rated, 2 times a week

  2. Classroom Observation Code: observer‐rated, 4 days for each treatment condition

  3. ADHD School Observation Code: observer, 3 to 4 days for each treatment condition

  4. Code for Observing Social Activity: observer, lunch and playground, 20 to 30 minutes, 4 days for each treatment condition

Non‐serious adverse events

  1. Side Effects Checklist: 13 items, rated by parents on Saturday and Sunday and rated by teacher twice a week

  2. Global Tic Rating Scale: rated by parents on Saturday and Sunday and rated by teacher twice a week

  3. Motor and vocal tic category: Observers coded presence or absence of tics in the classroom, lunchroom or playground, 4 times for each medication condition

Physician evaluations

  1. Yale Global Tic Severity Scale: rated every second week

  2. Tourette Syndrome Unidentified Rating Scale: rated every second week

  3. Global Tic Rating Scale (assessed in only 22 participants): rated every second week

  4. Shapiro Tourette Syndrome Severity Scale (assessed in only 22 participants): rated every second week

  5. Motor tic frequency tics: rated in 180 five‐second intervals in a simulated classroom; tics were coded as present or not present in each interval, rated every second week

  6. Weight: assessed every second week

  7. Heart rate: assessed every second week

  8. Blood pressure: assessed every second week

During 24‐month follow‐up

Physician evaluations

  1. All rated at minimally effective dose (right after RCT) 6 months, 12 months, 18 months and 24 months

  2. Yale Global Tic Severity Scale

  3. Shapiro Tourette Syndrome Severity Scale

  4. Three subscales from Tourette Syndrome Unified Rating Scale

  5. Total number of tics

  6. Number of tics observed in 2 minutes of quiet conversation with physician

  7. LeWitt Disability Scale, which assesses tics and symptoms of comorbidity

  8. Global Tic Rating Scale

  9. Blood pressure

  10. Heart rate

  11. Pulse

  12. Weight

Parent ratings

  1. Based on last 2 weeks and rated at MED (right after RCT) 6 months, 12 months, 18 months and 24 months

  2. Stimulant Side Effects Checklist

  3. Global Tic Rating Scale

Notes

Sample calculation: yes

Ethics approval: no information

Comments from study authors

  1. Magnitude of clinical improvement associated with 0.3 mg/kg dosage vs 0.5 mg/kg dosage was generally trivial for many children

  2. 0.5 mg/kg dosage was associated with more side effects, but fortunately they were generally of limited clinical significance

  3. Generalisability of findings from this study is subject to several qualifications. First, our data pertain to observed treatment effects over an 8‐week period and therefore cannot address the issue of tic exacerbation as a function of long‐term drug exposure. Furthermore, the findings pertain only to children with ADHD with tics of mild to moderate severity that occur frequently enough to be observed during 15‐minute intervals

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no; children were not excluded from participation in the study if they had prior experience with stimulant drug therapy, or if such therapy purportedly had exacerbated their tics

Any withdrawals due to adverse events: no

Key conclusions of study authors

  1. During the course of this short‐term drug evaluation, physician, teacher and parent ratings were in agreement that methylphenidate did not lead to worsening of the severity of children's tic disorders

  2. Methylphenidate is an effective drug for the treatment of ADHD and oppositional and aggressive behaviour

  3. Follow‐up study showed that long‐term treatment with methylphenidate seems to be safe and effective for the management of ADHD behaviours in many (but not necessarily all) children with mild to moderate tic disorders. Nevertheless, careful clinical monitoring is mandatory, to rule out the possibility of drug‐induced tic exacerbation in individual patients

Comments from study authors (limitations)

  1. Two‐year follow‐up component was not blinded. Although obstacles to creating and maintaining a long‐term double‐blind study with a placebo group are daunting, failure to do so does introduce the possibility of bias

  2. Absence of a no‐treatment group does not allow inferences about natural changes in tic status over time

Comments from review authors

  1. Well‐designed study

  2. No exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate. Twenty‐six children received stimulant medication throughout the follow‐up interval; of these children, 1 was switched to dextroamphetamine. However, we have chosen to use in our analyses results for all 26

  3. In the article, "Gadow 2001, Anxiety and Depression…", n = 38. We have included it because it is part of the study, but we have not used data from this article

  4. Sverd 1992 and Gadow 1992; preliminary results from Gadow 1996 are described

  5. All included articles include a mix of different protocols, so total numbers of included participants differ from article to article

Email correspondence with study authors: April 2013. We emailed study authors for supplemental information regarding cross‐over data. Data were not available. Also no further data for the interventions were available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Dose schedules were counterbalanced and assigned on a random basis

Allocation concealment (selection bias)

Low risk

Medication and identically matching placebos were dispensed to parents and school nurses in dated, sealed envelopes at 2‐week intervals

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, teachers, participants, observers and physicians were blinded to the identity of those conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Medication was administered under double‐blind conditions (i.e. no one involved in clinical management of the participant, data collection or interaction with the school knew the identity of treatment conditions)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Study describes how many people were included in the different analyses

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified or received

Vested interest bias

Low risk

Research grants from the Tourette Syndrome Association and the National Institute of Mental Health (NIMH)

Conflicts of interest: not stated

Gadow 2007

Methods

Double‐blind, randomised, cross‐over trial with interventions

  1. Methylphenidate

  2. Placebo

Phases

  1. Washout if previously medicated

  2. Eight‐week trial, 2 weeks on each arm. Performed in 2 cohorts (several years apart, same personnel)

Participants

Number of participants screened: not stated

Number of participants included: 71 (39 + 32, cohorts 1 and 2, respectively). Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 71 (57 boys, 14 girls)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R or DSM‐IV (subtype not stated)

Age: mean: 8.9 ± 1.9 years (range 6 to 12)

IQ: mean 103.8

Methylphenidate naive: not stated

Ethnicity: Caucasian (87%), African American (6%), Asian (1%), Hispanic (6%)

Country: USA

Setting: out‐patient clinic

Comorbidity: Tourette’s syndrome (96%), chronic motor tic disorder (4%), oppositional defiant disorder (56%), conduct disorder (7%), overanxious or generalized anxiety (30%), simple phobia (7%), obsessive‐compulsive disorder (11%)

Comedication: yes

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐III‐R or DSM‐IV diagnosis of ADHD

  2. Chronic motor tic disorder or Tourette’s syndrome

  3. ADHD clinical criteria at both school and home

Exclusion criteria

  1. Too severely ill (dangerous to self or others)

  2. Psychotic

  3. IQ < 70

  4. Seizure disorder

  5. Major organic brain dysfunction

  6. Major medical illness

  7. Medical or other contraindication to medication (other than tics)

  8. Pervasive developmental disorder

  9. Tics so severe at intake that the parent or the child requested immediate intervention

  10. Extremely mild tics at intake

Interventions

Participants were randomly assigned to placebo or to 1 of 3 possible drug condition orders of immediate‐release methylphenidate (0.1 mg/kg (mean 4.5 mg; SD 1.6), 0.3 mg/kg (mean 9.3 mg; SD 3.0) and 0.5 mg/kg (mean 14.3 mg; SD 3.3)), twice a day for 2 weeks, each under double‐blind conditions. Upper limit was 20 mg/d

Administration schedule: twice daily, 3.5 hours apart. Most days, a morning dose and a noon dose, 7 days a week

Duration of each medication condition: 2 weeks

Washout before study initiation: Minimum washout periods for children receiving medication at referral were as follows: 1 week for stimulants (n = 10), 3 weeks for neuroleptic or selective serotonin reuptake inhibitor (n = 2) and 2 weeks for clonidine (n = 1)

Medication‐free period between interventions: none

Titration period: not stated initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Teachers

    1. Abbreviated Teacher Rating Scale (total score (Hyperkinesis Index) and ADHD (factor 1) and Emotional lability (factor 2) subscales)

    2. IOWA Conners' Teacher Rating Scale (Inattention‐Impulsivity‐Overactivity and Oppositional Defiant subscales): twice a week, weekdays

  2. Parents

    1. Abbreviated Parent Rating Scale (Total score (Hyperkinesis Index) and ADHD (factor 1) and Emotional lability (factor 2) subscales): Saturdays and Sundays, every week

General behaviour

  1. IOWA Conners' Teacher Rating Scale (Oppositional Defiant subscale)

Non‐serious adverse events

  1. Physician

    1. Assessment of clinical status and heart rate, blood pressure and weight: biweekly (n = 57)

    2. Tics: Yale Global Tic Severity Scale (includes 4 behaviourally anchored subscales: Total Motor Tic score, Total Phonic Tic score, Overall Impairment Rating and Global Severity score), completed for all but the first 12 participants. Additional measures: Shapiro Tourette Syndrome Severity Scale (tic frequency, severity and impairment), Global Tic Rating Scale and Two‐Minute Tic and Habit Count, completed for first 12 participants only. Both rated at 2‐week intervals

  2. Parents and teachers

    1. Global Tic Rating Scale: twice a week for each week of the study

    2. Stimulant Side Effects Checklist: rated twice a week

Notes

Sample calculation: no

Ethics approval: yes

Comment from study authors

  1. Children were not excluded if previous treatment with stimulants had purportedly induced or exacerbated their tics

Key conclusion of study authors

  1. In this study, immediate‐release methylphenidate was found to be a safe and effective short‐term treatment for ADHD in children with chronic tic disorder (CTD), but complete normalisation of all problem behaviours often is not achieved at acceptable doses

Comments from review authors

  1. Given our concern about the possibility of a type II error (i.e. erroneously concluding that methylphenidate did not have an adverse effect on tics), follow‐up repeat‐measure ANOVAs were performed on each respondent category, even when the main effect of dose was not significant

  2. Use of a cross‐over design may result in carry‐over effects. However, differences in change scores for tic frequency (simulated classroom) and severity (Yale Global Tic Severity Scale ‐ Global Severity score), respectively, between baseline and placebo in children who received placebo first and last were non‐significant and minuscule (i.e. effect size = 0.08 and 0.19, respectively)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April 2014. We obtained supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Dose schedules were counterbalanced and assigned on a random basis

Allocation concealment (selection bias)

Low risk

Medication was administered twice daily, approximately 3.5 hours apart, 7 days per week, and was dispensed in dated, sealed envelopes at 2‐week intervals

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind conditions

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None required withdrawal of medication

Selective reporting (reporting bias)

Unclear risk

No information to assess this

Vested interest bias

Low risk

This study was supported in part by a research grant from the Tourette Syndrome Association Incorporated, and by Public Health Service (PHS) grant number MH45358 from the National Institute of Mental Health (NIMH)

Conflicts of interest: Study authors have no financial relationships to disclose

Gadow 2011

Methods

Eight‐week, placebo‐controlled, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 54 (42 boys, 12 girls). Participants were randomly assigned to the different possible drug condition orders: ADHD without anxiety 37, ADHD with anxiety 17

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III or DSM‐IV (subtype not stated)

Age: ADHD without anxiety 8.9 years, ADHD with anxiety 9.1 years (range 7 to 12)

IQ: ADHD without anxiety mean 103.5, ADHD with anxiety mean 103.1

Methylphenidate naive: 48 (89%)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: Tourette's (n = 52), chronic multiple tic disorder (n = 2), anxiety (n = 17), major depressive episode or dysthymia (n = 11), overanxious disorder or generalised anxiety disorder (n = 12), separation disorder (n = 6), social phobia (n = 1), oppositional defiant disorder (n = 36), conduct disorder (n = 4)

Comedication: not stated

Sociodemographics: ADHD without anxiety 37.1, ADHD with anxiety 35.4 (according to Hollingshead)

Inclusion criteria

  1. DSM‐III‐R or DSM‐IV diagnostic criteria for ADHD

  2. Chronic multiple tic disorder or Tourette’s disorder according to research diagnostic criteria

  3. Each child met ADHD clinical criteria at both school and home (the “and rule”)

Exclusion criteria

  1. Tics were the major clinical management concern

  2. Too severely ill (dangerous to self or others)

  3. Psychotic

  4. Mentally retarded (IQ < 70)

  5. Seizure disorder

  6. Major organic brain dysfunction

  7. Major medical illness

  8. Medical or other contraindication to medication (other than tics)

  9. Pervasive developmental disorder

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.1 mg/kg, 0.3 mg/kg and 0.5 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: 4.7 mg, 9.5 mg, 14.5 mg

Administration schedule: "administered twice daily, approximately 3.5 hours apart, 7 days a week"

Duration of each medication condition: 2 weeks

Washout before study initiation: not stated

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Abbreviated Teacher/Parent Rating Scale

  2. IOWA Conners' Teacher Rating Scale

  3. Conners' (48‐item) Parent Rating Scale

  4. Mothers’ Objective Method for Subgrouping

  5. Parent and Teacher Versions of the Child Symptom Inventory

General behaviour

  1. IOWA Conners' Teacher Rating Scale (Oppositional Defiant subscale)

Non‐serious adverse events

  1. Stimulant Side Effects Checklist

  2. Yale Global Tic Severity Scale, which includes 4 behaviourally anchored subscales: Total Motor Tic score, Total Phonic Tic score, Overall Impairment Rating and Global Severity score

Notes

Sample calculation: not stated

Ethics approval: yes; approved by a university Institutional Review Board (IRB)

Comments from study authors

  1. No evidence suggested that immediate‐release methylphenidate exacerbated tics in children with anxiety

  2. Teacher ratings actually indicated improvement in motor and vocal tic frequency with medication (placebo > 0.5 mg/kg)

Key conclusions of study authors

  1. Findings suggest that the co‐occurrence of diagnosed chronic multiple tic disorders and ADHD with anxiety represents a particularly troublesome clinical phenotype, at least in the home setting

  2. Comorbid anxiety disorder was not associated with a less favourable response to immediate‐release methylphenidate in children with ADHD and chronic multiple tic disorder, but replication with larger samples is warranted before firm conclusions can be drawn about potential group differences

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: May 2015. Emailed study authors requesting additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Dose schedules were counterbalanced and were assigned on a random basis

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"...dispensed in dated, sealed envelopes at two‐week intervals"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

"In order to increase sample homogeneity, an additional three children with diagnosed specific phobia (n = 2) or major depressive episode without comorbid anxiety disorder (n = 1) were excluded from data analyses"

Vested interest bias

Low risk

Study was funded by the National Institute of Mental Health (NIMH) and the Tourette Syndrome Association Incorporated. CIBA Pharmaceutical Company supplied methylphenidate placebos. Novartis supplied immediate‐release methylphenidate

Conflicts of interest: "Kenneth D. Gadow is a shareholder in Checkmate Plus, publisher of the Child Symptom Inventory‐4"

Garfinkel 1983

Methods

Double‐blind, randomised, placebo‐controlled, cross‐over experiment with 4 arms

  1. Methylphenidate

  2. Clomipramine

  3. Desipramine

  4. Placebo

Study lasted 20 weeks; first and last 2 weeks were baseline periods during which no medication was given. Different interventions lasted 3 weeks each (Monday to Friday) with a 7‐day washout between changes in drugs

Participants

Number of  participants screened: not stated

Number of participants included: 12 (all boys). Participants were randomly assigned to the different possible drug condition orders

Number of participants followed up: 12

Number of withdrawals: 0

Diagnosis: DSM‐III of ADD. Eight participants were day hospital patients and 4 were in‐patients

Age: mean 7.3 years (range 5.9 to 11.6)

IQ: > 70

Methylphenidate naive: 50%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic and patient ward

Comorbidity: no

Comedication: no comedication during study period

Sociodemographics: not stated. Children presented with remarkably similar clinical, family and educational histories. None of the children had localising neurological signs or met criteria for other psychiatric diagnoses

Inclusion criterion

  1. ADD according to DSM‐III

Exclusion criteria

  1. None mentioned directly

Interventions

Participants were randomly assigned to the different possible drug condition orders of methylphenidate, clomipramine, desipramine and placebo

Mean methylphenidate dosage: 18 mg/d

Administration schedule: b.i.d., morning and lunch

Duration of each medication condition: 3 weeks

Washout before study initiation: 2 weeks before study entry and 7 days between drug conditions

Titration period: first week of the drug condition after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale: rated daily by care workers (in settings away from the classroom) and teachers (in classroom)

General behaviour

  1. Werry‐Weiss‐Peters Activity Rating Scale: rated daily by parents for day patients and by evening child care staff for fully hospitalised children

Non‐serious adverse events

  1. Diastolic and systolic blood pressure, measured by the nurse while participants were sitting and standing at 09.00 AM and 12.00 PM

  2. Apical pulse, morning and afternoon

Notes

Sample calculation: no information

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusions of study authors

  1. Methylphenidate was significantly better for improving classroom and behavioural manifestations of ADD as compared with placebo, desipramine and clomipramine

  2. Tricyclic antidepressants may have a significant therapeutic effect as indicated by mood elevation and amelioration of non‐classroom behaviour in the evenings

  3. Results encourage further clinical and pharmacological investigation of ADD using various alternative treatments

Comment from review authors

  1. Only information and data extracted from the methylphenidate group and the placebo group

Email correspondence with study authors. October 2013. We obtained supplemental information regarding number of drop‐outs and SD for Conners' Rating Scale. We also wanted other data but were not able to obtain these, as the study took place several years ago and the data are no longer available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Children were randomly given methylphenidate, desipramine, clomipramine, placebo

A Latin square was followed to control for the order of presentation of drugs

Allocation concealment (selection bias)

Low risk

Children were randomly given MPH, DMI, CMI, placebo. A Latin square was followed to control for the order of presentation of drugs

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, parents, attending physicians, teachers, nursing staff and child care workers did not know which medication the child received, ensuring the double‐blind procedure. Medication was added to lactose powder and was placed in identical appearing gelatin capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants, parents, attending physicians, teachers, nursing staff and child care workers did not know which medication the child received, ensuring the double‐blind procedure. Medication was added to lactose powder and was placed in identical appearing gelatin capsules

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Low risk

No reporting bias

Vested interest bias

Low risk

Study was funded by Ontario Mental Health Foundation

Conflicts of interest: none

Gonzalez‐Heydrich 2010

Methods

Cross‐over trial with 2 interventions

  1. OROS methylphenidate

  2. Placebo

Phases: Number of phases varied by dose group. For all dose groups, after completing first phase, participants were on a 1‐week washout period before cross‐over. Participants assigned to 1 of 3 maximum OROS methylphenidate dose groups in randomly assigned order

  1. Group 1: 1 week low dose, 1 week placebo

  2. Group 2: 1 week low dose, 1 week medium dose, 1 week placebo

  3. Group 3: 1 week low dose, 1 week medium dose, 1 week high dose, placebo

Participants assigned to next group after 3 participants have successfully completed preceding group with no side effects

Participants

Number of participants screened: 40

Number of participants included: 33 (19 boys, 14 girls). Participants were randomly assigned to different possible drug condition orders

Number of participants followed up: 33

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV‐R (combined (51.1%), inattentive (48.5%))

Age: mean 10.5 years (SD 3.0, range 6.4 to 17.5)

IQ: mean 89.7 (SD 16.9, range 59 to 123)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: hospital ward

Comorbidity: epilepsy (100%), others (not stated)

Comedication: yes

Sociodemographics: not stated

Inclusion criteria

  1. Speaks English

  2. IQ > 35 and score > 35 on Scales of Independent Behavior Revised (SIB‐R) Broad Independence Scale (both IQ and adaptive functioning at the moderate mental retardation level or higher)

  3. Diagnosis of epilepsy by International League Against Epilepsy (ILEA) criteria 26 (repeated, afebrile, unprovoked seizures with a seizure within the past 5 years)

  4. Diagnostic and Statistical Manual of Mental Disorders (DSM)‐IV‐R diagnosis of ADHD

  5. Score ≥ 4 on Conners' Global Index (CGI) severity scale for ADHD

  6. Score > 90% on ADHD Rating Scale, Parent Version; investigator scored for age and sex on inattentive, hyperactive‐impulsive or total score at first visit

  7. Has not taken stimulants or alpha‐adrenergic medications for longer than 2 weeks before study entry

  8. If taking antidepressants, neuroleptics or lithium, doses have been stable for longer than 4 weeks

  9. Currently on an antiepileptic drug regimen with stable doses for longer than 4 weeks before study entry

  10. Seizure‐free for longer than 1 month before study entry

  11. Prescribing clinician for epilepsy anticipates the need for a stable antiepileptic drug regimen for the duration of the study

  12. Guardian gives permission for study personnel to communicate with prescribing epilepsy clinician

  13. Teacher agrees to fill out ADHD Rating Scale at baseline and at the end of each arm of the study

Exclusion criteria

  1. Has had a seizure within the month preceding study entry

  2. Change in antiepileptic drug regimen or dose within 4 weeks of study entry

  3. History of moderate or severe adverse event related to methylphenidate

  4. History of any psychotic disorder

  5. Current acute major depression or bipolar mania

  6. Current psychiatric disorder requiring pharmacotherapy (other than ADHD)

  7. Unstable significant medical condition other than epilepsy

  8. Any known conditions that may make treatment with methylphenidate medically inadvisable

  9. Not currently working with a physician for epilepsy treatment

  10. Previously participated in a trial that provided adequate treatment with extended‐release methylphenidate

  11. Weighs < 9 kg

  12. Pregnant

  13. Unwilling to use an effective form of contraception

  14. Child has taken a stimulant (methylphenidate, an amphetamine preparation or pemoline), an alpha‐adrenergic (clonidine or guanfacine) or other ADHD medication within 2 weeks of the screening telephone interview (children will not be withdrawn from psychotropic medications to be enrolled in the study)     

Interventions

Participants were randomly assigned to different possible drug condition orders of methylphenidate and placebo. Each patient was given 5 mg of immediate‐release methylphenidate in the morning and at noon for 1 day. If this dose was tolerated, 18 mg of OROS methylphenidate was administered in the morning for the remaining 6 days of the first week. For group 1, maximum dose remained 18 mg, and methylphenidate and placebo arms lasted 1 week. For group 2, a further 1 week of OROS methylphenidate at a dose of 36 mg was given in the morning for 1 week; this group was also administered placebo for 2 weeks. For group 3, maximum dose was 54 mg in the morning, and each arm of the cross‐over lasted 3 weeks

Mean methylphenidate dosage: 11 participants < 1 mg/kg/d, 13 participants 1 to 1.5 mg/kg/d, 9 participants 1.5 to 2 mg/kg/d

Administration schedule: 1 dose in the morning

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: yes; 1 week

Titration period: no

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition, Observer, Parent, Teacher Version; Conners' Global Index (CGI)‐ADHD‐Severity: clinician‐rated at baseline and at end of each cross‐over week

Serious adverse events

  1. Seizure Classification Interview: observer, baseline and during trial

Non‐serious adverse events

  1. Barkley Side Effects Checklist ‐ Modified: observer, each study visit

Notes

Sample calculation: no

Ethics approval: not stated

Comment from study authors

  1. Considering exposure time, we observed increased daily risk of seizures with increasing dose of OROS methylphenidate, suggesting that potential safety concerns require further study

Key conclusions of study authors

  1. No serious adverse events and no carry‐over effects were noted in the cross‐over trial

  2. A larger study is needed to assess the effect of OROS methylphenidate on seizure risk

  3. Cross‐over design, including participants with frequent seizures, could maximise power and address high participant heterogeneity and recruitment difficulties

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: April 2014. We obtained supplemental efficacy data from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation lists for each maximum dose group were prepared by a statistician and maintained by the research pharmacist

Allocation concealment (selection bias)

Low risk

Randomisation lists for each maximum dose group were prepared by a statistician and maintained by the research pharmacist

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants were randomly assigned to take OROS methylphenidate or placebo. Principal investigator was blind to medication status. In cases of seizure worsening... Data and Safety Monitoring Board (DSMB) and Institutional Review Board (IRB) were informed of these seizures. Study personnel, the DSMB and the IRB were not unblinded through this process, as the participant would not be exposed again to the same condition

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Principal investigator was blinded to medication status

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Five participants discontinued treatment while taking placebo and 14 while taking OROS methylphenidate; however, all were included in all analyses

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

High risk

All outcomes reported referred to parent‐ and teacher‐rated versions of ADHD Rating Scale, Fourth Edition, and to clinician‐rated ADHD Severity measured weeks 1 to 4 in the protocol. Barkley Total scores (although significantly different) were not reported, although individual effects were reported

Vested interest bias

Unclear risk

Supported by National Institute of Mental Health (NIMH) Grant, Number K23 MH066835

Conflicts of interest: Four study authors are involved in the pharmaceutical sector

Gorman 2006

Methods

Participants with ADHD took part in a randomly ordered, double‐blind, cross‐over clinical drug trial comprising 21 consecutive days of

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 43

Number of participants followed up: 41 (21 boys, 20 girls)

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (combined (n = 22), hyperactive‐impulsive (n = 0), inattentive (n = 19))

Age: mean 9.08 years (range 6.26 to 12.55)

IQ: mean 107.66

Methylphenidate naive: 37 (out of 41)

Ethnicity: Caucasian (92.67%)

Country: USA

Setting: out‐patient clinic

Comorbidity: > 2 anxiety disorders (12.16%), lifetime affective disorder (2.46%), oppositional defiant disorder or conduct disorder (49.28%)

Comedication: 0%

Sociodemographics: mean 50.43 (range 22 to 66) (Hollingsworth SES)

Inclusion criteria

  1. 6 to 12 years of age

  2. Diagnosis of ADHD according to DSM‐IV, and based on a combination of the Parent Interview for Child Symptoms‐4 and a semi structured interview

  3. IQ > 80

  4. Normal or corrected vision and hearing

  5. No current use of medicine

Exclusion criteria

  1. Physical disabilities; history of neurological disorder, chronic medical illness, bipolar disorder, schizophrenia or pervasive developmental disorder; and an episode of major depressive disorder within at least 6 months

Interventions

Participants were randomly assigned to 1 dose of methylphenidate and placebo

Mean methylphenidate dosage: final daily dose 33.12 mg ± 1.36 (SE), range 25 to 50 mg/d (0.94 ± 0.02 mg/kg)

Administration schedule: b.i.d.

Duration of each medication condition: 21 days

Washout before study initiation: not stated

Medication‐free period between interventions: from lunch to the following morning

Titration period: 0.25 mg/kg b.i.d. (breakfast and lunch) on days 1 to 2; 0.25 mg/kg b.i.d. plus 0.125 mg/kg at 4:00 PM on days 3 to 7; 0.3 mg/kg b.i.d. and 0.15 mg/kg at 4:00 PM on days 8 to 14; and 0.4 mg/kg b.i.d. and 0.2 mg/kg at 4:00 PM on days 15 to 21. All dosages were administered to the nearest 2.5 mg

Titration: took place after randomisation

Treatment compliance: not stated. Four participants with ADHD had undergone previous trials of stimulant therapy ranging from 2 weeks to 7 months

Outcomes

ADHD symptoms

  1. IOWA Conners' Scale, teacher‐ and parent‐rated: at the end of each treatment period (around day 21)

Serious adverse events

  1. Five participants with ‘serious side effects’ were referred to, but no further information was given

Non‐serious adverse events

  1. Barkley and Murphy Side Effects Rating Scale, rated by an investigator, at the end of each treatment

  2. Weight, in street clothes and without shoes on a professional scale, at the end of each treatment period

Notes

Sample calculation: not described.

Ethics approval: yes

Comment from study authors

  1. "The unusually high proportion of girls reflects increasing sensitivity by clinicians to the identification of girls with ADHD as well as the awareness of our referring sources that we were studying sex differences in ADHD"

Key conclusions of study authors

  1. ADHD subtypes benefited comparably from methylphenidate treatment with respect to inattention

  2. Children with ADHD‐combined subtype underwent greater reductions in hyperactivity/impulsivity, but children with ADHD‐inattentive subtype also benefited in this respect

  3. Only children with ADHD/C displayed a reduction in externalising problems under methylphenidate. ADHD subtypes reacted comparably with stimulant treatment with respect to arithmetic performance, valence of teacher and parent comments and task‐incompatible behaviours ‐ all measures that do not distinguish between inattention, hyperactivity/impulsivity or oppositionality

  4. Somatic effects of treatment were comparable for subtypes

Comment from review authors

  1. Chang et al. 2001 (in Gorman 2006) is a PhD thesis; the other 2 articles are based on data from this. We have not had access to the full thesis

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: January 2014. We obtained supplemental information from study authors. Study authors do not have the files anymore; therefore we are not able to obtain all of the data requested

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Answer from study author: "Table of random numbers were used to make the allocation process, such that numbers ending in an even digit corresponded to one order and those ending in an odd digit to another" (Krogh 2014c [pers comm]

Allocation concealment (selection bias)

Low risk

Answer from study author: "Table of random numbers were used to make the allocation process, such that numbers ending in an even digit corresponded to one order and those ending in an odd digit to another" (Krogh 2014c [pers comm]

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Placebo capsules, identical in appearance and taste to those containing methylphenidate, and administered on the same schedule

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Second author blinded. Placebo capsules, identical in appearance and taste to those containing methylphenidate, and administered on the same schedule

Incomplete outcome data (attrition bias)
All outcomes

Low risk

When parents reported serious side effects (n = 5), their children’s dosages were reduced, or planned increments were omitted. However, study authors noted "eliminating participants who could not tolerate their assigned dosage might potentially skew the sample"

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No

Answer from study author: "Event related potentials and performance measures on a cognitive task were not reported (I didn't attempt to publish the ERP data because the analyses I conducted did not yield significant results) ‐ And since we do not look at this outcome, we see it as low" (Krogh 2014c [pers comm])

Vested interest bias

Low risk

National Institute of Mental Health (NIMH)

Conflicts of interest: Study authors have no financial relationships to declare

Green 2011

Methods

One‐day, randomised, parallel trial with 2 arms

  1. Immediate‐release methylphenidate

  2. Placebo

Six‐month follow‐up of study participants continuing methylphenidate treatment

Participants

Number of participants screened: not stated

Number of participants included: 34 (20 boys, 14 girls)

Number of participants randomly assigned: methylphenidate 22, placebo 12

Number of participants followed up: methylphenidate 22, placebo 12

Number of patients continuing methylphenidate treatment beyond the 1‐day trial: 16

Number of participants followed up: 15

Number of withdrawals from extension trial: 1

Regarding the group of patients participating in the 1‐day trial

Diagnosis of ADHD: DSM‐IV‐TR (combined (33.3%), hyperactive‐impulsive (not stated), inattentive (50%), not otherwise specified (17.7%))

Age: mean 11.1 years (range 5 to 20)

IQ: mean: 81.4

Methylphenidate naive: 61.8%

Ethnicity: not stated

Country: Israel

Setting: hospital/out‐patient clinic

Comorbidity in total sample: velocardiofacial syndrome (100%), oppositional defiant disorder (23.5%), specific phobia (26.5%), generalised anxiety disorder (11.8%), social phobia(11.8%), dysthymic disorder (8.8%) and separation anxiety (5.9%)

Comorbidity in methylphenidate group: congenital anomalies of the heart and great vessels (54.5%)

Comedication: no

Sociodemographics: not stated. The 2 groups had similar baseline demographics

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

RCT: Participants were randomly assigned to methylphenidate or placebo

Mean methylphenidate dosage: 15.7 ± 5.6 mg (0.5 mg/kg)

Administration schedule: once

Duration of intervention: 1 day

Titration period: no mention, none

Washout before study initiation: 3 days

Treatment compliance: not stated

Follow‐up: Some participants continued methylphenidate treatment beyond the RCT

Mean MPH dosage: not stated

Administration schedule: not stated

Duration of treatment: 6 months

Treatment compliance: 1 withdrew because of poor compliance

Outcomes

Non‐serious adverse events

  1. Cardiologic evaluation (ECG, heart rate and blood pressure): immediately before taking the pill (methylphenidate or placebo), and again after 90 minutes

  2. Barkley Side Effects Rating Scale (modified Hebrew Version), parent‐rated: 24 hours after methylphenidate administration and at 6‐month follow‐up

Notes

Sample calculation: not stated

Ethics approval: Study protocol was approved by the Institutional Review Board of the Rabin Medical Center

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Comments from study authors

  1. We found that all participants (100%) with velocardiofacial syndrome treated with methylphenidate exhibited ≥ 1 side effect

  2. Rate of all side effects immediately observed following initiation of treatment remained similarly high after 6 months of treatment

  3. According to our findings, it seems that children with velocardiofacial syndrome did not develop tolerance to methylphenidate side effects

  4. None of the children withdrew because of side effects

  5. Limitations: short duration of parallel‐group trial and relatively small sample size

Key conclusions of study authors

  1. Use of methylphenidate in children with velocardiofacial syndrome appears to be effective and relatively safe

  2. Comprehensive cardiovascular evaluation for children with velocardiofacial syndrome before and during stimulant treatment is recommended

Email correspondence with study authors: November 2013. We obtained supplemental information regarding ADHD diagnostic criteria from study authors. Furthermore, we received safety data from the study sample, excluding participants older than 18 years or with IQ < 70 (or both), but we decided not to use these data in our analyses, as data were missing for 60% of the control group

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Participants were randomly assigned to drug or placebo in a 2:1 ratio, according to their order of recruitment

Allocation concealment (selection bias)

High risk

Participants were randomly assigned to drug or placebo in a 2:1 ratio, according to their order of recruitment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and their parents were blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

Study authors have confirmed that planned outcomes for the 1‐day (randomised controlled) trial were measured and reported

Vested interest bias

Low risk

This work was funded by the Basil O’Connor Starter Scholar Research Award of the March of Dimes, NARSAD (National Alliance for Research in Schizophrenia and Affective Disorders) Young Investigator Award, the Marguerite Stolz Award from the Sackler Faculty of Medicine and the National Institute on Drug Abuse (NIDA)

Conflicts of interest: Study authors have had no institutional or corporate/commercial relationships for the past 36 months that might pose a conflict of interest&&

Greenhill 2002

Methods

Three‐week, randomised, double‐blind, 32‐site, parallel trial with 2 arms

  1. Modified‐release methylphenidate

  2. Placebo

Participants

Number of participants screened: 507

Number of participants included: 321

Number of participants randomly assigned: methylphenidate 158, placebo 163

Number of participants followed up: methylphenidate 141, placebo 135

Number of withdrawals: methylphenidate 17, placebo 28

Diagnosis of ADHD: DSM‐IV (combined subtype or predominantly hyperactive‐impulsive subtype)

Sex: 257 boys, 57 girls

Age: mean 9 years (range 6 to 15)

IQ: > 80

ADHD treatment naive: 36%

Ethnicity: Caucasian (71%), African American (15%), Hispanic (10%), other (4%)

Country: USA

Setting: out‐patient clinic

Comorbidity: none

Comedication: concomitant use of clonidine, anticonvulsant drugs and medications known to affect blood pressure and heart rate was not allowed

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. 6 to 16 years of age

  2. Primary diagnosis of ADHD, combined subtype or predominantly hyperactive‐impulsive subtype, as defined in DSM‐IV

  3. Did not respond to placebo with a reduction of ADHD symptoms during washout period

  4. First‐grade or higher school setting in which a single teacher could assess behaviour

  5. Blood pressure, heart rate and oral temperature had to be within normal range

  6. Did not respond to placebo with a reduction of ADHD symptoms during washout period

  7. First‐grade or higher school setting in which a single teacher could assess behaviour

  8. Blood pressure, heart rate and oral temperature had to be within normal range

Exclusion criteria

  1. Comorbid psychiatric diagnosis

  2. History of seizure or tic disorder

  3. Family history of Tourette’s syndrome

  4. IQ < 80

  5. Inability to follow or understand study instructions

  6. Female who had undergone menarche

  7. Use of amphetamines, pemoline or an investigational drug within 30 days of study entry

  8. Concomitant use of clonidine, anticonvulsant drugs or medications known to affect blood pressure, heart rate or CNS function

  9. Hyperthyroidism or glaucoma

  10. Concurrent chronic or acute illness (e.g. allergic rhinitis, severe cold) or disability that could confound study results

  11. Failed a previous trial of stimulants for ADHD

  12. Requiring a third daily dose in the afternoon or evening

  13. Documented allergy or intolerance to methylphenidate

  14. Living with anyone who currently had substance abuse disorder (excluding dependency)

Interventions

Participants were randomly assigned to modified‐release methylphenidate or placebo

Mean methylphenidate dosage: 40.7 mg/d (1.28 mg/kg/d)

Administration schedule: once daily

Duration of intervention: 3 weeks

Titration period: none

Treatment compliance: Medication counts showed satisfactory adherence in both groups

Outcomes

General behaviour

  1. Conners’ Teacher Global Index: rated twice daily (morning and afternoon), 3 times a week

  2. Conners’ Parent Global Index, 1 day of each weekend during the morning, afternoon and evening

Non‐serious adverse events

  1. Pittsburgh (11‐item) Side Effects Questionnaire: parent‐ and teacher‐rated weekly

  2. Teachers completed a similar side effect questionnaire

Notes

Sample calculation: no

Ethics approval: yes; probably approved by an institutional review board

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; participants with a documented allergy or intolerance to methylphenidate were excluded

Comments from study authors (limitations)

  1. Because no dose‐response curves were collected on children in the study, we could not determine whether effects of modified‐release methylphenidate were dose‐related

  2. Study inclusion and exclusion criteria were selected for milder cases of ADHD

  3. Lower scores in this trial mean that findings may not be generalisable

  4. Three‐week duration of the trial did not allow investigators to determine whether dual‐phase effects of modified‐release methylphenidate persist with long‐term treatment

  5. Study design limited generalisability of the results because it excluded acute placebo responders and those who had failed to respond to any methylphenidate treatment before the start of the study

  6. Parents were aware that researchers had the option of stepping up the "dose" of placebo each week

Key conclusion of study authors

  1. Modified‐release methylphenidate administered once daily in the morning was well tolerated and was significantly more effective in a double‐blind comparison with placebo in controlling ADHD symptoms throughout the school day

Email correspondence with study authors: November and December 2013: not possible to get supplemental information regarding the study through personal email correspondence with study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stated "randomised". Stratification based on previous treatment before randomisation ensured equal distribution across the 2 treatment groups

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical appearing modified‐release methylphenidate and placebo capsules were packaged in blister cards

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Stated "double‐blinded"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

316 were included in the safety population, and 314 in the ITT efficacy population. All statistical summaries and analyses were conducted for the ITT population using the LOCF approach for children who withdrew prematurely

Selection bias (e.g. titration after randomisation → exclusion): yes; exclusion of placebo responders

Selective reporting (reporting bias)

Unclear risk

No protocol available. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

This study was funded by Celltech Pharmaceuticals Incorporated

Conflicts of interest: Dr. Greenhill is a consultant for Celltech‐Medeva and a member of its medical advisory board. Drs. Findling and Swanson are consultants for Celltech‐Medeva

Greenhill 2006

Methods

Seven‐week multi‐centre, randomised, double‐blind, placebo‐controlled, parallel trial with 2 arms

  1. Extended‐release dexmethylphenidate

  2. Placebo

To compare the efficacy and safety of extended‐release dexmethylphenidate vs placebo in paediatric patients with ADHD

Pre‐randomisation phase of up to 2 weeks followed by double‐blind treatment phase for 7 weeks (5 weeks of dose titration followed by 1 week of optimal constant dose)

Participants

Number of participants screened: not stated

Number of participants included: 103

Number of participants randomly assigned: methylphenidate 53, placebo 50

Number of participants followed up: methylphenidate 48, placebo 37 (ITT analyses: methylphenidate 52, placebo 45)

Number of withdrawals: methylphenidate 5, placebo 13
Diagnosis of ADHD: DSM‐IV, extended‐release dexmethylphenidate (combined (83%), hyperactive‐impulsive (1.9%), inattentive (15.1%))

Age: mean methylphenidate 9.6 years, placebo 10.4 years (range 6 to 17)

IQ: > 70 (age‐appropriate functioning levels academically)

Sex: 66 boys, 37 girls

Methylphenidate naive: 40

Ethnicity: Caucasian (60%), African American (23.3%), other (16.5%)

Country: USA

Setting: classroom setting

Comorbidity: no psychiatric comorbidity

Comedication: not stated

Sociodemographics: not stated. No significant difference in baseline demographics was noted between the 2 groups. See Tables 1 and 2

Inclusion criteria

  1. ADHD diagnosis as per DSM‐IV

  2. Baseline age of 6 to 17 years

  3. Conners' ADHD/DSM‐IV Scales ‐ Teacher, for boys 6 to 8 years ≥ 27, 9 to 11 years ≥ 24, 12 to 14 years ≥ 19, 15 to 17 years ≥ 14

  4. Conners' ADHD/DSM‐IV Scales ‐ Teacher, for girls 6 to 8 years ≥ 16, 9 to 11 years ≥ 13, 12 to 14 years ≥ 12; 15 to 17 years ≥ 6

  5. Age‐appropriate functioning levels academically

  6. Negative pregnancy test and adequate contraception

Exclusion criteria

  1. Clinically significant abnormalities in vital signs, physical examination findings or laboratory test results

  2. History of seizures or use of anticonvulsant medication

  3. Comorbid psychiatric conditions (obtained by clinical interview)

  4. Any medical condition that could interfere with study participation or assessments, or that may pose danger with administration of methylphenidate

  5. Psychotropic medications

  6. Initiation of psychotherapy within the past 3 months

  7. Positive urine drug screen

  8. History of poor response or intolerance to methylphenidate

  9. Pregnant or nursing

  10. Any other investigational drug within 30 days of study entry

Interventions

Participants were randomly assigned to an extended‐release formulation of dexmethylphenidate or placebo. Permitted doses were 5 mg/d for the first week; 5 or 10 mg/d for the second week; 5 mg/d, 10 mg/d or 15 mg/d for the third week; 5 mg/d, 10 mg/d, 15 mg/d or 20 mg/d for the fourth week; and 5 mg/d, 10 mg/d, 15 mg/d, 20 mg/d or 30 mg/d for the fifth through seventh weeks

Mean final methylphenidate dosage: 24.0 ± 7.1 mg/d

Administration schedule: once daily in the mornings

Duration of intervention: 5‐week titration period plus 2 weeks at a constant dose

Titration period: initiated after randomisation

Treatment compliance: At each study visit, compliance was assessed by investigator and study staff on the basis of pill count and participant report

Outcomes

ADHD symptoms

  1. Conners' ADHD/DSM‐IV Scales ‐ Teacher: rated weekly by teachers

  2. Conners' ADHD/DSM‐IV Scales ‐ Parent: rated weekly by parents

Quality of life

  1. Child Health Questionnaire, Parent Form 50: parent‐rated at final visit

Serious adverse events

  1. Spontaneous reporting recorded weekly: no deaths or serious adverse events

Non‐serious adverse events

  1. Vital signs were rated weekly

  2. Spontaneously reported adverse events were recorded weekly

Notes

Sample calculation: Assumptions for sample size and power included treatment difference of 9.0 and and SD of 13.5

Ethics approval: no information provided

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; participants with history of poor response or intolerance to methylphenidate were excluded

Withdrawals due to adverse events: methylphenidate 0, placebo 1

Comments from study authors

  1. It is of interest that no participant who received extended‐release dexmethylphenidate discontinued the study because of an adverse event. This may result in part from the fact that more than one‐third of participants in each treatment group had prior experience with ADHD medications (mainly methylphenidate and dexmethylphenidate) and in part from the flexible‐dose design of the study

  2. Another possible limitation is that patients with previous methylphenidate or dexmethylphenidate experience were enrolled only if they had not experienced moderate to severe adverse reactions. By excluding patients who could not tolerate methylphenidate or dexmethylphenidate, investigators may have inflated the apparent safety and tolerability of extended‐release dexmethylphenidate. However, only about one‐fourth of the participants in each treatment group had prior experience with methylphenidate or dexmethylphenidate, so this factor probably had little impact on adverse event rates during the study

Key conclusions of study authors

  1. In conclusion, results of this trial indicate that extended‐release dexmethylphenidate administered once daily in doses of 5 mg to 30 mg is safe and effective for treatment of paediatric patients with ADHD symptoms, as reflected by its significant superiority over placebo on Conners' ADHD/DSM‐IV Scales ‐ Teacher, Conners' ADHD/DSM‐IV Scales ‐ Parent, Clinical Global Impressions ‐ Improvement Scale and Clinical Global Impressions ‐ Severity Scale, and in the Child Health Questionnaire Psychosocial Component score

  2. Extended‐release dexmethylphenidate was well tolerated and had a safety profile consistent with those of other methylphenidate and dexmethylphenidate formulations, resulting in appetite reduction and reduced weight in some patients

Comments from review authors

  1. Statistically significant treatment by centre interaction in primary efficacy analysis

  2. Study authors defined baseline scores on Conners' ADHD/DSM‐IV Scales ‐ Teacher (to be included) as different scores for different age groups, but they informed efficacy as the difference from baseline to endpoint in all medicated individuals

  3. As precise information on dose (mg/kg/d) was lacking, it is difficult to evaluate whether 24 mg/d was enough for participants in this age range (6 to 17 years)

Email correspondence: July 2014. Wrote to Novartis to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further description provided

Allocation concealment (selection bias)

Unclear risk

No description provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further description provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind, no further description provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Study used an ITT analysis. LOCF analysis was used to impute missing values for all final visit analyses

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

High risk

Study authors defined baseline scores on Conners' ADHD/DSM‐IV Scales ‐ teacher‐rated, as inclusion criteria (different scores for age groups), but they informed efficacy as the difference from baseline to endpoint for all medicated individuals

Vested interest bias

High risk

Study funded by Novartis

Conflicts of interest: Two study authors are employed by Novartis. Only Roberta R. Ball has no conflicts of interest

Grizenko 2012

Methods

Double‐blind, cross‐over, placebo‐controlled, 2‐week trial of methylphenidate in children diagnosed with ADHD

  1. Methylphenidate (0.5 mg/kg, b.i.d.)

  2. Placebo

Phases: 2

Participants

Number of participants screened: not stated

Number of participants included: 430 (average IQ group). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 430 (334 boys, 96 girls)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (combined 96 (48.73%), hyperactive‐impulsive not stated for average IQ group alone, inattentive not stated for average IQ group alone)

Age: mean 9.45 years (range 6 to 12)

IQ: mean 96.68 ± 9.98 (range 80 to 120)

Methylphenidate naive: not stated for average IQ group alone

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: income group 4.62 (SD 1.56). Income groups set at 1 (< Can$ 6000), 2 (Can$ 6000 to 10,000), 3 (Can$ 10,000 to 20,000), 4 (Can$ 20,000 to 30,000), 5 (Can$ 30,000 to 40,000) and 6 (> Can$ 40,000)

Inclusion criteria

  1. Children 6 to 12 years of age

  2. Diagnosed with ADHD by a psychiatrist or a paediatrician participating in a double‐blind, placebo‐controlled trial. Children were recruited from the Disruptive Behavior Disorders Program and from the general sector out‐patient clinic at Douglas Hospital in Montreal, a psychiatric university teaching hospital

Exclusion criteria

  1. Children with IQ < 70 (as measured by the Wechsler Intelligence Scale for Children)

  2. Met DSM‐IV criteria for psychosis, pervasive developmental disorder or Tourette's syndrome

  3. Previous intolerance or allergic reaction to methylphenidate (Grizenko 2006 and Grizenko 2012 ‐ both in Grizenko 2012)

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 0.5 mg/kg/d

Administration schedule: b.i.d. morning and noon

Duration of each medication condition: methylphenidate 1 week, placebo 1 week (including weekends)

Washout before study initiation: 1 week before the start of the trial

Medication‐free period between interventions: not stated, appears to have been none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Global Index ‐ Teacher Version: once a week + 1 week before medication trial

  2. Conners' Global Index ‐ Parent Version: once a week (1 week before medication trial and on the Sunday after children were given their medication of the weekend (Grizenko 2006 ‐ in Grizenko 2012)

General behaviour

  1. Parents completed a Child Behavior Checklist 1 week before the start of the medication trial

Serious adverse events

  1. No important adverse events or side effects were noted

Non‐serious adverse events

  1. Barkley Side Effects Rating Scale

Notes

Sample calculation: not stated

Ethics approval: yes

Key conclusion of study authors

  1. Grizenko 2012: All children with ADHD within normal and high levels of intellectual functioning respond equally to psychostimulant treatment; proper medication management is necessary for all children with the disorder

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes (see summary of participants)

Any withdrawals due to adverse events: not stated

Email correspondence with study authors: October 2013. We received some data from study authors. We sent another email to ask for additional information but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was completed by a research psychologist who had no contact with participants

Allocation concealment (selection bias)

Low risk

"The sealed envelope containing the information about the order of MPH and placebo administration for the participant was opened after the CCR rating was assigned and recorded"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Both drug and placebo were prepared by a pharmacist in identical coloured gelatine capsules"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

See above

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

Estimated enrolment: 700

Study start date: November 1999

Estimated primary completion date: March 2018 (final data collection date for primary outcome measure)

Vested interest bias

Unclear risk

Study was supported by the Canadian Institutes of Health

Conflicts of interest: Dr. Joober has received consultation honoraria from Pfizer Canada and Janssen Ortho. Dr. Zappitelli receives research salary support and grant funding from the Kidney Research Scientist Core Education and National Training Programme, the Fondation de Recherche en Sante du Quebec and the Research Institute of the McGill University Health Centre, for research not related to this manuscript

Gruber 2007

Methods

Fourteen‐day, double‐blind, placebo‐controlled, cross‐over study with 2 interventions

  1. Placebo

  2. Methylphenidate

Participants

Number of participants screened: not stated

Number of participants included: 37 (31 boys, 6 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 37

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (70%), hyperactive‐impulsive (11%), inattentive (19%))

Age: mean 9.2 years (range 6 to 12)

IQ: mean 96.7

Methylphenidate naive: not stated

Ethnicity: Caucasian (94%), other (6%)

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (30%), conduct disorder (46%), major depressive disorder (5%), general anxiety disorder (3%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. ADHD according to DSM‐IV

  2. Patient at the Disruptive Behavior Disorders Program and in the out‐patient department of Douglas Mental Health University Institute in Montreal

  3. Between 6 and 12 years of age

Exclusion criteria

  1. Score < 80 on the Wechsler Intelligence Scale for Children, Third Edition

  2. Diagnosis of psychosis

  3. Diagnosis of Tourette's syndrome

  4. Pervasive developmental disorder

  5. Taking any medication other than methylphenidate

  6. Previous intolerance/allergic reaction to any psychostimulant

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 0.5 mg/kg/d

Administration schedule: twice daily, morning and noon

Duration of each medication condition: 7 days

Washout before study initiation: no

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: not stated

Outcomes

General behaviour

  • Child Behavior Checklist, daily

Non‐serious adverse events

  • Sleep assessment using miniature antigraphs, measured daily

Notes

Ethics approval: Study was approved by the Research Ethics Board of Douglas Mental Health University Institute

Key conclusion of study authors

  1. Findings of the present study support the hypothesis that sleep moderates performance on the Continuous Performance Test in children with ADHD receiving placebo or methylphenidate

Comment from review authors

  1. Unfortunately, the data are not useable because of how the study is set up

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes (see exclusion criteria)

Any withdrawals due to adverse events: no

Email correspondence with study authors: February and March 2014. Emailed study author twice but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Order of administration (methylphenidate or placebo) was determined by random assignment

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"MPH and placebo were prepared in identical coloured gelatin capsules by the hospital’s clinical pharmacist, who was not involved in the study in any other way. Capsules were sealed in individual, daily‐dose envelopes to help control accurate administration"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): yes

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Low risk

This was not an industry‐supported study

Conflicts of interest: Study authors have indicated no financial conflicts of interest

Hale 2011

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: baseline, placebo, low‐dose methylphenidate and high‐dose methylphenidate for 4 weeks per medication phase

Participants

Number of participants screened: 65

Number of participants included: 56 (39 boys, 17 girls). Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV‐TR (combined (58.9%), hyperactive‐impulsive (7.1%), inattentive (33.9%))

Age: mean 120.84 months (SD 30.85 months, range 6 to 16 years)

IQ: mean 99.56 (SD 6.84, n = 41)

Methylphenidate naive: number not stated ("All participants were either medication naive or received and appropriate wash‐out period")

Ethnicity: European American (82%), African American (18%)

Country: USA

Setting: out‐patient clinic

Comorbidity: specific learning disability (n = 13), oppositional defiant disorder/conduct disorder (n = 11), anxiety/depression (n = 6)

Comedication: not stated

Sociodemographics: middle class (n = 44), lower class (n = 12); urban (n = 36), suburban (n = 13), rural (n = 7)

Inclusion criteria

  1. Diagnosis based on DSM‐IV‐TR criteria by referring physician ‐ independent confirmation by licensed and/or certified psychologist

  2. Demonstrated significant attention, hyperactivity and/or impulse control problems interfering with major life function in both home and school settings

  3. ≥ 1.5 SD above the mean on ≥ 1 of the attention problems on the Child Behavior Checklist, Teacher Report Form; Inattention and/or Hyperactive‐Impulsive subscales of the Conners' Parent Rating Scale ‐ Revised and Long; or Conners' Teacher Rating Scale ‐ Revised

Exclusion criteria

  1. ≥ 1 comorbid secondary diagnosis

  2. History of mental retardation

  3. Seizure disorder ‐ brain injury

  4. Other medical condition affecting cognitive or neuropsychological performance ‐ missing or different instruments for measuring methylphenidate response (i.e. missing data)

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate (0.15 mg/kg, 0.30 mg/kg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: twice daily

Duration of each medication condition: 4 weeks

Washout before study initiation: 2 days

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale ‐ Revised and Long: teacher‐rated at baseline and at treatment follow‐up

General behaviour

  1. Schools Situation Questionnaire ‐ Revised: teacher‐rated at baseline and at treatment follow‐up

Non‐serious adverse events

  1. Side Effects Rating Scale, unclear who rated: at baseline and at treatment follow‐up

Notes

Sample calculation: no

Ethics approval: not stated

Comment from study authors

  1. Several study limitations are worth noting. First. age limitations. Second, neuropsychological tests that were not counterbalanced and analysed for order effects. Third, inter‐rater reliability during methylphenidate trials. Fourth, intelligence/cognitive screening

Key conclusion of study authors

  1. Robust cognitive and behavioural methylphenidate response was achieved for children with significant baseline executive working memory/self regulation (EWM/SR) impairment, yet response was poor for those with adequate EWM/SR baseline performance. Even for strong methylphenidate responders, the best dose for neuropsychological functioning was typically lower than the best dose for behaviour. Findings offer 1 possible explanation for why long‐term academic methylphenidate treatment gains in ADHD have not been realised

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: June to August 2014. Emailed study authors twice to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

All medications and placebos were prepared by the study pharmacist, who randomly assigned children to 1 of 6 trial orders in placebo (P), low‐dose (L) and high‐dose (H) conditions (P‐L‐H, P‐H‐L, L‐P‐H, L‐H‐P, H‐L‐P, H‐P‐L)

Allocation concealment (selection bias)

Low risk

Research assistants, teacher, parents and participants were blinded to the order of conditions. Ground methylphenidate tablet was placed in lactose‐filled opaque capsules for active drug conditions, with lactose included only for the placebo condition, and was administered twice per day

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Research assistants, teacher, parents and participants were blinded to the order of conditions. All medications and placebos were prepared by the study pharmacist. Ground methylphenidate tablet was placed in lactose‐filled opaque capsules for the active drug condition, with lactose included only for the placebo condition

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

After results were analysed, the order of conditions was revealed

Incomplete outcome data (attrition bias)
All outcomes

High risk

Participants for whom data were missing or different instruments were used for methylphenidate response were excluded

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

Research part funded by the Neuropsychiatric Research Institute, Fargo, North Dakota, USA

Conflicts of interest: Study authors disclose no conflicts of interest

Heriot 2008

Methods

Three‐month, parallel trial with 4 arms

  1. Methylphenidate and parent training programme

  2. Methylphenidate and supportive non‐training parent group

  3. Placebo and parent training

  4. Placebo and supportive non‐training parent group

Participants

Number of participants screened: 93

Number of participants included: 20 to 26 (NB: 6 withdrew, but unclear whether before or after random assignment)

Number of participants randomly assigned: not stated

Number of participants followed up: methylphenidate + parent training 4, methylphenidate + no training 4, placebo + training 4, placebo + no training 4

Number of withdrawals: not stated

Sex: 13 boys, 3 girls

Diagnosis of ADHD: DSM‐IV (combined (25%), hyperactive‐impulsive (56%), inattentive (19%))

Age: mean 4.78 years (range 3 to 6)

IQ: mean 97 (range 80 to 123)

Methylphenidate naive: 100%

Ethnicity: no information

Country: New Zealand

Setting: out‐patient clinic

Comorbidity (type: ODD 5/31%)

Comedication: no

Sociodemographics: SES group 1: 1, group 2: 0, group 3: 3, group 4: 4, group 5: 3, group 6: 5. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Between 3.0 and 5.9 years of age

  2. Resident with a primary caregiver for ≥ 6 months

  3. Meet diagnostic criteria for ADHD as defined in DSM‐IV

  4. Features of ADHD had to be present for ≥ 12 months and to a degree that was considered to be developmentally inappropriate and functionally inappropriate and functionally impairing across settings

  5. Above the 93rd percentile on the Global Index subscale of the Conners’ Rating Scales

Exclusion criteria

  1. Currently in hospital

  2. Currently in another treatment study

  3. Currently receiving treatment

  4. Full scale IQ < 80

  5. Pervasive developmental disorder or psychosis

  6. Major neurological or medical illness that would interfere with participation or require medications incompatible with methylphenidate

  7. Chronic serious tics or Tourette’s disorder

  8. History of child abuse

  9. Inability of parent to understand English

Interventions

Participants were randomly assigned to methylphenidate + parent training, methylphenidate + no parent training, placebo + parent training or placebo + no parent training

Mean methylphenidate dosage: 0.3 mg/kg

Administration schedule: twice a day ‐ morning and lunchtime

Duration of intervention: 3 months

Titration period: Dosage was built up over the first week

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale‐IV Parent and Teacher Versions: data not reported in an useable form ‐ not possible to obtain data from study author

  2. Conners’ Parent Rating Scale ‐ Revised, Long Version: no data provided in the article

  3. Conners’ Teacher Rating Scale ‐ Revised, Long Version: no data provided in the article

Non‐serious adverse events

  1. Stimulant drug, Barkley Side Effects Rating Scale

Notes

Sample calculation: no

Ethics approval: yes; obtained from the University of Waikato, Department of Psychology Ethical Review Committee and the Waikato Ethics Committee for Waikato Hospital

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Seven of the 16 children no longer met DSM‐IV (1994) diagnostic criteria for ADHD at the end of the study

  2. Ethnicity did not appear to affect outcomes, although it was unclear whether the content of the programme was equally appropriate for all ethnic groups

  3. Limitations

    1. Small number of participants

    2. Of those willing to participate, only 43% met eligibility criteria for the study

    3. No follow‐up data

Key conclusion of study authors

  1. Children were more likely to improve when treatment involved ≥ 1 active component (medication or parent training). However, variability in individual parental and child participant responses to all treatment conditions was notable, indicating the importance of interaction between treatment variables and other factors. Findings are discussed within the framework of a transactional model, and inferences are drawn about limitations of the idea that a "best treatment" exists that is universally applicable. Although improvement among children receiving treatment with methylphenidate was greater than for others, it is notable that all treatments were associated with improvement in some children

Email correspondence with study authors: January, February and May 2014. Emailed study author but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Participants were allocated to conditions sequentially, using RAND function (SPSS) to generate the sequence. Each parent was randomly assigned to attend the training programme group or the support group.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Throughout the period of data collection, participants and therapist were blinded to medication status

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Throughout the period of data collection, participants and therapist were blinded to medication status. All parents and teachers were blinded to the intervention

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only data for completing participants were reported

Selection bias (e.g. titration after randomisation → exclusion): unclear. Four were excluded after randomisation because of "treatment integrity problems"

Selective reporting (reporting bias)

Unclear risk

We were unable to identify a protocol

Vested interest bias

Low risk

No funding to conduct the study was received from any party

Conflicts of interest: None of the study authors are affiliated with pharmaceutical companies

Hicks 1985

Methods

Double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate, low and high doses

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 44. Participants were randomly assigned to different low and high doses of methylphenidate and to placebo

Number of participants followed up: 44 (36 boys, 8 girls; 20 in‐patients, 24 out‐patients)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 8.4 years (SD 1.75, range not reported)

IQ: mean 98.31 (SD 12.96)

Methylphenidate naive: not stated

Ethnicity: Caucasian (68.2%), African American (31.8%)

Country: not stated

Setting: out‐patient clinic and patient ward

Comorbidity: not stated

Comedication: not stated

Sociodemographics: mean 3.59 (SD 1.29) (Hollingshead 2‐factor SES index)

Inclusion criteria

  1. Diagnosis of ADHD according to DSM‐III

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to different orders of methylphenidate (low‐dose 0.3 mg/kg, high‐dose 0.6 mg/kg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: b.i.d. morning and noon

Duration of each medication condition: 12 for in‐patients, 19 for out‐patients

Washout before study initiation: not stated

Medication‐free period between interventions: 68 hours

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale: rated weekly

  2. Conners' Teachers Rating Scale: rated weekly

Non‐serious adverse events

  1. Pulse, measured once in each treatment condition, before and 1 hour after treatment

  2. Blood pressure, measured once in each treatment condition, before and 1 hour after treatment

  3. Blood samples, measured once in each treatment condition, before and 1 hour after treatment, including growth hormone, prolactin, thyroid‐stimulating hormone

Notes

Sample calculation: not stated

Ethics approval: yes

Key conclusion of study authors

  1. "MPH appears able to engage homeostatic mechanisms which take over and operate to return functioning to a more normal level"

Comment from review authors

  1. Study participants are both in‐patients and out‐patients; therefore the sample group is highly heterogeneous

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: March 2014. We obtained from study authors supplemental information regarding funding and ethics. Not able to obtain additional data as they are no longer available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Low risk

Funded by the National Institutes of Health (NIH)

Conflicts of interest: not stated

Hoeppner 1997

Methods

Double‐blind, cross‐over trial with 2 interventions

  1. High‐dose (H) methylphenidate

  2. Low‐dose (L) methylphenidate

  3. Placebo (P)

Four orders

  1. P, L, H

  2. H, P , L

  3. P, H, L

  4. L, P, H

Participants

Number of participants screened: 95

Number of participants included: 50. Participants were randomly assigned to different orders of low‐dose and high‐dose methylphenidate and placebo

Number of participants followed up: 50 (39 boys, 11 girls)

Number of withdrawals: none

Diagnosis of ADHD: DSM‐III‐R

Age: mean 9.6 years (range 6 to 18.1)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Meeting DSM‐III‐R ADHD, DSM‐III ADD or DSM‐III‐ADD/H criteria

  2. Score ≥ 1½ SD above the norm on the Conners’ Parent Rating Scale or the Conners’ Teacher Rating Scale

Exclusion criteria

  1. No information

Interventions

Participants were randomly assigned to different possible orders of high‐dose methylphenidate (0.3 mg/kg), low‐dose methylphenidate (0.15 mg/kg) and placebo (doses rounded up to the nearest 2.5 mg)

Mean methylphenidate dosage: no information

Administration schedule: twice a day ‐ 8:00 AM and 12:00 PM

Duration of each medication condition: 1 week

Washout before study initiation: "received an appropriate wash‐out period"

Medication‐free period between interventions: from lunchtime until next morning ‐ 20 hours

Titration period: none

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Conners’ Parent/Teacher Rating Scale: completed daily

Notes

Sample calculation: no

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Dependent measures in this study often failed to detect significant dose‐response effects when analysed individually, and placebo effect were found for 4 of the 9 measures. Although ratings from the Conners' Parent and Teacher Rating Scales were sensitive to dose‐response effects, findings were not uniform across raters and did not correspond with cognitive response

  2. Small sample size

Key conclusion of study authors

  1. According to cognitive rank order ratings, linear and quadratic methylphenidate response patterns were identified, with the best dose for each group significantly different from that obtained for all other conditions

Email correspondence with study authors: March 2014. Emailed study authors to request additional information but have received no reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information

Allocation concealment (selection bias)

Low risk

Pharmacists randomly assigned participants to 1 of 4 trial sequences

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Placebo consisted of an opaque capsule filled with lactose; active drug consisted of the same lactose‐filled capsule, to which methylphenidate was added

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Placebo consisted of an opaque capsule filled with lactose; active drug consisted of the same lactose‐filled capsule, to which methylphenidate was added

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No reporting bias

Vested interest bias

Unclear risk

No information

Horn 1991

Methods

Twelve‐week, double‐blind, randomised, parallel trial with 6 arms

  1. Placebo

  2. Low‐dose methylphenidate

  3. High‐dose methylphenidate

  4. Placebo + behavioural parent training + child self control instruction

  5. Low‐dose methylphenidate + behavioural parent training + child self control instruction

  6. High‐dose methylphenidate + behavioural parent training + child self control instruction.

Nine‐month follow‐up

Participants

Number of participants screened: 117

Number of participants included: 107 (83 boys, 24 girls)

Number of participants randomly assigned: 16 to each arm

Number of withdrawals after randomisation: 18

Number followed up: at end of study (12 weeks) 78, 9 months after termination of study 71

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 8.27 years (range 7 to 11)

IQ: > 70

Methylphenidate naive: not stated

Ethnicity: Caucasian (84.9%), African American (9.4%), Hispanic (3.8%), Asian American (1.9%)

Country: USA

Setting: out‐patient clinic

Comorbidity: conduct disorder (7.5%), disruptive oppositional disorder (15%), oppositional defiant disorder (43%)

Comedication: not stated

Sociodemographics: mean yearly family income USD $25,019. No significant differences in baseline demographics were noted between treatment groups or between treatment groups and treatment drop‐outs in any of the following parameters: child's age, grade, sex, IQ; parental marital status, annual family income and maternal education and age. However, a significantly greater number of non‐white children were included in the placebo alone condition than in remaining treatment conditions or among treatments drop‐outs

Inclusion criteria

  1. 7 to 11 years of age

  2. Exact agreement between a licensed clinical psychologist and a board certified paediatrician with respect to diagnosis of ADHD according to DSM‐III‐R criteria

  3. Score on Hyperkinesis Index of the Conners' Parent or Teacher Rating Scale ≥ 2 SD above published means

  4. Current psychostimulant therapy required to be withdrawn for the course of the study

Exclusion criteria

  1. Comorbid anxiety, depressive disorder or both

  2. Gross physical impairments, intellectual deficits or psychosis in child or parents

Interventions

Washout before study initiation: 2 weeks before initial diagnostic evaluation

Participants were randomly assigned to 1 of 6 treatment conditions, including placebo, low‐dose methylphenidate (0.4 mg/kg) and high‐dose methylphenidate (0.8 mg/kg)

Administration schedule: daily

Duration of intervention: 12 weeks

Titration period: no

Treatment compliance: 87.39% of parents in the non‐placebo medication conditions group reported anonymously that their child took the medications almost every day, whereas the remainder of families reported usage an average of 3 to 4 days a week. Stimulant medication was withdrawn immediately after post‐test assessments

Follow‐up: 9 months

Outcomes

ADHD symptoms

All dependent measures were administered at pre‐test, post‐test (within 1 week or 2 weeks of the end of the treatment phase of the study) and follow‐up

  1. Child Behaviour Checklist

  2. Swanson, Nolan and Pehlam Scale

  3. Conners' Parent Rating Scale

  4. Conners' Teacher Rating Scale

Adverse events

  1. Medication side effects, monitored by a board certified paediatrician

Notes

Sample calculation: no information

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: August 2013. Not possible to receive supplemental information or data through personal email correspondence with study authors. They do not recommend inclusion of the study in this review because of problems with the design and methods used at the time the study was carried out (Ramstad 2013a [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Two of 12 families were randomly assigned to each of the 6 treatment conditions. This procedure was repeated 8 times, so that by the end of the study, 16 families were randomly assigned to each of the 6 treatment conditions, for a total of 96 families

Allocation concealment (selection bias)

Unclear risk

No information

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. Note that assessors were blinded to treatment status at all times

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analyses were conducted on 4 samples: (1) participants completing the entire study and followed up at 9 months (n = 71), (2) participants completing the entire study (n = 78), (3) participants completing the entire study + participants completing ≥ 6 weeks (n = 90) and (4) all of the above + those who dropped out immediately after initiating treatment (n = 96). When no post‐test and/or follow‐up data were available for drop‐outs, pre‐test values or post‐test values, or both, were substituted. Given that analyses produced essentially the same results, only the analyses that include completers are reported

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

Unclear risk

No information

Ialongo 1994

Methods

Fourteen‐week, parallel trial with 3 arms

  1. Placebo

  2. Low‐dose stimulant therapy

  3. High‐dose stimulant therapy

Participants

Number of participants screened: not stated

Number of participants included: 48 (35 boys, 13 girls). 16 children randomly assigned to each of the following 3 treatment conditions: (1) medication placebo alone, (2) low‐dose (0.4 mg/kg) stimulant therapy, or (3) high‐dose (0.8 mg/kg) stimulant therapy. A sample of 21 non‐clinical controls (13 boys and 8 girls) was also included in the study

Number of withdrawals: Seven families dropped out from pre‐test to post‐test: high‐dose methylphenidate 3, placebo 4

Numper of participants followed up: low‐dose methylphenidate 16, high‐dose methylphenidate 13, placebo 12

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 7.97 years (SD 1.4, range 7 to 11)

IQ: ≤ 70

Methylphenidate naive: 98%

Ethnicity: Caucasian (78.0%), African American (12.2%), Hispanic (9.8%)

Country: USA

Setting: out‐patient clinic

Comorbidity: conduct disorder (n = 5), oppositional defiant disorder (n = 12)

Comedication: no

Sociodemographics: middle‐income families. No significant differences between treatment groups or between treatment groups and drop‐outs in any participant and/or demographic characteristics at pre‐test, with the exception of ethnicity. A significantly greater proportion of African American children were included in the placebo condition than in the high‐dose condition

Inclusion criteria

  1. Diagnosis of ADHD according to DSM‐III‐R

  2. Exact agreement between 2 assessors (paediatrician and psychologist) with respect to ADHD diagnosis

  3. Score on Hyperkinesis Indices of the Conners' Parent and Teacher Rating Scale ≥ 2 SD above published means

Exclusion criteria

  1. Comorbid anxiety and/or depressive disorder

  2. Gross physical impairment

  3. Intellectual deficits

  4. Psychosis in child or parents

  5. IQ < 70 (measured with the Peabody Picture Vocabulary Test ‐ Revised)

Interventions

Participants were randomly assigned to 3 treatment conditions: (1) medication placebo alone, (2) low‐dose (0.4 mg/kg) stimulant therapy or (3) high‐dose (0.8 mg/kg) stimulant therapy

Duration of intervention: 14 weeks

Titration period: no

Treatment compliance: One check on medication compliance consisted of periodic dispensing of medication over the course of the study during follow‐up visits to staff paediatricians. In addition, a medical compliance questionnaire was completed anonymously by parents 1 month after post‐test assessments. 91.9% of parents in the medication conditions indicated that their child took the stimulant medication almost every day throughout the study, whereas nearly all remaining families reported average usage of 3 to 4 days a week

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale

  2. Conners' Teacher Rating Scale

General behaviour

  1. Conners' Teacher Rating Scale, Conduct problems

Notes

Sample calculation: no information

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. In contrast to placebo, although methylphenidate resulted in significantly greater amelioration of many of the core features of ADHD, we found no evidence of a decrease in perceived competence, or of an increase in external or unknown explanations of control or dysphoria at post‐test (i.e. 14 weeks later)

Email correspondence with study authors: August 2013 and January 2014. We emailed study authors to request a copy of the protocol and additional information on, for example, the randomisation procedure and funding

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All medication was dispensed in a double‐blinded fashion

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All medication was dispensed in a double‐blinded fashion

Incomplete outcome data (attrition bias)
All outcomes

High risk

No description of imputation method

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Unclear risk

No information

Jacobi‐Polishook 2009

Methods

Single‐day, randomised, controlled, parallel, double‐blind study with 2 arms investigating postural stability in 24 children with ADHD

  1. Methylphenidate

  2. Placebo

Participants

Number of  participants screened: 80

Number of participants included: 24

Number randomly assigned: methylphenidate 12 (11 boys, 1 girl), placebo 12 (11 boys, 1 girl)

Number of participants followed up in each arm: methylphenidate 12, placebo 12

Number of withdrawals in each arm: methylphenidate 0, placebo 0

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: methylphenidate mean 10.06 years (range 7 to 16), placebo mean 10.88 years (range 7 to 16)

Methylphenidate naive: 0%

Ethnicity: Israeli

Country: Israel

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

IQ: > 70

Sociodemographics: not stated

Differences between groups

  1. No significant difference in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. DSM‐IV diagnosis

  2. 7 to 16 years of age

  3. Methylphenidate treatment on a daily basis for the past 3 months

  4. Only methylphenidate responders (improvement in ADHD symptoms after methylphenidate treatment according to parent and teacher reports on the ADHD Rating Scale, Fourth Edition, and according to paediatric neurologist follow‐up)

  5. IQ > 70

  6. ADHD symptoms had to be severe for ≥ 6 items on the DSM‐IV ADHD Rating Scale (ADHD RS‐IV), Parent Version

Exclusion criteria   

  1. Neurological, orthopaedic or psychiatric diagnoses according to DSM‐IV criteria that can affect motor control and postural stability: cerebral palsy, neuropathic disease, limb fracture or head trauma during the previous year

  2. Use of any medication other than methylphenidate during the study period

Interventions

Participants were randomly assigned to 5 mg of short‐acting methylphenidate or 5 mg placebo. They had been drug‐free for 24 hours before the start of the trial

Administration schedule: 5 mg × 1 (single dose)

Duration of intervention: 1 day

Titration period: none

Treatment compliance: 100%

Outcomes

Serious adverse events

  1. No serious adverse events of drug treatment were experienced

Non‐serious adverse events

  1. No non‐serious adverse events of drug treatment were experienced

Notes

Sample calculation: yes

Ethics approval: no information

Key conclusions of study authors

  1. Methylphenidate improves postural stability in ADHD, especially when an additional task is performed, probably through enhanced attention abilities, thus contributing to improved balance control during performance of tasks that require attention

  2. Methylphenidate remains to be studied as a potential drug treatment to improve balance control and physical functioning in other clinical populations

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; see 'Inclusion criteria'

Any withdrawals due to adverse events: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation using a table of random numbers

Allocation concealment (selection bias)

Low risk

Placebo pill was identical in appearance to the methylphenidate pill

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and tester administering the examination were blinded to group assignments

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants and tester administering the examination were blinded to group assignments

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs in either group

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

Unclear risk

No information

Jensen 1999 (MTA)

Methods

Multimodal Treatment Study of Children With ADHD (MTA) (Jensen 1999 (MTA)): 14‐month multi‐centre, randomised, parallel clinical trial with 4 arms

  1. Medication management

  2. Behavioural treatment

  3. Combined treatment (medication management + behavioural treatment)

  4. Community care (control group)

Phases: For the 2 groups receiving medication, an initial 28‐day titration period was provided. This titration phase was carried out as a randomised, double‐blind, placebo‐controlled, cross‐over trial with daily switching of methylphenidate doses (placebo, low, middle and high). Once delivery of randomly assigned treatments by MTA staff stopped at 14 months, the MTA became an observational study in which participants and families were free to choose their own treatment, but in the context of availability and barriers to care existing in their communities. The following follow‐up assessments took place after completion of the RCT at 10 months' follow‐up (24 months after randomisation), 3‐year follow‐up, 8‐year follow‐up and 10‐year follow‐up
In our review, we will compare combined behavioural treatment (RCT) according to our protocol and will look at the medication treatment group as a cohort (observational)

Participants

Titration period (Greenhill 2001) (Jensen 1999 (MTA))

28‐day RCT (combined treatment + medication management group)

Number of participants included: 289 (medication management 144, combined treatment 145)

Number of participants completed: 256

Number of participants not finishing titration: 33. Of the completers, 198 were assigned to an individually best dose of methylphenidate for the 14‐month trial, the rest to other medication or no medication

Main study (Jensen 1999) (Jensen 1999 (MTA))

Number of participants screened: 4541

Number of participants included: 579

Number of participants randomly assigned to methylphenidate + behavioural treatment (combined treatment): 145, MPH 144, behavioural treatment (BEH) 144

Number of participants followed up: combined treatment 142; medication 136; behavioural treatment 141

Number of withdrawals/drop‐outs: combined treatment 3; medication 8; behavioural treatment 3

Demographic data for combined treatment, behavioural treatment and medication management

Diagnosis of ADHD: DSM‐IV (combined (100%))

Age: mean 8.4 years (range 7 to 9.9)

IQ: mean 100.4

Sex: 346 boys, 87 girls

Methylphenidate naive: 177

Ethnicity: White (60.3%), African American (20.6%), Hispanic (8.8%), other (10.4%)

Country: USA and Canada

Setting: out‐patient clinic

Comorbidity: anxiety disorder (35.1%), conduct disorder (14.1%), oppositional defiant disorder (38.8%), affective disorder (3.5%), tic disorder (10.2%), mania/hypomania (3%)

Comedication: not stated

Sociodemographics: 130 families on welfare. Population ranges widely in socioeconomic status. No significant differences in baseline demographics were noted between the 3 groups

10‐Month follow‐up (MTA Group 2004) (Jensen 1999 (MTA))

Number of participants followed up in each arm: MGT 128, COMB 138, BEH 139; age (mean 8.4 years); sex (322 boys, 83 girls). No differences between demographic characteristics of the originally randomly assigned 579 MTA participants and participants assessed during 10‐month follow‐up. The only statistically significant difference among treatment groups was a trivial difference in age (MHT was 0.3 years older than BEH)

3‐Year follow‐up (Jensen 2007) (Jensen 1999 (MTA))

Number of participants followed up: medication 115, combined treatment 127

Age: mean 11.7 years (range 10 to 13)

Sex: 291 boys, 78 girls

No significant differences in baseline characteristics were noted between participants in the 36‐month assessment and those unable to be followed

8‐Year follow‐up (Molina 2008) (Jensen 1999 (MTA))

Number of participants followed up: medication management 101, combined treatment 119. 32.5% of the MTA sample was medicated more than 50% of days the previous year

Age: mean 16.8 years (range 13 to 16). At 8‐year assessment, 55 MTA participants had turned 18 years of age. Only 30% of the sample still fulfilled an ADHD diagnosis. Participants lost to 8‐year follow‐up, compared with those retained, more often were male; had younger mothers, less educated parents and lower parent income; and were more likely to have been on welfare at baseline

10‐Year follow‐up, blood pressure (Vitiello 2012) (Jensen 1999 (MTA))

Number of participants followed up: medication management 77, combined treatment 93. A comparison of participants retained through the year 10 (n = 346) vs those who were not (n = 233) showed a smaller proportion of males in the retained group. Furthermore, participants were divided into groups on the basis of the following criteria: "never medicated", currently medicated, previously medicated. For the currently medicated group, numbers of participants were, respectively, 184, 184, 108, 50 and 12 for 24 months, 36 months, 72 months, 96 months and 120 months of follow‐up, respectively. Medication use during the previous 30 days was the criterion for positive medication status

Growth studies, 24‐month assessment (Jensen 2004) (Jensen 1999 (MTA))

For growth outcomes, data from both the originally randomly assigned treatment groups and naturalistic subgroups were collected. Naturalistic subgroups consisted of those who had been medicated at all assessment points up to 24 months (medication use during previous 30 days was the criterion for positive medication status) and those who had not been medicated at assessment points

Participants with consistent use of medication (med/med): 255

Participants with no use of medication (NoMed/NoMed) 139; combined treatment 135, medication management 120

Growth studies, 36‐month assessment (Swanson 2007) (Jensen 1999 (MTA))

Naturalistic subgroups were established on the basis of patterns of treatment with stimulant medication. If medication was used within a 30‐day period before assessment, medication status was positive (Med); otherwise it was negative (NoMed). If an individual’s medication status changed at any assessment point, he or she was placed in the inconsistently medicated group

Participants with Med (n = 70). At baseline and at 14‐, 24‐ and 36‐month assessment points, respectively, percentages of medicated children taking methylphenidate were as follows: 85.4%, 79.7%, 76.8%, 73.5%. Naturalistic subgroups did not differ in initial size at birth (birth weight), age, parent or teacher ratings of ADHD symptoms, sex, expected adult size (mid‐parent size), welfare status or maternal smoking

Inclusion criteria

  1. Boys and girls

  2. 7 to 9. 9 years of age

  3. Grades 1 through 4

  4. In residence with the same primary caretaker(s) for the past 6 months or longer

  5. Meeting DSM‐IV criteria for ADHD, combined type.

Exclusion criteria

  1. Child currently in hospital

  2. Child currently in another study

  3. < 80 on all Weschler Intelligence Scales for Children, Third Edition, and on Severe Impairment Battery (SIB) (bipolar disorder, psychosis or personality disorder)

  4. Chronic serious tics or Tourette's syndrome

  5. Obsessive‐compulsive disorder serious enough to require separate treatment

  6. Neuroleptic medication in previous 6 months

  7. Major neurological or medical illness

  8. History of intolerance to MTA medications

  9. Ongoing or previously unreported abuse

  10. Missed one‐fourth of school days in previous 2 months

  11. Same classroom as child already in MTA study

  12. Parental stimulant abuse in previous 2 years

  13. Non‐English‐speaking primary caretaker

  14. Another child in same household in MTA study

  15. No telephone

  16. Suicidal or homicidal

Interventions

Titration period (Greenhill 2001) (Jensen 1999 (MTA))

Mean methylphenidate dose during titration period: combined treatment 32.1 mg/d, medication management 28.9 mg/d. Medication management started with a 4‐day single‐blind, safety lead‐in period, during which participants were exposed to 3 progressively higher daily methylphenidate doses given 3 times daily. This was followed by a 28‐day, double‐blind, daily, switch titration of methylphenidate hydrochloride, with 5 randomly ordered repeats each of placebo, 5 mg, 10 mg and 15 mg or 20 mg. Cross‐site teams of experienced clinicians blindly reviewed graphs portraying parent and teacher ratings of responses to each of the 4 doses and by consensus selected each child’s best dose

Compliance: not stated. 29 of 32 placebo responders had to go back to taking methylphenidate during the maintenance period

Main study (Jensen 1999) (Jensen 1999 (MTA))

Participants were randomly assigned to medication management, behavioural treatment or combined treatment

Mean methylphenidate dosage during main study: combined treatment 31.2 mg/d, medication management: 37.8 mg/d

Administration schedule: 3 times a day ‐ breakfast, lunch and in the afternoon

Duration of intervention: 14 months

Treatment compliance: monthly pill counts, intermittent saliva measurements to monitor intake of methylphenidate and encouragement for families to make up missed visits. "The study achieved a high degree of adherence to protocol." NB! For participants not attaining an adequate response to methylphenidate during titration, alternate medications were titrated openly in the following order until a satisfactory choice was found: dextroamphetamine, pemoline, imipramine and others, if necessary approved by a cross‐site panel. Thus, 256 participants successfully completed titration; of these, 198 of 289 participants were assigned to an individually titrated best dose of methylphenidate, and 26 were titrated to dextroamphetamine. 32 were given no medication because of a robust placebo response

10‐Month follow‐up (MTA Group 2004) (Jensen 1999 (MTA))

Duration of intervention: 24 months (10‐month follow‐up from 14‐month RCT)

Treatment compliance: not stated. From end of treatment to first follow‐up, percentage of participants taking medication decreased for combined treatment (87% vs 70%) and methylphenidate (93% vs 72%) but increased for behavioural intervention (23% to 38%)

3‐Year follow‐up (Jensen 2007) (Jensen 1999 (MTA))

Duration of intervention: 3 years (36‐month follow‐up from 14‐month RCT)

Treatment compliance: not stated. At 36 months, percentage of participants taking medication was 70% for the combined group, 72% for the methylphenidate group and 45% for the behavioural intervention group

8‐Year follow‐up (Molina 2009) (Jensen 1999 (MTA))

Duration of intervention: 8 years

Mean methylphenidate dosage: 44.93 mg

10‐Year follow‐up (Vitello 2012) (Jensen 1999 (MTA))

Duration of intervention: 10 years

Treatment compliance: not stated

Mean methylphenidate dosage: 54.3 mg

Outcomes

Titration period (Greenhill 2001) (Jensen 1999 (MTA))

  • Conners' Lonely and Milich Questionnaire (with Inattention/Overactive (I/O))

  • Aggressive/Defiant (A/D) and Mixed (I/O + A/D) subscales: assessed daily by parents and teachers

  • Swanson, Kotkin, Agler, M‐Flynn and Pelham (with Attention and Deportment subscales): rated daily by parents and teachers

Main study (Jensen 1999)

  • SNAP Inattention and Hyperactivity‐Impulsivity subscale, both parent and teacher: assessed at baseline and at 3, 9 and 14 months

  • SNAP Oppositional Defiant Disorder subscale, both parent‐ and teacher‐rated: assessed at baseline and at 3, 9 and 14 months.

