Scolaris Content Display Scolaris Content Display

Intervenciones farmacológicas para la apatía en la enfermedad de Alzheimer

Collapse all Expand all

References

Referencias de los estudios incluidos en esta revisión

CitAD trial {published data only}

Leonpacher AK, Peters ME, Drye LT, Makino KM, Newell JA, Devanand DP, et al. Effects of citalopram on neuropsychiatric symptoms in Alzheimer's dementia: evidence from the CitAD Study. American Journal of Psychiatry 2016;173(5):473‐80. [PUBMED: 27032628]CENTRAL
Porsteinsson AP, Drye LT, Pollock BG, Devanand DP, Frangakis C, Ismail Z, et al. CitAD Research Group. Effect of citalopram on agitation in Alzheimer disease: the CitAD randomized clinical trial. The Journal of the American Medical Association 2014;311(7):682‐91. CENTRAL

De Deyn 2004 {published data only}

De Deyn PP, Carrasco MM, Deberdt W, Jeandel C, Hay DP, Feldman PD, et al. Olanzapine versus placebo in the treatment of psychosis with or without associated behavioral disturbances in patients with Alzheimer's disease. International Journal of Geriatric Psychiatry 2004;19(2):115‐26. [PUBMED: 14758577]CENTRAL

Frakey 2012 {published data only}

Frakey LL, Salloway S, Buelow M, Malloy P. A randomized, double‐blind, placebo‐controlled trial of modafinil for the treatment of apathy in individuals with mild‐to‐moderate Alzheimer's disease. Journal of Clinical Psychiatry 2012;73(6):796‐801. [PUBMED: 22687392]CENTRAL

Herrmann 2005 {published data only}

Herrmann N, Rabheru K, Wang J, Binder C. Galantamine treatment of problematic behavior in Alzheimer disease: post‐hoc analysis of pooled data from three large trials. American Journal of Geriatric Psychiatry 2005;13(6):527‐34. [PUBMED: 15956273]CENTRAL

Herrmann 2007 {published data only}

Herrmann N, Lanctôt KL, Rothenburg LS, Eryavec G. A placebo‐controlled trial of valproate for agitation and aggression in Alzheimer's disease. Dementia and Geriatric Cognitive Disorders 2007;23(2):116‐9. [PUBMED: 17148938]CENTRAL

Herrmann 2008 {published data only}

Herrmann N, Rothenburg LS, Black SE, Ryan M, Liu BA, Busto UE, et al. Methylphenidate for the treatment of apathy in Alzheimer disease: prediction of response using dextroamphetamine challenge. Journal of Clinical Psychopharmacology 2008;28(3):296‐301. [PUBMED: 18480686]CENTRAL

Herrmann 2016 {published data only}

Herrmann N, O'Regan J, Ruthirakuhan M, Kiss A, Eryavec G, Williams E, et al. A randomized placebo‐controlled discontinuation study of cholinesterase inhibitors in institutionalized patients with moderate to severe Alzheimer disease. Journal of the American Medical Directors Association 2016;17(2):142‐7. [PUBMED: 26482056]CENTRAL

Kaufer 1998 {published data only}

Kaufer D. Beyond the cholinergic hypothesis: the effect of metrifonate and other cholinesterase inhibitors on neuropsychiatric symptoms in Alzheimer's disease. Dementia and Geriatric Cognitive Disorders 1998;9 Suppl 2:8‐14. [PUBMED: 9718229]CENTRAL

Lanctôt 2002 {published data only}

Lanctôt KL, Herrmann N, Van Reekum R, Eryavec G, Naranjo CA. Gender, aggression and serotonergic function are associated with response to sertraline for behavioral disturbances in Alzheimer's disease. International Journal of Geriatric Psychiatry 2002;17(6):531‐41. [PUBMED: 12112177]CENTRAL

Morris 1998 {published data only}

Morris JC, Cyrus PA, Orazem J, Mas J, Bieber F, Ruzicka BB, et al. Metrifonate benefits cognitive, behavioral, and global function in patients with Alzheimer's disease. Neurology 1998;50(5):1222‐30. [PUBMED: 9595967]CENTRAL

MSAD trial {published data only}

Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E. A 24‐week, randomized, double‐blind study of donepezil in moderate to severe Alzheimer's disease. Neurology 2001;57(4):613‐20. [PUBMED: 11524468]CENTRAL
Feldman H, Gauthier S, Hecker J, Vellas B, Xu Y, Ieni JR, et al. Efficacy and safety of donepezil in patients with more severe Alzheimer's disease: a subgroup analysis from a randomized, placebo‐controlled trial. International Journal of Geriatric Psychiatry 2005;20(6):559‐69. [PUBMED: 15920715]CENTRAL
Gauthier S, Feldman H, Hecker J, Vellas B, Ames D, Subbiah P, et al. Efficacy of donepezil on behavioral symptoms in patients with moderate to severe Alzheimer's disease. International Psychogeriatrics 2002;14(4):389‐404. [PUBMED: 12670060]CENTRAL

Padala 2017 {published data only}

Padala PR, Padala KP, Lensing SY, Ramirez D, Monga V, Bopp MM, et al. Methylphenidate for apathy in community‐dwelling older veterans with mild Alzheimer's disease: a double‐blind, randomized, placebo‐controlled trial. American Journal of Psychiatry 2017;175:appiajp201717030316. [PUBMED: 28945120]CENTRAL

Raskind 1999 {published data only}

Raskind MA, Cyrus PA, Ruzicka BB, Gulanski BI. The effects of metrifonate on the cognitive, behavioral, and functional performance of Alzheimer's disease patients. Metrifonate Study Group. Journal of Clinical Psychiatry 1999;60(5):318‐25. [PUBMED: 10362441]CENTRAL

Rosenberg 2013 {published data only}

Rosenberg PB, Lanctôt KL, Drye LT, Herrmann N, Scherer RW, Bachman DL, et al. Safety and efficacy of methylphenidate for apathy in Alzheimer's disease: a randomized, placebo‐controlled trial. Journal of Clinical Psychiatry 2013;74(8):810‐6. [PUBMED: 24021498]CENTRAL

Ruths 2008 {published data only}

Ruths S, Straand J, Nygaard HA, Aarsland D. Stopping antipsychotic drug therapy in demented nursing home patients: a randomized, placebo‐controlled study‐‐the Bergen District Nursing Home study (BEDNURS). International Journal of Geriatric Psychiatry 2008;23(9):889‐95. [PUBMED: 18306150]CENTRAL

Semgacestat trial {published data only}

Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. New England Journal of Medicine 2013;369(4):341‐50. [PUBMED: 23883379]CENTRAL
Rosenberg PB, Lanctôt KL, Herrmann N, Mintzer JE, Porsteinsson AP, Sun X, et al. Changes in neuropsychiatric inventory associated with semagacestat treatment of Alzheimer's disease. Journal of Alzheimer's Disease 2016;54(1):373‐81. [PUBMED: 27567808]CENTRAL

Sival 2002 {published data only}

Sival RC, Haffmans PM, Jansen PA, Duursma SA, Eikelenboom P. Sodium valproate in the treatment of aggressive behavior in patients with dementia‐‐a randomized placebo controlled clinical trial. International Journal of Geriatric Psychiatry 2002;17(6):579‐85. [PUBMED: 12112183]CENTRAL

Sultzer 2008 {published data only}

Sultzer DL, Davis SM, Tariot PN, Dagerman KS, Lebowitz BD, Lyketsos CG, et al. Clinical symptom responses to atypical antipsychotic medications in Alzheimer's disease: phase 1 outcomes from the CATIE‐AD effectiveness trial. American Journal of Psychiatry 2008;165(7):844‐54. [PUBMED: 18519523]CENTRAL

Tariot 2001 {published data only}

Tariot PN, Cummings JL, Katz IR, Mintzer J, Perdomo CA, Schwam EM, et al. A randomized, double‐blind, placebo‐controlled study of the efficacy and safety of donepezil in patients with Alzheimer's disease in the nursing home setting. Journal of the American Geriatrics Society 2001;49(12):1590‐9. [PUBMED: 11843990]CENTRAL