  • Abikoff Classroom Observational System (ADHD and oppositional/aggressive symptoms): blind ratings by blind observers

Serious adverse events

Main study (Jensen 1999)

  1. 6 of 11 reported severe side effects could have been due to non‐medication factors

  2. Three deaths were recorded among ADHD participants during 10 years of observation: a suicide at age 14 (participant was taking methylphenidate), a fatal car accident at age 17 (participant was the driver and was taking methylphenidate) and a sudden unexplained death at age 17 (participant was found dead in bed; no specific cause of death could be determined; he had been treated previously with methylphenidate and had been off medication for longer than 1 year when he died)

Non‐serious adverse events

Main study (Jensen 1999)

Participants were provided up to 8 additional sessions when needed to address clinical emergencies or instances of possible study attrition

  1. Pittsburgh Side Effects Rating Scale: monitored monthly, reviewed by the pharmacotherapist

  2. Internalising symptoms (anxiety and depression): measured with an internalising subscale from parent‐ and teacher‐completed Social Skills Responsive Scale (SSRS), measured at baseline and at 3, 9 and 14 months

  3. Children’s self ratings on the Multidimensional Anxiety Scale for Children: assessed at baseline and at 3, 9 and 14 months

Titration period (Greenhill 2001) (Jensen 1999 (MTA))

  1. Pittsburgh Side Effects Rating Scale (10 adverse events commonly associated with methylphenidate were rated)

3‐Year follow‐up (Molina 2007) (Jensen 1999 (MTA))

  1. Substance abuse: assessed at 24 and 36 months by a child‐reported substance use questionnaire (Molina and Pelham, 2003) adapted for the MTA. The measure included items for lifetime and current (past 6 months) use of licit substances (alcohol, cigarettes, chewing tobacco) and illicit drugs (marijuana and other street drugs). Also included were items for inappropriate or non‐prescribed use of medications, including stimulants

  2. Delinquency: assessed by the Self‐Reported Antisocial Behavior Questionnaire through 24‐month assessment and the Self‐Reported Delinquency Questionnaire at 36‐month assessment. Delinquency was coded along an ordinal scale on the basis of the most serious act committed during the past 6 months: 0 = no delinquency; 1 = minor delinquency only at home (e.g. theft of less than $5 or vandalism); 2 = minor delinquency outside the home (e.g. vandalism, cheating someone, shoplifting less than $5); 3 = moderately serious delinquency (e.g. vandalism, theft of $5 or more, weapon carrying); 4 = serious delinquency (e.g. breaking and entering, drug selling, attacking someone with the intent to seriously hurt or kill, rape); and 5 = engagement in 2 or more different Level 4 offences. Because only a small number of MTA children were coded 5 (n = 14 at baseline, n = 4 and 5 between 14 and 36 months), we grouped codes 4 and 5 for data analyses, making a 5‐level ordinal scale of 0 to 4

8‐Year follow‐up (Molina 2009) (Jensen 1999 (MTA))

  1. Delinquent behaviour coded on a 5‐point ordinal scale using parent and youth report across several measures

  2. Number of contacts with police and arrests using the Services for Children and Adolescents‐Parent Interview, parent reported

  3. Depression rated by the Children’s Depression Inventory, self rated

  4. Anxiety rated with by Multidimensional Anxiety Scale for Children, self rated

  5. Psychiatric hospitalisation by 8 years, parent reported

8‐Year follow‐up, substance abuse (Molina 2009) (Jensen 1999 (MTA))

  1. Substance use: assessed by a child/adolescent‐reported questionnaire adapted for the MTA. Substance use outcomes were measured at all interviews beginning with 24‐month assessment

For analysis of stimulant treatment duration in relation to substance use at 8‐year follow‐up, the primary outcome was the number of substances used in the past 6 months, to ensure that most stimulant treatment received would have preceded substance use. Component variables included the following: "drunk" once or more or drank alcohol 3 to 4 or more times; 1 or more cigarettes per day in the past month (time frame exception specific to tobacco); marijuana 2 or more times; and any other illicit drug use or prescription medication misuse. Secondary analyses explored each class of substances separately. For analysis of stimulant treatment exposure over time in relation to substance use at 8‐year follow‐up, the primary outcome variable was substance use disorder in the past year for any substance (excluding tobacco). Secondary analyses explored alcohol and marijuana/other drug use disorders separately

10‐Year follow‐up (Vitello 2012) (Jensen 1999 (MTA))

  1. Blood pressure and heart rate monitoring, measured after participants had been sitting for 5 minutes, adjusted for age and sex

  2. Height and weight

  3. Hospitalisation measured at each assessment point

  4. No symptomatic cardiovascular events leading to medical attention were reported during the period of observation, and no stimulant treatment discontinuation consequent to cardiovascular adverse effects occurred during the 10‐year period

Growth (Jensen 2004, Swanson 2007) (Jensen 1999 (MTA))

  1. Height in centimeters and weight in kilograms, assessed at baseline, 14 months, 24 months and 36 months

Notes

Sample calculation: yes; power analysis, with 576 participants required

Ethics approval: yes; approved by both local institutional review boards and National Institutes of Health Office for Protection From Research Risk

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any drop‐outs due to adverse events: Four participants were removed during the lead‐in (titration period) because of prohibitive side effects: 1 child with buccal movements; another with skin picking; a third with depression, crying, sleep delay and appetite loss; and a fourth who was anorexic, listless and emotionally constricted

Comments from study authors

  1. Main study (Jensen 1999)

    1. Recruitment, screening and selection procedures aimed to collect a carefully diagnosed sample of impaired children with ADHD and a wide range of comorbid conditions and demographic characteristics representative of patients seen in clinical practice

    2. The design did not include a no‐treatment or placebo group

    3. More than three‐fourths of participants given behavioural treatment were successfully maintained without medication throughout the study. Consequently, it should not be concluded that behavioural treatment interventions did not work

    4. Combined treatment and medication management were clinically and statistically superior to behavioural treatment and community care in reducing children’s ADHD symptoms. (...) For other areas of function (oppositional/aggressive behaviours, internalising symptoms, social skills, parent‐child relations and academic achievement), few differences among our treatments were noted, and when found, were generally of smaller magnitude

    5. Significantly lower total daily dose of methylphenidate in the combined treatment arm is noteworthy but was not unforeseen. The importance of this finding is unclear, and a rigorous test of the question would likely require a different design

  2. Titration period (Greenhill 2001) (Jensen 1999 (MTA))

    1. Short 28‐day duration limited generalisability to long‐term treatment

    2. Rates of response to methylphenidate ran between 70% and 80% within the expected range

    3. MTA titration study showed a steeper dose‐response curve for younger and lighter children with ADHD

    4. When ratings were collected under placebo‐controlled, double‐blind conditions, parents reported more adverse events than did teachers. For this reason, clinicians would be wise to collect methylphenidate side effect ratings from parents in the afternoons and evenings

  3. 10‐Month follow‐up (MTA Group 2004) (Jensen 1999 (MTA))

    1. At follow‐up, dose levels of medication management participants (in methylphenidate equivalents) were significantly higher than those of participants in the combined treatment group. Interesting results suggest the possibility that early combined treatment interventions might allow reduced overall medication requirements during later periods, consistent with findings reported by others

  4. 3‐Year follow‐up (Jensen 2007) (Jensen 1999 (MTA))

    1. By 36 months, none of the randomly assigned treatment groups differed significantly in any of the 5 clinical and functional outcomes. (..) However despite no significant group differences at 36‐month assessment, substantial improvement was manifested by all groups. As no untreated control groups were included, and because all treatment groups were improved in terms of relevant symptoms at 36 months compared with baseline, it is possible that all treatments worked but at different rates in different time periods

    2. It is interesting that both medication and educational services provided for 24 to 36 months were markers for poorer outcome at 36 months, suggesting that those who are doing poorly are given additional treatment, yet still do not do as well as those for whom treatment is not considered essential

  5. Vitiello 2001 (Jensen 1999 (MTA))

    1. Comorbid anxiety, oppositional defiant disorder or conduct disorder was not associated with statistically significant differences in methylphenidate dose at the end of titration or maintenance, the number of medication changes or the time to first change

    2. A short‐term response to placebo occurred in 32 children (of 256 who completed double‐blind titration) but was maintained in only 3 patients over the long term

  6. Conners 2001 (Jensen 1999 (MTA))

    1. It is clear that the treatment effect in this study depends on the choice of endpoint measure. Results highlight the fact that no "one true outcome" can be found for a randomised clinical trial, because different measures may be sensitive to different forms of treatment

  7. 8‐Year follow‐up (Molina 2008) (Jensen 1999 (MTA))

    1. Across time (to 8‐year follow‐up), 17.2% of children were medicated at every assessment, beginning with the 14‐month report

  8. 10‐Year follow‐up (Vitiello 2012) (Jensen 1999 (MTA))

    1. This clinical trial was not specifically designed to evaluate cardiovascular function

    2. Blood pressure and heart measurements were not conducted under double‐blind conditions, and measurement methods varied across clinical sites

    3. Abnormal blood pressure values were not systematically confirmed over 3 separate assessments, as required for a diagnosis of pre‐hypertension or hypertension

    4. Time of day when measurements were taken was variable

  9. Implications and applications for primary care providers (Jensen 2001) (Jensen 1999 (MTA))

    1. Behavioural treatments may help families actively cope with their child’s disorder while making necessary life accommodations to optimise family functioning, even when such treatments are not as effective as medication in reducing symptoms of children with ADHD

    2. Findings suggest that high‐quality treatments may have considerable impact on restoring children with ADHD to normal or near‐normal functioning at home and in the classroom. Because essentially none of the children with ADHD met the normal criteria met by 88% of comparison children drawn from the same classrooms at study outset, the notion that ADHD is just normal behaviour labelled by uninformed parents or overwhelmed teachers appears not only implausible, but preposterous

  10. W.E. Pelham 1999 (Jensen 1999 (MTA))

    1. Two major treatment modalities ‐ behavioural and pharmacological ‐ were assessed at different time points relative to the intensive phase of treatment. Specifically, the effects of pharmacological treatments were assessed at post treatment, while participants were actively medicated; in contrast, the effects of behavioural treatment were assessed after therapist involvement diminished. The intensive period of behavioural treatment ended in late December or early January, and endpoint measures typically were taken 4 to 6 months later ‐ usually several months after the last planned, face‐to‐face therapeutic contact. This design aspect has numerous implications for interpretation of study findings. For example, we cannot state that the medication (methylphenidate for the vast majority) had long‐term effects. Rather, results simply demonstrate that effects of methylphenidate given steadily for 14 months are the same at the end of that time as at the beginning (indeed the correlations between drug effects at these 2 points of the study are very high). When differences in outcomes between these groups (e.g. behavioural therapy, medication management) are analysed, data suggest it is likely that combined treatment for children whose parents and teachers continued the behavioural interventions they had been taught will have outcomes superior to those of medication management, and outcomes with combined treatment for those whose parents and teachers did not continue behavioural treatment will be equivalent to those with medication management alone (which would not be surprising, as functionally this is what they would be receiving)

  11. Growth, 24‐month outcomes (Jensen 2004) (Jensen 1999 (MTA))

    1. Growth suppression effects could be related to a medication effect, with the continuously treated subgroup showing slower growth than the untreated subgroup. Alternatively, the "Continuously treated subgroup", defined by unknown self selection factors, could have had a slower growth rate before the start of the study, which continued during treatment and follow‐up phases of the MTA. Our data cannot permit a determination of the validity of these alternative interpretations. At this first follow‐up, our observations were of children in the MTA when they were between the ages of 9 and 11 years, which is before the expected phase of accelerated growth in adolescence and before the age when growth is expected to slow and final height is approximated. Rate of growth and length of the growth phase together determine ultimate (adult) height, and it is possible that consistent treatment with medication may reduce the rate but lengthen the duration of growth, so final height would be delayed but not reduced. It is possible that the never‐medicated group was pared down to good responders and the medicated groups were enriched by poor behavioural responders. In the analysis of 14‐ to 24‐month change scores, "Medication status" was significant for both height (X² = 16.16, P value < 0.001) and weight (X² = 13.32, P value < 0.004)

  12. Growth, 36‐month assessment (Swanson 2007) (Jensen 1999 (MTA))

    1. We did not document a decrease in relative size among the group of participants with a history of treatment before entry into the MTA protocol during subsequent treatment with stimulant medication over 3 years. However, this group (the consistently medicated naturalistic subgroup) was smaller than the stimulant naive group (the newly medicated naturalistic subgroup at entry), suggesting that early treatment of children (before 7 to 9 years of age) with stimulant medication may have produced a reduction in growth rate before entry into the MTA protocol

Key conclusions of study authors

  1. Main study (Jensen 1999)

    1. For ADHD symptoms, our carefully crafted medication management was superior to behavioural treatment and to routine community care that included medication. Our combined treatment did not yield significantly greater benefits than medication management for core ADHD symptoms but may have provided modest advantages for non‐ADHD symptoms and positive functioning outcomes

  2. Vitiello 2001 (Jensen 1999 (MTA))

    1. For most children, initial titration found a dose of methylphenidate in the general range of the effective maintenance dose but did not prevent the need for subsequent maintenance adjustments. For optimal pharmacological treatment of individuals with ADHD, both careful initial titration and ongoing medication management are needed

  3. Titration period (Greenhill 2001) (Jensen 1999 (MTA))

    1. The MTA titration protocol validated the efficacy of weekend methylphenidate dosing and established a total daily dose limit of 35 mg of methylphenidate for children weighing < 25 kg. It replicated previously reported methylphenidate response rates (77%), distribution of best doses (10 mg/d to 50 mg/d) across participants, effect sizes on impairment and deportment and dose‐related adverse events. With t.i.d. dosing, the MTA titration trial showed that significant stimulant medication effects on ADHD symptom reduction and drug‐related adverse events could be detected by parents and teachers by using daily ratings under controlled conditions

  4. 10‐Month follow‐up (MTA Group 2004) (Jensen 1999 (MTA))

    1. The benefits of intensive medical intervention for ADHD extend 10 months beyond the intensive treatment phase only in symptom domains and diminish over time.

  5. 3‐Year follow‐up (Jensen 2007) (Jensen 1999 (MTA))

    1. By 36 months, the earlier advantage of having had 14 months of the medication algorithm was no longer apparent, possibly because of age‐related decline in ADHD symptoms, changes in medication management intensity, starting or stopping of medications altogether or other factors not yet evaluated. At 24 and 36 months, investigators assessed delinquency and substance abuse

  6. Molina 2007 (Jensen 1999 (MTA))

    1. Cause‐and‐effect relationships between medication treatment and delinquency are unclear; absence of associations between medication treatment and substance use must be re‐evaluated at older ages

    2. Findings underscore the need for continuous monitoring of these outcomes as children with attention deficit hyperactivity disorder enter adolescence. No statistically significant effects of randomly assigned treatment on individual rate of change in delinquency were observed between baseline and 36 months at the P value < .05 level. Results suggest that increasing delinquency between 24 and 36 months was associated with an increase in substance use during the same time period. We found no evidence of protective or adverse effects of medication treatment for ADHD in either study

  7. 8‐Year follow‐up (Molina 2009) (Jensen 1999 (MTA))

    1. Type or intensity of 14 months of treatment for ADHD in childhood (at age 7 to 9.9 years) does not predict functioning 6 to 8 years later. Rather, an early ADHD symptom trajectory regardless of treatment type is prognostic. This finding implies that children with behavioural and sociodemographic advantages, with the best response to any treatment, will have the best long‐term prognosis. As a group, however, despite initial symptom improvement during treatment that is largely maintained after treatment, children with combined‐type ADHD exhibit significant impairment in adolescence. Innovative treatment approaches targeting specific areas of adolescent impairment are needed

  8. 10‐Year follow‐up, blood pressure (Vitiello 2012) (Jensen 1999 (MTA))

    1. Stimulant treatment did not increase the risk for pre‐hypertension or hypertension over the 10‐year period of observation. However, stimulants had a persistent adrenergic effect on heart rate during treatment

  9. Growth studies, 24‐month follow‐up (Jensen 2004) (Jensen 1999 (MTA))

    1. In the MTA follow‐up, exploratory naturalistic analyses suggest that consistent use of stimulant medication was associated with maintenance of effectiveness but continued mild growth suppression

  10. Growth studies, 3‐year follow up (Swanson 2007) (Jensen 1999 (MTA))

    1. Combined‐type attention‐deficit hyperactivity disorders were, as a group, greater than expected from norms before treatment but show stimulant‐related decreases in growth rate after initiation of treatment, which appeared to reach asymptotes within 3 years without evidence of growth rebound

  11. W.E. Pelham 1999 (Jensen 1999 (MTA))

    1. Active medication for ADHD is better than withdrawn behavioural treatment (on some but not most measures)

    2. Combined treatment adds modestly to active medication but is superior to behaviour management alone

    3. Study treatments that include active medication are better than community treatments that include medication, and behavioural treatment is comparable with medication as delivered in the community

    4. Concurrent BT results ≥ 20% lower and increasing medication dosages relative to treatment with medication alone

  12. Comorbidity (Jensen 2001) (Jensen 1999 (MTA))

    1. Our findings suggest that children with ADHD with and without oppositional defiant disorder/conduct disorder and anxiety differed in many baseline characteristics, outcomes and response to treatment. Children with anxiety tended to be more treatment‐responsive than those with ADHD + oppositional defiant disorder/conduct disorder, and even ADHD‐only participants

  13. Anxiety (March 2000) (Jensen 1999 (MTA))

    1. Contravening earlier studies, no adverse effects of anxiety on medication response for core ADHD or other outcomes in anxious or non‐anxious ADHD children was demonstrated. When ADHD is treated, it is important for practitioners to search for comorbid anxiety and negative affectivity and to adjust treatment strategies accordingly

  14. Swanson 2007 (Jensen 1999 (MTA))

    1. Long‐term benefits of consistent treatment were not documented; selection bias was not shown to account for loss of relative superiority of medication over time; no evidence of "catch‐up" growth was found; early treatment with medication did not protect against later adverse outcomes. We expect that these challenges to views of practitioners will contribute to future controversy about long‐term outcomes of the MTA

  15. Molina 2012, substance use (Jensen 1999 (MTA))

    1. Our findings provide no evidence that ADHD medication protects from, or increases risk for, adolescent substance use, or SUD. This finding held for recent medication and for days of cumulative treatment with stimulants. Unmeasured confounders may have been operating because of the naturalistic follow‐up study design, and we did not statistically control for psychopathology and functioning at follow‐up assessment. Observed lack of association between stimulant exposure over time and adolescent substance use/SUD does not eliminate the possibility that brain‐based changes in neural mechanisms underlying addiction vulnerability are occurring as a function of prolonged stimulant treatment. Substance use/SUD outcomes for the MTA should be considered in the context of several unique study features and limitations. All children in the MTA were diagnosed with the combined type of DSM‐IV ADHD, and generalisation of study results generally should not extend beyond this subtype. Our follow‐up assessments, which relied on self report, often with 2‐year windows, might have missed episodes of substance use, and rates may be underestimated

  16. Pelham 2000 (Jensen 1999 (MTA))

    1. The Diagnostic Interview Schedule for Children interview shows that 75% of children in the behavioural treatment group were maintained without medication for 14 months, and 64% did not meet diagnostic criteria for ADHD at 14 months (MTA Cooperative Group, 1999, 1999a). Such findings highlight the fact that intensive behavioural treatments are a viable alternative to medication for treatment of individuals with ADHD

Comments from review authors

  1. Authors from the MTA study have written more than 70 articles describing different outcomes and challenges of the study. We have included only those found through our comprehensive literature search and others that we found relevant to include upon looking through article reference lists

  2. We have discussed whether to include the MTA study, as not all participants randomly assigned to medication (combined treatment and medication management group) received methylphenidate. Those who did not have an adequate response to methylphenidate were given other medication (e.g. dextroamphetamine, pemoline, imipramine) or no medication. Furthermore, some participants in the BEH group were also medicated during the 14‐month randomisation phase. From all other studies in this review, we have included only receivers of pure methylphenidate. Furthermore, lots of participants did not have an ADHD diagnosis at follow‐up assessment. At 8‐year follow‐up, only 30% of remaining participants still had a diagnosis of ADHD. However, we have chosen to use the data from MTA, as it is such a large and well‐known study. All MTA analyses will be included in the review as sensitivity analyses

  3. Regarding Molina 2012 (substance use): We have included/asked for additional data from this study, even though the medicated group received medication for a mean of only 2071.10 (SD 728.87) days of the 8 years the follow‐up took place

  4. The following articles from the MTA study have been assessed by only 1 review author: Pelham 2000, Carey 2000, Swanson and Hinshaw 2007, Galanter 2003, Hinshaw 1999, Molina 2013

Email correspondence with study authors: January 2014 to June 2014. We sent several emails to the MTA group to request additional information. However, we were not able to obtain additional data. We did receive an email from Dr. Hinshaw confirming that the data on ADHD symptoms, parent‐rated, were wrong ‐ instead of a mean of 1.85, the correct mean was 0.85 for combined treatment after 14 months. We also received an email from Dr. Swanson in June 2014 stating that he would help with data collection. We wanted to conduct a reanalysis of data excluding those few participants not receiving methylphenidate. Dr. Swanson proved several helpful comments on this and enclosed published articles, but we did not receive additional data, in part because of the time frame of this review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was done centrally by the National Institute of Mental Health (NIMH) Data Center, stratified by site in blocks of 16 (4 to each group). Stratified by 6 sites. Sealed, ordered envelopes were sent to sites for successive entries

Allocation concealment (selection bias)

Low risk

Sealed, ordered envelopes were sent to sites for successive entries. Treatment assignment was concealed until the family confirmed agreement to accept randomisation

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Treatment assignment was concealed until the family confirmed agreement to accept randomisation. After agreement on best dose, the blind was broken, and the agreed on dose (if not placebo) became the participant’s initial maintenance dose

Blinding of outcome assessment (detection bias)
All outcomes

High risk

After agreement on best dose, the blind was broken, and the agreed on dose (if not placebo) became the participant's initial maintenance dose. However, for some outcome measures, 3 strategies were devised to enlist blinded raters and objective observations

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analyses. Despite high compliance, we checked whether compliance with assessments (i.e. missing data) could have changed our findings. Random‐effects regression analyses were completed 2 ways: once with inclusion of all participants, and then including only participants who provided data over multiple time points during the study. No differences emerged from these 2 sets of analyses

Selective reporting (reporting bias)

Low risk

This study was supported by several grants from the National Institute of Mental Health (NIMH), Bethesda, Maryland

Vested interest bias

Low risk

This study was supported by several grants from the National Institute of Mental Health, Bethesda, Maryland

Conflicts of interest: Several study authors have affiliations with medical companies

Johnston 1988

Methods

Two‐week cross‐over trial with 3 interventions

  1. Methylphenidate

  2. Placebo

  3. Sustained‐release methylphenidate (we do not report on this group here)

Phases: to define rebound effects in 21 boys 4 to 10 years of age, with a DSM‐III diagnosis of ADD and treated with methylphenidate some days, and placebo other days

Participants

Number of participants screened: not stated

Number of participants included: 21. Participants were assigned to different orders of methylphenidate and placebo

Number of participants followed up: 21 (21 boys, 0 girls)

Number of withdrawals: 0

Diagnosis of ADD: DSM‐III‐R (subtype not stated)

Age: mean 7 years 7 months (range 4 to 10 years)

IQ: mean 101 (range 79 to 120)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: conduct disorder (n = 2), oppositional defiant disorder (n = 17), learning disability (n = 9)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐III‐R diagnosis of ADD

Exclusion criteria

  1. Not stated

Interventions

Random sequence allocation to

  1. 0.3 mg/kg methylphenidate twice daily (at breakfast and just before lunch; n = 21)

  2. 0.6 mg/kg methylphenidate twice daily (at breakfast and just before lunch; n = 16 of the same 21 participants)

  3. 20 mg sustained‐release methylphenidate (n = 8). Not reported here

  4. Placebo

Within‐participant random sequence, condition varied daily

Mean methylphenidate dosage: not stated

Duration of intervention: 2 weeks. Note: It is not clear for how many days each boy received either of the methylphenidate doses or placebo

Treatment compliance: not stated

Outcomes

ADHD symptoms

  • Modified Conners' Scale Parent, daily, and Teacher ACR, 2 to 3 ratings per treatment condition. Daily reports of social and academic behaviour. Not stated who did the rating for these reports

  • Swanson, Nolan and Pelham Scale, teacher‐rated

  • Abbreviated Conners' Rating Scale

  • IOWA Conners' Teacher Rating Scale

Non‐serious adverse events

  • Rebound/Modified Conners' Scale, Parent, for rebound effect assessment

  • Specific Behaviour Ratings by parent (5, specific individual problem behaviours, rated every night)

Notes

Sample calculation: none

Ethics approval: no information

Funding/vested interest: no information

Any withdrawals due to adverse events: no

Author affiliations: University. No conflicting interests stated

Comment from review authors

  1. Non‐validated endpoints used. No definition (defined cutoff score on scales) of what the authors considered to be ‘rebound’

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated

Email correspondence with study authors: Emailed study authors to request additional information. Also asked whether this study includes the Pelham 1989 reference. No answer from study author, so we extracted data as from 2 different studies

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Order of drug condition for each child was randomly assigned over days

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Child, parent, teacher and programme counsellors were blinded to the condition. Active medication and placebo were disguised in gelatin capsules and pre‐packaged in individually dated envelopes

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Low risk

Conflicts of interest: During the writing of this report, C. Johnston was supported by a Doctoral Fellowship from the Social Sciences and Humanities Research Council of Canada

Kaplan 1990

Methods

Three open trials, then a placebo‐controlled, double‐blind, cross‐over study with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 6. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 6 (all boys)

Number of withdrawals: 0. Three out‐patients were studied in an open‐label design

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 14.4 years (range 13 to 16)

IQ: mean 86 (range 76 to 97)

Methylphenidate naive: 5 (55%)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic and in‐patient ward

Comorbidity: aggressive conduct disorder (100%)

Comedication: yes; diphenhydramine 50 mg

Sociodemographics: not stated

Inclusion criteria

  1. Meeting DSM‐III criteria for both ADHD and aggressive conduct disorder

Exclusion criteria

  1. Consent from parent or custodial social service agency

  2. Psychosis or drug abuse history

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.6 mg/kg, with 30 mg as a ceiling for methylphenidate and placebo

Mean methylphenidate dosage: 0.47 mg/kg

Administration schedule: twice a day, 8:00 AM and noon

Duration of each medication condition: 3 weeks

Washout before study initiation: 1 week

Medication‐free period between interventions: 20 hours

Titration period: 1 week after randomisation

Treatment compliance: no information

Outcomes

ADHD symptoms

  • Conners’ Teacher Rating Scale: completed weekly by classroom teachers

  • In addition, for in‐patients, Conners' Rating Scale was completed weekly by the unit nurse

Non‐serious adverse events

  • Treatment Emergent Side Effect Scale: completed weekly by treating psychiatrists

  • Dizziness, appetite loss and headache were reported in 3 of the 9 youngsters

Notes

Sample calculation: no

Ethics approval: no information

Comment from study authors

  1. As the result of an oversight, assignment to methylphenidate‐first or placebo‐first condition was made in the absence of a specific randomisation formula. Five of the 6 participants received placebo for the first condition and methylphenidate for the second condition

Key conclusion of study authors

  1. Findings provide preliminary evidence of the efficacy of methylphenidate in reducing aggression among aggressive conduct‐disordered adolescents also diagnosed with ADHD

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: March 2014. Emailed study author twice to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

As the result of an oversight, assignment to methylphenidate or placebo as the first condition was made in the absence of a specific randomisation formula. Five of the 6 participants received placebo for the first condition and methylphenidate for the second condition

Allocation concealment (selection bias)

Low risk

Assignment to order condition was determined with no knowledge of the particular participants involved; thus participants did not receive 1 condition or the other based on symptoms or any other systematic bias

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

In 2 cases, school vacation at the in‐patient setting prevented teacher ratings from being obtained during 1 condition of the study. In those 2 instances, the decision was made to use Conners' ratings obtained from the unit nurse for both the condition for which no teacher ratings were provided and the condition for which teacher ratings were given; thus comparisons were consistent in terms of rater

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Unclear risk

No information

Kelly 1989

Methods

Double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Followed by long‐term follow‐up of 12 children out of 21 who continued to receive methylphenidate. Follow‐up for an average of 16 months

Phases

  1. Baseline

  2. Cross‐over study

  3. Follow‐up

Participants

Regarding the cross‐over trial

Number of participants screened: not stated

Number of participants included: 26, but 5 children dropped out, 3 were removed by parents and 2 were disqualified because of a death in the family in 1 and a protocol procedural error in the other

Number of participants randomly assigned: 21 (18 boys, 3 girls)

Number of participants followed up: 21, plus 2 participants who had been withdrawn but returned later for follow‐up

Number of withdrawals to follow‐up: 0, but data for a few variables were not obtained for all participants

Diagnosis of ADHD: DSM‐III

Age: mean 9.3 years (range 8 to 12)

IQ: mean 100.7

Methylphenidate naive: 100%

Ethnicity: White (62%), Hispanic/Oriental/Black (14%), mixed race (24%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional disorder (14%), enuresis (38%)

Comedication: not stated

Sociodemographics: 2‐parent household (95%)

Inclusion criteria

  1. ADHD diagnosis according to DSM‐III

  2. IQ > 80

  3. Free of major health problems, neurological disorders or psychosis

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to methylphenidate or placebo

Methylphenidate dosage: between 0.3 and 0.6 mg/kg/d in the short‐term phase

Administration schedule: 2 time points a day

Duration of each medication condition: not stated

Washout before study initiation: from noon to the following morning (i.e. the next day)

Titration period: no

Outcomes

ADHD symptoms

  1. Attention Deficit Disorder‐Hyperactivity: Comprehensive Teacher Rating Scale: rated before diagnosis, at cross‐over, at the conclusion of the short‐term protocol and again during long‐term follow‐up

  2. Conners' Parent Questionnaire: rated before diagnosis, at cross‐over, at the conclusion of the short‐term protocol and again during long‐term follow‐up

Notes

Sample calculation: not stated

Ethics approval: yes

Key conclusion of study authors

  1. Findings indicate that many pre‐adolescents with ADHD exhibit low self esteem. Despite clinical response to medication, short‐term improvement in self esteem may not occur; however, long‐term, multi‐modal management that includes medication does appear to improve self esteem

Comment from review authors

  1. The data that we used in the review were derived from the cross‐over period described

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: January 2014. Study authors not able to provide us with further information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Child assigned in a double‐blind, cross‐over format to methylphenidate followed by placebo or placebo followed by methylphenidate

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Because data for a few variables were not obtained for all participants, a procedure for unbalanced analysis of variance was required and was accomplished by using the general linear model (GLM) procedure in the Statistical Analysis System (SAS)

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Unclear risk

CIBA Geigy Pharmaceuticals provided placebos

Conflicts of interest: no information

Kent 1995

Methods

Double‐blind, cross‐over trial with 3 interventions

  1. Methylphenidate 10 mg

  2. Methylphenidate 15 mg

  3. Placebo

Phases: The first 2 doses (7:00 AM and noon) were unchanged from the open trial, whereas the 4:00 PM dose was 10 mg of methylphenidate, 15 mg of methylphenidate or placebo. Each of these 3 4:00 PM medication conditions was administered in random order during the 12‐day period. Each medication condition was administered for a total of 4 days

Participants

Number of participants screened: not stated

Number of participants included: 12 (11 boys, 1 girl). Participants were randomly assigned to different possible drug condition orders of methylphenidate and placebo

Number of participants followed up: not stated (but all 12 seem to appear in the results)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 9.0 years (SD 2, range 5.5 to 11.25)

IQ: not stated

Methylphenidate naive: 60%

Ethnicity: not stated

Country: USA

Setting: in‐patient ward

Comorbidity: oppositional defiant disorder (25%), learning disability and oppositional defiant disorder (50%), conduct disorder (8%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. In addition to fulfilling ADHD diagnostic criteria, patients were considered for the study only if they showed a beneficial response to an open trial of methylphenidate, as discussed below

Exclusion criteria

  1. Major depressive disorder

  2. Separation anxiety disorder

  3. Tics or history of tics

  4. Glaucoma

  5. Psychosis or history of psychosis

  6. Known hypersensitivity to methylphenidate

Interventions

Participants were randomly assigned to different possible drug condition orders of 10 mg or 15 mg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 7:00 AM, noon and 4:00 PM

Duration of each medication condition: 4 days

Washout before study initiation: none stated

Medication‐free period between interventions: none

Titration period: Open titration of methylphenidate was accomplished within 14 days of the child’s admission

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Child Behavior Rating Form, rated by nursing staff: after each day shift and each evening shift

Non‐serious adverse events

  1. Sleep latency, sleep adequacy, food intake and weight: recorded by nursing staff

Notes

Ethics approval: Informed consent for study participation was obtained from each child's parent or guardian. All procedures were approved by the study site's Human Subjects Research Review Committee

Comment from study authors

  1. Patient population studied was particularly disturbed, and data were obtained in the context of in‐patient treatment

Key conclusions of study authors

  1. This study's findings show that children with ADHD derive substantial symptom reduction from methylphenidate administered in late afternoon, with no untoward effects on sleep. Therefore, t.i.d. should be considered for those children who exhibit ADHD symptoms in the evening

  2. Adverse effects on sleep latency were not apparent in the sample overall

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; patients were excluded from consideration for study participation if they had known hypersensitivity to methylphenidate

Any withdrawals due to adverse events: none

Email correspondence with study authors: March 2014. Sent an email to study authors to request additional information but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Each of the 3 4:00 PM medication conditions was administered in random order during the 12‐day double‐blind cross‐over period

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The identity of each day’s 4:00 PM dose was known only to the hospital pharmacist, until the child completed the 12‐day protocol

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Results are provided for all 12 children

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

This work was supported by the John and Maxine Bendheim Fellowship and by the Leon Lowenstein Foundation

Conflicts of interest: not stated

Kent 1999

Methods

Cross‐over trial with 3 interventions

  1. Methylphenidate 0.3 mg/kg

  2. Methylphenidate 0.6 mg/kg

  3. Placebo

Phases: 3‐week, double‐blind, 2‐way cross‐over, long‐term (≥ 12 months) follow‐up

Participants

Number of participants screened: not stated

Number of participants included: 50 (38 boys, 12 girls). Participants were randomly assigned to different possible drug condition orders of methylphenidate and placebo

Number of participants followed up: 43

Number of withdrawals: 7

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean not reported (range 4 to 14 years)

IQ: "overall normal intelligence"

Methylphenidate naive: not stated

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: depression (37%), anxiety (37%), learning disability (51%), conduct disorder (5%), "psychiatric disorder" (2%), Tourette’s disorder (12%), "other" (23%)

Comedication: not stated

Sociodemographics: "Fifteen (30%) live in households that have a family income below the Canadian poverty line ($20,000/y)", 26 "rural", 9 "suburban", 14 "urban". 21 live with 1 biological parent, 24 live with both parents, 4 live with adoptive parents or "guardians"

Inclusion criteria

  1. ADHD diagnosis

  2. 4 to 14 years of age

  3. English or French speaking

  4. Living with caretakers with whom they had lived for longer than 6 months

  5. Presence of a teacher who could evaluate the child in class

Exclusion criteria

  1. History of significant developmental delay

  2. Previous diagnosis of pervasive developmental disorder

  3. Unwillingness of parents and/or school personnel to meet methylphenidate treatment requirements

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.3 mg/kg methylphenidate and 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: Each new condition started on a Saturday morning (to allow parents’ observation/evaluation on weekend). Capsules given at 8:00 AM and 12:00 PM. Conners' administered at baseline and on the last day of each week. A 30‐minute semi structured follow‐up interview was conducted ≥ 12 months after completion of the trial

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: 12:00 PM to 8:00 AM the following day

Titration period: none initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Questionnaire: baseline and on the last day of each week

  2. Conners' Teacher Questionnaire: baseline and on the last day of each week

Non‐serious adverse events

  1. Weekly reporting of side effects by parents and teachers. No use of standard side effects questionnaire

Notes

Sample calculation: not stated

Ethics approval: yes; the Research Ethics Board of the IWK Grace Health Centre approved the protocol

Comments from study authors

  1. "We found the MPT to be helpful, practical, and definitive for families of children with attention‐deficit/hyperactivity disorder to making a decision about medication use"

  2. "Regardless of the outcome, it was important for families to complete the MPT to understand, for their own child, the effect of methylphenidate on the child’s behaviour and the presence of any side effects"

Key conclusion of study authors

  1. "An 'N of 1' MPT was easily performed and permitted families to decide whether to use methylphenidate for long‐term treatment of attention‐deficit disorder or attention‐deficit/hyperactivity disorder. Regardless of methylphenidate use or lack of use, the condition of all of these children was improved at follow‐up"

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated
Email correspondence with study authors: August 2013. We received additional information from study authors, but they were not able to provide the additional data that we requested

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Once enrolled in the MPT, the non‐blinded hospital pharmacist randomly assigned each child to a particular dosing schedule". "The capsules contained, in random order: placebo of the prescribed dose of methylphenidate (Ritalin) hydrochloride (0.3 mg/kg or 0.6 mg/kg)"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Families (n = 50) with a child eligible for MPT were given 3 bottles of identical capsules". "The family, teacher, and physician were blinded for the order of medication"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"At the end of each trial the code was broken. The physician evaluated this information and made a clinical inference about the degree of response each week"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

High risk

Not stated

Conflicts of interest: Study authors sponsored by Pharmaceutical Manufacturers’ Association of Canada Studentship

Klorman 1990

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 2

Participants

Number of participants screened: not clear

Number of participants included: 48. Participants were randomly assigned to 1 of 1 dose of methylphenidate and placebo

Number of participants followed up: appears to have been 48

Number of withdrawals: not clear

Diagnosis of ADHD: DSM‐III

Age: mean not stated (range 12 to 18 years)

IQ: mean 108.62

Sex: 42 boys, 6 girls

Methylphenidate naive: 46/48; 2 had brief trials in childhood

Ethnicity: Caucasian (n = 46)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional and conduct disorder (n = 24), oppositional not conduct disorder (n = 12), anxiety (n = 5), drug or alcohol abuse (n = 2), depression (n = 1)

Comedication: no

Sociodemographics: double‐ or single‐parent family, predominantly middle class (mean Hollingshead socioeconomic status score of 48.8 (i.e. social class II))

Inclusion criteria

  1. Participants 12 to 18 years of age without previous stimulant therapy referred for evaluation of response to stimulants from 1984 to 1988

Exclusion criteria

  1. CNS involvement

  2. Childhood autism

  3. Psychosis

  4. Uncorrected visual or auditory problems

  5. IQ < 80

Interventions

Participants were randomly assigned to 3 weeks of methylphenidate hydrochloride and placebo

Mean methylphenidate dosage: 35.21 mg (± 5.94 (SD)). Range 15 mg (2 daily doses) to 40 mg (3 daily doses)

Administration schedule: Doses were gradually increased at the end of the first and second weeks

Washout before study initiation: not described

Titration period: after randomisation

Treatment compliance: not described

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Hyperactivity Questionnaire: rated by teacher and parent, weekly

  2. IOWA Inattention/Overactivity and Aggression Scales: rated by teacher and parent, weekly

Mean parent‐ and teacher‐reported Conners' hyperactivity and inattention scores were graphed but SD values were not. Also, the paper did not refer to measures of impulsivity or total scores. We could not use these data in our meta‐analyses because values were missing and study authors were not able to provide supplemental data on this outcome

Non‐serious adverse events

  1. Side effects reported in Table 3: appetite loss; increased thirst, dry mouth, stomachaches, nausea, headaches, sleep problems, shakiness, crying, anger, unhappiness, sadness

Notes

Sample calculation: not described

Ethics approval: not described

Key conclusion of study authors

  1. These results support the continued effectiveness of stimulant therapy for attention deficit disorder in adolescence. However, the magnitude of clinical effectiveness reported was smaller than was previously found in younger patients

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April 2014. We obtained supplemental information/data from study authors (Magnusson 2014a [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Received information from study author (Magnusson 2014a [pers comm])

Allocation concealment (selection bias)

Low risk

Received information from study author (Magnusson 2014a [pers comm])

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Substances were dispensed in capsules of identical appearance" (p 703)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parent and teacher ratings were used and participants were blinded to which capsule they were receiving

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

National Institute of Mental Health (NIMH) grant MH38118

Conflicts of interest: no corporate affiliations declared

Kolko 1999

Methods

Randomised, placebo‐controlled, cross‐over trial with 2 possible drug interventions and placebo, as well as 2 possible psychological interventions

  1. Methylphenidate at 0.3 mg/kg

  2. Methylphenidate at 0.6 mg/kg

  3. Placebo

and

  1. Behaviour modification

  2. No behavioural modification

Participants

Number of participants screened: 70

Number of participants included: 22 (all boys). Participants were randomly assigned to different possible drug condition orders

Number of participants followed up: 16

Number of withdrawals: 6

Diagnosis of ADHD: DSM‐III‐R (subtypes not stated)

Age: mean 9.6 years (range 6.9 to 12.9)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: African American (75%)

Country: USA

Setting: "partial hospitalisation" summer treatment programme

Comorbidity: conduct disorder (44%), oppositional defiant disorder (56%), anxiety disorder (18.8%), major depressive disorder (11.5%), dysthymia (6%), intermittent explosive disorder (6%), developmental articulation disorder (6%), asthma (12.5%)

Comedication: not stated

Sociodemographics: Three lived with 1 or both parents, 6 lived with grandparents, 1 lived with an aunt, 4 lived with non‐relatives, 2 lived with foster mother. 44% of families received welfare

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.3 mg/kg and 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 8:00 AM and 11:30 AM to 12.00 PM

Duration of each medication condition: 1 day. Each medication condition was administered once per week for a total of 6 days during the trial

Washout before study initiation: 2 weeks

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: not stated

Behavioural intervention: behaviour modification and no behaviour modification were alternated on a weekly basis for a total of 3 weeks per condition

Outcomes

ADHD symptoms

  1. Abbreviated IOWA Conners' Rating Scale, which includes an Inattentive/Overactive subscale and an Oppositional Defiant subscale

Non‐serious adverse events

  1. Barkley Stimulant Side Effects Rating Scale. Adapted by changing the rating scale to include only 4 points rather than 9 points

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusions of study authors

  1. Methylphenidate and behaviour modification had certain unique, main and incremental effects that extend findings supporting their combination and suggest that integrated studies should evaluate multiple dimensions of functioning in novel settings

  2. Incorporation of other intervention components in combined treatments may be warranted to enhance clinical efficacy

Comments from review authors

  1. Barkley Stimulant Side Effects Rating Scale was adapted by changing the rating scale to include only 4 points (rather than 9 points) after pilot‐testing. Therefore, we cannot be sure whether the scale is still valid

  2. Limitations: The number of participants studied was small, and only 22 of 70 screened met study eligibility criteria

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study author: January 2014. We were unable to obtain additional data (Nilausen 2014 [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Each methylphenidate condition was administered once per week for a total of 6 days during the trial, and behaviour modification and no behaviour modification were alternated on a weekly basis, for a total of 3 weeks per condition. Thus, daily methylphenidate conditions were crossed with 2 weekly behavioural intervention conditions

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

It is stated: "MPH or placebo was placed in identical opaque capsules"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Not stated

Selection bias: exclusion of 2 participants with challenging behaviour

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: no information

Kollins 2006 (PATS)

Methods

Eight‐phase, 70‐week, multi‐centre trial (phase III study) including

  1. Screening: varying time

  2. Uncontrolled parent training: 10 weeks

  3. Baseline: 2 to 4 weeks

  4. Open‐label, safety lead‐in: 1 week

  5. Random‐sequence, double‐blind, placebo‐controlled, cross‐over titration: 5 weeks. Optional pharmacogenetics study simultaneously

  6. Randomised, optimal dose, double‐blind, placebo‐controlled, parallel trial: 4 weeks

  7. Open‐label, uncontrolled maintenance: 10 months

  8. Randomised, double‐blind, placebo discontinuation: 6 weeks

If parents requested and clinicians agreed that participants were severely symptomatic, children could be moved directly into the medication phase that was concurrent with parent training. If participants did not tolerate the dosing in phase 4, they could enter the open‐label maintenance phase if they tolerated lower doses (e.g. 1.25 mg, 2.5 mg). If they tolerated all doses except 7.5 mg, they were eligible to enter phase 5, the cross‐over titration, with the planned week on a 7.5‐mg dose replaced by an additional 5‐mg week. After phase 5, cross‐over: (1) If the child showed the greatest clinical benefit during 1 of the 5 weeks of the cross‐over trial, with no room for improvement, the blind was broken and the child was randomly assigned to that methylphenidate dose or placebo in phase 6; (2) if the child was a placebo responder, phase 6 was skipped and the child was allowed to enter phase 7 while taking no medication and with monthly monitoring by the treating physician; (3) if a particular week was deemed best, with ongoing room for improvement, a 2‐week double‐blind trial of 7.5 mg and 10 mg t.i.d., each for 1 week, was implemented, and teacher and parent ratings and side effects data were subsequently blindly reviewed to determine the best dose; or (4) participants with no clinical benefit any week were not eligible to continue in phase 6 or 7 but were given 1‐month follow‐up and then were referred for community treatment

Participants

Number of participants screened: 1915

Number of participants included in the study: 303 (229 boys, 74 girls)

Number of participants who completed the last phase of the study: 8

Number of withdrawals during the study: 295

Diagnosis of ADHD: DSM‐IV (combined (75%), hyperactive‐impulsive (25%), inattentive (0%))

Age: mean 4.41 years (range not reported)

IQ: > 70 (mean 99.06)

Methylphenidate naive: 100%

Ethnicity: Caucasian (63%), African American (19%), Asian (2%), Hispanic or Latino (16%), American Indian or Alaskan Native (0.7%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (52%), communication disorder (22%), elimination disorder (i.e. encopresis, enuresis; 8%), specific phobia (8%), anxiety disorder (8%), developmental co‐ordination disorder (3%), conduct disorder (2%), pica (2%), adjustment disorder (1%), reactive attachment disorder (1%), obsessive‐compulsive disorder (0.7%), sleepwalking disorder (0.3%)

Comedication: no

Sociodemographics: double‐parent family (76%), single‐parent family (18%), mean Hollingshead socioeconomic status 47.20 (SD 9.56)

Phase 4, open‐label, safety lead‐in

Number of participants included: 183

Numberof participants followed up: 169

Number of withdrawals: 12

Number of participants leaving the study phase and entering maintenance (phase 7): 2

Phase 5, cross‐over

Number of participants included: 165 (122 boys, 43 girls)

Number of participants followed up: 147

Number of withdrawals: 14

Number of participants leaving the study phase and entering maintenance (phase 7): 4
Diagnosis of ADHD: DSM‐IV (combined (76%), hyperactive‐impulsive (24%), inattentive (0%))

Age: mean 4.74 years (range 3 to 5.5)

IQ: > 70 (mean 97.93)

Methylphenidate naive: 100%

Ethnicity: Caucasian (63%), African American (18%), Asian (1%), Hispanic or Latino (18%), American Indian or Alaskan Native (0.6%)

Country: USA

Comorbidity: oppositional defiant disorder (55%), communication disorder (20%), elimination disorder (i.e. encopresis, enuresis; 8%), specific phobia (7%), anxiety disorder (10%), developmental co‐ordination disorder (4%), conduct disorder (3%), pica (2%), adjustment disorder (0.6%), reactive attachment disorder (2%), obsessive‐compulsive disorder (0.6%), sleepwalking disorder (0.6%)

Comedication: no

Sociodemographics: double‐parent family (79%), single‐parent family (21%), mean Hollingshead socioeconomic status 47.01 (SD 9.58)

Phase 6, parallel

Number of participants included: 114 (85 boys, 29 girls)

Number of participants randomly assigned: methylphenidate 61, placebo 53

Number of participants followed up: 77

Number of withdrawals: 1

Number of participants leaving the study phase and entering maintenance (phase 7): 36

Diagnosis of ADHD: DSM‐IV (combined (75%), hyperactive‐impulsive (25%), inattentive (0%))

Age: mean 4.76 years (range not reported)

IQ: > 70 (mean 97.45)

Methylphenidate naive: 100%

Ethnicity: Caucasian (65%), African American (17%), Asian (0.9%), Hispanic or Latino (17%), American Indian or Alaskan Native (0.9%)

Country: USA

Comorbidity: oppositional defiant disorder (53%), communication disorder (22%), elimination disorder (i.e. encopresis, enuresis; 7%), specific phobia (7%), anxiety disorder (11%), developmental co‐ordination disorder (5%), conduct disorder (3%), pica (0.9%), adjustment disorder (0.9%), reactive attachment disorder (2%), obsessive‐compulsive disorder (0.9%), sleepwalking disorder (0.9%)

Comedication: no

Sociodemographics: double‐parent family (80%), single‐parent family (19%), mean Hollingshead socioeconomic status 47.61 (SD 9.45)

Phase 7, open‐label maintenance

Number of participants included: 140 (104 boys, 36 girls)

Number of participants followed up: 95

Number of withdrawals: 45

Diagnosis of ADHD: DSM‐IV (combined (76.4%), hyperactive‐impulsive (23.6%), inattentive (0%))

Age: mean 4.4 years

IQ: > 70

Methylphenidate naive: 100%

Ethnicity: Caucasian (65.0%), African American (17.1%), Asian (1.4%), Hispanic (15.7%), American Indian (0.7%)

Country: USA

Comorbidity: oppositional defiant disorder (52.9%), communication disorder (19.3%), anxiety disorder (11.4%)

Comedication: no

Sociodemographics: double‐parent family (81.4%), single‐parent family (18.6%), mean Hollingshead socioeconomic status 47.2 (SD 9.5)

Inclusion criteria

  1. 36 to 65 months (3 to 5.5 years)

  2. DSM‐IV criteria for ADHD, hyperactive/impulsive or combined subtype, on Parent Diagnostic Interview Schedule for Children, Fourth Edition, and clinical interview by experienced clinician; symptoms were required to be present for a minimum of 9 months

  3. Age‐ and sex‐adjusted T score ≥ 65 on the Hyperactive‐Impulsive subscale of Conners' Parent and Teacher Rating Scales

  4. Score < 55 on the Children's Global Assessment of Functioning Scale

  5. IQ > 70 as on the Differential Abilities Scale; children scoring < 70 were considered for inclusion if their composite score from the Vineland Adaptive Behavior Scale was > 70

  6. Enrolled in some type of day programme: day care, pre‐school, nursery school, kindergarten, for ≥ 2 half‐days/wk. School‐type programme, in which class included ≥ 8 same‐age peers; if children had been expelled from an eligible programme in the 3 months before screening, they could be considered for enrolment

  7. Teachers willing to complete rating scale

  8. Residing with primary caretaker for ≥ 6 months before screening

  9. Patients and parents willing to attend all visits required by the study

  10. Otherwise generally healthy, and systolic and diastolic blood pressure below 95th percentile for age and sex

  11. Stimulant‐naive

Additional inclusion criteria for phase 4, open‐label lead‐in

  1. Not showing substantial ADHD improvement after parent training (phase 2) (continued impairment, operationalised as < 30% reduction on Conners' Parent Rating Scale or Conners' Teacher Rating Scale, or a rating of less than "improved" by at least 2 of the 3 raters (parent, teacher, clinician) completing the Clinical Global Impressions Scale

  2. Parental consent to a medication trial

Additional inclusion criteria for phase 5, cross‐over

  1. Tolerating the dosing in phase 4 (i.e. children with moderate to severe adverse events at doses < 5 mg in phase 4 were not eligible to continue)

Exclusion criteria

  1. Children or their parent(s) could not understand or follow instructions given in the study

  2. Evidence of moderate to severe adverse events or evidence of a much improved response to any dose of methylphenidate or another stimulant

  3. > 5 weeks of exposure to ≥ 30 mg/d of methylphenidate or equivalent doses of other stimulants

  4. Use of any other psychotropic medication or an investigational drug in the past 30 days; episodic use of sympathomimetic decongestants for the common cold under the study physician's supervision was allowed

  5. History of motor or vocal tics or Tourette's syndrome

  6. Major medical conditions that would interfere with involvement in a long‐term study or could be affected negatively by methylphenidate

  7. Current evidence of adjustment disorder, pervasive developmental disorders, autism, psychosis, significant suicidality or other psychiatric disorder, in addition to ADHD that requires treatment with additional medication

  8. Evidence of current physical, sexual or emotional abuse

  9. Living with anyone who currently abuses stimulants or cocaine

  10. History of bipolar disorder in both biological parents

‐‐‐‐‐‐‐

All cases were presented to a cross‐site panel of clinicians, and only patients for whom consensus indicated that all inclusion (and no exclusion) criteria were met could be enrolled

Interventions

Phases 1 to 3

Enrolment, parent training; baseline: no medical intervention

Phases 4 to 8

Medical intervention (short‐acting, immediate‐release methylphenidate)

Phase 4, open‐label, safety lead‐in

Titration: starting dose of 1.25 mg twice daily; increased to 7.5 mg 3 times daily

Duration: 1 week

Treatment compliance: not stated

Phase 5, cross‐over

Randomly assigned sequence of doses of 1.25 mg, 2.5 mg, 5.0 mg or 7.5 mg immediate‐release methylphenidate and placebo administered 3 times daily for a week

Medication‐free period between interventions: none

Mean methylphenidate dose: not stated

Duration: 5 weeks

Phase 6, parallel

After a 24‐hour medication washout, 4 weeks of randomly assigned treatment with a participant's optimal methylphenidate dose as determined in phase 5, or placebo

Mean methylphenidate dose: 14.22 mg/d, 0.7 mg/kg/d

Duration: 4 weeks

Treatment compliance: not stated

Phase 7, open‐label, maintenance

Maintenance starting doses were based on the best dose decision from cross‐over titration. Phase 5 placebo responders were maintained without medication for ≥ 4 weeks, unless their condition deteriorated, in which case open‐label treatment could be initiated. For any participant whose condition deteriorated, the methylphenidate dose was gradually titrated for optimal response. The dosing regimen was adjusted to minimise some adverse events to 3 times daily (at breakfast, around noon after lunch and at 3:30 PM), 7 days a week

Mean methylphenidate dose: increased from 14.04 mg/d (0.71 mg/kg/d) at month 1 to 19.94 mg/d (0.92 mg/kg/d) at month 10

Duration: 10 months

Treatment compliance: not stated

Phase 8, discontinuation

Randomised, double‐blind, placebo discontinuation trial, in which an abrupt medication replacement consisted of placebo for half of the children, while others continued on their best methylphenidate dose from the end of phase 7. Children returned to active medication if they met relapse criteria, in other words, Conners' Parent Rating Scale or Conners' Teacher Rating Scale scores on the Hyperactivity/Impulsivity Index > 1.5 SD above age‐ and sex‐adjusted norms, or a clinician rating of 4 on the Clinical Global Impressions ‐ Severity Scale

Duration: 6 weeks

Treatment compliance: not stated. Those who opted out of the double‐blind phases would be allowed to continue on open‐label maintenance therapy. This greatly reduced incentive for families to remain in the double‐blind phases, especially if there was reason to suspect that a child had been randomly assigned to placebo

Outcomes

ADHD symptoms

  1. Conners', Loney and Milich Rating Scale, and Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale: teacher‐ and parent‐rated weekly in phase 5

  2. Swanson, Nolan and Pelham Scales, Fourth Edition, average of parent and teacher ratings: at the end of the last week (fourth) of phase 6

  3. Conners' Parent Rating Scale, Conners' Teacher Rating Scale: rated by parents and teachers weekly in phase 8

General behaviour

  1. Strengths and Weaknesses of Attention‐Deficit/Hyperactivity Disorder Symptoms and Normal Behaviour Scale, Early Childhood Inventory: teacher‐ and parent‐rated at baseline and at the end of phase 6

  2. Child Behavior Checklist (home functioning)

  3. Hillside Behaviour Rating Scale (school functioning)

Quality of life

  1. Child Global Assessment Scale: completed only during non‐randomised phases

Serious adverse events

  1. Medication‐related serious adverse events

Non‐serious adverse events

  1. Height and weight were measured without shoes or heavy clothes at each study visit. (Growth charts provided by the Centers for Disease Control and Prevention (CDC) were used to transform absolute units of measurement into Z‐scores.) Laboratory tests were performed at each study visit

  2. General clinician inquiry regarding the child's health problems at each study visit. Adverse events monitored by telephone, and parent‐ and teacher‐rated in phase 4. Side Effects Rating Scale, parent‐rated, weekly in phases 5, 6 and 8; monthly in phase 7. Side Effects Rating Scale, teacher‐rated, weekly in phases 5 and 8, and in the last week of phase 6, as well as in first and tenth months of phase 7

  3. Safety related to early and continued medication treatment evaluated at 6‐year follow‐up

  4. Adverse Events Checklist was based on the Pittsburgh Side Effects Rating Scale. The 4‐point (none, mild, moderate or severe) teacher‐rated scale included the following: buccal‐lingual movements; picking at skin or finger; lip or cheek chewing; other abnormal motor movements; worried, anxious appearance; dull, tired, listless appearance; headaches; stomachaches; crabby, irritable behaviour; tearful, sad, depressed behaviour; appetite loss; prone to crying; and uninterested in others with social withdrawal. The Parent Adverse Events Checklist also included trouble sleeping. Only adverse events rated as moderate or severe were counted as reportable in the open lead‐in, titration and parallel phases of the study, whereas adverse events rated as mild, moderate or severe were reported from the maintenance phase

  5. Blood pressure and pulse, at each study visit. Cardiovascular adverse events were based on age‐adjusted normative values. Tachycardia was defined as 2 measurements of resting heart rate above 120 beats per minute (bpm) at the same visit. Hypertension was defined as 2 blood pressure readings at the same visit that were above the 95th percentile for age and sex (systolic or diastolic), ranging from 110/72 for 3‐year‐olds to 115/74 for 6‐year‐olds. Blood pressure was checked again within 7 to 14 days. If the reading remained above the cutoff limit, an adverse event for hypertension was reported. Hypertension was rated as mild if < 10 mmHg, moderate if 11 mmHg to 20 mmHg and severe if > 20 mmHg above the limit

Notes

Sample calculation: yes. Target sample size stated in the online protocol 165. Sample to be randomly assigned 120

Ethics approval: yes; by institutional review boards at each study site. The study was monitored by the Data and Safety Monitoring Board of the National Institute of Mental Health
Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors (limitations)

  1. Design: The study included only immediate‐release methylphenidate, not an extended‐release preparation. The study may have included an order effect in the cross‐over phase, particularly for children who were randomly assigned to receive higher doses first. The PATS protocol did not provide a stimulant‐untreated clinical control group in non‐controlled phases. Comparisons of height and weight before and after treatment were made against population norms (i.e. children without an ADHD diagnosis and methylphenidate‐exposure). The follow‐up period was not sufficient for evaluation of the critical issue of long‐term effects of initial growth suppression observed in the first year of treatment

  2. Dose: Doses used in the PATS were relatively low and homogeneous, and the high end of the proposed dose range was truncated. This may have masked dose‐related effects

  3. Population: The rigorous procedures for diagnosis of ADHD enhanced the validity of the diagnostic process at the expense of excluding some children likely to have met the conventional criteria for ADHD in other settings. Children who showed substantial ADHD improvement after parent training were not eligible for medication phases. The sample was too small for the medication to be declared safe for this age group. Failure to meet remission criteria may be caused by severity of ADHD symptoms, not by possible ineffectivity of methylphenidate in pre‐schoolers with ADHD. A high attrition rate and differential attrition rates in the allocated groups were possibly due to a delayed study start, repeated consent procedures, an always available option to skip directly into maintenance and greater nervousness among parents about medication side effects in pre‐schoolers. Parents’ experience during the titration phase presumably heightened their awareness of behavioural differences associated with active and placebo medication

  4. Data: missing data in general. Adverse event data entry procedures may have inflated adverse event rates. Parents were told when double‐blind switches between the drug took place. This may have contributed to confounding of negative expectancy and reporting of adverse events

  5. Exploratory moderator analyses of efficacy data from phase 5 (cross‐over): No adjustments were made for multiple comparisons. Findings should be considered preliminary. The Diagnostic Interview Schedule for Children, Fourth Edition, Parent Version, has not been validated in pre‐school children. Sample sizes across different moderator categories/subgroups were relatively small. Data on compliance were missing

Key conclusions of study authors

  1. Phase 5 (cross‐over): Immediate‐release methylphenidate, delivered in 2.5‐mg, 5‐mg and 7.5‐mg doses t.i.d., produced significant reductions in ADHD symptom scale scores in pre‐schoolers compared with placebo, although effect sizes (0.4 to 0.8) were smaller than those cited for school‐aged children taking the same medication

  2. Pharmacogenetics study in phase 5 (cross‐over): Emerging evidence suggests the potential for understanding individual variability of responses to and side effects of ADHD medications through the study of genetics, although additional research is required before these findings can be proven to have clinical utility

  3. Exploratory moderator analyses of efficacy data from phase 5 (cross‐over): Of the 14 variables examined as potential moderators, only 1 (number of concurrent comorbid disorders) served as a moderator of methylphenidate dose response. In pre‐schoolers with ADHD, the presence of no or 1 comorbid disorder (primarily oppositional defiant disorder) predicted a large treatment response at the same level as has been found in school‐aged children, and 2 comorbid disorders predicted moderate treatment response; whereas the presence of ≥ 3 comorbid disorders predicted no treatment response to methylphenidate

  4. Phase 6 (parallel): Medication effects varied by informant and outcome measure. Parent measures and teacher scores on the Strengths and Weaknesses of Attention‐Deficit/Hyperactivity Disorder Symptoms and Normal Behaviour Scale did not differentially improve with methylphenidate. Parent‐rated depression (P value < 0.02) and dysthymia (P value < 0.001) on the Early Childhood Inventory worsened with methylphenidate, but scores were not in the clinical range. Significant medication effects were found on the clinician Clinical Global Impressions ‐ Severity Scale (P value < 0.0001) and in teacher ratings on the Social Competence Scale (P value < 0.03). Pre‐schoolers with ADHD treated with methylphenidate for 4 weeks improve in some aspects of functioning. Additional improvements might require longer treatment, higher doses and/or intensive behavioural treatment in combination with medication

  5. Phase 7 (maintenance): With careful monitoring and a gradual medication dose increase, most pre‐schoolers with ADHD maintained improvement during long‐term immediate‐release methylphenidate treatment. Variability in effective and tolerated dosing was substantial

  6. Entire study duration

    1. Adverse events during the study: 11% of pre‐schoolers discontinued treatment because of intolerable methylphenidate adverse events. Of the serious adverse events reported, 1 occurred at baseline, 2 at lead‐in, 3 in titration, 1 in parallel and 1 in maintenance. Only 1 was possibly related to methylphenidate

    2. Growth during the study: Average relative size at baseline was significantly greater than 0 for z height and z weight, indicating greater than expected height (by 2.04 cm) and weight (by 1.78 kg) for participants. During treatment, slopes were significantly less than 0 for z height and z weight, indicating reduction in growth rates. For 95 children who remained on medication, annual growth rates were 20.3% less than expected for height (‐1.38 cm/y) and 55.2% less (‐1.32 kg/y) for weight. Risks of reduced growth rates should be balanced against expected benefits when pre‐school‐aged children are treated with stimulant medication

Withdrawals due to adverse events: number of withdrawals due to adverse events during medication phases: 21 (i.e. 11%)

Number of participants leaving study phase and entering maintenance (phase 7): 4
Email correspondence with study authors: June 2014. We obtained supplemental information/data from the study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

  1. Phase 5 (cross‐over): Randomisation was done centrally at the co‐ordinating site, using a computerised stratified randomisation, a 1:1:1:1 starting dose allocation ratio and a randomised, balanced, cross‐over protocol designed to avoid order effects

  2. Phase 6 (parallel‐group): A second randomisation to active methylphenidate or to placebo was performed before entry into the parallel‐design, placebo‐controlled phase

  3. Phase 8 (discontinuation): Centralized randomisation used a computer programme; each child was allocated 1:1 to continuing methylphenidate or switching to placebo under double‐blind conditions

Allocation concealment (selection bias)

Low risk

  1. Phase 5 (cross‐over): central randomisation using a computer programme

  2. Phase 6 (parallel‐group): central randomisation using a computer programme

  3. Phase 8 (discontinuation): central randomisation using a computer programme

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

  1. Phase 5 (cross‐over): double‐blind. Placebo pills were identical to pills containing active medication capsules

  2. Phase 6 (parallel‐group): double‐blind. Placebo pills were identical to pills containing active medication capsules

  3. Phase 8 (discontinuation): double‐blind. Placebo pills were identical to pills containing active medication capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

  1. Phase 5 (cross‐over): double‐blind. Except in emergencies, clinicians remained blind to dose sequences. Blinding was maintained for primary dependent measures until after the best dose was determined, or as needed. Parent and teacher dose‐response rating scale graphs were prepared and were blindly evaluated by 2 study clinicians

  2. Phase 6 (parallel‐group): double‐blind. Except in emergencies, clinicians remained blind to dose sequences. Blinding was maintained for primary dependent measures until after the best dose was determined, or as needed. Parent and teacher dose‐response rating scale

  3. Phase 8 (discontinuation): double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

  1. Phase 5 (cross‐over): All analyses were run using the ITT principle (i.e. each observation obtained for the child was used in the analysis, including those from children who entered each of the 2 phases under consideration and did not complete the phase)

  2. Phase 6 (parallel‐group): All analyses were run using the ITT principle (i.e. each observation obtained for the child was used in the analysis, including those from children who entered each of the 2 phases under consideration and did not complete the phase)

  3. Phase 8 (discontinuation): ITT

Selective reporting (reporting bias)

Low risk

  1. Phase 5 (cross‐over): outcome measures reported in accordance with the published protocol

  2. Phase 6 (parallel‐group): outcome measures reported in accordance with the published protocol

  3. Phase 8 (discontinuation): not relevant

Vested interest bias

High risk

  1. Phase 5 (cross‐over): sponsored by the National Institute of Mental Health, Columbia/New York State Psychiatric Institute, Johns Hopkins University, Columbia University, University of California Irvine, Duke University Medical Center, New York University Child Study Center and University of California Los Angeles, Arizona Institute of Mental Health Research to J.K.G. Generic methylphenidate was purchased by grant funds

  2. Phase 6 (parallel‐group): sponsored by the National Institute of Mental Health, Columbia/New York State Psychiatric Institute, Johns Hopkins University, Columbia University, University of California Irvine, Duke University Medical Center, New York University Child Study Center and University of California Los Angeles, Arizona Institute of Mental Health Research to J.K.G. Generic methylphenidate was purchased by grant funds.

  3. Phase 8 (discontinuation): sponsored by the National Institute of Mental Health, Columbia/New York State Psychiatric Institute, Johns Hopkins University, Columbia University, University of California Irvine, Duke University Medical Center, New York University Child Study Center and University of California Los Angeles, Arizona Institute of Mental Health Research to J.K.G. Generic methylphenidate was purchased by grant funds

Conflicts of interest

  1. Phase 5 (cross‐over): Multiple study authors had relationships with several pharmaceutical companies for the period 2000 to 2007

  2. Phase 6 (parallel‐group): Multiple study authors had relationships with several pharmaceutical companies for the period 2000 to 2007. Placebo responders in phase 5 were excluded from phase 6. Participants with no clinical benefit any week were excluded from phase 6 (methylphenidate non‐responders)

  3. Phase 8 (discontinuation): Multiple study authors had relationships with several pharmaceutical companies for the period 2000 to 2007

Konrad 2004

Methods

Double‐blind, placebo‐controlled, within‐participant trial of cross‐over design, lasting 6 days with 2 possible drug interventions and placebo

  1. Methylphenidate 0.25 mg/kg (low dose)

  2. Methylphenidate 0.5 mg/kg (high dose)

  3. Placebo

The order of drug conditions was randomly assigned, with the restriction that higher doses should never be administered after placebo

Participants

Number of participants screened: not stated

Number of participants included: 60 (44 boys, 16 girls)

Number of participants randomly assigned: low‐dose methylphenidate 60; high‐dose methylphenidate 60; placebo 60

Number of participants followed up: 60 in each arm

Number of withdrawals: not stated for each arm

Diagnosis of ADHD: DSM‐IV (combined 47 (78%), inattentive 13 (22%))

Age: mean 10.8 years (SD 1.6, range 8 to 12)

IQ: mean 97.4 (SD 10.7, range not stated)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Germany

Setting: out‐patient clinic and in‐patient ward

Comorbidity: oppositional defiant disorder (n = 6; 10%), conduct disorder (n = 18; 30%), anxiety (n = 12; 18%), dyslexia (n = 19; 32%)

Comedication: no

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Only children without a prior history of stimulant treatment were included in the study protocol

Exclusion criteria

  1. General IQ < 80 (Wechsler Intelligence Scale for Children, Third Edition)

  2. Any potentially confounding diagnoses such as psychosis, mania, major depression, substance abuse, pervasive developmental disorders, receptive language disorders

  3. Use of any kind of additional medication (including selective serotonin reuptake inhibitors or anticonvulsants)

Interventions

Participants were randomly assigned to "low‐dose" methylphenidate (0.25 mg/kg), "high‐dose" methylphenidate (0.5 mg/kg) and placebo

Mean methylphenidate dosage: 9.2 mg (SD 2.2) for low‐dose group (0.25 mg/kg), 18.4 mg (SD 5.4) for high‐dose group (0.5 mg/kg)

Administration schedule: Medication was given between 7:00 AM and 8:00 AM for 6 days. "Cognitive testing began 60 minutes after medication ingestion and lasted 80 minutes". The order of drug conditions was randomly assigned, with the restriction that higher doses should never be administered after placebo. Thus, 11 orders were possible for the 6‐day procedure as a whole, and 6 orders were possible for the sequence of the 3 neuropsychological assessments. Children were assigned in equal numbers to the 6 orders

Duration of intervention: 6‐day intervention

Titration period: 1 week of 0.3 mg/kg methylphenidate for each participant "to ascertain tolerance"

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Primary outcomes

    1. German Teacher’s and Parental Report on ADHD symptoms (Fremdbeurteilungsbogen für Hyperkinetische Störungen)

    2. Parental questionnaire on ADHD symptoms (Diagnostiksystem für Psychische Störungen im Kindes‐ und Jugendalter nach ICD‐10 und DSM‐IV)

    3. Child Behavior Checklist

  2. Specific attentional outcome measures

    1. Baseline speed: assessed with a simple reaction time task

    2. Sustained attention: involved the continuous and consecutive presentation of 50 series of 12 different dot patterns (600 signals)

    3. Focused attention: Four letters were presented simultaneously, and the child was instructed to respond with the "yes" key to 1 target letter, but only if this occurred in 1 of the relevant diagonal positions

    4. Divided attention: dual task that combined optic and acoustic discrimination tasks

    5. Stop‐Signal paradigm: The "go" task in our stop‐signal task was a choice reaction task in which an unidentified flying object (UFO) appeared to the left or right of a fixation cross

    6. Visual set‐shifting: Task consisted of 3 parts

Notes

Sample calculation: not stated

Ethics approval: yes; "the study was approved by the Medical Ethical Committee of the University Hospital of Aachen"

Comments from study authors

  1. The present study investigated effects of day‐to‐day medication on attentional functions, which might differ from dose‐dependent effects in the long run

  2. Our study did not include a third methylphenidate dose, which would have allowed additional study of dose‐response curves for higher doses of methylphenidate

Key conclusions of study authors

  1. Results indicate that attentional functions are influenced differentially by methylphenidate; intensity‐dimension functions are best influenced by higher doses, executive functions by moderate doses and selectivity‐dimension functions by variable doses

  2. Divergent results from behaviour rating scales and from attentional paradigms emphasise that clinicians have to decide what constitutes an appropriate clinical response

  3. A more comprehensive assessment of attention may help to reveal an individually optimal dose for the treatment of attentional dysfunction

Comment from review authors

  1. This study focuses on aspects of attention in relation to methylphenidate

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: unclear; only children without a prior history of stimulant treatment were included in the study protocol

Any withdrawals due to adverse events: not stated

Email correspondence with study authors: January 2014. We sent an email to the study author to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Within the 6‐day protocol, the order of drug conditions was randomly assigned, with the restriction that higher doses should never be administered after placebo

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"The active medication and placebo were prepared by a study protocol physician who was not involved in the assessment. All capsules were identical opaque gelatin capsules and were administered in a double‐blinded manner. Capsules containing placebo (lactose) or 0.25 mg/kg or 0.5 mg/kg doses of methylphenidate were prepared for each participant"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

As above

Incomplete outcome data (attrition bias)
All outcomes

High risk

"Due to computer problems, data for four children in one task in one condition were missing. As recommended by Tabachnik and Fidell (1996), these data were replaced by the average of the group per condition"

Selection bias: yes; before testing, all children were given 0.3 mg/kg methylphenidate each day for ≥ 1 week to ascertain tolerance

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

This research was funded by the German Society for the Advancement of Scientific Research (DFG grant KFO112)

Conflicts of interest: none declared

Konrad 2005

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate (low and high doses)

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 44 (37 boys (84%), 7 girls (16%))

Number of participants followed up: 44

Number of withdrawals: not stated
Diagnosis of ADHD: DSM‐IV (combined type 100%)

Age: mean 10.3 years (SD 1.9, range 8 to 12)

IQ: mean 98.1

Methylphenidate naive: not stated

Ethnicity: not stated

Country: Germany

Setting: in‐patient ward

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD diagnosis using multiple measures and observation in playroom, etc.

  2. Children were included only if they also met criteria for ADHD diagnosis on teachers’ rating scale

Exclusion criteria

  1. General IQ < 80 (Wechsler Intelligence Scale for Children, Third Edition)

  2. Any pervasive developmental disorders, receptive language disorders, visual impairments

  3. Any kind of additional medication (including selective serotonin reuptake inhibitors (SSRIs) or anticonvulsants)

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of 0.25 mg/kg (low dose) or 0.5 mg/kg (high dose) methylphenidate and placebo

Mean methylphenidate dosage: 9.4 mg (SD 2.3) for the 0.25‐mg/kg dose; 18.6 mg (SD 5.3) for the 0.50‐mg/kg dose

Administration schedule: not stated

Time points: not stated

Duration of each medication condition: 2 days

Washout before study initiation: not stated

Medication‐free period between interventions: no

Titration period: Before testing, all children were given 0.30 mg/kg methylphenidate each day for ≥ 1 week to ascertain tolerance

Treatment compliance: not stated. Within the 6‐day protocol, the order of drug conditions was randomly assigned, with the restriction that the high dose never occurred right after placebo

Outcomes

ADHD symptoms

  1. Primary outcomes

    1. German Teachers’ Report on ADHD Symptoms (Fremdbeurteilungsbogen für Hyperkinetische Störungen) of the Parental Questionnaire of ADHD symptoms (Diagnostiksystem für Psychische Störungen im Kindes und Jugendalter nach ICD‐10 und DSM‐IV). Sum scores were calculated separately for both symptom scales (hyperactive‐impulsive symptoms and inattentive symptoms)

Notes

Sample calculation: not stated

Ethics approval: yes; informed parental consent was obtained for all participants, and the study was approved by the Medical Ethical Committee of the University

Key conclusions of study authors

  1. Trend tests revealed linear effects of methylphenidate dose on actigraph data in the test session (P value = 0.02) and at school (P value = 0.001), as well as on sustained attention (P value < 0.001); inhibitory control showed a quadratic dose‐response curve (P value < 0.001)

  2. Multi‐variate regression analyses revealed that changes in both hyperactive‐impulsive symptoms (28%) and inattentive symptoms (23%) could be explained by objective changes in motor activity. Thus, for clinical practice, it should be taken into account that behaviour ratings of ADHD symptoms seemed to be predominantly influenced by changes in motor activity

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; before testing, all children were given 0.30 mg/kg methylphenidate each day for ≥ 1 week to ascertain tolerance

Any withdrawals due to adverse events: not stated

Email correspondence with study author: July 2015. Email sent to study author to ask for additional information, but we have received no reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Within the 6‐day protocol, the order of drug conditions was randomly assigned, with the restriction that the high dose never was given immediately after placebo

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Active medication and placebo were prepared by a study protocol physician who was not involved in the assessment. All capsules were identical opaque gelatin capsules and were administered in a double‐blind manner

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

Funding for this research was provided through a grant from the German Research Foundation (DFG grant: KFO112–TP5)

Conflicts of interest: none declared

Leddy 2009

Methods

Nine‐week, randomised, controlled trial (RCT), cross‐over design, with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 1

Participants

Number of participants screened: 154

Number of participants included: 58 (sex not stated). Participants were randomly assigned to 1 of 3 possible drug condition orders

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (combined (95%), hyperactive‐impulsive (2%), inattentive (3%))

Age: mean not stated (range 6 to 12 years)

IQ: > 80

Methylphenidate naive: 19%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: oppositional defiant disorder (52%), conduct disorder (10.5%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 12 years of age

  2. ADHD (DSM‐IV)

  3. IQ ≧ 80

  4. Symptoms positive for both Disruptive Behavior Disorders Rating Scale and Diagnostic Interview Schedule for Children

  5. Impairment in 2 settings (Impairment Rating Scale)

Exclusion criteria

  1. Seizures/serious neurological problem

  2. Pervasive developmental disorder, schizophrenia or other psychotic disorder

  3. Necessity of psychotropic medication for treatment of a comorbid disorder

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.15 mg/kg (t.i.d.) methylphenidate, 0.3 mg/kg (t.i.d.) methylphenidate, 0.6 mg/kg (t.i.d.) methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 3 time points

Duration of each medication condition: 12 days for placebo, methylphenidate 0.15 mg and methylphenidate 0.3 mg; 9 days for methylphenidate 0.6 mg

Washout before study initiation: not stated

Medication‐free period between interventions: no

Titration period: not stated

Treatment compliance: not stated

Outcomes

General behaviour

  1. Disruptive Behavior Disorders Rating Scale

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale

Notes

Sample calculation: not stated

Ethics approval: yes

Key conclusion of study authors

  1. "among children with ADHD, those with DA‐related genotypes associated with greater brain DA signalling, DAT SLC6A3 9/9, and DRD2 A2/A2, showed a greater suppression of lunch meal intake as MPH dose increased in comparison to children with DA genotypes associated with lower brain DA signaling"

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated

Any withdrawals due to adverse events: not stated

Email correspondence with study author: June 2014. Emailed the study author to ask for raw data regarding side effects, data from the Disruptive Behavior Disorders Rating Scale and other data. Study author responded to say he did not have them (Holmskov 2014 [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Drug dose varied daily on a randomized basis and included four conditions (...)"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Double‐blind"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Double‐blind"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

Conflicts of interest: Dr. Waxmonsky has served on the Speakers' Board for Novartis, received an honorarium from Shire and received research support from Shire and Eli Lilly. Dr. Erbe has received educational and research support from Genzyme Corporation. Dr. Pelham was paid an honorarium by Shire Pharmaceuticals

Lehmkuhl 2002

Methods

Four‐week, randomised, double‐blind, parallel trial with 2 arms

  1. Sustained‐release methylphenidate 20 mg to 60 mg

  2. Placebo

Participants

Number of participants screened: 102

Number of participants included: 85 (75 boys, 10 girls)

Number of participants randomly assigned: methylphenidate 43, placebo 42

Number of participants followed up: methylphenidate 40, placebo 38

Number of withdrawals: methylphenidate 3, placebo 4

Diagnosis of ADHD: DSM‐IV (combined (74.1%), hyperactive‐impulsive (0%), inattentive (24.7%), unspecified (1.2%))

Age: mean 9.8 years (range 6 to 15)

Mean IQ: methylphenidate 104.8, placebo 102.7 (range 85 to 146)

Methylphenidate naive: 25 (29.4%)

Ethnicity: German (82), other (3)

Country: Germany

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (51.8%), conduct disorder (9.4%), unspecified conduct disorder (3.5%), dysthymia (1.2%)

Comedication: yes (n = 4)

Sociodemographics: not stated. The 2 groups were homogeneous in terms of sex distribution, school type, school class and nationality (At baseline, SERS‐D ratings for disturbed sleep, nightmares, sadness, weepiness, anxiety, drowsiness and nervous twitching were far more pronounced in group 1 than in group 2. These initial differences – except for sadness – levelled out during the 4‐week trial period)

Additional conduct disorders are less frequent in the group treated with medication (25 vs 30 participants), but this slight difference is not significant (Fisher's exact test, P value = 0.26)

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD, verified against the ADHD Diagnostic Checklist from the Diagnostic System for Mental Disorders in Childhood and Adolescence, as per ICD‐10 and DSM‐IV

  2. According to the class teacher, occurrence of considerable ADHD symptoms over the previous 3 school days (provided the mean score on the aggregate Fremdbeurteilungsbogen für Hyper kinetische Störunge rating scale was > 1.0)

  3. Patients between 6 and 16 years of age

  4. Attendance at an elementary or secondary school

  5. IQ > 85

  6. Body weight > 20 kg

Exclusion criteria

  1. Diagnosis of depression or anxiety

  2. Tics or Tourette’s syndrome or family occurrence of tic disorder

  3. Pervasive developmental disorder

  4. Psychosis

  5. History of seizures or evidence on the EEG of risk of seizures

  6. Pre‐treatment of patients with methylphenidate or other psychostimulants up to 3 weeks before the study

  7. Lack of knowledge of the German language of the patient or legal guardian

Interventions

Participants were randomly assigned to sustained‐release methylphenidate or placebo

Mean methylphenidate dosage: not stated

Administration schedule: once daily after breakfast

Duration of intervention: 4 weeks

Titration period: weekly dose titration initiated after randomisation. Initially, two 5 mg methylphenidate/placebo tablets/d for 2 days, then 20 mg (1 sustained‐release methylphenidate capsule/placebo) dosage increased to 40 mg and 60 mg, depending on weight and course of symptoms. Titration up to 40 mg and 60 mg modified‐release methylphenidate was possible in the second and third weeks of treatment, respectively (20 kg to 30 kg, maximum 20 mg modified‐release methylphenidate; 31 kg to 50 kg, maximum 40 mg modified‐release methylphenidate; < 50 kg, maximum 60 mg modified‐release methylphenidate)

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Moderation of ADHD symptoms according to teacher rating on the basis of the Fremdbeurteilungsbogen für Hyperkinetische Störungen (aggregate rating scale), an ADHD response‐symptom checklist for observers based on ICD‐10 and DSM IV

  2. Fremdbeurteilungsbogen für Hyper kinetische Störungen (aggregate rating scale) rated by observers, parents and teachers: rated each week (teacher rated in the morning, parents in the afternoon)

  3. Conners’ abbreviated questionnaire, teacher and parent/guardian rated: rated each week (teacher rated in the morning, parents in the afternoon)

Serious adverse events

  1. One serious adverse event ‐ appendicitis ‐ occurred in the methylphenidate group. A relationship between the adverse event and the medication product is considered unlikely by the study authors

Non‐serious adverse events

  1. Adverse events, documented by the investigating physician each week

  2. Vital parameters, assessed each week

  3. Blood analyses, assessed each week

  4. EEG, performed at screening and at the end of the study ‐ no remarkable changes were found during the study

  5. Physical and neurological examinations, performed at screening and at the end of the study ‐ no remarkable changes were found during the study

  6. Barkley Side Effect Rating Scale‐D, rated by parents and participants, time point not stated

  7. Neurological psychiatric adverse events were classified as follows

    1. Neurological disorders (e.g. headaches, disturbed sleep)

    2. Psychiatric disorders (e.g. sadness, aggressiveness)

Notes

Sample calculation:yes

Ethics approval: approved by local university ethics committees

Comments from study authors

  1. Participant and parent observations differ to some extent from investigating physicians' documentation of adverse events. Differences in disturbed sleep, sadness and weepiness between the 2 medication groups during week 1 thus seem more likely to be the result of initial differences and homogeneities before the start of the trial, as they improve over the course of the trial under treatment with methylphenidate

  2. No clinically relevant changes on laboratory measures before and after the study

Key conclusions of study authors

  1. This trial demonstrated clinically relevant and statistically significant benefits of medication over placebo during 4‐week therapy

  2. Lehmkuhl 2002: In both the confirmatory analysis (change in teacher‐rated aggregate Fremdbeurteilungsbogen für Hyper kinetische Störungen rating scale) and all secondary hypotheses of efficacy, medication 1 (methylphenidate) always proved far more effective than medication 2 (placebo). This effect is clinically relevant. Under medication 1, however, adverse events were considerably more frequent and severe

  3. Sinzig 2007 (in Lehmkuhl 2002): Long‐acting methylphenidate is effective in the treatment of oppositional defiant disorder and aggressive behaviour, especially with milder symptoms. Expected correlation between impulsivity and aggressiveness could be confirmed

Comments from review authors

  1. Lemkuhl 2002 and Döpfner 2003 (in Lehmkuhl 2002): Only one review author, who knew German, has extracted data from these 2 articles. Two review authors assessed the other 3 articles and extracted data

  2. No one chose to withdraw, but because of administrative mistakes, comedication, etc., participants were taken out per protocol

  3. Unpublished data from this study were received from HB Pharma

Any withdrawals due to adverse events: yes; 2 (appendicitis and aggression)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April to May 2014. We emailed study authors twice but received no further information

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to methylphenidate or placebo by the following 4 strata: age (6 to 8 years; 9 to 11 years; 12 to 16 years), sex, severity of the problem according to the teachers’ evaluation (Fremdbeurteilungsbogen für Hyper kinetische Störungen; sum‐score > 40 and < 40) and study centre attended. Central randomisation was performed

Allocation concealment (selection bias)

Low risk

Central randomisation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study drug (or placebo) was dispensed in packages containing a weekly supply that was blinded from medical personnel and parents

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Study drug (or placebo) was dispensed in packages containing a weekly supply that was blinded from medical personnel and parents

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No incomplete outcome data

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

High risk

Financial support for this study was provided by Medice Arzneimittel Pütter GmbH & Co. KG, Kuhloweg 37, D‐58638 Iserlohn

Conflicts of interest: Dr. Doepfner is a consultant for Lilly, Medice, Novartis and Union Chimique Belge; serves on the Advisory Boards of Lilly, Medice, Shire, Novartis and Union Chimique Belge; participates as a member of the Speakers' Bureaus of Lilly, Medice, Janssen‐Cilag and Union Chimique Belge; and has research contracts with Lilly, Medice, Novartis, Union Chimique Belge, the German Research Foundation and the Federal Ministry of Health. Dr. Lehmkuhl is on the Advisory Boards of Lilly and Medice. Dr. Sinzig has no financial relationships to disclose

Lijffijt 2006

Methods

Three‐week, randomised, double‐blind, cross‐over, within‐participant study conducted to test methylphenidate/placebo to assess correlations between measures of attention and inhibition with dopamine and norepinephrine blood levels

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 15 (13 boys, 2 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 15

Number of withdrawals: 0
Diagnosis of ADHD: DSM‐IV (combined (11%), hyperactive‐impulsive (2%), inattentive (2%))

Age: mean 10.74 years (range 7 to 13)

IQ: mean 97.60

Methylphenidate naive: 0%

Ethnicity: not stated

Country: the Netherlands

Setting: out‐patient clinic

Comorbidity: anxiety (n = 6), oppositional defiant disorder (n = 5)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD diagnosis according to DSM‐IV

  2. Participants familiar with intake of methylphenidate for at least a year

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to different possible drug condition orders of 0.5 mg/kg or 1.0 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: 22.67 mg

Administration schedule: not stated

Duration of each medication condition: 1 day

Washout before study initiation: 24 hours before testing

Medication‐free period between interventions: no

Titration period: none/duration

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale

  2. Conners' Teacher Rating Scale

General behaviour

  1. Child Behaviour Checklist; Teachers' Report Form

Non‐serious adverse events

  1. Paper mentioned only this: "Although side effects were minimal (a feeling of sleepiness), four participants were too fatigued after placebo or the 1.0 mg/kg dose to continue with the change task after they first completed the stop task"

Notes

Sample calculation: no

Ethics approval: yes; "The study was approved by the national medical ethical committee (CCMO)"

Key conclusion of study authors

  1. In children with ADHD, methylphenidate could act primarily on inhibitory control and is not influenced by task difficulty

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: "four participants were too fatigued after placebo or the 1.0 mg/kg dose to continue with the change task after they first completed the stop task"
Email correspondence with study authors: March 2014: We sent an email to the study author to request additional information but have received no reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs. "All participants were familiar with the intake of MPH for at least 1 year"

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: none declared

Lin 2014

Methods

Eight‐week, multi‐centre (31 sites in 5 countries), double‐blind, placebo‐controlled, comparator (MPH‐OROS), parallel study with 3 interventions

  1. Edivoxetine

  2. OROS methylphenidate

  3. Placebo

Phases

  1. Screening

  2. Clinical treatment

  3. Discontinuation

Participants

Number of participants screened: 448

Number of participants included: 340 (70.6% boys, 29.4% girls). Participants were randomly assigned to 1 of 5 possible drug condition orders

Number of participants followed up: 210

Number of withdrawals: 60
Diagnosis of ADHD: DSM‐IV‐TR (combined (70.9%), hyperactive‐impulsive (4.1%), inattentive (25%))

Age: mean 11.6 years (range 6 to 17)

IQ: not stated

Methylphenidate naive: All participants treated with methylphenidate were medication‐naive. 44% of placebo‐treated participants, 47% of edivoxetine‐treated participants in the 0.1 mg/kg/d arm and 49% of edivoxetine‐treated participants in each of the 0.2 mg/kg/d and 0.3 mg/kg/d arms had used stimulants previously

Ethnicity: Caucasian (72.6%), African American (not stated), Asian (not stated), Hispanic (not stated), other (not stated)

Country: USA, Canada, Taiwan, Mexico and Puerto Rico

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder ("less than 20%")

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD DSM‐IV‐TR diagnosis

  2. > 6 years and < 17 years and 9 months of age at the time of informed consent

  3. Diagnosis confirmed with the Kiddie Schedule for Affective Disorders and Schizophrenia, Present and Lifetime Versions, and ADHD Rating Scale, Fourth Edition, score > 1.5 SD above age/sex norms and Clinical Global Impressions ‐ ADHD ‐ Severity Scale score > 4

Exclusion criteria

  1. Body weight < 18 kg or > 75 kg

  2. History of bipolar I or II disorder, or psychosis, seizure disorder or pervasive developmental disorder; motor tics or a diagnosis of Tourette's syndrome; marked anxiety, tension or agitation sufficient to contraindicate treatment with OROS methylphenidate