Tariot 2011 {published data only}

Tariot PN, Schneider LS, Cummings J, Thomas RG, Raman R, Jakimovich LJ, et al. Chronic divalproex sodium to attenuate agitation and clinical progression of Alzheimer disease. Archives of General Psychiatry 2011;68(8):853‐61. [PUBMED: 21810649]CENTRAL

Trzepacz 2013 {published data only}

Trzepacz PT, Cummings J, Konechnik T, Forrester TD, Chang C, Dennehy EB, et al. Mibampator (LY451395) randomized clinical trial for agitation/aggression in Alzheimer's disease. International Psychogeriatrics 2013;25(5):707‐19. [PUBMED: 23257314]CENTRAL

Referencias de los estudios excluidos de esta revisión

Ballard 2004 {published data only}

Ballard CG, Thomas A, Fossey J, Lee L, Jacoby R, Lana MM, et al. A 3‐month, randomized, placebo‐controlled, neuroleptic discontinuation study in 100 people with dementia: the neuropsychiatric inventory median cutoff is a predictor of clinical outcome. Journal of Clinical Psychiatry 2004;65(1):114‐9. [PUBMED: 14744180]CENTRAL

Ballard 2005 {published data only}

Ballard C, Margallo‐Lana M, Juszczak E, Douglas S, Swann A, Thomas A, et al. Quetiapine and rivastigmine and cognitive decline in Alzheimer's disease: randomised double blind placebo controlled trial. The British Medical Journal (Clinical research ed.) 2005;330(7496):874. [PUBMED: 15722369]CENTRAL

Ballard 2008 {published data only}

Ballard C, Lana MM, Theodoulou M, Douglas S, McShane R, Jacoby R, et al. A randomised, blinded, placebo‐controlled trial in dementia patients continuing or stopping neuroleptics (the DART‐AD trial). PLoS Medicine 2008;5(4):e76. [PUBMED: 18384230]CENTRAL

Banerjee 2011 {published data only}

Banerjee S, Hellier J, Dewey M, Romeo R, Ballard C, Baldwin R, et al. Sertraline or mirtazapine for depression in dementia (HTA‐SADD): a randomised, multicentre, double‐blind, placebo‐controlled trial. Lancet 2011;378(9789):403‐11. [PUBMED: 21764118]CENTRAL

Breder 2004 {published data only}

Breder C, Swanink R, Marcus R, Kostic D, Iwamoto T, Carson W, et al. Dose‐ranging study of aripiprazole in patients with dementia of Alzheimer's disease. Neurobiology of Aging 2004;25(2):S190. CENTRAL

Bridges‐Parlet 1997 {published data only}

Bridges‐Parlet S, Knopman D, Steffes S. Withdrawal of neuroleptic medications from institutionalized dementia patients: results of a double‐blind, baseline‐treatment‐controlled pilot study. Journal of Geriatric Psychiatry and Neurology 1997;10(3):119‐26. [PUBMED: 9322135]CENTRAL

Brodaty 2003 {published data only}

Brodaty H, Ames D, Snowdon J, Woodward M, Kirwan J, Clarnette R, et al. A randomized placebo‐controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementia. Journal of Clinical Psychiatry 2003;64(2):134‐43. [PUBMED: 12633121]CENTRAL

Burns 1999 {published data only}

Burns A, Rossor M, Hecker J, Gauthier S, Petit H, Moller HJ, et al. The effects of donepezil in Alzheimer's disease ‐ results from a multinational trial. Dementia and Geriatric Cognitive Disorders 1999;10(3):237‐44. [PUBMED: 10325453]CENTRAL

Cohen‐Mansfield 1999 {published data only}

Cohen‐Mansfield J, Lipson S, Werner P, Billig N, Taylor L, Woosley R. Withdrawal of haloperidol, thioridazine, and lorazepam in the nursing home: a controlled, double‐blind study. Archives of Internal Medicine 1999;159(15):1733‐40. [PUBMED: 10448776]CENTRAL

Deberdt 2005 {published data only}

Deberdt WG, Dysken MW, Rappaport SA, Feldman PD, Young CA, Hay DP, et al. Comparison of olanzapine and risperidone in the treatment of psychosis and associated behavioral disturbances in patients with dementia. American Journal of Geriatric Psychiatry 2005;13(8):722‐30. [PUBMED: 16085789]CENTRAL

De Deyn 1999 {published data only}

De Deyn PP, Rabheru K, Rasmussen A, Bocksberger JP, Dautzenberg PL, Eriksson S, et al. A randomized trial of risperidone, placebo, and haloperidol for behavioral symptoms of dementia. Neurology 1999;53(5):946‐55. [PUBMED: 10496251]CENTRAL

De Deyn 2005 {published data only}

De Deyn P, Jeste DV, Swanink R, Kostic D, Breder C, Carson WH, et al. Aripiprazole for the treatment of psychosis in patients with Alzheimer's disease: a randomized, placebo‐controlled study. Journal of Clinical Psychopharmacology 2005;25(5):463‐7. [PUBMED: 16160622]CENTRAL

Devanand 2011 {published data only}

Devanand DP, Pelton GH, Cunqueiro K, Sackeim HA, Marder K. A 6‐month, randomized, double‐blind, placebo‐controlled pilot discontinuation trial following response to haloperidol treatment of psychosis and agitation in Alzheimer's disease. International Journal of Geriatric Psychiatry 2011;26(9):937‐43. [PUBMED: 21845596]CENTRAL

Devanand 2012 {published data only}

Devanand DP, Mintzer J, Schultz SK, Andrews HF, Sultzer DL, De la Pena D, et al. Relapse risk after discontinuation of risperidone in Alzheimer's disease. New England Journal of Medicine 2012;367(16):1497‐507. [PUBMED: 23075176]CENTRAL

De Vasconcelos 2007 {published data only}

De Vasconcelos Cunha UG, Lopes Rocha F, Avila de Melo R, Alves Valle E, De Souza Neto JJ, Mendes Brega R, et al. A placebo‐controlled double‐blind randomized study of venlafaxine in the treatment of depression in dementia. Dementia and Geriatric Cognitive Disorders 2007;24(1):36‐41. [PUBMED: 17495474]CENTRAL

Findlay 1989 {published data only}

Findlay DJ, Sharma J, McEwen J, Ballinger BR, MaClennan WJ, McHarg AM. Double‐blind controlled withdrawal of thioridazine treatment in elderly female inpatients with senile dementia. International Journal of Geriatric Psychiatry 1989;4(2):115‐20. CENTRAL

Holmes 2004 {published data only}

Holmes C, Wilkinson D, Dean C, Vethanayagam S, Olivieri S, Langley A, et al. The efficacy of donepezil in the treatment of neuropsychiatric symptoms in Alzheimer disease. Neurology 2004;63(2):214‐9. [PUBMED: 15277611]CENTRAL

Howard 2012 {published data only}

Howard R, McShane R, Lindesay J, Ritchie C, Baldwin A, Barber R, et al. Donepezil and memantine for moderate‐to‐severe Alzheimer's disease. New England Journal of Medicine 2012;366(10):893‐903. [PUBMED: 22397651]CENTRAL

Johannsen 2006 {published data only}

Johannsen P, Salmon E, Hampel H, Xu Y, Richardson S, Qvitzau S, et al. Assessing therapeutic efficacy in a progressive disease: a study of donepezil in Alzheimer's disease. CNS Drugs 2006;20(4):311‐25. [PUBMED: 16599649]CENTRAL

Katz 1999 {published data only}

Katz IR, Jeste DV, Mintzer JE, Clyde C, Napolitano J, Brecher M. Comparison of risperidone and placebo for psychosis and behavioral disturbances associated with dementia: a randomized, double‐blind trial. Risperidone Study Group. Journal of Clinical Psychiatry 1999;60(2):107‐15. [PUBMED: 10084637]CENTRAL