  3. History of electroencephalographic abnormalities

  4. Clinically significant abnormal ECG

  5. Serious or unstable medical illness

  6. Any medical condition that would markedly increase sympathetic nervous system activity (e.g. catecholamine‐secreting neural tumour)

  7. Requiring daily use of medications with sympathomimetic activity (e.g. albuterol, pseudoephedrine)

  8. Any medical condition that would be exacerbated by an increase in norepinephrine tone

  9. Current or past history of clinically significant hypertension

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of 18 mg, 36 mg or 54 mg OROS methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 1 a day

Duration of each medication condition: 8 weeks

Washout before study initiation: all methylphenidate naive

Medication‐free period between interventions: no

Titration period: none, initiated after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition, parent‐rated, Investigator‐administered and ‐scored: weekly, parent‐rated (administered and scored by qualified personnel at the investigative site based on an interview with the parent and the participant)

  2. Clinical Global Impressions ‐ ADHD ‐ Improvement Scale

  3. Clinical Global Impressions ‐ ADHD ‐ Severity Scale

  4. Swanson, Nolan and Pelham Scale, Fourth Edition

  5. Conners' Comprehensive Behavior Rating Scales

Non‐serious adverse events

  1. Adverse events, vital signs, clinical laboratory tests (e.g. chemistry, haematology, urinalysis), physical examination and ECGs

  2. Columbia Suicide Severity Rating Scale: occurrence, severity and frequency of suicide‐related thoughts and behaviours

Notes

Sample calculation: not stated

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes (3 participants)

Email correspondence with study authors: April 2015. We emailed study authors to ask for supplemental information/data but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Interactive voice response system was used for randomisation and to determine which study drug should be dispensed

Allocation concealment (selection bias)

Unclear risk

Not stated

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

High risk

Exclusion of methylphenidate non‐responders (after randomisation)

Selective reporting (reporting bias)

High risk

Yes

Vested interest bias

High risk

Study sponsored by Ely Lilly

Conflicts of interest: Five authors work for Lilly

Lopez 2003

Methods

Cross‐over trial with 3 interventions

  1. Methylphenidate (Ritalin)

  2. Concerta

  3. Placebo

Phases: 4

  1. Placebo

  2. Concerta 18 mg

  3. Concerta 38 mg

  4. Methylphenidate (Ritalin LA) 20 mg

Evaluated on day 0, randomly assigned to drug condition on days 7, 14, 21 and 28. One practice visit for a study duration of 5 weeks in total

Participants

Number of participants screened: not stated

Number of participants included: 36 (29 boys, 7 girls). Participants were randomly assigned to 1 of 24 (4 × 3 × 2 × 1) possible drug condition orders

Number of participants followed up: 36

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9 years (range 6 to 12)

IQ: not stated

Methylphenidate‐naive: no

Ethnicity: Caucasian (36%), African American (27%), Hispanic or other (36%)

Country: USA

Setting: out‐patient clinic

Co‐morbidity: not stated

Co‐medication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Met ADHD criteria based on Diagnostic Interview Schedule for Children

  2. Parents consented to participation

Exclusion criteria

  1. Concurrent significant medical or psychiatric illness or substance use disorder

Interventions

Participants were randomly assigned to 1 of 24 possible drug condition orders of methylphenidate (20 mg), (18 mg) Concerta, (36 mg) Concerta and placebo

Mean methylphenidate dosage: not stated

Administration schedule: not stated

Duration of each medication condition: 1 day

Washout before study initiation: not stated

Medication‐free period between interventions: On the morning following each study period, participants resumed their regularly prescribed medication up to Thursday evening before the next study period day on Saturday

Titration period: All participants had been stabilised on an equivalent dose of 10 mg twice daily of methylphenidate before study entry

Treatment compliance: No participants discontinued the study prematurely

Outcomes

ADHD symptoms

  1. Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: observer, during each study period

Non‐serious adverse events

  1. Physical exam, vital signs, haematology, blood chemistries, urinalysis ‐ screening

  2. Adverse events: self reported, each study period

  3. Vital signs: observer, measured every 2 hours at each study period

  4. Adverse effects did occur in < 3% of participants exposed to each agent and dose, and 1 participant from each treatment group experienced a single mild adverse event that included abdominal pain, nausea and dyspnoea

Notes

Sample calculation: yes

Ethics approval: yes

Comment from study authors

  1. Although single blinding of raters added to the objectivity of the observations, lack of medication blinding had both negative and positive implications. On the negative side, participants may have noticed the difference in the appearance of agents administered to them, thus producing bias …[...]... another issue to contemplate is that all participants in the study had previously been stabilised on methylphenidate and, irrespective of blinding, may well have been able to identify when they were receiving placebo

Key conclusion of study authors

  1. Although both methylphenidate (Ritalin LA) and Concerta were shown to be effective, the different release profile for each formulation can result in distinct differences between effects on measures of attention and deportment

Comment from review authors

  1. Not able to use reported data

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April 2014. We received supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Order of medication assignment was determined by random assignment by a computer programme

Allocation concealment (selection bias)

Unclear risk

Participants were randomly assigned to 4 treatment periods. For purposes of this study, with the exception of the medicating nurse, all study personnel were blinded to the medication administered to the child

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Single‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

For purposes of this study, with the exception of the medicating nurse, all study personnel were blinded to the medication administered to the child

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None reported. No participants discontinued the study prematurely

Selection bias: no, but all patients were stabilised previously on methylphenidate

Selective reporting (reporting bias)

Unclear risk

Protocol not identified

Vested interest bias

High risk

The study was funded by Novartis

Conflicts of interest: Dr. Silva is a consultant and a member of the Speakers' Bureau for Novartis. Dr. Lopez is a consultant for Eli Lilly, Novartis and Shire. He is also a member of the Speakers' Bureaus for Novartis and Shire

Lufi 1997

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: Participants randomly assigned to 3 weeks of methylphenidate, 3 weeks of placebo. No washout. Assessment at baseline and by the end of each phase

Participants

Number of participants screened: not stated

Number of participants included: 20 (18 boys, 2 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 20

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.23 years (± 1.62, range 7.17 to 12.42)

IQ: > 70

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Israel

Setting: out‐patient clinic

Comorbidity: none

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. IQ > 70

  2. Treatment‐naive

Exclusion criteria

  1. Implicitly stated: gross physical impairment, intellectual deficits, major disease or serious psychological problems, and not receiving any psychological treatment prior to taking part in the research

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 10 mg methylphenidate and placebo

Mean methylphenidate dosage: 10 mg/d

Administration schedule: mornings

Duration of each medication condition: 3 weeks

Washout before study initiation: All participants were treatment‐naive

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners’ Teacher Rating Scale

General behaviour

  1. Global Teacher Ratings: rating scale from 1 to 10 constructed specifically for this study (assessment before ingestion of medication, after 3 weeks of first medication period, after 3 weeks of second medication period)

Notes

Sample calculation: not stated

Ethics approval: not stated

Comment from study authors

  1. Strong placebo effect

Key conclusions of study authors

  1. Methylphenidate improved classroom behaviour as compared with no treatment

  2. Placebo influence had almost the same effect as medication

  3. Neither of these treatments significantly improved cognitive functioning and personality characteristics of the child with ADHD

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: September 2013. We obtained supplemental information/data from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"everyone involved in the assessment phase ...was blind to the type of medication..."
All medications (both placebo and methylphenidate) were given in identical capsules to prevent recognition of the true medication

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"...the parents, the teacher, the child and the psychologist who tested the child did not know what kind of medication the child was taking..."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Unclear risk

No information

Lufi 2007

Methods

Six‐week, randomised, double‐blind, placebo‐controlled, cross‐over study conducted to detect the effects of methylphenidate on co‐ordination and handwriting

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 19

Number of participants included: 19 (12 boys, 7 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 19

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.51 years (range 7.08 to 13.83)

IQ: > 70

Methylphenidate naive: 0%

Ethnicity: not stated

Country: Israel

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

Sociodemographics: "Participants were from a medium level social‐economic status"

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 0.4 mg/kg

Administration schedule: not stated

Duration of each medication condition: 3 weeks

Washout before study initiation: no

Medication‐free period between interventions: yes

Titration period: not stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners’ Abbreviated Symptom Questionnaire (teacher): assessed at baseline, weeks 3 and 6

General behaviour

  1. Achenbach’s Teacher Report: assessed at baseline, weeks 3 and 6

Notes

Sample calculation: not stated

Ethics approval: not stated

Comments from study authors (limitations)

  1. Limited group of participants

  2. Use of only 1 dosage level

Key conclusions of study authors

  1. Results show that methylphenidate improved some cognitive functions of eye‐hand co‐ordination slightly better than placebo

  2. Behaviour variables assessed by teachers improved only under the influence of methylphenidate

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no drop‐outs

Email correspondence with study authors: October 2013. We obtained additional information on IQ from the first study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomized"

Allocation concealment (selection bias)

Unclear risk

"The study was designed as a double‐blind, randomized, crossover, placebo‐control procedure"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Each participant was given exactly 0.4 mg/kg of MPH in special capsules of MPH and a placebo to avoid recognition of the medication"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: not stated

Manos 1999

Methods

Four‐week, double‐blind titration, placebo‐controlled protocol. Cross‐over trial with 2 interventions in 4 doses

  1. Methylphenidate 5 mg × 2 daily

  2. Methylphenidate 10 mg × 2 daily

  3. Methylphenidate 15 mg × 2 daily

  4. Placebo × 2 daily

Phases

  1. Cross‐over trial with Adderall and methylphenidate. First phase was not blinded. (The child’s paediatrician or family physician determined whether methylphenidate or Adderall should be prescribed; criteria were the physician’s familiarity with the agent and whether he or she wanted the participant to receive a single dose (Adderall) or a twice‐daily dose (methylphenidate) of medication treatment for ADHD.) Second phase: A 1‐of‐3 medication dose sequence was randomly assigned. 15 mg methylphenidate was always given after 10 mg methylphenidate

  2. 42 of the 117 participants receiving methylphenidate were matched, with 42 receiving Adderall

  3. Best dose was compared with placebo. Best dose was assigned by consensus of a clinical child psychologist and a board‐certified child and adolescent psychiatrist before the medication blind was broken

Manos 1999: Seven youths given methylphenidate and 4 given Adderall did not receive the 15 mg dose of medication. The decision to forego the 15 mg condition was based on the paediatrician’s assessment that the child was too young or underweight for this high dose and our assessment that the best dose had already been achieved at a lower dose

Participants

Number of participants screened: 195

Number of participants included: 177. Participants were randomly assigned to different possible drug condition orders

Number of participants followed up: 134

Number of withdrawals: 43

Diagnosis of ADHD: DSM‐IV (combined (55%), inattentive (45%))

Age: mean 10.1 years (SD not stated, range 5 to 17)

IQ: > 70

Sex: 33 boys, 9 girls

Methylphenidate naive: not stated

Ethnicity: Caucasian (93%), African American (7%), Asian (0%), Hispanic (0%), others (0%)

Country: USA

Setting: out‐patient clinic

Comorbidity: no significant comorbid disorders. Although no formal comorbidity data are available, it appears that psychiatric comorbidity was modest in this cohort

Comedication: not stated

Sociodemographics: predominantly well educated

Manos 1999 and Faraone 2002: 42 were participants matched to the Adderall group in order of diagnostic category, age and sex. Only these 42 of 117 receiving methylphenidate were compared with the Adderall group

Findling 2001a: 195 youths entered the study. Data for a best dose were provided for 177 participants: 111 in the methylphenidate group, 66 in the mixed amphetamine salts group

Diagnosis of ADHD: inattentive (47%), combined (53%). Inattentive subtype is overrepresented in the older age group

Age group: 4 to 8 years (mean 6.35) 69 (57 boys); 8 to 11 years (mean 9.47) 56 (45 boys); 11 to 17.59 years (mean 13.64) 52 (41 boys)

Sociodemographics: No significant differences in baseline demographics were noted between groups

Findling 2001b

Number of participants included: 195

Number of participants completed: 137: methylphenidate 82, Adderall 55

Diagnosis of ADHD: DSM‐IV (combined (57%), inattentive (43%))

Age: mean 10 years (SD not stated, range 4 to 17)

IQ: > 70

Sex: 66 boys, 16 girls

Ethnicity: Caucasian (84%), African American (6%), other (10%)

Country: USA

Comorbidity: without significant comorbid disorders

Comedication: possible, but not recorded

Sociodemographics: predominantly well educated. No differences in sex, ethnicity or ADHD subtype were found between methylphenidate and Adderall groups. No participants had a history of hypertension, hypotension or clinically significant cardiovascular disease

Inclusion criteria

  1. All children diagnosed with ADHD met full DSM‐IV diagnostic criteria for this disorder. Criteria include (1) presence of ≥ 6 symptoms for inattention and/or ≥ 6 symptoms for hyperactivity/impulsivity; (2) symptoms significantly interfering with functioning at home and at school as noted during structured or semi structured clinical interviews with the Computerized Diagnostic Interview for Children; (3) symptom severity on broad‐band (i.e. Conners’ Abbreviated Symptoms Questionnaire (Conners, 1969)) and narrow‐band (e.g. ADHD Rating Scale (DuPaul, 1991)) rating scales at threshold or above (i.e. rated 2 or 3); (4) multiple raters (e.g. parents, teachers) who agreed to the presence of the symptoms; and (5) empirical comparison with norms indicating ≥ 1/5 SD cutoff on ≥ 1 rating scale. It should be noted that in identifying the presence of symptoms, behaviours across informants were not pooled observations. Behaviours were considered significant only if 2 informants agreed to the presence of the symptom on rating scales or in interviews

Exclusion criteria

  1. No patients were excluded from the trial per se

Interventions

Participants were randomly assigned to different orders of 5 mg, 10 mg or 15 mg methylphenidate and placebo

Mean methylphenidate dosage: best dose 9.1 mg to 10.4 mg

Administration schedule: morning (at 8.00 AM) and noon

Duration of each medication condition: 1 week

Washout before study initiation: none

Titration period: none

Treatment compliance: 11 terminated because of adverse events

Outcomes

ADHD symptoms

Findling 2001: compared treatment for children and adolescents and weight‐adjusted dosing of methylphenidate. Measurement instruments include the following

  1. ADHD Rating Scale

  2. Abbreviated Symptoms Questionnaire (Conners)

  3. Composite Rating and School Situations Questionnaire Revised, parent and teacher. Composite Rating, also observer‐rated. Rating every seventh day of each week’s dose and baseline. Best dose evaluated and compared with placebo and Adderall

Best dose based only on Abbreviated Symptoms Questionnaire ‐ Teacher. Does not separate methylphenidate and mixed amphetamine salts in tables

Non‐serious adverse events

  1. Side Effects Behavior Monitoring Scale by parents every week (symptoms were considered problematic if parents rated them as ≥ 5)

  2. Blood pressure and pulse every week

Findling 2001b

  1. Blood pressure and pulse

Notes

Sample calculation: Not stated

Ethics approval: Not stated

Key conclusions of study authors

  1. Manos 1999: Both methylphenidate and Adderall have been shown to be effective treatments for children with ADHD

  2. Farone 2002: The present report extends this prior work by applying drug–placebo response curve methods to the data reported by Manos et al. (1999). Results show that the efficacy of Adderall and methylphenidate in improving functioning is seen throughout the full range of improvement scores. Both drugs prevent worsening and, for most patients, lead to improvements that are well into the normal range

  3. Findling 2001a: Data suggest that psychostimulants are equally effective in treating children and adolescents with ADHD. Adolescents with ADHD may not necessarily require more medication than younger children to achieve a similar therapeutic response

  4. Findling 2001b: Short‐term cardiovascular effects of both Adderall and methylphenidate were modest. No participants experienced any clinically significant change in these cardiovascular measures during the course of this brief trial

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; 15 youths in the Adderall sample had been tried on methylphenidate before enrolment in this medication trial. Because of lack of response or serious side effects, these children discontinued use of methylphenidate. A total of 37% of the Adderall sample subsequently was composed of children who had unsuccessfully used methylphenidate but successfully responded to Adderall

Any withdrawals due to adverse events: yes; Findling 2001a (11/195), due to multiple adverse events

Email correspondence with study authors: April 2014. Emailed study authors twice to request additional data but have received no answer

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

First phase not blinded. Methylphenidate is a selected group

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Clinician, teacher and parent were blinded only to dose, not to medication

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Clinician, teacher and parent were blinded only to dose, not to medication

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Only best dose compared with placebo. Findling (Abbreviated Symptoms Questionnaire) + 30 participants. Of 43 participants with < 4 weeks of data, 30 had only 3 weeks of data because physicians considered them too young or too small to receive 15 mg before initiating protocol

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

Study was supported in part by funding from Shire Pharmaceutical Development Incorporated to Dr. Faraone

Conflicts of interest: Study authors acknowledge partial support to the second author from the National Institute on Drug Abuse (NIDA) (grants R01‐DA07957 and MCJ‐390592) and from the Maternal and Child Health Program, Health Resources and Service Administration, Department of Health and Human Services (grant 390715), and to the third author from the Stanley Foundation

Martins 2004

Methods

Four‐week‐long, double‐blind, randomised, parallel trial with 2 interventions

  1. Methylphenidate group: methylphenidate, 7 days a week

  2. Placebo group: methylphenidate Monday through Friday, and placebo on weekends

Participants

Number of participants screened: not stated

Number of participants included: 40 (all boys)

Number of participants randomly assigned: methylphenidate 21, placebo 19

Number of participants followed up: methylphenidate 19, placebo 19

Number of withdrawals: methylphenidate 2, placebo 0
Diagnosis of ADHD: DSM‐IV (combined (92.5%))

Age: mean methylphenidate 9 years (SD 2.2, range 6 to 14), placebo 9.6 years (SD 2.8, range 6 to 14)

IQ: mean, methylphenidate 97.3, placebo 93.5

Methylphenidate naive: not stated

Ethnicity: European‐Brazilian: methylphenidate 16 (76.2%), placebo 15 (78.9%)

Country: Brazil

Setting: out‐patient clinic

Comorbidity: yes; conduct or oppositional defiant disorder (methylphenidate 57.2%, placebo 57.9%)

Comedication: not stated, no psychiatric medication

Sociodemographics: Monthly family income was calculated according to the following formula: total monthly income received by all members of the family (expressed in number of minimum wages) divided by the number of persons in the family. A value lower than 0.7 (approximately US$ = 68 per family member per month) is usually an indicator of poverty in Brazil. Methylphenidate 3.3, placebo 2.4. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. ADHD diagnosis according to DSM‐IV criteria

  2. Between 6 and 14 years of age

  3. Male

  4. Education level between first and eighth elementary grades

Exclusion criteria

  1. Presence of significant neurological or clinical disease

  2. Presence of bipolar disorder or substance abuse/dependence disorder

  3. Use of any psychiatric medication in the past 6 months, including methylphenidate

  4. Estimated IQ < 70

Interventions

Participants were randomly assigned to methylphenidate 7 days a week or to methylphenidate on weekdays and placebo on weekends

Mean methylphenidate dosage: Initial dose of methylphenidate was 0.3 mg/kg/d the first week. Dose was raised to 0.5 mg/kg/d the second week and to 0.70 mg/kg/d the third and fourth weeks

Administration schedule: twice a day: breakfast and lunch

Duration of intervention: 4 weeks

Titration period: none

Treatment compliance: Only 7 of 160 blister packs were returned with unused pills

Outcomes

ADHD symptoms

  1. 10‐item Conners’ Abbreviated Rating Scale (Conners 1985). Rated every Monday after school by both teachers and parents

Serious adverse events

  1. Barkley Side Effect Rating Scale

    1. Number of adverse effects reported

    2. Mean severity of reported adverse effects

(Completed only by parents for assessment of side effects on weekends)

Notes

Sample calculation: no

Ethics approval: approved by the Ethical Committee of the Hospital de Clínicas de Porto Alegre (HCPA) (approved as an International Review Board (IRB) by the Office for Human Research Protections, United States of America)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Comments from study authors

  1. Context established by the home setting on the weekend may have created conditions in which the effects of methylphenidate were minimal or insignificant, because children may have been involved in play activities much of the time

  2. It is reasonable to suggest that during the weekend, when networks related to ADHD neurobiology might be demanded less often, differences between drug and placebo would be more difficult to detect

  3. It is possible that parental tolerance of ADHD symptoms on weekends might be greater than on weekdays. Our findings may not be generalisable to females

Key conclusion of study authors

  1. Our findings suggest that weekend holidays during methylphenidate administration reduce the side effects of insomnia and appetite suppression without causing a significant increase in ADHD symptoms, on weekends or on the first day of school after the weekend (Monday)

Email correspondence with study authors: We have contacted study authors several times to ask for additional information about data, but we have received no data from this study that we can use

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised, double‐blind, parallel‐group design was used. Participants were randomly assigned to 1 of 2 groups, according to a computer‐derived algorithm (EPIINFO.06)

Allocation concealment (selection bias)

Low risk

Computer‐derived algorithm

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Methylphenidate and placebo pills were of the same shape and colour

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind. No other information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Measured adherence to protocol by assessing returned, unopened blister packs. None of the findings in the analyses was significantly affected by the 2 participants (they did not follow the protocol as stated by researchers) from the methylphenidate group who did not receive a few doses appropriately on weekends or on Monday. Some teacher ratings (8.5%) were missed because of the child’s absence from school on a specific day of evaluation or because of a school holiday

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No reporting bias

Vested interest bias

Unclear risk

Methylphenidate and placebo pills were supplied by Novartis Pharmaceuticals (São Paulo, Brazil) at no cost and without restrictions. No additional funding was requested or received from Novartis or any other commercial entity

Conflicts of interest: Study authors have reported no conflicts of interest

McBride 1988a

Methods

Individual, double‐blind, cross‐over trial for 4 weeks with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 73. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 70 (53 boys, 17 girls)

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐III (77% met the criteria for ADD with hyperactivity)

Mean age: methylphenidate responders 8.5 (SD not stated), methylphenidate non‐responders 9.5 (SD not stated). Range 6 to 17 years

Mean IQ: methylphenidate responders 102 (SD 21), methylphenidate non‐responders 89 (SD 23)

Methylphenidate naive: 71 (97%)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: carbamazepine or phenytoin or valproic acid or mephobarbital (6.4 receiving a combination of these drugs). Clonidine (n = 1)

Sociodemographics: no information

Inclusion criteria

  1. Referred because the child’s academic performance was below that expected on the basis of his abilities, as documented by psychological testing, or because his behavioural dysfunction was interfering with self‐image and socialisation, or for both reasons

  2. 6 to 17 years of age

  3. No child was excluded from the trial on the basis of low intelligence, history of seizures or concurrent medication

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.3 mg/kg methylphenidate and placebo. Methylphenidate was rounded to the nearest 1.25 mg

Mean methylphenidate dosage: no information, but mean dose during follow‐up was 0.36 mg/kg/dose

Administration schedule: morning and 4 hours later

Duration of each medication condition: 2 weeks

Washout before study initiation: none

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Teacher and Parent Questionnaire

  2. Parent Questionnaires was rated at the end of each weekend and at the end of each school week. Teacher Questionnaires were filled out at the end of the week

Serious adverse events

  1. No serious side effects during the trial

Non‐serious adverse events

  1. No information about how data on side effects were obtained

None of the parents of responders who had experienced side effects during the trial thought the effects were significant enough that they should not treat their child with methylphenidate, and no side effects other than appetite suppression continued during regular therapy after the trial

Follow‐up 6 months after: n = 33. 15 had no change in weight curves. 1 gained 7 kg beyond his original percentile

Notes

Sample calculation: no information

Ethics approval: no information

Comments from study authors

  1. Participants in this study were not a randomly selected group of children with ADD but, rather, a referral population already screened by their school psychologists and primary care physicians

  2. Individual children with differing absorption, metabolism or underlying neurochemical abnormality may have different response thresholds, and potential responders may have been overlooked because they did not consistently try a higher dose

  3. 15% of children with ADD in this study were adopted

  4. The finding that non‐responders were older may reflect the development of secondary characteristics such as decreased motivation and poor study habits in long‐term ADD ‐symptoms not easily reversed during a short trial

  5. A problem inherent in this trial, as in an open trial, is dependence on the observations of teachers and parents who have variable observational skills, variable tolerance for symptoms of ADD and different perspectives on medication

  6. Lower Conners' scores may be explained by the fact that many characteristics rated on Conners' questionnaires reflect hyperactivity, and some of these children were more inattentive than hyperactive

Key conclusions of study authors

  1. 51 (of 70) children showed improvement during 1 of the 2‐week periods, and that period corresponded with methylphenidate therapy in 48

  2. 6 of the 22 who did not respond to methylphenidate experienced worsening of function while taking the drug. No serious side effects were reported during the trial

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The pharmacist labelled the 2 sets of capsules as “Medicine A” and “Medicine B” in either order by coin flip for the first 22 trials, and then by using a random numbers table

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The manner of labelling was sealed by the pharmacist in an envelope that was not opened until the trial had ended and findings had been discussed with the parents

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objectivity was lessened for a few parents because the decreased appetite associated with methylphenidate led them to suspect which capsules contained the drug

Incomplete outcome data (attrition bias)
All outcomes

Low risk

In the few trials for which 1 to 3 of 10 items on Conners' questionnaire had not been scores, the score was prorated based on 30 points maximum

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

None

Vested interest bias

Unclear risk

No information

McGough 2006

Methods

Phase II, randomised, double‐blind, placebo‐controlled, laboratory‐classroom, cross‐over study with a lead‐in open‐label dose‐optimisation phase

  1. Mehtylphenidate transdermal system

  2. Placebo

Participants

Number of participants screened: 93

Number of participants included: 80

Number of participants randomly assigned: methylphenidate 80, placebo 80 (cross‐over)

Number of withdrawals: 7 "adverse events", 3 consent withdrawn, 2 lost to follow‐up, 1 because of "lack of efficacy". 1 participant discontinued because of "protocol violation"

Diagnosis of ADHD: DSM‐IV‐R (combined 62 (79%), hyperactive‐impulsive 4 (5%), inattentive 13 (17%))

Age: mean 9.1 years (SD 1.7, range 6 to 12)

IQ: > 70

Sex of baseline participants included in ITT: 57 boys (72%), 22 girls (28%)

Methylphenidate naive: 37%

Ethnicity: Caucasian 55 (70%), African American 8 (10%), Asian 2 (3%), other 14 (18%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 12 years of age inclusive

  2. ADHD DSM‐IV diagnosis using Kiddie Schedule for Affective Disorders and Schizophrenia and psychiatric assessment

  3. ADHD Rating Scale score ≥ 26 at baseline/unmedicated

  4. Normal laboratory parameters and vital signs including ECG

Exclusion criteria

  1. Comorbid psychiatric diagnosis (apart from oppositional defiant disorder)

  2. History of seizures

  3. History of tic disorders

  4. Mental retardation

  5. Any illness or skin disorder that might jeopardise safety or compromise study assessments

  6. No clonidine, atomoxetine, antidepressants, investigational medications, hepatic P450 enzyme‐altering agents, medications with central nervous system effects, sedatives, anxiolytics or antipsychotics within the 30 days before screening

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: "At the end of the dose optimization phase of the study, the majority of patients (78%) were optimized to either the 16 mg or 20 mg dosage strengths"

Administration schedule: once daily

Duration of each medication condition: 1 week

Washout before study initiation: "up to 28 days"

Medication‐free period between interventions: 4:00 PM to 7:00 AM the next day (15 hours)

Titration period: 5‐week dose‐optimisation phase

Treatment compliance: "During the laboratory classroom period, 97% and 96% of participants were compliant with [methylphenidate transdermal system] MTS and placebo treatments respectively"

Outcomes

ADHD symptoms

  1. Primary outcome

    1. Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale at multiple time points: pre‐dose and 2, 3, 4, 5, 6, 7.5, 9. 10.5 and 12 hours post dose

    2. ADHD Rating Scale, Fourth Edition (administered at each visit)

    3. Conners' Parent Rating Scale ‐ Revised Short Version (Conners 1997a). Completed at 11:00 AM and 3:00 PM on the Sunday before the first visit and subsequently, before each visit to the centre

"The mean values of the CPRS‐R over the 11:00 AM and 3:00 PM time points were used in the analysis"

General behaviour

  1. Clinical Global Impressions ‐ Severity and ‐ Improvement scales as well as the Parent Global Assessment

Non‐serious adverse outcomes

  1. Vital signs, blood pressure, pulse, oral temperature, respiratory rate, height, weight, laboratory measures, physical examination, dermal evaluation

Notes

Sample calculation: yes

Ethics approval: yes

Key conclusions of study authors

  1. Treatment with methylphenidate transdermal system resulted in statistically significant improvement in all efficacy measures

  2. Time course and therapeutic effects of methylphenidate transdermal system suggest that this novel methylphenidate delivery system provides efficacious once‐daily treatment for ADHD

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; "Participants were either known to be responsive to stimulants or naive to stimulant treatment"

Any withdrawals due to adverse events: yes; 7/93 participants were withdrawn because of adverse effects during the dose‐optimisation period (i.e. before randomisation)

Email correspondence with study authors: June 2014. We obtained supplemental information regarding risk of bias. Additional data were not available (Ramstad 2013a [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Participants were randomized into either 1 week of [methylphenidate transdermal system] MTS or 1 week of [placebo transdermal system] PTS (in their individually optimized dose) and were crossed over to the opposite treatment the following week"

From correspondence: "Participants were randomized centrally for each of the study conditions. Randomization codes were not available to site study staffs, but were provided to research pharmacies at each site which corresponded to a particular dose pack" (Ramstad 2014 [pers comm])

Allocation concealment (selection bias)

Low risk

See above

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Phase‐II randomized, double‐blind, placebo‐controlled laboratory classroom, crossover study with a lead‐in open‐label dose optimization phase"

From correspondence: "Active and inactive patches were identical in appearance" (Ramstad 2014 [pers comm])

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

See above

Incomplete outcome data (attrition bias)
All outcomes

High risk

Safety population according to Table Two is n = 93. Table Six lists safety data for n = 80 participants only. Safety data for 13 participants appear to be "missing" ‐ including those from participants withdrawn before randomisation as the result of adverse events

Selective reporting (reporting bias)

High risk

Safety population according to Table Two is n = 93. Table Six lists safety data for n = 80 participants only. Safety data for 13 participants appear to be "missing" ‐ including those from participants withdrawn before randomisation as the result of adverse events

Vested interest bias

High risk

"This study was supported by funding from Shire US Inc"

Conflicts of interest: Two medical writers acknowledged (Amy M. Horton & Michelle Roberts) but were unclear about where they came from or what their role was in the publication

McInnes 2007

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate at low, medium and high doses

  2. Placebo

Phases: 4

Participants

Number of participants screened: 17

Number of participants included: 16 (12 boys, 4 girls). Participants were randomly assigned to 1 of 12 possible drug condition orders

Number of participants followed up: 16

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV‐TR (combined (63%), hyperactive‐impulsive (6%), inattentive (31%))

Age: mean 9.2 years (range 7 to 12)

IQ: mean 107.7 (range not reported)

Methylphenidate naive: ˜ 80%

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (19%), conduct disorder (25%), generalised anxiety disorder (31%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children 7 to 12 years of age with ADHD referred to an out‐patient neuropsychiatry clinic for evaluation of response to methylphenidate

Exclusion criteria

  1. Children with low cognitive performance (IQ < 80)

Interventions

Participants were randomly assigned to 1 of 12 possible drug condition orders of low, medium and high methylphenidate and placebo

Mean methylphenidate dosage: low (mean 0.21 mg/kg to 0.33 mg/kg, SD 0.07 to 0.02); medium (mean 0.31 mg/kg to 0.43 mg/kg, SD 0.09 to 0.03); and high (mean 0.42 mg/kg to 0.65 mg/kg, SD 0.13 to 0.15) methylphenidate Administration schedule: 1 per day at 9:00 AM

Duration of each medication condition: 1 day

Washout before study initiation: 48 hours before study

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Inattention/overactivity symptoms based on 5/10 items from the Iowa Conners’ Rating Scale, rated by observer 90 to 120 minutes after ingestion of capsule

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. With respect to the medication protocol, we cannot predict that similar results would hold with longer‐term treatment with methylphenidate, given that these findings were obtained under a single acute drug challenge

  2. Our findings for a predominantly male group of children with ADHD may not be generalisable to other groups of children with ADHD, for example, community samples that involve more girls and different rates of occurrence of comorbid conditions

  3. Small sample size places limitations on conclusions that can be drawn from our findings

Key conclusions of study authors

  1. Findings provide preliminary evidence that methylphenidate affects higher‐level language comprehension skills, which require sustained attention and mental effort

  2. If generalisable to classroom listening skills, these findings have implications for clinicians and teachers involved with children with ADHD

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

'Multiple blind procedures', capsules identically packaged by pharmacists

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

'Examiner, who was kept blind to child’s medication status'

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No LTFU

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

High risk

Symptom data not reported

Vested interest bias

High risk

Supported by funds from The Psychiatric Endowment Fund

Conflicts of interest: Study authors had received funding from Eli Lilly, Shire Pharmaceuticals, Janssen‐Ortho and McNeil Pharmaceuticals

Moshe 2012

Methods

Two‐week, randomised, double‐blind, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 78

Number of participants included: 57 (all boys). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 57

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined and hyperactive‐impulsive (53%), inattentive (47%))

Age: mean 9.5 years (range 7 to 12)

IQ: normal

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Israel

Setting: out‐patient clinic

Comorbidity: no

Comedication: no

Sociodemographics: representing all socioeconomic strata

Inclusion criteria

  1. Male

  2. 7 to 12 years

  3. ADHD, DSM‐IV

  4. Drug‐naive and with no other intervention

  5. Suitable candidate for methylphenidate treatment

  6. Attention disorder was associated with a significant effect on daily life, and scores on 1 of the Attention subscales of both parent and teacher questionnaires were 1.5 SD or above the mean as suggested in clinical guidelines

Exclusion criteria

  1. Chronic psychiatric and neurological disorders, for example, obsessive‐compulsive disorder

  2. Tourette's syndrome

  3. Seizure disorder

  4. Severe learning disability (defined by special education enrolment)

  5. Definitive primary diagnosis of an anxiety disorder (DSM‐IV) or sensory impairment

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.3 mg/kg immediate‐release methylphenidate and placebo

Methylphenidate dose range: 6 mg to 12 mg

Administration schedule: once daily

Duration of each medication condition: 1 week

Washout before study initiation: no (drug‐naive)

Titration period: no

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale, revised: weekly (after each intervention period)

Notes

Sample calculation: yes

Ethics approval: yes

Comments from study authors

  1. Results of the present study should be interpreted with caution

  2. As only boys were included, the results might not be valid for girls

  3. Children were clinic referrals and therefore might not be representative of the population of children with ADHD at large

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April to October 2013. We received supplemental data from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned with a table of random numbers

Allocation concealment (selection bias)

Low risk

Placebo (prepared as look‐alike capsules by the hospital pharmacy)

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 information

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

According to study authors, all planned outcomes were assessed and analysed

Vested interest bias

Low risk

Not funded

Conflicts of interest: none declared

Muniz 2008

Methods

Randomised, multi‐centre, double‐blind, 5‐period, cross‐over trial in a laboratory classroom setting with 3 interventions

  1. Extended‐release dexmethylphenidate

  2. Extended‐release racemic methylphenidate hydrochloride

  3. Placebo

Phases: extended‐release dexmethylphenidate 20 mg/d, extended‐release dexmethylphenidate 30 mg/d, extended‐release racemic methylphenidate hydrochloride 36 mg/d, extended‐release racemic methylphenidate hydrochloride 54 mg/d and placebo

Participants

Number of participants screened: 84

Number of participants included: 84 (55 boys, 29 girls). Participants were randomly assigned to 1 of 5 possible drug condition orders

Number of participants followed up: 81

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐IV (combined (89.3%), inattentive (10.7%))

Age: mean 9.5 years (SD 1.7, range 6 to 12)

IQ: not stated

Methylphenidate naive: 0%

Ethnicity: Caucasian (42.9%), African American (27.4%), Hispanic (28.6%), other (1.2%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD using Diagnostic Interview Schedule for Children

  2. On stabilised total daily dose or nearest equivalent dose of 40 mg to 60 mg of extended‐release racemic methylphenidate hydrochloride or 20 mg to 30 mg extended‐release dexmethylphenidate for ≥ 2 weeks before screening visit

Exclusion criteria

  1. Tic disorder or Tourette’s syndrome

  2. History of a seizure disorder

  3. Psychiatric illness

  4. Substance abuse disorder

  5. Taking prohibited concomitant medications or ADHD medication other than methylphenidate

  6. Taking antidepressant or psychotropic medications

  7. Had begun psychotherapy within 3 months before randomisation

  8. Home‐schooled children

  9. Females of child‐bearing potential with positive urine pregnancy test before enrolment (or, if sexually active, not using adequate and reliable contraception)

Interventions

Participants were randomly assigned to 1 of 5 possible drug condition orders of extended‐release dexmethylphenidate 20 mg/d, 30 mg/d, 36 mg/d, 54 mg/d and placebo

Mean methylphenidate dosage: not stated

Administration schedule: once daily. Morning dosing as 2 capsules Sunday to Saturday. 6 doses were administered at home (Sunday to Friday), and the Saturday dose was administered by research staff. This was repeated until all 5 treatments had been administered. Mean duration of exposure to study medication was 7 days for all 5 treatments

Washout before study initiation: 6 days medication‐free

Titration period: "On stabilized total daily dose or the nearest equivalent dose of 40 to 60 mg of d,l‐MPH or 20 to 30 mg d‐MPH for at least 2 weeks prior to the screening visit" initiated before randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Primary efficacy variable

    1. Change from pre‐dose Swanson, Kotkit, Agler, M‐Flynn and Pelham (SKAMP) Rating Scale‐Combined score at 2 hours post dose with extended‐release dexmethylphenidate 20 mg/d compared with extended‐release racemic methylphenidate hydrochloride 36 mg. This change was calculated by subtracting the pre‐dose value (hour 0 score) from the post‐dose value

  2. Secondary outcome measures

    1. Change from pre‐dose (0 hour) on Combined, Attention and Deportment subscores of the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale, at specified intervals post dose (0.5, 1, 2, 3, 4, 6, 8, 10, 11 and 12 hours), and area under the score vs time curve (AUC) of the change from pre‐dose in Combined score from hour 0 to hour 4 (AUC 0 to 4), and from hour 0 to hour 12 (AUC 0 to 12)

General behaviour

  1. "The Conners’ Parent Rating Scale (CPRS), a 27‐item questionnaire designed to evaluate children’s behaviour (Conners 1998a), was completed by parents on the Practice Day to assess behaviour without medication and at each subsequent assessment day to rate the child’s behaviour during the previous week"

Serious adverse events

  1. Safety assessments consisted of monitoring and recording of all adverse events

Non‐serious adverse events

  1. Safety assessments consisted of monitoring and recording all adverse events and recording vital signs and body weight at each visit

  2. Laboratory parameters (including haematology, blood chemistry and urinalysis), ECGs and results of physical examinations were assessed for abnormalities at screening and final visits (no final visit assessments were carried out for ECGs and physical examinations)

Notes

Sample calculation: yes; "It was determined that approximately 90 patients were required to detect a 0.05‐level treatment difference at 84% power assuming a difference and standard deviation of 3.5 and 11 for SKAMP‐Combined score, at 2 hours post‐dose using a paired t‐test"
Ethics approval: no information

Key conclusions of study authors

  1. "The results of this study demonstrated that all active treatments generally provided significant improvement in ADHD symptoms over placebo over 11 to 12 hours postdose in children 6–12 years old"

  2. "The primary efficacy variable, adjusted mean change in SKAMP‐Combined score from pre‐dose to 2 hours post‐dose, was significantly greater during treatment with d‐MPH‐ER 20 mg/day than d,l‐MPH‐ER 36 mg/day (adjusted mean change 10.65 and 5.94, respectively; p 0.001). Similar results at two hours post‐dose were noted for the secondary measure of SKAMP‐Combined score comparing d‐MPH‐ER 30 mg/day with d,l‐MPH‐ER 54 mg/day (adjusted mean change 11.17 and 7.52, respectively; p0.001)."

Comment from review authors

  1. We did not include the Swanson pencil and paper math test of "academic productivity" among ADHD outcome measures. This may be seen as an ADHD outcome measure, but it does not specifically measure the 3 key ADHD core signs of inattention, hyperactivity and impulsivity

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; "Children recruited for this study had been stabilized on a total daily dose or the nearest equivalent dose of 40 to 60 mg of d,l‐MPH or 20 to 30 mg d‐MPH for at least 2 weeks prior to the screening visit" ‐ so presumably non‐responders to methylphenidate and those experiencing intolerable adverse events while taking methylphenidate were not included

Email correspondence with study authors: July 2014. Emailed study authors to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"A total of 84 subjects were randomized to receive treatment and were included in the efficacy and safety analyses"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"After the Practice Day, the first assigned treatment was dispensed to the parents as blinded capsules according to their child’s randomized sequence. To maintain blinding, all treatments were over‐encapsulated and the same number of capsules were given once daily for each sequence"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The ratings were based on the frequency and quality of behaviours as observed by three independent, blinded raters in each class. To maintain consistency throughout the study, the blinded observers were responsible for observing and rating the same 6 children at specified intervals throughout the 12‐hour testing period at each center"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"The intent‐to‐treat population included all randomized patients who took at least 1 dose of study medication and had at least 1 post‐dose efficacy measurement"

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No selective outcome reporting

Vested interest bias

High risk

"This study was funded by Novartis Pharmaceuticals Corporation and reports the following involvement: design and conduct of the study; collection, management, analysis, and interpretation of data; preparation, review, and approval of the manuscript"

Conflicts of interest: Dr. Muniz is an employee of Novartis Pharmaceuticals Corporation. He has no other relationships to disclose. Dr. Brams reports the following relationships: serves as speaker, consultant and Advisory Board member for Novartis and Shire; receives grant research support from Novartis, Shire and Eli Lilly. Dr. Mao reports the following relationships: speaker for Novartis, Eli Lilly, Bristol‐Myers Squibb, AstraZeneca and Shire; consultant for Eli Lilly, Novartis and Shire; receives grant research support from Novartis. Mr. McCague is an employee of Novartis Pharmaceuticals Corporation. He has no other relationships to disclose. Ms. Pestreich is an employee of Novartis Pharmaceuticals Corporation. She has no other relationships to disclose. Dr. Silva reports the following relationships: none since 15 December 2006; before that, she was a speaker for Novartis, AstraZeneca and Janssen; received grant/research support from Novartis and Celgene

Murray 2011

Methods

Phase IV, double‐blind, randomised, cross‐over, analogue classroom trials with 2 interventions

  1. OROS methylphenidate

  2. Placebo

Phases

  1. Screening/washout phase: up to 28 days

  2. Titration: up to 6 weeks

  3. Double‐blind assessment period, with the following subperiods

    1. Open‐label OROS methylphenidate: school day 1, OROS methylphenidate or placebo

    2. Open‐label OROS methylphenidate: ≥ 7 days

    3. School day 2: OROS methylphenidate or placebo

Participants

Number of participants screened: not stated

Number of participants included: 89. Participants randomly assigned to 1 of 2 possible drug condition orders: 68

Number of withdrawals before randomisation: 21

Number of withdrawals after randomisation: 2

Diagnosis of ADHD: DSM‐IV‐TR (combined (59%), hyperactive‐impulsive (4%), inattentive (37%))

Age: mean 10.75 years (range 5 to 15)

IQ: > 80

Sex: 45 boys, 23 girls

Methylphenidate naive: 65%

Ethnicity: Caucasian (62%), African American (28%), Asian (not stated), Hispanic (not stated), other (10%)

Country: USA

Setting: out‐patient clinic

Comorbidity: anxiety (9%), depressive disorders (1%), learning disability (38%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. 9 to 12 years of age

  2. DSM‐IV‐TR diagnosis of ADHD

  3. Parent‐completed ADHD‐RS‐IV total or subscale scores ≥ 90th percentile for their age and sex

  4. Attendance at a public or private school

  5. Ability to read and understand English

  6. Patients currently receiving ADHD medication must be inadequately managed on their current stimulant dose

  7. To be eligible for the double‐blind, randomised assessment period, participants had to reach their individualised OROS methylphenidate dose, defined as

    1. ADHD Rating Scale, Fourth Edition (as scored by parent or guardian): ≤ 75th percentile for age and sex

    2. ADHD Rating Scales, Fourth Edition (as scored by parent or guardian): between 75th and 85th percentiles for age and sex after either (1) a dose decrease for tolerability (1 dose decrease by 18 mg to a minimum of 18 mg/d was allowed), or (2) having reached a dosage of 54 mg/d

Exclusion criteria

  1. Estimated full‐scale IQ score ≤ 80, as determined by the 4‐subtest version of the Wechsler Abbreviated Scale of Intelligence

  2. Severe learning disability, defined as ≥ 2 SD below the mean score for their age on Gray Oral Reading Test, Test of Phonological Processing or Wechsler Individual Achievement Test, Second Edition

  3. History or current diagnosis of a neurological or psychiatric disorder that might compromise the participant’s welfare or ability to comply with study requirements

  4. Inability to take or tolerate OROS methylphenidate

  5. History of or current primary diagnosis of severe anxiety disorder, conduct disorder, psychotic disorders, pervasive developmental disorder, eating disorder, obsessive‐compulsive disorder, sleep disorder, major depressive disorder, bipolar disorder, substance use disorder, chronic tic disorder, personal or family history of Tourette's syndrome

  6. Weight < 3rd percentile for age

  7. History of hospitalisation for treatment of a mood, anxiety or psychotic disorder

  8. History of failed response to methylphenidate

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of OROS methylphenidate and placebo

Mean OROS methylphenidate daily dosage: 47.6 mg

Administration schedule: once daily, morning

Average duration of OROS methylphenidate treatment: not stated

Duration of placebo intervention: 1 day

Washout before study initiation: up to 28 days

Medication‐free period between interventions: no

Titration period: before randomisation, up to 6 weeks

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale, observer‐rated: 4 hours post dose (at the 2 laboratory days)

Serious adverse events

  1. Serious adverse effects assessed at the 2 laboratory days and during open‐label periods

Non‐serious adverse events

  1. Adverse effects, vital signs and body weight, at the 2 laboratory days

  2. Adverse effects were collected during the open‐label phase

Notes

Sample calculation: yes

Ethics approval: yes

Key conclusion of study authors

  1. OROS methylphenidate improves performance on measures of attention and vigilance, behaviour and working memory in a laboratory school setting in 9‐ to 12‐year‐olds with ADHD

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; a history of failed response to methylphenidate was an exclusion criterion

Any withdrawals due to adverse events: 2

Email correspondence with study authors: June 2013 to June 2014. We have attempted to obtain supplemental efficacy data (Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale) and safety data from study authors. We are awaiting data from the Yale Open Data Access Project

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

Low risk

Double‐blind. Placebo and OROS methylphenidate were matched in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

3 participants received OROS methylphenidate on both laboratory school days in error; therefore, only data for their first laboratory school assessment were included in the analyses

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

High risk

Supported by Ortho‐McNeil Janssen Scientific Affairs, LLC

Conflicts of interest: Several study authors had affiliations with pharmaceutical companies producing methylphenidate

Musten 1997

Methods

Double‐blind, randomised, placebo‐controlled, cross‐over trial with 3 interventions

  1. Low dose of methylphenidate

  2. High dose of methylphenidate

  3. Placebo

Participants

Number of participants screened: 109

Number of participants included: 54 met inclusion criteria; of these, the parents of 13 children refused methylphenidate treatment. In the final sample, 41 children were included. Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: 31

Number of withdrawals: 10

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Regarding participants completing the study

Age: mean 58.07 months (range 48 to 70)

IQ: mean 99.26

Sex: 26 boys, 5 girls

Methylphenidate naive: 93.5%

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (84%), conduct disorder (19%), mood disorder (0%), obsessive‐compulsive disorder (0%), overanxious disorder (0%), somatisation disorder (0%), psychotic symptoms (0%)

Comedication: not stated

Sociodemographics: combined parental income Canadian $42,000: 2‐parent home (74%), single‐parent home (26%). Significant differences were observed between the treatment refused group (n = 13) and the treatment completed group (n = 31) on baseline symptoms assessed by Diagnostic Interview for Children and Adults‐Parents; Swanson, Nolan and Pelham scale; and Conners' hyperactivity ratings

Inclusion criteria

  1. 4 to 6 years of age

  2. DSM‐III‐R diagnosis of ADHD (assessed by parent reports in the Diagnostic Interview for Children and Adults‐Parents and a score > 1 of 8 on the 14 DSM‐III‐R items on the parent‐rated Swanson, Nolan and Pelham Scale)

  3. Standard score 80 on the Peabody Picture Vocabulary Test if unilingual English, 72 if bilingual

  4. Mean score equal to 1.5 SD above age and sex means on the Hyperkinesis Index of Conners' Parent Rating Scale‐Revised, as completed by the parent. Reports from day care providers or pre‐schools were also required to indicate problem behaviours

  5. Attention score < 88 seconds on the parent‐supervised attention task. This criterion is 1.5 SD above the mean for attention on the task as performed by normal pre‐school children

  6. Parents and children fluent in English

Exclusion criteria

  1. Attending or entering first grade at the time of assessment or for the duration of the study

  2. Sensory or physical handicaps, developmental disorders (e.g. autism), neurological disease or obvious CNS dysfunction as assessed by a paediatrician

  3. Had been receiving methylphenidate longer than 6 months, or daily dose was above dose specified in the research protocol

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of low‐dose (0.3 mg/kg) and higher‐dose (0.5 mg/kg) methylphenidate and placebo

Administration schedule: twice daily, morning and lunch

Duration of each medication condition: 7 to 10 days

Washout before study initiation: 48 hours before screening assessment

Titration period: none

Treatment compliance: Treatment compliance was determined by counting the number of pills returned to the researcher at the end of each assessment week

Data on compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale ‐ Revised

Non‐serious adverse events

  1. Side Effects Rating Scale (Barkley 1990) rated by parent at the end of each treatment period

Notes

Sample calculation: no

Ethics approval: no information

Comment from study authors

  1. Data on side effects are limited because of the age group of the population under investigation (4 to 6 years) ‐ pre‐schoolers cannot always articulate medication‐related sensations, and this may have interfered with parents’ ability to detect medication side effects

Key conclusions of study authors

  1. Musten 1997 (Musten 1997): Results suggest that methylphenidate can be used to improve the functioning of pre‐school‐aged children with ADHD, in a manner similar to their school‐age counterparts

  2. Firestone 1998 (Musten 1997): Results indicate that methylphenidate has relatively low toxicity in pre‐school children (over the first 7 to 10 days), that some behavioural changes that might be viewed as side effects of methylphenidate are actually normal behaviours or ADHD behaviours in pre‐school children (e.g. sociability), that these "side‐effect" behaviours are more common in pre‐school than school‐aged children, that some "side effects" of methylphenidate are associated with improvements in behaviour and that pre‐school and school‐aged children may experience different side effects of methylphenidate (e.g. mood changes, anxiety)

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: June 2013. Personal email correspondence with study author did not provide supplemental data and information as requested because of author's retirement, and because he has no access to the data because the study took place 15 years ago

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Treatment was presented in a fully randomised order as prepared by the hospital's pharmacy department

Allocation concealment (selection bias)

Low risk

Methylphenidate and placebo were placed in orange gelatin capsules to disguise taste differences

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants, research personnel and medical personnel were unaware of the order of medication conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All participants, research personnel and medical personnel were unaware of the order of medication conditions

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only data for children completing the entire study were analysed

Selective reporting (reporting bias)

Unclear risk

Not possible to get a copy of the protocol

Vested interest bias

Low risk

Supported by Health Canada grant

Conflicts of interest: none declared

Newcorn 2008

Methods

Twenty‐site, randomised, placebo‐controlled, double‐blind, 6‐week, parallel trial with 3 arms

  1. Extended‐release methylphenidate

  2. Placebo

  3. Atomoxetine

Participants

Number of participants screened: 635

Number of participants included: 516

Number of participants randomly assigned: methylphenidate 220, placebo 74

Number of participants followed up: methylphenidate 180, placebo 57

Number of withdrawals: methylphenidate 40, placebo 17

DSM‐IV diagnosis of ADHD (combined (67%), hyperactive‐impulsive (1%), inattentive (32%))

Age: MPH mean 10.2, P mean 10.1 (range 6 to 16)

IQ: not stated

Sex: 211 boys, 83 girls

MPH‐naive: 41%/121

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (36%)

Comedication: not stated

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Children 6 to 16 years of age

  2. Meeting DSM‐IV criteria for ADHD, any subtype

  3. Symptom severity at entry was required to be ≥ 1.5 SD above US age and sex norms, as assessed by the ADHD Rating Scale, Fourth Edition ‐ Parent Version

  4. ADHD as the primary diagnosis

Exclusion criteria

  1. Seizures, bipolar disorder, psychotic illness or pervasive developmental disorder

  2. Taking concomitant psychoactive medications.

  3. Anxiety or tic disorders or both

  4. Previously treated with an adequate trial of methylphenidate or amphetamine and either did not experience at least some improvement in ADHD signs and symptoms (non‐responders) or did experience intolerable adverse events

Interventions

Participants were randomly assigned to OROS methylphenidate or placebo

Mean methylphenidate dosage: 39.9 mg/d (SD 14.6) or 1.26 mg/kg/d (SD 0.55)

Administration schedule: single morning dose

Duration of intervention: 6 weeks

Titration period: none

Treatment compliance: not stated. Patients were required to discontinue any psychoactive medication for ≥ 5 days before entering the study

Outcomes

ADHD symptoms

  1. ADHD Rating Scale: observer‐rated at baseline and at weeks 1, 3 and 6

  2. Conners' Parent Rating Scale, ADHD Index: rated at baseline and at weeks 1, 3 and 6

  3. General behaviour

Quality of life

  1. Child Health Questionnaire: parent/teacher/observer‐rated at baseline and at weeks 3 and 6

  2. Serious adverse events

Non‐serious adverse events

  1. Open‐ended questioning for adverse events and vital signs: observer‐rated at baseline and at weeks 1, 3 and 6

  2. No differences were observed in mean change in systolic blood pressure between placebo and methylphenidate

  3. Weight loss was significantly greater with methylphenidate than with placebo

Notes

Sample calculation: no

Ethics approval: yes; approved by each site’s ethical review board

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Comments from study authors (limitations)

  1. It is likely that the methylphenidate dose was suboptimal for some adolescent participants

  2. Restricting the dose of methylphenidate to 54 mg could also have limited response in some younger participants, as OROS methylphenidate sometimes is prescribed at doses higher than the FDA‐recommended maximum for children

Key conclusion of study authors

  1. Both treatments produced robust improvement, with a statistically significant difference in response favouring OROS methylphenidate

Email correspondence with study authors: November 2013 and January 2014. We requested additional data from study authors but never received them

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study drugs were administered according to a double‐dummy design. Identically appearing capsules were used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Study drugs were administered according to a double‐dummy design. Identically appearing capsules were used

Incomplete outcome data (attrition bias)
All outcomes

Low risk

NNTB was calculated for each treatment in relation to placebo and for atomoxetine in relation to methylphenidate. The number of participants was chosen to have 90% power to declare non‐inferiority on the basis of a comparison of response rates, with a non‐inferiority margin of 15%

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Supported by Eli Lilly and Company

Conflicts of interest: Dr. Newcorn receives grant support from Eli Lilly and McNeil; is a consultant and/or advisor for Eli Lilly, McNeil, Shire, Novartis and Sanofi‐Aventis; and is a member of Speakers' Bureaus for Eli Lilly and Novartis. Dr. Kratochvil receives grant support from Abbott, Cephalon, Eli Lilly, McNeil, Pfizer, Shire and Somerset; receives from Eli Lilly study medication for an NIMH (National Institute of Mental Health)‐funded study; is a consultant for Abbott, AstraZeneca, Eli Lilly and Pfizer; and is a member of the Eli Lilly Speakers' Bureau. Dr. Casat receives research funding from Eli Lilly, Novartis and Abbott, and serves on an advisory board for Eli Lilly. Dr. Allen and Dr. Ruff are employees and shareholders of Eli Lilly. Dr. Michelson and Dr. Moore are former employees of Eli Lilly

Nikles 2006

Methods

Cross‐over trial with 2 interventions

  1. Short‐acting stimulants (methylphenidate and dexamphetamine)

  2. Placebo

Phases: Each trial included 3 pairs of treatment periods (n‐of‐1). Each pair contained the stimulant and the comparator stimulant or placebo. The child’s doctor individualised the dosing, and the order of drugs was randomly assigned within each pair. Two treatment periods were included in a school week, and 2 days per treatment period (not including Wednesdays and weekends)

Participants

Number of participants screened: 108 (85 boys, 21 girls). Two boys repeated n‐of‐1 trials; 22 (20%) trials were not completed

Number of participants included: 86. Participants were randomly assigned to 1 of 3 possible drug condition orders

Number of participants followed up: 86

Number of withdrawals: 10 (12%) trials were not completed
Diagnosis of ADHD: DSM‐IV (subgroups not stated)

Age: median 10 years (range 5 to 16)

IQ: not stated

Sex: 66 boys, 18 girls

Methylphendaite‐naive: 36 were taking methylphenidate, 47 dexamphetamine, 3 unknown pre‐trial medication

Ethnicity: All were Caucasian (Nikles 2007 in Nikles 2006)

Country: Australia

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: caregiver's occupation: full‐time work 21%, part‐time or casual work 33%, unemployed or retired 5%, unpaid homemakers 25%, other 10%, unknown 5%

Inclusion criteria

  1. Clinical diagnosis of ADHD according to DSM‐IV criteria

  2. Stable on an apparently optimal dose of stimulant

  3. Informed consent from parent and school teacher (children 12 years of age provided assent)

  4. 5 to 16 years of age

  5. Uncertainty about treatment effectiveness

Exclusion criteria

  1. No teachers available and willing to provide observations

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of individual doses of methylphenidate, dexamphetamine and placebo

Mean methylphenidate dosage: not stated
Administration schedule and time points: different for different individuals, as this is a series of n‐of‐1 trials

Duration of each medication condition: 2 days each of methylphenidate and placebo per week

Washout before study initiation: 40 hours (from 4:00 PM Tuesday to 8:00 AM Thursday) and 64 hours (4:00 PM Friday to 8:00 AM Monday)

Titration period: not stated, but all were stabilised on an "optimal" dose of stimulant, initiated before randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

First 41 participants used at the end of each treatment period

  1. Conners’ Teacher and Parent Self Reported ‐ Revised, short form

  2. Conners‐Wells Adolescent Rating Scales

From 42nd participant onwards used at the end of each treatment period

  1. Changed to ADHD du Paul Rating Scale IV parent and teacher questionnaires, "because they were less expensive but adequately reliable and valid for monitoring response to treatment"

  2. Conners‐Wells Adolescent Rating Scales

Non‐serious adverse events

  1. Assessment of adverse effects by parent and teacher during and at the end of each treatment period

Notes

Sample calculation: irrelevant

Ethics approval: yes

Key conclusion of study authors

  1. Attention deficit hyperactivity disorder n‐of‐1 trials can be implemented successfully by mail and telephone communication. This type of trial can be valuable in clarifying treatment effect when it is uncertain, and in this series, treatments had a noticeable impact on short‐term management

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes (n = 1, insomnia and depression)

Comments from review authors

  1. This study was more about the use of n‐of‐1 trials in determining optimal drug treatment in cases for which this was uncertain

  2. Nikles 2007 (in Nikles 2006) does not add any relevant outcome data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The order of drugs was randomly assigned within each pair"; "There were three pairs of treatment periods, with the order of drugs randomly assigned by a computer‐generated randomisation schedule within each"

Allocation concealment (selection bias)

Low risk

See above

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Patients, parents, doctors, and the research assistant were all blinded to medication order"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"A hospital pharmacy encapsulated the medication (crushing of tablets and production of identical capsules containing either medication or placebo)"; "Active medication was encapsulated and identical placebo capsules were produced by a hospital pharmacy"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Low risk

All outcomes are reported

Vested interest bias

Unclear risk

No information

Conflicts of interest: Study authors have indicated that they have no financial relationships relevant to this article to disclose

Oesterheld 1998

Methods

Cross‐over trial with 3 interventions

  1. Methylphenidate

  2. Lactose placebo

  3. Vitamin C placebo

Phases: Five‐day trial lasting for 3 consecutive weeks, with 2 medication‐free days between phases

Participants

Number of participants screened: 30

Number of participants included: 4. Participants were randomly assigned to different possible drug condition orders of methylphenidate, lactose placebo and vitamin C placebo

Number followed up: 4 (2 boys, 2 girls)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (66.7%), hyperactive‐impulsive (0%), inattentive (33.3%))

Age: mean 8.25 years (range 5 to 11)

IQ: mean 72.5 (range 63 to 79)

Methylphenidate naive: 100%

Ethnicity: Native American (100%)

Country: USA

Setting: out‐patient clinic (residential school)

Comorbidity: not stated

Comedication: no

Sociodemographics: not living with family

Inclusion criteria

  1. Native Americans

  2. 5 to 12 years of age

  3. Residing at the residential school within 6 months

  4. DSM‐IV ADHD diagnosis

  5. Diagnosis of foetal alcohol syndrome, partial foetal alcohol syndrome or alcohol‐related birth defects according to criteria from the Fetal Alcohol Study Group of the Research Society of Alcoholism (1989)

Exclusion criteria

  1. Pregnancy

  2. Lactose intolerance

  3. Prior psychotropic medication use

  4. Bipolar disorder

  5. Acute and chronic medical or neurological disorders

  6. Current history of seizures

  7. Lead levels > 9 mcg/dL

  8. Height and weight at or below 3rd percentile

  9. IQ < 60

Interventions

Participants were randomly assigned to different possible drug condition orders of methylphenidate (0.6 mg/kg), lactose placebo and vitamin C placebo

Mean methylphenidate dosage: not stated (range 10 mg/d to 17.5 mg/d)

Administration schedule: 7:30 AM, 11:00 AM and 2:00 PM

Duration of each medication condition: 5 days for 3 consecutive weeks

Washout before study initiation: not stated

Medication‐free period between interventions: 2 days

Titration period: none. Fixed dose. Initiated before/after randomisation

Treatment compliance: medication given by nurse (directly observed treatment)

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale (48 items) and Conners' Teacher Rating Scale (39 items): both completed daily in each trial period by caretakers and teachers, respectively

Non‐serious adverse events

  1. Barkley Side Effects Questionnaire was completed by both teacher and caretaker before treatment

Notes

Sample calculation: no

Ethics approval: yes; Human Subjects Committee of the University of South Dakota's School of Medicine and the Research Committee of the Black Hills Children's Home Society

Key conclusion of study authors

  1. When fetal alcohol syndrome and ADHD co‐exist, ADHD symptoms of native children may respond to standard treatment with methylphenidate without major side effects

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from review authors

  1. Methylphenidate was prepared to the nearest 2.5 mg using regular 5 mg and 10 mg methylphenidate tablets that had been crushed and placed within gelatin capsules, which may result in uncertainty of actual dose administered

Email correspondence with study authors: not able to find email addresses for study authors because information is missing from the paper. Not possible to find on the Internet, etc.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The order of the trials was randomly determined

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

School caretakers, nurses, teachers and researchers were blinded as to whether treatment consisted of placebo or active agent. All agents were placed in identical capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

On each day of each trial, a teacher, blinded to evaluation data, rated the child's behaviour in school using Conners' Teacher Rating Scale (39 items). The caregiver, blinded to evaluation data, completed Conners' Parent Rating Scale (48 items) on a daily basis. School caretakers, nurses, teachers and researchers were blinded as to whether treatment consisted of placebo or active agent. All agents were placed in identical capsules

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

Research was supported by a University of South Dakota/USF‐Mini Grant

Conflicts of interest: none declared

Overtoom 2003

Methods

Double‐blind, randomised, cross‐over trial with 4 interventions

  1. Methylphenidate

  2. Desipramine

  3. L‐dopa

  4. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 16. Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: 16 (all boys)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (combined (100%))

Age: mean 10.4 years (range 7 to 12)

IQ: 95.4

Methylphenidate naive: 0 (0%)

Ethnicity: not stated

Country: the Netherlands

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 6), comorbid anxiety disorder (n = 1), specific developmental disorders (n = 3)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosis of ADHD according to DSM‐III‐R criteria

  2. Scored in the clinical range on both the Child Behavior Checklist Inattention factor (t scores > 70, i.e. 98th percentile) and Conners' Teacher Rating Scale Hyperactivity factor (mean factor score > 2.2)

Exclusion criteria

  1. Diagnosis of tic disorder or pervasive developmental disorder

  2. Abnormal values (1 week before medication study) of ECG and blood measures that contraindicate desipramine medication

  3. Family history of severe heart problems

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of methylphenidate, desipramine, L‐dopa and placebo

Mean methylphenidate dosage: 15 mg

Administration schedule: once, in the afternoon

Duration of each medication condition: 1 day

Washout before study initiation: 3 days before for methylphenidate users

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: 100%

Outcomes

Non‐serious adverse events

  1. Sleepiness reported by 1 participant

Notes

Sample calculation: no
Ethics approval: yes

Key conclusions of study authors

  1. Inhibition of performance improved under desipramine but not under methylphenidate or L‐dopa. Response time to the stop signal was marginally shortened after intake of desipramine

  2. Methylphenidate decreases omission and choice errors and causes faster reaction times in trials without the stop tone

  3. No effects of L‐dopa whatsoever were noted

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; all participants were already stable on methylphenidate

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

Not specified

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"A double‐blind randomised design was used"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Low risk

Research supported by Netherlands Organisation for Scientific Research (NWO) Grant 575‐63‐082

Conflicts of interest: not declared

Palumbo 2008

Methods

Multi‐centre, randomised, double‐blind, 16‐week, parallel trial with 2 × 2 factorial design

  1. Methylphenidate alone

  2. Methylphenidate and clonidine

  3. Clonidine alone

  4. Placebo

Two successive 4‐week titration periods followed by an 8‐week maintenance period

Participants

Number of participants screened: 205

Number of participants included: 122 (98 boys, 24 girls)

Number randomly assigned: methylphenidate 29, placebo 30, methylphenidate + clonidine 32, clonidine 31

Number followed up: methylphenidate 18, placebo 10, methylphenidate + clonidine 24, clonidine 31

Number of withdrawals: methylphenidate 11, placebo 20, methylphenidate + clonidine 8, clonidine 5

DSM‐IV diagnosis of ADHD (combined (76%), hyperactive‐impulsive (4%), inattentive (20%))

Age: mean 9.5 (range not reported)

IQ: > 70

Methylphenidate‐naive: 57 (46.7%)

Ethnicity: Caucasian (77.9%), African American (10.7%), Hispanic (6.6%), other (4.9%)

Country: USA

Setting: out‐patient clinic

Co‐morbidity: oppositional defiant disorder (47%), conduct disorder (9%)

Co‐medication: no. Protocol‐based behavioural interventions

Sociodemographics: not stated. Participant groups were similar except for a higher percentage of whites in the clonidine group and some minor differences with regard to family history of ADHD and tics

Inclusion criteria

  1. Children 7 through 12 years of age in school

  2. DSM‐IV ADHD, any subtype: indication of a sufficient number of ADHD symptoms on the Disruptive Behavior Disorders Rating Scale as rated by a teacher

  3. Rating of ADHD symptoms above specified cutoff scores (boys: grade 2 to 3 = 10; grade ≥ 4 = 9; girls: grade 2 to 3 = 7, grade ≥ 4 = 6) on the Iowa Conners' Teacher Rating Scale

  4. Indication of the presence of sufficient ADHD symptoms at home on the Iowa Conners' Parent Rating Scale

  5. Investigators rating of global functioning on the Child Global Assessment Scale (CGAS) ≤ 70, with difficulty evident in ≥ 2 areas, such as school and home

  6. ADHD must be viewed as worthy of treatment with medications, as judged by the parent and the site investigator

  7. Informed consent/assent signed

  8. Designated school for each participant agrees to participate in the study by completing all required questionnaires and following all specified procedures

  9. Child must be able to swallow the tablets and capsules used in this study

Exclusion criteria

  1. Evidence of a tic disorder

  2. Major depression

  3. Pervasive developmental disorder

  4. Autism

  5. Psychosis

  6. Mental retardation

  7. Anorexia nervosa

  8. Bulimia

  9. Serious cardiovascular (e.g. significant hypotension, congenital heart disease) or other medical disorder that would preclude safe use of methylphenidate or clonidine

  10. Impaired renal function or pregnancy

  11. Family history of long QT syndrome, cardiomyopathy or premature (age 45 years) sudden death

  12. Prolonged QTc interval (> 440 milliseconds), high‐grade ventricular ectopy, atrioventricular block beyond first degree, bundle branch block, intraventricular conduction block (> 100 milliseconds), pacemaker rhythm or HR < 60 bpm on the ECG, significant hypotension, cardiomyopathy, congenital heart disease, aortic or pulmonary stenosis, history of syncope

  13. Blood pressure ≥ 2 SD above or below the age‐ and sex‐adjusted mean

  14. Stimulants had to be discontinued ≥ 2 weeks before enrolment

  15. Any other psychotropic medications, anxiolytics or hypnotics had to be discontinued ≥ 6 weeks before enrolment

Previous use of methylphenidate or clonidine was permitted

Interventions

Participants were randomly assigned to immediate‐release methylphenidate or placebo

Mean methylphenidate dosage: 0.76 ± 0.54 mg/kg/d (30.2 ± 18.9 mg/d (methylphenidate‐only group) and 25.4 ± 18.2 mg/d (methylphenidate + clonidine))

Administration schedule: 1 to 3 times daily (morning, noon and afternoon)

Duration of intervention: 12 to 16 weeks

Washout before study initiation: 2 weeks

Titration period: 8‐week flexible‐dose titration period (4 weeks for clonidine, then 4 weeks for methylphenidate) initiated after randomisation

Maintenance period of optimal dose: 8 weeks

Treatment compliance: monitored using pill counts (results of monitoring not stated)

Outcomes

ADHD symptoms

  1. ADHD Rating Scale (ADHD RS‐IV, 18‐item): child psychiatrist‐rated at baseline and at 16 weeks

  2. ASQ: teacher‐rated at baseline and at 4, 8, 12 and 16 weeks

  3. ASQ: parent‐rated at baseline and at 4, 8, 12 and 16 weeks

  4. IOWA Conners' Rating Scale: teacher‐rated at baseline and at 4, 8, 12 and 16 weeks

Quality of life

  1. Child Global Assessment Scale: rated at baseline and at 4, 8, 12 and 16 weeks

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale (20 items, modified from the original 13 items to include potential clonidine side effects): parent‐ and teacher‐rated at baseline and at 4, 8, 12 and 16 weeks.

  2. Spontaneous self reports of adverse events: parent‐ and participant‐rated at baseline and at 4, 8, 12 and 16 weeks, or by telephone calls conducted between visits

  3. Weight, ECG, supine and standing blood pressures and pulse at visits 4, 8, 12 and 16

Notes

Sample calculation: yes; sample size of 140 participants (35 per treatment group) was determined to provide between 80% and 90% power to detect a group difference (effects of clonidine)
Ethics approval: yes; approved by the institutional review board at each site

Comments from study authors

  1. Overall, findings should be viewed cautiously in the light of the relatively small sample size and differential rates of attrition across groups

  2. Findings are limited by the exclusion of children with certain co‐morbid disorders such as mood and anxiety disorders, known cardiac problems or abnormal ECGs

  3. Also, given that all participants received psychoeducational and behavioural interventions as part of the protocol, these results may be limited to settings in which such behavioural interventions are applied

  4. Study relied largely on parent and teacher questionnaires to identify possible side effects of medications. However, such rating scales may overestimate rates of medication side effects because sometimes such complaints reflect other factors, such as underlying conditions or co‐morbid psychopathology

Key conclusions of study authors

  1. Based on Conners' Teachers Abbreviated Symptom Questionnaire; methylphenidate offers best combination of efficacy and tolerability for ADHD

  2. Clonidine, used alone or with methylphenidate, appears safe and well tolerated in childhood ADHD

  3. Study provides evidence that measures of quality of life for the family are sensitive to pharmacological treatment for ADHD

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: Moderate to severe adverse events were cited as a reason for withdrawal in 8 participants: methylphenidate + clonidine 5, clonidine 2, methylphenidate 1

Methylphenidate + clonidine: irritability (n = 1), tearfulness and irritability (n = 1), headaches (n = 1), itching (n = 1), asymptomatic ECG abnormalities (prolonged QTc) (n = 1) Methylphenidate: tachycardia and palpitations (n = 1)

Email correspondence with study authors: March 204 to June 2014. We attempted to obtain supplemental efficacy data from the study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation plan included stratification by centre (investigator) and by sexual maturity status (Prepubertal: Tanner I‐II, Pubertal: Tanner III‐V)

Allocation concealment (selection bias)

Low risk

Only the programmer in the Biostatistics Centre and the pharmacist knew the allocation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Methylphenidate (or matching placebo) powder packaged in gelatin capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All investigators, study co‐ordinators, teachers, parents and children were blinded to treatment assignments

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Statistical analyses were performed according to the ITT principle, and last available observations were carried forward and imputed when needed for both efficacy and safety measures. However, only data from the ADHD Rating Scale for children who completed the titration period were analysed

Selective reporting (reporting bias)

Low risk

Protocol published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

This project was supported by NIH (National Institutes of Health) and NINDS (National Institute of Neurological Disorders and Stroke)

Conflicts of interest: Some study authors are on the ADHD Advisory Board and the Speakers' Bureau of; are scientific consultants or principal or site investigators for; and/or have received educational or funding support from several pharmaceutical companies

Pearson 2013

Methods

Four‐week, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases

  1. 1‐week placebo

  2. 1‐week low‐dose methylphenidate

  3. 1‐week medium‐dose methylphenidate

  4. 1‐week high‐dose methylphenidate

Participants

Number of participants screened: 94

Number of participants included: 24 (19 boys, 5 girls). Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 24

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined 19, inattentive 5)

Age: mean 8.8 years (range 7.1 to 12.7)

IQ: mean 85 (range 46 to 112)

Methylphenidate naive: 13

Ethnicity: Caucasian (n = 13), African American (n = 4), Asian (n = 1), Hispanic (n = 5), other (n = 1)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 5), obsessive‐compulsive disorder (n = 2), separation anxiety disorder (n = 1)

Comedication: yes

Sociodemographics: Hollingshead 4 Factor Social Class 1.7 (0.9); Hollingshead 4 Factor SES Score 52.3 (10.8)

Inclusion criteria

  1. Not clearly stated

Exclusion criteria

  1. Serious neurological disorders (e.g. stroke, seizures)

  2. Down syndrome

  3. Fragile X syndrome

  4. Tourette's syndrome

  5. Psychosis

  6. Mood disorders

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of (low, medium and high) methylphenidate and placebo

Mean methylphenidate dosage: low = 0.21 mg/kg; medium = 0.35 mg/kg; high = 0.48 mg/kg

Administration schedule: b.i.d.

Duration of each medication condition: 1 week

Washout before study initiation: yes

Medication‐free period between interventions: not stated

Titration period: 1 week before randomisation

Treatment compliance: "Parents completed a medication administration form, were asked about missing or late doses at weekly interviews and teachers were asked as well. Forms were verified by number of pills in returned vials. Families were asked again in case of discrepancy. Parents and teachers were also asked about unanswered items om questionnaires"

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale ‐ Revised

  2. Conners' Teacher Rating Scale ‐ Revised

  3. Swanson, Nolan and Pelham Scale, Fourth Edition

  4. ADD/H Comprehensive Teacher Rating Scale (ACTeRS), Parent and Teacher Forms

Non‐serious adverse events

  1. Nine of 24 parents (38%) reported insomnia at the high dose, compared with 5 (21%) while taking placebo

  2. Nine parents reported loss of appetite at the high dose, compared with only 1 during placebo

Notes

Sample calculation: no

Ethics approval: yes

Key conclusions of study authors

  1. It is ideal for clinicians to assess behavioural response in both home and school settings when titrating methylphenidate treatment for children with ASD and significant ADHD symptoms

  2. It is important to monitor each child for side effects (e.g. increases in stereotypies or irritability)

  3. Methylphenidate formulations are efficacious and well tolerated in children with ASD and significant ADHD symptoms

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

We used the Digram balanced randomisation method that Dr. David Lane (a professor of both psychology and statistics at nearby Rice University) used to create the 8 balanced drug dose orders to which our 24 participants were assigned

Allocation concealment (selection bias)

Low risk

Central allocation

Blinding of participants and personnel (performance bias)
All outcomes

High risk

One‐week, single‐blind, "lead‐in‐dosing"; all study personnel with participant contact were blind with respect to dosages given during the drug trial

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The study medication was prepared by the University of Texas Psychiatry Research Pharmacy: the Ritalin LA beads were mixed with (inert) placebo beads and placed in two opaque gelatin capsules, and the white generic IR‐MPH was crushed and mixed with cornstarch and placed in two size 1 gelatin capsules"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Blank was left for missing data

Selection bias: yes; exclusion of placebo responder during single‐blind placebo washout week

Selective reporting (reporting bias)

Low risk

All outcomes are reported

Vested interest bias

Low risk

Funded by grant number MH072263 from National Institute of Mental Health (NIMH)

Conflicts of interest: none declared&&

Pelham 1989

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases

  1. No clear method description: "The procedure was double‐blind placebo‐controlled in which each child received, in random order with condition varied daily, placebo b.i.d. and 0.3 mg MPH/kg b.i.d." ‐ "5 to 9 days of data were gathered"

Participants

Number of participants screened: not stated

Number of participants included: 24. Participants were randomly assigned to 1 of 2 possible drug condition orders; condition varied daily: placebo b.i.d. and 0.3 mg MPH/kg b.i.d.

Number followed up: not stated

Number of withdrawals: not stated
Diagnosis of ADHD: DSM‐III (combined (not stated), hyperactive‐impulsive (19/24), inattentive (2/24))

Age: mean not reported (range: boys 5 years 6 months to 11 years; girls 5 years 8 months to 11 years 3 months)

IQ: boys 100.8 (SD 14.23), girls 104.0 (SD16.52)

Sex: 12 boys, 12 girls

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: attention deficit disorder (1/24), conduct disorder (5/24), oppositional defiant disorder (15/24), learning disability (6/25)

Comedication: yes; other doses of stimulants reported but not on study days

Sociodemographics: not stated

Inclusion criteria

  1. None stated

Exclusion criteria

  1. Children who were mentally retarded and those who had gross neurological disorders were not included in the study

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.3 mg/kg methylphenidate and placebo. "Each child received, in random order with condition varied daily, placebo b.i.d. and 0.3 mg MPH/kg b.i.d."
Mean methylphenidate dosage: boys 9.8 mg (range 5.3 to 16.9), girls 9.5 mg (range 5.2 to 13.1)

Administration schedule: at breakfast and at lunchtime

Duration of each medication condition: not stated

Washout before study initiation: not stated

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Abbreviated Conners’ Teacher Rating Scale (CTRS): 1 to 3 times per condition

  2. Revised Behaviour Problem Checklist: counsellor‐rated, 1 to 3 times per condition

  3. IOWA Conners' Teacher Rating Scale (IOWA CTRS)

General behaviour

  1. Daily frequencies (following rules, non‐compliance, positive peer behaviours, conduct problems, negative verbalisations, numbers of time‐outs per day): daily

  2. Time‐out: daily

  3. Classroom measures, teacher recorded: daily

  4. Daily report card

  5. Observed peer interaction using modification of RECESS code: daily

Notes

Sample calculation: no
Ethics approval: no information.

Comment from study authors

  1. Study involved only 12 boys and 12 girls; 1 dose of methylphenidate in the context of a highly structured summer programme: results must be considered preliminary

Key conclusions of study authors

  1. Results revealed equivalent and beneficial effects of methylphenidate for both boys and girls

  2. Methylphenidate therefore would appear to be a treatment that is as useful for girls with ADD as for boys with ADD

Comments from review authors

  1. Data reported from this study cannot be considered robust

  2. No information on ethics committee approval, on randomisation, on primary endpoint (multiple endpoints), on sample size calculation, on safety, etc. Concomitant behavioural treatment was provided, but no details are given in the Methods section

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no information
Any withdrawals due to adverse events: no information

Email correspondence with study authors: October 2014. We received no supplemental information/data from study authors. We asked authors whether this article was part of the Johnston 1988 study but received no response, so we extracted the data as from 2 separate studies

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Each child received, in random order with condition varied daily, placebo and methylphenidate

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Active medication and placebo were disguised in gelatin capsules and were pre‐packaged in individual, dated envelopes

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Unclear risk

Not stated

Pelham 1990a

Methods

Double‐blind, placebo‐controlled, cross‐over trial

  1. Immediate‐release methylphenidate

  2. Sustained‐release methylphenidate

  3. Sustained‐release dextroamphetamine

  4. Pemoline

  5. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 22 (all boys). Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: 22

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 10.39 years (range 8.08 to 13.17)

IQ: mean 105.68

Methlyphenidate‐naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (summer treatment programme)

Comorbidity: oppositional/defiant disorder (n = 9), conduct disorder (n = 4), "suggesting the presence of a learning disability", but IQ > 80 (n = 13)

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. Participating in the Summer Treatment Program of the 1988 Western Psychiatric Institute and Clinic Attention Deficit Disorder Program

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned different possible drug condition orders of methylphenidate (10 mg b.i.d., SR‐20 q.a.m.) and placebo

Mean methylphenidate dosage: immediate‐release methylphenidate 10 mg = 0.29 mg/kg

Administration schedule: twice daily: immediate‐release methylphenidate twice daily: morning and lunchtime; sustained‐release methylphenidate once daily: morning

Duration of each medication condition: 1 day, but in total 3 to 6 days

Washout before study initiation: none

Medication‐free period between interventions: not stated

Titration period: no

Treatment compliance: none

All completed study: not stated

Outcomes

ADHD symptoms

  1. Teacher ratings on the Abbreviated Conners' Teacher Rating Scale: 2 to 4 times in each medication condition

  2. Counsellor ratings on the Abbreviated Conners' Teacher Rating Scale: 2 to 4 times in each medication condition

Non‐serious adverse events

  1. Parent, teacher and counsellor side effect checklist: at least once per condition

Notes

Sample calculation: no

Ethics approval: not stated

Comments from study authors

  1. It should be noted that the results we have presented apply to the short‐term effects of these medications

  2. Results of this study might not predict long‐term response to the initial dose

Key conclusions of study authors

  1. The 3 long‐acting stimulants and immediate‐release methylphenidate were superior to placebo

  2. All 4 had similar time courses with effects from 1 to 9 hours after ingestion

  3. Individual differences in drug responsivity were noted

Comments from review authors

  1. The article states that comorbidity is diagnosed according to DSM‐III‐R, but study authors do not mention whether the ADHD diagnosis is based on DSM‐III‐R

  2. We have chosen in our data extraction to assume that this is so

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: January 2014. No contact made through author correspondence

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

Low risk

Active medication and placebo were disguised in gelatin capsules and were pre‐packaged in individual daily pill reminders

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: not stated

Pelham 1993a

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate 0.3 mg/kg and 0.6 mg/kg

  2. Placebo

Phases: 3

Participants

Number of participants screened: not stated

Number of participants included: 31. Participants were randomly assigned to 1 of 3 possible drug condition orders

Number of participants followed up: 31 (all boys)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (therefore no subtype)

Age: mean 98.8 months (range 5.42 to 9.92)

IQ: mean 110.7 (range not stated)

Methylphenidate naive: not stated

Ethnicity: Caucasian (93.5%), African American (6.5%)

Country: USA

Setting: hospital (Summer Treatment Program)

Comorbidity: oppositional defiant disorder (32%), conduct disorder (48%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criterion

  1. Boys with ADHD attending a Psychiatric Institute and Clinic Summer Treatment Program

Exclusion criteria

  1. None described

Interventions

Participants were randomly assigned to 1 (of not stated) possible drug condition order of 0.3 mg/kg and 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage (SD): low 8.1 mg (range 5 to 15); high 16.0 mg (range 10 to 22.5)

Administration schedule: twice a day morning and midday; conditions were changed daily over 6 weeks

Duration of each medication condition: 2 weeks overall (individual treatment condition 1 day)

Washout before study initiation: not described

Titration period: none

Treatment compliance: not reported

Outcomes

ADHD symptoms

  1. Teachers rated inattention/overactivity daily with the IOWA Conners' Teacher Rating Scale

Notes

Sample calculation: no

Ethics approval: not stated

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Relatively small incremental value was gained by the higher dose of medication or by the addition of behaviour modification compared with the effects of the low dose of methylphenidate

Email correspondence with study author: June 2014. Emailed study author to ask for additional information but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Referred to as a randomised study but sequence generation not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Capsules were identically packaged

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Referred to as double‐blind but not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Appears to be 100% follow‐up

Selective reporting (reporting bias)

Unclear risk

Protocol not identified

Vested interest bias

Unclear risk

None reported

Conflicts of interest: no information

Pelham 1999

Methods

Six‐week, within‐participant, double‐blind, placebo‐controlled, cross‐over design

  1. Methylphenidate

  2. Placebo

  3. D‐ and L‐AMPH

Phases: 5

Participants

Number of participants screened: 26

Number of participants included: 25 (21 boys, 4 girls). Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 23

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.6 years (range 5.8 to 12.7)

IQ: average intelligence

Methylphenidate naive: not stated

Ethnicity: Caucasian (88%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 13), conduct disorder (n = 8)

Comedication: not stated

Sociodemographics: "Median family income was US $40 000, with incomes ranging widely (from US $10 000 per year to US $100 000 per year)"

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 10 mg or 17.5 mg methylphenidate and placebo

Administration schedule: 2 time points

Duration of each medication condition: 5‐day period

Washout before study initiation: not stated

Medication‐free period between interventions: no

Titration period: none initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale (parent)

  2. IOWA Conners' Rating Scale (teacher)

Non‐serious adverse events

  1. Tics

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusions of study authors

  1. "Both drugs were routinely superior to placebo and produced dramatic improvements in rates of negative behaviour, academic productivity, and staff/parent ratings of behaviour"

  2. DEX‐AMPH AMPH produced greater improvement than methylphenidate. Doses of DEX‐AMPH AMPH used were more potent than those of methylphenidate

  3. Both drugs produced low levels of side effects. 25% of study participants were judged by the clinical team to be non‐responders

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes; 1 (exacerbation of tic disorder)

Email correspondence with study authors: April 2014. We requested additional information from study authors but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomized"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"The clinical team was not blind to medication condition when making their recommendations. Therefore, as a reliability check of the clinical team’s recommendations, one of the authors (J.W.) who was not involved in the clinical team meetings made independent recommendations based on the same data given to the clinical team. This rater was blind to drug condition except placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"The clinical team was not blind to medication condition when making their recommendations. Therefore, as a reliability check of the clinical team’s recommendations, one of the authors (J.W.) who was not involved in the clinical team meetings made independent recommendations based on the same data given to the clinical team. This rater was blind to drug condition except placebo"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

Grants from the Shire Richwood Pharmaceutical Company and National Institute of Mental Health (Grants MH53554, MH45576 and MH50467)

Pelham 2001a

Methods

Cross‐over trial with 3 interventions

  1. Concerta

  2. Immediate‐release methylphenidate

  3. Placebo

Phases: 3

Participants

Number of participants screened: 70

Number of participants included: 68. Participants were randomly assigned to 1 possible drug condition order

Number of participants followed up: 68 (66 for ADHD symptoms)

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.1 years (range 6 to 12)

IQ: mean 104.8

Sex: 89% boys, 11% girls

Methylphenidate naive: 0%

Ethnicity: Caucasian (94%), other (6%)

Country: USA

Setting: out‐patient clinic (Summer Treatment Program)

Comorbidity: oppositional defiant disorder (43%), conduct disorder (37%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. Children 6 to 12 years of age with ADHD were recruited via several sources, including advertisement; physician, agency and school referral; and parent referral

  2. All participants were required to be medicated with methylphenidate and to receive a stable dose for ≥ 4 weeks before the start of the study

Exclusion criteria

  1. Medical condition that would contraindicate the use of stimulants

  2. Physical condition or severe learning difficulty that would interfere with participation in study, including IQ < 80

  3. Receiving additional medication for ADHD

  4. Any medication with CNS effects, anticonvulsants or investigational medications

  5. Reached menarche

  6. Blood pressure at or above the 95th age and height percentile

Interventions

Participants were randomly assigned to different possible drug condition orders of immediate‐release methylphenidate 3 times a day (t.i.d.); Concerta methylphenidate once a day (qd); and placebo

Mean methylphenidate dosage: 0.75 mg/kg (SD 0.34) (Three dosing levels were used: 5 mg immediate‐release methylphenidate t.i.d./18 mg Concerta q.d.; 10 mg immediate‐release methylphenidate t.i.d./36 mg Concerta q.d.; and 15 mg immediate‐release methylphenidate t.i.d./54 mg Concerta q.d. The dose level used for each child was based on that child’s methylphenidate dosing before the start of the study

Administration schedule: immediate‐release methylphenidate 3 times a day; Concerta once daily

Duration of each medication condition: 7 days

Washout before study initiation: not described

Titration period: none

Treatment compliance: "virtually 100%"

Outcomes

ADHD symptoms

  1. Teachers and parents completed weekly symptom ratings using the IOWA Conners’ Rating Scale

  2. Teachers and parents rated oppositional defiant behaviour weekly using the Abbreviated Conners' Rating Scale

  3. Teacher ratings on the Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale, daily

Non‐serious adverse events

  1. Reports of adverse events were collected via spontaneous reports over the course of the study. Additionally, each week, parents provided responses to questions on adverse events, sleep quality, appetite and tics

Notes

Sample calculation: no

Ethics approval: yes

Comment from study authors

  1. All participants also received a behavioural intervention

Key conclusion of study authors

  1. This investigation clearly supports the efficacy of the Concerta long‐acting formulation of methylphenidate for parents who wish to have medication benefits for their child throughout the day and early evening

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; all participants were required to be medicated with methylphenidate and were receiving a stable dose for ≥ 4 weeks before the start of the study

Email correspondence with study author: June 2014. Emailed study author to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Referred to as double‐blind; capsules were identical

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Referred to as double‐blind; capsules were identical

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Data reported for 66/70 for ADHD and behaviour

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

High risk

Research was supported by ALZA Corporation, the manufacturers of Concerta

Conflicts of interest: Dr. Pelham is a member of the ALZA advisory committee on Concerta and its development. Drs. Hoffman and Lock are members of the ALZA paediatric advisory board

Pelham 2002

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 136 (all boys) (110 for 30‐day follow‐up). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 136 (106 for 30‐day follow‐up)

Number of withdrawals: 0 (4 for 30‐day follow‐up)

Diagnosis of ADHD: DSM‐III‐R (therefore no subtype)

Age: mean 9.7 years (range 7.6 to 12.7)

IQ: mean 104.5

Methylphenidate naive: not stated

Ethnicity: Caucasian (81%), African American (15%)

Country: USA

Setting: Summer Treatment Program

Comorbidity: oppositional defiant disorder (53%), conduct disorder (24%)

Comedication: not stated

Sociodemographics: median family income: US $25,000, (range US $10,000 to > $100,000)

Inclusion criterion

  1. Boys attending a psychiatric institute and clinic intensive Summer Treatment Program over 8 weeks

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 possible drug condition order of 0.3 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: 10 mg (SD 2.7)

Administration schedule: twice daily at 7:45 AM and 11:45 AM

Duration of each medication condition programme: 12 days (order randomly assigned daily over 6 weeks, doses administered over weekdays, except on Fridays); follow‐up: 30 days

Washout before study initiation: 2 weeks medication free baseline in programme, unclear for follow‐up

Titration period: none

Treatment compliance: not reported

Outcomes

ADHD symptoms

  1. Programme: daily ratings of inattention/hyperactivity using IOWA Conners' Rating Scale completed by counsellor

  2. Follow‐up: daily ratings of inattention/hyperactivity using IOWA Conners' Rating Scale completed by teacher

General behaviour

  1. Programme: daily ratings of oppositional defiant behaviour using IOWA Conners' Rating Scale completed by counsellor

  2. Follow‐up: daily ratings of oppositional defiant behaviour using IOWA Conners' Rating Scale, completed by teacher

Notes

Sample calculation: no

Ethics approval: yes

Comment from study authors

  1. Boys were told 50% of the time whether they were receiving placebo or medication and incorrectly 50% of the time; all boys received behavioural interventions over the course of the programme

Key conclusion of study authors

  1. Expectancy (of treatment effectiveness) did not improve behaviour; only active medication improved behaviour

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: June 2014. Emailed study authors to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Capsules were identical

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Raters were blinded to medication condition

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No loss to follow‐up apparent during programme but data reported for 106/110 followed up in the community

Selective reporting (reporting bias)

Unclear risk

Although measures were rated daily, how data were aggregated/reported as a single result per phase is not clear

Vested interest bias

High risk

Supported by NIMH (Grant MH48157)

Conflicts of interest: Pelham served as an advisor for ALZA Corporation (see Pelham 2001a)

Pelham 2005

Methods

Eight‐day, multi‐centre, double‐blind, randomised, dose‐ranging, cross‐over trial with 2 interventions

  1. Methylphenidate transdermal system

  2. Placebo

From a Summer Treatment Program

Participants

Number of participants screened: 36

Number of participants included: 36 (33 boys, 3 girls). Participants were randomly assigned to 1 of 8 possible drug conditions

Number of participants followed up: 36

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.6 years (range 6 to 13)

IQ: mean 105.3 (SD 18)

Stimulant‐naive: 15 (42%)

Ethnicity: Caucasian (75%), African American (19%), Hispanic (3%), mixed African American/White (3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no; antidepressants were withdrawn from 2 participants before study enrolment

Sociodemographics: Parents were manual workers (6%), clerical/sales workers (32%), technicians/semi professionals (23%) and executives/major professionals (32%)

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Medical history prohibiting patients from taking stimulants or participating

  2. Involvement in Summer Treatment Program activities

  3. Skin problems or allergies to ingredients in the patches

Interventions

Participants were randomly assigned to 1 of 8 possible drug conditions of methylphenidate transdermal system and placebo

Methylphenidate transdermal system dosage: 6.25 cm² (0.45 mg/h), 12.5 cm² (0.9 mg/h), 25 cm² (1.8 mg/h)

Administration schedule: 1×/d (at 6:00 AM and 7:00 AM). Application sites were alternated each day between left and right hips

Duration of each medication condition (crossed with time of application): 1‐day; patch worn for ≥ 12 hours/d

Duration of study: 8 days

Washout before study initiation: no

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: 100%. Parents returned patches and dosing records to the study site

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale and Abbreviated Conners' Rating Scale were rated daily by parents, counsellors and teachers

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale completed daily by parents, counsellors and teachers

  2. Any other adverse events that the child experienced were recorded

  3. Skin irritation at application sites rated each day by parents for skin reactions or irritations before application, before removal and the following morning. Presence of erythema: none (0), very slight (1), well defined (2), moderate (3) or severe (4). Presence of discomfort: none (0), mild (1), moderate but tolerable (2) or severe (3)

Notes

Sample calculation: yes (36 to 48)

Ethics approval: yes; institutional review board at each site approved the study

Comments from study authors

  1. No children experienced a skin reaction severe enough that the study physician recommended discontinuing the patch

  2. Limitations: short study duration, no controlled time‐course evaluation

Key conclusions of study authors

  1. Methyphenidate transdermal system produced significant effects that were similar to those previously reported with comparable methylphenidate doses

  2. Substantial effect of application time on total daily functioning not apparent in this setting; additional controlled time‐course studies will be necessary to fully evaluate the question of morning onset

  3. Further study will be necessary to establish long‐term efficacy and safety of the methylphenidate transdermal system

Comment from review authors

  1. Families received monetary compensation to participate

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: February to March 2014. We attempted to obtain supplemental information regarding randomisation, allocation concealment, washout period and efficacy and safety data from study authors but without success

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study sponsor produced random orders and prepared medication kits in numbered containers for each participant

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Treatment sequences were concealed until completion of the study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Treatment sequences were concealed until completion of the study

Incomplete outcome data (attrition bias)
All outcomes

High risk

Evaluable participant data were analysed. As the result of record‐keeping difficulties at 1 site, efficacy data were excluded for 5 participants

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

High risk

Study was supported by Noven Pharmaceuticals. Furthermore, Dr. Pelham was supported by grants from NIAAA, NIDA, NIMH and NINDS

Conflicts of interest: Several study authors have received consulting fees and research funding and have been consultants and/or served on the Speakers' Bureaus of several pharmaceutical companies in the past year

Pelham 2011

Methods

Three‐week, randomised, double‐blind, double‐dummy, cross‐over trial with 3 interventions

  1. Immediate‐release methylphenidate

  2. Methylphenidate transdermal system

  3. Placebo

Parents and children spent Friday evening to Sunday morning in a laboratory setting, where data collection took place

Participants

Number of participants screened: not stated

Number of participants included: 10 (all boys). Participants were randomly assigned to different drug orders of immediate‐release methylphenidate, methylphenidate transdermal system and placebo

Number of participants followed up: 9

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (combined (80%), hyperactive‐impulsive (0%), inattentive (20%))

Age: mean 8.6 years (range 6.4 to 9.7)

IQ: mean 95.3 (range 83 to 109)

Methylphenidate naive: none; all receiving a stable dose of immediate‐release methylphenidate before enrolment

Ethnicity: Caucasian (50%), African American (20%), Asian (0%), Hispanic (0%), Native American (10%), other (20%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder/conduct disorder (80%)

Comedication: no other psychotropics

Sociodemographics: not stated

Inclusion criteria

  1. ADHD diagnosis according to DSM‐IV

  2. IQ > 80

Exclusion criteria

  1. Skin sensitivity or any significant dermatological disease

  2. Atypical electrocardiogram results or hypertension

  3. Atypical blood and urine test results or evidence of other medical condition that could be worsened by stimulant usage

  4. Participants taking other psychotropics besides methylphenidate

  5. Participants with psychopathology other than ADHD or oppositional defiant disorder or conduct disorder severe enough to merit additional treatment

Interventions

Participants were randomly assigned to immediate‐release methylphenidate, methylphenidate transdermal system or placebo

Immediate‐release methylphenidate

Mean dosage: 30 mg/24 h

Administration schedule: 10 mg t.i.d. morning, lunch, afternoon

Methylphenidate transdermal system

Mean dosage: 33 mg/24 h

Administration schedule: two 10 cm² methylphenidate transdermal system patches worn for 24 hours at the buttock, with sides alternated daily

Time point: applied in the morning

Duration of each medication condition: 1 week

Washout before study initiation: 48 hours before study (no washout between treatment periods)

Titration period: none

Treatment compliance: Dosing and adhesion records were collected each week, but no data were available in the article

Outcomes

ADHD symptoms

  1. Iowa Conners' Scale, rated by the laboratory classroom teacher at each treatment condition

Non‐serious adverse events

  1. Vital signs (temperature, weight, blood pressure, pulse) were measured Friday night by staff. Furthermore, vital signs were taken at 0, 4, 8 and 24 hours after the first dose on Saturdays

  2. Adverse events were measured Friday night by staff and during weekdays by parents, and were reported spontaneously on Saturday evening and Sunday morning

  3. Skin was examined Friday night by staff. Furthermore, nurses assessed each methylphenidate transdermal system application for skin irritation during the laboratory day before application and at 0.5, 12 and 24 hours after removal

  4. Sleep: Parents rated the child’s sleep during weekdays. On Friday and Saturday nights in the laboratory setting, nursing staff monitored children hourly between 9:00 PM and midnight and recorded sleep onset

  5. Appetite: Parents rated this during weekdays

Notes

Sample calculation: no

Ethics approval: yes

Key conclusion of study authors

  1. Methylphenidate transdermal system demonstrates efficacy and tolerability comparable with t.i.d. immediate‐release methylphenidate

Comments from study authors

  1. All participants had been receiving methylphenidate previously, and this may explain the small number of side effects

  2. Furthermore, tolerability findings cannot be generalised to stimulant‐naive children

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no; however, all participants had been receiving methylphenidate previously, and this may explain the small number of side effects

Email correspondence with study authors: July 2014. Emailed study authors to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random assignment; no information about how

Allocation concealment (selection bias)

Low risk

Double‐dummy procedure: All children received patches and capsules each day, with applicable placebos

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

Low risk

One participant discontinued the study during the first week because of worsening behaviour while taking placebo and was not included in the analyses: all other participants completed the study

Adverse events: All participants receiving ≥ 1 dose of medication were included in the safety analysis

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

Not possible to find the protocol

Vested interest bias

High risk

Sponsored by a grant from Noven Pharmaceuticals (manufacturer of the MTS)

Conflicts of interest: Dr. Pelham has served as a consultant for Shire, McNeil, Noven, Celltech/Medeva, Novartis and Abbott Laboratories; has received honoraria from Shire and Janssen and research support from Shire, Alza, Eli Lilly, Noven and Cephalon; and holds common stock in Abbott Laboratories. Dr. Waxmonsky has served on the Speakers' Bureau for Novartis and has received research support from Eli Lilly and Shire Incorporated. Dr. Hoffman has served on the advisory board and Speakers’ Bureau for Shire Pharmaceuticals and on the Speakers' Bureau for McNeil. Dr. Ballow has received research support from GlaxoSmithKline, Panacos, Boehringer Ingelheim, Pharmasset, Jacobus and Pharmena. Dr. Schentag has served as a consultant for or received support from Noven, Wyeth, Daiichi, Targanta Therapeutics and Astellas. Dr. Gonzalez is a full‐time employee of P’Kinetics International Incorporated. No other conflicts of interest are known

Pelham 2014

Methods

Three‐week, randomised, controlled, cross‐over trial with 2 factors ‐ medication and behavioural intervention ‐ with daily medication changes

  1. Methylphenidate

  2. Placebo

  3. Behavioural treatment

Participants

Number of participants screened: not stated

Number of participants included: 48 (44 boys, 4 girls). Participants were randomly assigned to 1 of 3 different doses of drug condition orders

Number of participants followed up: 47

Number of withdrawals: 1
Diagnosis of ADHD: DSM‐VI (subtypes not stated)

Age: mean 9.35 years (range 5 to 12)

IQ: mean 106.33 (SD 14.61; range not stated)

Methylphenidate naive: not stated

Ethnicity: Caucasian (79%), African American (12.5%)

Country: USA

Setting: Summer Treatment Program

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV ADHD

  2. IQ ≥ 80

  3. No documented adverse response to, or medical conditions that would contraindicate use of, methylphenidate

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of 0.15 mg/kg/dose methylphenidate t.i.d.; 0.3 mg/kg/dose methylphenidate t.i.d.; 0.6 mg/kg/dose methylphenidate t.i.d. and placebo

Mean methylphenidate dosage: not clearly stated: "average doses were 5.4 mg (range = 2.5 – 10), 11 mg (range = 6.25 – 20), and 21 mg (range = 11.25 – 30), respectively"

Administration schedule: 3 time points

Duration of each medication condition: 0.15 mg dose for 4 days, 0.30 mg dose for 4 days and 0.6 mg dose for 3 days, switched daily

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: not stated.

Treatment compliance: "One child’s parents withdrew from the study after 2 days because of their concerns about possible side effects of the medication"

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale: rated by counsellors, daily

General behaviour

  1. IOWA Conners' Rating Scale O/D: rated by counsellors, daily

Non‐serious adverse events

  1. Counsellors completed the Pittsburgh Side Effects Rating Scale, daily

  2. Study staff monitored ratings for clinically significant adverse events, daily

Notes

Sample calculation: no
Ethics approval: yes

Comments from study authors

  1. The prototypic child with ADHD could be treated with the equivalent of 0.15 mg/kg methylphenidate (5 mg per dose in the current sample) twice daily—a dose lower than that used in studies of stimulant treatment in the past 30 years—if he or she is receiving moderate‐ to high‐intensity behavioural treatment

  2. Our data show that stimulant doses can be reduced dramatically if a child is treated with behaviour modification

Key conclusion of study authors

  1. "Results illustrate the importance of taking dosage/intensity into account when evaluating combined treatments; there were no benefits of combined treatments when the dosage of either treatment was high but combination of the low‐dose treatments produced substantial incremental improvement over unimodal treatment"

Comment from review authors

  1. Very difficult to understand the real effect of medication because of daily oscillation of the dose

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; only participants not having any documented adverse response to methylphenidate were included
Any withdrawals due to adverse events: yes (n = 1)
Email correspondence with study authors: April 2015. We emailed study authors to request supplemental information but have not received a response

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

High risk

"The children, their parents, and clinical staff members were uninformed of medication condition and only the research coordinator, pharmacist and medical director had access to the medication order. The medical director could reveal medication conditions in cases of severe side‐effect reports"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Observers were independent staff members who were not involved in the children’s treatment"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No access to protocol. No description in clinicaltrials.gov

Vested interest bias

Low risk

Funded by a grant from the National Institute of Mental Health (MH62946). Dr. Pelham was funded by grants from the National Institutes of Health (MH62946, MH69614, MH53554, MH69434, MH65899, MH78051, MH062946, NS39087, AA11873, DA12414, HD42080) and the Institute of Education Sciences (L03000665A). Dr. Fabiano was supported in part by a Ruth S. Kirschstein National Research Service Award Predoctoral Fellowship (1F31MH064243‐01A1) and by the Department of Education, Institute of Education Sciences (R324J06024, R324B06045)

Conflicts of interest: not declared

Perez‐Alvarez 2009

Methods

Twelve‐month, randomised, controlled, parallel study with 2 different treatment strategies

A) Participants with a score ≧ 2.5/1.8 on the Swanson, Nolan and Pelham Scale, Fourth Edition (teacher/parents), were randomly assigned to

  1. Concerta

  2. Humanistic psychology

  3. Concerta + psychology

B) Participants with score < 2.5/1.8 on the Swanson, Nolan and Pelham Scale, Fourth Edition, were randomly assigned to

  1. Humanistic psychology

  2. Concerta + psychology

Participants

Number of participants screened: not stated

Number of participants included: 150

Number randomly assigned: Concerta + psychology 59, humanistic psychology 59

Number followed up: Concerta + psychology 59, humanistic psychology 59

Number of withdrawals: Concerta + psychology 0, humanistic psychology 0

Diagnosis of ADHD: DSM‐IV‐TR (combined (67%), hyperactive‐impulsive (0%), inattentive 33%))

Age: mean 10 years (range 7 to 14)

Sex: boy:girl 4:1 for those with combined type; 1:3 for those with inattentive type

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Spain

Setting: out‐patient clinic

Comorbidity: no

Comedication: no

IQ: > 70

Sociodemographics: not stated

Inclusion criteria

  1. ADHD DSM‐IV‐TR, combined or inattentive type

  2. 7 to 15 years

  3. IQ > 70

Exclusion criteria

  1. Previous medication or therapy

  2. Comorbidity

Interventions

Participants were randomly assigned to Concerta + psychology or humanistic psychology

OROS methylphenidate dose: not stated

Administration schedule: not stated

Duration of intervention: 12 months

Titration period: not stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanons, Nolan and Pelham, Fourth Edition (18‐item): rated at baseline, 6 months and 12 months by teachers and parents

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Key conclusion of study authors

  1. In summary, while further confirmation is awaited, the cognitive PASS assessment may be a useful tool for better diagnostic and prognostic classification in comparison with behavioural phenotyping

Comment from review authors

  1. In study methods and data extraction, all participants receiving Concerta + psychology (3A + 2B) were analysed together, as were all participants receiving humanistic psychology (2A + 1B)

Email correspondence with study authors: June 2014. We received supplemental information regarding funding, ethics approval and protocol from the study authors, although they were not able to send data from the Swanson, Nolan and Pelham Scale, Fourth Edition

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were randomly assigned, but no information was given to explain how assignment was done

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No; all planned analyses are described in the paper

Vested interest bias

Low risk

Study was not funded. Research was part of the workday, participants were voluntary and no funding was needed to implement the study

Conflicts of interest: none. Investigators are staff members at institutions (affiliations) reported in the paper

Pliszka 1990

Methods

Four‐week, double‐blind, cross‐over trial, in which children were randomly assigned to the following

  1. Two doses of methylphenidate (low: 0.25 mg/kg to 0.40 mg/kg; and high: 0.45 mg/kg to 0.70 mg/kg)

  2. Placebo

Participants

Number of participants screened: 79

Number of participants included: 46

Number of participants followed up: 43 (30 without anxiety, 13 with anxiety)

Number of withdrawals: 3. Participants were randomly assigned to different orders of methylphenidate low dose, methylphenidate high dose or placebo

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 9 years (range not reported)

IQ: > 70

Sex: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: anxiety (30%), oppositional defiant disorder (56.0%), conduct disorder (23.3%) (Participants who expressed transient anxiety or depression about consequences of punishment for misbehaviour were considered to not meet the criteria for an overanxious disorder)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐III‐R criteria for ADHD

  2. Candidate for a stimulant trial

Exclusion criteria

  1. None stated

No patients met the criteria for any psychotic or depressive disorder. Participants were free of medication, as well as any other medical disorder

Interventions

A total of 4 weeks of medication. First week always placebo; remaining 3 weeks, participant were randomly assigned to different orders of placebo and low‐ (0.25 mg/kg to 0.40 mg/kg) and high‐dose (0.45 mg/kg to 0.70 mg/kg) methylphenidate. Doses were as follows: 5 mg and 10 mg for weight < 25 kg; for those overweight, the 2 dose conditions were 10 mg and 20 mg

Administration schedule: b.i.d., 7:00 AM and 12 noon. Saturday, the dose was adjusted so that it was given 90 minutes before measurements were taken

Duration of each medication condition: 1 week

Washout before study initiation: not relevant; did not take methylphenidate before

Medication‐free period between interventions: 17 hours

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA Conners' Teacher Rating Scale (both Inattention/Overactivity and Aggressive subscales): rated weekly by teachers at the end of each week

Notes

Sample calculation: no information

Ethics approval: no information

Comments from study authors

  1. Data strongly suggest that children who met criteria for ADHD and anxiety are not children who simply have become "demoralised" by frequent conflicts with parents and teachers. They represent a distinct subgroup

  2. It is unclear from these data whether children with comorbid ADHD and anxiety form a separate subtype of ADHD, similar to the DSM‐III diagnosis of ADD without hyperactivity, or whether this is a group of children with primary anxiety that may lead to oppositional behaviour and temper tantrums, and the presence of these symptoms may lead parents and teachers to report inattention and overactivity when the symptoms may not be objectively present

  3. This study strongly suggests that it is important to control for the presence of anxiety disorders in research on ADHD

  4. Analyses were made on the basis of whether anxiety was present with ADHD. To meet criteria for anxiety, children had to report the symptoms themselves

Key conclusions of study authors

  1. A total of 43 participants completed a double‐blind trial of methylphenidate; participants with comorbid anxiety had a significantly poorer response

  2. Results suggest that ADHD with comorbid anxiety may characterise children with primary anxiety who develop secondary inattentiveness, or may represent a different subtype of ADHD, perhaps similar to the condition of attention deficit disorder without hyperactivity, as under DSM‐III

  3. ADHD participants with anxiety were rated significantly lower on the IOWA Conners' Teacher Rating Scale, Inattention/Overactivity subscale, than those without anxiety, although the mean score for both groups was well into the disturbed range

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: November 2013. We received from the study authors supplemental information regarding participants' intellectual function and funding and additional data. The raw data no longer exist; therefore we cannot analyse data from different periods in the cross‐over trial, only endpoint data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned to placebo and low‐dose and high‐dose methylphenidate

Allocation concealment (selection bias)

Unclear risk

Randomly assigned to placebo and low‐dose and high‐dose methylphenidate. Not stated how investigators allocated participants to the intervention

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The child, his or her parents, teachers and research assistants were all blinded to drug status

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The child, his or her parents, teachers and research assistants were all blinded to drug status

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Only 3 drop‐outs. All other participants were included in the final analysis

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

The entire outcome as stated in the methods was reported in the results

Vested interest bias

Low risk

National Institute of Mental Health (NIMH)

Conflicts of interest: not declared

Pliszka 2000

Methods

Three‐week, randomised, double‐blind, placebo‐controlled, parallel trial with 3 arms

  1. Adderall

  2. Methylphenidate

  3. Placebo

Participants

Number of participants screened: 73

Number of participants included: 58

Number randomly assigned: methylphenidate 20, placebo 18

Number followed up: methylphenidate 19, placebo 16

Number of withdrawals: methylphenidate 1, placebo 2

Diagnosis of ADHD: DSM‐IV. Diagnostic Interview Schedule for Children diagnosis of ADHD (subtype not stated)

Age: mean 9 years (range not reported)

IQ: > 75

Sex: not stated

Methylphenidate naive: methylphenidate 5 (25%), placebo 1 (6%)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (methylphenidate 14%, placebo 10%), conduct disorder (methylphenidate 1%, placebo 2%), anxiety disorder (methylphenidate 20%, placebo 5%)

Comedication: not stated

Sociodemographics: not stated. No significant difference in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Children in grades 1 to 5

  2. ADHD according to Diagnostic Interview Schedule for Children

  3. IQ > 75

  4. ≥ 1.5 SD above the mean for his/her age and sex on the IOWA Conners' Teacher Rating Scale, Inattention/Overactivity factor

Exclusion criteria

  1. Other medical illness

  2. Meeting Diagnostic Interview Schedule for Children criteria for major depression episode, manic episode or tic disorder

  3. History of psychosis or signs of psychosis or significantly depressed mood on the mental status examination

  4. Current treatment consists of non‐stimulant psychotropic medication

Interventions

Participants were randomly assigned to immediate‐release methylphenidate or placebo

Mean methylphenidate dosage: total dose 25.2 mg/d

Administration schedule: 17 (85%) received 2 or more doses

Time points: 1 to 3 times a day: morning, after school and additional noon if needed

Duration of intervention: 3 weeks

Titration period: none

Treatment compliance: not stated. Dosage was adjusted at the end of weeks 1 and 2 via an algorithm based on teacher and parent ratings

Outcomes

ADHD symptoms

  1. IOWA Conners' Teacher Rating Scale: rated twice daily (morning and afternoon) Mondays through Thursdays

General behaviour

  1. Conners' Global Index: parent‐rated

Non‐serious adverse events

  1. Multi‐Modality Treatment of ADHD (MTA) side effects scale: parent‐rated weekly (Thursday evenings)

Notes

Sample calculation: no

Ethics approval: no

Comment from study authors

  1. Mean mg/kg dose for methylphenidate non‐responders was 0.43, which is less than the 0.3 mg/kg to 0.8 mg/kg dose known to be required for adequate response in some children with ADHD

Key conclusion of study authors

  1. Both medications were superior to placebo for reducing inattentive and oppositional symptoms in the classroom and on the Conners' Global Index

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: January 2014. Supplemental information received from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned: "We used a random number generator to determine which of the 3 groups the child was assigned to"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, children, teachers and treating physicians were blind to medication status. Medication was crushed, mixed with a blue food powder and placed in opaque capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Placebo participants were randomly assigned to follow methylphenidate or Adderall treatment algorithm. The blinded psychiatrist could not determine the child’s medication status simply by knowing which algorithm was being followed. Principal Investigator knew the medication status of participants

Incomplete outcome data (attrition bias)
All outcomes

High risk

Morning and afternoon IOWA scores were averaged at the end of the week

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

All planned outcomes were reported

Vested interest bias

High risk

Research funded by Shire Richwood Incorporated

Conflicts of interest: Dr. Browne is currently with Watson Pharmaceuticals, Corona, California

Pliszka 2007

Methods

Five‐week double‐blind, cross‐over, placebo‐controlled trial, conducted to examine electrophysiological effects of methylphenidate on inhibitory control in children with ADHD

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 12

Number of participants included: 12 (8 boys, 4 girls). Participants were randomly assigned to 1 of (not stated) possible drug condition orders

Number of participants followed up: 12

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (100%))

Age: mean 12.3 years (range 9 to 15)

IQ: not stated

Methylphenidate naive: 10

Ethnicity: Caucasian (67%), African American (8%), Hispanic (25%)

Country: USA

Setting: not stated

Comorbidity: oppositional defiant disorder (45%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD, combined type

  2. Conners' Global Index: restless/impulsive ratings ≥ 1.5 SD above the mean for child’s age and sex on both parent and teacher ratings

Exclusion criteria

  1. General Cognitive Ability on DIfferential Abilities Scale > 85

  2. Any substance abuse/dependence

  3. Any neurological disease

  4. Long‐term use of any medicine

  5. Learning disability

Interventions

Participants were randomly assigned to different possible drug condition orders of 5 mg, 10 mg, 15 mg, 20 mg methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 3 time points

Duration of each medication condition: 7 days

Washout before study initiation: not stated

Medication‐free period between interventions: 0

Titration period: none initiated before/after randomisation

Outcomes

General behaviour

  1. Parents and Teacher Conners' Global Index (CGI)

Notes

Sample calculation: no

Ethics approval: yes; study approved by the Institutional Review Board of the University

Comments from study authors

  1. Several limitations of the study should be noted

  2. Our sample was relatively small and was composed entirely of children with ADHD, combined type alone, and no other significant comorbidity other than oppositional defiant disorder

  3. All participants were positive responders to methylphenidate

  4. Our sample was too small to examine for effects of sex and ethnicity, and results should be replicated in a larger sample

Key conclusions of study authors

  1. Methylphenidate may improve inhibitory control by enhancing brain mechanisms that trigger the inhibitory process and make stopping a motor act more probable (reflected by increased N200) and by increasing attentional resources to the task when unsuccessful inhibitions occur (as reflected by increased NoGo‐P3)

  2. These results are consistent with functional imaging studies, suggesting a role for the right frontal inferior cortex and the cingulate cortex in the pathophysiology of ADHD

Comment from review authors

  1. Although this study includes only good responders to methylphenidate, this occurred by chance and not by design, as 10 out of 12 were treatment naive

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: May 2014. Study authors could not give us the necessary data (e.g. separate data for each intervention period); therefore we could not use Conners' Global Index data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Double‐blind, crossover, placebo‐controlled"

Allocation concealment (selection bias)

Low risk

From email: One investigator was assigned on the basis of chance; the other remained blinded

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind. One investigator assigned on the basis of chance; the other remained blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

One investigator assigned on the basis of chance; the other remained blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

Low risk

Funded by the National Institute of Mental Health Grant R01 MH63986

Conflicts of interest: Pliszka received honoraria and research support from Shire and MacNeil and research support from Ely Lilly and Cephalon

Quinn 2004

Methods

Cross‐over trial with 3 interventions

  1. Dex,l‐methylphenidate

  2. Dexmethylphenidate

  3. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 32. Participants were randomly assigned to possible drug condition orders

Number of participants followed up: 31 (all boys)

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (combined (87.1%), hyperactive‐impulsive (9.7%), inattentive (3.2%))

Age: not stated (range 9 to 12 years)

IQ: > 80

Methylphenidate naive: 0%

Ethnicity: not stated

Country: USA and Canada

Setting: out‐patient clinic

Comorbidity: (0%)

Comedication: 0% for other medications for ADHD

Sociodemographics: not stated

Inclusion criteria

  1. Male, between 9 and 12 years of age

  2. Meet DSM‐IV criteria for ADHD, as confirmed by the Diagnostic Interview Schedule for Children

  3. Clinical history of positive response to treatment with ≥ 20 mg/d of dex,l‐MPH for ≥ 1 month

  4. Rating > 90th percentile on Parent and Teacher Versions of the Swanson, Nolan and Pelham ADHD Rating Scale

  5. IQ ≥ 80 as assessed by a validated intelligence test

Exclusion criteria

  1. Any associated CNS, cardiovascular, renal or respiratory disorder

  2. Known sensitivity to methylphenidate or receiving other medication for ADHD

  3. Comorbid clinical disorders reported during clinical interview or identified during administration of the Diagnostic Interview Schedule for Children

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of dex,MPH, dex,I‐MPH and placebo. Both the order of drugs and the dose sequence were randomly assigned

Mean methylphenidate dosage: d‐MPH 2.5 mg, 5 mg or 10 mg; dex,l‐MPH 5 mg, 10 mg or 20 mg

Administration schedule: once at 8.30 AM

Duration of each medication condition: 1 day. Interventions were separated by ≥ 6 days

Washout before study initiation: 24 hours

Medication‐free period between interventions: ≥ 24 hours

Titration period: none

Treatment compliance: administered at the laboratory

Outcomes

ADHD symptoms

  1. Conners, Loney and Milich (CLAM) Scale: teacher‐rated, at 2 hours, 3.5 hours and 6 hours (CLAM aggressive/defiant, CLAM inattention/overactivity)

  2. Conners’ Hyperactivity Index: teacher‐rated, at 2 hours, 3.5 hours and 6 hours

Non‐serious adverse events

  1. Adverse events

  2. Average pulse rate also measured

Notes

Sample calculation: no

Ethics approval: approved by each centre’s institutional review board

Comments from study authors (limitations)

  1. Study was conducted in a laboratory school setting (no healthy participants, high level of staffing, short and repetitive classroom period compared with a regular classroom setting)

  2. Relatively small sample size

  3. Narrow age range

Key conclusions of study authors

  1. Efficacy of methylphenidate resides in the d‐isomer. Elimination of the i‐isomer does not diminish the efficacy of an acute dose of methylphenidate

  2. This study demonstrates that single low (2.5 mg), medium (5 mg) and high (10 mg) doses of dexmethylphenidate match the efficacy of equimolar single low (5 mg), medium (10 mg) and high (20 mg) doses of dex,l‐methylphenidate over a 6‐hour period, based on repeated measurements of a surrogate measure of academic performance

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; included only children stable on methylphenidate

Email correspondence with study authors: July to August 2014. We contacted study authors to obtain safety data and supplemental information regarding randomisation, allocation concealment, blinding, handling of incomplete outcome data and outcomes, but we were not successful

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised. Both the order of drugs and the dose sequence were randomly assigned

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Except for first practice day, on which only participants were blinded, administration of doses was double‐blinded throughout the study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind. Blinded observers rated behaviour

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Study authors report only 1 respondent (LTFU), not related to side effects

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

All outcomes reported. No protocol available

Vested interest bias

High risk

Study was funded by Celgene

Conflicts of interest: All study authors disclosed that they have past and present affiliations with the pharmaceutical industry

Ramtvedt 2013

Methods

Six‐week, cross‐over trial with 3 interventions

  1. Methylphenidate

  2. Placebo

  3. Dextroamphetamine

Phases: 3

Participants

Number of participants screened: not stated

Number of participants included: 36. Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 36 (29 boys, 7 girls) (n = 34 for adverse event data)

Number of withdrawals: 0
Diagnosis of ADHD: DSM‐IV‐TR (combined (69%), hyperactive‐impulsive (3%), inattentive (28%))

Age: mean 11.4 years (range 9 to 14)

IQ: mean 90.9

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Norway

Setting: out‐patient clinic

Comorbidity: anxiety/depressive disorder (n = 9, 25%), oppositional defiant disorder (n = 20, 55%), learning disability (n = 22, 61%) and Asperger syndrome (n = 1, 3%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. ADHD diagnosis rated as > 2 SD above the mean on the Conners’ Rating Scale, DSM‐IV Inattention and/or DSM‐IV Hyperactivity‐Impulsivity subscales

  2. Between 9 and 14 years of age

  3. No prior treatment with stimulants

  4. Stimulant treatment that has been approved by a paediatrician or psychiatrist

Exclusion criteria

  1. Moderate to severe mental retardation

  2. Psychosis

  3. Brain injury

  4. Sensory deficits, motor impairment or both

  5. Epilepsy

  6. Factors that would substantially reduce the possibility of obtaining reliable observations from a parent or teacher

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate (30 mg/d to 40 mg/d), dextroamphetamine (10 mg/d to 20 mg/d) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: methylphenidate, 3 time points (morning, lunch and afternoon); dextroamphetamine, 2 time points (morning and afternoon)

Duration of each medication condition: 2 weeks (first week: low dose (30 mg/d), second week: high dose (40 mg/d))

Washout before study initiation: no; all were stimulant‐naive

Medication‐free period between interventions: yes; Saturday and Sunday (48 hours)

Titration period: 2 weeks initiated after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. DSM‐IV Inattention and Hyperactive‐Impulsive subscales from Conners’ Rating Scale – Revised, Long Version, Parent and Teacher Forms

  2. 21‐item ADHD questionnaire was developed for this study (8 items reflecting inattention, 6 items reflecting hyperactive‐impulsive behaviour and 4 items reflecting oppositional defiant behaviour), completed daily from Monday to Friday, every week by parents and teachers (Ramtvedt 2013)

  3. Children's self‐report scale ‐ developed for this study: 8 items, rated weekly by the children themselves

General behaviour

  1. Child Behavior Checklist

  2. Teacher Rating Form

  3. ADHD questionnaire ‐ Inattentive subscale, teacher‐rated

Non‐serious adverse events

  1. Barkley Side Effect Rating Scale: Parents were instructed to rate side effects in co‐operation with their child at the end of each week during the trial

Notes

Sample calculation: not stated

Ethics approval: yes

Comments from study authors

  1. Drugs were not camouflaged in identical capsules, increasing the risk for identification of drug order

  2. In only 1 case, a parent identified the drug order with certainty. That particular child was removed from the study

  3. ADHD questionnaire, used to rate ADHD symptoms during the stimulant trial, was developed for this study and cannot be considered equivalent to well‐established ADHD Rating Scales

  4. Sample size might not have been sufficient to detect subtle differences between stimulants at the group level

Key conclusions of study authors

  1. Methylphenidate and dextroamphetamine were significantly effective. No significant superiority of 1 stimulant over the other was detected at the group level

  2. Adverse events associated with dextroamphetamine vs methylphenidate appear similar at the group level but may differ substantially in individual children

  3. Overall, insomnia and decreased appetite were significantly associated with stimulants

Comments from review authors

  1. This does not seem to be a particularly useful paper because the main point (not explicitly stated) was to obtain an equivalence for the QbTest (measures the 3 core signs of ADHD). As a result of this, the medication regimen was dedicated to find the best effects of both medications

  2. Study authors stated that the primary outcome measure was developed for the study and cannot be considered equivalent to known ADHD Rating Scales. Therefore, we have not used the data on ADHD symptoms in our analyses

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: April 2015. We obtained supplemental information or data regarding comedication, randomisation and blinding

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly and evenly assigned to each of 6 possible drug orders. "The children were assigned to the six possible drug condition orders by drawing numbers"

Allocation concealment (selection bias)

Unclear risk

Not clear

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Participants, parents, teachers and test administrators were blinded to drug order. One parent identified the drug order. Tablets of similar colours, shapes and textures were administered, but the drugs were not camouflaged in identical capsules

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

No access to protocol

Vested interest bias

High risk

First phase was conducted as part of ordinary clinical practice at Neuropsychiatric Unit, Østfold Hospital Trust. Second and third phases, data analysis and preparation of manuscript were sponsored by South‐Eastern Norway Regional Health Authority, and also by Østfold Hospital Trust and National Resource Centre for ADHD, both under the umbrella of South‐Eastern Norway Regional Health Authority

Conflicts of interest: Henning Aabech is a member of the Strattera Advisory Board, Eli Lilly Norway

Rapport 1985

Methods

Triple‐blind, randomised, cross‐over trial with 4 interventions

  1. Placebo

  2. 5 mg methylphenidate

  3. 10 mg methylphenidate

  4. 15 mg methylphenidate

Phases

  1. Week 1: baseline assessment

  2. Weeks 2 to 5: cross‐over trial

Participants

Number of participants screened: 22

Number of participants included: 14 (12 boys, 2 girls)

Number of participants followed up: 11

Number of withdrawals: 1

Number of participants excluded during the study: 2

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: 8.3/7.75 years (range 6 to 10)

IQ: > 100

Methylphenidate naive: not stated

Ethnicity: Caucasian (86%), not stated (14%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

Sociodemographics: Low to middle socioeconomic status

Inclusion criteria

  1. DSM‐III diagnosis of ADHD by the child's paediatrician and the clinic's directing clinical psychologist

  2. Maternal report of a developmental history consistent with ADD‐H (Barkley 1981)

  3. Maternal rating of ≥ 2 SD above the mean for the child's age on the Werry‐Weiss‐Peters Activity Scale

  4. Teacher rating > 15 on the Abbreviated Conners' Teacher Rating Scale

  5. Children showing a favourable response to methylphenidate (≥ 25% mean increase in classroom on‐task behaviour and decrease ≥ 2 SD on the Abbreviated Conners' Teacher Rating Scale (compared with baseline levels) during any of the active medication weeks)

  6. Performance on the Matching Familiar Figures Test characteristic of a "fast‐inaccurate or impulsive" responder (i.e. faster than average responses and higher than average error rates for the child`s age)

Exclusion criteria

  1. Any gross neurological, sensory or motor impairment

  2. Those currently taking medication

  3. Those classified as non‐responders, defined as neither improved (methylphenidate‐induced facilitation ≥ 25%) on the Paired Associates Learning test nor declined in relation to baseline and placebo

Interventions

Participants were randomly assigned to 1 of 24 possible drug condition orders of fixed doses of methylphenidate (5 mg, 10 mg, 15 mg) and placebo. As a result of the small sample size, not all drug orders were used. Post hoc comparison showed that doses were distributed approximately equally across different positions in the sequence

Administration schedule: once daily, in the morning

Duration of each medication condition: 7 days

Washout before study initiation: none, but all weekly dosage changes occurred on Saturdays to control for potential rebound effects

Titration period: none

Treatment compliance: Both used and unused envelopes with capsules were returned to control for medication compliance. No results regarding compliance were reported

Outcomes

ADHD symptoms

  1. Abbreviated Conners; Teacher Rating Scale: Teacher rated symptoms each Friday, in the morning, or at the end of each treatment condition

  2. Dosage changes occurred on Saturdays

No useable data

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusion of study authors

  1. Results of the present investigation demonstrate a functional relationship between psychoactive medication and impulsivity, attention and behaviour of children with ADD in classroom settings

Comment from review authors

  1. Although study authors refer in 1 of the articles to the other 2 articles as a recent study and a past investigation, it seems to us that these articles discuss the exact same study

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; included only methylphenidate‐favourable responders

Any withdrawals due to adverse events:no

Email correspondence with study authors: July 2013. Study authors informed us that original records were shredded after 20 years, and that they could not provide the requested data (Ramstad 2013c [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Order of drug administration was determined by randomly assigning each child to 1 of 24 possible drug disorders

Allocation concealment (selection bias)

Low risk

Methylphenidate was packaged by the pharmacist in coloured gelatin capsules to avoid detection of dose and taste. Capsules were placed in individually dated envelopes to ensure accurate dose administration

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Triple‐blind design

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Teachers and observers were blind to when medication was administered and what specific doses were given

Incomplete outcome data (attrition bias)
All outcomes

High risk

Data from 2 of 12 children in the present investigation were not included in the statistical analyses because they were classified as non‐responders according to results of the Paired Associates Learning test

Selective reporting (reporting bias)

Unclear risk

Not possible to receive a copy of the protocol

Vested interest bias

Unclear risk

No information regarding funding

Conflicts of interest: not declared

Rapport 1987

Methods

Double‐blind, placebo‐controlled, cross‐over trial with 2 interventions

  1. Methylphenidate (5 doses)

  2. Placebo

Phases: Five methylphenidate doses were given in a randomly assigned, counterbalanced sequence. Children received each dose for 6 consecutive days

Participants

Number of participants screened: 134. Children were screened for inclusion after referrals from paediatricians, psychiatrists and school personnel over a 5‐year period. Different publications reported different numbers of included children, ranging from 22 to 76 children. Study authors stated: "this is a sample of children who were recruited across several years with publications presenting findings over that time period. Thus, there were slight differences in sample composition as more participants were added. But essentially these studies included the same sample". Participants were randomly assigned to 1 of 4 possible drug condition orders. Different publications reported different figures for missing data, number of withdrawals and other important information. This was included in our 'Risk of bias' assessment and is reported in the 'Risk of bias' table. One‐day washout (Saturdays) was described in each publication

Age: 10 to 12 years. Reported ages varied across publications but, in most publications, ranged from 6 to 11 years

DSM‐III diagnosis of ADHD (subtype not stated)

IQ: mean 101. DuPaul 1993 stated that children were of average or above average intelligence

Sex: 37 boys and 5 girls (Rapport 1987)

Methylphenidate naive: 100% (in both Rapport 1987 + DuPaul 1993). In Rapport 1987, 8 had experienced brief trials of stimulants within the previous 4 years

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: No children were taking medication before participating in the study (DuPaul 1993 in Rapport 1987)

Sociodemographics: low to middle socioeconomic status (Hollingshead) (Rapport 1987 and DuPaul 1993 in Rapport 1987)

Inclusion criteria

  1. Independent diagnosis by the child's paediatrician and the Rhode Island Construction Leadership Council (CLC), directing clinical psychologists using DSM‐III criteria for ADHD

  2. Maternal report of a developmental history consistent with ADHD and problems in ≥ 50% of situations on the Home Situations Questionnaire

  3. Maternal rating ≥ 2 SD above the mean for the child's age on the Werry‐Weis‐Peters Activity Scale

  4. Teacher rating on the Abbreviated Conners' Teacher Rating Scale > 15 ‐ the designated cutoff score for hyperactivity

  5. Performance on the Matching Familiar Figures Test characteristic of a "fast inaccurate or impulsive" responder (i.e. faster than average responses and higher than average error rates for the child's age)

  6. Absence of any gross neurological, sensory or motor impairment as determined by paediatric examination

We believe that this study consists of 13 articles. When study authors were asked about that, Dr. DuPaul answered that many of the publications based on the Rhode Island Construction Leadership Council (CLC) Clinic "were fairly the same study". Even though some publications describe 6 inclusion criteria and others describe 5, we believe that the publications are based on the same study (Ramstad 2013b [pers comm])

Interventions

Participants were randomly assigned to 1 of 5 possible drug condition orders of methylphenidate and placebo. Methylphenidate was described by each child's paediatrician in the following doses: placebo, 5 mg, 10 mg, 15 mg and 20 mg. Fixed doses were prescribed (rather than mg/kg)

Administration schedule: Children were seen once a week at the Rhode Island Construction Leadership Council (CLC) Clinic. Baseline measures were obtained during the child's second clinic visit to allow familiarisation with clinic personnel and testing procedures. On subsequent testing days, children were administered a capsule of the active agent (methylphenidate) or placebo. This procedure continued until each child received each dose for 6 consecutive days. All weekly dosage changes occurred on Sundays, and no medication was administrated on Saturdays, to allow for needed washout due to inter‐individual variation in serum and blood plasma levels following acute administration of methylphenidate. All children described in the present study were classified as favourable responders, whereas those whose performance did not improve to this extent were classified as non‐responders. These criteria were established a priori. All children described in the present study were favourable responders based on the given criteria. (Children showing drug‐induced facilitation of performance ≥ 25% (i.e. a 25% drop in error rate) compared with baseline or placebo were classified as favourable responders, whereas those whose performance did not improve to this extent were classified as non‐responders)

Treatment compliance: Rapport 1987 and DuPaul 1993 (in Rapport 1987): Both used and unused envelopes were returned to the Rhode Island Construction Leadership Council (CLC) on a weekly basis to assess medication compliance. DuPaul 1993 and Denney 1999 (in Rapport 1987): Medication was properly administered nearly 100% of the time, with "make‐up" observation days scheduled after rare occasions when compliance was not obtained

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Teacher Rating Scale: once each week (1.5 to 2 hours after morning medication)

Non‐serious adverse events

  1. Data from the Subjective Treatment Emergent Symptom Scale (no data reported on this scale. Additional data cannot be obtained from study authors)

  2. Kelly 1988 (in Rapport 1987) measured heart rate. Each child’s resting heart rate (average beats per minute) was measured for 5‐minute periods at 3 intervals (immediately before oral ingestion of methylphenidate or placebo (IV), 120 minutes post ingestion and 180 minutes post ingestion). Each of these measurements was taken at the same relative time of day (between 2:00 PM and 6:00 PM) and on the same day of each week throughout the 6‐week course of the study

Notes

Sample calculation: no

Ethics approval: not stated. From Kelly 1988 in Rapport 1987: believe the study was approved by the Institutional Review Board IRB at the University of Rhode Island (the site of the study), but Dr. Rapport could confirm this (Ramstad 2013c [pers comm])

Key conclusions of study authors

  1. Group results showed significant medication effects on classroom percentage of on‐task behaviour, academic efficiency, teacher ratings of attention and continuous performance task omission errors

  2. Results indicate that higher dosages are linearly related to increasing levels of rate, and that these effects are dependent upon both the initial heart rate value and the time course of medication

  3. Methylphenidate had a significant effect on classroom measures of attention and academic efficiency, which were similar to those of normal control children. Still 25% of the sample failed to show normalised levels of classroom performance

  4. Results of the present investigation show that methylphenidate effects were generally rate‐dependent in the classical fashion (i.e. negative linear relationship between control response rate and output ratio) for responding that was controlled by schedules, which engendered both low and high response rates

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; excluded methylphenidate non‐responders

Any withdrawals due to adverse events: not stated

Email correspondence with study author: We contacted Dr. Rapport by email (Ramstad 2013c [pers comm]). He has stated that he does not have any supplementary data except those provided in these articles. We also contacted Dr. DuPaul (Ramstad 2013b [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Rapport 1987: All children received each of the 5 methylphenidate doses in a randomly assigned counterbalanced sequence

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

All methylphenidate and placebo doses were packaged in coloured gelatin capsules by the clinic's pharmacist. Capsules were sealed in individual daily‐dated envelopes. All teachers were blinded as to time of administration and specific dose

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): yes (4 participants)

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Unclear risk

Study was not supported by any funding, either external or internal. This project was supported in part by a Biomedical Research Support Grant (no. S07 RR05712), which was awarded to the first study author by the Biomedical Research Support Grant Program, Division of Research Resources, National Institutes of Health

Conflicts of interest: no information

Rapport 2008

Methods

Double‐blind, placebo‐controlled, 5‐week, cross‐over trial with 2 interventions

  1. Methylphenidate (5 mg, 10 mg, 15 mg and 20 mg)

  2. Placebo

Phases: baseline (1 week) and 5 cross‐over phases (1 week placebo and 1 week methylphenidate each)

Participants

Number of participants screened: not stated

Number of participants included: 65. Participants were randomly assigned to 1 (of not stated) possible drug condition order

Number of participants followed up: 65 (58 boys, 7 girls)

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV (all were of the combined subtype)

Age: mean 8.56 years (SD 1.25; range 6 to 11)

IQ: mean 102.8 (SD 10.0; range not stated)

Methylphenidate naive: Eight had experienced brief trials of stimulant therapy within the previous 4 years. None were prescribed psychostimulants immediately before the start of the current study

Ethnicity: Caucasian (100%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: low to middle

Inclusion criteria

  1. ADHD diagnosis confirmed by the Kiddie Schedule for Affective Disorders and Schizophrenia

  2. Problems in ≥ 50% of the situations on the Barkley Home Situations Questionnaire

  3. Maternal rating ≥ 2 SD above the mean on the Werry‐Weiss‐Peters Activity Scale

  4. Teacher rating on the ADD/H Comprehensive Teacher Rating Scale ≥ 2 SD above the mean

Exclusion criteria

  1. Conduct disorder; gross neurological, sensory or motor impairment

Interventions

Participants were randomly assigned to different possible drug condition orders of immediate‐release methylphenidate (5 mg/d, 10 mg/d, 15 mg/d, 20 mg/d) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: once daily, in the morning

Duration of each medication condition: 1 week

Washout before study initiation: None were prescribed psychostimulants immediately before the start of the current study

Medication‐free period between interventions: 1 day

Titration period: none

Treatment compliance: "nearly 100%; envelopes were returned on a weekly basis"

Outcomes

Non‐serious adverse events

  1. Subject's Treatment Emergent Symptoms Scale of National Institute of Mental Health (NIMH) (adjusted to children for the children): rated weekly by observer interviewing parent and child

Notes

Sample calculation: not stated

Ethics approval: yes

Comments from study authors

  1. Increased frequency and/or severity of emergent symptoms reported by or observed in children receiving psychostimulant therapy are probable to the extent that dosing regimens differ from the parameters reported herein, particularly for symptoms highly specific to methylphenidate, that is, children receiving multiple doses per day, single doses exceeding 20 mg, different methylphenidate formulations and methylphenidate over a longer duration of time are likely to experience greater frequency and/or severity of emergent symptoms

  2. Immediate‐release methylphenidate formulary is currently used less frequently than that of newer formulations, and the generalisability of present findings to long‐duration, sustained‐release and other variants is unknown

Key conclusion of study authors

  1. Collectively, our findings point to a clear need to develop psychometrically sound treatment‐emergent symptom rating scales for the purposes of monitoring physical and behavioural complaints of children treated with psychostimulants

Comment from review authors

  1. Study authors used a design with a once‐daily, immediate‐release preparation of methylphenidate. This seems to be exceptional compared with other studies

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: April 2014. Obtained supplemental information regarding data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

By email: We used a computer‐generated random assignment (without replacement) procedure with an additional stipulation that a nearly equal number of children had to be assigned to each of the possible drug condition orders. Children were entered onto the list as they entered the study and followed that particular order (of which coders, parents, teachers and evaluators were unaware throughout the study) (Magnusson 2014b [pers comm])

Allocation concealment (selection bias)

Low risk

Same as above

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Methylphenidate and placebo dosages were packaged in coloured gelatin capsules by the clinic's pharmacist to avoid detection of dose and taste

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Rater was blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Information received by email: no drop‐outs (Magnusson 2014b [pers comm])

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

All outcomes are reported

Vested interest bias

Low risk

None

Conflicts of interest: no financial, corporate or commercial relationships to disclose

Riggs 2011

Methods

Sixteen‐week, randomised, 11‐centre, parallel‐group trial with 2 arms

  1. OROS methylphenidate

  2. Placebo

Participants in both medication groups received manual‐standardised, individual cognitive‐behavioural therapy through motivational enhancement approaches throughout the 16‐week medication trial

Participants

Number of participants screened: 1334

Number of participants included: 303 (239 boys, 64 girls)

Number of participants randomly assigned: methylphenidate 151, placebo 152

Number of participants followed up in each arm: methylphenidate 151, placebo 152

Number of withdrawals in each arm: methylphenidate 33, placebo 43

Diagnosis of ADHD: DSM‐IV (combined (68.6%), hyperactive‐impulsive (2.6%), inattentive (28.1%))

Age: mean 16.5 years (SD 1.3; range 13 to 18)

IQ: All participants were cognitively normal

Methylphenidate naive: not stated

Ethnicity: Hispanic (15.2%)

Race: Caucasian (61.7%), African American (23.2%), Asian (1.3%), Native American/Alaskan (1.0%), other (12.7%)

Country: USA

Setting: out‐patient clinic

Comorbidity: major depressive disorder (12.5%), conduct disorder (32.3%), non‐nicotine substance use disorder (100%)

Comedication: drug/alcohol use

Sociodemographics: not stated

Differences between groups

  1. No statistically significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Meets DSM‐IV diagnostic criteria for ADHD

  2. Meets DSM‐IV diagnostic criteria for ≥ 1 non‐nicotine substance use disorder

  3. DSM‐IV ADHD Symptom Checklist score ≥ 22 derived from the adolescent‐completed checklist

Exclusion criteria

  1. Serious medical illness or cardiac illness

  2. History of tic disorder*

  3. Pregnant or breastfeeding

  4. Meets DSM‐IV criteria for current or lifetime psychotic disorder

  5. Meets DSM‐IV criteria for current or lifetime bipolar disorder

  6. Requires/or prescribed other concurrent psychotropic medication

  7. Taking any medications that may produce interactions with OROS methylphenidate

Opiate dependence

  1. Methamphetamine abuse, dependence or past month use

  2. Suicidal risk

  3. Enrolled in an in‐patient, residential, day treatment or out‐patient substance abuse programme within 28 days before signing consent

  4. Participation in other substance or mental health treatment

Exploratory analyses of treatment response (ADHD‐RS)

  1. ADHD subtypes

    1. Defined according to DSM‐IV criteria (i.e. inattentive subtype with ≥ 6 inattentive symptoms and not ≥ 6 hyperactive‐impulsive symptoms)

      1. Number of participants excluded: 13 because they did not meet criteria for a subtype, 14 because they did not meet criteria for hyperactive‐impulsive subtype

      2. Number of participants included: inattentive subtype 103, combined subtype 173. Subtypes did not differ with regard to age or ethnicity but did differ with regard to sex, comorbid conduct disorder, inattentive and hyperactive‐impulsive symptoms and dependence diagnoses

    2. More strictly defined subtypes (i.e. strict inattentive subtype with ≥ 6 inattentive symptoms and ≤ 3 hyperactive‐impulsive symptoms; strict combined type with ≥ 6 inattentive symptoms and ≥ 8 hyperactive‐impulsive symptoms)

      1. Number of participants included: inattentive subtype 52, combined subtype 97

  2. Major depressive disorder comorbidity

    1. 38 participants with major depressive disorder, 265 without major depressive disorder

    2. Number of participants randomly assigned

      1. Methylphenidate: 19 with major depressive disorder, 133 without major depressive disorder

      2. Placebo: 19 with major depressive disorder, 132 without major depressive disorder

    3. No significant differences between groups were noted with regard to ADHD symptom severity, comorbid conduct disorder or substance abuse or dependence diagnoses. Differences between groups were noted with regard to age, sex, court mandate to substance treatment and days of past‐month non‐nicotine substance use

  3. Comorbid conduct disorder

    1. 299 participants included in this analysis. Four were excluded because they did not have a non‐tobacco substance use disorder or did not meet inclusion criteria for ADHD

    2. Number of participants randomly assigned: methylphenidate 48 with conduct disorder, placebo 49 with conduct disorder

Interventions

Participants were randomly assigned to OROS methylphenidate or placebo

Mean methylphenidate dosage at the end of treatment: 68 mg

Administration schedule: single dose in the morning

Duration of intervention: 16 weeks

Titration period: 2 weeks post randomisation

Treatment compliance: 79% (pill counts), 82.3% (self reports)

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, clinician‐administered, adolescent informant: rated at baseline and weekly for 16 weeks

  2. ADHD Rating Scale, clinician‐administered, parent informant: rated at weeks 8 and 16

Serious adverse events

  1. Systematically assessed by medical clinicians during weekly visits

Non‐serious adverse events

  1. Systematically assessed by medical clinicians during weekly visits

  2. Laboratory assessments ascertained at baseline and at 16 weeks included liver function testing, complete blood count with differential and urinalysis

  3. Massachusetts General Hospital Abuse and Diversion Questionnaire, self administered and completed monthly

  4. Massachusetts General Hospital Liking Scale completed at study weeks 4, 8, 12 and 16. Relevant subscales: feeling high, feeling depressed, craving medication, craving substances

Notes

Sample calculation: yes; sample size of 300 participants was calculated for ADHD Rating Scale

Ethics approval: yes; Institutional Review Boards approved the protocol before participant enrolment

Comments from study authors

  1. One participant did not meet diagnostic criteria for ADHD, and 1 did not have a score ≥ 22 on the ADHD Rating Scale, but both were included in analyses

  2. Two participants did not meet diagnostic criteria for a non‐tobacco substance use disorder but were included in analyses

  3. Results of this study add to the growing suspicion that CBT (for SUD) may contribute to ADHD treatment response and warrants further investigation

  4. Limitations: This study was not powered to address safety, and no current consensus exists regarding the most valid outcome measures for ADHD in adolescents with or without substance use disorder

Key conclusions of study authors

  1. OROS methylphenidate did not show greater efficacy than placebo for ADHD or greater reduction in substance use among adolescents concurrently receiving individual cognitive‐behavioural therapy for co‐occurring substance use disorders

  2. OROS methylphenidate was relatively well tolerated and was associated with modestly greater clinical improvement on some secondary ADHD and substance outcome measures

  3. With good monitoring, and in the context of substance abuse treatment, OROS methylphenidate can be used safely in adolescents with a substance use disorder despite non‐abstinence

  4. Higher baseline use of alcohol and cannabis was associated with increased risk of experiencing a treatment‐related adverse event with OROS methylphenidate, but baseline use did not increase the risk of serious adverse events or of any particular category of adverse events

  5. For adolescent misuse/diversion of OROS methylphenidate, results suggest that OROS methylphenidate was not misused/diverted to a greater extent than placebo and was not impacted by baseline substance use severity

  6. Despite baseline differences, both inattentive and combined subtypes responded equally to treatment, suggesting limited relevance for subtype designation in treatment planning

  7. ADHD symptom severity (based on DSM‐IV ADHD Rating Scale) followed a slightly different course of improvement, although with no differences between the group with comorbid major depressive disorder and the group without major depressive disorder in baseline or 16‐week symptom severity, or in 16‐week reduction: Presence of major depressive disorder was not associated with ADHD treatment response in this sample

  8. Interaction effects showed that OROS methylphenidate improved substance use disorder outcomes among adolescents with comorbid conduct disorder compared with placebo

  9. Although severe substance use disorder may require more intensive psychosocial treatment, OROS methylphenidate may improve substance treatment outcomes among adolescents with comorbid attention and conduct problems

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from review authors

  1. Data are influenced by drug abuse, especially data on adverse events, but no statistically significant differences between OROS methylphenidate and placebo in self reported medication abuse were noted

Email correspondence with study authors: December 2013. We obtained supplemental information (IQ, allocation concealment and detailed description of 1 of the serious adverse events)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to OROS methylphenidate or matching placebo in a 1:1 ratio, stratified by site and completed by computer at a centralised location

Allocation concealment (selection bias)

Low risk

Product was supplied in pre‐randomly assigned kits containing individual bottles. Kits and bottles were labelled with the protocol number and treatment/randomisation number. Labeling protected the study, ensuring that it remained blinded, and indicated that the medication was investigational

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Sites were provided blinded medication bottles for each randomly assigned participant

Masking: double‐blinded (participant, caregiver, investigator, outcomes assessor)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Masking: double‐blinded (participant, caregiver, investigator, outcomes assessor)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Primary analyses were ITT, including all randomly assigned study participants

Completers (N = 227) were not different from non‐completers (N = 76) in baseline demographic or clinical characteristics, and the proportion of completers did not differ by treatment assignment

Selective reporting (reporting bias)

Low risk

No major violation compared with CT.gov

All pre‐specified outcomes of interest have been reported

Vested interest bias

Unclear risk

OROS methylphenidate and matching placebo were supplied to the Clinical Trials Network contract pharmacy (EMINENT Services Corporation) by McNeil Consumer and Specialty Pharmaceuticals (distributor for Concerta), at no cost

Principal investigators are NOT employed by the organisation sponsoring the study. NO agreement between principal investigators and study sponsor (or its agents) restricts the principal investigator's rights to discuss or publish trial results after the trial is complete

Conflicts of interest: Some study authors have received research support from, served on Speakers' Bureaus of or acted as consultants for pharmaceutical companies

Rubinsten 2008

Methods

Five‐day, randomised, double‐blind, placebo‐controlled, cross‐over trial with 4 interventions and 11 possible orders

  1. Methylphenidate, in 3 different dosages

  2. Placebo

Participants

Number of participants screened: 170

Number of participants included: 18. Participants were randomly assigned to 1 of 11 possible drug condition orders

Number of participants followed up: 18 (15 boys, 3 girls)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐IV (combined (28%), hyperactive‐impulsive (0%), inattentive (72%))

Age: mean 9.73 years (range not reported)

IQ: mean 104.44

Methylphenidate naive: 0%

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: no

Sociodemographics: no information

Inclusion criteria

  1. Completed the same research protocol in an acute, randomised placebo‐controlled, cross‐over trial

  2. IQ ≥ 82

  3. No current evidence on or history of neurological dysfunction, poor physical health, uncorrected sensory impairments or history of psychosis

  4. At least average reading scores

  5. ADHD according to DSM‐IV

  6. One group of patients needed to have a score < 79 (n = 6)

    1. One group had a score between 80 and 89 on the Wide Range Achievement Test (n = 6 of 9 meeting criterion)

    2. One group had a score ≥ 90 on the Wide Range Achievement Test (n = 6 of 83 meeting criterion)

  7. Free of medication for minimum of 24 hours before diagnostic assessment and participation in the medical trial

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 11 possible drug condition orders of methylphenidate (10 mg, 15 mg and 20 mg) and placebo

Mean methylphenidate dosage: 0.27 mg/kg, 0.42 mg/kg and 0.58 mg/kg, respectively

Administration schedule: not stated

Duration of each medication condition: 1 day

Washout before study initiation: 24 hours

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Examiner observed the child for possible side effects of medication

Notes

Sample calculation: no

Ethics approval: not stated

Comment from study authors

  1. This was a sample of small children, so we do not know whether the same results would hold for adolescents or adults

Key conclusion of study authors

  1. We found clear dissociation of methylphenidate functions: Methylphenidate improved working memory functions but did not improve specific cognitive functions such as quantity manipulation. Moreover, methylphenidate showed decreased efficacy for arithmetic performance in ADHD + developmental dyscalculia, highlighting the need for additional intervention in this subgroup

Comment from review authors

  1. Study authors selected 12 patients from a larger population but did not describe how this was done

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Master randomisation tables were prepared by the research support pharmacist at the hospital using simple randomisation with restrictions. A balanced block 22 design was used

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Examiner, psychiatrist, child and child’s family were not informed about the child’s randomisation order or daily medication status until trial completion. Placebo and active medication were prepared by the hospital pharmacist as powdered and packaged in an opaque gelatin capsule to prevent identification of content by colour, taste or volume. Each child’s medication was placed in an individually named and dated envelope to ensure accurate administration

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Examiner, psychiatrist, child and child’s family were not informed about the child’s randomisation order or daily medication status until trial completion.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Low risk

Research was completed while Dr. Rubinsten was a post‐doctoral fellow at the Hospital for Sick Children (HSC), in Toronto, Canada, and was supported by the Rothschild Fellowship from Israel. It was undertaken, in part, through funding received from the Canadian Institutes of Health (CIHR: Grant #MOP 64312), a CIHR post‐doctoral fellowship (A‐CB), and the Canada Research Chairs Program (RT)

Samuels 2006

Methods

Double‐blind, randomised, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 2

Participants

Number of participants screened: not stated

Number of participants included: 6. Participants were randomly assigned to 1 (not stated) possible drug condition order

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean (not stated)

IQ: mean (not stated)

Sex: not stated

Methylphenidate naive: not stated

Ethnicity: Caucasian (100%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children 5 to 15 years of age

  2. Long‐term stable dose of stimulant medicine for treatment of ADHD

  3. Recruited from local private paediatrician and psychiatric offices

Exclusion criteria

  1. Receiving concomitant medication that might increase blood pressure

  2. Documented hypertension requiring antihypertensive therapy

Interventions

Participants were randomly assigned to 1 (not stated) possible drug condition order of (not stated) methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: Each child was kept on his or her previous regimen of treatment and a corresponding placebo

Duration of each medication condition: 24 hours

Washout before study initiation: 3‐day run‐in

Titration period: Children were kept on stable medication doses and schedules

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. 24‐hour ambulatory blood pressure monitoring (ABPM) was performed on and off medication

No separate data were available for methylphenidate

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusion of study authors

  1. This study provides evidence of a possible negative cardiovascular effect of stimulant medications in children with ADHD. This potential cardiovascular risk should be balanced against the beneficial behavioural effects of this class of medication

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: July 2014. Emailed study authors to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described; "randomized in double‐blind fashion"

Allocation concealment (selection bias)

Unclear risk

"Randomized in double‐blind fashion"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as "double‐blind". All medications and placebo were identically packaged by the research pharmacy

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only participants who successfully underwent an ambulatory blood pressure monitoring study at the end of each phase were included in the primary analyses

Selective reporting (reporting bias)

Unclear risk

Protocol not found

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: not declared

Schachar 1997a

Methods

Twelve‐month, parallel RCT with 4 arms

  1. Methylphenidate + parent training

  2. Methylphenidate + parent support

  3. Placebo + parent training

  4. Placebo + parent support

Follow‐up at baseline (before treatment), at end of titration (3 to 4 weeks) and after 4, 8 and 12 months of treatment

Follow‐up study of cohort completing the 12‐month RCT: Participants were evaluated annually for 5 years

Participants

Number of participants screened: 302

Number of participants included: 91 included in the RCT (methylphenidate: 37 boys, 9 girls; placebo: 37 boys, 8 girls)

Number of participants randomly assigned: methylphenidate 46, placebo 45

Number of participants followed up: methylphenidate 45, placebo 18

Number of withdrawals: methylphenidate 1, placebo 27

Diagnosis of ADHD: DSM‐III‐R diagnosis of ADHD (subtype not stated)

Mean age: methylphenidate 8.4 years, placebo 8.3 years (range 6 to 12)

IQ: mean (methylphenidate 110.3; placebo 107.6)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Canada

Setting: research unit at hospital.

Comorbidity: oppositional defiant disorder (methylphenidate 56.5%, placebo 44.4%), conduct disorder (methylphenidate 6.5%, placebo 20.0%), anxiety (methylphenidate 21.7%, placebo 24.4%)

Comedication: no

Sociodemographics: Placebo group had a higher score at baseline for psychosocial adversity than those assigned to the methylphenidate group

Psychosocial risk index: methylphenidate 1.5%, placebo 2.7%

Inclusion criteria

Screening

  1. Age between 6 and 12 years

  2. ≥ 6 of 14 ADHD symptoms on the ADHD Rating Scale (DuPaul 1991a)

  3. Parental report of some ADHD symptoms at school

  4. No previous treatment with methylphenidate

  5. Willingness to participate in a study that involved random assignment to treatments for ADHD (both pharmacological and non‐pharmacological)

  6. ≥ 1 parent able to communicate in English.

Diagnostic evaluation

  1. Exhibit pervasive ADHD, defined as ≥ 8 of the 14 DSM‐III‐R criteria for ADHD, in 1 setting (at home on the PICS, or at school on the Teacher Telephone Interview (prepared by study authors)) and ≥ 5 ADHD criteria in the other setting

  2. History of ADHD symptoms of ≥ 6 months’ duration before the age of 7 years

Final inclusion criteria

  1. Exhibit pervasive ADHD, defined as ≥ 8 of the 14 DSM‐III‐R criteria for ADHD in 1 setting and ≥ 5 ADHD criteria in the other setting

  2. History of ADHD symptoms of ≥ 6 months' duration before the age of 7 years

  3. Estimated full‐scale IQ > 80

  4. No primary anxiety or affective disorder

Exclusion criteria

  1. Attending a full‐time residential or day treatment programme

  2. Has received regular medication for a medical problem

  3. Chronic medical condition including a severe motor or vocal tic disorder and Tourette's syndrome

  4. Prior treatment for tics

Interventions

Randomly assigned to 1 of the 4 treatment groups after stratification based on comorbid, conduct or oppositional disorder: methylphenidate + parent training/parent support, placebo + parent training/parent support

Titration period: 3 to 4 weeks (depending on child's weight) after randomisation

Mean methylphenidate dosage: target dose 0.7 mg/kg body weight, twice daily

Administration schedule: breakfast and lunch

Duration of intervention: 12 months

Treatment compliance: methylphenidate 80%, placebo 64%

Outcomes

ADHD symptoms

  1. Parent‐ and teacher‐rated IOWA Conners' Rating Scale at baseline, at end of titration and at 4 months

General behaviour

  1. Parent‐ and teacher‐rated telephone interview probe (TIP) (Corkum, personal communication, 1992) questionnaire at baseline and at 4 months. Grouped into 4 factors: inattention, hyperactivity‐impulsivity, oppositional behaviour, difficulty experienced by rater in dealing with typical daily problem situations

Non‐serious adverse effects

  1. Parent‐ and teacher‐rated questionnaire with 14 common side effects (modified from Barkley 1990) administered by telephone at baseline, at end of titration and at 4 months. Grouped into 4 factors: affective, overfocusing, physiological and tics. Parent‐rated questionnaire with 16 possible adverse effects (also modified from Barkley 1990) at annual evaluations (telephone interviews)

  2. Height and weight were measured at baseline, at week 3 to 4 and at 4 months in the RCT, and annually in the follow‐up study. Standing height was measured in centimeters without shoes from floor to vertex of head. Weight in indoor clothing, without shoes, was measured in kilograms. Baseline and annual measures of height and weight were ascertained with the same stadiometer

  3. Presence and severity of tics were rated at baseline, end of titration, 4 months, 8 months and 12 months by research assistants interviewing parents and teachers. Moderate and severe Tourette's‐like symptoms were assessed and diagnosed by the supervising psychiatrist

Notes

Sample calculation: yes

Ethics approval: yes

Comments from study authors

  1. Current RCT is limited by sample size and by medium statistical power

  2. This cannot be generalised to situations in which higher doses of methylphenidate may be used, or to children with more than moderate tics

  3. Instrument used to measure adverse effects was originally designed for short‐term pharmacological studies and could fail to identify potentially long‐term health concerns in the follow‐up study

  4. Sample in the follow‐up study of previous participants in an RCT would be biased in favour of adherence relative to a cohort chosen and followed from the time of identification

  5. Infrequent contact with participants and lack of an untreated control group are additional limitations of the follow‐up study

  6. Study did not include pubertal staging

Key conclusions of study authors

RCT

  1. Positive effects of methylphenidate on behaviour are evident in the classroom, but with methylphenidate given twice daily, parents did not report that methylphenidate improved behaviour at home

  2. Comorbid anxiety did not appear to influence development of side effects or behavioural response to methylphenidate when dose was titrated as in standard clinical practice

  3. Doses of methylphenidate based on the typical clinical titration procedure did not produce significantly more tics than placebo in children with or without pre‐existing (mild to moderate) tics

Follow‐up study

  1. Psychostimulants improve ADHD symptoms for up to 5 years, but adverse effects persist

  2. Long‐term use of high doses of stimulants during a period of 1 to 5 years is likely to have measurable effects on rate of growth in school‐aged children with attention deficit hyperactivity disorder

Comments from review authors

  1. Thiruchelvam 2001 reported no relevant outcome measures for our review; the article was therefore excluded during the full‐text reading process

  2. Corkum 1999 was excluded during the abstract reading process

  3. Data in both Schachar 1997 and Diamond 1999 are preliminary (interim results of first 4 months of treatment). Data from the 2 placebo groups (cointervention: parent training and parent support) and the 2 methylphenidate groups (same 2 cointerventions) are combined (placebo vs methylphenidate). No final report focuses on ADHD symptoms

  4. As participants are allowed to switch from the allocated intervention group during the study, we can use only data regarding adverse effects

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: September to October 2013. Unfortunately, they were not able to provide supplemental data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned after stratification based on the presence of comorbid conduct or oppositional disorder and assigned to 1 of the 4 treatment groups. Randomly assigned based on a random number table

Allocation concealment (selection bias)

Low risk

Concealed from the physician and participants before randomisation and maintained throughout the study

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants (child, research staff, parents and teachers) were blinded to the medication assignment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All participants (child, research staff, parents and teachers) were blinded to the medication assignment

Incomplete outcome data (attrition bias)
All outcomes

High risk

No imputation method: The effect of treatment was analysed among participants who adhered to their original medication assignment. At the 4‐month point, children not taking any medication were grouped with those taking placebo. When a participant switched from placebo to methylphenidate treatment, they were counted within the methylphenidate group

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not possible to find protocol

Vested interest bias

Unclear risk

Medical Research Council of Canada, National Health Research Development Program of Canada and the Department of Psychiatry, The Hospital for Sick Children, Toronto. Placebo pills were provided by Ciba Geigy, Canada, Ltd

Conflicts of interest: Two study authors have reported working as consultants for pharmaceutical companies, and 1 has furthermore received industry‐sponsored research grants

Schachar 2008

Methods

Single‐centre, randomised, double‐blind, 3‐way cross‐over trial with 2 interventions

  1. Methylphenidate (immediate‐release or extended‐release)

  2. Placebo

Phases: immediate‐release and multi‐layer‐release

Participants

Number of participants screened: not stated

Number of participants included: 18

Number of participants followed up: 17 (15 boys, 2 girls)

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV diagnosis of ADHD (combined (100%))

Age: mean 11.3 years (range 6.8 to 15.3)

IQ: ≥ 85

Methylphenidate naive: 5 (29.4)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: no

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 15 years of age

  2. IQ ≥ 85 within previous 12 months

  3. Mentally and physically competent to provide written informed consent

  4. Ability to read, speak and understand English

  5. Otherwise able to comply with study protocol

Exclusion criteria

  1. Allergic to methylphenidate or amphetamines, or had a history of serious adverse reactions to methylphenidate or lack of response to methylphenidate

  2. Serious or unstable medical illness, comorbid psychiatric illness of sufficient severity to require treatment or currently receiving psychotropic medications or herbal treatments

  3. Disorders of the sensory organs (particularly deafness), autism, psychosis or any unstable psychiatric conditions

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of immediate‐release and extended‐release methylphenidate and placebo

Mean methylphenidate dosage: 31.2 mg/d (SD 11.7 mg/d; range 20 to 60 mg/d), 1.2 mg/kg

Administration schedule: twice daily, in the morning and at noon (4 hours apart)

Duration of each medication condition: 6 days

Washout before study initiation: 1 day

Titration period: none. Participants who were receiving methylphenidate at the time of study entry received the dose of methylphenidate that they were taking before entry into the study/duration initiated before/after randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA: parent‐rated weekly (8 times in 1 day)

  2. Conners' Continous Performance Task: rated weekly (8 times in 1 day)

Non‐serious adverse events

  1. Clinical Assessment of Side Effects Scale: parents and teachers separately, rated weekly 1 day before other assessments

  2. Spontaneously reported adverse events: investigator, collected weekly

Notes

Sample calculation: no

Ethics approval: yes

Key conclusion of study authors

  1. Both MLR methylphenidate and immediate‐release methylphenidate compared with placebo demonstrated significant improvement on IOWA‐C average total score, with onset of action observable by 1 hour

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: May 2014. Emailed study authors to ask for supplementary information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned to a treatment sequence according to a Latin square

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind; not further described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind; not further described

Incomplete outcome data (attrition bias)
All outcomes

High risk

All enrolled participants with data from all 3 treatment phases who did not have major protocol violations were considered evaluable for efficacy; all participants were evaluated for safety

Selective reporting (reporting bias)

Low risk

No protocol was published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Study was funded by Purdue Pharma (Canada)

Conflicts of interest: Some study authors are working for Purdue Pharma

Schulz 2010

Methods

Double‐blind, randomised, multi‐centre, triple cross‐over design with 3 interventions

  1. Methylphenidate ‐ Ritalin LA 20 mg once daily

  2. Methylphenidate ‐ Medikinet Retard 20 mg once daily

  3. Placebo

Participants

Number of patients screened: 147

Number of participants included: 147

Number of participants randomly assigned: not stated

Number of participants followed up: 139

Number of withdrawals: 1
Diagnosis of ADHD: DSM‐IV diagnosis (combined (55%), hyperactive‐impulsive (8%), inattentive (37%))

Age: mean 10.2 years (range 6 to 14)

IQ: not stated

Sex: 119 boys, 28 girls

Methylphenidate naive: 0%

Ethnicity: not stated

Country: Germany

Setting: out‐patient clinic

Comorbidity: 6.8% (disturbance in social behaviour (2.7%), initial insomnia (0.7%), oppositional defiant disorder (1.36%), dysphemia (0.68%), encopresis (0.68%))

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD confirmed by Kiddie Schedule for Affective Disorders and Schizophrenia in the German Version (Delmo et al. 2000)

  2. All children had to be on a stable and well‐tolerated dose of 20 mg‐equivalent methylphenidate for ≥ 1 month before screening

Exclusion criteria

  1. Patients with known previous non‐response to methylphenidate

  2. Children with relevant somatic or psychiatric comorbidity requiring pharmacological treatment (e.g. psychosis, major depression)

  3. Patients with warnings as described in the prescribing information for Ritalin LA, including tic disorders

Interventions

Pre‐randomisation phase and 3 treatment periods of 7 days each. Participants were randomly assigned to 1 of 6 possible drug condition orders of 20 mg Ritalin LA methylphenidate, 20 mg Medikinet Retard methylphenidate and placebo

Mean methylphenidate dosage: 20 mg daily

Administration schedule: once daily, morning

Time points: 9:00 AM

Duration of each medication condition: 7 days

Washout before study initiation: none

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham (SKAMP) Scale (Wigal 1998) by 3 treatment‐blinded observers: rated at 1.5, 3.0, 4.5, 6.0 and 7.5 hours after drug intake at the end of each treatment week

General behaviour

  1. Nisonger Child Behavior Rating Form (NCBRF‐TIQ) to assess behaviour in children (Aman 2002): applied once at the end of each treatment period

Serious adverse events

  1. One serious adverse event was reported: a case of acute appendicitis during treatment with Ritalin LA. This was judged to be not treatment related

  2. Four events were reported as "severe" and drug‐related: placebo: 2× aggressive behaviour; placebo: 1× lack of attention; Medikinet: 1× aggressive behaviour

Non‐serious adverse events

  1. Monitoring and recording of all adverse events (AEs) by

    1. "Non‐directive questioning" at every visit

    2. "Regular monitoring of vital signs"

    3. Participant were called by phone every midweek and were asked about adverse events

    4. Clinical abnormality in blood pressure defined as increase or decrease ≥ 20 mmHg from age‐dependent normal values (ages 6 to 9: systolic blood pressure (SBP) 90 mmHg, diastolic blood pressure (DBP) 60 mmHg; age > 10 years: SBP 110 mmHg, DBP 75 mmHg)

Clinically notable increased values for SBP were recorded under all 3 treatments: 15% on placebo, 17% on Ritalin LA and 18% on Medikinet. Abnormal increases in DBP were noted in 5% of patients taking placebo and Ritalin and in 3% of patients given Medikinet. Abnormal values in SBP among participants who had normal values at screening and at baseline were recorded in only 7 participants (3 given placebo, 3 taking Medikinet and 2 taking Ritalin LA ‐ as reported in the article). Changes in vital signs were attributed to sympathomimetic effects of methylphenidate and were not considered clinically relevant

Notes

Sample calculation: yes
Ethics approval: yes

Comments from study authors

  1. Another factor to be considered a limitation is that all participants were known methylphenidate responders. This decision was made to ensure that study objectives regarding efficacy were met. However, it might have led to overestimation of treatment effect and tolerability in the overall population, as no treatment‐naive participants were included in this trial

  2. Individual dose titration as a standard approach in everyday practice was not allowed during this trial; therefore treatment response might not have been optimal for all children

  3. Inclusion of many other children, all with ADHD, as well as other experimental factors might have influenced the behaviour of the individual child

  4. Encapsulation might have altered the pharmacokinetics of active substances

Key conclusions of study authors

  1. Compared with placebo, both Ritalin LA and Medikinet Retard demonstrated robust treatment effects on core inattentive and hyperactive symptoms for up to 7.5 hours in children with ADHD in a laboratory classroom

  2. Treatment with Ritalin LA and Medikinet Retard was generally well tolerated

  3. No participant had to discontinue participation in the trial because of adverse events

  4. Although almost every third participant was affected by ≥ 1 adverse event, almost all adverse events were of mild intensity

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; efficacy trial to prove non‐inferiority of Ritalin LA vs Medikinet Retard. Highly restrictive inclusion criterion ‐ responders only. Study authors acknowledge "it might overestimate the treatment effect and the tolerability in the overall population, as no treatment naive patients have been included in this trial"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization was centrally generated with a validated system that automated the random assignment of treatment sequences to randomization numbers in a specified ratio"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All treating physicians, other site staff, and patients, as well as data analysts and Novartis in‐house personnel, remained blinded from the time of randomization until database lock. The raters also did not have access to information about adverse events to maintain the blind"; "To ensure blinding of the study, all capsules were overencapsulated in an identical optical design"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The scale was rated by three treatment‐blinded observers in the classroom who were specifically trained on the SKAMP scale"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomly assigned participants were included in the intention‐to‐treat (ITT) population. Missing values for the primary endpoint were replaced by the worst value observed in another participant under the same treatment at the same assessment time. For non‐inferiority comparisons, the per‐protocol (PP) population consisting of participants without major protocol violations was considered primary

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

High risk

Trial aimed at showing efficacy of Ritalin LA with purpose of obtaining marketing authorisation

Conflicts of interest: Almost all study authors have received grants, research support or other kinds of financial support from the medical industry

Schwartz 2004

Methods

Two‐week, double‐blind, placebo‐controlled, cross‐over, randomised clinical trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 44 (37 boys, 7 girls)

Number of participants followed up: 44

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.2 years (range 6 to 12)

IQ: > 70

Methylphenidate naive: 16

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: conduct disorder (39%), oppositional defiant disorder (32%), separation anxiety disorder (16%), major depression (7%)

Comedication: no

Sociodemographics: income level category, mean 3.7

Inclusion criteria

  1. ADHD diagnosis

Exclusion criteria

  1. IQ < 70

  2. Tourette's syndrome

  3. Pervasive developmental disorder

  4. Psychosis.

  5. Taking any medication other than methylphenidate

  6. Previous intolerance or allergic reaction to any psychostimulant

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of (0.5 mg/kg) methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: twice per day: morning/noon time points

Duration of each medication condition: 7 days

Washout before study initiation: yes (2 weeks)

Medication‐free period between interventions: not stated

Titration period: none stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Rating Scales ‐ both teacher‐ and parent‐rated

  2. Restricted Academic Situation Scale

Non‐serious adverse events

  1. Actigraphic monitoring of sleep

Notes

Sample calculation: post hoc power calculation

Ethics approval: yes

Comment from study authors

  1. Good responder participants showed improvement in behaviour when taking methylphenidate but no difference in sleep‐onset latency compared with those given placebo (average 49 minutes). However, this latency was rather long, and this was an open trial with no comparison group with poor or no response to methylphenidate

Key conclusions of study authors

  1. Methylphenidate induces a 4% reduction in actigraph (slight but significant sleep disturbance)

  2. Children who responded well to methylphenidate did not exhibit increased motor activity in sleep compared with those who did not respond or who responded poorly

  3. Therapeutic efficiency does not come with more sleep side effects

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; exclusion of children with history of intolerance

Email correspondence with study authors: March to April 2014. We emailed study authors twice to ask for supplemental information regarding data on ADHD symptoms but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"After baseline assessments, children randomly received either placebo or (...)"