Lyketsos 2003 {published data only}

Lyketsos CG, DelCampo L, Steinberg M, Miles Q, Steele CD, Munro C, et al. Treating depression in Alzheimer disease: efficacy and safety of sertraline therapy, and the benefits of depression reduction: the DIADS. Archives of General Psychiatry 2003;60(7):737‐46. [PUBMED: 12860778]CENTRAL

Magai 2000 {published data only}

Magai C, Kennedy G, Cohen CI, Gomberg D. A controlled clinical trial of sertraline in the treatment of depression in nursing home patients with late‐stage Alzheimer's disease. American Journal of Geriatric Psychiatry 2000;8(1):66‐74. [PUBMED: 10648297]CENTRAL

Mintzer 2006 {published data only}

Mintzer J, Greenspan A, Caers I, Van Hove I, Kushner S, Weiner M, et al. Risperidone in the treatment of psychosis of Alzheimer disease: results from a prospective clinical trial. American Journal of Geriatric Psychiatry 2006;14(3):280‐91. [PUBMED: 16505133]CENTRAL

Nyth 1992 {published data only}

Nyth AL, Gottfries CG, Lyby K, Smedegaard‐Andersen L, Gylding‐Sabroe J, Kristensen M, et al. A controlled multicenter clinical study of citalopram and placebo in elderly depressed patients with and without concomitant dementia. Acta Psychiatrica Scandinavica 1992;86(2):138‐45. [PUBMED: 1529737]CENTRAL

Petracca 1996 {published data only}

Petracca G, Teson A, Chemerinski E, Leiguarda R, Starkstein SE. A double‐blind placebo‐controlled study of clomipramine in depressed patients with Alzheimer's disease. Journal of Neuropsychiatry and Clinical Neurosciences 1996;8(3):270‐5. [PUBMED: 8854297]CENTRAL

Petracca 2001 {published data only}

Petracca GM, Chemerinski E, Starkstein SE. A double‐blind, placebo‐controlled study of fluoxetine in depressed patients with Alzheimer's disease. International Psychogeriatrics 2001;13(2):233‐40. [PUBMED: 11495397]CENTRAL

Raskind 2000 {published data only}

Raskind MA, Peskind ER, Wessel T, Yuan W. Galantamine in AD: A 6‐month randomized, placebo‐controlled trial with a 6‐month extension. The Galantamine USA‐1 Study Group. Neurology 2000;54(12):2261‐8. [PUBMED: 10881250]CENTRAL

Reifler 1989 {published data only}

Reifler BV, Teri L, Raskind M, Veith R, Barnes R, White E, et al. Double‐blind trial of imipramine in Alzheimer's disease patients with and without depression. American Journal of Psychiatry 1989;146(1):45‐9. [PUBMED: 2643356]CENTRAL

Rosenberg 2010 {published data only}

Rosenberg PB, Drye LT, Martin BK, Frangakis C, Mintzer JE, Weintraub D, et al. Sertraline for the treatment of depression in Alzheimer disease. American Journal of Geriatric Psychiatry 2010;18(2):136‐45. [PUBMED: 20087081]CENTRAL

Rosler 1999 {published data only}

Rosler M, Anand R, Cicin‐Sain A, Gauthier S, Agid Y, Dal‐Bianco P, et al. Efficacy and safety of rivastigmine in patients with Alzheimer's disease: international randomised controlled trial. The British Medical Journal (Clinical research ed.) 1999;318(7184):633‐8. [PUBMED: 10066203]CENTRAL

Roth 1996 {published data only}

Roth M, Mountjoy CQ, Amrein R. Moclobemide in elderly patients with cognitive decline and depression: an international double‐blind, placebo‐controlled trial. British Journal of Psychiatry 1996;168(2):149‐57. [PUBMED: 8837903]CENTRAL