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

Grants from Le Fonds de la Recherche en Santé du Québec and the Canadian Institutes of Health Research

Conflicts of interest: Yes. Dr. Joober is a principal investigator on a clinical trial not related to this study that is sponsored by AstraZeneca Canada Incorporated, and receives no direct compensation for this trial. Dr. Boivin has the following industry financial ties: The Litebook Company Ltd., Medicine Hat, Alberta, Canada; and Pulsar Informatics Inc., Vancouver, British Columbia, Canada

Sharp 1999

Methods

Cross‐over trial with 3 interventions

  1. Methylphenidate

  2. Dextroamphetamine

  3. Placebo

Phases

  1. Three‐week medical‐free baseline, randomised administration of methylphenidate, dextroamphetamine and placebo followed by breakfast and lunch daily

  2. Stepwise increase in stimulant dose each week

  3. Each phase lasted 3 weeks

  4. Dosage range: methylphenidate 10 mg/d to 70 mg/d, dextroamphetamine 5 mg/d to 30 mg/d

Participants

Number of participants screened: 150 (girls)

Number of participants included: 42 girls and 56 comparison boys

Number of participants followed up: 42

Number of withdrawals: 1 girl given placebo at each phase

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Mean age: 9.1 years (range 6.0 to 12.7)

Mean IQ: boys 109.3, girls 105.2

Sex: 56 boys, 42 girls

Methylphenidate naive: Elia 1991: 18 out of 48 had no previous stimulant drug treatment; Castellanos 1996: 80% had been previously treated with stimulants

Ethnicity: girls: Caucasian (67%), African American (19%), Hispanic (14%); boys: Caucasian (73%), African American (21%), Hispanic (4%), Asian (2%)

Country: USA

Setting: out‐patient clinic

Comorbidity: 71% boys, 69% girls; oppositional defiant disorder (33% boys, 50% girls), conduct disorder (7% boys, 2% girls), major depression (0% boys, 7% girls), separation anxiety (0% boys, 2% girls), specific phobias (0% boys, 7% girls), trichotillomania (2% boys, 0% girls), tic disorders not otherwise specified (13% boys, 2% girls), enuresis (18% boys, 12% girls), reading disorder (5% boys, 8% girls)

Comedication: not stated

Sociodemographics: SES mean score 48.0 ± 25.8 (girls), 52.4 ± 26.9 (boys)

Inclusion criteria

  1. Girls with a history of severe hyperactivity, impulsivity and inattentiveness that interfered with home and school functioning

  2. Symptoms of ADHD present in ≥ 2 settings

  3. Conners' Hyperactivity factor scores from home teachers were ≥ 2 SD greater than age and sex norms

Exclusion criteria

  1. Full‐scale IQ < 80 on Wechsler Intelligence Scale for Children ‐ Revised

  2. Chronic medical or neurological disease, including Tourette’s disorder and chronic tic disorders

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of methylphenidate, dextroamphetamine and placebo

Sharp 1999

Mean methylphenidate dosage: week 1 mean dose 0.45 mg/kg, week 2 mean dose 0.85 mg/kg and week 3 mean dose 1.28 mg/kg

Administration schedule: breakfast and lunch ‐ 5 days per week administered by nurse; administered by parent on weekends

Elia 1991

Mean methylphenidate dosage: week one 0.9 mg/kg, week two 1.5 mg/kg and week three 2.5 mg/kg

Administration schedule: 9:00 AM and 1:00 PM throughout the entire week (7 days)

Duration of each medication condition: 3 weeks

Washout before study initiation: 3‐week medication‐free period before study

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Hyperactivity and Conduct factors score: teacher‐rated

  2. Wender Utah Rating Scale (WURS): parent‐rated

  3. Conners' Abbreviated Teacher Rating Scale (ABTRS): completed weekly

  4. Conners' Teacher Rating Scale (CTRS): completed weekly

General behaviour

  1. Child Behavior Checklist (CBCL): parent‐rated

  2. Woodstock‐Johnson Achivement Battery: observer (psychologist)‐rated

  3. Teachers Report Form (TRF): teacher‐rated

  4. Conners' Parent Questionnaire (CPQ): completed weekly by parents

  5. Children’s Psychiatric Rating Scale (CPRS): completed weekly by psychiatric staff

  6. Activity monitor (measuring truncal motor activity): from 9:00 AM to 4:00 PM

Serious adverse events

Mean beneficial and adverse effects of dextroamphetamine and methylphenidate were nearly identical for all ratings, including ratings of appetite problems. However, objectively verified significant decreases in body weight (drug main effect, F 10.27; P value = 0.0002) were significantly greater for dextroamphetamine (mean change ‐1.1 ± 1.0 kg from baseline; P value = 0.02) than for methylphenidate (‐0.4 ± 1.1 kg; not significant)

Stimulant‐related adverse effects and body weight

Non‐serious adverse events

  1. Subject Treatment Emergent Symptom Scale (STESS): physicians and parents

  2. Some items on the CPRS, such as nervous mannerisms and obsessive thinking: rated as adverse effects (Elia 1991)

  3. 24‐hour urine and blood studies: at baseline and at third week for each treatment

Notes

Sample calculation: no

Ethics approval: no information available

Key conclusion of study authors

  1. Sharp 1999

    1. Our primary conclusion is that our sample of girls demonstrated very similar patterns of comorbidity and impairment and identical patterns of drug response

Comment from review authors

  1. We need a lot of information on the girls

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study authors: June 2014. We received supplemental information from Dr. Castellanos

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Order of medication (methylphenidate, placebo or dextroamphetamine) was randomly assigned on the basis of a table administered by the research pharmacy. Dose escalation was fixed, with 2 ranges, depending on body weight (> or < 30 kg). Three weeks (1 dose per week) in each phase. For most children, this was followed by random assignment to pemoline vs placebo after another washout period

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Methylphenidate, dextroamphetamine and placebo were packaged in identical capsules by the NIH pharmacy and were administered by NH nurses in double‐blinded randomly assigned order

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All ratings were performed blind to treatment assignment

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Did not describe what they did with the missing data

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol published. All outcomes discussed were reported

Vested interest bias

Unclear risk

None mentioned

Conflicts of interest: not declared

Shiels 2009

Methods

"3‐day, double‐blind, placebo‐controlled medication assessment"

Participants

Number of participants screened: not stated

Number of participants included: 49

Number of participants randomly assigned: not stated

Number of participants followed up: not stated

Number of withdrawals: methylphenidate 0, placebo 0

Diagnosis of ADHD: DSM‐IV (combined 35 (71%), hyperactive‐impulsive 3 (6%), inattentive 11 (23%))

Age: mean 10.5 years (range 9 to 12)

IQ: mean 104 (range not stated)

Sex: 80% boys, 20% girls

Methylphenidate naive: not stated

Ethnicity: Caucasian (76%), African American (14%), other (10%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (43%), conduct disorder (22%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV ADHD diagnosis

  2. Attending ADHD summer research programme

Exclusion criteria

  1. Full‐scale IQ < 80

  2. History of seizures or other neurological problems or medication to prevent seizures

  3. History of other medical problems for which psychostimulant treatment may involve considerable risk

  4. Current use of psychotropic medications other than for ADHD (i.e. antipsychotics, mood stabilisers, antidepressants and anxiolytics)

  5. History or concurrent diagnosis of pervasive developmental disorder, schizophrenia or other psychotic disorders

  6. Absence of functional impairment

  7. Vision or hearing problems that would make it difficult to complete discounting tasks (or other tasks; data not reported)

Interventions

Participants were assigned to extended‐release methylphenidate in "high" or "low" dose (range 18 to 90 mg/d)

Mean methylphenidate: low dose 39 mg Concerta, high dose 73 mg Concerta

Administration schedule: once daily

Time points: in the morning, 90 minutes before cognitive task

Duration of intervention: 7:30 AM to 5:00 PM, Monday through Thursday

Titration period: not stated

Treatment compliance: not stated

Washout before study initiation: methylphenidate 1 week if taking atomoxetine, 24 hours if taking methylphenidate

Outcomes

General behaviour

Impairment Rating Scale (IRS; Fabiano 2006)

Non‐serious adverse events

Pittsburgh Side Effects Rating Scale, blood pressure and heart rate assessed daily during times of peak medication effects

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: none

Key conclusions of study authors

  1. These findings provide initial evidence that stimulant medication reduces delayed discounting among those with the disorder

  2. Methylphenidate, an effective pharmacological treatment for ADHD, reduced the preference for smaller immediate rewards over larger delayed rewards when delays and rewards were actually experienced by the child. This raises the possibility that stimulant medication improves real‐world behaviour of children with ADHD by altering delay‐related impulsivity

Comment from review authors

  1. This study focused on effects of methylphenidate on "delay discounting", a function of impulsivity. However, no data are available on the effects of methylphenidate on ADHD symptoms more generally, nor are data available regarding side effects or changes in blood pressure and pulse.

Email correspondence with study authors: July 2014. Emailed study authors to request additional information but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"To maintain blinding, participants were given the same number of opaque capsules per day regardless of actual MPH dose"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not described

Vested interest bias

High risk

Not described

Conflicts of interest: "In the past 3 years, James G. Waxmonsky has served on the Speakers Bureau for Novartis, received honoraria from Scepter, and received research support from Eli Lilly"

Silva 2005a

Methods

Six‐week, randomised, single‐blind, placebo‐controlled, cross‐over trial with 5 interventions

  1. Extended‐release methylphenidate 20 mg

  2. Extended‐release methylphenidate 40 mg

  3. OROS methylphenidate 18 mg

  4. OROS methylphenidate 36 mg

  5. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 54 (34 boys, 20 girls)

Number of participants followed up: 53

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (combined (70.4%), hyperactive‐impulsive (1.9%), inattentive (27.8%))

Age: mean 9.4 years (range 6 to 12)

IQ: > 80

Methylphenidate naive: 0%

Ethnicity: Caucasian (63.0%), African American (14.8%), Asian (0%), other (22.2%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children 6 to 12 years old

  2. Meeting DSM‐IV criteria for primary diagnosis of ADHD

  3. Written consent from parents

  4. Treated and stabilised on total daily dose of 20 mg to 40 mg methylphenidate for ≥ 2 weeks before enrolment

  5. Females are required to be pre‐menarchal, sexually abstinent or using an approved method of contraception

  6. Females of childbearing potential are required to have a negative pregnancy test before enrolment

Exclusion criteria

  1. IQ level ≤ 80

  2. Diagnosed with Tourette's syndrome or a tic disorder

  3. Deemed by investigator to be unable to comply with study instructions

  4. Significant concurrent medical or psychiatric illness or substance abuse disorder

  5. History of sensitivity to methylphenidate

  6. History of substance abuse

  7. Currently taking atomoxetine

  8. Have taken, or is currently taking or planning to take, any investigational drug within 30 days of study start date

Interventions

Participants were randomly assigned to 1 of 10 possible drug condition orders of 20 mg extended‐release methylphenidate, 40 mg extended‐release methylphenidate, 18 mg OROS methylphenidate, 36 mg OROS methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: once

Duration of each medication condition: 1 day

Washout before study initiation: 1 day

Medication‐free period between interventions: Participants were instructed to continue to take their regularly prescribed medication Sundays through Thursdays between study days; no medication was administered on Fridays to avoid possible carry‐over effects during the Saturday treatment period

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale: by blinded raters, ‐0.5, 0.5, 1, 2, 3, 4, 6, 8, 10 and 12 hours post dose. Primary analysis time point was 2 hours post dose

Non‐serious adverse events

  1. Adverse events were assessed throughout the classroom period day

Only 1 adverse event was considered to be drug‐related

Notes

Sample calculation: yes; 46

Ethics approval: approved by an independent investigational review board

Comments from study authors

  1. As available formulations of extended‐release methylphenidate contain 11% more methylphenidate than their OROS methylphenidate counterparts, outcomes might appear to be biased toward extended‐release methylphenidate

  2. OROS methylphenidate releases a third bolus of active drug between 8 and 12 hours after dosing, whereas extended‐release methylphenidate releases its last bolus at 4 hours post dose. Hence, outcomes during the last 4 hours of the day might, theoretically, be biased towards OROS methylphenidate

  3. Study was conducted in a controlled laboratory classroom

  4. These results are representative of participants who are known responders to methylphenidate

  5. All participants had been previously stabilised on ADHD medication, so they may have been able to detect a difference between active treatment and placebo

Key conclusions of study authors

  1. Efficacy of extended‐release methylphenidate 20 mg is similar to that of OROS methylphenidate 18 mg and 36 mg during the first 8 hours post dose

  2. Statistically greater benefits are observed with extended‐release methylphenidate 40 mg than with OROS methylphenidate 36 mg and persist through hour 8

  3. Active treatments show comparable efficacy from 8 to 12 hours post dose

  4. Both doses of each methylphenidate formulation are well tolerated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: April 2014. Sent an email to study authors to ask for additional data but have not receive a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All study personnel, with the exception of the nurse, pharmacist or physician dispensing medication, were blinded

Study medications were administered in an opaque container with a small aperture, so participants could not see them during administration

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale was completed by blinded raters

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol found

Vested interest bias

High risk

Funding from Novartis Pharmaceuticals Corporation

Conflicts of interest: All study authors have been consultants, have received honoraria or have worked for Novartis

Silva 2006

Methods

Cross‐over trial with 2 interventions

  1. D‐MPH‐ER 20 mg once daily

  2. Placebo

Phases: 2 cross‐over phases

Participants

Number of participants screened: not stated

Number of participants included: 54 (38 boys, 16 girls)

Number of participants followed up: 53

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (combined (90.7%), hyperactive‐impulsive (0%), inattentive (9.3%))

Age: mean 9.4 years (range 6 to 12)

IQ: not stated

Methylphenidate naive: none

Ethnicity: predominantly Caucasian

Country: USA

Setting: laboratory classroom

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Boys and girls

  2. Age 6 to 12 years

  3. Diagnosed with ADHD

  4. Must have been stabilised on methylphenidate 20 mg/d to 40 mg/d for ≥ 1 month

  5. For girls: pre‐menarchal, sexually abstinent or using reliable contraceptive and have negative urine pregnancy test

Exclusion criteria

  1. Investigator deemed IQ below average or evidence of IQ < 80; home‐schooled

  2. Tourette's syndrome

  3. Tic disorder

  4. Significant medical illness

  5. Significant psychiatric illness (schizophrenia, bipolar disorder, autism)

  6. Parents or guardians unable to understand or follow instructions

  7. Taking antidepressants

  8. Initiated psychotherapy 3 months before screening

  9. Positive urine drug screening

  10. Poor response to methylphenidate

  11. Currently taking other medications for ADHD

  12. Taking or planning to take another investigational drug within 30 days of study start; previously participated in d‐MPH‐ER study

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 20 mg/d d‐MPH‐ER and placebo

Mean methylphenidate dosage: 20 mg/d

Administration schedule: once daily; morning

Duration of each medication condition: 1 week

Washout before study initiation: 1 day

Medication‐free period between interventions: 1 day

Titration period: none

Treatment compliance: 100% in laboratory classroom sessions

Outcomes

ADHD symptoms

  1. SKAMP: once weekly, observer‐rated

Non‐serious adverse events

  1. Recorded adverse events described by parents and children, as well as in laboratory school setting, 1 day each week

Notes

Sample calculation: yes

Ethics approval: not stated

Comments from study authors

  1. One of the inclusion criteria in this study was that participants were known to be currently stable on another methylphenidate preparation

  2. We listed all side effects, irrespective of whether they were reported by parent or child. These were gathered both during the week preceding laboratory observation days and during the laboratory classroom day

  3. We did not systematically analyse compliance data during the week preceding the laboratory classroom

Key conclusion of study authors

  1. In this study, once‐daily d‐MPH‐ER 20 mg was a safe and effective treatment for paediatric participants with ADHD symptoms

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Any withdrawals due to adverse events: yes; 1 (during placebo)

Email correspondence with study authors: June 2014. Sent twice to study author to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned by computerised random number assignment

Allocation concealment (selection bias)

Low risk

Study medication comprised 1 bottle of 5 capsules of blinded study medication or 1 bottle of 5 matching placebo capsules

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. Study medication comprised 1 bottle of 5 capsules of blinded study medication or 1 bottle of 5 matching placebo capsules

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Three independent blinded raters

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Safety population consisted of all participants who received ≥ 1 dose of study medication. Efficacy population comprised all randomly assigned participants who provided valid efficacy measurements for both treatment periods

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

Financed by Novartis

Conflicts of interest: Some study authors have affiliations with medical companies

Silva 2008

Methods

Randomised, multi‐centre, double‐blind, placebo‐controlled, cross‐over design with 2 interventions

  1. d‐MPH‐ER

  2. Placebo

Participants

Number of participants screened: 68 (45 boys, 23 girls)

Number of participants included: 68

Number of participants followed up: 67

Number of withdrawals: 1 (from the placebo group)

Diagnosis of ADHD: DSM‐IV (combined (82.4%), hyperactive‐impulsive (0%), inattentive (17.6 %))

Age: mean 9.5 years (range 6 to 12)

IQ: > 70

Methylphenidate naive: none

Ethnicity: Caucasian (50%), African American (22.1%), Asian (0%), Hispanic (19.1%), other (8.8%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 12 years of age

  2. DSM‐IV diagnosis of ADHD

  3. Participants had to be clinically and behaviourally stable in the opinion of the referring physician and the site's principal investigator

  4. Have taken their current dose of medication without adjustment for ≥ 2 weeks (this was required to be a total daily dose or nearest equivalent of methylphenidate 40 mg or immediate‐release d‐MPG 20 mg (Concerta 36 mg was allowed))

  5. Parents and/or guardians had to provide informed consent

  6. Female participants were required to be pre‐menarcheal or sexually abstinent, or had to be using an adequate and reliable contraceptive method (e.g. double‐barrier method), which was documented in the medical record. Girls who were sexually active were required to have a negative result on a urine pregnancy screening test

Exclusion criteria

  1. Children were excluded if they or their parents/guardians were unable to understand or follow instructions necessary to participate in the study

  2. If they were deemed by the investigator to have below‐normal cognitive capacity, or if they were home schooled

  3. Diagnosed with Tourette's disorder or a tic disorder, or had a history of, or concurrent, significant medical or psychiatric illness or substance abuse disorder

  4. Children taking an antidepressant medication, those who initiated psychotherapy within the 3 months preceding screening and those with a positive urine drug screen

  5. Also excluded were children with poor response or intolerance to methylphenidate, who were currently taking other medications for ADHD or were taking or planning to take any other investigational drug within 30 days of study start or who had previously participated in d‐MPH‐ER studies

Interventions

Participants were randomly assigned to d‐ER‐MPH or placebo

Mean methylphenidate dosage: 20 mg/d

Administration schedule: once daily, in the morning

Duration of each medication condition: 7 days

Washout before study initiation: 2 days

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Combined score on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale: measured at time points 1, 3, 4, 5, 7, 9, 10, 11 and 12 hours post dose by independent blinded raters. Rated at practice day (0), period 1 (visit day 7) and period 2 (visit day 14), and at final visit (visit day 15)

  2. Attention and deportment scores on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale, obtained from 0.5 hours up to 12 hours by independent blinded raters. Rated at practice day (0), period 1 (visit day 7) and period 2 (visit day 14), and at final visit (visit day 15)

  3. Combined score on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale, measured at time point 0.5 by independent raters. Rated at practice day (0), period 1 (visit day 7) and period 2 (visit day 14), and at final visit (15)

Non‐serious adverse events

  1. Vital signs obtained at practice day (0), period 1 (visit day 7) and period 2 (visit day 14), and at final visit (15)

  2. Recording of spontaneously reported adverse events

Notes

Sample calculation: no

Ethics approval: yes

Comment from study authors

  1. Unequal carry‐over effects: As a result of the design used in this study, a test for carry‐over effects could not be performed. Instead, tests of sequence effects in the analysis of the co‐variance model were examined. If tests on sequence factors among time points were statistically significant (P value = 0.05), analyses were performed by each period

Limitations

  1. Each participant took d‐MPH‐ER for only 1 week

  2. Only 1 dose (20 mg/d) was used, meaning that results may not be generalisable to other doses

  3. Study was confined to school‐aged children, so applicability of results to pre‐school children, adolescents or adults is unknown

  4. All participants in this study had been previously shown to respond to and tolerate methylphenidate or D‐MPH

  5. Children who received placebo during the week before the laboratory classroom day showed statistically better pre‐dose performance than children who received active medication on all measures except the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale

Key conclusions of study authors

  1. In this study, once‐daily d‐MPH‐ER 20 mg was effective in treating both inattentive and behavioural symptoms in paediatric patients over a 12‐hour laboratory classroom day

  2. Primary efficacy variable ‐ combined score on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale ‐ showed significant superiority over placebo at all time points from 1 to 12 hours

  3. Secondary efficacy variables indicated that onset of effect was rapid (0.5 hours) and duration of effect was relatively long (12 hours post dose)

  4. In this sample, the drug was safe and well tolerated

  5. Changes in vital signs were comparable with those of placebo

Inclusion of methylphenidate responders only/exclusion of MPH non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Email correspondence with study authors: October 2013. We received supplemental information from study authors regarding ethics approval and data. We were not able to obtain first period data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random assignment

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All study medications were identical in appearance for blinding purposes; double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Safety population consisted of all participants who took ≥ 1 dose of study medication. Efficacy population included all randomly assigned participants who provided valid efficacy measurements for both treatment periods

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Outcomes according to protocol

Vested interest bias

High risk

Funded by Novartis Pharmaceuticals Corporation

Conflicts of interest: Some study authors have affiliations with medical companies

Smith 1998

Methods

Main study

Eight‐week intensive Summer Treatment Program, including a 6‐week, double‐blind, cross‐over trial with 4 interventions

  1. Three different doses of methylphenidate

  2. Placebo

Follow‐up study

Retrospective follow‐up study of 16 individuals who completed double‐blind, placebo‐controlled, cross‐over studies during 2 separate Summer Treatment Programs

Participants

Main study

Number of participants screened: not stated

Number of participants included: 49

Number of participants followed up: 46 (41 boys, 5 girls)

Number of withdrawals: 3
Diagnosis of ADHD: DSM‐III‐R

Age: mean 13.8 years (range 12 to 17)

IQ: mean 101 (range 65 to 129)

Methylphenidate naive: 28%

Ethnicity: Caucasian (85%), African American (15%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (50%), conduct disorder (15%)

Comedication: no

Sociodemographics: median family income US $38,500

Inclusion criteria

  1. Meeting DSM‐III‐R diagnostic criteria for ADHD

  2. 12th birthday before the protocol began

  3. Verbal IQ > 80

  4. No conditions that precluded a trial of stimulant medication or full participation in Summer Treatment Program academic and athletic activities

Exclusion criteria

  1. No information

Follow‐up study

Number of participants included: 16 (all boys)

Number of participants followed up: 16

Number of withdrawals: none

Diagnosis of ADHD: DSM‐III‐R

Age: mean (children) 10.2 years (range 8 to 11); mean (adolescents) 12.7 years (range 12 to 14.5)

IQ: mean (children) 109; mean (adolescents) 107

Methylphenidate naive: 4 (25%)

Ethnicity: Caucasian (100%)

Country: USA

Comorbidity: not stated

Comedication: not stated

Sociodemographics: family income (children US $37,943; adolescents US $49,650)

Inclusion criteria

  1. Meeting DSM‐III‐R diagnostic criteria for ADHD

  2. Younger than 12 years old when they participated in the children’s Summer Treatment Program

  3. Older than 12 years old when they participated in the adolescent Summer Treatment Program

  4. Full‐scale IQ > 80

  5. No learning disabilities or physical condition that precluded full participation in Summer Treatment Program classroom activities, athletic activities or a trial of stimulant medication

Exclusion criteria

  1. No information

Interventions

Main study

Participants were randomly assigned to 1 of the possible drug condition orders of 25 mg/d, 50 mg/d or 75 mg/d methylphenidate and placebo

Mean methylphenidate dosage: 0.17 mg/kg

Administration schedule: 3 times a day: 7:45 AM, 11:45 AM, 3:45 PM

Duration of each medication condition: 6 days

Washout before study initiation: 2 weeks

Medication‐free period between interventions: 16 hours

Titration period: none

Treatment compliance: not stated

Follow‐up study

Participants were randomly assigned to 1 of the possible drug condition orders of 0.3 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: 0.3 mg/kg

Administration schedule: twice a day: 8:00 AM and 12:00 PM

Duration of each medication condition: 1 day

Washout before study initiation: 2 weeks

Medication‐free period between interventions: 20 hours

Titration period: none

Treatment compliance: not stated. Adolescents were evaluated on a protocol that included placebo and 3 doses of methylphenidate. To facilitate comparison, doses for adolescents were converted to milligrams per kilogram, and the dose closest to 0.3 mg/kg was used in this study

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale (subscale: Inattention/Overactivity): completed every day by counsellors and classroom teachers

General behaviour

  1. IOWA Conners' Rating Scale (subscale: Oppositional Defiant): completed every day by counsellors and classroom teachers

Non‐serious adverse events

  1. Smith 1998: Side effects rating form (rating 12 potential side effects associated with stimulant medication on a 4‐point scale. A side effect rating of 3 (severe) was defined as troubling enough to contraindicate that dose of medication): completed every day by counsellors and parents during medication assessment

  2. Evans 1997: Ratings on major side effects associated with MPH: completed each day by classroom teachers

Notes

Sample calculation: no
Ethics approval: not stated

Comments from study authors

Main study

  1. One participant had a full‐scale IQ of 65 but was judged to be sufficiently intelligent to understand the behavioural contingencies, activity rules and social skills training provided in the programme

  2. Comment on high response rate in this study compared with other studies of adolescents with ADHD

    1. Higher response rate in this study may be due to greater statistical and methodological power to detect medication effects compared with previous studies, including (1) a larger sample, (2) a broader range of doses, (3) measurement in a well‐controlled, naturalistic setting, (4) repeated replications of medication conditions and (5) a statistical cutoff of 0.5 to define a positive response to medication

Follow‐up study

  1. For data compared in this study, randomisation and medication administration procedures were identical for children and adolescents, except that adolescents were evaluated on a protocol that included placebo and 3 doses of methylphenidate. To facilitate comparison, doses for adolescents were converted to milligrams per kilogram, and the dose closest to 0.3 mg/kg was used in this study

  2. Sample included only Caucasian males

  3. Only one third of participants in the adolescent programme had completed the summer treatment programme for children

  4. Students exhibited a much higher than expected positive response to stimulant medication

Key conclusions of study authors

Main study

  1. Results show that the shape of the dose‐response curve is influenced by the measurement method; most adolescents exhibited improved social behaviour when treated with methylphenidate, most positive effects of methylphenidate were achieved at the lowest dose and diminishing positive effects and increasing risk of negative effects were noted with successively higher doses

Follow‐up study

  1. Stimulant medication is equally effective with children and adolescents with ADHD who are engaged in similar activities

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no
Email correspondence with study authors: September 2014. We contacted study authors twice to ask for supplemental information/data but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Medication conditions were randomly assigned daily, with each condition occurring once a week. Thus, adolescents received a mode of 6 replications of each medication condition

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind; no further information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind; no further information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

"Some missing data was caused by holidays or absences from the program"

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

Low risk

Supported by grants from the National Institute on Drug Abuse, the National Institute of Mental Health, the National Institute on Alcohol Abuse and Alcoholism and the National Institute of Child Health and Human Development

Conflicts of interest: not declared

Smith 2004

Methods

n‐of‐1 randomised, double‐blind, cross‐over trial investigating the effects of Ritalin on the disruptive behaviour of a child diagnosed with ADHD. One‐day antecedent analysis in the clinic followed by an extended school‐based trial, during which participants received methylphenidate or placebo before evaluation

Duration: 15 days

Participants

Number of participants screened: not stated

Number of participants included: 1 boy

Number of participants followed up: 1

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV (subtype: not stated)

Age: 11 years

IQ: 124

Methylphenidate naive: no

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participant was randomly assigned to 1 of 2 possible drug condition orders of (20 mg/d) immediate‐release methylphenidate and placebo.

Out‐patient clinic setting, duration 1 day: ingestion of 20 mg methylphenidate or placebo 45 minutes before evaluation (randomised)

Cross‐over 4 hours later followed by second evaluation

School setting: 15 days (3 school weeks)

Administration schedule: once daily, mornings, 45 minutes before first evaluation

Medication‐free periods: on weekends; a total of 9 days with medication and 6 days without, randomly assigned throughout the trial period

Titration period: no

Washout period before study initiation: none other than weekends

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition ‐ School Version (18 items): completed by the teacher and an independent observer in the classroom. Classroom assessments were conducted within normal classroom activities in the morning (first 4 hours of school). Mean inter‐rater reliability 79% (range 56% to 100%)

Notes

Sample calculation: no

Ethics approval:not stated

Comments from study authors

  1. A major problem with single data point analyses is the difficulty involved in generalising outcomes to other settings and times

  2. Another difficulty involved inclusion of only 1 participant. However, the objectivity of clinic data in combination with checklist outcomes provides generalisable results, which reduces some of these limitations

Key conclusion of study authors

  1. The investigation suggests that the 1‐day antecedent analysis procedure could be used as an initial evaluation of the use of Ritalin

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: October 2013. We received from study authors supplemental information regarding diagnostic criteria

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A third party, who was not involved in the clinical procedure, determined the selection of actual medication packs

Allocation concealment (selection bias)

Low risk

A third party, who was not involved in the clinical procedure, determined the selection of actual medication packs

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

A third party, not involved in the clinical procedures, determined the selection of actual medication packs. Thus, persons conducting the direct evaluation were blinded to all medication manipulations (i.e. the child, parents, therapists and data collectors). However, no description was provided about whether medication and placebo pills were identical. "Prior to clinical assessment, two packets of medication were provided to the participant's parent. One package contained 20 mg of Ritalin and the other package contained a placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

A third party, who was not involved in the clinical procedure, determined the selection of actual medication packs. Thus, persons conducting the direct evaluation were blinded to all medication manipulations (i.e. the child, parents, therapists and data collectors)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol identified and no email from study author

Vested interest bias

Unclear risk

Not stated

Smithee 1998

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 26 (20 boys, 6 girls)

Number of participants followed up: not stated

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐IV (combined (77% ), inattentive (23%))

Age: mean 9.63 years (range 6.5 to 12)

IQ: mean 99.71

Methylphenidate naive: 24

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant/conduct disorder (n = 13a), anxiety disorders (n = 13), nocturnal enuresis (n = 4), vocal tics (n = 4), motor tics (n = 3)

Comedication: not stated

Sociodemographics: middle class status; mean social class of 2.12

Inclusion criteria

  1. 6.5 to 12 years of age

  2. Full‐scale, verbal or performance IQ ≧ 85

  3. Normal or corrected vision and hearing

  4. No physical handicaps

  5. No history of psychotic or neurological disorder

  6. No previous psychotropic treatment except for brief interventions of at most a couple of months (n = 2)

Exclusion criteria

  1. None stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 0.78 mg/kg/d

Administration schedule: 3 times daily for 14 days per medication conditions

Time points: morning, noon and 4:00 PM

Duration of each medication condition: 14 days

Washout before study initiation: not stated

Medication‐free period between interventions: no

Titration period: on days 1 to 3, participants received a dose 2.5 mg below their target dose of 0.3 mg/kg b.i.d. On days 4 to 7, dose was raised to 0.3 mg/kg b.i.d., and on day 8, the 4:00 PM dose was added

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Hyperactivity Questionnaire, parent and teacher: before trial and at each phase of 14 days

  2. IOWA, parent and teacher: before trial and at each phase of 14 days

  3. Hyperactivity Attention and Aggression Scales of the TOTS, parent and teacher: before trial and at each phase of 14 days

Non‐serious adverse events

  1. Weight with clothes but without shoes: observer, before and last day of each phase

  2. Interview to assess the presence of side effects: 11 somatic side effects and 3 mood problems

  3. Appetite decrease, increased thirst, dry mouth, stomachaches, nausea, headaches, dizziness, tremors, drowsiness, sleep problems, tics, crying, anger and sadness, EEG

Relatively few side effects were reported, probably in part because of adjustment in dosages in response to emergent symptoms

Notes

Sample calculation: no

Ethics approval: not stated

Comment from study authors

  1. Teachers tended to detect greater improvement with medication than did parents. Nevertheless, both parents and teachers detected significant improvement with treatment

Key conclusion of study authors

  1. Stimulant treatment increased accuracy and speed among younger children and curtailed variability of reaction time for the sample as a whole. However, methylphenidate did not affect ERPs. In combination, results imply that enhancement of performance by methylphenidate does not involve the demands of response selection examined in this trial

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with trial authors: March 2014. We received supplemental data from trial authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Medications were administered in random order

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications were administered in random order, under double‐blind conditions; capsules of identical appearance and taste

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

At the end of each phase, an investigator blind to pharmacological conditions administered a structured interview to the child’s parent concerning emergent side effects in the preceding period

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reports of side effects led to bind reduction of dosages or omission of planned increments for 3 participants under methylphenidate

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Table with numbers of reported somatic effects not given

Vested interest bias

Low risk

Research supported by National Institute of Mental Health (NIMH) Grant MH 38228; Rafael Klorman

Conflicts of interest: not declared

Solanto 2009

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: week‐long exposure to placebo and to each of 3 different dosage regimens of immediate‐release methylphenidate

Participants

Number of participants screened: not stated

Number of participants included: 30

Number of participants followed up: 25

Number of withdrawals: 5

Diagnosis of ADHD: DSM‐IV (combined (60%), inattentive (40%))

Age: not stated

IQ: not stated

Sex: not stated

Methylphenidate naive: All but 1 were stimulant‐naive

Ethnicity: not stated

Country: USA

Setting: hospital

Comorbidity: oppositional defiant disorder (combined 13%, predominantly inattentive 20%); learning disability (combined 40%, predominantly inattentive 20%); anxiety (combined 0%, predominantly inattentive 10%)

Comedication: no

Sociodemographics: minority representation (combined 53%, predominantly inattentive 20%)

Inclusion criteria

  1. Between 7 and 12 years of age

  2. Concordant reports: Conners' Parent Rating Scale and Conners' Teacher Rating Scale ‐ Combined group T scores ≧ 65 on both DSM‐IV Inattentive and DSM‐IV Hyperactive‐Impulsive Scales

  3. Predominantly Inattentive group T scores ≧ 65 on the DSM‐IV Inattentive Scale and < 65 on the DSM‐IV Hyperactive‐Impulsive scale

  4. Diagnosis of ADHD, combined or predominantly inattentive according to a structured diagnostic interview of the parent: Diagnostic Interview Schedule for Children and Adolescents

  5. Expert clinical diagnosis of ADHD based on review of all information collected

Exclusion criteria

  1. Currently receiving psychotropic medication

  2. Wechsler Intelligence Scale for Children, Third Edition: < 80

  3. Mood disorder, Tourette's disorder or psychotic disorder

  4. Sensory impairment or chronic medical or neurological condition including asthma that required systemic medication

  5. Colour blindness

Interventions

Participants were randomly assigned to 1 of 24 possible drug condition orders of low‐dose, medium‐dose and high‐dose methylphenidate and placebo

Mean methylphenidate dosage (Combined vs predominantly Inattentive): low dose 0.50 ± 0.12 vs 0.44 ± 0.13; medium dose 0.83 ± 0.20 vs 0.73 ±‐0.21; high dose 1.54 ± 0.31 vs 1.40 ± 0.38

Administration schedule: 3 times daily: morning, midday and 3:00 PM

Duration of each medication condition: 1 week

Washout before study initiation: not required

Medication‐free period between interventions: no

Titration period: open‐label lead‐in week, not specified whether before or after randomisation of treatment

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale, parent and teacher: baseline and end of each medication phase

  2. Conners' Teacher Rating Scale, parent and teacher: baseline and end of each medication phase

  3. ADHD Rating Scale, Fourth Edition: observer, weekly

Non‐serious adverse events

  1. Height, weight and vital signs: observer, weekly

  2. Side Effects Rating Scale: observer, weekly

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusion of study authors

  1. Results support the clinical utility of methylphenidate in the treatment of predominantly inattentive subtype and provide no evidence of differences in response between subtypes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: April 2014. We received supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Double‐blind, cross‐over with week‐long exposure to placebo and each of 3 different dosage regimens of immediate‐release methylphenidate in randomly assigned order

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, cross‐over with week‐long exposure to placebo and each of 3 different dosage regimens of immediate‐release methylphenidate in randomly assigned order. Study medications were prepared and coded by the hospital pharmacy using identical gelatin capsules for active medication and placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Across the 4 placebo and drug conditions, data points were missing as follows: Parent Conners' 1%, Teacher Conners' 6%, Parent SKAMP 1%, Teacher SKAMP 4%, Side Effects 0%. Missing scores were replaced by the mean of scores for that group in that condition

Selection bias: titration conducted but unclear whether before or after randomisation. No participants were excluded after the 1‐week titration period

Selective reporting (reporting bias)

Low risk

Protocol identified; all outcomes reported

Vested interest bias

High risk

No information

Conflicts of interest: Three study authors have served or received grants from pharmaceutical companies in the past

Stein 1996

Methods

Five‐week, triple‐blind, placebo‐controlled, cross‐over trial with 4 interventions

  1. Methylphenidate twice daily

  2. Methylphenidate 3 times daily

  3. Methylphenidate titration

  4. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 25 (all boys)

Number of participants followed up: 24

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐III‐R (combined (88%), inattentive (12 %))

Age: mean 8.0 years (range 6 to 12)

IQ: not stated

Methylphenidate naive: 44%

Ethnicity: Caucasian (96%), Hispanic (4%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (28%), conduct disorder (12%)

Comedication: no

Sociodemographics: Hollingshead SES category I: 8 (33%), II: 8 (33%), III: 3 (12.5%), IV: 5 (20.8%)

Inclusion criteria

  1. DSM‐III‐R ADHD diagnosis from parent interview using Disruptive Behavior Disorders module of the NIMH Diagnostic Interview Schedule for Children ‐ Parent‐rated (DISC‐P)

  2. Clinically significant ratings on Conners' Parent Rating Scale for Impulsivity/Hyperactivity factor (T > 65) or on Child Behavior Checklist (parent form)

  3. Attention factor (T > 65)

  4. Teacher ratings below 20th percentile for attention or hyperactivity problems on the ADD‐H Teacher Rating Scale (ACTeRS)

Exclusion criteria

  1. History of significant developmental delay (indicated by IQ testing or special educational services)

  2. Diagnosis of pervasive developmental disorder

  3. Unwillingness of parents or school personnel to meet study requirements

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 8.8 mg, 0.3 mg/kg/dose

Administration schedule: twice/3 times daily: 8:00 AM, 12:00 PM (and 2:00 PM)

Duration of each medication condition: 1 week

Washout before study initiation: ≥ 5 days

Medication‐free period between interventions: none

Titration period: initiated after randomisation

Treatment compliance: "there was generally good compliance, with a mean of 1.5 missed doses per child over the 5‐week course of the study"

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale (CPRS), 48 items; factors include Conduct Problem, Learning Problem, Psychosomatic Problems, Impulsivity/Hyperactivity and Anxiety. Collected on a weekly basis for 5 consecutive weeks, including baseline assessment

  2. ADD‐H Comprehensive Teacher Rating Scale (ACTeRS): collected on a weekly basis for 5 consecutive weeks, including baseline assessment

Serious adverse events

  1. None observed

Non‐serious adverse events

  1. Stimulant Side Effects Rating Scale (SSERS): collected on a weekly basis for 5 consecutive weeks, including baseline assessment

  2. Sleep log: Parents completed a sleep log to record the time when the child was sent to bed and fell asleep, as well as total sleep duration

  3. Actigraph: For 2 consecutive 18‐hour periods during each week, children wore an actigraph wrist monitor from 4:00 PM to 8:00 AM to record activity level, latency to sleep onset (time sent to bed to first minute of sleep), duration of sleep and number and duration of awakenings

Notes

Sample calculation: no

Ethics approval: approved by the Institutional Review Board of the University of Chicago

Comments from study authors

  1. Analysis revealed no significant effects of order on any of the measures of ADHD symptoms, sleep variables and side effects

  2. Our sample size and resultant statistical power were moderate, limiting our ability to detect mild or subtle effects

Key conclusions of study authors

  1. For many children with ADHD, t.i.d. dosing may be optimal

  2. Few differences in acute side effects have been noted between b.i.d. and t.i.d. methylphenidate dosing

  3. Dosing schedule should be selected according to severity and time course of ADHD symptoms, rather than in anticipation of dosing schedule‐related side effects

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: July 2014. Supplemental information regarding randomisation and whether all planned outcomes were measured and reported was received from study authors. Unfortunately, study authors no longer had access to the dataset; therefore it has not been possible to receive supplemental data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to 1 of 4 different orders of drug administration. The research pharmacist used a random numbers table to assign participants to different orders of administration

Allocation concealment (selection bias)

Low risk

"All medication and placebos were prepared by the study pharmacist and placed in opaque gelatin capsules" In addition to the pharmacist, 1 investigator had access to the dosage code in the event of a medical emergency that necessitated breaking the blind

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Triple‐blinded: Participants always took 3 capsules daily during the study. "Active drug phase (b.i.d. or three t.i.d.) was always preceded by a titration phase. The purpose of the titration phase was to introduce a typical dose of MPH gradually, so that any observation of side effects during the b.i.d. and t.i.d. dosing phases could not be attributed to rapid introduction of MPH"; "All medication and placebos were prepared by the study pharmacist and placed in opaque gelatin capsules"; "In addition to the pharmacist, one investigator had access to the dosage code in the event of a medical emergency that necessitated breaking the blind"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Triple‐blinded: In addition to the pharmacist, one investigator had access to the dosage code in the event of a medical emergency that necessitated breaking the blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

One drop‐out. Data collected on this participant were used in the descriptive information for baseline, t.i.d. and titration phases

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

All planned outcomes were measured and reported

Vested interest bias

Low risk

None declared. The work was supported by the Smart Family Foundation

Conflicts of interest: no affiliations with pharmaceutical companies stated

Stein 2003

Methods

Four‐week, double‐blind, placebo‐controlled, cross‐over trial in which participants were randomly assigned to

  1. Three doses of methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 47 (33 boys, 14 girls)

Number of participants followed up: 39

Number of withdrawals: 8

Diagnosis of ADHD: DSM‐IV (combined (68%), hyperactive‐impulsive (0%), inattentive (32%))

Age: mean 9.02 years (range 5 years 11 months to 16 years)

IQ: mean 106.8

Stimulant‐naive: 70%

Ethnicity: Caucasian (89.4%), African American (4.3%), Hispanic (2.1%), other (4.3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (17%), encopresis/enuresis (10.6%), tic disorder (2.1%)

Comedication: no

Sociodemographics: predominantly mid to upper socioeconomic status referral base of the clinic

Inclusion criteria

  1. DSM‐IV criteria for ADHD

Exclusion criteria:

  1. Mental retardation

  2. Severe mood disorders (requiring antidepressant or concurrent psychotropic medications)

  3. Tourette's syndrome, seizure disorders or other medical disorders associated with symptoms that may mimic ADHD (e.g. thyroid disorder)

  4. Children taking systemic medications

Interventions

Participants were randomly assigned to different orders of OROS methylphenidate (18 mg, 36 mg, 54 mg) and placebo. No child could start with the 54‐mg dose, and 1 child who weighed < 40 kg did not receive the 54‐mg dose to minimise potential side effects in smaller children

Administration schedule: once daily

Duration of each medication condition: 7 days

Washout before study initiation: 2‐week washout period for children who took stimulant medication before study start

Medication‐free period between interventions: no

Titration period: none

Treatment compliance: 92% of all study medications were given

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, Fourth Edition: parent‐rated, at baseline and weekly during interventions

  2. ACTeRS: teacher‐rated at baseline and weekly during interventions

Non‐serious adverse events

  1. Side Effect Rating Scale (SERS; Barkley 1990): parent‐rated at baseline and weekly during interventions

  2. Vital signs (weight, height, blood pressure, pulse and temperature): obtained weekly by clinical staff

  3. Children’s Sleep Questionnaire

Notes

Sample calculation: no

Ethics approval: approved by the Institutional Review Boards of The University of Chicago, Children’s National Medical Center and the General Clinical Research Center Advisory Council

Comments from study authors

  1. Forced titration procedure deserves some comment

  2. The advantage of this procedure is increased potential to determine optimal response. However, this procedure is likely to result in increased reports of stimulant side effects compared with a more gradual titration procedure

In interpreting the results, several limitations should be kept in mind

  1. Potential expectancy biases or placebo effects need to be considered because each medication differed in appearance, size and colour, and, in the case of the 54‐mg condition, number of capsules

  2. Other limitations include the short‐term nature of the study, the relatively small number of participants with ADHD predominantly inattentive subtype and the measures of ADHD symptoms used that were rating scales rather than behavioural observations or laboratory measures of attention

Key conclusions of study authors

  1. In children with ADHD combined subtype ‐ the most common subtype of ADHD ‐ increasing doses of stimulant medication were associated with increased improvement in inattention and hyperactivity symptoms

  2. In children with ADHD predominantly inattentive subtype, symptom improvement occurred at lower doses and less benefit was derived from higher doses

  3. In both ADHD subtypes, higher doses were associated with parent ratings of increased insomnia and decreased appetite

  4. Children who were homozygous for the less common, 9‐repeat DAT1 30‐UTR genotype displayed a distinct dose–response curve from that of other genotype groups, with absence of typical linear improvement when the dose was increased from 18 mg to 36 mg and 54 mg

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Correspondence with study authors: August 2014. We contacted study authors to request additional data. Study authors no longer have access to the data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Dosing schedules were assigned from a randomly ordered list of all dosing schedules

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Each medication differed in appearance, size and colour and, in the case of the 54‐mg condition, number of capsules. The placebo capsule was slightly larger than the methylphenidate preparations. Study authors conducted several analyses to investigate the impact of these differences in appearance on the findings of the study (almost no impact on the results) but still considered risk of bias to be high. Parents and clinicians who were blinded to genotype and medication status rated ADHD symptoms, impairment and stimulant side effects each week

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Parents and clinicians who were blinded to genotype and medication status rated ADHD symptoms, impairment and stimulant side effects each week

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Any child who could not complete a treatment phase was classified as a premature discontinuation. Their data were included for all phases that were completed. No description on how many participants the analyses were based on

Selective reporting (reporting bias)

High risk

Data were included for all phases that were completed. There is no description that reveals how many people the analyses were based on. In the abstract belonging to the study (Stein et al. 2004), researchers write that they will "prospectively evaluate the effects of different doses of stimulant medication on these and other sleep problems". This is not provided in any of the articles included in the Stein 2003 study

Vested interest bias

High risk

This study was supported by the National Institute of Mental Health, the General Clinical Research Center Program of the National Center for Research Resources and the National Institutes of Health, Department of Health and Human Services

Conflicts of interest: Drs. Stein, Robb, Conlon and Newcorn participate in the Speakers' Bureau for McNeil Consumer and Specialty Pharmaceuticals, and Drs. Stein and Newcorn are members of the Concerta National Advisory Committee

Stein 2011

Methods

Eight‐week, double‐blind, cross‐over trial comparing the following

  1. Extended‐release d‐MPH (10 mg, 20 mg, 25 to 30 mg)

  2. Extended‐release MAS (10 mg ,20 mg, 25 to 30 mg)

with a week of randomly assigned placebo within each drug period

Participants

Number of participants screened: 77

Number of participants included: 65

Number of participants followed up: 56

Number of withdrawals: 9

Demographic data on the 56 participants

Diagnosis of ADHD: DSM‐IV (combined (67%), hyperactive‐impulsive (0%), inattentive (33%))

Age: mean 11.78 years (range 9 to 17)

IQ: > 70

Sex: 41 boys, 15 girls

MPH‐naive: 35.7%

Ethnicity: Caucasian (41.0%), African American (41.0%), Asian (2%), Hispanic (7%), Bi‐racial/mixed (9%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD, any subtype

  2. Signed informed consent and assent

  3. Clincal Global Impressions ‐ Severity for ADHD (CGI‐S‐ADHD) rating ≥ 4

  4. Findings on physical exam, laboratory studies, vital signs and ECG are judged to be normal for age

  5. Pulse and blood pressure are within 95% of age and sex means

  6. Able to complete study instruments and swallow capsules

  7. Willing to commit to the entire visit schedule for the study, including ≥ 1 visit to UIC Medical Center

Exclusion criteria

  1. Mental retardation, autism, severe mood disorders, Tourette's disorder, seizure disorders or other medical disorders that were contraindications of stimulant treatment that mimic ADHD (e.g. thyroid disorder)

  2. Non‐responder to either medication at doses offered in the study in an adequate trial. Must not have experienced disabling adverse effects with either medication

  3. Concomitant psychotropic medications or medications that might have a CNS effect are required

  4. Any other medical condition that represents a contraindication for either treatment

  5. History of alcohol or drug abuse in the past 3 months, or a positive urinary toxic screen on initial evaluation that is not explained by a time‐limited medical circumstance

  6. Females of childbearing age who are sexually active, do not use acceptable birth control (double protection method) and, after counselling, are unwilling to do so

  7. History of allergic reactions to multiple medications

  8. History of psychosis

  9. Diagnosis of bipolar disorder

Interventions

Participants were randomly assigned to 3 dose conditions of ER d‐MPH and ER MAS administered sequentially from lowest to highest dose with a randomly assigned week of placebo during each period

Methylphenidate dosage: 10 mg, 20 mg and 25 to 30 mg. Maximum dose was 25 mg in smaller children (i.e. < 35 kg) to minimise potential side effects

Administration schedule: once daily

Duration of each medication condition: 1 week

Washout before study initiation: 2‐day washout period before beginning of the trial. No washout period between treatment periods

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Parent Rating Scale, Fourth Edition, rated weekly

Non‐serious adverse events

  1. Stimulant Side Effects Rating Scale (Barkley 1990): rated weekly by parents

  2. Vital signs (weight, height, blood pressure, pulse and temperature): obtained weekly

  3. ECG before and after study

  4. Sleep measured by actigraphy and questionnaires (from abstract)

Notes

Sample calculation: no

Ethics approval: approved by the Institutional Review Board of University of Illinois at Chicago

Comment from study authors

  1. Short duration of time children were maintained on each dose and fixed dose titration

Key conclusions of study authors

  1. Both ER d‐MPH and ER MAS were associated with significant, dose‐dependent reductions in ADHD symptoms

  2. Decreased appetite and insomnia were more common and were seen at higher dose levels for both stimulants

  3. Dose level, rather than stimulant class, was strongly related to medication response

  4. Although most children responded similarly to both stimulants, 14.3% of total samples were responders to ER d‐MPH only, and 12.5% responded only to ER MAS

  5. Future comparative effectiveness studies with multiple informants and larger samples over longer time periods are necessary to develop a data‐driven, personalised approach to ADHD treatment

Comment from review authors

  1. Protocol states that an exclusion criterion was "Non‐responder to either medication at the doses offered in the study in an adequate trial". This is not written in the full text of the study. Asked study author about this issue, who replied, "Although non response was an exclusion [criterion] a priori, in fact this did not come up and no cases were excluded from participation based upon this". Even though this means that children in the trial were not only responders, we still choose to analyse this study as part of the studies that included only responders

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; see comments above

Email correspondence with study authors: November and December 2013. We received supplemental data from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Order of drug was randomly assigned so that 50% started with ER d‐MPH and 50% started with ER MAS; also a randomly assigned week of placebo during each period

Allocation concealment (selection bias)

Low risk

Weekly blister packs containing capsules of study drug, which were indistinguishable from each other

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participant, caregivers, outcome assessors

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant, caregivers, outcome assessors

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants who received ≥ 2 weeks of study drug to ensure that all participants had been exposed to ≥ 1 week of active drug were analysed

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No. Regarding the Wiebe et al. abstract, a paper is almost ready for submission

Vested interest bias

High risk

Investigator‐initiated study sponsored by Novartis Pharmaceuticals, with additional support provided by the University of Illinois at Chicago (UIC) Center for Clinical and Translational Science (CCTS)

Conflicts of interest: Some study authors are affiliated with pharmaceutical companies

Stoner 1994

Methods

Two n‐of‐1, double‐blind, placebo‐controlled, cross‐over, randomised controlled trial. Both cases received 4 levels of the following

  1. Methylphenidate 5 mg, 10 mg and 15 mg

  2. Placebo

After the double‐blind trial, the code was broken, and the best dose was administered to the participant. Follow‐up data on the best dose were also measured. Follow‐up data were recorded anecdotally for the first case (Dan) and systematically for the second case (Bill)

Participants

Number of participants screened: 2 boys

Number of participants included: 2

Number of participants followed up: 2

Number of withdrawals: none

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: Dan 9 years, Bill 13 years

IQ: not stated

Methylphenidate naive: 100%

Ethnicity: not stated

Country: USA

Setting: not stated

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to different orders of 3 doses of methylphenidate and placebo

Mean methylphenidate dosage: not relevant

Administration schedule: once daily at breakfast

Duration of each medication condition: Dan received placebo for 3 days, and 5 days of treatment with 5 mg, 10 mg or 15 mg methylphenidate consecutively. Bill received placebo for 3 days, and 3 days of treatment with 5 mg, 10 mg or 15 mg methylphenidate consecutively

Washout before study initiation: treatment‐naive

Medication‐free period between interventions: 24 hours between doses

Titration period: none

Treatment compliance: Parents were instructed to initial a monitoring form each time they gave a dose to their child after breakfast. No further description of this was provided

Regarding the follow‐up trial: Dan 15 mg; Bill 10 mg (5 mg twice daily)

Outcomes

General behaviour

  1. Child Attention Problems Scale (CAP, 12‐items; Barkley 1990): teacher‐rated, at the end of each medication trial

Non‐serious adverse events

  1. Stimulants Drug Side Effect Rating Scale: rated by participants, parents and Bill’s teacher at the end of each medication trial

No relevant outcomes measured in the follow‐up phase

Notes

Sample calculation: not relevant

Ethics approval: not stated

Comments from study authors

  1. Although results from both studies are promising, they must be interpreted with great caution because various methodological and design features introduced threats to internal validity. For example, inclusion of a no‐medication day between trial phases would be more in keeping with standard practice of clinical research trials of medication

  2. Even though methylphenidate has a relative low half‐life (4 to 6 hours), it is thought to be completely eliminated from the body within 24 hours of ingestion; a carry‐over of effect from one trial to another may have occurred

Key conclusion of study authors

  1. Curriculum‐based measurement data collected during short medication trials can be used to select a dose of methylphenidate that is likely to be beneficial for a student’s ongoing academic growth

Comments from review authors

  1. As we do not know whether any of the children were intellectually disabled, we can use the study results only in sensitivity analyses

  2. The article reports some side effects, but we could not obtain more information on this from the study authors; therefore, these data are not used in the review

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: no

Email correspondence with study author: January 2014. Wrote to study author to request additional information regarding ethics approval, intellectual disability, etc., but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The order of medication levels was determined randomly. In packing the envelopes, 1 of the study authors arranged the coded envelopes according to the randomly determined order of trials

Allocation concealment (selection bias)

Low risk

Each medication level was prepared and packed in separate envelopes by a pharmacist

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Each dose of methylphenidate and placebo was grounded into a powder, mixed with an inert compound, and was sealed in a small coloured drug capsule, so that doses were identical in appearance and taste. Thus, the pharmacist, the data collectors, the participants and their families did not know the order of drug administration

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Each dose of methylphenidate and placebo was grounded into a powder, mixed with an inert compound, and was sealed in a small coloured drug capsule, so that doses were identical in appearance and taste. Thus, the pharmacist, the data collectors, the participants and their families did not know the order of drug administration

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All data were provided

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol was identified, and no answer was received from the study author

Vested interest bias

Low risk

National Association of School Psychologists

Conflicts of interest: not declared

Sumner 2010

Methods

Three‐week, triple‐blind, randomised, cross‐over trial with 2 interventions

  1. Methylphenidate or atomoxetine

  2. placebo

Participants

Number of participants screened: not stated

Number of participants included: 31

Number of participants followed up: not stated

Number of withdrawals: 1
Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean not stated (range 6 to 14 years)

IQ: not stated

Sex: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: none

Comedication: none

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 14 years

  2. ADHD, any subtype, according to DSM‐IV criteria

  3. ADHD symptoms of sufficient severity to warrant pharmacotherapy, but patient had never received the active medications evaluated in this study or had not exhibited significant treatment‐limiting adverse effects while using them

Exclusion criteria

  1. Enrolment in self contained special education classes

  2. Females with prior onset of menses: positive urine pregnancy tests at enrolment

  3. Structural cardiac abnormalities, cardiomyopathy, serious heart arrhythmias or other serious cardiac abnormalities

  4. Considered by the investigator to be medically inappropriate for inclusion in a study using placebo

  5. Prior or present use of alcohol or illicit drugs

  6. Major medical or neurological disorders that could affect motor activity, attention, school attendance or ability to follow the study protocol

  7. Diagnosis of a current anxiety disorder, tics, Tourette's syndrome, major depressive disorder

  8. History of bipolar disorder, dysthymia or psychosis, or a family history of Tourette's syndrome

  9. Treatment with a monoamine oxidase inhibitor within 14 days before screening

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders: placebo, low, medium doses of OROS methylphenidate; or low, medium doses of OROS methylphenidate, placebo

Doses: low (18 mg), medium (participants < 50 kg: 27 mg; participants > 50 kg: 36 mg)

Administration schedule: not stated

Duration of each medication condition: 1 week

Washout before study initiation: 1 week

Titration period: no

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. ADHD Rating Scale: clinician‐rated, weekly

  2. Conners' Parent Rating Scale ‐ Revised: short form, parent‐rated, weekly

Notes

Sample calculation: no
Ethics approval: yes

Comments from study authors (limitations)

  1. Small participant sample size

  2. Short medication washout

  3. Lack of information about prior ADHD medication use

  4. Absence of a true placebo arm in a parallel design

Key conclusions of study authors

  1. Use of an objective measure, coupled with more stringent treatment response thresholds, may better inform treatment decisions by clinicians and may reduce costs and enhance assay sensitivity in ADHD clinical trials

  2. Longer‐term, multi‐centre, prospective, parallel‐group studies incorporating true placebo arms and more heterogeneous participant populations are warranted to confirm these preliminary findings

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; exclusion of participants who have exhibited significant treatment‐limiting adverse effects while treated with methylphenidate
Any withdrawals due to adverse events: not stated
Email correspondence with study authors: August to September 2013. We attempted to obtain supplemental information regarding characteristics of participants, withdrawals, funding and efficacy data from study authors but without success

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned to 1 of 2 treatment sequence groups

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Triple‐blinded: Study participants, their parents/guardians and clinicians were blinded to study assignment in terms of medications and dosing sequences. An unblinded prescribing physician and an unblinded study co‐ordinator did not participate in clinical ratings. Placebo was matched to blinded medication, so that the 2 were indistinguishable

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

See above

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

One participant withdrew before the placebo visit and was not included in these analyses

Selective reporting (reporting bias)

Unclear risk

No protocol was available

Vested interest bias

Unclear risk

It was not clear who sponsored the study, but someone did (see authors' affiliations)

Conflicts of interest: Calvin R. Sumner is an employee of and an equity holder for the study sponsor. Virginia S. Haynes, PhD, is an employee of 3i Global (Basking Ridge, NJ) and a paid consultant for the study sponsor. Martin H. Teicher, MD, PhD, served as paid consultant and clinical investigator for the sponsor. Jeffrey H. Newcorn, MD, serves as advisor and consultant for Lilly, Ortho‐McNeil Janssen, Schering‐Plough and Shire. He receives research support from Lilly, Ortho‐McNeil Janssen and Shire

Sunohara 1999

Methods

Cross‐over trial with 3 arms

  1. Methylphenidate: low dose

  2. Methylphenidate: high dose

  3. Placebo

Phases: 3

Participants

Number of participants screened: not stated

Number of participants included: 20 (16 boys, 4 girls)

Number of participants followed up: 20

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 10.5 years (range 10 to 12)

IQ: < 80

Methylphenidate naive: none

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 4), learning disability (n = 8)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children 10 to 12 years of age meeting DSM‐II‐R criteria for ADHD recruited from Child Development Centre at Hospital for Sick Children, Canada

Exclusion criteria

  1. Conduct disorder, externalising disorder, anxiety

  2. IQ < 80

Interventions

Participants were randomly assigned to low‐ (mean 0.28 mg/kg) and high‐dose (mean 0.56 mg/kg) methylphenidate and placebo in the morning and afternoon

Duration of each medication condition: 2 days

Washout before study initiation: no

Titration period: no

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Reported as adverse effects. There were 0 adverse effects at the higher dose for all children

Notes

Sample calculation:not stated

Ethics approval:no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: unclear

Key conclusion of study authors

  1. No adverse effects of the higher dose were reported for any children

Email correspondence with study authors: April 2014. Wrote to study authors to ask for additional data but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

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

Not stated

Selective reporting (reporting bias)

High risk

Not clear

Vested interest bias

High risk

Research was supported by a RESTRACOM graduate studentship for The Hospital for Sick Children Research Institute and Novartis Pharmaceuticals

Swanson 1998

Methods

Randomized, double‐blind, cross‐over study with 6 interventions

  1. Methylphenidate

  2. Placebo

Adderall in 4 different doses

  1. 5 mg

  2. 10 mg

  3. 15 mg

  4. 20 mg

Participants

Number of participants screened: 36.

Number of participants included: 33

Number of participants followed up: 29

Number of withdrawals: 4

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 10.58 years (range 7 to 14)

IQ: > 80

Sex: 26 boys, 7 girls

Methylphenidate naive: 0%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (laboratory classroom)

Comorbidity: none

Comedication: none

Sociodemographics: not stated

Inclusion criteria

  1. Age between 7 and 14 years

  2. Diagnosis of ADHD by DSM‐IV

  3. History of clinically significant response to typical doses of methylphenidate (5 mg to 20 mg, b.i.d. or t.i.d.)

Exclusion criteria

  1. Blood pressure outside 95th percentiles for age and sex

  2. Wechsler Intelligence Scale for Children ‐ Second Edition: score < 80

  3. Abnormalities noted upon physical examination

  4. Current treatment with a non‐stimulant medication for ADHD

  5. Comorbid disorder

  6. History of aggressive behaviour serious enough to preclude participation in regular classroom activities

Interventions

Participants were randomly assigned to 1 of 36 possible drug condition orders of unknown dose of methylphenidate, placebo and 4 doses of Adderall

Mean methylphenidate dosage: not stated

Administration schedule: once, morning

Duration of each medication condition: 7 days

Washout before study initiation: not stated

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: 30 of 33 participants completed the 7‐week trial

Outcomes

ADHD symptoms

  1. SKAMP Teacher Rating Scale, as modified by Greenhill into 2 domains of Attention and Deportment (i.e. positive behaviour): completed by 1 of the teachers after each 45‐minute class period (i.e. every 1.5 hours on the Saturday of each week, the teacher monitoring behaviour rated each student on each item)

Serious adverse events

  1. No unusual or serious side effects were noted in this study

Non‐serious adverse events

  1. After each classroom period (6 times per day), teachers assessed each student on the MTA 10‐Item Stimulant Side Effects (SSE) rating scale

  2. Parents completed a side effects rating scale that included SSE items plus an additional item (trouble sleeping) 3 times per day, on Monday, Wednesday and Friday of each week

Notes

Sample calculation: no

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; only known methylphenidate responders were included

Comments from study authors

  1. Laboratory classroom experience may represent a novel experience for participants, and this could alter response to medication

  2. Group interactions in a classroom with 16 or 17 students with ADHD may be different from those in the typical classroom of 20 to 30 students including 1 to 2 children with ADHD

  3. Behaviour of 16 to 17 students with ADHD in a classroom with a student:teacher ratio of about 8:1 may be different from that in a classroom with the typical ratio of 20:1 to 30:1

Key conclusions of study authors

  1. This documentation of efficacy in a controlled study supports the addition of Adderall to the armamentarium of psychotropic medications for treatment of ADHD

  2. Differences in time‐response patterns of Adderall and methylphenidate may help tailor treatment to meet specific clinical needs of different children with ADHD

Email correspondence with study authors: January 2014. No reply has been received

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Latin‐square design (with provisions for 36 participants) was used to determine the within‐participant order of administration of the 6 medication conditions, so that for each of the first 6 weeks, approximately one sixth of participants would be assigned to each of the 6 conditions. Separate randomisation was used to determine assignment of conditions across participants for week 7, to provide an opportunity to make up missed weeks

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

For each condition, a pharmacist prepared a set of 7 identical capsules, all containing placebo (lactose), 1 of the 4 doses of Adderall or methylphenidate

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

30 of 33 participants completed the 7‐week trial; 98% of data were collected as planned. For each individual, missing data (1.1% for SKAMP, 0% for Stimulant Side Effects) were replaced by the average of values from adjacent time points (or adjacent values for session 1 or 6)

Selective reporting (reporting bias)

Low risk

No information

Vested interest bias

High risk

Supported by a grant from Richwood Pharmaceutical Company

Swanson 1999

Methods

Four‐period, double‐blind, randomised, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases

  1. b.i.d.: 2 doses of immediate‐release methylphenidate 4,5 hours apart

  2. Flat: morning dose of 80% of b.i.d. morning dose and the rest at 30‐minute intervals over 6 hours, starting 1.5 hours after first dose

  3. Ascending: morning dose of 40% of b.i.d. morning dose and that rest as increasing doses at 30‐minute intervals over 5 hours, starting 1.5 hours after first dose

Participants

Number of participants screened: not stated

Number of participants included: 38 (33 boys, 5 girls)

Number of participants randomly assigned: 34. Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 31

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 9.2 years (range 7 to 12)

IQ: not stated

Methylphenidate naive: 0%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (laboratory classroom)

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosis of ADHD

  2. Onset by 7 years of age

  3. Receiving current treatment with methylphenidate doses of 5 to 15 mg administered 2 or 3 times per day

  4. 7 to 12 years of age

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of methylphenidate and placebo

Mean MPH dosage: not stated. Nominal dose: 20 mg/d

Administration schedule: 30‐minute intervals from 7:30 AM to 3:00 PM

Duration of each medication condition: 1 day

Washout before study initiation: none

Titration period: none (participants were kept on current standard treatment in‐between trial days)

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale (SKAMP): teacher‐rated, 4 times during the day

  2. Conners, Loney and Milich Scale (CLAM): teacher‐rated, 4 times during the day

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Of the 38 children recruited for this trial, only 34 entered

Key conclusion of study authors

  1. Acute tolerance to methylphenidate appears to exist

Comment from review authors

  1. Publication includes 2 studies: In study I, relative efficacy was determined for 3 dosing patterns of methylphenidate vs placebo. In study II, tolerance was assessed by comparison of 3‐times‐a‐day regimens of methylphenidate only. The latter is not part of the data extraction

Email correspondence with study authors: May 2014. Emailed study authors to ask for additional information. No reply has been received

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

On subsequent Saturdays, each child received (in random order) 1 of 3 possible drug condition orders of methylphenidate and placebo

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All treatments were administered in identical capsules

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only completers were followed up

Selective reporting (reporting bias)

Low risk

No protocol was published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Supported by ALZA Corporation, Palo Alto, California

Swanson 2002a

Methods

Randomised, double‐blind, cross‐over design

  1. Immediate‐release methylphenidate t.i.d. or methylphenidate experimental administration

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 32 (28 boys, 4 girls)

Number of participants followed up: 30

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (combined (93.8%), hyperactive‐impulsive (6.2%), inattentive (0%))

Age: mean 9.9 years (range 7 to 13)

IQ: not stated

Methylphenidate naive: 0%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic (laboratory classroom)

Comorbidity: 0%

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD

  2. Normal blood pressure

  3. Not physically ill

  4. Able to understand that they could withdraw from the study at any time

Exclusion criteria

  1. Oppositional defiant disorder

  2. Conduct disorder

  3. Mood disorders

  4. Anxiety disorders

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of 5 mg, 10 mg or 15 mg t.i.d., or 18 mg/d, 36 mg/d or 54 mg/d administered in bolus at 7:30 AM and once every 30 minutes for 8 hours of methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 3 time points

Duration of each medication condition: 1 day

Washout before study initiation: not stated

Medication‐free period between interventions: none

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale (SKAMP)

General behaviour

  1. Actigraph activity measurements

Non‐serious adverse events

  1. Proof of Concept Study (n = 32)

  2. Teachers: appetite loss (t.i.d., n = 6; ascending, n = 7)

  3. Parent reports of somatic complaints (primarily headache or stomachache): t.i.d. methylphenidate (n = 9), ascending (n = 5)

  4. Sleep onset was delayed slightly in the medication conditions (ascending, 0.65 h; t.i.d., 0.55 h)

Notes

Sample calculation: not stated

Ethics approval: yes

Comments from study authors (limitations)

  1. Lack of normal control participants

  2. Use of subjective rating measures

  3. Use of different actigraph modes of operation

  4. Lack of a systematic evaluation of whether baseline levels of behaviour were predictive of medication effects

Key conclusions of study authors

  1. Combination of an initial bolus of methylphenidate and an ascending pattern of small doses significantly decreased hyperactivity and reduced inappropriate behaviour

  2. Studies showed acute tolerance to clinical doses of methylphenidate and application of this: creation of an ascending drug delivery pattern (OROS) of rapid onset and with long duration of effect

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: All children in the trial were undergoing treatment with methylphenidate when they were enrolled

Email correspondence with study authors: Study authors were contacted twice by email and were asked for much supplemental information regarding data, but we have received no data; therefore, most of the data from this study cannot be used in this review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomized, 3‐way, crossover trial in which a double‐blind, double‐dummy procedure was used"

Allocation concealment (selection bias)

Unclear risk

Not enough information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not enough information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not enough information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not enough information

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no, but all children in the trial were undergoing treatment with methylphenidate when they were enrolled

Selective reporting (reporting bias)

Low risk

"Two additional items developed (from SKAMP) for the NIMH Collaborative Multisite Multimodal Treatment Study of Children With ADHD (MTA) (Greenhill et al., 2001) were included in the classroom ratings but were not included in the analyses of this study"

Vested interest bias

High risk

"Supported by the ALZA Corporation"

Swanson 2002b

Methods

Randomized, 3‐way, cross‐over trial with 2 interventions

  1. Methylphenidate (t.i.d. or OROS extended release)

  2. Placebo

Phases: 3

Participants

Number of participants screened: not stated

Number of participants included: 64

Number of participants followed up: 59

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐IV (combined (82.8%), hyperactive‐impulsive (not stated), inattentive (not stated))

Age: mean 9.2 years (range 6 to 12)

IQ: not stated

Sex: 81.3% boys

Methylphenidate naive: none

Ethnicity: Caucasian (82.8%), African American (not stated), Asian (not stated), Hispanic (not stated)

Country: USA

Setting: out‐patient clinic (laboratory classroom)

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV criteria for ADHD diagnosis, treated with immediate‐release methylphenidate 5 mg to 15 mg b.i.d. or t.i.d.

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of 5 mg, 10 mg or 15 mg t.i.d., or 18 mg, 36 mg or 54 mg OROS methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: t.i.d. 7:30 AM, 11:30 AM and 3:30 PM; OROS 7:30 AM

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: no measure

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale (SKAMP): rated by laboratory school teacher on attendance on Saturdays

  2. Conners, Loney and Milich Scale (CLAM): rated by parent and community teacher on Fridays

  3. Swanson, Nolan and Pelham (SNAP): rated by community teacher and parent at the end of each week

Non‐serious adverse events

  1. "In addition to the effectiveness and efficacy measures, adverse effects were actively solicited and assessed and information about sleep, appetite, and tics was collected using a parent questionnaire"

  2. Vital signs (blood pressure, pulse) after each classroom session on Saturdays in the laboratory school setting

  3. Sleep: activity monitoring and parental assessment

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusion of study authors

  1. Pharmacokinetic/Pharmacodynamic modelling provided the target for a novel drug delivery pattern to overcome these shortcomings: an initial bolus to elicit a rapid response and an ascending pattern of drug delivery to maintain constant effects

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; "The children with ADHD in the proof‐of‐concept and proof‐of‐product studies were selected based on a history of clinical response to stimulant medication, and this limits the extrapolation of the findings to the drug‐naive population"

Any withdrawals due to adverse events: no

Email correspondence with study authors: March 2014. We contacted study authors twice by email and asked for supplemental information regarding data, but we have received no data; therefore most of the data from this study cannot be used in this review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Double‐blind procedures were implemented by administration of methylphenidate or placebo in capsules

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, double‐dummy procedure was used to disguise the 3 treatments

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Children with ADHD in proof‐of‐concept and proof‐of‐product studies were selected on the basis of a history of clinical response to stimulant medication; this limits extrapolation of findings to the drug‐naive population

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

High risk

Supported by ALZA Corporation

Swanson 2004b

Methods

Multi‐centre, double‐blind, double‐dummy, placebo‐controlled, cross‐over study in an analogue classroom setting comparing 3 treatment conditions

  1. Metadate CD

  2. Concerta

  3. Placebo

Each treatment intervention lasted a week

Participants

Number of participants screened: 214

Number of participants included: 184

Number of participants followed up: 157/184 ITT

Number of withdrawals: 27

Diagnosis of ADHD: DSM‐IV (combined (82.2%), hyperactive‐impulsive (4.8%), inattentive (13.0 %))

Age: mean 9.6 years (range 6 to 12)

IQ: > 80

Sex: 131 boys, 48 girls

Methylphenidate naive: none

Ethnicity: Caucasian (70%), African American (11.5%), Asian (1.7%), Hispanic (12.5%), other (5.3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: Approximately 25% had a comorbid condition; anxiety and oppositional defiant disorder were most frequent. Females had a greater rate of comorbid anxiety disorder (from Sonuga‐Barke 2007). Anxiety: 20.8 girls, 5.9 boys. Oppositional defiant disorder: 12.5 girls, 8.8 boys. Insomnia: 2.1 girls, 5.1 boys.

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children (6 to 12 years old) who had clinical diagnoses of a DSM‐IV subtype of ADHD (inattentive type, hyperactive‐impulsive type or combined type) were recruited

  2. Treatment with methylphenidate in doses of 10 mg/d to 60 mg/d (5 mg to 20 mg per administration, 1 to 3 times a day)

  3. Children were deemed otherwise healthy by means of a medical history, physical examination, vital signs measurement (blood pressure, heart rate, respiration and temperature) and clinical laboratory assessments (haematology and urinalysis)

  4. In addition, children had to demonstrate the ability to swallow placebo study treatment capsules whole and without difficulty

  5. Receiving an approved form of methylphenidate with demonstrated clinical improvement during this treatment

Exclusion criteria

  1. Intelligence quotient < 80 or inability to follow or understand study instructions

  2. Pregnancy

  3. History of seizure or tic disorder

  4. Family history of seizure or Gilles de La Tourette’s syndrome

  5. Congenital cardiac abnormality

  6. History of cardiac disease including myocardial infarction within 3 months of study entry

  7. Glaucoma

  8. Hyperthyroidism

  9. History of substance abuse or caretaker with history of substance abuse

  10. Concurrent chronic or acute illness or other condition that might confound study rating measures

  11. Documented allergy or intolerance to methylphenidate

  12. Use of an investigational drug within 30 days of study entry

  13. Use of concomitant medication that could interfere with assessment of efficacy and safety of study treatments

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of Metadate CD, Concerta and placebo

Dose: Children treated with low doses (20 mg/d) of methylphenidate were randomly assigned to receive Metadate CD 20, Concerta 18 or placebo; those treated with medium doses (20 to 40 mg/d) were randomly assigned to receive Metadate CD 40, Concerta 36 or placebo; children treated with high doses (40 mg/d) were randomly assigned to receive Metadate CD 60, Concerta 54 or placebo

Administration schedule: once daily in the morning

Duration of each medication condition: 7 days

Washout before study initiation: no

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: Regarding the medicine, not stated. 157 received all 3 levels of treatment and participated in all 7 classroom sessions

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale (SKAMP): tested on day 7 in the laboratory school by 2 trained observers. Time points for the test (after ingestion of medication/placebo): h = 0, h = 1.5, h = 3.0, h = 4.5, h = 6, h = 7, h = 12.0

  2. Swanson, Nolan and Pelham (SNAP): parent‐rated, administered twice during each treatment week on days 3 and 6

Non‐serious adverse events

  1. Adverse events reported by participant or parent (guardian). Reported adverse events were characterised (by the investigator at each site) as mild, moderate or severe

  2. Barkley Side Effects Rating Scale: rated symptoms during past week and completed weekly by parent (guardian) on day 6

  3. Heart rate: measured before doses were taken and at the following time points after ingestion: h = 1.5, h = 3.0, h = 4.5, h = 6, h = 7, h = 12.0

  4. Blood pressure: measured before doses were taken and at the following time points after ingestion: h = 1.5, h = 3.0, h = 4.5, h = 6, h = 7, h = 12.0

Notes

Sample calculation: yes

Ethics approval: yes

Comments from study authors

  1. In ANOVA of measures of blood pressure and heart rate, only 2 statistically significant differences related to treatment emerged: systolic blood pressure at hour 7.5, and pulse rate at hour 1.5

  2. Site differences in this study deserve some comment because this is a common finding in multi‐site studies. Site difference was most prominent for subjective outcome measures on the SKAMP Rating Scale, which depends on the training of observers (difficult to equate across sites) and the context of the classroom (controlled but still may vary across sites because of class size, physical space and other factors that may not be standardised)

  3. Regarding analysis of sex (Sonuga‐Barke 2007): Despite the relatively large number of females in the sample, power was insufficient to include dose level as a factor in the analysis

  4. Given the manner in which sex effects appeared to vary across the day, present results may be consistent with pharmacokinetic or pharmacodynamic explanations, or a combination of the 2. Early and late sex‐related differences in clinical effect may be independent of one another or linked. Several possible explanations seem worth testing. The superior methylphenidate response shown by females in the early part of the day may result from greater sensitivity to methylphenidate or from higher methylphenidate plasma concentrations due to increased rates or efficiency of absorption of immediate‐release components. The steeper decline in methylphenidate response shown by females may be a consequence of earlier but normative clearance of methylphenidate following more rapid absorption, or may be indicative of more rapid clearance in females than in males

Limitations

  1. Laboratory setting lacks many features of the natural environment of the home and school. Thus, it is not certain whether the patterns reported here would be observed in school settings in which an ADHD student would be in a classroom in which most students not affected by this disorder

  2. Study was designed to contrast total absorbed daily doses that were approximately equal, although this resulted in differences in initial bolus doses of the 2 active treatments (Metadate CD and Concerta). In this study, doses were not evaluated that were equal to the initial bolus doses of immediate‐release methylphenidate, which would provide another test of the PK/PD model

  3. Effects of both Metadate CD and Concerta in the low‐dose subgroup were smaller than in the high‐dose subgroup, but we do not know whether a higher dose in the low‐dose subgroup would have increased the ES. Lack of tailoring to achieve rigorous experimental control may be another limitation of this study

  4. This study included only patients who already were being treated successfully with methylphenidate. This means that severe and marked adverse events are unlikely to be seen in this study, and it is possible that lack of effect on side effects rating scale factors other than sleep/appetite may occur more readily in medication‐naive patients. Furthermore, it is important to recognise that this was a secondary analysis of a study powered to show non‐equivalence between the 2 methylphenidate formulations in terms of efficacy, not in terms of adverse events

  5. Sonuga‐Barke 2009: This study included only patients who already were being treated successfully with methylphenidate. This means that severe and marked adverse events are unlikely to be seen in this study

  6. Trial was also underpowered for detecting rare events that could be severe. Measures of adverse events were derived only from parent ratings, not from direct observations of behaviour

  7. Significant side effects not measured by the Barkley Side Effects Rating Scale may have occurred

Key conclusions of study authors

  1. Once‐daily doses of Metadate CD and Concerta produced statistically significantly different PD effects on surrogate measures of behaviour and performance among children with attention deficit hyperactivity disorder in the laboratory school setting

  2. As predicted by the PK/PD model, superiority at any point in time was achieved by the formulation with the highest expected plasma methylphenidate concentration

Regarding sex differences

  1. Dose titration of once‐daily formulations of methylphenidate ideally should be based on systematic evidence of response at different periods across the day

  2. Responses of female patients may require additional assessments later in the day to determine optimal dose

Comment from review authors

  1. Compared ratings on sex from SKAMP. However only compared the 2 medication conditions vs each other, not medication conditions vs placebo; therefore we cannot use these analyses

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation:yes

Any withdrawals due to adverse events: Three study participants discontinued because of adverse events that were judged to be unrelated to medications (gastroenteritis: Concerta (n = 1); fever: placebo (n = 1); sunburn: placebo (n = 1))

Email correspondence with study authors: June 2014. We emailed study authors to obtain supplemental data. Unfortunately, data in the correct format are no longer accessible

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

Randomised

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, double‐dummy. Treatments were packaged according to a double‐dummy design. Each treatment pack contained a 1‐week supply of study treatment, with each day’s supply consisting of 1 large capsule to accommodate the size of any dose level of Concerta (containing Concerta or placebo) and, depending on dose level, between 1 and 3 smaller Metadate CD‐sized capsules (containing Metadate CD or placebo)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data from participants finishing the whole study (n = 157) and from the ITT population (n = 184)

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

High risk

Study was funded by Celltech Pharmaceuticals Incorporated

Conflicts of interest: Some study authors are consultants for pharmaceutical companies

Symons 2007

Methods

Prospective, randomised, double‐blind, placebo‐controlled, cross‐over, single‐case designs were used to evaluate methylphenidate administration for 3 school‐aged children with cerebral palsy and comorbid ADHD symptoms

  1. Low‐dose methylphenidate

  2. High‐dose methylphenidate

  3. Placebo

Phases: 4 (including baseline)

Participants

Number of participants screened: not stated

Number of participants included: 2 (1 boy, 1 girl)

Number of participants followed up: 2

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV (combined (100%), hyperactive‐impulsive (0%), inattentive (0%))

Age: mean 10.4 years (range 9 to 11)

IQ: > 70. Participant 2 = 92 non‐verbal and 96 performance non‐verbal (range not stated)

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: cerebral palsy (100%), mild cognitive impairment (100%), autism spectrum disorder (50%), physical impairment (50%), learning disability (50%), other health impairment (50%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD

  2. Cerebral palsy

  3. Attending public elementary schools

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of methylphenidate (low or high dose) and placebo

Mean methylphenidate dosage: 2 doses of methylphenidate administered: low (0.3 mg/kg) and high (0.5 mg/kg)

Administration schedule: once daily, mornings, before the child’s school arrival

Duration of each medication condition: Order of drug/placebo administration was randomly assigned, and administration was provided for 5 consecutive school days in each condition. Drug treatment was not administered on weekends

For participant 2, the order of administration was as follows: baseline, low dose, placebo, high dose

For participant 3, the order of administration was as follows: baseline, placebo, low dose, high dose

Washout before study initiation: not stated

Medication‐free period between interventions: weekends

Titration period: not stated

Treatment compliance: not stated

Outcomes

General behaviour

  1. Direct observation protocol based on a 10‐second partial interval (1/0) for stereotyped and disruptive behaviours and time sampling for task‐related behaviour was used to code data collected directly from each child’s classroom for two 30‐minute sessions on 2 separate days for each week of the trial. Student and teaching staff behaviours were recorded using handheld digital cameras. An observer designated as the primary observer coded all videotaped sessions for that target student. A secondary observer independently coded 20% of randomly selected videotaped sessions for each student

Non‐serious adverse events

  1. No parent or teacher reports described significant side effects associated with either dose level (e.g. changes in sleep, eating, etc.)

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusions of study authors

  1. Low‐dose but not high‐dose methylphenidate administration resulted in clinically significant reductions in directly observed stereotyped and disruptive behaviours for 3 elementary school–aged children with cerebral palsy

  2. For 2 of the children, stereotyped behaviour was exacerbated during high‐dose administration

  3. Finally, no change in attending, as measured by direct observation of task‐related behaviour, was noted

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: May 2014. Emailed study author to request additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Order of drug/placebo administration was randomly assigned

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Parents, school personnel (i.e. teachers, teacher assistants) and research assistants responsible for observational data collection and coding were kept blind to drug/placebo conditions

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

Unclear risk

Not stated

Conflicts of interest: This work was supported, in part, by a McKnight Land‐Grant Professorship to Frank Symons

Szobot 2004

Methods

Four‐day, double‐blind, placebo‐controlled, randomised, fixed dose‐escalating of methylphenidate, parallel‐group trial

Participants

Number of participants screened: not stated

Number of participants included: 36 (all boys)

Number of participants randomly assigned: methylphenidate 19, placebo 17

Number of participants followed up: methylphenidate 19, placebo 17

Number of withdrawals: none

Diagnosis of ADHD: DSM‐IV, combined (methylphenidate 84.6%, placebo 76.5%)

Age: mean 11.6 years

IQ: mean 94.7

Methylphenidate naive: no psychiatric medication for the past 6 months

Ethnicity: European‐Brazilian (89%)

Country: Brazil

Setting: out‐patient clinic

Comorbidity: Conduct disorder and oppositional defiant disorder (methylphenidate 58.8%, placebo 68.4%), depressive disorder (methylphenidate 5.2%, placebo 5.9%), multiple anxiety disorder (methylphenidate 5.2%, placebo 0%), tic disorder (methylphenidate 0%, placebo 5.9%)

Comedication: not stated

Sociodemographics: low‐ to middle‐income families

Inclusion criteria

  1. Diagnosis of ADHD according to DSM‐IV criteria (American Psychiatric Association, 1994)

  2. Between 8 and 17 years old

  3. Male sex

Exclusion criteria

  1. Presence of any neurological or significant clinical disease

  2. Presence of bipolar disorder or any substance abuse/dependence disorder

  3. Use of any psychiatric medication in the previous 6 months

  4. Estimated IQ < 70

Interventions

Participants were randomly assigned to methylphenidate or placebo

Mean methylphenidate dosage: 0.72 mg/kg/d

Administration schedule: twice a day: morning and lunchtime

Duration of intervention: 4 days

Titration period: first day 0.35 mg/kg

Treatment compliance: good

Outcomes

ADHD symptoms

  1. Conners' Abbreviated Rating Scale (ABRS)

Quality of life

  1. Children’s Global Assessment Scale (CGAS). CGAS was scored by a child psychiatrist

Non‐serious adverse events

  1. A research assistant called each family to check side effects. No participants had to interrupt the protocol because of side effects

Notes

Sample calculation: no

Ethics approval: approved by the Ethical Committee of the HCPA (approved as an IRB by the Office for Human Research Protections, USA)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Our results extend the efficacy of methylphenidate for ADHD core symptoms as extensively demonstrated in clinical trials with samples from developed countries to samples from developing countries, for whom a diverse culture may modulate clinical presentation of the disorder

  2. This study’s rate of robust response with methylphenidate might reflect the short duration (4 days) of the clinical trial

  3. Results may not generalise to other different sociocultural groups or to patients from the community

  4. Side effects were not objectively registered

Key conclusion of study authors

  1. Methylphenidate group had a significantly greater decrease in ABRS scores and a significantly greater increase in CGAS scores when compared with the placebo group (P value < 0.01)

Email correspondence with study authors: October 2013. We received from the study authors supplemental information regarding blinding and allocation concealment

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned on the basis of a computer‐derived algorithm (EPIINFO6)

Allocation concealment (selection bias)

Low risk

Only the doctor performing the randomisation knew the allocation, and he had nothing to do with data collection

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind: Participants, parents and all research team members who had contact with participants. Both methylphenidate and placebo pills were manufactured by a single pharmaceutical company; they had the same format and colour and were given to participants in 4 different blisters

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All research team members who had contact with participants

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo‐responders): no

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

High risk

This work was supported by research funds from Hospital de Clínicas de Porto Alegre, FAPERGS and NOVARTIS

Szobot 2008

Methods

Cross‐over trial with 2 interventions

  1. SODAS methylphenidate

  2. Placebo

Phases: 2‐ or 3‐week phases, with each group receiving placebo or SODAS methylphenidate

Participants

Number of participants screened: 25 from a previous ADHD/substance misuse study and 15 through advertising

Number of participants included: 16

Number of participants followed up: 16

Number of withdrawals: 2 (both from group A; withdrawal rate 12.5%)

Diagnosis of ADHD: DSM‐IV (combined 12 (75%), inattentive 3 (18.75%), hyperactive/impulsive (n = 1))

Mean age: group A 17.5 years (SD 2.33), group B 17.38 (SD 2.2)

IQ: group A 79.43 (SD 16.66), group B 84.75 (SD 21.16)

Sex: all boys (100%)

Methylphenidate naive: 100%

Ethnicity: European‐Brazilian (group A 3 (37.5%), group B 7 (87.5%))

Country: Brazil

Setting: out‐patient clinic

Comorbidity: conduct disorder (group A 100%, group B 75%); oppositional defiant disorder (group A 25%, group B 37.5%); depression (group A 12.5%, group B 25%)

Comedication: yes (marijuana and cocaine); group A: marijuana (100%) and cocaine (50%); group B: marijuana (87.5%) and cocaine (37.5%)

Sociodemographics: divorced parents (group A 37.5%, group B 50%); socioeconomic group A + B + C (group A 50%, group B 87.5%); group D + E (group A 50%, group B 12.5%)

Inclusion criteria

  1. 15 to 21 years of age

  2. Male

  3. Current diagnosis of abuse of marijuana or cocaine (K‐SADS‐E and MINI)

  4. Current diagnosis of ADHD: Kiddie Schedule for Affective Disorders and Schizophrenia for School‐Age Children, Epidemiological Version (K‐SADS‐E)

  5. Stimulant naive

Exclusion criteria

  1. Absence of responsible adult ‐ to inform on childhood psychopathology and to take responsibility for medication and/or the need for in‐patient care for substance misuse or psychiatric comorbidity

  2. Primary psychiatric condition requiring immediate treatment (e.g. moderate/severe depression)

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean SODAS methylphenidate dosage: 0.3, 0.7 and 1.2 mg/kg/d for weeks 1, 2 and 3 for group A, and for weeks 4, 5 and 6 for group B

Administration schedule: morning dose time points

Duration of each medication condition: 3 weeks

Washout before study initiation: no

Medication‐free period between interventions: 24 hours

Titration period: none

Treatment compliance: "Study compliance was assessed by self‐report, mother’s report and pill counting"

Outcomes

ADHD symptoms

  1. Kiddie Schedule for Affective Disorders and Schizophrenia for School‐Age Children, Epidemiological Version (K‐SADS‐E), for diagnosis

  2. Swanson, Nolan and Pelham scale, Fourth Edition (SNAP‐IV)

General behaviour

  1. Clinical Global Impression (CGI) Scale

Serious adverse events

  1. Barkley Side Effect Rating Scale (SERS)

Non‐serious adverse events

  1. Barkley Side Effect Rating Scale (SERS)

Notes

Sample calculation: not stated

Ethics approval:"The project was approved by the Institutional Review Board (IRB) of Hospital de Clinicas de Porto Alegre (approved as an IRB by the Office for Human Research Protections, United States of America, IRB 00000921"

Comments from study authors

  1. In the present study, SODAS methylphenidate was significantly superior to placebo in reducing ADHD symptoms and improving global functioning for all main outcome measures (SNAP‐IV and CGI scores)

  2. No treatment effect on illicit substance use disorders was noted, and methylphenidate‐SODAS was well tolerated, despite causing greater appetite reduction than was seen with placebo

Key conclusions of study authors

  1. SODAS methylphenidate was more effective than placebo in reducing ADHD symptoms in a non‐abstinent out‐patient sample of adolescents with comorbid substance use disorders

  2. Randomised clinical trials, with larger samples and substance use disorder interventions, are recommended

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: One withdrawal resulted from a participant feeling "worse", "more restless"

Email correspondence with study authors: May 2014. We received supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

"One of the investigators (LAR) randomized the 16 subjects into groups A or B and prepared weekly blisters of medications for each participant"

Allocation concealment (selection bias)

High risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"A pharmacist packaged mPH‐SODAS and matching placebo in capsules so that the MPH‐SODAS and placebo could not be visually differentiated"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

From author correspondence: All persons who evaluated outcome measures were blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): unclear

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

High risk

"The ADHD outpatient program receives research support from Bristol‐Myers Squibb, Eli‐Lilly, Janssen‐Cilag and Novartis"

Conflicts of interest: Study authors are consultants and speakers for various companies

Tannock 1989

Methods

Within‐participant, cross‐over trial with 3 interventions

  1. Methylphenidate 0.3 mg/kg

  2. Methylphenidate 1 mg/kg

  3. Placebo

Two different drug conditions each day

Participants

Number of participants screened: 16

Number of participants included: 19

Number of participants followed up: 12 (10 boys, 2 girls)

Number of withdrawals: none

Diagnosis of ADHD: DSM‐III ADD‐H (equivalent to 'combined' in later classifications)

Age: mean 8.4 years (range 6 to 11)

IQ: mean 105

Methylphenidate naive: Five received methylphenidate previously. Three were taking methylphenidate at the time of referral and had a 48‐hour pre‐trial washout

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (n = 4), learning difficulties (n = 8; according to school record and defined as < 25th percentile on 1 or more subtests within the Wide Range Achievement Test (WRAT‐R))

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Thought by referring physician to have ADHD

  2. Confirmed through assessment by 2 child psychiatrists using the Parent Interview Child Symptoms Questionnaire

  3. Confirmed by teacher information on Conners' Teacher Questionnaire, Rutter B Questionnaire and the Swanson, Nolan and Pelham Questionnaire

Exclusion criteria

  1. Full‐scale WISC‐R < 80

  2. Exclusive diagnosis of emotional or conduct disorder; major neurological, physical or sensory impairment; and/or any contraindication for use of methylphenidate (e.g. tics, seizures, heart disease)

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of 0.3 mg/kg or 0.6 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: not stated.

Administration schedule: Interval of 4 hours separated morning and afternoon doses

Duration of each medication condition: not stated

Washout before study initiation: 4 hours

Titration period: none

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Treatment‐emergent side effects (e.g. stomach distress, pallor, mood swings, tics)

  2. Pulse and blood pressure readings taken in sitting position immediately before medication and again 1 hour after administration of dose

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated

Key conclusions of study authors

  1. Behavioural and academic improvements produced by a dose of 0.3 mg/kg in the morning were no longer evident in the afternoon

  2. 1 mg/kg methylphenidate produced behavioural improvements that were clinically and statistically discernible in the afternoon, although academic improvements had dissipated

  3. Carryover effects of methylphenidate into the afternoon were discernible with 1.0 mg/kg but not with 0.3 mg/kg

Comment from review authors

  1. Outcomes for ADHD symptoms were not appropriate for this study, so only adverse effects were examined

Email correspondence with study authors: sent email to study authors to ask for additional information but have not received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The order of medication condition was randomized with the restrictions that each child receive two different medication conditions each day (e.g. high, low) occur with equal frequency in the morning and afternoon" "The order of these six combinations was randomized for each child"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"The methylphenidate and placebo were packaged in coloured gelatin capsules by the hospital pharmacist to avoid detection of dose and taste, packaged in individual envelopes and dispensed by project staff 1 hour before testing"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

Jointly funded by Ontario Mental Health Foundation (Grant No. 963‐86/88) and Health and Welfare Canada (Grant No. 6606‐3166‐42)

Conflict of interest: Study authors' affiliations were not declared

Tannock 1992

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate low dose

  2. Methylphenidate high dose

  3. Placebo

Phases: 3 separate drug conditions: placebo, low‐dose (0.3 mg/kg) and high‐dose (1.0 mg/kg) medication, with 2 test sessions each. Drug conditions changed on a daily basis: 6 test sessions plus baseline practice session

Participants

Number of participants included: 26

Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 23

Number of withdrawals: 3

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 9.2 years (range not stated)

IQ: mean 105.9 (SD 10.5)

Sex: 24 boys, 2 girls

Methylphenidate naive (not clear; "9 children receiving stimulant medication prior to the present study")

Ethnicity: not stated

Country: Canada

Setting: hospital/out‐patient department

Comorbidity: 12

Comedication: not clear

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosis of ADHD confirmed

  2. Child had to be scheduled to receive a trial with methylphenidate independent of the present investigation

Exclusion criteria

  1. Wechsler Intelligence Scale for Children ‐ Revised: < 80

  2. Exclusive DSM‐III diagnosis of Conduct disorder or an Emotional disorder

  3. Major neurological, physical or sensory impairment

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: N/A (modal dose at 0.3 mg/kg = 7.5 mg (range 5.0 mg to 15 mg), modal dose at 1.0 mg/kg = 27.5 mg (range 17.5 mg to 47.5 mg))

Administration schedule: daily single dose

Duration of each medication condition: 1 day

Washout before study initiation: 48 hours before study initiation

Medication‐free periods between interventions: no

Titration period: none

Treatment compliance: capsules administered by project staff

Outcomes

General behaviour

  1. Wisconsin Card Sorting Test: observer, daily, 90 minutes after treatment

Non‐serious adverse events

  1. Decreased cognitive flexibility (repetitive actions with obsessive quality)

  2. Movement stereotypes

  3. Facial motor tics

  4. Topic perseveration

  5. Excessive pre‐occupation with the task at hand and persistent talkativeness: observer, during test session

  6. Treatment‐emergent side effects conducted routinely during medication trial in our laboratory

  7. Monitored throughout sessions

Three children exhibited unusual side effects leading to trial termination. One exhibited hypersensitivity (skin rash, urticaria and throat clearing), and 2 became somewhat disoriented and confused and complained of odd sensations in their limbs

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusion of study authors

  1. Results indicate that methylphenidate increased perseverative errors on the first assessment but decreased them on the second; clinical symptoms of perseveration occurred at both assessments. Findings suggest that methylphenidate may reduce cognitive flexibility temporarily in some children with ADHD

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Any withdrawals due to adverse events: yes; 3

Email correspondence with study authors: April 2014. We emailed study authors twice to ask for supplemental information/data but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not clear

Allocation concealment (selection bias)

Low risk

Double‐blind, placebo‐controlled design. Drug order for the first 3 test sessions was randomly assigned, and each child retained the same order for the remaining 3 sessions

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled design. Medication packaged in coloured gelatin capsules to avoid detection of dose and taste. Each child followed the same routine for every test session

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not clear

Incomplete outcome data (attrition bias)
All outcomes

High risk

Incomplete outcome data for 3 respondents with unusual side effects

Selective reporting (reporting bias)

Unclear risk

No information presented on numbers of participants with side effects or outcomes for data on nausea, blood pressure, etc.

Vested interest bias

Low risk

Research supported in part by a grant from the Canadian Psychiatric Research Foundation and a Post‐Doctoral Fellowship by the Ontario Mental Health Foundation

Conflicts of interest: not declared

Tannock 1993

Methods

Cross‐over trial with 2/3 interventions

  1. Methylphenidate low dose

  2. Methylphenidate high dose

  3. Placebo

Phases

  1. Each child in ADHD group participated in a 6‐day medication trial, consisting of six 3‐hour test sessions

  2. Each child was tested on 2 separate occasions when given placebo, low‐dose methylphenidate (0.3 mg/kg) and high‐dose methylphenidate (1.0 mg/kg) in a randomly assigned counterbalanced sequence

Participants

Number of participants screened: not stated

Number of participants included: methylphenidate 22, control 16. Participants were randomly assigned to 1 of 6 possible drug condition orders

Number of participants followed up: 22 (21 boys, 1 girl)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 9.4 years (range not stated)

IQ: mean 105.5 (SD 11.3)

Methylphenidate naive: not clear: "Thus all these children would have received a trial with psychostimulants independently of this study"; "8 children had been receiving stimulant medication prior to this study")

Ethnicity: not stated

Country: Canada

Setting: out‐patient clinic

Comorbidity: 55%; oppositional disorder (n = 5), conduct disorder (n = 3), emotional disorder as well as oppositional or conduct disorder (n = 4), major depression (n = 2), avoidant disorder (n = 1), separation anxiety disorder (n = 1) and learning disorder (n = 6)

Comedication: not clear

Sociodemographics: not stated

Inclusion criteria

  1. Confirmed diagnosis of ADHD: Child demonstrated ≥ 3 symptoms of inattentiveness, 3 of impulsiveness and 2 of hyperactivity, with a history of these symptoms before 6 years of age, based on diagnostic interview

  2. Diagnosis was made if the child received a Rutter B total score ≥ 9 and fulfilled any 2 of the following criteria: score ≥ 15 on the Conners' Abbreviated Teacher Questionnaire; ≥ 4 inattentive, 4 impulsive and 3 hyperactivity symptoms on the Swanson, Nolan and Pelham Scale; or score of 5 or 6 on the Rutter‐B hyperactivity factor

Exclusion criteria

  1. Wechsler Intelligence Scale for Children ‐ Revised: score < 80 with psychosis, or with any major neurological, physical or sensory impairment

Interventions

Participants were randomly assigned to 1 of 6 possible drug condition orders of (dose not stated) methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: 6 separate medication administrations: 2 placebo, 2 low dose (0.3 mg/kg) and 2 high dose (1.0 mg/kg). First 3 test sessions were ordered randomly, than were ordered repeatedly for last 3 sessions: daily for 6 days

Duration of each medication condition: 3 hours

Washout before study initiation: 48 hours

Medication‐free period between interventions: not clear

Titration period: none

Treatment compliance: "Medication was administered by project staff"

Outcomes

Non‐serious adverse events

  1. Blood pressure and pulse readings: before and 1 hour after ingestion

  2. Reduced social responsivity: during each test session, observer

  3. Intense concentration: during each test session, observer

  4. Stereotypy: during each test session, observer

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusions of study authors

  1. Results indicate that only high‐dose treatment had a specific effect on focused attention, and this effect was delayed relative to the more salient but non‐specific effects on overall efficiency of information processing

  2. Task performance at high dose was related to concurrent clinical manifestations of intense concentration, but no evidence suggested that methylphenidate produced overfocusing

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated
Email correspondence with study authors: April 2014. Emailed study authors twice for supplemental information/data but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Double‐blind, placebo‐controlled, within‐participant design was used ...randomly assigned, counterbalanced sequence. Medications were prepared individually for each child and were packaged in coloured gelatin capsules to avoid detection of dose and taste

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled, within‐participant design was used ...randomly assigned, counterbalanced sequence. Medications were prepared individually for each child and were packaged in coloured gelatin capsules to avoid detection of dose and taste

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No withdrawals were noted

Selection bias (e.g. titration before randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

All outcomes reported

Vested interest bias

Low risk

Study funded by the Canadian Psychiatric Research Foundation and the Medical Research Council of Canada

Conflicts of interest: not declared

Tannock 1995a

Methods

Four‐day, randomised, double‐blind, placebo‐controlled, cross‐over trial with

  1. Three doses of methylphenidate (0.3 mg/kg, 0.6 mg/kg and 0.9 mg/kg)

  2. Placebo

With initial 1‐day open‐label trial

Participants

Number of participants screened: 28

Number of participants included: 28 (25 boys, 3 girls). Participants were randomly assigned to 3 doses of methylphenidate and placebo

Number of participants followed up: 28

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 8.9 years (range 7 to 11)

IQ: mean 106.5 (15.6)

Methylphenidate naive: 80%

Ethnicity: Caucasian (90%), other (10%)

Country: Canada

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder or conduct disorder (35%), learning disabilities (39%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. ADHD, DSM‐III‐R

Exclusion criteria     

  1. IQ: Wechsler Intelligence Scale for Children ‐ Revised: score < 80

  2. Anxiety disorder (DSM‐III‐R)

  3. Major neurological, physical or sensory impairment

Interventions

Participants were randomly assigned to 3 doses of methylphenidate (0.3 mg/kg, 0.6 mg/kg, 0.9 mg/kg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: once daily

Duration of each medication condition: 1 day

Washout before study initiation: For children receiving methylphenidate before trial, washout period ≥ 48 hours before trial, with no washout between periods

Titration period: All children participated in an initial 1‐day open trial with a 0.3 mg/kg dose of methylphenidate to ascertain tolerance, before proceeding with the double‐blind, placebo‐controlled trial

Treatment compliance: not stated

Outcomes

Non‐serious adverse events

  1. Cardiovascular function (heart rate). Radial pulse, taken for 1 minute. with the child seated, was measured 3 times during each session: immediately before medication (time 0) and again at 1 hour (time 1) and 2 hours (time 2) following administration of the oral dose. From time 0 to time 1, children were seated at a table, colouring or playing quietly with puzzles or board games: Children completed the cognitive task from time 1 to time 2

Notes

Sample calculation: yes

Ethics approval: yes; Institutional Research Ethics Board

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; 1‐day open‐label trial to ascertain tolerance towards medication, but all participants were analysed

Key conclusions of study authors

  1. Results indicate that methylphenidate enhanced cognitive flexibility, although the high dose was less effective than lower doses in enhancing response inhibition

  2. Dissociations of dose effects on cognitive function and behaviour were demonstrated: Dose‐response functions for changes in behaviour were linear, whereas the function for response inhibition was U‐shaped

Email correspondence with study authors: August 2013. We received from the first study authors supplemental information regarding drop‐outs, screening and ethics approval

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Low risk

All medication was prepared by the hospital pharmacy and was packaged in opaque gelatin capsules to avoid detection of dose and taste

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Children, parents, teachers and research staff were all blinded to medication conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Children, parents, teachers and research staff were all blinded to medication conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data from all 28 participants were analysed

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

Low risk

Medical Research Council of Canada and Health and Welfare Canada

Conflicts of interest: nothing to declare

Tannock 1995b

Methods

Cross‐over trial with 1 interventions

  1. Methylphenidate (0.3 mg/kg, 0.6 mg/kg, 0.9 mg/kg)

  2. Placebo

Phases: washout (if applicable), baseline, trial
Two groups ‐ ADHD and ADHD + anxiety ‐ were subjected to the same trial

Participants

Number of participants screened: 50

Number of participants included: 40 (34 boys, 6 girls). Participants were randomly assigned to 1 of 12 possible drug condition orders

Number of participants followed up: 40

Number of withdrawals: none

Diagnosis of ADHD: DSM‐III‐R (no subtype)

Age: mean not stated (range 7 to 11 years); ADHD mean 9.4; ADHD + anxiety mean 9.1

IQ: > 80

Methylphenidate naive: not stated

Ethnicity: Caucasian (90%), African American or Asian (10%)

Country: Canada

Setting: out‐patient clinic

Comorbidity (type: overanxious 27.5%, separation anxiety 5%, overanxiety and separation anxiety 10%, avoidant disorder with overanxious traits 2.5%, ODD 55%, conduct disorder 10%)

Comedication: not stated

Sociodemographics: predominantly from middle‐class families

Inclusion criteria

  1. Meeting DSM‐III‐R criteria

  2. At least 2 of the following

    1. ≥ 15 on the Conners' Abbreviated Symptom Questionnaire

    2. ≥ 4 inattentive, 4 impulsive and 3 hyperactive symptoms rated as "pretty much" or "very much" on Swanson, Nolan and Pelham Questionnaire

    3. Score of 5 or 6 on the Hyperactivity factor of the Rutter Child Scales

Exclusion criteria

  1. Full scale IQ < 80

  2. Evidence of major neurological, physical or sensory impairment

  3. Medical or neurological contraindications for stimulant medication

  4. Not knowing the number facts of 10. Needing to use concrete materials (i.e. fingers) to add numbers

Inclusion criteria for the ADHD + anxiety group

One or more of the following

  1. DSM‐III‐R criteria for overanxious, separation anxiety or avoidant disorder based on parent interview

  2. Score > 1 SD above the mean for age and sex on the Revised Children’s Manifest Anxiety Scale

  3. Score > 1 SD for age and sex on the Trait Scale‐C2 of the State‐Trait Anxiety Inventory for Children

Children in the ADHD group did not meet any of these criteria for anxiety

Interventions

Participants were randomly assigned to 1 of 12 possible drug condition orders of 0.3 mg/kg, 0.6 mg/kg or 0.9 mg/kg methylphenidate and placebo

Mean methylphenidate dosage: 0.3 mg/kg = 8.75 (± 2.2); 0.6 mg/kg = 17.75 (± 4.2); 0.9 mg/kg = 27.25 (± 5.6)

Administration schedule: once per day

Duration of each medication condition: 1 day

Washout before study initiation: 48 hours when applicable

Medication‐free period between interventions: not stated

Titration period: 1‐day open trial on 0.3 mg/kg before proceeding to ascertain tolerance. Not stated whether initiated before/after randomisation

Treatment compliance: Medication was administered at the laboratory

Outcomes

Non‐serious adverse events

  1. Heart rate and radial pulse, once at baseline and 3 times during each session

Notes

Sample calculation: not stated

Ethics approval: not stated

Key conclusion of study authors

  1. High levels of trait anxiety in children with ADHD predict poor (but not adverse) response to methylphenidate in terms of working memory, and add to growing evidence that ADHD with anxiety constitutes a distinct and clinically meaningful subtype of ADHD

Comment from review authors

  1. Main trial was preceded by a 1‐day trial of the lowest dose to assess tolerance to methylphenidate

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; see above

Any withdrawals due to adverse events: no

Email correspondence with study authors: June 2014. Emailed study authors twice to request supplemental information/data but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Drug order was counterbalanced and determined by random assignment, such that an approximately equal numbers of children were assigned to each of 12 possible orders

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Active medication and placebo were prepared by the hospital pharmacy, packaged in identical opaque gelatin capsules and administered in a double‐blind manner

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): 1‐day open trial to assess tolerance before main trial, but no children were excluded

Selective reporting (reporting bias)

Low risk

All outcomes are reported

Vested interest bias

Low risk

Research was supported in part by the Ontario Mental Health Foundation and the National Health Research and Development Program, Health Canada

Taylor 1987

Methods

Double‐blind, randomised, placebo‐controlled, cross‐over trial with 2 arms

  1. Methylphenidate

  2. Placebo

Each treatment period lasted for 3 weeks with a washout period of 1 week planned between treatments and up‐titration to optimum dosage during the 3 weeks

Participants

Number of participants screened: 64

Number of participants included: 39. Of these, 26 had an ADHD diagnosis according to DSM‐III. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 38

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐III (n = 26; type not stated)

The following data reflect the whole trial (n = 38)

Age: mean 8.6 years (range 6 to 10)

IQ: all > 65; mean 93.4

Sex: all boys

Methylphenidate naive: 100%

Ethnicity: not stated

Country: England

Setting: out‐patient clinic

Comorbidity: no attentive or restless behaviour, but antisocial, disruptive or aggressive in conduct (n = 6), hyperactive but not antisocial or aggressive (n = 9), with both hyperactive and disruptive behaviour (n = 23), conduct disorder (n = 7), relationship problems (n = 2) and disturbance of emotions specific to childhood (n = 3)

Comedication: not stated

Sociodemographics: 40% from broken homes

Inclusion criteria

  1. IQ > 65

  2. Free of autistic features

  3. Lives in a family home, not an institution

  4. Attending primary school

  5. Problems assessed at the clinic as severe enough to warrant psychiatric treatment

  6. Free of contraindications to stimulant medication

  7. No treatment with stimulant drugs

  8. No psychotropic drugs given for ≥ 6 months previously

Exclusion criteria

  1. Children with severe intellectual retardation or neurological disease

  2. Autistic and psychotic children

  3. Pre‐school children

  4. Adolescents

  5. Female

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Mean methylphenidate dosage: 9 received doses from 0.2 mg/kg to 0.5 mg/kg, 21 from 0.5 mg/kg to 0.9 mg/kg and 8 from 0.9 mg/kg to 1.4 mg/kg

Administration schedule: time points not stated

Duration of each medication condition: 3 weeks

Washout before study initiation: All were stimulant‐naive

Medication‐free period between interventions: 1 week

Titration period: A flexible dosage regimen was used after randomisation. Each child began with 5 mg daily, with dosage adjustments made at 2‐ to 3‐day intervals. The optimum dosage was assessed for each child, in the light of clinical response and the occurrence of side effects, to a maximum of 30 mg daily

Treatment compliance: Compliance with medication was rated as good or very good in 89%

Outcomes

ADHD symptoms

  1. Conners’ Teacher Rating Scale: rated at the end of each treatment period, that is, day 21

  2. Parental Account of Childhood symptoms (PACS), hyperactivity scale: rated at the end of each treatment period, that is, day 21

Serious adverse events

  1. No serious physical effects of medication were encountered

Non‐serious adverse events

  1. Unwanted effects of medication were assessed by using the physician’s ratings of 26 possible symptoms with full physical examination, rated at the end of each treatment period, that is, day 21

Notes

Sample calculation: no

Ethics approval: not stated

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no; all methylphenidate treatment‐naive

Comment from study authors (limitation)

  1. Long‐term treatment with stimulant drugs could not be assessed in this short‐term trial

Key conclusion of study authors

  1. A good response to methylphenidate was predicted by higher levels of inattentive and restless behaviour, impaired performance on tests of attention, clumsiness, younger age and absence of symptoms of overt emotional disorder. DSM‐III and ICD‐9 diagnoses of "hyperactivity" were not good predictors

Comments from review authors

  1. Long‐term treatment with stimulant drugs could not be assessed in this short‐term trial

  2. Children with tics or cardiovascular disease were excluded

  3. Demographic characteristics and mean dose include the whole sample (n = 38). However, upon receipt of correspondence from the trial author, outcomes measured included the 26 individuals diagnosed with ADHD (DSM‐III)

  4. Invetigators tested both for methylphenidate vs placebo (effects of treatment) and for end of first treatment period vs end of second treatment period (effects of occasion), as well as the interaction between treatment and occasion. "The two order groups were combined together for those measures that had shown no significant effects of test occasion or interaction of occasion with treatment. For measures which did show main or interactive effects of occasion, the two order groups were considered separately"

Email correspondence with trial authors: October 2013. We received from trial authors no supplemental information regarding data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Children were randomly allocated to receive drug first or placebo first. This was carried out by pharmacy staff, who knew only the name and identifying number of each case

Allocation concealment (selection bias)

Low risk

Allocation was carried out by pharmacy staff, who knew only the name and identifying number of each case

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Tablets were dispensed to a trial member, who did not know what they contained and handed them to parents

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Psychiatrist supervising treatment also assessed and recorded side effects, physical findings and parents’ general impressions at the end of each treatment; other assessors of outcomes were blinded to the treatment given but also to possible clues arising from physical effects of the drug. Behavioural measures were carried out independently of one another by investigators blind to results of the other tests and to teacher ratings

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data were missing for 3 children: One child had missing data on the Parental Account of Childhood Symptoms because he had been taken into care, and two children had missing data on the Conners’ Teacher Rating Scale hyperactivity factor because they had been excluded from school. Method of imputation not described

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

High risk

Partially funded by grant from CIBA Ltd., which provided medicine and placebo

Conflicts of interest: Dr. Schachar was supported during this period by a fellowship from the Medical Research Council of Canada&&

Taylor 1993

Methods

Four‐week, double‐blind, cross‐over trial with 3 interventions

  1. Two different doses of methylphenidate

  2. Placebo

Each medication dose/placebo was given for 1 week

Participants

Number of participants screened: 57

Number of participants included: 32 (27 boys, 5 girls) were randomly assigned to possible drug condition orders

Number of participants followed up: 32

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R

Age: mean 10.26 years (range 7.0 to 12.9)

IQ: mean 107 (SD 16.2; range 78 to 139)

Methylphenidate naive: 12 (37.5%)

Ethnicity: no information

Country: Canada

Setting: out‐patient clinic

Comorbidity: no information

Comedication: no information

Sociodemographics: no information

Inclusion criteria

  1. 7 to 12 years of age

  2. Verbal or performance IQ ≥ 85

Exclusion criteria

  1. No information

Interventions

Participants were randomly assigned to possible drug condition orders of, respectively, mean 6.72 mg (5 mg to 10 mg) and mean 11.88 mg (10 mg to 15 mg) methylphenidate and placebo

Administration schedule: morning and noon

Duration of each medication condition: 1 week

Washout before study initiation: none

Titration period: none

Treatment compliance: no information

Outcomes

Non‐serious adverse events

  1. Decreased appetite, trouble falling asleep, stomachache, headache, dysphoria; withdrawn, tearful, anxious, licking lips, picking at skin; facial grimacing, repetitive movements

Notes

Sample calculation: no information

Ethics approval: no information

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Key conclusion of study authors

  1. Only ERPs (event‐related potentials) reflected slowed processing in children with ADHD that normalised with appropriate medication

Email correspondence with study authors: January 2014. No supplemental information was received. We therefore have no more information on numbers of participants with non‐serious adverse events

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised. All medication and placebo testing was conducted under double‐blind conditions with randomly assigned testing order

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Active medication and placebo substances were placed in identical red and white capsules in powder form. Matched on both taste and appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

All medication and placebo testing was conducted under double‐blind conditions with randomly assigned testing order, but how blinding was done was not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Selection bias: This series of 32 children with ADHD does not include an additional group of 12 children with ADHD, who after drug trials were deemed "non‐responders"

Selective reporting (reporting bias)

High risk

Did not report adverse event data

Vested interest bias

Unclear risk

Not stated

Tervo 2002

Methods

Triple‐blind, 3‐way, cross‐over trial with 2 interventions

  1. Methylphenidate at high and low doses

  2. Placebo

Phases

  1. High: 0.3 mg/kg b.i.d.

  2. Low: 0.05 mg/kg b.i.d.

Participants

Number of participants screened: not stated

Number of participants included: 63 (49 boys, 14 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 49

Number of withdrawals: 14

Diagnosis of ADHD: DSM‐IV (combined (70%), hyperactive‐impulsive (16%), inattentive (5%), other (9%))

Age: mean 9 years 10 months (SD 2 years 10 months; range not stated)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: motor dysfunction (35%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible dose levels of immediate‐release methylphenidate and placebo

Mean methylphenidate dosage: not stated

Administration schedule: twice daily; time points not stated

Duration of each medication condition: 6 days

Washout before study initiation: 1 day

Titration period: not stated

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' Parent Rating Scale: parent‐rated at baseline and at end of period

  2. Conners' Teacher Rating Scale: teacher‐rated at baseline and at end of period

General behaviour

  1. Child Behavior Checklist (CBCL): parent‐rated at baseline and at end of period

  2. Home Situations Questionnaire: parent‐rated at baseline and at end of period

  3. Teacher report form (similar to CBCL): teacher‐rated at baseline and at end of period

  4. School Situations Questionnaire: teacher‐rated at baseline and at end of period

Non‐serious adverse events

  1. Side Effects Rating Scale: parent, at baseline and at end of period

Notes

Sample calculation: no

Ethics approval: no

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. A limitation of this study is that neuropsychological functioning (e.g. intellect, ability, memory, visual perceptual functioning) was not measured in all children

  2. All children with MD had substantially impaired motor skills and "soft neurological signs" such as mixed laterality, mirror or overflow movements or choreiform movements

Key conclusions of study authors

  1. Children with ADHD‐MD were more likely to have severe ADHD combined type and other neurodevelopmental and behavioural problems

  2. Both groups of children had a linear dose response to medication (placebo, low, high), and no evidence was found of a group‐by‐dose interaction or an overall group effect at home or at school

Comments from review authors

  1. IQ not reported

  2. Four children withdrew from the study as the result of adverse reactions ‐ all taking high dose

Email correspondence with study authors: April 2013. We received from study authors the full dataset in SPSS

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Low risk

Only the clinical pharmacist knew the sequence of phases

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Capsules of medication and placebo were made and dispensed as described by Barkley (1988). Tablets were crushed and placed within orange opaque gelatin capsules. Capsules disguised the taste of methylphenidate and lactose placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Triple‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

14 of the 63 trials were excluded from the analysis because of inadequately completed outcome measures. Four of the 14 children did not complete the trial because of adverse reactions to medication (e.g. irritability, headache, stomachache). These children received high‐dose medication in the first trial interval

Selective reporting (reporting bias)

Low risk

No protocol was published. All pre‐specified outcomes of interest have been reported

Vested interest bias

Unclear risk

No conflicts of interest have been disclosed

Tirosh 1993a

Methods

16‐day, double‐blind, cross‐over, counterbalanced study with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 20 (16 boys, 4 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 20

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (ADD 30%, ADHD 70%)

Age: mean 9.3 years (range 7 to 12)

IQ: mean 102 (SD 11)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Israel

Setting: out‐patient clinic

Comorbidity: no

Comedication: no

Sociodemographics: middle (12), upper‐middle (5) and low (3) socioeconomic status of parents

Inclusion criteria

  1. DSM‐III diagnosis of ADHD

  2. 7 to 12 years old

Exclusion criteria

  1. Methylphenidate before study

  2. Neurological, sensory or physical health problems

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo

Methylphenidate dose range: 0.3 mg/kg to 0.5 mg/kg

Administration schedule: twice daily

Duration of each medication condition: 8 days

Washout before study initiation: no (participants were methylphenidate naive)

Titration period: no

Treatment compliance: Parents were asked to bring their packages of tablets back for pill count; no data

Outcomes

ADHD symptoms

  1. Abbreviated Parent Rating Scale (APRS), weekly

  2. Teacher Rating Scale (TRS), weekly

Notes

Sample calculation: no

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Funding: no funding

Key conclusions of study authors

  1. Teacher Rating Scale: Placebo‐drug difference correlated more significantly with outcome measures than did the baseline drug difference

  2. Study underlines the validity of a multi‐measure placebo/drug trial in evaluating the efficacy of methylphenidate for children with attention deficit disorder

Email correspondence with study authors: August 2013. Study author stated that data were discarded (Ramstad 2013d [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation with a table of random numbers

Allocation concealment (selection bias)

Low risk

Look‐alike placebo tablets were supplied by the hospital pharmacy and were similarly administered

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 information

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

No information

Tirosh 1993b

Methods

Double‐blind, controlled, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: unknown

Number of participants included: 11 (8 boys, 3 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 10

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: median 9 years 8 months (range 6.9 to 12.3 years)

IQ: median 106 (range 92 to 118)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Israel

Setting: out‐patient clinic

Comorbidity: none

Comedication: none

Sociodemographics: low middle class (n = 2), middle class (n = 8)

Inclusion criteria

  1. Healthy; no neurological deficit

  2. Living with their natural parents

  3. Medication‐naive

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of 0.3 mg/kg to 0.4 mg/kg methylphenidate hydrochloride (total dose 10 mg to 15 mg) and placebo

Mean methylphenidate dosage: 0.3 mg/kg to 0.4 mg/kg

Administration schedule: once daily at 7:30 AM on 6 of the 8 days. During the other 2 days, a second dose was administered at 2:00 PM

Duration of each medication condition: 8 days

Washout before study initiation: not stated

Medication‐free period between interventions: 3 days

Titration period: none

Treatment compliance: As measured by returned package pill counts, this was rated as full compliance for the 10 children remaining in the study

Outcomes

ADHD symptoms

  1. Conners’ Teacher and Parent Rating Scale: before therapy and after each intervention

Non‐serious adverse events

  1. Sleep measurements, 10 minutes before "lights out" until morning awakening for 4 successive nights during each of the 3 respective periods

Notes

Sample calculation: no

Ethics approval: not stated

Key conclusions of study authors

  1. Results support the notion that ADHD is a centrally generated disorder attributable to hypoarousal, which subsequently stimulates motor overactivity

  2. Methylphenidate does not appear to affect sleep patterns adversely and possibly normalises them in individuals with ADHD

Comment from review authors

  1. No useful data

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: December 2013. We were unable to receive supplemental data from study authors because the study is 20 years old (Ramstad 2013d [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

A drug‐placebo sequence was used for children assigned odd numbers in the study and vice versa

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Look‐alike placebo tablets were supplied

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Investigator who analysed the data was unaware of the drug‐placebo sequence

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol available

Vested interest bias

Unclear risk

No information

Tourette's Syndrome Study Group 2002

Methods

Sixteen‐week, multi‐centre, randomised, double‐blind, placebo‐controlled, parallel trial to assess the efficacy of clonidine and methylphenidate for children and adolescents with ADHD and chronic tic disorder

Weeks 1 to 4: clonidine/placebo dose titration; week 5 to 8: addition of methylphenidate/placebo dose titration; week 9 to 16: maintenance therapy

Participants

Number of participants screened: not stated

Number of participants included: 136

Number of participants randomly assigned: methylphenidate 37, placebo 32, clonidine 34, methylphenidate + clonidine 33

Number of participants followed up: methylphenidate 33, placebo 25, clonidine 30, methylphenidate + clonidine 33

Number of withdrawals: methylphenidate 4, placebo 7, clonidine 4, clonidine + methylphenidate 4

Diagnosis of ADHD: DSM‐IV: methylphenidate: combined 32%, hyperactive‐impulsive 3%, inattentive 65%; placebo: combined 19%, hyperactive‐impulsive 0%, inattentive 81%; clonidine: combined 21%, hyperactive‐impulsive 3%, inattentive 76%; clonidine + methylphenidate: combined 33%, hyperactive‐impulsive 3%, inattentive 64%

Mean age: methylphenidate 10.7 years (SD 2.0); placebo 9.7 years (SD 1.8); clonidine 9.7 years (SD 1.8); clonidine + methylphenidate 10.6 years (SD 1.9). Total age range 7 to 14

Sex: methylphenidate 34 boys, 3 girls; placebo 29 boys, 3 girls; clonidine 29 boys, 5 girls; clonidine + methylphenidate 24 boys, 9 girls

Stimulant‐naive: 42%

Ethnicity: methylphenidate 81% Caucasian; placebo 94% Caucasian; clonidine 94% Caucasian; clonidine + methylphenidate 85% Caucasian

Country: USA

Setting: out‐patient clinic

Comorbidity: tic disorder diagnosis 100%; primarily obsessive‐compulsive disorder (OCD) and oppositional defiant disorder (ODD). Furthermore, conduct disorder, generalised anxiety disorder and major depressive disorder. Methylphenidate: obsessive‐compulsive disorder (11%), oppositional defiant disorder (33%). Placebo: obsessive‐compulsive disorder (22%), oppositional defiant disorder (41%). Clonidine: obsessive‐compulsive disorder (15%), oppositional defiant disorder (48%). Clonidine + methylphenidate: obsessive‐compulsive disorder (16%), oppositional defiant disorder (31%)

Comedication: not stated

IQ: > 70

Sociodemographics: not stated. Participant groups were similar, except that participants assigned to methylphenidate (alone or in combination with clonidine) are approximately 1 year older, present a higher proportion of pubertal cases, show underrepresentation of the inattentive subtype of ADHD (and overrepresentation of the combined subtype) and have lower baseline Conners' Abbreviated Symptom Questionnaire (ASQ)‐Teacher scores. A higher proportion of girls was found in the combined treatment group

Inclusion criteria

  1. DSM‐IV for ADHD

  2. Designated teacher in daily direct contact with the participant had to indicate the presence of a sufficient number of ADHD symptoms (rated as "pretty much" or "very much") in the classroom setting using the Disruptive Behavior Disorders Rating Scale (updated to DSM‐IV) to meet DSM‐IV criteria; and had to rate the severity of ADHD symptoms above specified cutoff scores (boys: grade 2 to 3 = 10, grade ≥ 4 = 9; girls: grade 2 to 3 = 7, grade ≥ 4 = 6) on the Iowa Conners' Teacher Rating Scale

  3. Investigator’s rating of global functioning on the Child‐Global Assessment Scale (C‐GAS) had to be 70 (indicating difficulty in ≥ 1 area, such as school)

  4. DSM‐IV for Tourette's disorder, chronic motor tic disorder or chronic vocal tic disorder

Exclusion criteria

  1. Secondary tic disorder (tardive tics, neuroacanthocytosis, Huntington disease)

  2. Major depression, pervasive developmental disorder, autism, psychosis, anorexia nervosa, bulimia, serious cardiovascular or other medical disorder that would preclude the safe use of methylphenidate or clonidine, impaired renal function, pregnancy

  3. Mental retardation

  4. The following cardiac conditions: prolonged QTc interval (> 440 milliseconds), high‐grade ventricular ectopy, AV block > 1 degree, bundle branch block, intraventricular conduction block (100 milliseconds), pacemaker rhythm or heart rate < 60 on the electrocardiogram, cardiomyopathy, complex heart disease, aortic or pulmonary stenosis, family history of long QT syndrome, cardiomyopathy or premature sudden death (age 45 years), history of syncope and blood pressure < 2 SD from age‐ and sex‐adjusted mean

  5. Other medications for treatment of ADHD, tics or other associated behavioural symptoms. Any such treatment had to be discontinued ≥ 6 weeks (2 weeks for methylphenidate) before enrolment

Interventions

Participants were randomly assigned to methylphenidate (Ritalin; Novartis) alone, clonidine alone, clonidine + methylphenidate or placebo

Methylphenidate dosage: methylphenidate alone 25.7 mg/d, methylphenidate + clonidine 26.1 mg/d

Administration schedule: 2 to 3 times daily

Duration of intervention: 12 weeks (4‐week titration of methylphenidate, 8‐week maintenance phase)

Titration period: 4‐week initial dose titration period for clonidine, after which came 4‐week dose titration for methylphenidate. Both titration periods took place after randomisation

Treatment compliance: Pill count monitored compliance

Outcomes

ADHD symptoms

  1. ADHD Conners' Abbreviated Symptom Questionnaire for Teachers (ASQ‐Teacher): rated at baseline and at week 16

  2. Iowa Conners' Teacher Rating Scale: rated at baseline and at week 16

  3. Conners' Abbreviated Symptom Questionnaire for Parents (ASQ‐Parent): rated at baseline and at weeks 8, 12 and 16

General behaviour

  1. Iowa Conners' Teacher Rating Scale: rated at baseline and at week 16

Quality of life

  1. Children's Global Assessment Scale: rated by site investigator at baseline and at weeks 8, 12 and 16

Non‐serious adverse events

  1. Yale Global Tic Severity Scale (YGTSS): rated by site investigator at weeks 8, 12 and 16

  2. Tic Symptom Self‐Report Scale (TSSR): parent/participant‐ and teacher‐rated at weeks 8, 12 and 16 (teacher ratings only at week 16)

  3. Global Tic Rating Scale: parent/participant‐ and teacher‐rated at weeks 8, 12 and 16 (teacher ratings only at week 16)

  4. Vital signs, ECG: rated at weeks 8, 12 and 16

  5. Side Effects Rating Scale: rated at weeks 8, 12 and 16 (teacher ratings only at week 16)

  6. Independent safety monitoring committee, consisting of child psychiatrist, paediatric cardiologist, paediatrician and statistician; reviewed data regarding adverse events throughout the study

Notes

Sample calculation: yes; 120 participants

Ethics approval: yes; the protocol was approved by the institutional review board at each site

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Our study did exclude participants with known cardiac problems, so the safety of combined clonidine and methylphenidate in this group was not addressed

Key conclusions of study authors

  1. Our study indicates that prior concerns that methylphenidate worsens tics and that the drug should be avoided in patients with tics may be unwarranted

  2. The most effective treatment for ADHD in our trial was the combination of clonidine and methylphenidate, although the incremental benefit of adding clonidine to methylphenidate came at the expense of additional side effects, particularly sedation

Comments from review authors

  1. Data extracted from week 4 to week 16 (from when methylphenidate was introduced to participants)

  2. Not able to use study data in our analyses

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation. Stratification by centre (investigator) and sexual maturity status (prepubertal: Tanner stage I to II; pubertal: Tanner stage III to V) Blocking was used to ensure approximate balance among treatment groups within each stratum

Allocation concealment (selection bias)

Low risk

Sealed envelopes that contained participants' treatment assignments

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinded: participants, clinicians, data collectors, outcome assessors, data analysts, data safety and monitoring committee, manuscript writers

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Only the programmer in the Biostatistics Center who generated the plan and the pharmacist in the Pharmacy Center who packaged and labeled the drug were aware of treatment assignments

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Primary statistical analyses were performed according to the ITT principle and were based on all randomly assigned participants, as randomised. For analysis of outcome variables for efficacy, if a participant was missing a response at a particular visit, the last available observation for that participant was carried forward and imputed for that visit

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): no

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

Unclear risk

Vested interest: the National Institute of Neurological Disorders and Stroke, the General Clinical Research Center, the National Center for Research Resources, the Tourette Syndrome Association

Boeringer Ingelheim Inc. (particularly Dr. Virgil Dias), for supplying clonidine and matching placebo; Bausch and Lomb, Inc., for supplying small gifts for our study participants

Conflicts of interest: none declared

Tucker 2009

Methods

Multi‐centre, open‐label RCT including behaviour treatment as cointervention with 2 arms

  1. Extended‐release methylphenidate and behavioural treatment

  2. Behavioural treatment

Participants

Number of participants screened: 142

Number of participants included: 109

Number of participants followed up: 104 (66 boys, 38 girls)

Number of withdrawals: 5

Diagnosis of ADHD: DSM‐IV (subtypes not described)

Age: mean 8.4 years (range 6 to 12)

IQ: not stated; all age‐appropriate cognitive functioning

Methylphenidate naive: 100%

Ethnicity: Caucasian (73.1%), African American (24.0%), other (2.9%)

Country: USA

Setting: out‐patient clinic

Comorbidity: none

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐IV diagnosis of ADHD

  2. Age‐appropriate cognitive functioning

Exclusion criteria

  1. Previous exposure to methylphenidate or any amphetamine‐based medication

  2. Positive urine drug screen

  3. Clinically significant abnormality in the screening assessment (physical exam, vital signs, laboratory tests)

  4. Cardiac abnormality

  5. History of seizures or schizophrenia; current diagnosis of mood disorder or anxiety disorder

Interventions

Participants were randomly assigned to extended‐release methylphenidate plus behavioural treatment or to behavioural treatment alone

Mean methylphenidate dosage: not stated

Administration schedule: once daily

Duration of intervention: 3 months

Titration period: initiated after randomisation. Methylphenidate was started at 10 mg/d and could be increased weekly in intervals of 10 mg/d to a maximum of 60 mg/d. Methylphenidate was administered to achieve the desired clinical effect with minimum or no side effects

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Conners' ADHD/DSM‐IV Scales for Parents: measured at baseline and at end of treatment

Non‐serious adverse events

  1. Investigating for potential genotoxic effects: No significant differences were found

  2. Chromosomal aberrations (CAs), including micronuclei (MN) and sister chromatid exchanges (SCEs), in cultured peripheral blood lymphocytes. Blood samples were obtained from all participating patients for evaluation of cytogenetic status at baseline and after 3 months of treatment. These data are not used in the review

Notes

Sample calculation: yes

Ethics approval: yes; approved by the ethics committees for all 17 study centres

Key conclusion of study authors

  1. These findings support the notion that methylphenidate does not induce chromosomal alterations nor other types of genetic damage in children treated for ADHD

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no; all participants were Methylphenidate naive

Email correspondence with study authors: December 2013 and January 2014. Not able to contact study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised; no further description

Allocation concealment (selection bias)

Unclear risk

Randomisation was stratified by age group (6 to 8 years and 9 to 12 years) and by centre

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Evaluation of cytogenetic damage by blinded slide readers

Incomplete outcome data (attrition bias)
All outcomes

High risk

Outcome data reported for 68 of 109 randomly assigned participants

Selective reporting (reporting bias)

Unclear risk

No protocol identified

Vested interest bias

High risk

This work was funded by Novartis Pharmaceuticals Corporation

Conflicts of interest: Some study authors were employed by Novartis (5 of 8 had a Novartis email address)

Ullmann 1985

Methods

Eight‐week, cross‐over trial

  1. Three doses of methylphenidate (0.3 mg/kg, 0.5 mg/kg, 0.8 mg/kg)

  2. Placebo

Two phases

  1. Phase 1: 4‐week fixed increase in methylphenidate dose

  2. Phase 2: 4‐week randomised, cross‐over trial

The trial was conducted over 3 years

Participants

Number of participants screened: not stated

Number of participants included: 86. Participants were randomly assigned to 3 doses of methylphenidate and placebo

Number of participants followed up: 86 (67 boys, 19 girls)

Number of withdrawals: 0
Diagnosis of ADHD: DSM‐III (ADD (15.1%), ADD‐H (70.9%), other (14.0%))

Age: mean 8.6 years (SD 1.8; range not stated)

IQ: > 70

Methylphenidate naive: not stated

Ethnicity: African American (17.4%), other (82.6%)

Country: USA

Setting: not stated

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria  

  1. ADHD diagnosis according to DSM‐III

Exclusion criteria

  1. Not described

Interventions

Participants were randomly assigned to different doses of methylphenidate (0.3 mg/kg, 0.5 mg/kg and 0.8 mg/kg) and placebo

Administration schedule: once daily, before school

Duration of each medication condition: 1 week

Washout before study initiation: dose taken in the morning to the next day's dose

Titration period: 4 weeks

Treatment compliance: "compliance were probably high"

Outcomes

ADHD symptoms

  1. ADD/H Comprehensive Teacher Rating Scale: completed by teachers on a weekly basis, on the last day before treatment switching  

Notes

Sample calculation:no information

Ethics approval:no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comment from study authors

  1. Order effects were non‐significant, that is, ratings were similar for the first and second weeks on each dose, regardless of the order in which doses were given

Key conclusion of study authors

  1. Results from the study show that methylphenidate has a major effect in improving attention and is helpful in decreasing activity levels but often has only a minor effect on deficient social skills and oppositional (aggressive) behaviour

Email correspondence with study authors: not able to find first or second study author's contact information; therefore not able to request supplemental data necessary for meta‐analyses

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random assignment

Allocation concealment (selection bias)

High risk

No description; only "methylphenidate in opaque capsules"

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No description

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Teacher rating under blinded conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data were pooled for a total of 86 participants

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Unable to obtain protocol

Vested interest bias

Unclear risk

National Institutes of Mental Health (NIMH). Ciba‐Geigy provided medication and placebo

Conflicts of interest: no information

Ullmann 1986

Methods

Double‐blind, cross‐over trial in which participants were randomly assigned to the following conditions

  1. Three different doses of methylphenidate (0.3 mg/kg, 0.5 mg/kg and 0.8 mg/kg)

  2. Placebo

Participants

Number of participants screened: not stated

Number of participants included: 118. Participants were randomly assigned to 3 different doses of methylphenidate and placebo

Number of participants followed up: 118 (92 boys, 26 girls)

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 8.6 years (range 6 to 14)

IQ: > 70

Methylphenidate naive: not stated

Ethnicity: Caucasian (81.4%), African American (18.6%)

Country: USA

Setting: school setting and out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosis of ADHD

Exclusion criteria

  1. None mentioned

Interventions

Participants were randomly assigned to 3 different doses of methylphenidate (0.3 mg/kg, 0.5 mg/kg and 0.8 mg/kg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: not stated

Duration of each medication condition: 1 week

Washout before study initiation: no information

Titration period: none

Treatment compliance: excellent compliance in all but a few children

Outcomes

ADHD symptoms

  1. ADD/H Comprehensive Teacher Rating Scale: rated by teachers at the end of each school week

Non‐serious adverse events

  1. Teacher Checklist: rated by teachers at the end of each school week

Unable to obtain data on adverse events, as could not locate contact details of the first or second study author (see notes)

Notes

Sample calculation: no

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Double‐blind placebo evaluation of children with ADD can and should be done by practitioners to avoid medicating children who are responding to non‐specific effects of drugs

Comment from study authors (on side effects)

  1. Very few children were reported completely free of adverse effects; however, these were, on the whole, mild effects. In fact, a slightly higher proportion of children were reported to show adverse psychological effects at baseline (0.60) and on baseline (0.53) than on medication (0.46), but these differences were not significant

Comment from review authors

  1. Used data from MPH responders in our analyses, which yielded a highly biased result

Email correspondence with study authors: not able to find first or second study author's contact information; therefore not able to obtain additional data (e.g. data (table) on adverse effects) from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information

Allocation concealment (selection bias)

Low risk

Daily doses of methylphenidate or placebo were placed in gelatin capsules to disguise taste and dose differences; medication for each week was packaged in a dated envelope

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Children, parents and teachers were blinded to dose order, as was the assistant who recorded data sent in by the teachers

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Children, parents and teachers were blinded to dose order, as was the assistant who recorded data sent in by the teachers

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No incomplete outcome data

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested interest bias

Unclear risk

Supported in part by a National Institute of Mental Health (NIMH) grant. Ciba‐Geigy provided medication and placebo

Conflicts of interest: not declared

Urman 1995

Methods

Double‐blind, placebo‐controlled, within‐participant (cross‐over) trial looking at cardiovascular effects of methylphenidate (0.3 mg/kg, 0.6 mg/kg and 0.9 mg/kg) doses at baseline and after 60 and 120 minutes post methylphenidate challenge in 2 groups of participants with ADHD

  1. ADHD without anxiety

  2. ADHD with anxiety

Participants

Number of participants screened: not stated

Number of participants included: 63 (58 boys, 5 girls)

Number of participants followed up: 63 (ADHD without anxiety 34, ADHD with anxiety 29)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R (but "82% of the ADHD and 93% of the ADHD/ANX group" would meet the DSM‐IV criteria for a diagnosis of ADHD combined type)

Age: ADHD without anxiety, mean 9.1 years (SD 1.3; range 6 to 12); ADHD with anxiety, mean 8.7 years (SD 1.4) (overall range 6 to 12)

IQ: not stated

Methylphenidate naive: 63 (100%)

Ethnicity: Caucasian (90%), "African or Asian descent" (10%)

Country: Canada

Setting: not stated

Comorbidity: ADHD group: oppositional defiant disorder (14; 42%), conduct disorder (5; 15%); ADHD with anxiety group: oppositional defiant disorder (11; 38%), conduct disorder (8; 28%). Also, in the ADHD with anxiety group, 3 met the DSM‐III criteria for overanxious disorder, 2 met criteria for separation anxiety disorder and 5 met criteria for both overanxious and separation anxiety disorders)

Sociodemographics: "The children tended to come from middle‐class families"

Inclusion criteria

ADHD with anxiety group

  1. T score ≤ 1 SD above the mean for age and sex on both the RCMAS and the Trait Scale of the STAIC

ADHD without anxiety group

  1. T score > 1 SD above the mean for age and sex on the RCMAS and/or the Trait Scale of the STAIC

  2. "The children could also meet diagnostic criteria for any anxiety disorder based on the parent interview"

Exclusion criteria

ADHD without anxiety group

  1. "Any child who scored within this range but met diagnostic criteria for any anxiety disorder on the basis of parent diagnostic interview was excluded from the analysis"

Interventions

Participants were randomly assigned to 1 of 4 possible drug condition orders of placebo and methylphenidate (0.3 mg/kg, 0.6 mg/kg or 0.9 mg/kg), given once daily over a 4‐day period. Dosage was based on a child’s body weight to the nearest 2.5 mg. Children followed the same routine for every test session with pre‐methylphenidate measurements and with measurements at 60 minutes and again at 120 minutes after taking methylphenidate
Mean methylphenidate dosage: not stated

Duration of each medication condition: once daily, administered over a 4‐day period ‐ each medication condition given once to each child

Washout before study initiation: All were methylphenidate naive

Medication‐free period between interventions: not stated

Treatment compliance: "Any child with missing data for a particular measure was excluded from the analysis of that measure", but study authors did not indicate the overall quantity of missing data. "Medication was administered by a research nurse"

Outcomes

Non‐serious adverse events

  1. Blood pressure (systolic and diastolic)

  2. Heart rate (radial pulse measured for 60 seconds)

Notes

Sample calculation: no information

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not stated

Comment from study authors

  1. From a clinical perspective, stimulant‐related increases in heart rate and blood pressure in both groups of children with ADHD were modest and generally of little clinical concern. Nonetheless, the present findings of an exaggerated cardiovascular response to methylphenidate in the ADHD with anxiety group should alert clinicians to the possibility of a differential medication response in anxious ADHD children

Key conclusions of study authors

  1. This study yielded 2 major findings. First, it demonstrated that baseline cardiovascular function did not differ between children with ADHD and children with ADHD and anxiety. Second, it revealed the presence of an exaggerated diastolic blood pressure response to methylphenidate in children with ADHD and anxiety

  2. Data from the present study add to growing evidence that children with ADHD and anxiety constitute a distinct subgroup of children with ADHD. Moreover, the exaggerated response of children with ADHD and anxiety to methylphenidate suggests the presence of abnormal regulation of norepinephrine (NE) function in this group, compared with that observed in the anxiety disorder population

Comments from review authors

  1. Inadequate description of method for blood pressure and heart rate measurement

  2. Measurement of heart rate was presumably carried out by palpating radial pulse ‐ pulse oximetry or ECG would produce more reliable results

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"The order of medications was counterbalanced and determined by random assignment such that an approximately equal number of children received each dose on a given day in the trial"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All medication was prepared by the hospital pharmacy and packaged in opaque gelatin capsules, to avoid detection of dose and taste"; "Children, parents and research staff were blind to the medication conditions"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Medication was administered by a research nurse who was blind to children’s medication condition and classification of anxiety"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not stated

Vested interest bias

Low risk

This work was supported in part by funds from the Medical Research Council of Canada and the Research Institute of the Hospital for Sick Children

Conflicts of interest: not declared

Van der Meere 1999a

Methods

Seven‐week, randomised, double‐blind, placebo‐controlled, parallel‐group trial with 3 arms

  1. Methylphenidate

  2. Placebo

  3. Clonidine

Participants

Number of participants screened: not stated

Number of participants included: 72 (62 boys, 10 girls)

Number of participants randomly assigned: methylphenidate 24, placebo 24, clonidine 24

Number of participants followed up in each arm: methylphenidate 23, placebo 24

Number of withdrawals in each arm: methylphenidate 1, placebo 0

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 8.8 years (range 7 to 12)

IQ: > 70

Methylphenidate naive: 100%

Ethnicity: mainly Caucasian

Country: the Netherlands

Setting: out‐patient clinic

Comorbidity: conduct disorder (11%), oppositional defiant disorder (33%), depressive disorder (3%), overanxious disorder (1%), dysthymia (3%), generalized tonic‐clonic seizures (1%), ventricular septal defect (1%), congenital hypothyroidism (1%), precocious puberty (1%), deaf in right ear (1%), atresia in 1 ear (1%)

Comedication: yes; for participants who had diseases that required it, for example, hypothyroidism

Sociodemographics: 44% from lower socioeconomic families

Differences between groups

  1. No significant differences in age and IQ were noted between the 2 groups

Inclusion criteria

  1. Boys and girls

  2. 6 to 15 years of age

  3. IQ > 70

  4. Living in a family home and attending school

  5. DSM‐III‐R diagnosis of ADHD, ADHD symptoms impeding development and psychological/educational treatments with insufficient effect

  6. No earlier use of stimulant drugs or clonidine and no psychoactive medications of any kind in the last 6 months

  7. No medical contraindications

  8. No important changes expected for the course of the trial

  9. Iit was considered clinically meaningful by both parents and the attending physician to try "hyperactivity medication"

Exclusion criteria

  1. Additional psychoactive drugs during the trial

  2. Pervasive developmental disorder or tic disorder. These participants were included in other trial groups

Interventions

Participants were randomly assigned to methylphenidate, placebo or clonidine

Total oral daily methylphenidate dosage: 0.06 mg/kg

Mean of the absolute methylphenidate dose: 9.85 mg (2.26)

Administration schedule: breakfast and lunchtime

Duration of intervention: 7 weeks

Titration: no, but dosage adjustments were made/allowed during first weeks

Titration period: none

Treatment compliance: maintained and checked by instructions (both oral and written) to both parents and child, and by counting tablets remaining at the end of treatment. Only 1 participant in the methylphenidate group showed poor compliance

Outcomes

ADHD symptoms

  1. Conners' Teacher Rating Scale: baseline and at week 7

General behaviour

  1. Parent and Teacher Versions of the abbreviated Groninger Behaviour Observation Scale (GOO and GBO, respectively): baseline and at weeks 3, 5 and 7

  2. Parent and Teacher Versions of the abbreviated Groninger Behaviour Checklists (GGGS and GGBS, respectively): baseline and at week 7

  3. GPO rating: baseline and at week 7

Non‐serious adverse events

  1. Parent ratings of drowsiness, insomnia, decreased appetite, nausea, headache, nervousness, motor restlessness, feelings of dizziness, dry mouth, nightmares, apathy, irritability and "other complaints"

  2. Child and parents were asked about all kinds of physical and behavioural complaints or changes at study visits

Notes

Sample calculation: yes (≥ 15 participants in each group)

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. We concluded that the state regulation problem in ADHD is resistant to methylphenidate and clonidine

Email correspondence with study authors: September 2013. We obtained supplemental information regarding a publication with information about randomisation and supplemental data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The statistician sent the randomisation list to the pharmacist. Participants were then randomly assigned by a research pharmacist. To ensure blinding, pharmacists applied randomisation blocks at random at a length of 2 or 4 participants. Methylphenidate and matching placebos were used

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Teacher, parent, clinician, child and experimenter were blind to treatment conditions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Teacher, parent, clinician, child and experimenter were blind to treatment conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT

Selection bias: no, but dosage adjustments were made for several participants because of annoying adverse events

Selective reporting (reporting bias)

Low risk

No

Vested interest bias

High risk

This study was supported by grants from the Sophia Foundation for Medical Research and Boehringer Ingelheim BV, the Netherlands

Wallace 1994

Methods

Double‐blind, single‐participant, randomised, cross‐over trial

  1. Methylphenidate

  2. Placebo

Conducted in 11 hospitalised children with ADHD

Participants

Number of participants screened: not stated

Number of participants included: 11. Participants were randomly assigned to 1 of 2 conditions: methylphenidate and placebo

Number followed up: 11

Sex: not reported

Number of withdrawals: 0

Diagnosis of ADHD: DSM‐III‐R (subtype not described)

Age: mean 9 years 5 months (range 4 to 13 years)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Setting: hospital/out‐patient clinic

Comorbidity type: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Children diagnosed with ADHD who were hospitalised at a psychiatric hospital

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to methylphenidate and placebo on a daily basis

Mean methylphenidate dosage: not stated

Administration schedule: daily

Duration of each medication condition: methylphenidate 2 to 7 days, placebo 3 to 6 days

Washout before study initiation: not described

Medication‐free period between interventions: none

Titration period: Optimal dose was based on maximal effectiveness and minimal side effects, but this appears to reflect daily response to methylphenidate

Treatment compliance: not described

Outcomes

ADHD symptoms

  1. Abbreviated Conners’ Teacher Rating Scale (15‐item): rated by teachers and by day and night nursing staff

Serious adverse events

  1. Side effects mentioned but not reported

Non‐serious adverse events

  1. Side effects mentioned but not reported

Notes

Sample calculation: not stated

Ethics approval: not stated

Comment from study authors

  1. Assigning highly consistent rates would reduce variance in measurements

Key conclusions of study authors

  1. n‐of‐1 trial is useful for evaluating the effectiveness of methylphenidate in individual patients with ADHD

  2. Short duration of MPH action allows for multiple cross‐over trials over a brief time

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Comment from review authors

  1. Data are not reported in a suitable form for meta‐analysis; therefore we could use no data from this study

Email correspondence with study authors: We could find no contact information; therefore, we could not get additional data through personal email correspondence with study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence was determined by "coin toss method"

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"After completion of each trial, the treatment code was broken", but it was not clear whether the psychiatry physician and the fellow who made the decision to cease methylphenidate were blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

All data appear to have been reported, but methylphenidate was discontinued on the basis of results; therefore, data collection did not continue for some participants

Selective reporting (reporting bias)

Unclear risk

Protocol not identified

Vested interest bias

Unclear risk

Veterans Administration Medical Center, Vermont

Conflicts of interest: not declared

Wallander 1987

Methods

Double‐blind, placebo‐controlled, cross‐over trial with 3 interventions

  1. Methylphenidate 0.3 mg/kg

  2. Methylphenidate 0.6 mg/kg

  3. Placebo

Participants

Number of participants screened: 28 (20 boys, 8 girls)

Number of participants included: not clear. Participants were randomly assigned to 1 of possible drug condition orders

Number of participants followed up: unclear

Number of withdrawals: unclear

Diagnosis of ADHD: DSM‐III (subtype not stated)

Age: mean 8.4 years (range 6 to 12)

IQ: mean 79.64

Methylphenidate naive: 28 (100%)

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic and in‐patient ward

Comorbidity: not stated

Comedication: not stated

Sociodemographics: 3.52 (Hollingshead‐Redlich Index)

Inclusion criteria

  1. Meeting DSM‐III criteria for the diagnosis of ADHD, as assessed by an experienced psychiatrist and paediatrician

  2. Scoring 2 SD above the mean for age and sex on both hyperactivity and distractibility factors of the Conners' Teacher Rating Scale

  3. Naive to stimulant medication

Exclusion criteria

  1. No information

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of methylphenidate (0.3 mg/kg and 0.6 mg/kg) and placebo

Mean methylphenidate dosage: no information

Administration schedule: twice a day, 8:30 AM and noon

Duration of each medication condition: 12 days for in‐patients and 19 days for out‐patients (mean 15.25 days)

Washout before study initiation: none

Medication‐free period between interventions: 68 hours

Titration period: none

Treatment compliance: no information

Outcomes

General behaviour

  1. Oppositional behaviour

Notes

Sample calculation: none

Ethics approval: no information

Key conclusions of study authors

  1. Results indicate no change in social behaviours, as participants decreased their display of problem behaviours as a function of stimulants

  2. Peers and teachers responded and attendees less to them, however, when they received stimulants as compared with placebo

Comment from review authors

  1. Data could not be used in meta‐analyses because some were missing

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Email correspondence with study authors: December 2013. No supplemental information available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Receiving interventions in counterbalanced orders

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Agreement of 90% across all categories had to be reached with this validity observer

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Protocol not identified

Vested interest bias

Low risk

Funded in part by Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) grants and the University of Southern California Faculty Research and Innovation Fund

Conflicts of interest: not declared

Waxmonsky 2008

Methods

Nine‐week therapeutic summer camp consisting of a cross‐over trial with 4 interventions

  1. Placebo

  2. Methylphenidate 0.15 mg/kg t.i.d.

  3. Methylphenidate 0.3 mg/kg t.i.d.

  4. Methylphenidate 0.6 mg/kg t.i.d.

Varied daily within a cross‐over design of 3 intensities of behaviour modification therapy

  1. No

  2. Low

  3. High

Each lasted 3 weeks

Participants

Number of participants screened: 106 participants in the 2003 and 2004 Summer Treatment Program (University of Buffalo)

Number of participants included: 101. ADHD subgroup 33, ADHD + severe mood dysregulation (SMD) subgroup 68

Number of participants followed up: 99

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐IV (combined (92%), hyperactive‐impulsive (not stated), inattentive (not stated))

Age: mean 8.5 years (range 5 to 12)

IQ: mean105

Sex: 82 boys, 19 girls

Methylphenidate naive: not stated

Ethnicity: predominantly Caucasian

Country: USA

Setting: out‐patient clinic (Summer Treatment Program)

Comorbidity: oppositional defiant disorder (54%), conduct disorder (12%)

Comedication: not stated

Sociodemographics: predominantly middle class

Inclusion criteria

  1. 5 to 12 years of age

  2. Participants were required to stop all psychotropic medication 1 week before intake

  3. DSM‐IV diagnosis of ADHD

  4. ADHD‐related impairment in ≥ 2 realms according to Parent and Teacher Versions of the Impairment Rating Scale (IRS)

  5. IQ > 80

  6. Subgroups

    1. ADHD: not meeting National Institutes of Mental Health criteria for severe mood dysregulation

    2. ADHD plus severe mood dysregulation: meeting National Institutes of Mental Health criteria for severe mood dysregulation, and having Young Mania Rating Scale (YMRS) score ≥ 12 based on the last month's behaviour and Conners' Global Index (CGI) mania severity score ≥ 3

Exclusion criteria

  1. History of seizures or other neurological problems

  2. Medical history that would involve considerable risk in taking stimulant medication

  3. History or concurrent diagnosis of any of the following disorders: pervasive developmental disorder, schizophrenia or other psychotic disorders, sexual disorder, organic mental disorder or eating disorder

  4. Documented serious adverse reaction to methylphenidate

  5. Significant developmental delays or autistic spectrum illness

  6. Active use of psychotropic medication for disorders besides ADHD, including use of antidepressants or mood‐stabilising medications

  7. Meeting full criteria for the narrow‐phenotype criteria of bipolar disorder or in need of urgent psychiatric treatment (active suicidal ideation). Participants newly identified with major depressive disorder or bipolar disorder on the Diagnostic Interview Schedule for Children were directly assessed by an M.D.‐ or Ph.D.‐level clinician

Interventions

Participants attended a Summer Treatment Program each Monday through Friday for 9 weeks. Participants were randomly assigned possible drug condition orders of 0.15 mg/kg t.i.d., 0.3 mg/kg t.i.d. and 0.6 mg/kg t.i.d. and placebo

Average methylphenidate dosages: 5 mg, 10 mg and 18 mg for 0.15 mg/kg, 0.3 mg/kg and 0.6 mg/kg doses, respectively

Administration schedule: 7:45 AM, 11:45 AM and 3:45 PM

Duration of each medication condition: Each dose varied daily and was repeated 3 or 4 times within each behavioural treatment condition

Study duration: Monday through Friday for 9 weeks, totalling 45 days

Washout before study initiation: 1 week

Medication‐free period between interventions: 0 to 2 days

Titration period: none

Treatment compliance: not stated

Cointervention: Three behavioural conditions (no behaviour modification, low‐intensity behaviour modification and high‐intensity behaviour modification) are delivered in random order, with each condition lasting 3 weeks. Parents attended training sessions and implemented behaviour programmes at home

Outcomes

Non‐serious adverse events

  1. Pittsburgh Side Effects Rating Scale: completed daily by camp staff and parents

Notes

Sample calculation: not stated

Ethics approval: yes; by the Health Sciences Institutional Review Board (IRB) of the University of Buffalo

Comment from study authors

  1. Limitations: Trial duration of only 9 weeks, lack of daily completion of mood assessments and limited generalisation potential due to a population of predominantly Caucasian and middle‐class participants

Key conclusion of study authors

  1. Methylphenidate and behaviour modification therapy are tolerable and effective treatments for children with ADHD and severe mood dysregulation, but additional treatments may be needed to optimise their functioning

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking MPH before randomisation: yes; excluded patients with prior serious reactions to methylphenidate
Any withdrawals due to adverse events: 1 withdrew because of tic‐like movements

Email correspondence with study authors: August 2014. Obtained supplemental information regarding randomisation, allocation concealment and handling of missing data. Not possible to retrieve safety data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random orders were generated by computer with the restrictions that each condition occurred at least once in each week. Children were assigned to previously generated codes at enrolment

Allocation concealment (selection bias)

Low risk

Because this was a cross‐over study in which all children received all conditions multiple times, each child's entire 9‐week schedule was assigned at once. Treatment orders were concealed in an opaque envelope and were stored in a locked cabinet in the medication lab. Only authorised staff members had access to this cabinet

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind. Children, parents and staff were blinded to medication conditions. Placebo and methylphenidate were packaged in identical opaque capsules to maintain blinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind. Parents and staff were blinded to medication conditions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

High risk

This study was funded by National Institute of Mental Health (NIMH) Grant MH62946 and a Klingenstein Third Generation Foundation Fellowship in Child and Adolescent Depression Research

Conflicts of interest: Several authors have affiliations with pharmaceutical companies

Whalen 1990

Methods

Two cross‐over trials with 3 interventions conducted 3 years apart

  1. Methylphenidate 0.3 mg/kg

  2. Methylphenidate 0.6 mg/kg (maximum 25 mg)

  3. Placebo

Participants

Number of participants screened: unknown

Number of participants included: study one 24, study two 25. Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: study one 24 (22 boys, 2 girls), study two 25 (25 boys)

Number of withdrawals: study one 0, study two 0

ADHD diagnosis: study 1 DSM‐III‐R, study 2 DSM‐III

Age: study one mean 9 years 8 months (range 6.4 to 13.2 years), study two mean 9 years 1 month (range 6.4 to 12.5 years)

IQ: no mental retardation (both studies)

Methylphenidate naive: 0 % (both studies)

Ethnicity: study one: Caucasian (92%), African American (4%), Hispanic (4%); study 2: Caucasian (72%), African American (12%), Asian (8%), Hispanic (8%)

Country: USA

Setting: out‐patient clinic (Summer Treatment Program)

Comorbidity: study one not stated, study two no gross neurological dysfunction

Comedication: not stated

Sociodemographics: All were from middle‐ or low middle‐income backgrounds (both studies)

Inclusion criteria

  1. Study one

    1. Primary diagnosis of ADDH

    2. Taking methylphenidate on a regular basis before the programme

    3. Conners' ADHD Stigma Questionnaire: ratings from parents > 15

  2. Study two

    1. Diagnosis of hyperactivity, ADD or ADDH

Exclusion criteria

  1. No information

Interventions

Participants were randomly assigned to 1 of the possible drug condition orders of 0.3 mg/kg (study one) or 0.6 mg/kg (study two) of methylphenidate and placebo. As regards study two, a single dose of 25 mg was set as the upper limit

Mean methylphenidate dosage: 8.75 mg

Administration schedule: twice a day, morning and lunchtime

Duration of each medication condition: 1 week

Washout before study initiation: unknown

Medication‐free period between interventions: 20 hours

Titration period: none

Treatment compliance: good (2 staff dispensed the medication for ingestion)

Outcomes

General behaviour

  1. UC‐Conners' Child Behavior Scale (UC‐CCBS): rated once by staff and twice by naive university undergraduates

Notes

Sample calculation: no

Ethics approval: no information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; only participants taking maintenance dosage of methylphenidate or well titrated

Comment from study authors

  1. Studies used similar procedures, with a few exceptions. The 2 sessions (i.e. study two) occurred between 1 and 2 weeks apart, rather than on consecutive days. Thus, the opportunity for medication washout was greater. Study two teams consisted of 3 rather than 4 players. In study two, 2 boys received scores below the hyperactivity cutoff on the maternal ratings, and paternal ratings yielded scores below the cutoff for 2 different boys

Key conclusions of study authors

  1. In both studies, double‐blind ratings done by naive and staff observers demonstrated nearly identical medication effects, that is, placebo‐related increases in behaviour problems and methylphenidate‐related increases in dysphoria

  2. Ratings of medication effects proved remarkably resilient, showing an invulnerability to biasing influences introduced by general knowledge about research design, diagnostic status or treatment effects; or by specific knowledge about and experience with participating children

Email correspondence with study authors: January 2014. No supplemental information has been received

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Study one: no information (unclear risk of bias)

Study two: Two thirds of the boys received medication first, and the remaining third received placebo first. (This unequal split resulted from the boys’ simultaneous participation in a randomised 3‐week dose‐response assessment that was not pertinent to the present study) (high risk of bias)

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Both active medication and placebo were placed in opaque gelatin capsules. Two staff, blinded to medication status, dispensed medication (low risk of bias)

Study 2: All staff were familiar with the boys, and some knew which boys had been diagnosed with ADHD, but none knew medication dosage or status (high risk of bias)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Study 2: All staff were familiar with the boys, and some knew which boys had been diagnosed with ADHD, but none knew medication dosage or status

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No drop‐outs

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

None

Vested interest bias

Unclear risk

No information

Wigal 2003

Methods

Double‐blind, 2‐stage, cross‐over, pharmacokinetic and pharmacodynamic trial with 4 interventions

  1. Ritalin 10 mg

  2. Immediate‐release or extended‐release methylphenidate 40:60 (treatment C)

  3. Immediate‐release or extended‐release methylphenidate 30:70 (treatment D)

  4. Placebo

Phases: initial screening week, stage 1 cross‐over of Ritalin vs placebo, stage 2 cross‐over of treatment C and treatment D. Four weeks, with each study period lasting 1 week

Participants

Number of participants screened: not stated

Number of participants included: 27. Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 25 (21 boys, 4 girls)

Number of withdrawals: 4

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 10 ± 1.4 years (range 7 to 12)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: Caucasian (88%), African American (8%), Asian (4%), Hispanic (0%), other (0%)

Country: USA

Setting: out‐patient clinic (laboratory and community)

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. One of 3 DSM‐IV ADHD diagnostic criteria as specified in the Diagnostic Interview Schedule for Children or the Swanson, Nolan and Pelham, or both

  2. 7 to 12 years of age

  3. In need of methylphenidate treatment

  4. Positive methylphenidate response

  5. Prior successful treatment of ADHD symptoms with a methylphenidate product without adverse events

  6. Methylphenidate products restricted to immediate‐release methylphenidate twice daily, with the first daily dose required to be between 7.5 and 15 mg, and the second daily dose required to be between 5 and 15 mg, yielding a total daily dose of between 12.5 and 30 mg; or a sustained‐release methylphenidate product taken once daily, with the single daily dose required to be 20 mg

  7. Oral or written consent by both children and parents

  8. Normal blood pressure, pulse rate and temperature

  9. Girls had to be premenarchal

Exclusion criteria

  1. Participation in another drug study during the preceding 30 days

  2. Concurrent illness or condition with symptoms that could affect performance of any of the tests performed

  3. Family history of drug abuse

  4. Unable to follow instructions given in the study

  5. Individuals who were severely depressed, psychotic, anxious, tense or agitated; or who had seizures or a family history of Tourette’s syndrome, with primary diagnosis of oppositional defiant disorder or conduct disorder

  6. Participants taking a medication in addition to methylphenidate for ADHD

  7. Documented allergy or intolerance to methylphenidate

  8. Individuals who were diagnosed with hyperthyroidism, were lactose‐intolerant or had glaucoma

  9. Participants unable to comply with blood drawing procedures during initial screening

  10. Use of any of the following medications: amphetamines, pemoline, tricyclic antidepressants, MAO (monoamine oxidase) inhibitors, serotonin reuptake inhibitors, neuroleptics, benzodiazepines or benzodiazepine derivatives, clonidine, anticonvulsant medications, cough/cold preparations containing stimulants or sedatives

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate and placebo in each of the 2 stages, 1 and 2, with each treatment period lasting 1 week

Stage 1

  1. Treatment A

    1. Participants were given 1 encapsulated tablet of Ritalin (10 mg) and 1 capsule of placebo after breakfast, and 1 encapsulated tablet of Ritalin (10 mg) after lunch for 7 days

  2. Treatment B

    1. Participants were given 2 placebo capsules after breakfast and 1 placebo capsule after lunch for 7 days

Stage 2

Half of the participants in each treatment group were randomly assigned to 20 mg/d dosage of methylphenidate, the other half to 40 mg/d dosage

  1. Treatment C

    1. Participants were given a daily morning dose (after breakfast) of two 20 mg capsules of the 40:60 prototype formulation or one 20 mg capsule of the 40:60 prototype formulation and 1 capsule of placebo; and a midday dose (after lunch) of 1 capsule of placebo for 7 days

  2. Treatment D

    1. Participants were given a daily morning dose (after breakfast) of two 20 mg capsules of the 30:70 prototype formulation or one 20 mg capsule of the 30:70 prototype formulation and 1 capsule of placebo; and a midday dose (after lunch) of 1 capsule of placebo for 7 days

    2. Blood samples (3 mL) were collected pre‐dose and at 0.5, 1.5, 2, 3, 4.5, 6, 7.5 and 9 hours after the morning dose on the last day (Saturday) of each treatment week

Mean methylphenidate dosage: 20 mg/d or 40 mg/d

Administration schedule: once daily or twice daily

Time points: mornings and after lunch

Duration of each medication condition: 1 week

Washout before study initiation: not stated

Medication‐free period between interventions: 1 day (Sundays)

Titration period: none

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham (10‐item) Scale: deportment and attention ratings performed every 1.5 hours (0 to 9 hours) at the end of each treatment week, by laboratory classroom teachers

  2. Swanon, Kotkin, Agler, M‐Flynn and Pelham (10‐item) Scale: rated on weekdays by community classroom teachers

  3. Conners, Loney and Milich (16‐item): rated Monday, Wednesday and Friday by community classroom teachers and parents

General behaviour

  1. Conners, Loney and Milich (16‐item) Scale: ratings performed once every Monday, Wednesday and Friday of each treatment week by the regular community classroom teacher and the parents

Non‐serious adverse events

  1. Side Effects Rating form completed during all treatments by the children’s regular community classroom teacher on Monday, Wednesday and Friday of each treatment week, daily by parents and each Saturday by the University of California at Irvine Child Developmental School (UCI‐CDC) classroom teacher

  2. All adverse events occurring during the study were reviewed by the investigator to assess their relationship to drug treatment (unrelated, unlikely, possibly, probably, almost certainly)

  3. In addition, each sign or symptom reported was graded on a 3‐point scale (mild, moderate or severe), and date and time of onset, time relationship to drug dosing, duration and outcome were noted

  4. Clinical laboratory tests were performed at the local laboratory at baseline and at the end of the trial, and included blood count with differential, biochemistry (Na (sodium), K (potassium), Ca (calcium), PO4 (phosphate), total protein, glucose, alkaline phosphatase, AST (aspartate aminotransferase), ALT (alanine aminotransferase), total bilirubin, creatinine, albumin) and urinalysis (osmolality, pH (power of hydrogen) level, glucose, protein, white blood cells and casts)

  5. Vital signs, including blood pressure and heart rate, were measured at each visit to the laboratory classroom

  6. Body weight: 36.9 ± 7.6 kg

  7. Height: 142 ± 8.4 cm

Notes

Sample calculation: not stated

Ethics approval: yes; study protocol and participant's informed consent form were approved by the Institutional Review Board (Office of the Vice Chancellor for Research, University of California at Irvine)

Key conclusion of study authors

  1. Both methylphenidate formulations, given once in the morning, were superior to placebo and comparable with Ritalin b.i.d. treatment on all primary efficacy measures

Comments from review authors

  1. Prototype formulation used ‐ this may not be bioequivalent to the marketed formulation

  2. Selective reporting of safety findings ‐ omission of data obtained by adverse events rating form. Pre‐specified primary outcome measure for efficacy (of prototype formulations versus placebo) ‐ regular community classroom teacher. Conners’ Global Index scores from Collegiate Learning Assessment (CLA) results presented only in Figure 4; no mean (SD) values provided in publication

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; methylphenidate tolerability was an inclusion criterion. Serious adverse events were an exclusion criterion

Email correspondence with study authors: July 2014. Emailed study authors twice for supplemental information but have received no response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Qualified participants were randomly assigned to a 2‐stage, double‐blind study sequence consisting of 4 study treatments, each lasting for a period of 1 week

Allocation concealment (selection bias)

Low risk

Ritalin (10 mg) tablets were placed into capsule shells by Eurand Americas Incorporated. Resulting capsules were tested according to US Pharmacopeial Convention (USP) dissolution conditions for methylphenidate tablets and showed a dissolution profile comparable with intact Ritalin tablets. All medications were supplied in white, opaque, size 3, hard gelatin capsule shells, and were packaged in blister cards to enhance compliance

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Ritalin (10 mg) tablets were placed into capsule shells by Eurand Americas Incorporated. Resulting capsules were tested according to US Pharmacopeial Convention (USP) dissolution conditions for methylphenidate tablets and showed a dissolution profile comparable with intact Ritalin tablets. All medications were supplied in white, opaque, size 3, hard gelatin capsule shells, and were packaged in blister cards to enhance compliance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): yes

Selective reporting (reporting bias)

Unclear risk

"Because the administration of the Side Effects Rating Form involved queries about specific adverse events, the frequency of adverse events reported in the parents’ and teachers’ Side Effect Rating Form was higher than that obtained from the reports elicited by general inquire (data not shown)"

Vested interest bias

High risk

Funding for this study was provided by Celltech Americas Incorporated

Conflicts of interest: Some study authors are working for Celltech Americas Incorporated

Wigal 2004

Methods

Four‐week, randomised, double‐blind, placebo‐controlled, ITT, parallel trial conducted at 12 US centres with 3 arms

  1. Dexmethylphenidate

  2. Dex,I‐methylphenidate

  3. Placebo

Participants

Number of participants screened: 174

Number of participants included: 132 (116 boys, 16 girls)

Number of participants randomly assigned: dexmethylphenidate 44, dex,l‐methylphenidate 46, placebo 42

Number of participants followed up: dexmethylphenidate 42, dex,l‐methylphenidate 40, placebo 37

Number of withdrawals: dexmethylphenidate 2, dex,l‐methylphenidate 6, placebo 5

Diagnosis of ADHD: DSM‐IV (combined (64%), hyperactive‐impulsive (1%), inattentive (35%))

Age: mean 9.8 years (range 6 to 17)

IQ: not stated. No mental retardation

Methylphenidate naive: 95 (72%)

Ethnicity: Caucasian (78%), African American (14%), other (8%)

Country: USA

Setting: out‐patient clinic

Comorbidity: no

Comedication: no

Sociodemographics: not stated. No significant differences in baseline demographics were noted between the 2 groups

Inclusion criteria

  1. Enrolled in elementary school

  2. Within 30% of normal body weight

  3. Anticipated as available for the entire length of the study

  4. Female participants were required to be pre‐menarche

Exclusion criteria

  1. History or evidence of cardiovascular, renal, respiratory (other than asthma/allergy), endocrine or immune system disease

  2. History of substance abuse

  3. Hypersensitivity to dex,l‐methylphenidate or other stimulants

  4. Treatment with any investigational drug within 30 days of screening

  5. Any other significant central nervous system disorders such as mental retardation, Tourette’s or chronic tic disorder, psychosis, pervasive developmental disorder, eating disorders, obsessive‐compulsive disorder, impulse control disorder or sleep disorders requiring medication, major depressive disorder or generalised anxiety disorder

  6. Treatment with antidepressants (tricyclic antidepressants, serotonin reuptake inhibitors and monoamine oxidase inhibitors), sedative/hypnotics (e.g. barbiturates, benzodiazepine), neuroleptic/antipsychotics, mood stabilisers; anticonvulsants, beta‐blockers, α2‐agonists, thyroid medications and long‐term oral steroids

Interventions

Participants were randomly assigned to immediate‐release methylphenidate (dex‐ or d,l‐ racemats) or placebo

Mean methylphenidate dosage: dexmethylphenidate 18.25 mg/d, dex,i‐methylphenidate 32.14 mg/d

Administration schedule: twice daily in the morning (7:00 AM to 8:00 AM) and at noon (11:30 AM to 2:30 PM)

Duration of intervention: 4 weeks

Titration period: maximum 3 weeks initiated after randomisation, included in the 4 weeks of intervention

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Nolan and Pelham ADHD Rating Scale ‐ Teacher Version: teacher‐rated, at baseline and twice weekly for 4 weeks, in the afternoon

  2. Swanson, Nolan and Pelhan ADHD Rating scale ‐ Parent Version: parent‐rated, at baseline and daily on the weekend for 4 weeks, at 3:00 PM and 6:00 PM

Non‐serious adverse events

  1. Occurrence and severity, monitored by investigator, at weekly visits

  2. Laboratory tests, physical examination findings and vital signs

Notes

Sample calculation: yes

Ethics approval: yes; approved by the institutional review board at each centre

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Comments from study authors

  1. Post hoc contrasts of small differences between dexmethylphenidate and dex,l‐methylphenidate conditions were not statistically significant in this between‐participant design

  2. Three participants who did not meet the criteria set for a placebo response during the lead‐in period were inadvertently entered into the double‐blind phase (2 randomly assigned to dex,l‐methylphenidate, 1 to placebo), which was a violation of the protocol

Key conclusions of study authors

  1. For treatment of ADHD, an average titrated dose of 18.25 mg/d of dexmethylphenidate is as efficacious and safe as an average titrated dose of 32.14 mg/d of dex,l‐methylphenidate

  2. Both active treatments have large effect sizes. Thus, dexmethylphenidate and dex,l‐methylphenidate appear to provide similar efficacy

Comment from review authors

  1. We have not used these data, as reported data could not be used in our meta‐analyses

Email correspondence with study authors: January 2014. Not possible to obtain data from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomised"

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Drug (dexmethylphenidate and dex,l‐methylphenidate) and placebo were identical in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Sample size calculations were based on a clinically meaningful effect size of 0.75 in the change in score over 4 weeks on the teacher‐rated Swanson, Nolan and Pelham Scale between dexmethylphenidate and placebo groups. Efficacy parameters were performed on the ITT sample, which included participants who received medication, had a baseline efficacy evaluation and had ≥ 1 post‐baseline efficacy evaluation

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No protocol published

All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

This study was supported by Celgene Corporation

Conflicts of interest: Dr. Wigal reports extensive disclosure

Wigal 2013

Methods

Four‐ to six‐week, open‐label treatment (dose optimisation), cross‐over, 2‐week double‐blind trial with 2 interventions

  1. Methylphenidate (NWP06 ‐ liquid formulation of extended‐release methylphenidate)

  2. Placebo

Phases: 2

Participants

Number of participants screened: 45 (32 boys, 12 girls)

Number of participants included: 44. Participants were randomly assigned to 1 of the possible drug condition orders

Number of participants followed up: 39

Number of withdrawals: 6

Diagnosis of ADHD: DSM‐IV (combined (70.5%), hyperactive‐impulsive (2.3%), inattentive (27.3%))

Age: mean 8.8 years (range 6 to 12)

IQ: not stated

Methylphenidate naive: 0 (0%)

Ethnicity: White 35 (79.5%), Black/African American 4 (9.1%), Asian 3 (6.8%), other 2 (4.5%)

Country: USA

Setting: out‐patient clinic

Comorbidity: elimination disorder (4; 9.1%), oppositional defiant disorder (8; 18.2%), specific phobias (2; 4.5%)

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. ADHD diagnosis by psychiatrist, psychologist, developmental paediatrician or paediatrician

  2. Pharmacological treatment for ADHD and has experienced suboptimal efficacy or a safety or tolerability issue with current regimen, or has been in need of a long‐acting liquid formulation

  3. Conners' Global Index Scale score > 3

  4. ADHD Rating Scale score (Hyperactive‐Impulsive or Inattentive subscale) > 90th percentile for age and sex

Exclusion criteria

  1. Comorbidity (DSM‐IV Axis I), with the exceptions of specific phobia, motor skills disorders, oppositional defiant disorder, sleep disorders, elimination disorders, adjustment disorders, learning disorders or communication disorders

  2. IQ < 80

  3. Chronic disease: seizure disorder, thyroid disease, Tourette’s disorder or family history of Tourette’s disorder or tics, serious cardiac conditions, cardiomyopathy, serious arrhythmias, structural cardiac disorders, glaucoma or severe hypertension

  4. Any investigational medication 15 days before screening

  5. Atomeoxetine (ATX) inhibitor 30 days before screening

Interventions

Participants were randomly assigned to different sequences of methylphenidate and placebo

Mean methylphenidate dosage: 32.8 mg/d

Administration schedule: q.i.d.