Satterlee 1995 {published data only}

Satterlee WG, Reams SG. Burns PR, Hamilton S, Tran PV, Tollefson GD. A clinical update on olanzapine treatment in schizophrenia and in elderly Alzheimer's disease patients. Psychopharmacology Bulletin 1995;31:534. CENTRAL

Schneider 2006 {published data only}

Schneider LS, Tariot PN, Dagerman KS, Davis SM, Hsiao JK, Ismail MS, et al. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer's disease. New England Journal of Medicine 2006;355(15):1525‐38. [PUBMED: 17035647]CENTRAL

Seltzer 2004 {published data only}

Seltzer B, Zolnouni P, Nunez M, Goldman R, Kumar D, Ieni J, et al. Efficacy of donepezil in early‐stage Alzheimer disease: a randomized placebo‐controlled trial. Archives of Neurology 2004;61(12):1852‐6. [PUBMED: 15596605]CENTRAL

Street 2000 {published data only}

Street JS, Clark WS, Gannon KS, Cummings JL, Bymaster FP, Tamura RN, et al. Olanzapine treatment of psychotic and behavioral symptoms in patients with Alzheimer disease in nursing care facilities: a double‐blind, randomized, placebo‐controlled trial. The HGEU Study Group. Archives of General Psychiatry 2000;57(10):968‐76. [PUBMED: 11015815]CENTRAL

Streim 2004 {published data only}

Steim J, Breder C, Swanink R, McQuade R, Iwamoto T, Carson W, et al. Flexible‐dose aripiprazole in psychosis of Alzheimer's disease. Neurobiology of Aging 2004;25(2):S191. CENTRAL

Tariot 2004a {published data only}

Tariot PN, Profenno LA, Ismail MS. Efficacy of atypical antipsychotics in elderly patients with dementia. Journal of Clinical Psychiatry 2004;65 Suppl 11:11‐5. [PUBMED: 15264966]CENTRAL

Tariot 2004b {published data only}

Tariot P. Medication helps quell the agitation of dementia. www.urmc.rochester.edu/pr/news/story.cfm?id=5932004. CENTRAL

Van Reekum 2002 {published data only}

Van Reekum R, Clarke D, Conn D, Herrmann N, Eryavec G, Cohen T, et al. A randomized, placebo‐controlled trial of the discontinuation of long‐term antipsychotics in dementia. International Psychogeriatrics 2002;14(2):197‐210. [PUBMED: 12243210]CENTRAL

Wilcock 2000 {published data only}

Wilcock GK, Lilienfeld S, Gaens E. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer's disease: multicentre randomised controlled trial. Galantamine International‐1 Study Group. The British Medical Journal (Clinical research ed.) 2000;321(7274):1445‐9. [PUBMED: 11110737]CENTRAL

Winblad 2001 {published data only}

Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G, Wimo A, et al. A 1‐year, randomized, placebo‐controlled study of donepezil in patients with mild to moderate AD. Neurology 2001;57(3):489‐95. [PUBMED: 11502918]CENTRAL

Abe 1975

Abe T, Morita M, Kawai K, Misawa S, Kanai H, Hirose G, et al. Transmission of a t(13q22q) chromosome observed in three generations with segregation of the translocation D1‐trisomy syndrome. Humangenetik 1975;30(3):207‐15.

Albert 2013

Albert PR, Benkelfat C. The neurobiology of depression‐‐revisiting the serotonin hypothesis. II. Genetic, epigenetic and clinical studies. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 2013;368(1615):20120535.

Allard 1990

Allard P, Alafuzoff I, Carlsson A, Eriksson K, Ericson E, Gottfries CG, et al. Loss of dopamine uptake sites labeled with [3H]GBR‐12935 in Alzheimer's disease. European Neurology 1990;30(4):181‐5.

Andrade 2015

Andrade 2015. The primary outcome measure and its importance in clinical trials. Journal of Clinical Psychiatry 2015;76(10):e1320‐3.

APA 2013

American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th Edition. Arlington, VA: American Psychiatric Association, 2013.