Duration of each medication condition: 1 week

Washout before study initiation: yes (1 day for stimulants)

Medication‐free period between interventions: no

Titration period: 3 weeks before randomisation

Treatment compliance: 2 withdrawals of assent/consent, 2 adverse events, 1 lack of efficacy, 1 LTFU

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham Scale, ADHD Rating Scale (open‐label phase)

Non‐serious adverse events

42 participants (93.3%) experienced a treatment‐emergent adverse event. Three (6.7%) participants experienced severe adverse effects (affect lability, aggression and initial insomnia), and 2 (4.4%) participants had to discontinue medication (affect lability and aggression)

  1. Open‐label phase: Participants experienced decreased appetite (55.6%), abdominal pain upper (42.2%), affect lability (26.7%), initial insomnia (22.2%), insomnia (17.8%) and headache (17.8%). Other adverse events reported in more than 5% of participants included vomiting, diarrhoea, logorrhoea, aggression, dizziness, irritability, fatigue, upper respiratory tract infection, cough and flushing

  2. Double‐blind phase: 11 (24.4%) participants had an adverse event while receiving NWP06, and 5 (11.1%) participants had an adverse event while receiving placebo

Notes

Sample calculation: no

Ethics approval: yes

Comments from study authors

  1. "This study of NWP06 allowed inclusion of patients who were either treatment naive or had previously been treated with stimulants (...)"; "Subjects were required to have been in need of pharmacological treatment for ADHD (...)"

  2. "Our population more closely reflects a real‐world population and provides a more rigorous test of the study drug"

Key conclusion of study authors

  1. NWP06 resulted in significant improvement in the Swanson, Kotkin, Agler, M‐Flynn and Pelham‐combined score at 4 hours post dose as compared with placebo among completers. This study shows that NWP06 significantly improved ADHD symptoms in school‐aged children and was well tolerated

Comments from review authors

  1. Laboratory school environment and lack of the ADHD Rating Scale

  2. Race/ethnicity does not reflect a real‐world population

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: not clear

Any withdrawals due to adverse events: yes (n = 2)

Email correspondence with study authors: emailed study authors to request additional information but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Results from open‐label phase

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Results from open‐label phase

Incomplete outcome data (attrition bias)
All outcomes

High risk

Participants had to be in treatment with suboptimal efficacy (no inclusion of placebo responders)

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): Six participants were LTFU during the open‐label titration phase, 2 as a result of adverse events. These participants are not included in our analyses

Selective reporting (reporting bias)

Low risk

ADHD Rating Scale not reported (used only in the open phase)

Vested interest bias

High risk

Study received funds from NextWave Pharmaceutics (Belden and Berry are with NextWave)

Conflicts of interest: All study authors are affiliated with NextWave Pharmaceuticals

Wigal 2014

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate‐MLR

  2. Placebo

Phases: 4

  1. Screening/washout period (< 4 weeks)

  2. Open‐label dose‐optimisation period

  3. Randomised, double‐blind, placebo‐controlled, cross‐over design

  4. 30‐day safety follow‐up period

Participants

Number of participants screened: 32

Number of participants included: 26 (in open‐label phase). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 20

Number of withdrawals: 6

Diagnosis of ADHD: DSM‐IV‐TR (combined (n = 11), hyperactive‐impulsive (n = 3), inattentive (n = 12))

Age: mean 8.7 years (range 6 to 12)

IQ: mean not stated (range 86 to 133)

Sex: 12 boys, 10 girls

Methylphenidate naive: none

Ethnicity: White (82%), Black (9%), Asian (5%), Hispanic or Latino (23%), other (5%)

Country: USA

Setting: out‐patient clinic

Comorbidity: 11; generalised anxiety disorder, enuresis, oppositional defiant disorder, chronic motor or vocal tic disorder, transient tic disorder

Comedication: no

Sociodemographics: not stated

Inclusion criteria

  1. Children (male or female) 6 to 12 years of age

  2. Any of the 3 subtypes of ADHD as defined by DSM‐IV‐TR, ADHD‐RS‐IV total or subscale score > 90th percentile relative to the general population of children by age and sex

  3. Naive to treatment for ADHD or inadequately managed on current treatment regimen

  4. Negative illicit drug and alcohol test results at screening and at each visit to the research site

Exclusion criteria

  1. IQ > 80

  2. Any severe psychiatric or significant comorbid condition

  3. Use of a monoamine oxidase inhibitor or any psychotropic medication with central nervous system effects < 14 days of screening, or any experimental drug or medical device > 30 days of screening

  4. Clinically significant electrocardiogram (ECG), or any laboratory abnormality

  5. Any participant unable or unwilling to follow directions and complete study assessments or take oral capsules

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate (15 mg, 20 mg, 30 mg or 40 mg) and placebo

Mean methylphenidate dosage: 32 mg

Administration schedule: once a day

Time points: in the morning

Duration of each medication condition: 1 week

Washout before study initiation: 2 days

Medication‐free period between interventions: not stated

Titration period: yes; 2 to 4 weeks before randomisation

Treatment compliance: yes; verified at scheduled study visits by study personnel who examined documentation of drug dispensed, drug consumed and remaining drug, and recorded the information on the drug reconciliation form

Compliance was calculated to be > 82% throughout the study

Outcomes

ADHD symptoms

  1. Swanson, Kotkin, Agler, M‐Flynn and Pelham: total scores, trained observers, post dose over time points 1.0, 2.0, 3.0, 4.5, 6.0, 7.5, 9.0, 10.5 and 12.0 hours

  2. Swanson, Kotkin, Agler, M‐Flynn, and Pelham: attention and deportment scores averaged over all post‐dose time points

  3. ADHD Rating Scale, Fourth Edition: clinician‐rated, 3.0 hours post dose each laboratory day

Non‐serious adverse events

  1. Safety and tolerability assessments

Notes

Sample calculation: yes

Ethics approval: yes

Comment from study authors

  1. Limitations of this study: Study was slightly underpowered and produced a study population that may not be reflective of the general population of children and adolescents with ADHD

Key conclusions of study authors

  1. In this study, methylphenidate‐MLR administered to children 6 to 12 years of age demonstrated a significant decrease in scores on the Swanson, Kotkit, Agler, M‐Flynn and Pelham Scale compared with placebo

  2. Onset of action in this population is 1 hour, and duration of efficacy is sustained to 12 hours post dose

  3. Future studies that include measurement of efficacy earlier than hour 1.0 and extend beyond hour 12.0 would add clarity to the precise onset and duration of clinical efficacy

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; 2 withdrew during the open‐label phase as the result of lack of efficacy

Any withdrawals due to adverse events: yes (n = 1)

Email correspondence with study authors: April 2015. Obtained supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Randomisation was determined by using a table of random numbers. Randomisation tables were provided to the randomisation monitor, who created unblinding envelopes and packaged the drug with blinded labels

Allocation concealment (selection bias)

High risk

One participant received placebo at 2 periods ‐ the method was not efficacious

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All sponsor representatives, investigators, participants and independent raters remained blinded until after data were locked

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Yes; this has been done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT were used

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo‐responders): no; no exclusion of methylphenidate non‐responders after randomisation

Selective reporting (reporting bias)

Low risk

No selective reporting bias

Vested interest bias

High risk

Study was funded by Rhodes Pharmaceuticals L.P.

Conflicts of interest: Several study authors work for, or have received grant and research support or both from pharmaceutical companies

Wilens 2006b

Methods

Two‐week, randomised, double‐blind, 15‐centre, parallel trial with 2 arms

  1. OROS methylphenidate

  2. Placebo

Phases: preceded by a 4‐week, open‐label, dose‐titration phase, and followed by an 8‐week, open‐label, follow‐up phase

Participants

Number of participants screened: not stated

Number of participants included: 220 in the 4‐week dose‐titration phase

Number of participants randomly assigned: 177 (142 boys, 35 girls); methylphenidate 87, placebo 90

Number of withdrawals: methylphenidate 16, placebo 28

Number of participants followed up: 171; number completing follow‐up: 135

Diagnosis of ADHD: DSM‐IV (subtype not stated)

Age: mean 14.6 years (range 13 to 18)

IQ: not stated

Methylphenidate naive: not stated

ADHD treatment‐naive: 24

Ethnicity: Caucasian (75.1%), African American (13.6%), other (11.3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated. The 2 groups were similar demographically, but the placebo group had a greater ratio of males (P value < 0.04)

Inclusion criteria

  1. Diagnosis of attention deficit hyperactivity disorder (ADHD), as defined by the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM‐IV)

  2. Children's Global Assessment Scale rating of 41 to 70 at baseline (screening phase)

  3. Between 8 and 13 years of age

Exclusion criteria

  1. Participants who are known to not respond to methylphenidate

  2. Adverse experiences from methylphenidate or hypersensitivity to Concerta or its components

  3. Marked anxiety, tension or agitation

  4. Psychiatric comorbidity requiring additional or different medication

  5. Glaucoma, ongoing seizure disorder, psychotic disorder, Tourette's disorder or family history of Tourette's disorder, bipolar disorder, an eating disorder

  6. Treatment with theophylline, coumarin, anticonvulsants

  7. Severe gastrointestinal narrowing

  8. Systolic or diastolic blood pressure at the 95th percentile or greater for age, sex and height at screening

Interventions

Participants were randomly assigned to OROS methylphenidate or placebo

Mean methylphenidate dosage: 0.84 mg/kg

Administration schedule: once daily

Duration of intervention: 2 weeks

Titration period: 4 weeks initiated before randomisation. All participants initiated therapy at 18 mg/d, and clinical response was measured after 1 week. If response to treatment was inadequate, as per the a priori study definition, the dose was titrated upward (in 18‐mg increments) at 1‐week intervals for up to 4 weeks, with maximum dose of 72 mg/d

Treatment compliance: not stated; 8‐week, open‐label follow‐up on individualised dosage

Outcomes

ADHD symptoms

  1. ADHD Rating Scale, clinician‐ and parent‐rated: completed at baseline and weekly during double‐blind phase

Serious adverse events

  1. Reported in only 1 participant during the open‐label dose‐titration phase of the study. While being treated with OROS methylphenidate 18 mg/d, a 16‐year‐old female participant with a history of depression and suicidal ideation threatened suicide on the third day of medication use after an argument with her mother. Decision was made to discontinue study medication, and symptoms resolved

  2. No serious adverse events were reported during the double‐blind phase

Non‐serious adverse events

  1. Heart rate and blood pressure, recorded by a clinician weekly throughout the whole study

  2. ECG at screening and at end of double‐blind phase of the study

  3. Spontaneous reports to the investigator of adverse events recorded at weekly visits

  4. Safety assessments made at monthly visit and every 2 weeks between monthly visits during follow‐up

  5. Height and weight assessed at baseline and at weeks 4 and 8 in the follow‐up study

No participants experienced clinically important effects on ECG indexes, heart rate or blood pressure during the study

Notes

Sample calculation: yes

Ethics approval: yes; study was approved by the institutional review boards for all participating centres before the start of the study

Inclusion of methylphenidate responders only/exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; criteria of response, defined as ≥ 30% improvement from baseline on the investigator‐scored ADHD Rating Scale. Participants who successfully completed the open‐label dose‐titration phase were assigned a randomisation number

Comments from study authors

  1. Exclusion criteria were significant and possibly limited the generalisability of results

  2. Participants may have had improved ADHD symptoms or psychosocial carry‐over effects as a consequence of participation in the study, medication titration and regular meeting with study personnel

  3. Our study was conducted partially during the summer, and this may have resulted in less stress on adolescents and overall improvement in both study groups. More sensitive measures of attention may be needed for adolescents with ADHD than for children

  4. Participants were titrated to their individualised dosage before the double‐blind phase of the study. This may have biased the results toward a positive response in the double‐blind phase

The short duration of the double‐blind phase may have decreased the likelihood of detecting potential rare adverse events. Rates of adverse events reported for OROS methylphenidate have been underestimated because participants entering this study phase were already stabilised on an affective tolerated dosage of medication

Key conclusions of study authors

  1. In adolescents, once‐daily OROS methylphenidate significantly reduced ADHD symptoms and was well tolerated at dosages up to 72 mg/d

  2. Adolescents required, on average, a higher absolute dose but a lower weight‐adjusted dose (mg/kg) of OROS methylphenidate than was previously reported in children

  3. The incidence of adverse events was not related to dose

Email correspondence with study authors: December 2013 and Janurary 2014. Not able to make contact with study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, but method was not described

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Investigators were supplied with packages containing medication for each participant, as identified by randomisation number. Therefore, investigators and participants were blinded to whether a participant was receiving active medication or placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Investigators were supplied with packages containing medication for each participant, as identified by randomisation number. Therefore, investigators and participants were blinded to whether a participant was receiving active medication or placebo

Incomplete outcome data (attrition bias)
All outcomes

High risk

Non‐responders not included. LOCF technique was used for all assessments in the double‐blind phase. Of 182 (83%) participants who successfully achieved the criteria for improvement at the dose‐titration phase, only 177 were randomly assigned to the double‐blind phase, because 5 reached the criteria after the double‐blind phase was closed. Of 177 randomly assigned participants, 1 did not enter the double‐blind phase, and efficacy data were not collected for another participant. Therefore, 175 participants were included in the efficacy analysis of the double‐blind phase, but 177 were included in the dosage and safety analysis

Selective reporting (reporting bias)

Low risk

No major violation compared with what is reported in ClinicalTrials.gov

All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

Funded by McNeil Consumer and Specialty Pharmaceuticals

Conflicts of interest: Several study authors have had commitments (e.g. speakers, consultants, advisors) with various pharmaceutical companies

Wilens 2008

Methods

Open‐label, 5‐week, dose‐optimisation period. This dose was maintained during the subsequent 3 weeks of the trial, except for 1 day per week of 3‐way cross‐over assessment

Randomised, double‐blind, 8‐centre, 3‐way, 3‐week, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: 148

Number of participants included: 128 in open‐label, dose‐titration; 120 randomly assigned to 1 of 3 possible drug orders

Number followed up: 127 for safety and 117 for efficacy

Number of withdrawals: 2

Characteristics of the 127 followed up for safety

Sex: 84 boys, 42 girls

Diagnosis of ADHD: DSM‐IV (combined or hyperactive‐impulsive (92.1%) inattentive (not stated))

Age: mean 8.8 years (SD 1.84; range 6 to 12)

IQ: > 80

Methylphenidate naive: not stated

Ethnicity: Caucasian (63.2%), African American (15.4%), Asian (not stated)

Country: USA

Comorbidity: not allowed

Comedication: not stated

Sociodemographics: not stated

Characteristics of the 117 followed up for efficacy

Sex: 75 boys, 42 girls

Diagnosis of ADHD: DSM‐IV (type not stated)

Age: mean 8.8 years (range 6 to 12)

IQ: > 80

MPH‐naive: not stated

Ethnicity: Caucasian (63.2%), African American (15.4%), Asian (0%), other (21.4%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not allowed

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Diagnosed with ADHD according to DSM‐IV‐T

  2. Minimum IQ score of 80

Exclusion criteria

  1. Conduct disorder or comorbid illnesses that contraindicated or could confound methylphenidate transdermal system treatment

  2. History of failing to respond to psychostimulant treatment

  3. Taken another investigational product within 30 days of screening or participated in other research trials involving drug treatment during the course of the study

  4. Safety population consisted of 127 participants, who received ≥ 1 dose of study medication

11 participants discontinued before the double‐blind randomisation phase, resulting in an ITT population of 117 participants

Seven participants discontinued before the double‐blind randomisation phase, 3 were randomly assigned but did not undergo methylphenidate transdermal system (MTS) treatment

Two participants did not complete the analogue classroom phase of the study: 1 because of an application site reaction, and 1 because of an adverse event (conjunctivitis), resulting in a total of 115 study completers

Interventions

Participants were randomly assigned to 1 of 3 possible drug orders of methylphenidate (4 and 6 hours) and placebo

Mean methylphenidate dosage: 10 mg patch (n = 15), 15 mg patch (n = 34), 20 mg patch (n = 32) and 30 mg patch (n = 36)

Administration schedule: once daily in the morning; patch worn for 9 hours daily, and for 4 or 6 hours for cross‐over assessments

Duration of each medication condition: 1 day

Washout before study initiation: none; in‐between treatment with optimal dose of methylphenidate

Titration period: 5 weeks before randomisation

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. Swanson, Kotkit, Agler, M‐Flynn and Pelham: teacher‐rated at randomisation (week 5) and 2 hours after patch application at end of treatment (week 6, 7, 8)

Quality of life

  1. ADHD Impact Module‐Child ‐ Child Impact Scale and Family Impact Scale: rated at baseline, at randomisation (week 5) and at end of study (week 8)

Non‐serious adverse events

  1. Vital signs were evaluated at screening, at baseline and at weeks 1 to 8

  2. Erythema, edema, papules and vesicles, discomfort, haematology, urinalysis and electrocardiographic measures were completed at screening, at baseline and at weeks 5 and 8

No clinically meaningful changes from baseline were observed in vital signs, ECG, urinalysis and haematological results or physical examinations. Adverse events were recorded from the time informed consent was signed until 30 days (week 12) after the last drug treatment

Notes

Sample calculation: yes; assuming a mean difference in Swanson, Kotkit, Agler, M‐Flynn and Pelham deportment score of 2.0 between active treatment and placebo, an SD of 5.0, a between correlation of 0.2, 90% power and a probability level of .05 (2‐sided), it was estimated that approximately 102 participants were needed to complete the double‐blind, cross‐over phase of the study

Ethics approval: yes; institutional review board at each site approved the study

Comments from study authors

  1. Important to note that participants who failed to respond to psychostimulants in the past and those with conduct disorder were excluded from the study. Therefore, results of this study should not be extrapolated to these patient populations

  2. From Manos in Wilens 2008: Lack of placebo comparison has the potential to confound the findings of this study. The relatively short study duration (about 2 months) may not be sufficient to capture some emerging changes in health‐related quality of life

Key conclusion of study authors

  1. All efficacy measures indicated that 4‐ and 6‐hour wear times improved ADHD symptoms

Comments from review authors

  1. Treatment period is 1 day, 1 week apart for the 3 phases, and all participants are treated with methylphenidate at optimal titrated dose in‐between

  2. Quality of life assessments are aggregated for all (not possible to assess methylphenidate vs placebo)

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; participants with a history of failing to respond to psychostimulant treatment were also excluded

Any withdrawals due to adverse events: yes; 1 (conjunctivitis)

Email correspondence with study authors in April to June 2014. Emailed study authors twice to ask for additional data but never received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Single randomisation schedule prepared by an independent statistician using computer software that generated random numbers

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Each participant wore 2 patches prepared by an unblinded pharmacist to maintain treatment blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

ITT population (more than 1 application of a study drug and pre‐dose efficacy assessment at week 6). Participants who did not reach an optimal dose by week 5 were discontinued from the study

Selection bias (e.g. titration after randomisation → exclusion of methylphenidate non‐responders or placebo responders): yes; participants who did not reach an optimal dose by week 5 were discontinued from the study

Selective reporting (reporting bias)

Unclear risk

Protocol published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

This study was funded by Shire Development Incorporated

Conflicts of interest: Several study authors have affiliations with medical companies

Wilens 2010

Methods

Blind, randomised, 4‐week, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Participants

Number of participants screened: unknown

Number of participants included: 36. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 30 (25 boys, 5 girls)

Number of withdrawals: 4

Diagnosis of ADHD: DSM‐IV (combined (53%), hyperactive‐impulsive (3%), inattentive (43%))

Age: mean 9.17 years (SD 1.84; range 6 to 12)

IQ: > 70

Methylphenidate naive: 14 (47%)

Ethnicity: Caucasian (90%), African American (not stated), Asian (3%), Hispanic (not stated), other (7%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional disorder (70%), conduct disorder (7%), major depressive disorder (3%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Male and female out‐patients

  2. 6 to 12 years of age

  3. Diagnosis of ADHD by DSM‐IV, as manifested in clinical evaluation and confirmed by structured interview

  4. Participation in structured morning routine (e.g. school, camp, other organized activities)

Exclusion criteria

  1. Mental retardation (IQ < 75)

  2. Participants with a medical condition, or treatment that will jeopardise participant safety or affect the scientific merit of the study

  3. Participants with moderate to severe dermatological atopy

  4. Participants with known structural cardiac abnormalities

  5. Organic brain disorders

  6. Seizure disorder

  7. Participants with Tourette's syndrome or a history of psychosis or bipolar disorder

  8. Participants with current comorbid psychopathology that, in the investigator's opinion, will warrant immediate treatment or will interfere with safe execution of the protocol (i.e. anxiety or major depressive disorder rated as moderate on Conners' Global Index)

  9. Participants with a history of intolerable adverse effects or non‐response to methylphenidate

  10. Pregnant or nursing females

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate (20 mg) and placebo

Mean methylphenidate dosage: not stated

Administration schedule: patch applied in morning and worn for 9 hours

Duration of each medication condition: 2 weeks

Washout before study initiation: none

Titration period: 1 week after randomisation

Treatment compliance: > 80% for all participants

Outcomes

ADHD symptoms

  1. ADHD Rating Scale: clinician‐rated at baseline and weekly for 4 weeks

  2. Conners' Global Index: parent‐rated at baseline and weekly for 4 weeks (no data)

Non‐serious adverse events

  1. Adverse events and vital signs: clinician‐rated at baseline and weekly for 4 weeks

  2. ECG: clinician‐rated at baseline and at end of study

Notes

Sample calculation: no

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; participants with a history of no response or intolerability to methylphenidate were excluded for ethical reasons

Comment from study authors

  1. Doses used are lower than US FDA‐approved dose of 30 mg. It is unclear whether results of this study represent optimal response

Key conclusion of study authors

  1. Early administration of methylphenidate transdermal system was associated with improved ADHD symptoms and before‐school functioning in children with ADHD

Email correspondence with study authors: March/April 2014. We contacted the first study author twice to ask for supplemental data but have not received a response

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Low risk

Prescriptions filled by hospital pharmacy

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No additional information from study author

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind (participant, caregiver, investigator)

Incomplete outcome data (attrition bias)
All outcomes

High risk

ITT: participants who completed 1 week of treatment; LOCF

Selection bias: yes. Excluded participants not following 1 week of treatment

Selective reporting (reporting bias)

Low risk

Protocol published. All pre‐specified outcomes of interest have been reported

Vested interest bias

High risk

This study and medication/placebo were funded by a grant through Shire Pharmaceuticals. Shire had no role in design, collection, analysis, interpretation, writing or decision to submit

Conflicts of interest: Some study authors have received research support from medical companies

Wilkison 1995

Methods

Double‐blind, placebo‐controlled, cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 2

Participants

Number of participants screened: not stated

Number of participants included: 16. Participants were randomly assigned to 1 possible drug condition order

Number of participants followed up: 16 implied (16 boys, 0 girls)

Number of withdrawals: not stated

Diagnosis of ADHD: DSM‐III‐R (subtype not stated)

Age: mean 10.2 years (range 8 to 13)

IQ: mean 112.6

Methylphenidate naive: 0%

Ethnicity: not stated

Country: USA

Setting: out‐patient clinic

Comorbidity: no

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Boys

  2. 8 to 13 years of age

  3. ADHD

  4. Recruited from an out‐patient clinic at a large metropolitan children’s hospital in the USA

  5. Conners' Hyperactivity Index score 1.5 SD above the norm on parent ratings

  6. ≥ 6 months of treatment with methylphenidate

  7. Confirmed DSM‐III‐R diagnosis

  8. History of > 6 months of methylphenidate treatment

Exclusion criteria

  1. No physical or psychiatric diagnosis other than ADHD

  2. Evidence of learning disability

  3. "Each parent and prescribing physician reported that the subject responded positively to MPH"

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate (normal dose) and placebo

Mean methylphenidate dosage: 0.030 mg/kg (range 0.08 to 1.10 mg/kg/d)

Administration schedule: not stated

Duration of each medication condition: 36 hours

Washout before study initiation: not stated

Titration period: not stated

Treatment compliance: not stated

Outcomes

General behaviour

  1. Behaviour problems ‐ Child Behavior Checklist, parent ratings. Data not reported

Notes

Sample calculation: not described

Ethics approval: not described

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no

Key conclusion of study authors

  1. Stimulant effects on child’s motivation to perform a task may compensate for deficits in other areas

Comment from review authors

  1. All participants had been treated previously with methylphenidate for longer than 6 months; each parent and physician reported good response

Email correspondence with study authors: May 2014. We obtained supplemental information from study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

From correspondence: generated by pharmacist (Magnusson 2014c [pers comm])

Allocation concealment (selection bias)

Low risk

From correspondence: Recruiter had no involvement in and was blinded to allocation sequence (Magnusson 2014c [pers comm])

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

From correspondence: Pharmacist instructed parents (via instructions on pill bottle) regarding which pills should be administered and at what time (Magnusson 2014c [pers comm])

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

See above

Incomplete outcome data (attrition bias)
All outcomes

Low risk

From correspondence: Approximately 2% of skin conductance and heart rate data were affected by participant movement and were replaced with interpolated values Physiological data were lost for 2 participants with ADHD and for 2 participants without ADHD. For 3 of the remaining 28 participants, 1 of the 4 repeated measurements of interbeat intervals was replaced with the mean of the participant's diagnostic group because his ECG recordings were inadequate (Magnusson 2014c [pers comm])

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

No reporting bias

Vested interest bias

Low risk

Research was supported by University of Utah Biomedical Sciences Research Grant and a grant from the University Research Committee

Conflicts of interest: no corporate affiliations described

Wodrich 1998

Methods

Three‐week cross‐over trial with 3 interventions

  1. Methylphenidate 5 mg, twice daily

  2. Methlphenidate 15 mg, twice daily

  3. Placebo

Phases

  1. Each drug condition lasted 7 days (Thursday through Wednesday)

  2. "No washout period was deemed necessary, given the brief half‐life and absence of carry‐over effects of MPH [methylphenidate]"

Participants

Number of participants screened: 123 ("treated in the clinic")

Number of participants included: 57 (47 boys, 10 girls)

Number of participants followed up: 57

Number of withdrawals: 66. Away on summer vacation (n = 30), teacher failed to complete all rating forms (n = 29), teacher marked 2 values indicated on a single dimension (n = 3), adverse medication side effect (n = 2), dropped out before child’s medication trial was completed (n = 2)

Diagnosis of ADHD: DSM‐III‐R

Age: mean 8.5 years (SD 2.1; range 6 to 14)

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: Caucasian "Anglo" 54 (95%), Hispanic 3 (5%)

Country: USA

Setting: out‐patient clinic

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. DSM‐III‐R Diagnosis of ADHD, confirmed by a clinical psychologist

Exclusion criteria

  1. No primary disorder "better explained the child’s presenting symptoms" (e.g. mood disorder, anxiety disorder, adjustment disorder, pervasive developmental disorder)

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders (5 mg methylphenidate or 15 mg methylphenidate) and placebo, given at 8:00 AM and 12:00 PM

Mean methylphenidate dosage: 10 mg/d, 30 mg/d and placebo

Administration schedule: 3‐week, triple‐blind, cross‐over medication trial consisting of placebo, 5 mg of methylphenidate and 15 mg of methylphenidate, twice a day. Each drug condition lasted 7 days (Thursday through Wednesday)

Duration of each medication condition: 3 weeks

Washout before study initiation: approximately 20 hours (lunchtime until "immediately before school" the next day)

Titration period: not stated

Treatment compliance: not stated

Number of withdrawals: 2. Parents dropped out of the study before their child’s medication trial was completed

Outcomes

ADHD symptoms

  1. Abbreviated Conners' Rating Form

  2. School Situations Questionnaire: "As our research concern was to locate tools helpful to school psychologists, we chose to address only the utility of using SSQ. For statistical analysis, scores from zero (no problem) to 9 (severe problem) were entered for each situation" (p 84)

General behaviour

  1. Personality Inventory for Children

Serious adverse events

  1. Side Effects Questionnaire

Non‐serious adverse events

  1. Side Effects Questionnaire

Notes

Sample calculation: not stated

Ethics approval: not stated

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes

Key conclusions of study authors

  1. School Situation Questionnaire ratings improved with methylphenidate treatment in all situations related to task performance (i.e. arriving at school; during individual seatwork, small group activities and lectures) but less so in non‐task or unstructured situations

  2. Many change scores were large enough to be clinically meaningful

  3. Use of School Situations Questionnaire by school psychologists was discussed as a means of efficiently providing contextual information not available from ADHD dimensional rating scales

Email correspondence with study authors: June 2014. We obtained supplemental information from study authors. We contacted study author to ask for missing data, but s/he was not able to supply the data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Subjects then underwent a three‐week, triple‐blinded, cross‐over medication trial consisting of placebo, 5 mg, and 15 mg of MPH dose twice a day (immediately before school and at lunchtime)" (p 83); "Order of administration was counterbalanced so that equivalent numbers of children received each sequence" (p 83)

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Subjects then underwent a three‐week, triple‐blinded, cross‐over medication trial consisting of placebo, 5 mg, and 15 mg of MPH dose twice a day (immediately before school and at lunchtime). Each drug condition lasted seven days (Thursday through Wednesday)" (p 83); "Doses were prepared and packaged for the three week trial by a licensed pharmacist who split MPH tablets and placed them with lactose into re‐sealable capsules. The placebo dose consisted of lactose‐filled capsules only. Equivalent numbers of capsules (three) were dispensed at each dosing time to maintain uniformity and disguise presence/amount of

medication, as three capsules were required to encompass the largest dose (15 mg)" (p 83)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"This procedure was followed for three weeks, with the medication code being broken the final week and determination about medication efficacy and decision to continue addressed at that time" (p 84)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No information

Vested interest bias

Unclear risk

No information

Wolraich 2001

Methods

Twenty‐eight‐day, randomised, parallel, double‐blind clinical trial with 3 arms

  1. OROS methylphenidate

  2. Immediate‐release methylphenidate

  3. Placebo

Participants

Number of participants screened: 405

Number of participants included: 312; OROS methylphenidate 94, immediate‐release methylphenidate 94, placebo 89

Number of participants followed up: OROS methylphenidate 79, immediate‐release methylphenidate 81, placebo 46

Number of withdrawals: OROS methylphenidate 15, immediate‐release methylphenidate 13, placebo 43

Diagnosis of ADHD: DSM‐IV (combined (73.4%), hyperactive‐impulsive (7.1%), inattentive (19.5%))

Age: mean 9.0 years (range 6 to 12)

IQ: > 70

Sex: 233 boys, 49 girls

Methylphenidate naive: 20.2%

Ethnicity: Caucasian (84.4%), African American (7.4%), Asian (0.4%), Hispanic (3.5%), other (4.3%)

Country: USA

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (41.8%), conduct disorder (11.3%), tic disorder (5.3%), anxiety disorder (1.4%), depression (0.7%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. 6 to 12 years of age

  2. Clinical diagnosis of any subtype of ADHD ‐ had to be confirmed by the Diagnostic Interview Schedule for Children (Version 4), administrated by a trained interviewer

  3. Patients who were taking methylphenidate or had taken it in the past had to have been on a total daily dose of methylphenidate (immediate‐release or a combination of immediate‐release/extended‐release) of ≥ 10 mg but ≤ 60 mg

  4. Patients had to agree to take the supplied study drug as the only medication for ADHD during the 4‐week study period

  5. IQ > 70

Exclusion criteria

  1. Acute or serious chronic disease

  2. Hypersensitivity to methylphenidate

  3. Significant adverse experiences from methylphenidate

  4. Taking a medication that would interfere with safe administration of methylphenidate

  5. Glaucoma, Tourette’s syndrome, ongoing seizure disorder or psychotic disorder

  6. Girls who had reached menarche

During the course of the study, participants were allowed to receive behavioural interventions as long as the interventions had been initiated before the start of the study and did not change during the study. New behavioural therapy was not allowed during the course of the study

Interventions

Average total daily dose: immediate‐release methylphenidate 29.5 mg per day (0.90.4 mg/kg/d), OROS methylphenidate 34.3 mg per day (1.1 to 0.5 mg/kg/d)

Administration schedule: 3 times a day

Duration of intervention: 4 weeks

Titration period: 4‐week titration period before randomisation (open‐label) for study participants who had not received methylphenidate for ADHD from their own practitioner in the 4 weeks before study entry. Participants who had taken methylphenidate during the 4 weeks before study entry were assigned to a dose level based on their pre‐study therapeutic dose and regimen

Treatment compliance: not stated

Outcomes

ADHD symptoms

  1. IOWA Conners' Rating Scale: teacher‐ and parent‐rated on day 27

  2. Swanson, Nolan and Pelham Scale, Fourth Edition: teacher‐ and parent‐rated, week 4

Quality of life

  1. Child Global Assessment Scale

Notes

Sample size calculation: yes

Ethics approval: yes

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: yes; 111 who had not received methylphenidate before the study initially were enrolled into a dose‐titration study. One of these did not enrol in the randomised study because the 54‐mg dose was found to be ineffective

Key conclusion of study authors

  1. Results of this study show that OROS methylphenidate administered once a day and immediate‐release methylphenidate administered 3 times a day were significantly better than placebo and were not significantly different from each other for the primary efficacy measure, teacher IOWA Conners' Rating Scale, Inattention‐Impulsivity‐Overactivity subscale score, which evaluated attention and behaviour at school. Furthermore, significant improvement in attention and behaviour was seen in the first week for participants who were taking OROS methylphenidate qd or immediate‐release methylphenidate t.i.d. compared with placebo, and this improvement was maintained throughout the 4 weeks of the study. These results were consistent across settings (home and school), raters (parents, teachers, clinical investigators) and measures (IOWA Conners' Rating Scale; Swanson, Nolan and Pelham, Fourth Edition; Peer Interaction; Global Assessments; Parent Satisfaction) and were statistically significant

Comments from review authors in July 2013:corresponded with first study author, Wolraich, who answered all of our questions (Krogh 2013c [pers comm])

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Within each dose level, participants were randomly assigned equally to OROS methylphenidate qd, immediate‐release methylphenidate (overencapsulated Ritalin) t.i.d. or placebo in a 3‐group parallel design. Stratified randomisation was conducted centrally at ALZA Corporation

Allocation concealment (selection bias)

Low risk

Double‐dummy

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind; double‐dummy

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind; double‐dummy

Incomplete outcome data (attrition bias)
All outcomes

Low risk

LOCF

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Unclear risk

Not able to get study protocol

Vested interest bias

High risk

Funded by ALZA Corporation (medical company)

Conflicts of interest: Study authors are part of the Concerta Study Group

Zeiner 1999

Methods

Cross‐over trial with 2 interventions

  1. Methylphenidate

  2. Placebo

Phases: 7 weeks (3 weeks of medication, 1 week washout, 3 weeks of placebo)

Zeiner 1995 (in Zeiner 1999): extended treatment (mean duration 634 ± 130 days)

Participants

Number of participants screened: 46

Number of participants included: 38. Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 36 (36 boys, 0 girls)

Number of withdrawals: 2

Diagnosis of ADHD: DSM‐III‐R or DSM‐IV (combined type (> 75%), hyperactive‐impulsive and inattentive types not stated)

Age: mean 8,7 years (range 7 to 11)

IQ: mean 102 (range 79 to 139)

Methylphenidate naive: 100%

Ethnicity: not stated

Country: Norway

Setting: out‐patient clinic

Comorbidity: oppositional defiant disorder (23; 64%)

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Male

  2. Between 7 and 12 years of age

  3. Fulfilled diagnostic criteria for ADHD

  4. IQ ≥ 70

Exclusion criteria

  1. Pervasive developmental disorder

  2. Psychosis or mood disorder

  3. Any acute or chronic medical or neurological disease

  4. Used stimulants or any other psychotropic drug

Interventions

Participants were randomly assigned to 1 of 2 possible drug condition orders of methylphenidate (0.5 mg/kg) and placebo. Thirty‐one children received a morning dose of 10 mg or 15 mg (and 5 young boys received 7.5 mg)

Mean methylphenidate dosage: not stated; 0.55 mg/kg to 0.56 mg/kg daily across the entire duration of the extended treatment period (Zeiner 1995 in Zeiner 1999)

Administration schedule: Capsules were given in 2 doses (at 8 AM and 11.30 AM)

Duration of each medication condition: 3 weeks

Washout before study initiation: not relevant

Methylphenidate naive: all

Medication‐free period between interventions: 1 week

Titration period: none

Treatment compliance: no information

Outcomes

ADHD symptoms

  1. Parent Account of Childhood Symptoms, at home

  2. Conners’ Teacher Rating, in classroom

Assessments of the child were made during the last week of each trial period

Non‐serious adverse events

  1. Height, weight, heart rate, systolic blood pressure, diastolic blood pressure

Notes

Sample calculation: no
Ethics approval: no information

Comment from study authors

  1. This study has obvious limitations. Sample size was limited, and heterogeneity was noted with regard to behavioural characteristics and test performance. This heterogeneity may conceal associations found in subgroups of children with ADHD. Boys between 7 and 11 years of age were chosen. They represent the majority of children admitted with hyperactivity and inattention, but boys of other age groups may show a different clinical picture

Key conclusions of study authors

  1. Response to methylphenidate was examined in 36 boys, 7 to 11 years of age, with ADHD in a double‐blind, placebo‐controlled trial of cross‐over design

  2. Hyperactivity and conduct problems were significantly reduced during methylphenidate treatment

  3. Stimulant medication was associated with improvement on tests of sustained attention, working memory and motor steadiness

  4. When individual changes were studied, it was found that 83% showed significant improvement in their hyperactivity at home or at school, and for 60%, levels of hyperactive behaviour were within the normal range

  5. High levels of hyperactivity at school at a relatively low age was a significant predictor of normalisation of hyperactivity in ≥ 1 setting. However, these predictors could classify correctly only 71% of children

  6. In clinical practice, a trial with stimulants is indicated for children with ADHD who show symptoms that are sufficiently severe to cause impairment at home and at school

Comments from review authors

  1. Generation of allocation sequence is unclear

  2. No sample size calculation was performed

  3. Number of screened patients is unclear

  4. In Zeiner 1995 (in Zeiner 1999), this was written: No sociodemographic information

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: no
Email correspondence with study authors: May to June 2014. We emailed Dr. Zeiner on 05 May 2014 and 02 June 2014 but never received a reply

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Children were randomly allocated to receive methylphenidate first or placebo first

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"A randomized crossover, double‐blind design with methylphenidate and placebo". Placebo and methylphenidate capsules that were used were identical, to ensure blindness to the drug condition

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"All raters were blind to the drug condition"

Incomplete outcome data (attrition bias)
All outcomes

High risk

"The report from a teacher during one of the periods was missing for one child. Owing to technical problems or unwillingness to participate data were missing on some of the test measures [which measured side effects] for a few children" (Zeiner 1995 in Zeiner 1999)

Selection bias: yes; only responders from the 7‐week trial were included in the extended‐treatment methylphenidate group

Selective reporting (reporting bias)

Low risk

No selective outcome reporting

Vested interest bias

Low risk

This research was supported by the Norwegian Medical Research Council, the Norwegian Public Health Association and the Legacy of Haldis and Josef Andresen

Conflicts of interest: not declared

Zeni 2009

Methods

Randomised, cross‐over trial with 2 interventions

  1. Methylphenidate and aripiprazole

  2. Placebo and aripiprazole

Phases: 2; randomisation, cross‐over

Participants

Number of participants screened: 30

Number of participants included: 16 (9 boys, 7 girls). Participants were randomly assigned to 1 of 2 possible drug condition orders

Number of participants followed up: 14

Number of withdrawals: 1

Diagnosis of ADHD: DSM‐IV (inattentive (7.1%), hyperactive (14.3%), combined (78.6%), out of n = 14)

Age: mean 10.71 years (SD 1.86; range 8 to 17)

IQ: > 70

Methylphenidate‐naive: not stated

Ethnicity: European‐Brazilian (71.4%), other (28.6%)

Country: Brazil

Setting: out‐patient clinic

Co‐morbidity: bipolar disorder (71.4%), borderline personality disorder (28.6%), anxiety disorder (57.1%), conduct disorder (57.1%), oppositional defiant disorder (78.6%), psychosis (50%), out of n = 14

Co‐medication: aripiprazole (100%)

Sociodemographics: divorced parents (57.1%); socioeconomic level A + B + C: 92.9%; D + E: 7.1%

Inclusion criteria

  1. 8 to 17 years of age

  2. Diagnosis of borderline personality disorder, Axis I or II, co‐morbid with ADHD (DSM‐IV)

  3. ADHD symptom onset preceding mood symptoms

  4. ≥ 30% improvement in mood symptoms in previous trial of aripiprazole

  5. Residual attention hyperactivity and opposition symptoms defined as score > 1.5 on the Swanson, Nolan and Pelham Scale, Fourth Edition

Exclusion criteria

  1. IQ < 70

  2. Use of any medication other than aripiprazole 10 weeks before the study

  3. Diagnoses of pervasive developmental disorder

  4. Schizophrenia

  5. Substance abuse or dependence

  6. Severe suicide/homicide risk counterindicating out‐patient treatment

  7. History of hypersensitivity to aripiprazole or methylphenidate

  8. Any other acute or chronic disease that may interfere with the study

  9. Pregnancy

Interventions

Participants were randomly assigned to different possible drug condition orders of methylphenidate (0.3 mg/kg for the first week, 0.7 mg/kg for the second week) and placebo, both alongside aripiprazole

Mean methylphenidate dosage: 15 mg in the first week (10 mg in the morning and 5 mg in the afternoon), 35 mg in the second week (20 mg in the morning and 15 mg in the afternoon)

Administration schedule: b.i.d. (morning and afternoon)

Duration of each medication condition: 2 weeks

Washout before study initiation: none (but recruited from 6‐week aripiprazole study)

Medication‐free period between interventions: not stated

Titration period: none

Treatment compliance: self report, mother’s report, pill counting in returned blister packs

Outcomes

ADHD symptoms

  1. Swanson, Nolan and Pelham Scale, Fourth Edition (Brazilian Version): rated weekly by parents

General behaviour

  1. Oppositional index of the Swanson, Nolan and Pelham Scale, Fourth Edition (Brazilian Version): rated weekly by parents

Non‐serious adverse events

  1. Serious Adverse Event Rating Scale (SAERS)

  2. Open question

Notes

Sample calculation: no

Ethics approval: yes

Comments from study authors

  1. The small sample size may not have allowed us to detect significant differences between placebo and methylphenidate

  2. The short‐term duration of this trial may be an important limitation for the observation of significant differences in adverse events between therapeutic agents

Key conclusions of study authors

  1. Methylphenidate was not more effective than placebo in reducing attention or hyperactivity symptoms in this short‐term trial

  2. Secondary finding suggests that depressive symptoms can be ameliorated by the addition of methylphenidate to aripiprazole

  3. Although methylphenidate was found to not de‐stabilise borderline personality disorder, its use should be considered with caution because 1 participant presented a severe mixed episode needing hospitalisation during methylphenidate treatment

Exclusion of methylphenidate non‐responders/children who have previously experienced adverse events while taking methylphenidate before randomisation: Only participants who reported improved borderline personality disorder in the previous aripiprazole trial were included. However, participants who reported that ADHD improved too much (as indicated by a score < 1.5 on the Swanson, Nolan, and Pelham Scale, Fourth Edition) in the previous aripiprazole trial were excluded (see 'Exclusion criteria')

Any withdrawals due to adverse events: 1; exacerbation of borderline personality disorder and ADHD requiring hospitalisation

Email correspondence with study authors: May 2014. We obtained supplemental information regarding data. Study authors provided us with data from the first period

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An independent third party randomly assigned participants to groups A and B

Allocation concealment (selection bias)

Low risk

A pharmacist packaged methylphenidate and matching placebo in capsules, so they could not be differentiated by shape, colour, smell, weight or taste

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A pharmacist packaged methylphenidate and matching placebo in capsules, so they could not be differentiated by shape, colour, smell, weight or taste

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

After all 4 assessments were completed, study blinding was broken

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analyses of primary and secondary outcome measures were performed using a mixed‐effects model (MEM) approach, which provides a flexible framework for analysis of repeated measures, while accounting for missing data (i.e. lost to follow‐up)

Selection bias (e.g. titration after randomisation → exclusion): no

Selective reporting (reporting bias)

Low risk

Outcomes reported according to protocol

Vested interest bias

Unclear risk

This work was fully supported by research grants from Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq, Brazil) (Grant 471761=03‐6) and Hospital de Clinicas de Porto Alegre (GPPG 03‐325). Aripiprazole was provided by Bristol‐Myers Squibb without restriction

Conflicts of interest: stated, "this is an independent investigator trial". However some study authors have affiliations with medical companies

ADD: attention deficit disorder.
ADD‐H: attention deficit disorder with hyperactivity.
ADHD: attention deficit hyperactivity disorder.
b.i.d.: twice a day.
CNS: central nervous system.
DSM‐III‐R: Diagnostic and Statistical Manual of Mental Disorders, Third Edition, Revised.
DSM‐IV: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition.
DSM‐IV‐TR: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision.
ECG: electrocardiogram.
EEG: electroencephalogram.
EKG: electrocardiogram.
FDA: Food and Drug Administration.
ITT: intention‐to‐treat.
LLC: limited liability company.
LOCF: last observation carried forward.
LTFU: loss to follow‐up.
MedDRA: Medical Dictionary for Regulatory Activities.
MPH: methylphenidate.
MPT: methylphenidate trial.
MTA: Multimodal Treatment Study of Children With ADHD.
MTS: methylphenidate transdermal system.
NNTB: number needed to treat for an additional beneficial outcome.
OROS: Osmotic Release Oral System.
PTS: placebo transdermal system.
q.a.m.: every morning.
qd: once a day.
RCT: randomised controlled trial.
SD: standard deviation.
SODAS: spheroidal oral drug absorption system.
t.i.d.: three times a day.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

An 2013

N = No information

Likely no relevant outcome for our review

Outcomes reported: Resting state brain function

Anderson 2002

N = 10 boys

Likely no relevant outcome for our review

Outcomes reported: Functional magnetic resonance relaxometry

Barkley 1988a

N = 28

Likely no relevant outcome for our review

Outcomes reported: Home Situations Questionnaire; Parenting Stress Index; Beck Depression Inventory

Barkley 1997

N = No information

Likely no relevant outcome for our review

Outcomes reported: "2 questionnaires"; "Electronic apparatus"

Bart 2013

N = 24

Likely no relevant outcome for our review

Outcomes reported: Movement Assessment Battery for Children – Second edition; Online Continuous Performance Test

Bedard 2002

N = 31

Likely no relevant outcome for our review

Outcomes reported: Response interference; Stroop Naming Speed

Bedard 2003

N = 59

Likely no relevant outcome for our review

Outcomes reported: Parent Interview for Child Symptoms; Teacher Telephone Interview–IV; Selective Stop‐Signal Task

Bedard 2004

N = 26

Likely no relevant outcome for our review

Outcomes reported: Parent Interview for Child Symptoms; Teacher Telephone Interview–IV; Reading subtest/Wide Range Achievement Test ‐ 3; Word Attack and Word Identification subtests of the Woodcock Reading Mastery; Test‐Revised Cambridge Neuropsychological Testing Automated Battery

Bedard 2007

N = 40

Likely no relevant outcome for our review

Outcomes reported: Working memory; Test of Word and Language Efficiency; Wide Range Achievement Test ‐ 3; Woodcock‐Johnson Tests of Achievement; Cambridge Neuropsychological Test Automated Battery (CANTAB) Spatial Span

Beery 1994

N = 70

Likely no relevant outcome for our review

Outcomes reported: Behavioural management, behavioural disinhibition

Ben‐Pazi 2006

N = 55

Likely no relevant outcome for our review

Outcomes reported: Hastening phenomena

Bental 2008

N = 25

Likely no relevant outcome for our review

Outcomes reported: Reading measures

Brown 1984b

N = 20

Likely no relevant outcome for our review

Outcomes reported: Children's Checking Task

Buhrmester 1992

N = 19

Likely no relevant outcome for our review

Outcomes reported: Prosocial behavior

Campbell 1996

N = 30

Likely no relevant outcome for our review

Outcomes reported: Reactions time on Tachistoscopic Task

Carlson 1991

N = 13

Likely no relevant outcome for our review

Outcomes reported: Reaction times; cognitive tasks

Carlson 1992

N = 24

Likely no relevant outcome for our review

Outcomes reported: On task and disruptive behaviour; academic work completion and accuracy

Cox 2004b

N = 6

Likely no relevant outcome for our review

Outcomes reported: Driving performance ‐ measured by computer

Dawson 1998

N = 13

Likely no relevant outcome for our review

Outcomes reported: Mirsky's proposed factors of attention: sustained attention, focus/execute, encode, and stability of attention

De Sonneville 1991

N = 17

Likely no relevant outcome for our review

Outcomes reported: Specific attention function: sustained attention, information processing, response organization

DeVito 2008

N = 21

Likely no relevant outcome for our review

Outcomes reported: Cambridge Gamble Task; measures of response inhibition and reflection‐impulsivity on the Information Sampling Task

Evans 1986

N = No information

Likely no relevant outcome for our review

Outcomes reported: Verbal memory and learning

Fox 2014

N = 14

Likely no relevant outcome for our review

Outcomes reported: Memory tasks

Francis 2001

N = 50

Likely no relevant outcome for our review

Outcomes reported: Story telling and story grammar analysis

Gan 1982

N = 20

Likely no relevant outcome for our review

Outcomes reported: Performance on paired‐associate learning task

Granger 1996

N = 26

Likely no relevant outcome for our review

Outcomes reported: Social behaviour

Grizenko 2010

N = 371

Likely no relevant outcome for our review

Outcomes reported: Academic behaviour; sustained attention; impulse inhibition control

Günther 2010

N = 54

Likely no relevant outcome for our review

Outcomes reported: Sustained attention measured by computer attention tests

Halliday 1983

N = No information

Likely no relevant outcome for our review

Outcomes reported: Event‐related potentials

Hanisch 2004

N = 45

Likely no relevant outcome for our review

Outcomes reported: Computerized attention tasks

Hazel‐Fernandez 2006

N = 19

Likely no relevant outcome for our review

Outcomes reported: Paired Associates Learning Task; Tower of Hanoi

Hinshaw 1989

N = 25

Likely no relevant outcome for our review

Outcomes reported: Prosocial behavior

Hinshaw 1993

N = 22

Likely no relevant outcome for our review

Outcomes reported: Antisocial behaviour

Humphries 1979

N = 24

Likely no relevant outcome for our review

Outcomes reported: Maze‐tracking performance

King 2009a

N = 75

Likely no relevant outcome for our review

Outcomes reported: Social information processing

King 2009b

N = 32

Likely no relevant outcome for our review

Outcomes reported: Laboratory provocation task: measuring hostile, instrumental, reactive, and proactive aggression

Lange 2007

N = 58

Likely no relevant outcome for our review

Outcomes reported: Reaction time; alertness; vigilance; divided attention

Leitner 2007b

N = 16

Likely no relevant outcome for our review

Outcomes reported: Gait; stride to stride variability, memory, visual‐spatial, verbal, and attention domains

Malone 1988

N = 31

Likely no relevant outcome for our review

Outcomes reported: Word processing; reaction time; cognitive decision task

Malone 1993

N = 26

Likely no relevant outcome for our review

Outcomes reported: Impulsive responding

Malone 1994

N = 17

Likely no relevant outcome for our review

Outcomes reported: Right hemisphere dysfunction

Martin 2007

N = 24

Likely no relevant outcome for our review

Outcomes reported: Depression; addiction rate; Continuous Performance Task; heart rate and blood pressure

Mehta 2004

N = 14

Likely no relevant outcome for our review

Outcomes reported: Working memory

Milich 1989

N = 26

Likely no relevant outcome for our review

Outcomes reported: Continuous Performance Task

Milich 1991

N = 21

Likely no relevant outcome for our review

Outcomes reported: Task persistence

Novak 1995

N = 16

Likely no relevant outcome for our review

Outcomes reported: Reaction time; visuospatial attention

O'Toole 1997

N = 23

Likely no relevant outcome for our review

Outcomes reported: Non‐verbal learning

Peeke 1984

N = 9

Likely no relevant outcome for our review

Outcomes reported: Verbal information processing

Pelham 1985

N = 29

Likely no relevant outcome for our review

Outcomes reported: Classroom academic and social behaviour

Pelham 1990b

N = 17

Likely no relevant outcome for our review

Outcomes reported: Attention during baseball game; on task behaviour; ability to answer question about the status of the game

Pelham 1992

N = 28

Likely no relevant outcome for our review

Outcomes reported: Self‐reported attribution and evaluation of behaviour

Pelham 1997

N = 60

Likely no relevant outcome for our review

Outcomes reported: Performance, self‐evaluation, persistence, and attributions on cognitive task

Pelham 2001b

N = 68

Likely no relevant outcome for our review

Outcomes reported: A large number of different measures of behaviour

Rapport 1995

N = 45

Likely no relevant outcome for our review

Outcomes reported: Paired Associates Learning Task

Richardson 1988

N = 42

Likely no relevant outcome for our review

Outcomes reported: Reading achievement

Rubia 2003

N = 13

Likely no relevant outcome for our review

Outcomes reported: Motor timing

Sangal 2006

N = 58

Likely no relevant outcome for our review

Outcomes reported: Auditory amplitude

Sengupta 2008

N = 188

Likely no relevant outcome for our review

Outcomes reported: Task oriented behaviour

Silk 2012

N = No information

Likely no relevant outcome for our review

Outcomes reported: Neural substrates

Smith 2013

N = 20

Likely no relevant outcome for our review

Outcomes reported: Functional magnetic resonance imaging

Solanto 1986

N = 12

Likely no relevant outcome for our review

Outcomes reported: Attention during play measured by locomotor activity; Children's Checking Test; and fine motor control

Solanto 1997

N = 22

Likely no relevant outcome for our review

Outcomes reported: Continuous Performance Test

Srinivas 1992

N = 9

Likely no relevant outcome for our review

Outcomes reported: Sustained attention measured by computer attention tests

Strand 2012

N = 17

Likely no relevant outcome for our review

Outcomes reported: Working memory

Stray 2009

N = 25

Likely no relevant outcome for our review

Outcomes reported: Motor functions

Swanson 1993

N = 26

Likely no relevant outcome for our review

Outcomes reported: Impulsive responding

Szobot 2003

N = 36

Likely no relevant outcome for our review

Outcomes reported: Cerebral blood flow

Tannock 2000

N = 47

Likely no relevant outcome for our review

Outcomes reported: Naming speed and academic measures

Teicher 2003

N = 19

Likely no relevant outcome for our review

Outcomes reported: Computerized Contiuous Performance Test and functional magnetic resonance imaging

Teicher 2006

N = 14

Likely no relevant outcome for our review

Outcomes reported: Rate‐dependent behavioural effects

Teicher 2007

N = 11

Likely no relevant outcome for our review

Outcomes reported: McLean Motion Attention Test

Tillery 2000

N = 32

Likely no relevant outcome for our review

Outcomes reported: Auditory performance

Trommer 1991

N = 44

Likely no relevant outcome for our review

Outcomes reported: Go‐No‐GO performance

Tsang 2012

N = 134

Likely no relevant outcome for our review

Outcomes reported: Cognitive tasks; emotional functions

Tucha 2006

N = 58

Likely no relevant outcome for our review

Outcomes reported: Reaction time tasks

Verbaten 1994

N = 12

Likely no relevant outcome for our review

Outcomes reported: Continuous Performance Test

Waschbusch 2007

N = 61

Likely no relevant outcome for our review

Outcomes reported: Academic oriented tasks

Whalen 1987

N = 24

Likely no relevant outcome for our review

Outcomes reported: Social behaviour

Characteristics of studies awaiting assessment [ordered by study ID]

Drtilkova 1997

Methods

RCT

14 days

Studied all aspects of methylphenidate, amphetaminil, mesocarb and placebo in a group of 118 children

Participants

Interventions

Outcomes

Notes

Article is written in Czech. We have not yet been able to have the article translated

Short 2004

Methods

The purpose of this trial was to examine the efficacy of psychostimulant medication in a naturalistic sample of pre‐school children. Researchers examined the benefits and side effects of methylphenidate and mixed amphetamine salts (Adderall)

Participants

Interventions

Outcomes

Notes

RCT: randomised controlled trial.

Characteristics of ongoing studies [ordered by study ID]

Bottelier 2014

Trial name or title

Methods

16‐week, placebo‐controlled RCT with methylphenidate in 100 medication‐naive participants with ADHD

Participants

Interventions

Outcomes

Starting date

Contact information

Notes

This article describes methods and design. We are awaiting additional publications

Gendron 2012

Trial name or title

Methods

Placebo‐controlled trial of methylphenidate involving 30 medication‐naive children with ADHD

Participants

Interventions

Outcomes

Starting date

Contact information

Notes

Correspondence with trial authors in August 2013. Trial will be submitted for review over the next few months

NCT00483106

Trial name or title

Clinical and pharmacogenetic study of attention deficit hyperactivity disorder (ADHD)

Methods

RCT

Participants

Children with ADHD, aged 6 to 12 years

Interventions

Ritalin and placebo

Outcomes

Conners' Global Index (CGI)

Starting date

Contact information

Ridha Jobber, McGill University

Notes

NCT02039908

Trial name or title

Examining tolerance to CNS stimulants in ADHD

Methods

RCT

Participants

Children with ADHD, aged 6 to 12 years

Interventions

Methylphenidate

Outcomes

Starting date

Contact information

Notes

Nurmi 2013

Trial name or title

Pharmacogenetics of growth effects complicating ADHD treatment (Neuropsychopharmacology)

Methods

8‐week RCT: double‐blind, randomised, placebo‐controlled trial with 4 interventions and 16‐month extension

  1. Guanfacine

  2. Dexmethylphenidate

  3. Combination guanfacine + dexmethylphenidate (16‐month)

  4. Placebo

Phases: 8‐week, double‐blind; 16 months open

Participants

Number of participants screened: not stated

Number of participants included: 209 (sex not stated). Participants were randomly assigned to 1 of 4 possible drug condition orders

Number of participants followed up: 99

Number of withdrawals: not stated

Diagnosis of ADHD: not stated

Age: not stated

IQ: not stated

Methylphenidate naive: not stated

Ethnicity: not stated

Country: USA

Comorbidity: not stated

Comedication: not stated

Sociodemographics: not stated

Inclusion criteria

  1. Not stated

Exclusion criteria

  1. Not stated

Interventions

Participants were randomly assigned to 1 of 3 possible drug condition orders of dexmethylphenidate and placebo

Administration schedule: not stated

Duration of each medication condition: not stated

Washout before trial initiation: not stated

Medication‐free period between interventions: not stated

Titration period: not stated

Treatment compliance: not stated

Outcomes

Outcomes: adverse events

Both dexmethylphenidate monotherapy (Z‐score decrease of ‐0.12 for height and ‐0.84 for BMI) and combination treatment (Z‐score decrease of ‐0.19 for height and ‐0.83 for BMI) were associated with slowing of growth. In the methylphenidate‐only condition, DRD3 rs3732790 minor allele homozygotes showed a 2.0 greater Z‐score weight loss than was seen with common allele carriers (P value = 1.09 × 10–8)

Starting date

2012 (date on which conference abstract published). Study end date unknown

Contact information

Erika L Nurmi: [email protected]

James T McCracken: [email protected]. Department of Psychiatry and Biobehavioral Sciences, UCLA NPI‐Semel Institute, 760 Westwood Plaza, Los Angeles, CA 90024‐1759, USA

Notes

Sample calculation: no

Ethics approval: not stated

Results of this trial have not yet been published. Trial authors expect to publish their findings within the year

ADHD: Attention deficit hyperactivity disorder.
BMI: Body mass index.
CNS: Central nervous system.
RCT: randomised controlled trial.

Data and analyses

Open in table viewer
Comparison 1. Teacher‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

Analysis 1.1

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

19

1698

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

‐0.77 [‐0.90, ‐0.64]

2 Subgroup analysis: types of scales Show forest plot

19

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

Subtotals only

Analysis 1.2

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

2.1 Conners' Teacher Rating Scale (CTRS)

8

518

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

‐0.64 [‐0.82, ‐0.47]

2.2 Abbreviated Conners' Rating Scale (ACRS) ‐ Teacher

2

105

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

‐0.75 [‐1.79, 0.29]

2.3 Conners' Abbreviated Symptom Questionnaire for Teachers (ASQ‐Teacher)

1

59

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

‐0.28 [‐0.79, 0.23]

2.4 IOWA Conners' Teacher Rating Scale (IOWA CTRS) ‐ hyperactivity

2

193

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

‐1.08 [‐1.39, ‐0.77]

2.5 Schedule for Non‐adaptive and Adaptive Personality (SNAP) ‐ Teacher

2

328

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

‐0.61 [‐0.96, ‐0.25]

2.6 Teacher ratings of attention

1

20

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

‐0.55 [‐1.45, 0.35]

2.7 Teacher ratings of impulsivity

1

20

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

0.04 [‐0.83, 0.92]

2.8 IOWA Conners' Teacher Rating Scale ‐ Inattention/Overactivity (IOWA‐I/O)

2

197

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

‐1.03 [‐1.36, ‐0.69]

2.9 Fremdbeurteilungsbogen für Hyperkinetische Störungen (FBB‐HKS)

1

85

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

‐1.06 [‐1.52, ‐0.61]

2.10 Conners’ ADHD/DSM‐IV Scales ‐ Teacher (CADS‐T)

2

254

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

‐1.05 [‐1.31, ‐0.78]

2.11 Strengths and Weaknesses of ADHD Symptoms and Normal Behaviour (SWAN) Scale

1

64

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

‐0.33 [‐0.82, 0.17]

3 Medication status: medication naive versus not medication naive Show forest plot

6

717

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

‐0.79 [‐1.08, ‐0.50]

Analysis 1.3

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 3 Medication status: medication naive versus not medication naive.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 3 Medication status: medication naive versus not medication naive.

3.1 Medication naive

4

431

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

‐0.63 [‐0.94, ‐0.31]

3.2 Not medication naive

2

286

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

‐1.06 [‐1.33, ‐0.79]

4 Subgroup analysis: duration of treatment Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

Analysis 1.4

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 4 Subgroup analysis: duration of treatment.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 4 Subgroup analysis: duration of treatment.

4.1 Short term (up to 6 months)

18

1445

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

‐0.81 [‐0.94, ‐0.68]

4.2 Long term (over 6 months)

1

253

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

‐0.47 [‐0.72, ‐0.22]

5 Subgroup analysis: dose Show forest plot

19

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

Subtotals only

Analysis 1.5

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 5 Subgroup analysis: dose.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 5 Subgroup analysis: dose.

5.1 Low dose

8

493

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

‐0.64 [‐0.82, ‐0.46]

5.2 High dose

7

688

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

‐0.81 [‐1.08, ‐0.54]

5.3 Unknown dose

6

669

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

‐0.87 [‐1.06, ‐0.68]

6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

Analysis 1.6

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

6.1 Parallel‐group trials

17

1506

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

‐0.80 [‐0.95, ‐0.65]

6.2 First‐period cross‐over trials

2

192

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

‐0.58 [‐0.87, ‐0.29]

7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

Analysis 1.7

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

7.1 Trials with cohort selection bias of all participants

7

994

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

‐0.79 [‐1.01, ‐0.57]

7.2 Trials without cohort selection bias of all participants

12

704

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

‐0.76 [‐0.93, ‐0.59]

8 ADHD symptoms, cross‐over trial (endpoint data) Show forest plot

59

5145

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

‐0.93 [‐1.06, ‐0.80]

Analysis 1.8

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trial (endpoint data).

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trial (endpoint data).

8.1 Low risk of bias

4

204

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

‐0.64 [‐0.97, ‐0.30]

8.2 High risk of bias

55

4941

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

‐0.95 [‐1.09, ‐0.82]

9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

59

6821

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

‐0.85 [‐0.96, ‐0.74]

Analysis 1.9

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

9.1 Low dose

42

3408

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

‐0.73 [‐0.89, ‐0.57]

9.2 High dose

36

3413

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

‐0.98 [‐1.13, ‐0.84]

10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

75

6344

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

‐0.91 [‐1.01, ‐0.80]

Analysis 1.10

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

10.1 All parallel‐group trials and first‐period cross‐over trials

19

1698

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

‐0.77 [‐0.90, ‐0.64]

10.2 Cross‐over trials (endpoint data)

56

4646

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

‐0.95 [‐1.09, ‐0.82]

11 All parallel‐group trials and cross‐over trials: risk of bias Show forest plot

75

6344

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

‐0.91 [‐1.01, ‐0.80]

Analysis 1.11

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 11 All parallel‐group trials and cross‐over trials: risk of bias.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 11 All parallel‐group trials and cross‐over trials: risk of bias.

11.1 Low risk of bias

4

204

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

‐0.64 [‐0.97, ‐0.30]

11.2 High risk of bias

71

6140

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

‐0.92 [‐1.03, ‐0.81]

Open in table viewer
Comparison 2. Independent assessor‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

Analysis 2.1

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

10

1907

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

‐0.64 [‐0.89, ‐0.39]

2 Subgroup analysis: types of scales Show forest plot

10

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

Subtotals only

Analysis 2.2

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

2.1 Swanson, Kotkin, Agler, M‐Glynn and Pelham (SKAMP) Scale

1

164

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

‐0.72 [‐1.04, ‐0.41]

2.2 ADHD Rating Scale (ADHD‐RS )

7

1442

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

‐0.71 [‐1.09, ‐0.32]

2.3 Swanson, Nolan and Pelham (SNAP) Scale

1

221

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

‐0.35 [‐0.61, ‐0.08]

2.4 Unknown

1

78

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

‐0.94 [‐1.41, ‐0.47]

3 Subgroup analysis: duration of treatment Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

Analysis 2.3

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

3.1 Short term (up to 6 months)

9

1686

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

‐0.68 [‐0.95, ‐0.40]

3.2 Long term (over 6 months)

1

221

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

‐0.35 [‐0.61, ‐0.08]

4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

Analysis 2.4

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

4.1 Parallel‐group trials

7

1609

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

‐0.60 [‐0.91, ‐0.28]

4.2 First‐period cross‐over trials

3

298

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

‐0.76 [‐0.99, ‐0.52]

5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

Analysis 2.5

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

5.1 Trials with cohort selection bias of all participants

4

630

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

‐0.51 [‐0.73, ‐0.29]

5.2 Trials without cohort selection bias of all participants

6

1277

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

‐0.72 [‐1.11, ‐0.33]

6 Subgroup analysis: dose Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

Analysis 2.6

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 6 Subgroup analysis: dose.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 6 Subgroup analysis: dose.

6.1 Low dose

0

0

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

0.0 [0.0, 0.0]

6.2 High dose

6

1380

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

‐0.55 [‐0.91, ‐0.20]

6.3 Unknown dose

4

527

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

‐0.80 [‐0.98, ‐0.61]

7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias Show forest plot

19

2471

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

1.00 [‐1.16, ‐0.84]

Analysis 2.7

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias.

7.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

7.2 High risk of bias

19

2471

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

1.00 [‐1.16, ‐0.84]

8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

19

3874

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

‐0.89 [‐1.02, ‐0.75]

Analysis 2.8

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

8.1 Low dose

16

2021

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

‐0.69 [‐0.82, ‐0.55]

8.2 High dose

12

1853

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

‐1.13 [‐1.31, ‐0.95]

9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

28

4215

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

‐0.83 [‐0.98, ‐0.67]

Analysis 2.9

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

9.1 All parallel‐group trials and first‐period cross‐over trials

10

1907

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

‐0.64 [‐0.89, ‐0.39]

9.2 Cross‐over trials (endpoint data)

18

2308

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

‐0.95 [‐1.13, ‐0.77]

Open in table viewer
Comparison 3. Parent‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

Analysis 3.1

Comparison 3 Parent‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

21

2187

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

‐0.66 [‐0.82, ‐0.51]

2 Subgroup analysis: types of scales Show forest plot

21

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

Subtotals only

Analysis 3.2

Comparison 3 Parent‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

2.1 Conners' Parent Rating Scale (CPRS)

7

751

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

‐0.58 [‐0.87, ‐0.30]

2.2 ADHD Rating Scale ‐ Fourth Edition (ADHD‐RS‐IV)

2

194

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

‐0.30 [‐0.58, ‐0.02]

2.3 Fremdbeurteilungsbogen für Hyperkinetische Störungen (FBB‐HKS)

1

85

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

‐0.91 [‐1.36, ‐0.46]

2.4 Conners’ ADHD/DSM‐IV Scales ‐ Parent (CADS‐P)

1

120

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

‐1.26 [‐1.65, ‐0.86]

2.5 CADS‐P Inattentive subscale

1

109

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

‐0.78 [‐1.17, ‐0.39]

2.6 CADS‐P Hyperactivity subscale

1

109

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

‐0.93 [‐1.32, ‐0.53]

2.7 Clinican's Manual for the Assesment of Disruptive Behavior Disorders Rating Scale for Parents (Barkley)

1

41

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

‐0.21 [‐0.82, 0.41]

2.8 Abbreviated Conners' Rating Scale (ACRS) ‐ Parent

2

121

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

‐0.62 [‐0.99, ‐0.25]

2.9 Swanson, Nolan, and Pelham, Fourth Edition ‐ Parent (SNAP‐IV‐Parent) Scale

4

430

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

‐0.60 [‐0.79, ‐0.40]

2.10 Strengths and Weaknesses of ADHD Symptoms and Normal Behavior (SWAN) Scale

1

86

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

‐0.43 [‐0.86, 0.00]

2.11 IOWA Conners' Rating Scale ‐ Inattention/Overactivity (IOWA‐I/O)

3

352

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

‐0.78 [‐1.35, ‐0.21]

3 Subgroup analysis: duration of treatment Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

Analysis 3.3

Comparison 3 Parent‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

3.1 Short term (up to 6 months)

20

1925

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

‐0.67 [‐0.84, ‐0.50]

3.2 Long term (over 6 months)

1

262

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

‐0.58 [‐0.82, ‐0.33]

4 Subgroup analysis: dose Show forest plot

21

2335

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

‐0.64 [‐0.79, ‐0.48]

Analysis 3.4

Comparison 3 Parent‐rated ADHD symptoms, Outcome 4 Subgroup analysis: dose.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 4 Subgroup analysis: dose.

4.1 Low dose

5

329

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

‐0.53 [1.00, ‐0.07]

4.2 High dose

10

1132

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

‐0.60 [‐0.86, ‐0.33]

4.3 Unknown dose

8

874

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

‐0.72 [‐0.92, ‐0.52]

5 Medication status: medication naive versus not medication naive Show forest plot

7

795

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

‐0.76 [‐1.05, ‐0.48]

Analysis 3.5

Comparison 3 Parent‐rated ADHD symptoms, Outcome 5 Medication status: medication naive versus not medication naive.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 5 Medication status: medication naive versus not medication naive.

5.1 Medication naive

4

492

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

‐0.69 [‐1.03, ‐0.35]

5.2 Not medication naive

3

303

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

‐0.88 [‐1.33, ‐0.42]

6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

Analysis 3.6

Comparison 3 Parent‐rated ADHD symptoms, Outcome 6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

6.1 Trials with selection bias of all participants

10

1559

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

‐0.68 [‐0.86, ‐0.51]

6.2 Trials without cohort selection bias of all participants

11

628

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

‐0.62 [‐0.90, ‐0.33]

7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

Analysis 3.7

Comparison 3 Parent‐rated ADHD symptoms, Outcome 7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

7.1 Parallel‐group trials

19

2094

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

‐0.66 [‐0.83, ‐0.50]

7.2 First‐period cross‐over trials

2

93

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

‐0.65 [‐1.07, ‐0.23]

8 ADHD symptoms, cross‐over trials (endpoint data) Show forest plot

41

3734

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

‐0.78 [‐0.90, ‐0.67]

Analysis 3.8

Comparison 3 Parent‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data).

Comparison 3 Parent‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data).

8.1 Low risk of bias

4

204

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

‐0.54 [‐0.96, ‐0.13]

8.2 High risk of bias

37

3530

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

‐0.81 [‐0.92, ‐0.69]

9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

41

4918

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

‐0.69 [‐0.79, ‐0.58]

Analysis 3.9

Comparison 3 Parent‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

9.1 Low dose

26

2272

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

‐0.65 [‐0.82, ‐0.48]

9.2 High dose

28

2646

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

‐0.72 [‐0.84, ‐0.60]

10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

59

5861

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

‐0.74 [‐0.83, ‐0.65]

Analysis 3.10

Comparison 3 Parent‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 3 Parent‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

10.1 All parallel‐group trials and first‐period cross‐over trials

21

2215

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

‐0.66 [‐0.82, ‐0.51]

10.2 Cross‐over trials (endpoint data)

39

3646

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

‐0.79 [‐0.90, ‐0.67]

Open in table viewer
Comparison 4. Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparision of raters Show forest plot

31

5697

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

‐0.69 [‐0.79, ‐0.59]

Analysis 4.1

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparision of raters.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparision of raters.

1.1 Teacher‐rated

19

1689

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

‐0.78 [‐0.93, ‐0.63]

1.2 Independent assessor‐rated

9

1829

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

‐0.61 [‐0.87, ‐0.35]

1.3 Parent‐rated

21

2179

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

‐0.65 [‐0.81, ‐0.50]

2 Age Show forest plot

6

1039

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

‐0.44 [‐0.74, ‐0.14]

Analysis 4.2

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Age.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Age.

2.1 2 to 6 years

1

64

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

‐0.33 [‐0.82, 0.17]

2.2 7 to 11 years

2

278

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

‐0.59 [‐1.03, ‐0.15]

2.3 12 to 18 years

3

697

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

‐0.38 [‐0.88, 0.12]

3 Comorbidity versus no comorbidity Show forest plot

20

2310

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

‐0.72 [‐0.91, ‐0.53]

Analysis 4.3

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Comorbidity versus no comorbidity.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Comorbidity versus no comorbidity.

3.1 ADHD with comorbidity

18

1981

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

‐0.72 [‐0.94, ‐0.51]

3.2 ADHD without comorbidity

2

329

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

‐0.69 [‐0.92, ‐0.46]

4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated) Show forest plot

2

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

Subtotals only

Analysis 4.4

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated).

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated).

4.1 Combined ADHD

2

559

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

0.65 [‐1.30, 2.60]

4.2 Inattentive ADHD

1

204

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

‐1.31 [‐1.61, ‐1.01]

5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated) Show forest plot

4

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

Subtotals only

Analysis 4.5

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated).

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated).

5.1 First‐period data

4

372

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

‐0.64 [‐0.85, ‐0.44]

5.2 Endpoint data

4

372

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

‐0.91 [‐1.18, ‐0.65]

Open in table viewer
Comparison 5. Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of serious adverse events (SAE) Show forest plot

9

1532

Risk Ratio (IV, Random, 95% CI)

0.98 [0.44, 2.22]

Analysis 5.1

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Number of serious adverse events (SAE).

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Number of serious adverse events (SAE).

2 Nervous system Show forest plot

6

2280

Risk Ratio (IV, Random, 95% CI)

0.87 [0.30, 2.53]

Analysis 5.2

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Nervous system.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Nervous system.

2.1 Aggression

1

303

Risk Ratio (IV, Random, 95% CI)

0.50 [0.05, 5.49]

2.2 Concussion

1

303

Risk Ratio (IV, Random, 95% CI)

0.34 [0.01, 8.17]

2.3 Loss of consciousness

1

221

Risk Ratio (IV, Random, 95% CI)

0.33 [0.01, 8.02]

2.4 Psychosis

4

712

Risk Ratio (IV, Random, 95% CI)

1.78 [0.19, 16.96]

2.5 Syncope

3

741

Risk Ratio (IV, Random, 95% CI)

1.39 [0.23, 8.47]

3 Digestive system: gastrointestinal disorders Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 5.3

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Digestive system: gastrointestinal disorders.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Digestive system: gastrointestinal disorders.

4 Urinary system: kidney infection Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 5.4

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Urinary system: kidney infection.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Urinary system: kidney infection.

5 Circulatory and respiratory systems: asthma Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 5.5

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems: asthma.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems: asthma.

6 Immune system: cyst rupture Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 5.6

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Immune system: cyst rupture.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Immune system: cyst rupture.

7 Other: drug toxicity Show forest plot

1

303

Risk Ratio (IV, Random, 95% CI)

0.34 [0.01, 8.17]

Analysis 5.7

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Other: drug toxicity.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Other: drug toxicity.

Open in table viewer
Comparison 6. Number of serious adverse events: cross‐over trials (endpoint data)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of serious adverse events (SAE) Show forest plot

8

1721

Risk Ratio (IV, Random, 95% CI)

1.62 [0.34, 7.71]

Analysis 6.1

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 1 Number of serious adverse events (SAE).

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 1 Number of serious adverse events (SAE).

2 Hallucinations/psychosis Show forest plot

4

187

Risk Ratio (IV, Random, 95% CI)

1.10 [0.18, 6.72]

Analysis 6.2

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 2 Hallucinations/psychosis.

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 2 Hallucinations/psychosis.

Open in table viewer
Comparison 7. Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of non‐serious adverse events Show forest plot

21

3132

Risk Ratio (IV, Random, 95% CI)

1.29 [1.10, 1.51]

Analysis 7.1

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Total number of non‐serious adverse events.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Total number of non‐serious adverse events.

2 Subgroup analysis: total number of non‐serious adverse events according to dose Show forest plot

21

3135

Risk Ratio (IV, Random, 95% CI)

1.28 [1.11, 1.49]

Analysis 7.2

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

2.1 Low dose

2

151

Risk Ratio (IV, Random, 95% CI)

1.09 [0.82, 1.46]

2.2 High dose

10

1761

Risk Ratio (IV, Random, 95% CI)

1.22 [1.10, 1.35]

2.3 Unknown dose

10

1223

Risk Ratio (IV, Random, 95% CI)

1.37 [1.01, 1.87]

3 Nervous system Show forest plot

21

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 7.3

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Nervous system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Nervous system.

3.1 Affective

4

390

Risk Ratio (Random, 95% CI)

2.39 [0.48, 11.96]

3.2 Aggression

2

417

Risk Ratio (Random, 95% CI)

1.16 [0.17, 7.80]

3.3 Apathy

1

59

Risk Ratio (Random, 95% CI)

0.80 [0.19, 3.33]

3.4 Confusion

2

548

Risk Ratio (Random, 95% CI)

1.01 [0.22, 4.73]

3.5 Depression

1

59

Risk Ratio (Random, 95% CI)

0.83 [0.22, 3.10]

3.6 Dizziness

3

683

Risk Ratio (Random, 95% CI)

2.50 [0.70, 8.99]

3.7 Drowsiness

4

811

Risk Ratio (Random, 95% CI)

1.27 [0.82, 1.98]

3.8 Emotional lability

1

132

Risk Ratio (Random, 95% CI)

2.41 [0.27, 21.32]

3.9 Fatigue

7

858

Risk Ratio (Random, 95% CI)

0.76 [0.36, 1.63]

3.10 Headache

17

2724

Risk Ratio (Random, 95% CI)

1.22 [0.90, 1.64]

3.11 Insomnia

3

349

Risk Ratio (Random, 95% CI)

1.31 [0.35, 4.93]

3.12 Irritability

11

1721

Risk Ratio (Random, 95% CI)

1.11 [0.77, 1.60]

3.13 Nervousness

2

362

Risk Ratio (Random, 95% CI)

2.52 [0.82, 7.76]

3.14 Pain

1

132

Risk Ratio (Random, 95% CI)

1.91 [0.21, 17.60]

3.15 Picking at skin or fingers, nail biting, lip or cheek chewing

1

316

Risk Ratio (Random, 95% CI)

1.01 [0.60, 1.70]

3.16 Sad, tearful or depressed

4

707

Risk Ratio (Random, 95% CI)

1.41 [0.86, 2.29]

3.17 Somnolence

2

173

Risk Ratio (Random, 95% CI)

0.59 [0.11, 3.11]

3.18 Trouble sleeping or sleep problems

13

2416

Risk Ratio (Random, 95% CI)

1.60 [1.15, 2.23]

3.19 Tics or nervous movements

8

1231

Risk Ratio (Random, 95% CI)

0.85 [0.26, 2.79]

3.20 Worried or anxious

3

596

Risk Ratio (Random, 95% CI)

1.37 [0.84, 2.25]

4 Digestive system Show forest plot

18

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 7.4

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Digestive system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Digestive system.

4.1 Decreased appetite

16

2962

Risk Ratio (IV, Random, 95% CI)

3.66 [2.56, 5.23]

4.2 Decreased weight

6

859

Risk Ratio (IV, Random, 95% CI)

3.89 [1.43, 10.59]

4.3 Diarrhoea

5

857

Risk Ratio (IV, Random, 95% CI)

1.07 [0.41, 2.74]

4.4 Dyspepsia

2

159

Risk Ratio (IV, Random, 95% CI)

1.80 [0.71, 4.54]

4.5 Increased appetite

1

179

Risk Ratio (IV, Random, 95% CI)

0.07 [0.00, 1.43]

4.6 Nausea

11

1995

Risk Ratio (IV, Random, 95% CI)

1.30 [0.85, 1.99]

4.7 Stomachache

13

2341

Risk Ratio (IV, Random, 95% CI)

1.30 [1.00, 1.69]

4.8 Vomiting

11

1916

Risk Ratio (IV, Random, 95% CI)

1.17 [0.76, 1.79]

5 Circulatory and respiratory systems Show forest plot

8

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 7.5

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems.

5.1 ECG: prolonged QT‐interval

2

466

Risk Ratio (Random, 95% CI)

0.81 [0.13, 5.00]

5.2 ECG: tachycardia

1

245

Risk Ratio (Random, 95% CI)

1.04 [0.11, 10.18]

5.3 Cough

4

996

Risk Ratio (Random, 95% CI)

0.95 [0.41, 2.18]

5.4 Nasal congestion

2

479

Risk Ratio (Random, 95% CI)

1.19 [0.59, 2.41]

5.5 Pharyngolaryngeal pain

1

303

Risk Ratio (Random, 95% CI)

1.12 [0.59, 2.13]

5.6 Supraventricular extrasystoles

1

17

Risk Ratio (Random, 95% CI)

3.00 [0.11, 84.55]

5.7 Upper respiratory tract infection

1

217

Risk Ratio (Random, 95% CI)

1.07 [0.42, 2.76]

6 Skeletal and muscular systems Show forest plot

2

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 7.6

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Skeletal and muscular systems.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Skeletal and muscular systems.

6.1 Arthralgia

1

303

Risk Ratio (IV, Random, 95% CI)

0.67 [0.24, 1.84]

6.2 Asthenia

1

177

Risk Ratio (IV, Random, 95% CI)

0.21 [0.01, 4.25]

6.3 Back pain

1

303

Risk Ratio (IV, Random, 95% CI)

0.81 [0.39, 1.66]

6.4 Myalgia

1

303

Risk Ratio (IV, Random, 95% CI)

0.60 [0.23, 1.62]

6.5 Toothache

1

303

Risk Ratio (IV, Random, 95% CI)

1.01 [0.43, 2.35]

7 Immune system Show forest plot

7

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 7.7

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Immune system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Immune system.

7.1 Gastroenteritis

3

435

Risk Ratio (IV, Random, 95% CI)

4.63 [0.99, 21.72]

7.2 Influenza

3

624

Risk Ratio (IV, Random, 95% CI)

0.65 [0.20, 2.10]

7.3 Nasopharyngitis

5

979

Risk Ratio (IV, Random, 95% CI)

1.15 [0.70, 1.87]

7.4 Otitis media

1

100

Risk Ratio (IV, Random, 95% CI)

1.77 [0.17, 18.94]

7.5 Pharyngitis

2

293

Risk Ratio (IV, Random, 95% CI)

2.43 [0.49, 12.05]

7.6 Pyrexia

2

400

Risk Ratio (IV, Random, 95% CI)

1.02 [0.01, 87.72]

7.7 Rhinitis

1

132

Risk Ratio (IV, Random, 95% CI)

1.28 [0.43, 3.79]

7.8 Upper respiratory tract infection ‐ not otherwise specified (NOS)

5

917

Risk Ratio (IV, Random, 95% CI)

1.19 [0.68, 2.06]

7.9 Viral infection NOS

3

614

Risk Ratio (IV, Random, 95% CI)

0.70 [0.23, 2.15]

8 Height Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 7.8

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 8 Height.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 8 Height.

9 Weight Show forest plot

5

805

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

‐1.12 [‐1.55, ‐0.70]

Analysis 7.9

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 9 Weight.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 9 Weight.

10 BMI Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 7.10

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 10 BMI.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 10 BMI.

11 Vital signs Show forest plot

9

3374

Mean Difference (IV, Random, 95% CI)

1.41 [0.30, 2.52]

Analysis 7.11

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 11 Vital signs.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 11 Vital signs.

11.1 Diastolic blood pressure (mmHg)

8

1067

Mean Difference (IV, Random, 95% CI)

0.94 [‐0.12, 2.01]

11.2 Systolic blood pressure (mmHg)

8

1067

Mean Difference (IV, Random, 95% CI)

‐0.05 [‐1.25, 1.16]

11.3 Pulse or heart rate (bpm)

8

1240

Mean Difference (IV, Random, 95% CI)

3.41 [0.87, 5.94]

12 Other Show forest plot

5

1815

Risk Ratio (IV, Random, 95% CI)

1.20 [0.56, 2.57]

Analysis 7.12

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 12 Other.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 12 Other.

12.1 Accidental injury

3

656

Risk Ratio (IV, Random, 95% CI)

0.99 [0.48, 2.07]

12.2 Epistasis

1

132

Risk Ratio (IV, Random, 95% CI)

4.25 [0.23, 77.22]

12.3 Excoriation

1

303

Risk Ratio (IV, Random, 95% CI)

3.52 [1.19, 10.46]

12.4 Overdose

1

221

Risk Ratio (IV, Random, 95% CI)

2.97 [0.12, 72.20]

12.5 Skin disorder (rash)

2

200

Risk Ratio (IV, Random, 95% CI)

0.52 [0.01, 26.44]

12.6 Skin laceration

1

303

Risk Ratio (IV, Random, 95% CI)

0.42 [0.15, 1.16]

Open in table viewer
Comparison 8. Number of non‐serious adverse events: cross‐over trials (endpoint data)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of non‐serious adverse events Show forest plot

21

2072

Risk Ratio (Random, 95% CI)

1.33 [1.11, 1.58]

Analysis 8.1

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 1 Total number of non‐serious adverse events.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 1 Total number of non‐serious adverse events.

2 Subgroup analysis: total number of non‐serious adverse events according to dose Show forest plot

21

2859

Risk Ratio (Random, 95% CI)

1.26 [1.11, 1.44]

Analysis 8.2

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

2.1 Low dose

16

1539

Risk Ratio (Random, 95% CI)

1.11 [0.94, 1.31]

2.2 High dose

12

1080

Risk Ratio (Random, 95% CI)

1.57 [1.22, 2.01]

2.3 Unknown dose

2

240

Risk Ratio (Random, 95% CI)

0.98 [0.51, 1.86]

3 Nervous system Show forest plot

50

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 8.3

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 3 Nervous system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 3 Nervous system.

3.1 Aggression

2

589

Risk Ratio (Random, 95% CI)

0.52 [0.17, 1.60]

3.2 Agitation

1

62

Risk Ratio (Random, 95% CI)

1.18 [0.38, 3.60]

3.3 Anger

3

264

Risk Ratio (Random, 95% CI)

0.45 [0.26, 0.77]

3.4 Behavioural complaints

1

82

Risk Ratio (Random, 95% CI)

0.55 [0.35, 0.86]

3.5 Buccal or lingual movements

4

302

Risk Ratio (Random, 95% CI)

1.06 [0.62, 1.79]

3.6 Compulsive acts

1

90

Risk Ratio (Random, 95% CI)

2.57 [1.45, 4.56]

3.7 Daydreaming

3

222

Risk Ratio (Random, 95% CI)

0.66 [0.44, 0.98]

3.8 Dizziness

9

746

Risk Ratio (Random, 95% CI)

1.17 [0.89, 1.55]

3.9 Drowsiness: dull, tired, listless or sleepy

21

1350

Risk Ratio (Random, 95% CI)

0.97 [0.73, 1.28]

3.10 Euphoria

6

405

Risk Ratio (Random, 95% CI)

1.06 [0.72, 1.57]

3.11 Headache

37

3752

Risk Ratio (Random, 95% CI)

1.21 [1.01, 1.45]

3.12 Insomnia or sleep problems

31

3270

Risk Ratio (Random, 95% CI)

1.57 [1.20, 2.06]

3.13 Irritability

23

2238

Risk Ratio (Random, 95% CI)

0.92 [0.66, 1.27]

3.14 Nightmares

10

686

Risk Ratio (Random, 95% CI)

0.97 [0.66, 1.42]

3.15 Overly meticulous

1

96

Risk Ratio (Random, 95% CI)

40.77 [2.35, 706.72]

3.16 Obsessive thinking

1

90

Risk Ratio (Random, 95% CI)

2.35 [1.53, 3.62]

3.17 Picking at skin or fingers, nail biting, lip or cheek chewing

15

888

Risk Ratio (Random, 95% CI)

1.12 [0.88, 1.41]

3.18 Repetitive language

1

48

Risk Ratio (Random, 95% CI)

1.0 [0.32, 3.10]

3.19 Sad, tearful or depressed

23

1849

Risk Ratio (Random, 95% CI)

1.15 [0.94, 1.41]

3.20 Socially withdrawn ‐ decreased interaction with others

12

771

Risk Ratio (Random, 95% CI)

1.24 [0.82, 1.87]

3.21 Sleep efficiency (SEF)

2

108

Risk Ratio (Random, 95% CI)

0.48 [0.02, 14.28]

3.22 Stares a lot

9

904

Risk Ratio (Random, 95% CI)

1.03 [0.75, 1.40]

3.23 Tics or nervous movements

19

1403

Risk Ratio (Random, 95% CI)

1.33 [1.03, 1.72]

3.24 Unusual blinking

1

48

Risk Ratio (Random, 95% CI)

3.13 [0.12, 80.68]

3.25 Worried or anxious

20

1673

Risk Ratio (Random, 95% CI)

0.82 [0.60, 1.12]

4 Digestive system Show forest plot

42

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 8.4

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 4 Digestive system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 4 Digestive system.

4.1 Decreased appetite or loss of appetite

35

3862

Risk Ratio (Random, 95% CI)

3.04 [2.35, 3.94]

4.2 Diarrhoea

3

402

Risk Ratio (Random, 95% CI)

0.58 [0.19, 1.74]

4.3 Dry mouth

5

342

Risk Ratio (Random, 95% CI)

1.25 [0.54, 2.90]

4.4 Dyspepsia

1

62

Risk Ratio (Random, 95% CI)

0.22 [0.02, 2.14]

4.5 Nausea

9

768

Risk Ratio (Random, 95% CI)

1.52 [1.00, 2.30]

4.6 Increased appetite

1

136

Risk Ratio (Random, 95% CI)

0.20 [0.08, 0.50]

4.7 Stomachache

33

3777

Risk Ratio (Random, 95% CI)

1.61 [1.27, 2.04]

4.8 Vomiting

4

710

Risk Ratio (Random, 95% CI)

0.90 [0.26, 3.11]

5 Urinary system: urinary incontinence Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 8.5

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 5 Urinary system: urinary incontinence.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 5 Urinary system: urinary incontinence.

6 Skeletal and muscular system: somatic complaints Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.6

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 6 Skeletal and muscular system: somatic complaints.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 6 Skeletal and muscular system: somatic complaints.

7 Immune system Show forest plot

7

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 8.7

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 7 Immune system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 7 Immune system.

7.1 Allergic rhinitis

4

475

Risk Ratio (IV, Random, 95% CI)

1.38 [0.35, 5.51]

7.2 Fever

2

91

Risk Ratio (IV, Random, 95% CI)

1.39 [0.09, 20.56]

7.3 Lymphadenitis

2

296

Risk Ratio (IV, Random, 95% CI)

3.93 [0.44, 35.11]

7.4 Pharyngolaryngeal pain

1

160

Risk Ratio (IV, Random, 95% CI)

2.0 [0.19, 21.62]

7.5 Pharyngitis

4

754

Risk Ratio (IV, Random, 95% CI)

0.71 [0.19, 2.62]

7.6 Upper respiratory tract infection

5

888

Risk Ratio (IV, Random, 95% CI)

0.61 [0.27, 1.37]

8 Skin Show forest plot

3

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 8.8

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 8 Skin.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 8 Skin.

8.1 Rash

2

208

Risk Ratio (IV, Random, 95% CI)

0.96 [0.14, 6.41]

8.2 Skin laceration

1

167

Risk Ratio (IV, Random, 95% CI)

2.96 [0.12, 71.75]

9 Vital signs Show forest plot

14

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.9

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 9 Vital signs.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 9 Vital signs.

9.1 Diastolic blood pressure (mmHg)

11

755

Mean Difference (IV, Random, 95% CI)

1.23 [‐0.39, 2.86]

9.2 Systolic blood pressure (mmHg)

11

755

Mean Difference (IV, Random, 95% CI)

0.53 [‐1.30, 2.36]

9.3 Pulse or heart rate (bpm)

14

939

Mean Difference (IV, Random, 95% CI)

5.06 [2.88, 7.24]

10 Height (cm) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.10

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 10 Height (cm).

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 10 Height (cm).

11 Weight Show forest plot

6

530

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

‐0.06 [‐0.23, 0.11]

Analysis 8.11

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 11 Weight.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 11 Weight.

Open in table viewer
Comparison 9. Teacher‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials: risk of bias Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

Analysis 9.1

Comparison 9 Teacher‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials: risk of bias.

Comparison 9 Teacher‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials: risk of bias.

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

5

668

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

‐0.87 [‐1.04, ‐0.71]

2 Subgroup analysis: types of scales Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

Analysis 9.2

Comparison 9 Teacher‐rated general behaviour, Outcome 2 Subgroup analysis: types of scales.

Comparison 9 Teacher‐rated general behaviour, Outcome 2 Subgroup analysis: types of scales.

2.1 Conners' Global Index ‐ Teacher (CGI‐T)

1

314

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

‐0.91 [‐1.14, ‐0.68]

2.2 Groninger Behaviour Observation Scale (GBOS)

1

43

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

‐0.84 [‐1.46, ‐0.21]

2.3 Conners' Teacher Rating Scale ‐ Conduct problems

1

25

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

‐0.67 [‐1.48, 0.14]

2.4 IOWA Conners' Rating Scale ‐ Oppositional/Defiant (IOWA‐O/D)

2

286

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

‐0.86 [‐1.12, ‐0.59]

3 Subgroup analysis: dose Show forest plot

5

696

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

‐0.85 [‐1.02, ‐0.69]

Analysis 9.3

Comparison 9 Teacher‐rated general behaviour, Outcome 3 Subgroup analysis: dose.

Comparison 9 Teacher‐rated general behaviour, Outcome 3 Subgroup analysis: dose.

3.1 Low dose

2

71

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

‐0.67 [‐1.16, ‐0.19]

3.2 High dose

3

466

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

‐0.89 [‐1.08, ‐0.70]

3.3 Unknown dose

1

159

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

‐0.84 [‐1.21, ‐0.46]

4 Subgroup analysis: duration of treatment Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

Analysis 9.4

Comparison 9 Teacher‐rated general behaviour, Outcome 4 Subgroup analysis: duration of treatment.

Comparison 9 Teacher‐rated general behaviour, Outcome 4 Subgroup analysis: duration of treatment.

4.1 Short term (up to 6 months)

5

668

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

‐0.87 [‐1.04, ‐0.71]

4.2 Long term (over 6 months)

0

0

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

0.0 [0.0, 0.0]

5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

Analysis 9.5

Comparison 9 Teacher‐rated general behaviour, Outcome 5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials.

Comparison 9 Teacher‐rated general behaviour, Outcome 5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials.

5.1 Parallel‐group trials

5

668

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

‐0.87 [‐1.04, ‐0.71]

5.2 First‐period cross‐over trials

0

0

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

0.0 [0.0, 0.0]

6 General behaviour, cross‐over trials (endpoint data) Show forest plot

16

2014

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

‐0.69 [‐0.78, ‐0.60]

Analysis 9.6

Comparison 9 Teacher‐rated general behaviour, Outcome 6 General behaviour, cross‐over trials (endpoint data).

Comparison 9 Teacher‐rated general behaviour, Outcome 6 General behaviour, cross‐over trials (endpoint data).

6.1 Low dose

13

1110

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

‐0.60 [‐0.72, ‐0.48]

6.2 High dose

12

904

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

‐0.82 [‐0.95, ‐0.68]

7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

16

1308

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

‐0.75 [‐0.87, ‐0.63]

Analysis 9.7

Comparison 9 Teacher‐rated general behaviour, Outcome 7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 9 Teacher‐rated general behaviour, Outcome 7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

7.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

7.2 High risk of bias

16

1308

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

‐0.75 [‐0.87, ‐0.63]

8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data) Show forest plot

21

1976

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

‐0.79 [‐0.88, ‐0.70]

Analysis 9.8

Comparison 9 Teacher‐rated general behaviour, Outcome 8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data).

Comparison 9 Teacher‐rated general behaviour, Outcome 8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data).

8.1 All parallel‐group trials and first‐period cross‐over trials

5

668

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

‐0.87 [‐1.04, ‐0.71]

8.2 Cross‐over trials (endpoint data)

16

1308

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

‐0.75 [‐0.87, ‐0.63]

Open in table viewer
Comparison 10. Independent assessor‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 General behaviour, cross‐over trials (endpoint data) Show forest plot

8

1241

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

‐0.60 [‐0.75, ‐0.46]

Analysis 10.1

Comparison 10 Independent assessor‐rated general behaviour, Outcome 1 General behaviour, cross‐over trials (endpoint data).

Comparison 10 Independent assessor‐rated general behaviour, Outcome 1 General behaviour, cross‐over trials (endpoint data).

1.1 Low dose

7

903

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

‐0.56 [‐0.76, ‐0.36]

1.2 High dose

5

338

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

‐0.71 [‐0.93, ‐0.49]

2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

8

951

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

‐0.69 [‐0.88, ‐0.49]

Analysis 10.2

Comparison 10 Independent assessor‐rated general behaviour, Outcome 2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 10 Independent assessor‐rated general behaviour, Outcome 2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

2.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

2.2 High risk of bias

8

951

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

‐0.69 [‐0.88, ‐0.49]

3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data) Show forest plot

8

951

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

‐0.69 [‐0.88, ‐0.49]

Analysis 10.3

Comparison 10 Independent assessor‐rated general behaviour, Outcome 3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data).

Comparison 10 Independent assessor‐rated general behaviour, Outcome 3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data).

3.1 Parallel‐group trials and first‐period cross‐over trials

0

0

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

0.0 [0.0, 0.0]

3.2 Cross‐over trials (endpoint data)

8

951

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

‐0.69 [‐0.88, ‐0.49]

Open in table viewer
Comparison 11. Parent‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.1

Comparison 11 Parent‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 11 Parent‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

2 Subgroup analysis: risk of bias Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.2

Comparison 11 Parent‐rated general behaviour, Outcome 2 Subgroup analysis: risk of bias.

Comparison 11 Parent‐rated general behaviour, Outcome 2 Subgroup analysis: risk of bias.

2.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

2.2 High risk of bias

6

670

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

‐0.53 [‐0.78, ‐0.27]

3 Subgroup analysis: types of scales Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.3

Comparison 11 Parent‐rated general behaviour, Outcome 3 Subgroup analysis: types of scales.

Comparison 11 Parent‐rated general behaviour, Outcome 3 Subgroup analysis: types of scales.

3.1 The Weekly Parent Ratings of Evening and Morning Behaviour (WPREMB) ‐ Revised

1

17

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

0.50 [‐0.47, 1.47]

3.2 Conners' Global Index (CGI) ‐ Parent

2

352

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

‐0.41 [‐0.63, ‐0.20]

3.3 Swanson, Nolan and Pelham, Fourth Edition ‐ Oppositional (SNAP‐IV‐Oppositional)

1

15

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

‐0.86 [‐1.95, 0.23]

3.4 IOWA Conners' Rating Scale ‐ Oppositional/Defiant (IOWA‐I/O)

2

286

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

‐0.75 [‐1.01, ‐0.49]

4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.4

Comparison 11 Parent‐rated general behaviour, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 11 Parent‐rated general behaviour, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

4.1 Parallel‐group trials

5

655

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

‐0.51 [‐0.78, ‐0.23]

4.2 First‐period cross‐over trials

1

15

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

‐0.86 [‐1.95, 0.23]

5 Subgroup analysis: duration of treatment Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.5

Comparison 11 Parent‐rated general behaviour, Outcome 5 Subgroup analysis: duration of treatment.

Comparison 11 Parent‐rated general behaviour, Outcome 5 Subgroup analysis: duration of treatment.

5.1 Short term (up to 6 months)

6

670

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

‐0.53 [‐0.78, ‐0.27]

5.2 Long term (over 6 months)

0

0

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

0.0 [0.0, 0.0]

6 Subgroup analysis: dose Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

Analysis 11.6

Comparison 11 Parent‐rated general behaviour, Outcome 6 Subgroup analysis: dose.

Comparison 11 Parent‐rated general behaviour, Outcome 6 Subgroup analysis: dose.

6.1 Low dose

0

0

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

0.0 [0.0, 0.0]

6.2 High dose

4

496

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

‐0.42 [‐0.77, ‐0.08]

6.3 Unknown dose

2

174

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

‐0.73 [‐1.08, ‐0.38]

7 General behaviour, cross‐over trials (endpoint data) Show forest plot

6

550

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

‐0.75 [‐0.93, ‐0.56]

Analysis 11.7

Comparison 11 Parent‐rated general behaviour, Outcome 7 General behaviour, cross‐over trials (endpoint data).

Comparison 11 Parent‐rated general behaviour, Outcome 7 General behaviour, cross‐over trials (endpoint data).

7.1 Low dose

5

248

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

‐0.65 [‐0.93, ‐0.38]

7.2 High dose

4

302

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

‐0.83 [‐1.07, ‐0.60]

8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

6

384

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

‐0.84 [‐1.05, ‐0.63]

Analysis 11.8

Comparison 11 Parent‐rated general behaviour, Outcome 8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 11 Parent‐rated general behaviour, Outcome 8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

8.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

8.2 High risk of bias

6

384

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

‐0.84 [‐1.05, ‐0.63]

9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data) Show forest plot

12

1054

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

‐0.68 [‐0.86, ‐0.50]

Analysis 11.9

Comparison 11 Parent‐rated general behaviour, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data).

Comparison 11 Parent‐rated general behaviour, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data).

9.1 All parallel‐group trials and first‐period cross‐over trials

6

670

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

‐0.53 [‐0.78, ‐0.27]

9.2 Cross‐over trials (endpoint data)

6

384

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

‐0.84 [‐1.05, ‐0.63]

Open in table viewer
Comparison 12. Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisions of raters Show forest plot

8

1338

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

‐0.69 [‐0.86, ‐0.52]

Analysis 12.1

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparisions of raters.

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparisions of raters.

1.1 Teacher‐rated

5

668

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

‐0.87 [‐1.04, ‐0.71]

1.2 Independent assessor‐rated

0

0

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

0.0 [0.0, 0.0]

1.3 Parent‐rated

6

670

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

‐0.53 [‐0.78, ‐0.27]

2 Comorbidity versus no comorbidity Show forest plot

7

579

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

‐0.70 [‐0.98, ‐0.43]

Analysis 12.2

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Comorbidity versus no comorbidity.

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Comorbidity versus no comorbidity.

2.1 ADHD with comorbidity

6

265

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

‐0.59 [‐0.95, ‐0.23]

2.2 ADHD without comorbidity

1

314

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

‐0.91 [‐1.14, ‐0.68]

3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 12.3

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated).

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated).

3.1 First‐period data

1

16

Mean Difference (IV, Random, 95% CI)

‐0.81 [‐1.75, 0.13]

3.2 Endpoint data

1

14

Mean Difference (IV, Random, 95% CI)

0.14 [‐0.71, 1.00]

Open in table viewer
Comparison 13. Quality of life: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Subgroup analysis: types of scales Show forest plot

3

514

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

0.61 [0.42, 0.80]

Analysis 13.1

Comparison 13 Quality of life: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Subgroup analysis: types of scales.

Comparison 13 Quality of life: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Subgroup analysis: types of scales.

1.1 Child Health Questionnaire (CHQ)

1

257

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

0.54 [0.25, 0.83]

1.2 Children´s Global Assessment Scale (CGAS)

1

36

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

0.79 [0.10, 1.47]

1.3 Child Health and Illness Profile, Child Edition: Parent Report Form (CHIP‐CE:PRF)

1

221

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

0.64 [0.37, 0.91]

Flow chart.
Figuras y tablas -
Figure 1

Flow chart.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

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

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial.
Figuras y tablas -
Figure 3

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

Funnel plot of comparison: 1. Teacher‐rated ADHD symptoms, outcome: 1.8 All data at low and high risk of bias (parallel‐group and cross‐over trials).
Figuras y tablas -
Figure 4

Funnel plot of comparison: 1. Teacher‐rated ADHD symptoms, outcome: 1.8 All data at low and high risk of bias (parallel‐group and cross‐over trials).

Trial Sequential Analysis: serious adverse events.
Figuras y tablas -
Figure 5

Trial Sequential Analysis: serious adverse events.

Trial Sequential Analysis: non‐serious adverse events.
Figuras y tablas -
Figure 6

Trial Sequential Analysis: non‐serious adverse events.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.
Figuras y tablas -
Analysis 1.1

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 1.2

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 3 Medication status: medication naive versus not medication naive.
Figuras y tablas -
Analysis 1.3

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 3 Medication status: medication naive versus not medication naive.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 4 Subgroup analysis: duration of treatment.
Figuras y tablas -
Analysis 1.4

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 4 Subgroup analysis: duration of treatment.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 5 Subgroup analysis: dose.
Figuras y tablas -
Analysis 1.5

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 5 Subgroup analysis: dose.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.
Figuras y tablas -
Analysis 1.6

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.
Figuras y tablas -
Analysis 1.7

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trial (endpoint data).
Figuras y tablas -
Analysis 1.8

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trial (endpoint data).

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.
Figuras y tablas -
Analysis 1.9

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 1.10

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 11 All parallel‐group trials and cross‐over trials: risk of bias.
Figuras y tablas -
Analysis 1.11

Comparison 1 Teacher‐rated ADHD symptoms, Outcome 11 All parallel‐group trials and cross‐over trials: risk of bias.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.
Figuras y tablas -
Analysis 2.1

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 2.2

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.
Figuras y tablas -
Analysis 2.3

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.
Figuras y tablas -
Analysis 2.4

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.
Figuras y tablas -
Analysis 2.5

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 6 Subgroup analysis: dose.
Figuras y tablas -
Analysis 2.6

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 6 Subgroup analysis: dose.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias.
Figuras y tablas -
Analysis 2.7

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.
Figuras y tablas -
Analysis 2.8

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 2.9

Comparison 2 Independent assessor‐rated ADHD symptoms, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 3 Parent‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.
Figuras y tablas -
Analysis 3.1

Comparison 3 Parent‐rated ADHD symptoms, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 3.2

Comparison 3 Parent‐rated ADHD symptoms, Outcome 2 Subgroup analysis: types of scales.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.
Figuras y tablas -
Analysis 3.3

Comparison 3 Parent‐rated ADHD symptoms, Outcome 3 Subgroup analysis: duration of treatment.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 4 Subgroup analysis: dose.
Figuras y tablas -
Analysis 3.4

Comparison 3 Parent‐rated ADHD symptoms, Outcome 4 Subgroup analysis: dose.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 5 Medication status: medication naive versus not medication naive.
Figuras y tablas -
Analysis 3.5

Comparison 3 Parent‐rated ADHD symptoms, Outcome 5 Medication status: medication naive versus not medication naive.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.
Figuras y tablas -
Analysis 3.6

Comparison 3 Parent‐rated ADHD symptoms, Outcome 6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.
Figuras y tablas -
Analysis 3.7

Comparison 3 Parent‐rated ADHD symptoms, Outcome 7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 3.8

Comparison 3 Parent‐rated ADHD symptoms, Outcome 8 ADHD symptoms, cross‐over trials (endpoint data).

Comparison 3 Parent‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.
Figuras y tablas -
Analysis 3.9

Comparison 3 Parent‐rated ADHD symptoms, Outcome 9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose.

Comparison 3 Parent‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 3.10

Comparison 3 Parent‐rated ADHD symptoms, Outcome 10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data).

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparision of raters.
Figuras y tablas -
Analysis 4.1

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparision of raters.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Age.
Figuras y tablas -
Analysis 4.2

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Age.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Comorbidity versus no comorbidity.
Figuras y tablas -
Analysis 4.3

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Comorbidity versus no comorbidity.

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated).
Figuras y tablas -
Analysis 4.4

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated).

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated).
Figuras y tablas -
Analysis 4.5

Comparison 4 Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated).

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Number of serious adverse events (SAE).
Figuras y tablas -
Analysis 5.1

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Number of serious adverse events (SAE).

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Nervous system.
Figuras y tablas -
Analysis 5.2

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Nervous system.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Digestive system: gastrointestinal disorders.
Figuras y tablas -
Analysis 5.3

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Digestive system: gastrointestinal disorders.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Urinary system: kidney infection.
Figuras y tablas -
Analysis 5.4

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Urinary system: kidney infection.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems: asthma.
Figuras y tablas -
Analysis 5.5

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems: asthma.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Immune system: cyst rupture.
Figuras y tablas -
Analysis 5.6

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Immune system: cyst rupture.

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Other: drug toxicity.
Figuras y tablas -
Analysis 5.7

Comparison 5 Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Other: drug toxicity.

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 1 Number of serious adverse events (SAE).
Figuras y tablas -
Analysis 6.1

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 1 Number of serious adverse events (SAE).

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 2 Hallucinations/psychosis.
Figuras y tablas -
Analysis 6.2

Comparison 6 Number of serious adverse events: cross‐over trials (endpoint data), Outcome 2 Hallucinations/psychosis.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Total number of non‐serious adverse events.
Figuras y tablas -
Analysis 7.1

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Total number of non‐serious adverse events.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.
Figuras y tablas -
Analysis 7.2

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Nervous system.
Figuras y tablas -
Analysis 7.3

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Nervous system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Digestive system.
Figuras y tablas -
Analysis 7.4

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 4 Digestive system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems.
Figuras y tablas -
Analysis 7.5

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 5 Circulatory and respiratory systems.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Skeletal and muscular systems.
Figuras y tablas -
Analysis 7.6

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 6 Skeletal and muscular systems.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Immune system.
Figuras y tablas -
Analysis 7.7

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 7 Immune system.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 8 Height.
Figuras y tablas -
Analysis 7.8

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 8 Height.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 9 Weight.
Figuras y tablas -
Analysis 7.9

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 9 Weight.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 10 BMI.
Figuras y tablas -
Analysis 7.10

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 10 BMI.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 11 Vital signs.
Figuras y tablas -
Analysis 7.11

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 11 Vital signs.

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 12 Other.
Figuras y tablas -
Analysis 7.12

Comparison 7 Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials, Outcome 12 Other.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 1 Total number of non‐serious adverse events.
Figuras y tablas -
Analysis 8.1

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 1 Total number of non‐serious adverse events.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.
Figuras y tablas -
Analysis 8.2

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 2 Subgroup analysis: total number of non‐serious adverse events according to dose.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 3 Nervous system.
Figuras y tablas -
Analysis 8.3

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 3 Nervous system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 4 Digestive system.
Figuras y tablas -
Analysis 8.4

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 4 Digestive system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 5 Urinary system: urinary incontinence.
Figuras y tablas -
Analysis 8.5

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 5 Urinary system: urinary incontinence.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 6 Skeletal and muscular system: somatic complaints.
Figuras y tablas -
Analysis 8.6

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 6 Skeletal and muscular system: somatic complaints.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 7 Immune system.
Figuras y tablas -
Analysis 8.7

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 7 Immune system.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 8 Skin.
Figuras y tablas -
Analysis 8.8

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 8 Skin.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 9 Vital signs.
Figuras y tablas -
Analysis 8.9

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 9 Vital signs.

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 10 Height (cm).
Figuras y tablas -
Analysis 8.10

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 10 Height (cm).

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 11 Weight.
Figuras y tablas -
Analysis 8.11

Comparison 8 Number of non‐serious adverse events: cross‐over trials (endpoint data), Outcome 11 Weight.

Comparison 9 Teacher‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials: risk of bias.
Figuras y tablas -
Analysis 9.1

Comparison 9 Teacher‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials: risk of bias.

Comparison 9 Teacher‐rated general behaviour, Outcome 2 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 9.2

Comparison 9 Teacher‐rated general behaviour, Outcome 2 Subgroup analysis: types of scales.

Comparison 9 Teacher‐rated general behaviour, Outcome 3 Subgroup analysis: dose.
Figuras y tablas -
Analysis 9.3

Comparison 9 Teacher‐rated general behaviour, Outcome 3 Subgroup analysis: dose.

Comparison 9 Teacher‐rated general behaviour, Outcome 4 Subgroup analysis: duration of treatment.
Figuras y tablas -
Analysis 9.4

Comparison 9 Teacher‐rated general behaviour, Outcome 4 Subgroup analysis: duration of treatment.

Comparison 9 Teacher‐rated general behaviour, Outcome 5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials.
Figuras y tablas -
Analysis 9.5

Comparison 9 Teacher‐rated general behaviour, Outcome 5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials.

Comparison 9 Teacher‐rated general behaviour, Outcome 6 General behaviour, cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 9.6

Comparison 9 Teacher‐rated general behaviour, Outcome 6 General behaviour, cross‐over trials (endpoint data).

Comparison 9 Teacher‐rated general behaviour, Outcome 7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.
Figuras y tablas -
Analysis 9.7

Comparison 9 Teacher‐rated general behaviour, Outcome 7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 9 Teacher‐rated general behaviour, Outcome 8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 9.8

Comparison 9 Teacher‐rated general behaviour, Outcome 8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data).

Comparison 10 Independent assessor‐rated general behaviour, Outcome 1 General behaviour, cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 10.1

Comparison 10 Independent assessor‐rated general behaviour, Outcome 1 General behaviour, cross‐over trials (endpoint data).

Comparison 10 Independent assessor‐rated general behaviour, Outcome 2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.
Figuras y tablas -
Analysis 10.2

Comparison 10 Independent assessor‐rated general behaviour, Outcome 2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 10 Independent assessor‐rated general behaviour, Outcome 3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 10.3

Comparison 10 Independent assessor‐rated general behaviour, Outcome 3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data).

Comparison 11 Parent‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.
Figuras y tablas -
Analysis 11.1

Comparison 11 Parent‐rated general behaviour, Outcome 1 All parallel‐group trials and first‐period cross‐over trials.

Comparison 11 Parent‐rated general behaviour, Outcome 2 Subgroup analysis: risk of bias.
Figuras y tablas -
Analysis 11.2

Comparison 11 Parent‐rated general behaviour, Outcome 2 Subgroup analysis: risk of bias.

Comparison 11 Parent‐rated general behaviour, Outcome 3 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 11.3

Comparison 11 Parent‐rated general behaviour, Outcome 3 Subgroup analysis: types of scales.

Comparison 11 Parent‐rated general behaviour, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.
Figuras y tablas -
Analysis 11.4

Comparison 11 Parent‐rated general behaviour, Outcome 4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials.

Comparison 11 Parent‐rated general behaviour, Outcome 5 Subgroup analysis: duration of treatment.
Figuras y tablas -
Analysis 11.5

Comparison 11 Parent‐rated general behaviour, Outcome 5 Subgroup analysis: duration of treatment.

Comparison 11 Parent‐rated general behaviour, Outcome 6 Subgroup analysis: dose.
Figuras y tablas -
Analysis 11.6

Comparison 11 Parent‐rated general behaviour, Outcome 6 Subgroup analysis: dose.

Comparison 11 Parent‐rated general behaviour, Outcome 7 General behaviour, cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 11.7

Comparison 11 Parent‐rated general behaviour, Outcome 7 General behaviour, cross‐over trials (endpoint data).

Comparison 11 Parent‐rated general behaviour, Outcome 8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.
Figuras y tablas -
Analysis 11.8

Comparison 11 Parent‐rated general behaviour, Outcome 8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias.

Comparison 11 Parent‐rated general behaviour, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data).
Figuras y tablas -
Analysis 11.9

Comparison 11 Parent‐rated general behaviour, Outcome 9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data).

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparisions of raters.
Figuras y tablas -
Analysis 12.1

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Comparisions of raters.

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Comorbidity versus no comorbidity.
Figuras y tablas -
Analysis 12.2

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 2 Comorbidity versus no comorbidity.

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated).
Figuras y tablas -
Analysis 12.3

Comparison 12 Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials, Outcome 3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated).

Comparison 13 Quality of life: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Subgroup analysis: types of scales.
Figuras y tablas -
Analysis 13.1

Comparison 13 Quality of life: parallel‐group trials and first‐period cross‐over trials, Outcome 1 Subgroup analysis: types of scales.

Summary of findings for the main comparison. Methylphenidate compared with placebo or no intervention for ADHD

Methylphenidate compared with placebo or no intervention for ADHD

Patient or population: children and adolescents (up to and including 18 years of age) with ADHD
Settings: out‐patient clinic, in‐patient hospital ward and summer school
Intervention: methylphenidate
Comparison: placebo or no intervention

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo or no intervention

Methylphenidate

ADHD symptoms: all parallel‐group trials and first‐period cross‐over trials
ADHD Rating Scale (Teacher‐rated)

Average study duration: 74.8 days

Mean ADHD symptom score in the intervention groups corresponds to a mean difference of 9.6 (95% CI 11.25 to 8.00) on ADHD Rating Scale

SMD

‐0.77 (‐0.90 to ‐0.64)

1698
(19 studies)

⊕⊝⊝⊝
Very lowa,b

The analysis was conducted on a standardised scale with data from studies that used different teacher‐rated scales of symptoms (Conners' Teacher Rating Scale (CTRS), Strengths and Weaknesses of ADHD Symptoms and Normal Behaviour (SWAN) Scale, Schedule for Non‐adaptive and Adaptive Personality (SNAP) ‐ Teacher, Fremdbeurteilungsbogen für Hyperkinetische Störungen (FBB‐HKS)). The effect size has been translated on to the ADHD Rating Scale from the SMD

Total number of serious adverse events

Trial population

RR 0.98
(0.44 to 2.22)

1532
(9 studies)

⊕⊝⊝⊝
Very lowa,c

16 per 1000

16 per 1000
(7 to 36)

Total number of non‐serious adverse events

Trial population

RR 1.29
(1.10 to 1.51)

3132
(21 studies)

⊕⊝⊝⊝
Very lowa,b

408 per 1000

526 per 1000
(448 to 615)

General behaviour: all parallel‐group trials and first‐period cross‐over trials
General behaviour rating scales (Teacher‐rated)

Mean general behaviour score in the intervention groups was 0.87 standard mean deviations lower (95% CI 1.04 to 0.71 lower)

SMD

‐0.87 (‐0.71 to ‐1.04)

668
(5 studies)

⊕⊝⊝⊝
Very lowa,d

Quality of life

(Parent‐rated)

Mean quality of life score in the intervention groups corresponds to a mean difference of 8.0 (95% CI 5.49 to 10.46) on the Child Health Questionnaire

SMD 0.61
(0.42 to 0.80)

514
(3 studies)

⊕⊝⊝⊝
Very lowa,e

*The basis for the assumed risk (e.g. median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ADHD: Attention deficit hyperactivity disorder; CI: Confidence interval; RR: Risk ratio; SMD: Standardised mean difference

GRADE Working Group grades of evidence.
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

aDowngraded two levels due to high risk of bias (systematic errors causing overestimation of benefits and underestimation of harms) in several risk of bias domains, including lack of sufficient blinding and selective outcome reporting (many of the included trials did not report on this outcome).
bDowngraded one level due to inconsistency: moderate statistical heterogeneity.
c Downgraded one level due to imprecision: wide confidence intervals.
dDowngraded one level due to indirectness: children's general behavior was assessed by different types of rating scales with different focus on behavior.
e Downgraded one level due to indirectness: children's quality of life was assessed by their parents.

Figuras y tablas -
Summary of findings for the main comparison. Methylphenidate compared with placebo or no intervention for ADHD
Table 1. ADHD symptoms ‐ rating scales

Name of scale

Abbreviation

Reference

Abbreviated Conners’ Rating Scales, Parent (ACPRS) and Teacher (ACTRS),

including Abbreviated Parent Rating Scale (APRS) and Teacher Rating Scale, Hyperkinesis Index and ADHD and Emotional Lability subscales

ACRS

Conners 1997a

Abbreviated Symptom Questionnaire, including ASQ Teacher and ASQ Parent

ASQ

Conners 1995

Academic Performance Rating Scale

APRS

DuPaul 1991a

The ADD/H Comprehensive Teacher Rating Scale

ACTeRS

Ullmann 1984

ADHD/ODD Rating Scale, Parent‐ and Teacher‐Rated

ADHD‐RS

Barkley 1998

ADHD Rating Scale, including ADHD Rating Scale Parent and Teacher Ratings

ADHD‐RS

DuPaul 1991a

ADHD Rating Scale‐IV, including ADHD Rating Scale‐IV Parent and Teacher Versions

ADHD‐RS‐IV

DuPaul 1991a

Brief Psychiatric Rating Scale for Children

BPRS

Gale 1986

Child Attention Problems Rating Scale

CAP

Achenbach 1986

Child Attention Profile

CAP

Barkley 1988b

Child Behavior Rating Form

NCBHF

Aman 1996

Child Symptom Inventory

CSI

Gadow 1994

Children’s Psychiatric Rating Scale

CPRS

Pfefferbaum‐Levine 1983

Conners’ Abbreviated Hyperactivity Questionnaire

C‐HI

Conners 1997a

Conners’ Abbreviated Questionnaire

ASQ

Conners 1995

Conners’ Abbreviated Parent Teacher Questionnaire

APTQ

Rowe 1997

Conners’ Abbreviated Rating Scale

ABRS

Conners 1997a

Conners’ Abbreviated Symptom Questionnaire

ASQ

Conners 1995

Conners Abbreviated Symptom Questionnaire for Parents

ASQ‐Parent

Conners 1995

Conners’ Abbreviated Symptom Questionnaire for Teachers

ASQ‐Teacher

Conners 1997a

Conners’ Abbreviated Teacher Rating Scale

ABTRS

Conners 2001

Conners’ ADHD/DSM‐IV Scales Adolescent

CADS‐A

Conners 1997b

Conners’ ADHD/DSM‐IV Scales Parent

CADS–P, CADS‐P DSM‐IV

Conners 1997a

Conners’ ADHD/DSM‐IV Scale Teacher, including Inattentive and Hyperactive‐Impulsive subscales

CADS‐T, CADS‐T DSM‐IV

Conners 1997a

Conners’ Rating Scale ‐ Revised, Parent and Teacher: Hyperactivity and Conduct Factors score

CPRS‐R and CTRS‐R

Goyette 1978

Conners’ Hyperactivity Index, Parent and Teacher, including abbreviated versions

CPRS/CTRS‐Hyperactivity index

Conners 1997a

Conners’ Hyperkinesis Index

Milich 1980

Conners, Loney and Milich Scale

CLAM

Milich 1980

Conners’ Parent and Teacher Rating Scale ‐ Revised, Short Form

CRS‐R:S

Conners 1997a

Conners’ Parent Rating Scale, including abbreviated versions

CPRS

Conners 1998b

Conners’ Parent Rating Scale ‐ Revised

CPRS‐R

Conners 1997a

Conners’ Parent Rating Scale ‐ Revised, Short Form

CPRS‐R:S

Conners 1997a

Conners’ Parent Rating Scale ‐ Revised, Long Version

CPRS‐R:L

Conners 1997a

Conners’ Rating Scale ‐ Revised

CRS‐R

Conners 1997a

Conners’ Short Form Rating Scale, Parent and Teacher

Conners 1997a

Conners’ Teacher Rating Scale

CTRS

Conners 1998a

Conners’ Teacher Rating Scale ‐ Revised, Long Version

CTRS‐R:L

Conners 1998a

Diagnostic and Statistical Manual of Mental Disorders Total

DSM‐IV

APA 1994

Diagnostiksystem für Psychische Störungen im Kindes ‐ und Jugendalter nach ICD‐10 und DSM‐IV,

Parental Questionnaire of ADHD symptoms

DISYPS

Döpfner 2000

Fremdbeurteilungsbogen für Hyperkinetische Störungen

FBB‐HKS

Döpfner 2008

German Teacher’s report on ADHD symptoms

FBB‐HKS of the DISYPS

Döpfner 2000

Hyperactivity Index of the Revised Conners Parent and Teacher Rating Scales

Goyette 1978

IOWA Conners Parent Rating Scale, including abbreviated versions

IOWA CPRS

Loney 1982

IOWA Conners Teacher Rating Scale, including abbreviated versions

IOWA CTRS

Loney 1982

IOWA Conners Teacher Rating Scale, Inattention/Overactivity (I/O) and Oppositional/Defiant (O/D) subscales

IOWA‐I/O and O/D subscales

Loney 1982

IOWA Inattention/Overactivity and Aggression/Noncompliance scales ‐ Parent and Teacher rating

IOWA

Loney 1982

Lehrer‐Fragenbogen von Steinhausen

LF

Steinhausen 1993

Loney’s Time on Task Scale, Hyperactivity, Attention and Aggression subscales

TOTS

Fitzpatrick 1992b

Modified Conner Scale Parent and Teacher

ACR

Conners 1997a

Mothers’ Objective Method for Subgrouping

MOMS

Loney 1984

Parent Symptom Checklist

PSC ADHD

Döpfner 2000

Parental Account of Children’s Symptoms

PACS

Chen 2006

Restricted Academic Situation Scale

RASS

Fischer 1998

Schedule for Affective Disorders and Schizophrenia

K‐SADS/ K‐SADS‐E for diagnosis

Chambers 1985

Schedule for Non‐adaptive and Adaptive Personality

SNAP

Clark 1993; Clark 1996

Swanson, Nolan, and Pelham ‐ IV SNAP‐ADHD Rating scale

SNAP‐ADHD

Swanson 1992

Swanson, Nolan, and Pelham ‐ IV SNAP‐IV (Brazilian Version)

SNAP‐IV

Clark 1993; Clark 1996

Swanson, Kotkin, Atkins, M‐Flynn, Pelham Scale (SKAMP combined, SKAMP attention, and SKAMP deportment)

SKAMP (SKAMP combined, SKAMP attention, and SKAMP deportment)

Wigal 1998; Murray 2009

Teacher Self‐control Rating Scale

SCRS

Kendall 1979

Turgay ‐ DSM‐IV Scale, Parent

T‐DSM‐IV Scale, Parent

Turgay 1994; Ercan 2001

Turgay ‐ DSM‐IV Scale, Teacher

T‐DSM‐IV Scale, Teacher

Turgay 1994; Ercan 2001

Teacher Hyperactivity Index

THI

Achenbach 1991b

Teacher Symptom Checklist

TSC

Döpfner 2000

Vanderbilt ADHD Rating Scale

VADP(T)RS

Wolraich 2003

Wender Utah Rating Scale

WURS

Ward 1993

Wide Range Achievement Test

WRAT‐4

Wilkinson 2006

Wide Range Achievement Test Revised

WRAT‐R

Woodcock 2001

ADD/H: Attention deficit disorder with hyperactivity.
ADHD: Attention deficit hyperactivity disorder.
DSM‐IV: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition.
ODD: Oppositional defiant disorder.

Figuras y tablas -
Table 1. ADHD symptoms ‐ rating scales
Table 2. Table 2: General behaviour rating scales

Name of scale

Abbreviation

Reference

Achenbach Child Behaviour Checklist

CBCL

Achenbach 1991a

Achenbach’s Teacher Report

ATRF

Achenbach 1991b; Achenbach 2001

ADHD Rating Scale

ADHD‐RS

DuPaul 1991a

ADHD School Observation Code

ADHD‐SOC

Gadow 1996

Barkley Scales, Disruptive Behavior Disorders Rating Scale

Barkley 1991a

Child Attention Problems Scale

CAP

Barkley 1991

Child Attention Profile

CAP

Barkley 1988b

Child Behavior Checklist

CBCL

Achenbach 1991a

Child Health Questionnaire

CHQ

Landgraf 1998

Child and Adolescent Psychiatric Assessment, selected items

CAPA

Angold 1995

Children’s Psychiatric Rating Scale

CPRS

Fish 1985

Classroom Observation Code (Abikoff Classroom Observational System)

COC

Abikoff 1980

Code for Observing Social Activity

COSA

Sprafkin 1986

Conners' Child Behavior Scale

UC‐CCBS

Ladd 1996

Conners' Global Index Scale

CGI‐S

Conners 1998a

Conners’ Global Index ‐ Parent

CGI‐P

Conners 1997a

Conners' Global Index ‐ Teacher

CGI‐T

Conners 1998a

Conners', Loney and Milich Scale

CLAM

Milich 1980

Conners’ Parent Questionnaire

CPQ

Conners 1995

Conners’ Parent Rating Scale

CPRS

Conners 1998b

Conners’ Teacher Rating Scale

CTRS

Conners 1998a

Conners’ Teacher Rating Conduct Problems

Miller 1997

Disruptive Behavior Disorders Rating Scale, Parent‐ and Teacher‐Rated

DBS

Mendelsohn 1978

Disruptive Behavior Disorders Rating Scale

DBD

Silva 2005b

Groninger Behaviour Observation Scale

GOO and GBO

Van der Meere 1999b

Groninger Behaviour Checklists, Parent and Teacher Versions of the abbreviated Groninger

GGGS and GGBS

Van der Meere 1999b

Hillside Behavior Rating Scale

HBRS

Gittleman‐Klein 1976

Home Situations Questionnaire

HSQ

Barkley 1987

Home Situations Questionnaire ‐ Revised

HSQ‐R

DuPaul 1992

Humphrey’s Teacher Self‐Control Rating Scale

TSCRS

Humphrey 1982

Hyperactivity Index from the Conners Revised Teacher Rating Scale

CTRS‐R‐Hyperactivity Index

Goyette 1978

Impairment Rating Scale

IRS

Fabiano 2006

Inpatient Global Rating Scale, Revised

IGRS

Conners 1985

Inpatient Global Rating Scale, Somatic factor

IGRS‐S

Conners 1985

IOWA Conners' Rating Scale, Oppositional/Defiant (O/D) subscales

IOWA‐O/D subscales

Loney 1982

Nisonger Child Behavior Rating Form

NCBRF

Aman 1996

Paired Associates Learning

PAL

Wechsler 1945

Parent Global Assessment for Improvement

PGA

McGough 2006a

Peer Conflict Scale

PCS

Marsee 2007

Personality Inventory for Children

PIC

Lachar 1986

School Situations Questionnaire

SSQ

Barkley 1987

School Situations Questionnaire ‐ Revised

SSQ‐R

DuPaul 1992

Schedule for Nonadaptive and Adaptive Personality

SNAP

Clark 1993; Clark 1996

Strengths and Weaknesses of ADHD Symptoms and Normal Behavior Scale, Parent and Teacher

SWAN

Swanson 2006; Polderman 2007

Subjective Treatment Emergent Symptom Scale

STESS‐R

Guy 1976

Swanson, Nolan and Pelham, Fourth Edition

SNAP‐IV

Bussing 2008

Teachers Report Form

TRF

Achenbach 1991b

Telephone Interview Probe (Parent and Teacher)

TIP

Corkum 2007

Vanderbilt ADHD rating scales: Vanderbilt ADHD Diagnostic Parent Rating Scale and Vanderbilt ADHD Diagnostic Teacher Rating Scale

VADPRS and VADTRS

Wolraich 2003

Wahler, House and Stambaugh’s Ecobehavioral Assessment System

ECO

Wahler 1976

The Weekly Parent Ratings of Evening and Morning Behaviour

WREMB‐R

Kelsey 2004

Werry‐Weiss‐Peters Activity Rating Scale

WWP

Routh 1978

Woodcock‐Johnson Achievement Battery

WJ‐III Ach

Woodcock 2001

ADHD: attention deficit hyperactivity disorder.

Figuras y tablas -
Table 2. Table 2: General behaviour rating scales
Table 3. Table 3: Quality of life ratings scales

Name of scale

Abbreviation

Reference

ADHD Impact Module‐Child

AIM‐C

AIM‐C 2013

Child Impact Scale and Home Impact Scale

CIS/HIS

Landgraf 2002

Child Health and Illness Profile, Child Edition: Parent Report Form

CHIP‐CE:PRF

Riley 2004

Child Health Questionnaire

CHQ‐P

Landgraf 1998

Children's Global Assessment Scale

CGAS

Shaffer 1983

Comprehensive Psychopathological Rating Scale

CPRS

Aasberg 1978

Health Utilities Index ‐ 2

HUI‐2

Torrance 1982

ADHD: attention deficit hyperactivity disorder.

Figuras y tablas -
Table 3. Table 3: Quality of life ratings scales
Comparison 1. Teacher‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

19

1698

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

‐0.77 [‐0.90, ‐0.64]

2 Subgroup analysis: types of scales Show forest plot

19

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

Subtotals only

2.1 Conners' Teacher Rating Scale (CTRS)

8

518

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

‐0.64 [‐0.82, ‐0.47]

2.2 Abbreviated Conners' Rating Scale (ACRS) ‐ Teacher

2

105

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

‐0.75 [‐1.79, 0.29]

2.3 Conners' Abbreviated Symptom Questionnaire for Teachers (ASQ‐Teacher)

1

59

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

‐0.28 [‐0.79, 0.23]

2.4 IOWA Conners' Teacher Rating Scale (IOWA CTRS) ‐ hyperactivity

2

193

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

‐1.08 [‐1.39, ‐0.77]

2.5 Schedule for Non‐adaptive and Adaptive Personality (SNAP) ‐ Teacher

2

328

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

‐0.61 [‐0.96, ‐0.25]

2.6 Teacher ratings of attention

1

20

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

‐0.55 [‐1.45, 0.35]

2.7 Teacher ratings of impulsivity

1

20

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

0.04 [‐0.83, 0.92]

2.8 IOWA Conners' Teacher Rating Scale ‐ Inattention/Overactivity (IOWA‐I/O)

2

197

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

‐1.03 [‐1.36, ‐0.69]

2.9 Fremdbeurteilungsbogen für Hyperkinetische Störungen (FBB‐HKS)

1

85

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

‐1.06 [‐1.52, ‐0.61]

2.10 Conners’ ADHD/DSM‐IV Scales ‐ Teacher (CADS‐T)

2

254

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

‐1.05 [‐1.31, ‐0.78]

2.11 Strengths and Weaknesses of ADHD Symptoms and Normal Behaviour (SWAN) Scale

1

64

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

‐0.33 [‐0.82, 0.17]

3 Medication status: medication naive versus not medication naive Show forest plot

6

717

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

‐0.79 [‐1.08, ‐0.50]

3.1 Medication naive

4

431

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

‐0.63 [‐0.94, ‐0.31]

3.2 Not medication naive

2

286

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

‐1.06 [‐1.33, ‐0.79]

4 Subgroup analysis: duration of treatment Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

4.1 Short term (up to 6 months)

18

1445

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

‐0.81 [‐0.94, ‐0.68]

4.2 Long term (over 6 months)

1

253

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

‐0.47 [‐0.72, ‐0.22]

5 Subgroup analysis: dose Show forest plot

19

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

Subtotals only

5.1 Low dose

8

493

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

‐0.64 [‐0.82, ‐0.46]

5.2 High dose

7

688

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

‐0.81 [‐1.08, ‐0.54]

5.3 Unknown dose

6

669

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

‐0.87 [‐1.06, ‐0.68]

6 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

6.1 Parallel‐group trials

17

1506

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

‐0.80 [‐0.95, ‐0.65]

6.2 First‐period cross‐over trials

2

192

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

‐0.58 [‐0.87, ‐0.29]

7 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

19

1698

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

‐0.77 [‐0.90, ‐0.64]

7.1 Trials with cohort selection bias of all participants

7

994

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

‐0.79 [‐1.01, ‐0.57]

7.2 Trials without cohort selection bias of all participants

12

704

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

‐0.76 [‐0.93, ‐0.59]

8 ADHD symptoms, cross‐over trial (endpoint data) Show forest plot

59

5145

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

‐0.93 [‐1.06, ‐0.80]

8.1 Low risk of bias

4

204

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

‐0.64 [‐0.97, ‐0.30]

8.2 High risk of bias

55

4941

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

‐0.95 [‐1.09, ‐0.82]

9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

59

6821

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

‐0.85 [‐0.96, ‐0.74]

9.1 Low dose

42

3408

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

‐0.73 [‐0.89, ‐0.57]

9.2 High dose

36

3413

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

‐0.98 [‐1.13, ‐0.84]

10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

75

6344

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

‐0.91 [‐1.01, ‐0.80]

10.1 All parallel‐group trials and first‐period cross‐over trials

19

1698

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

‐0.77 [‐0.90, ‐0.64]

10.2 Cross‐over trials (endpoint data)

56

4646

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

‐0.95 [‐1.09, ‐0.82]

11 All parallel‐group trials and cross‐over trials: risk of bias Show forest plot

75

6344

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

‐0.91 [‐1.01, ‐0.80]

11.1 Low risk of bias

4

204

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

‐0.64 [‐0.97, ‐0.30]

11.2 High risk of bias

71

6140

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

‐0.92 [‐1.03, ‐0.81]

Figuras y tablas -
Comparison 1. Teacher‐rated ADHD symptoms
Comparison 2. Independent assessor‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

10

1907

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

‐0.64 [‐0.89, ‐0.39]

2 Subgroup analysis: types of scales Show forest plot

10

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

Subtotals only

2.1 Swanson, Kotkin, Agler, M‐Glynn and Pelham (SKAMP) Scale

1

164

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

‐0.72 [‐1.04, ‐0.41]

2.2 ADHD Rating Scale (ADHD‐RS )

7

1442

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

‐0.71 [‐1.09, ‐0.32]

2.3 Swanson, Nolan and Pelham (SNAP) Scale

1

221

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

‐0.35 [‐0.61, ‐0.08]

2.4 Unknown

1

78

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

‐0.94 [‐1.41, ‐0.47]

3 Subgroup analysis: duration of treatment Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

3.1 Short term (up to 6 months)

9

1686

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

‐0.68 [‐0.95, ‐0.40]

3.2 Long term (over 6 months)

1

221

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

‐0.35 [‐0.61, ‐0.08]

4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

4.1 Parallel‐group trials

7

1609

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

‐0.60 [‐0.91, ‐0.28]

4.2 First‐period cross‐over trials

3

298

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

‐0.76 [‐0.99, ‐0.52]

5 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

5.1 Trials with cohort selection bias of all participants

4

630

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

‐0.51 [‐0.73, ‐0.29]

5.2 Trials without cohort selection bias of all participants

6

1277

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

‐0.72 [‐1.11, ‐0.33]

6 Subgroup analysis: dose Show forest plot

10

1907

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

‐0.64 [‐0.89, ‐0.39]

6.1 Low dose

0

0

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

0.0 [0.0, 0.0]

6.2 High dose

6

1380

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

‐0.55 [‐0.91, ‐0.20]

6.3 Unknown dose

4

527

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

‐0.80 [‐0.98, ‐0.61]

7 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: risk of bias Show forest plot

19

2471

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

1.00 [‐1.16, ‐0.84]

7.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

7.2 High risk of bias

19

2471

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

1.00 [‐1.16, ‐0.84]

8 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

19

3874

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

‐0.89 [‐1.02, ‐0.75]

8.1 Low dose

16

2021

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

‐0.69 [‐0.82, ‐0.55]

8.2 High dose

12

1853

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

‐1.13 [‐1.31, ‐0.95]

9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

28

4215

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

‐0.83 [‐0.98, ‐0.67]

9.1 All parallel‐group trials and first‐period cross‐over trials

10

1907

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

‐0.64 [‐0.89, ‐0.39]

9.2 Cross‐over trials (endpoint data)

18

2308

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

‐0.95 [‐1.13, ‐0.77]

Figuras y tablas -
Comparison 2. Independent assessor‐rated ADHD symptoms
Comparison 3. Parent‐rated ADHD symptoms

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

21

2187

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

‐0.66 [‐0.82, ‐0.51]

2 Subgroup analysis: types of scales Show forest plot

21

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

Subtotals only

2.1 Conners' Parent Rating Scale (CPRS)

7

751

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

‐0.58 [‐0.87, ‐0.30]

2.2 ADHD Rating Scale ‐ Fourth Edition (ADHD‐RS‐IV)

2

194

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

‐0.30 [‐0.58, ‐0.02]

2.3 Fremdbeurteilungsbogen für Hyperkinetische Störungen (FBB‐HKS)

1

85

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

‐0.91 [‐1.36, ‐0.46]

2.4 Conners’ ADHD/DSM‐IV Scales ‐ Parent (CADS‐P)

1

120

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

‐1.26 [‐1.65, ‐0.86]

2.5 CADS‐P Inattentive subscale

1

109

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

‐0.78 [‐1.17, ‐0.39]

2.6 CADS‐P Hyperactivity subscale

1

109

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

‐0.93 [‐1.32, ‐0.53]

2.7 Clinican's Manual for the Assesment of Disruptive Behavior Disorders Rating Scale for Parents (Barkley)

1

41

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

‐0.21 [‐0.82, 0.41]

2.8 Abbreviated Conners' Rating Scale (ACRS) ‐ Parent

2

121

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

‐0.62 [‐0.99, ‐0.25]

2.9 Swanson, Nolan, and Pelham, Fourth Edition ‐ Parent (SNAP‐IV‐Parent) Scale

4

430

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

‐0.60 [‐0.79, ‐0.40]

2.10 Strengths and Weaknesses of ADHD Symptoms and Normal Behavior (SWAN) Scale

1

86

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

‐0.43 [‐0.86, 0.00]

2.11 IOWA Conners' Rating Scale ‐ Inattention/Overactivity (IOWA‐I/O)

3

352

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

‐0.78 [‐1.35, ‐0.21]

3 Subgroup analysis: duration of treatment Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

3.1 Short term (up to 6 months)

20

1925

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

‐0.67 [‐0.84, ‐0.50]

3.2 Long term (over 6 months)

1

262

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

‐0.58 [‐0.82, ‐0.33]

4 Subgroup analysis: dose Show forest plot

21

2335

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

‐0.64 [‐0.79, ‐0.48]

4.1 Low dose

5

329

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

‐0.53 [1.00, ‐0.07]

4.2 High dose

10

1132

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

‐0.60 [‐0.86, ‐0.33]

4.3 Unknown dose

8

874

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

‐0.72 [‐0.92, ‐0.52]

5 Medication status: medication naive versus not medication naive Show forest plot

7

795

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

‐0.76 [‐1.05, ‐0.48]

5.1 Medication naive

4

492

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

‐0.69 [‐1.03, ‐0.35]

5.2 Not medication naive

3

303

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

‐0.88 [‐1.33, ‐0.42]

6 Subgroup analysis: trials with cohort selection bias of all participants compared with trials without cohort selection bias of all participants Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

6.1 Trials with selection bias of all participants

10

1559

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

‐0.68 [‐0.86, ‐0.51]

6.2 Trials without cohort selection bias of all participants

11

628

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

‐0.62 [‐0.90, ‐0.33]

7 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

21

2187

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

‐0.66 [‐0.82, ‐0.51]

7.1 Parallel‐group trials

19

2094

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

‐0.66 [‐0.83, ‐0.50]

7.2 First‐period cross‐over trials

2

93

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

‐0.65 [‐1.07, ‐0.23]

8 ADHD symptoms, cross‐over trials (endpoint data) Show forest plot

41

3734

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

‐0.78 [‐0.90, ‐0.67]

8.1 Low risk of bias

4

204

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

‐0.54 [‐0.96, ‐0.13]

8.2 High risk of bias

37

3530

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

‐0.81 [‐0.92, ‐0.69]

9 ADHD symptoms, cross‐over trials (endpoint data), subgroup analysis: dose Show forest plot

41

4918

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

‐0.69 [‐0.79, ‐0.58]

9.1 Low dose

26

2272

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

‐0.65 [‐0.82, ‐0.48]

9.2 High dose

28

2646

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

‐0.72 [‐0.84, ‐0.60]

10 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials compared with cross‐over trials (endpoint data) Show forest plot

59

5861

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

‐0.74 [‐0.83, ‐0.65]

10.1 All parallel‐group trials and first‐period cross‐over trials

21

2215

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

‐0.66 [‐0.82, ‐0.51]

10.2 Cross‐over trials (endpoint data)

39

3646

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

‐0.79 [‐0.90, ‐0.67]

Figuras y tablas -
Comparison 3. Parent‐rated ADHD symptoms
Comparison 4. Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparision of raters Show forest plot

31

5697

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

‐0.69 [‐0.79, ‐0.59]

1.1 Teacher‐rated

19

1689

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

‐0.78 [‐0.93, ‐0.63]

1.2 Independent assessor‐rated

9

1829

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

‐0.61 [‐0.87, ‐0.35]

1.3 Parent‐rated

21

2179

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

‐0.65 [‐0.81, ‐0.50]

2 Age Show forest plot

6

1039

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

‐0.44 [‐0.74, ‐0.14]

2.1 2 to 6 years

1

64

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

‐0.33 [‐0.82, 0.17]

2.2 7 to 11 years

2

278

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

‐0.59 [‐1.03, ‐0.15]

2.3 12 to 18 years

3

697

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

‐0.38 [‐0.88, 0.12]

3 Comorbidity versus no comorbidity Show forest plot

20

2310

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

‐0.72 [‐0.91, ‐0.53]

3.1 ADHD with comorbidity

18

1981

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

‐0.72 [‐0.94, ‐0.51]

3.2 ADHD without comorbidity

2

329

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

‐0.69 [‐0.92, ‐0.46]

4 Subtypes ADHD: ADHD Rating Scale (parent‐, teacher‐ or independent assessor‐rated) Show forest plot

2

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

Subtotals only

4.1 Combined ADHD

2

559

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

0.65 [‐1.30, 2.60]

4.2 Inattentive ADHD

1

204

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

‐1.31 [‐1.61, ‐1.01]

5 Cross‐over trials: first‐period data versus endpoint data (parent‐, independent assessor‐ and teacher‐rated) Show forest plot

4

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

Subtotals only

5.1 First‐period data

4

372

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

‐0.64 [‐0.85, ‐0.44]

5.2 Endpoint data

4

372

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

‐0.91 [‐1.18, ‐0.65]

Figuras y tablas -
Comparison 4. Additional subgroup analyses of ADHD symptoms: parallel‐group trials and first‐period cross‐over trials
Comparison 5. Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of serious adverse events (SAE) Show forest plot

9

1532

Risk Ratio (IV, Random, 95% CI)

0.98 [0.44, 2.22]

2 Nervous system Show forest plot

6

2280

Risk Ratio (IV, Random, 95% CI)

0.87 [0.30, 2.53]

2.1 Aggression

1

303

Risk Ratio (IV, Random, 95% CI)

0.50 [0.05, 5.49]

2.2 Concussion

1

303

Risk Ratio (IV, Random, 95% CI)

0.34 [0.01, 8.17]

2.3 Loss of consciousness

1

221

Risk Ratio (IV, Random, 95% CI)

0.33 [0.01, 8.02]

2.4 Psychosis

4

712

Risk Ratio (IV, Random, 95% CI)

1.78 [0.19, 16.96]

2.5 Syncope

3

741

Risk Ratio (IV, Random, 95% CI)

1.39 [0.23, 8.47]

3 Digestive system: gastrointestinal disorders Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

4 Urinary system: kidney infection Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

5 Circulatory and respiratory systems: asthma Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

6 Immune system: cyst rupture Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Subtotals only

7 Other: drug toxicity Show forest plot

1

303

Risk Ratio (IV, Random, 95% CI)

0.34 [0.01, 8.17]

Figuras y tablas -
Comparison 5. Number of serious adverse events: parallel‐group trials and first‐period cross‐over trials
Comparison 6. Number of serious adverse events: cross‐over trials (endpoint data)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of serious adverse events (SAE) Show forest plot

8

1721

Risk Ratio (IV, Random, 95% CI)

1.62 [0.34, 7.71]

2 Hallucinations/psychosis Show forest plot

4

187

Risk Ratio (IV, Random, 95% CI)

1.10 [0.18, 6.72]

Figuras y tablas -
Comparison 6. Number of serious adverse events: cross‐over trials (endpoint data)
Comparison 7. Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of non‐serious adverse events Show forest plot

21

3132

Risk Ratio (IV, Random, 95% CI)

1.29 [1.10, 1.51]

2 Subgroup analysis: total number of non‐serious adverse events according to dose Show forest plot

21

3135

Risk Ratio (IV, Random, 95% CI)

1.28 [1.11, 1.49]

2.1 Low dose

2

151

Risk Ratio (IV, Random, 95% CI)

1.09 [0.82, 1.46]

2.2 High dose

10

1761

Risk Ratio (IV, Random, 95% CI)

1.22 [1.10, 1.35]

2.3 Unknown dose

10

1223

Risk Ratio (IV, Random, 95% CI)

1.37 [1.01, 1.87]

3 Nervous system Show forest plot

21

Risk Ratio (Random, 95% CI)

Subtotals only

3.1 Affective

4

390

Risk Ratio (Random, 95% CI)

2.39 [0.48, 11.96]

3.2 Aggression

2

417

Risk Ratio (Random, 95% CI)

1.16 [0.17, 7.80]

3.3 Apathy

1

59

Risk Ratio (Random, 95% CI)

0.80 [0.19, 3.33]

3.4 Confusion

2

548

Risk Ratio (Random, 95% CI)

1.01 [0.22, 4.73]

3.5 Depression

1

59

Risk Ratio (Random, 95% CI)

0.83 [0.22, 3.10]

3.6 Dizziness

3

683

Risk Ratio (Random, 95% CI)

2.50 [0.70, 8.99]

3.7 Drowsiness

4

811

Risk Ratio (Random, 95% CI)

1.27 [0.82, 1.98]

3.8 Emotional lability

1

132

Risk Ratio (Random, 95% CI)

2.41 [0.27, 21.32]

3.9 Fatigue

7

858

Risk Ratio (Random, 95% CI)

0.76 [0.36, 1.63]

3.10 Headache

17

2724

Risk Ratio (Random, 95% CI)

1.22 [0.90, 1.64]

3.11 Insomnia

3

349

Risk Ratio (Random, 95% CI)

1.31 [0.35, 4.93]

3.12 Irritability

11

1721

Risk Ratio (Random, 95% CI)

1.11 [0.77, 1.60]

3.13 Nervousness

2

362

Risk Ratio (Random, 95% CI)

2.52 [0.82, 7.76]

3.14 Pain

1

132

Risk Ratio (Random, 95% CI)

1.91 [0.21, 17.60]

3.15 Picking at skin or fingers, nail biting, lip or cheek chewing

1

316

Risk Ratio (Random, 95% CI)

1.01 [0.60, 1.70]

3.16 Sad, tearful or depressed

4

707

Risk Ratio (Random, 95% CI)

1.41 [0.86, 2.29]

3.17 Somnolence

2

173

Risk Ratio (Random, 95% CI)

0.59 [0.11, 3.11]

3.18 Trouble sleeping or sleep problems

13

2416

Risk Ratio (Random, 95% CI)

1.60 [1.15, 2.23]

3.19 Tics or nervous movements

8

1231

Risk Ratio (Random, 95% CI)

0.85 [0.26, 2.79]

3.20 Worried or anxious

3

596

Risk Ratio (Random, 95% CI)

1.37 [0.84, 2.25]

4 Digestive system Show forest plot

18

Risk Ratio (IV, Random, 95% CI)

Subtotals only

4.1 Decreased appetite

16

2962

Risk Ratio (IV, Random, 95% CI)

3.66 [2.56, 5.23]

4.2 Decreased weight

6

859

Risk Ratio (IV, Random, 95% CI)

3.89 [1.43, 10.59]

4.3 Diarrhoea

5

857

Risk Ratio (IV, Random, 95% CI)

1.07 [0.41, 2.74]

4.4 Dyspepsia

2

159

Risk Ratio (IV, Random, 95% CI)

1.80 [0.71, 4.54]

4.5 Increased appetite

1

179

Risk Ratio (IV, Random, 95% CI)

0.07 [0.00, 1.43]

4.6 Nausea

11

1995

Risk Ratio (IV, Random, 95% CI)

1.30 [0.85, 1.99]

4.7 Stomachache

13

2341

Risk Ratio (IV, Random, 95% CI)

1.30 [1.00, 1.69]

4.8 Vomiting

11

1916

Risk Ratio (IV, Random, 95% CI)

1.17 [0.76, 1.79]

5 Circulatory and respiratory systems Show forest plot

8

Risk Ratio (Random, 95% CI)

Subtotals only

5.1 ECG: prolonged QT‐interval

2

466

Risk Ratio (Random, 95% CI)

0.81 [0.13, 5.00]

5.2 ECG: tachycardia

1

245

Risk Ratio (Random, 95% CI)

1.04 [0.11, 10.18]

5.3 Cough

4

996

Risk Ratio (Random, 95% CI)

0.95 [0.41, 2.18]

5.4 Nasal congestion

2

479

Risk Ratio (Random, 95% CI)

1.19 [0.59, 2.41]

5.5 Pharyngolaryngeal pain

1

303

Risk Ratio (Random, 95% CI)

1.12 [0.59, 2.13]

5.6 Supraventricular extrasystoles

1

17

Risk Ratio (Random, 95% CI)

3.00 [0.11, 84.55]

5.7 Upper respiratory tract infection

1

217

Risk Ratio (Random, 95% CI)

1.07 [0.42, 2.76]

6 Skeletal and muscular systems Show forest plot

2

Risk Ratio (IV, Random, 95% CI)

Subtotals only

6.1 Arthralgia

1

303

Risk Ratio (IV, Random, 95% CI)

0.67 [0.24, 1.84]

6.2 Asthenia

1

177

Risk Ratio (IV, Random, 95% CI)

0.21 [0.01, 4.25]

6.3 Back pain

1

303

Risk Ratio (IV, Random, 95% CI)

0.81 [0.39, 1.66]

6.4 Myalgia

1

303

Risk Ratio (IV, Random, 95% CI)

0.60 [0.23, 1.62]

6.5 Toothache

1

303

Risk Ratio (IV, Random, 95% CI)

1.01 [0.43, 2.35]

7 Immune system Show forest plot

7

Risk Ratio (IV, Random, 95% CI)

Subtotals only

7.1 Gastroenteritis

3

435

Risk Ratio (IV, Random, 95% CI)

4.63 [0.99, 21.72]

7.2 Influenza

3

624

Risk Ratio (IV, Random, 95% CI)

0.65 [0.20, 2.10]

7.3 Nasopharyngitis

5

979

Risk Ratio (IV, Random, 95% CI)

1.15 [0.70, 1.87]

7.4 Otitis media

1

100

Risk Ratio (IV, Random, 95% CI)

1.77 [0.17, 18.94]

7.5 Pharyngitis

2

293

Risk Ratio (IV, Random, 95% CI)

2.43 [0.49, 12.05]

7.6 Pyrexia

2

400

Risk Ratio (IV, Random, 95% CI)

1.02 [0.01, 87.72]

7.7 Rhinitis

1

132

Risk Ratio (IV, Random, 95% CI)

1.28 [0.43, 3.79]

7.8 Upper respiratory tract infection ‐ not otherwise specified (NOS)

5

917

Risk Ratio (IV, Random, 95% CI)

1.19 [0.68, 2.06]

7.9 Viral infection NOS

3

614

Risk Ratio (IV, Random, 95% CI)

0.70 [0.23, 2.15]

8 Height Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

9 Weight Show forest plot

5

805

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

‐1.12 [‐1.55, ‐0.70]

10 BMI Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

11 Vital signs Show forest plot

9

3374

Mean Difference (IV, Random, 95% CI)

1.41 [0.30, 2.52]

11.1 Diastolic blood pressure (mmHg)

8

1067

Mean Difference (IV, Random, 95% CI)

0.94 [‐0.12, 2.01]

11.2 Systolic blood pressure (mmHg)

8

1067

Mean Difference (IV, Random, 95% CI)

‐0.05 [‐1.25, 1.16]

11.3 Pulse or heart rate (bpm)

8

1240

Mean Difference (IV, Random, 95% CI)

3.41 [0.87, 5.94]

12 Other Show forest plot

5

1815

Risk Ratio (IV, Random, 95% CI)

1.20 [0.56, 2.57]

12.1 Accidental injury

3

656

Risk Ratio (IV, Random, 95% CI)

0.99 [0.48, 2.07]

12.2 Epistasis

1

132

Risk Ratio (IV, Random, 95% CI)

4.25 [0.23, 77.22]

12.3 Excoriation

1

303

Risk Ratio (IV, Random, 95% CI)

3.52 [1.19, 10.46]

12.4 Overdose

1

221

Risk Ratio (IV, Random, 95% CI)

2.97 [0.12, 72.20]

12.5 Skin disorder (rash)

2

200

Risk Ratio (IV, Random, 95% CI)

0.52 [0.01, 26.44]

12.6 Skin laceration

1

303

Risk Ratio (IV, Random, 95% CI)

0.42 [0.15, 1.16]

Figuras y tablas -
Comparison 7. Number of non‐serious adverse events: parallel‐group trials and first‐period cross‐over trials
Comparison 8. Number of non‐serious adverse events: cross‐over trials (endpoint data)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of non‐serious adverse events Show forest plot

21

2072

Risk Ratio (Random, 95% CI)

1.33 [1.11, 1.58]

2 Subgroup analysis: total number of non‐serious adverse events according to dose Show forest plot

21

2859

Risk Ratio (Random, 95% CI)

1.26 [1.11, 1.44]

2.1 Low dose

16

1539

Risk Ratio (Random, 95% CI)

1.11 [0.94, 1.31]

2.2 High dose

12

1080

Risk Ratio (Random, 95% CI)

1.57 [1.22, 2.01]

2.3 Unknown dose

2

240

Risk Ratio (Random, 95% CI)

0.98 [0.51, 1.86]

3 Nervous system Show forest plot

50

Risk Ratio (Random, 95% CI)

Subtotals only

3.1 Aggression

2

589

Risk Ratio (Random, 95% CI)

0.52 [0.17, 1.60]

3.2 Agitation

1

62

Risk Ratio (Random, 95% CI)

1.18 [0.38, 3.60]

3.3 Anger

3

264

Risk Ratio (Random, 95% CI)

0.45 [0.26, 0.77]

3.4 Behavioural complaints

1

82

Risk Ratio (Random, 95% CI)

0.55 [0.35, 0.86]

3.5 Buccal or lingual movements

4

302

Risk Ratio (Random, 95% CI)

1.06 [0.62, 1.79]

3.6 Compulsive acts

1

90

Risk Ratio (Random, 95% CI)

2.57 [1.45, 4.56]

3.7 Daydreaming

3

222

Risk Ratio (Random, 95% CI)

0.66 [0.44, 0.98]

3.8 Dizziness

9

746

Risk Ratio (Random, 95% CI)

1.17 [0.89, 1.55]

3.9 Drowsiness: dull, tired, listless or sleepy

21

1350

Risk Ratio (Random, 95% CI)

0.97 [0.73, 1.28]

3.10 Euphoria

6

405

Risk Ratio (Random, 95% CI)

1.06 [0.72, 1.57]

3.11 Headache

37

3752

Risk Ratio (Random, 95% CI)

1.21 [1.01, 1.45]

3.12 Insomnia or sleep problems

31

3270

Risk Ratio (Random, 95% CI)

1.57 [1.20, 2.06]

3.13 Irritability

23

2238

Risk Ratio (Random, 95% CI)

0.92 [0.66, 1.27]

3.14 Nightmares

10

686

Risk Ratio (Random, 95% CI)

0.97 [0.66, 1.42]

3.15 Overly meticulous

1

96

Risk Ratio (Random, 95% CI)

40.77 [2.35, 706.72]

3.16 Obsessive thinking

1

90

Risk Ratio (Random, 95% CI)

2.35 [1.53, 3.62]

3.17 Picking at skin or fingers, nail biting, lip or cheek chewing

15

888

Risk Ratio (Random, 95% CI)

1.12 [0.88, 1.41]

3.18 Repetitive language

1

48

Risk Ratio (Random, 95% CI)

1.0 [0.32, 3.10]

3.19 Sad, tearful or depressed

23

1849

Risk Ratio (Random, 95% CI)

1.15 [0.94, 1.41]

3.20 Socially withdrawn ‐ decreased interaction with others

12

771

Risk Ratio (Random, 95% CI)

1.24 [0.82, 1.87]

3.21 Sleep efficiency (SEF)

2

108

Risk Ratio (Random, 95% CI)

0.48 [0.02, 14.28]

3.22 Stares a lot

9

904

Risk Ratio (Random, 95% CI)

1.03 [0.75, 1.40]

3.23 Tics or nervous movements

19

1403

Risk Ratio (Random, 95% CI)

1.33 [1.03, 1.72]

3.24 Unusual blinking

1

48

Risk Ratio (Random, 95% CI)

3.13 [0.12, 80.68]

3.25 Worried or anxious

20

1673

Risk Ratio (Random, 95% CI)

0.82 [0.60, 1.12]

4 Digestive system Show forest plot

42

Risk Ratio (Random, 95% CI)

Subtotals only

4.1 Decreased appetite or loss of appetite

35

3862

Risk Ratio (Random, 95% CI)

3.04 [2.35, 3.94]

4.2 Diarrhoea

3

402

Risk Ratio (Random, 95% CI)

0.58 [0.19, 1.74]

4.3 Dry mouth

5

342

Risk Ratio (Random, 95% CI)

1.25 [0.54, 2.90]

4.4 Dyspepsia

1

62

Risk Ratio (Random, 95% CI)

0.22 [0.02, 2.14]

4.5 Nausea

9

768

Risk Ratio (Random, 95% CI)

1.52 [1.00, 2.30]

4.6 Increased appetite

1

136

Risk Ratio (Random, 95% CI)

0.20 [0.08, 0.50]

4.7 Stomachache

33

3777

Risk Ratio (Random, 95% CI)

1.61 [1.27, 2.04]

4.8 Vomiting

4

710

Risk Ratio (Random, 95% CI)

0.90 [0.26, 3.11]

5 Urinary system: urinary incontinence Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

6 Skeletal and muscular system: somatic complaints Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

7 Immune system Show forest plot

7

Risk Ratio (IV, Random, 95% CI)

Subtotals only

7.1 Allergic rhinitis

4

475

Risk Ratio (IV, Random, 95% CI)

1.38 [0.35, 5.51]

7.2 Fever

2

91

Risk Ratio (IV, Random, 95% CI)

1.39 [0.09, 20.56]

7.3 Lymphadenitis

2

296

Risk Ratio (IV, Random, 95% CI)

3.93 [0.44, 35.11]

7.4 Pharyngolaryngeal pain

1

160

Risk Ratio (IV, Random, 95% CI)

2.0 [0.19, 21.62]

7.5 Pharyngitis

4

754

Risk Ratio (IV, Random, 95% CI)

0.71 [0.19, 2.62]

7.6 Upper respiratory tract infection

5

888

Risk Ratio (IV, Random, 95% CI)

0.61 [0.27, 1.37]

8 Skin Show forest plot

3

Risk Ratio (IV, Random, 95% CI)

Subtotals only

8.1 Rash

2

208

Risk Ratio (IV, Random, 95% CI)

0.96 [0.14, 6.41]

8.2 Skin laceration

1

167

Risk Ratio (IV, Random, 95% CI)

2.96 [0.12, 71.75]

9 Vital signs Show forest plot

14

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 Diastolic blood pressure (mmHg)

11

755

Mean Difference (IV, Random, 95% CI)

1.23 [‐0.39, 2.86]

9.2 Systolic blood pressure (mmHg)

11

755

Mean Difference (IV, Random, 95% CI)

0.53 [‐1.30, 2.36]

9.3 Pulse or heart rate (bpm)

14

939

Mean Difference (IV, Random, 95% CI)

5.06 [2.88, 7.24]

10 Height (cm) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

11 Weight Show forest plot

6

530

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

‐0.06 [‐0.23, 0.11]

Figuras y tablas -
Comparison 8. Number of non‐serious adverse events: cross‐over trials (endpoint data)
Comparison 9. Teacher‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials: risk of bias Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

1.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

1.2 High risk of bias

5

668

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

‐0.87 [‐1.04, ‐0.71]

2 Subgroup analysis: types of scales Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

2.1 Conners' Global Index ‐ Teacher (CGI‐T)

1

314

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

‐0.91 [‐1.14, ‐0.68]

2.2 Groninger Behaviour Observation Scale (GBOS)

1

43

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

‐0.84 [‐1.46, ‐0.21]

2.3 Conners' Teacher Rating Scale ‐ Conduct problems

1

25

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

‐0.67 [‐1.48, 0.14]

2.4 IOWA Conners' Rating Scale ‐ Oppositional/Defiant (IOWA‐O/D)

2

286

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

‐0.86 [‐1.12, ‐0.59]

3 Subgroup analysis: dose Show forest plot

5

696

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

‐0.85 [‐1.02, ‐0.69]

3.1 Low dose

2

71

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

‐0.67 [‐1.16, ‐0.19]

3.2 High dose

3

466

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

‐0.89 [‐1.08, ‐0.70]

3.3 Unknown dose

1

159

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

‐0.84 [‐1.21, ‐0.46]

4 Subgroup analysis: duration of treatment Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

4.1 Short term (up to 6 months)

5

668

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

‐0.87 [‐1.04, ‐0.71]

4.2 Long term (over 6 months)

0

0

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

0.0 [0.0, 0.0]

5 Subgroup analysis: parallel‐group trials versus first‐period cross‐over trials Show forest plot

5

668

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

‐0.87 [‐1.04, ‐0.71]

5.1 Parallel‐group trials

5

668

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

‐0.87 [‐1.04, ‐0.71]

5.2 First‐period cross‐over trials

0

0

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

0.0 [0.0, 0.0]

6 General behaviour, cross‐over trials (endpoint data) Show forest plot

16

2014

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

‐0.69 [‐0.78, ‐0.60]

6.1 Low dose

13

1110

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

‐0.60 [‐0.72, ‐0.48]

6.2 High dose

12

904

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

‐0.82 [‐0.95, ‐0.68]

7 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

16

1308

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

‐0.75 [‐0.87, ‐0.63]

7.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

7.2 High risk of bias

16

1308

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

‐0.75 [‐0.87, ‐0.63]

8 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (teacher‐rated) versus cross‐over trials (endpoint data) Show forest plot

21

1976

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

‐0.79 [‐0.88, ‐0.70]

8.1 All parallel‐group trials and first‐period cross‐over trials

5

668

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

‐0.87 [‐1.04, ‐0.71]

8.2 Cross‐over trials (endpoint data)

16

1308

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

‐0.75 [‐0.87, ‐0.63]

Figuras y tablas -
Comparison 9. Teacher‐rated general behaviour
Comparison 10. Independent assessor‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 General behaviour, cross‐over trials (endpoint data) Show forest plot

8

1241

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

‐0.60 [‐0.75, ‐0.46]

1.1 Low dose

7

903

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

‐0.56 [‐0.76, ‐0.36]

1.2 High dose

5

338

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

‐0.71 [‐0.93, ‐0.49]

2 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

8

951

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

‐0.69 [‐0.88, ‐0.49]

2.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

2.2 High risk of bias

8

951

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

‐0.69 [‐0.88, ‐0.49]

3 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (independent assessor‐rated) compared with cross‐over trials (endpoint data) Show forest plot

8

951

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

‐0.69 [‐0.88, ‐0.49]

3.1 Parallel‐group trials and first‐period cross‐over trials

0

0

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

0.0 [0.0, 0.0]

3.2 Cross‐over trials (endpoint data)

8

951

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

‐0.69 [‐0.88, ‐0.49]

Figuras y tablas -
Comparison 10. Independent assessor‐rated general behaviour
Comparison 11. Parent‐rated general behaviour

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All parallel‐group trials and first‐period cross‐over trials Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

2 Subgroup analysis: risk of bias Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

2.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

2.2 High risk of bias

6

670

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

‐0.53 [‐0.78, ‐0.27]

3 Subgroup analysis: types of scales Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

3.1 The Weekly Parent Ratings of Evening and Morning Behaviour (WPREMB) ‐ Revised

1

17

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

0.50 [‐0.47, 1.47]

3.2 Conners' Global Index (CGI) ‐ Parent

2

352

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

‐0.41 [‐0.63, ‐0.20]

3.3 Swanson, Nolan and Pelham, Fourth Edition ‐ Oppositional (SNAP‐IV‐Oppositional)

1

15

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

‐0.86 [‐1.95, 0.23]

3.4 IOWA Conners' Rating Scale ‐ Oppositional/Defiant (IOWA‐I/O)

2

286

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

‐0.75 [‐1.01, ‐0.49]

4 Subgroup analysis: parallel‐group trials compared with first‐period cross‐over trials Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

4.1 Parallel‐group trials

5

655

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

‐0.51 [‐0.78, ‐0.23]

4.2 First‐period cross‐over trials

1

15

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

‐0.86 [‐1.95, 0.23]

5 Subgroup analysis: duration of treatment Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

5.1 Short term (up to 6 months)

6

670

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

‐0.53 [‐0.78, ‐0.27]

5.2 Long term (over 6 months)

0

0

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

0.0 [0.0, 0.0]

6 Subgroup analysis: dose Show forest plot

6

670

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

‐0.53 [‐0.78, ‐0.27]

6.1 Low dose

0

0

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

0.0 [0.0, 0.0]

6.2 High dose

4

496

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

‐0.42 [‐0.77, ‐0.08]

6.3 Unknown dose

2

174

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

‐0.73 [‐1.08, ‐0.38]

7 General behaviour, cross‐over trials (endpoint data) Show forest plot

6

550

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

‐0.75 [‐0.93, ‐0.56]

7.1 Low dose

5

248

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

‐0.65 [‐0.93, ‐0.38]

7.2 High dose

4

302

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

‐0.83 [‐1.07, ‐0.60]

8 Subgroup analysis: general behaviour, cross‐over trials (endpoint data): risk of bias Show forest plot

6

384

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

‐0.84 [‐1.05, ‐0.63]

8.1 Low risk of bias

0

0

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

0.0 [0.0, 0.0]

8.2 High risk of bias

6

384

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

‐0.84 [‐1.05, ‐0.63]

9 Subgroup analysis: all parallel‐group trials and first‐period cross‐over trials (parent‐rated) compared with cross‐over trials (endpoint data) Show forest plot

12

1054

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

‐0.68 [‐0.86, ‐0.50]

9.1 All parallel‐group trials and first‐period cross‐over trials

6

670

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

‐0.53 [‐0.78, ‐0.27]

9.2 Cross‐over trials (endpoint data)

6

384

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

‐0.84 [‐1.05, ‐0.63]

Figuras y tablas -
Comparison 11. Parent‐rated general behaviour
Comparison 12. Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisions of raters Show forest plot

8

1338

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

‐0.69 [‐0.86, ‐0.52]

1.1 Teacher‐rated

5

668

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

‐0.87 [‐1.04, ‐0.71]

1.2 Independent assessor‐rated

0

0

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

0.0 [0.0, 0.0]

1.3 Parent‐rated

6

670

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

‐0.53 [‐0.78, ‐0.27]

2 Comorbidity versus no comorbidity Show forest plot

7

579

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

‐0.70 [‐0.98, ‐0.43]

2.1 ADHD with comorbidity

6

265

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

‐0.59 [‐0.95, ‐0.23]

2.2 ADHD without comorbidity

1

314

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

‐0.91 [‐1.14, ‐0.68]

3 Cross‐over trials: first‐period data versus endpoint data (teacher‐, parent‐, and independent assessor‐rated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 First‐period data

1

16

Mean Difference (IV, Random, 95% CI)

‐0.81 [‐1.75, 0.13]

3.2 Endpoint data

1

14

Mean Difference (IV, Random, 95% CI)

0.14 [‐0.71, 1.00]

Figuras y tablas -
Comparison 12. Additional subgroup analyses of general behaviour: parallel‐group trials and first‐period cross‐over trials
Comparison 13. Quality of life: parallel‐group trials and first‐period cross‐over trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Subgroup analysis: types of scales Show forest plot

3

514

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

0.61 [0.42, 0.80]

1.1 Child Health Questionnaire (CHQ)

1

257

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

0.54 [0.25, 0.83]

1.2 Children´s Global Assessment Scale (CGAS)

1

36

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

0.79 [0.10, 1.47]

1.3 Child Health and Illness Profile, Child Edition: Parent Report Form (CHIP‐CE:PRF)

1

221

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

0.64 [0.37, 0.91]

Figuras y tablas -
Comparison 13. Quality of life: parallel‐group trials and first‐period cross‐over trials