Attems 2014

Attems J, Jellinger KA. The overlap between vascular disease and Alzheimer's disease‐‐lessons from pathology. BioMed Central Medicine 2014;12:206.

Ballard 2006

Ballard C, Waite J, Birks. Atypical antipsychotics for the treatment of aggression and psychosis in Alzheimer's disease. Cochrane Database of Systematic Reviews 2006, Issue 1. [DOI: 10.1002/14651858.CD003476.pub2; PUBMED: 16437455]

Barnhart 2004

Barnhart WJ, Makela EH, Latocha MJ. SSRI‐induced apathy syndrome: a clinical review. Journal of Psychiatric Practice 2004;10(3):196‐9.

Bartus 1982

Bartus RT, Dean RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982;217(4558):408‐14.

Berman 2012

Berman K, Brodaty H, Withall A, Seeher K. Pharmacologic treatment of apathy in dementia. American Journal of Geriatric Psychiatry 2012;20(2):104‐22. [PUBMED: 21841459]

Bhandari 2004

Bhandari M, Busse JW, Jackowski D, Montori VM, Schunemann H, Sprague S, et al. Association between industry funding and statistically significant pro‐industry findings in medical and surgical randomized trials. Canadian Medical Association Journal 2004;170(4):477‐80. [PUBMED: 14970094]

Birks 2006

Birks J. Cholinesterase inhibitors for Alzheimer's disease. Cochrane Database of Systematic Reviews 2006, Issue 1. [DOI: 10.1002/14651858.CD005593; PUBMED: 16437532]

Boyle 2003

Boyle PA, Malloy PF, Salloway S, Cahn‐Weiner DA, Cohen R, Cummings JL. Executive dysfunction and apathy predict functional impairment in Alzheimer disease. American Journal of Geriatric Psychiatry 2003;11(2):214‐21.

Bressan 2005

Bressan RA, Crippa JA. The role of dopamine in reward and pleasure behaviour‐‐review of data from preclinical research. Acta Psychiatrica Scandinavica. Supplement 2005;427:14‐21.

Brodaty 2012

Brodaty H, Arasaratnam C. Meta‐analysis of nonpharmacological interventions for neuropsychiatric symptoms of dementia. American Journal of Psychiatry 2012;169(9):946‐53.

Callegari 2016

Callegari I, Mattei C, Benassi F, Krueger F, Grafman J, Yaldizli O, et al. Agomelatine Improves apathy in frontotemporal dementia. Neuro‐Degenerative Diseases 2016;16(5‐6):352‐6. [PUBMED: 27229348]

Clarke 2011

Clarke DE, Ko JY, Kuhl EA, Van Reekum R, Salvador R, Marin RS. Are the available apathy measures reliable and valid? A review of the psychometric evidence. Journal of Psychosomatic Research 2011;70(1):73‐97. [PUBMED: 21193104]

Cummings 1994

Cummings JL, Mega M, Gray K, Rosenberg‐Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology 1994;44(12):2308‐14. [PUBMED: 7991117]

Cummings 2001

Cummings JL, Nadel A, Masterman D, Cyrus PA. Efficacy of metrifonate in improving the psychiatric and behavioral disturbances of patients with Alzheimer's disease. Journal of Geriatric Psychiatry and Neurology 2001;14(2):101‐8.

Cummings 2015

Cummings J, Friedman JH, Garibaldi G, Jones M, Macfadden W, Marsh L, et al. Apathy in neurodegenerative diseases: recommendations on the design of clinical trials. Journal of Geriatric Psychiatry and Neurology 2015;28(3):159‐73. [PUBMED: 25809634]

David 2008

David R, Koulibaly M, Benoit M, Garcia R, Caci H, Darcourt J, et al. Striatal dopamine transporter levels correlate with apathy in neurodegenerative diseases A SPECT study with partial volume effect correction. Clinical Neurology and Neurosurgery 2008;110(1):19‐24.

De Boer 2005

De Boer SF, Koolhaas JM. 5‐HT1A and 5‐HT1B receptor agonists and aggression: a pharmacological challenge of the serotonin deficiency hypothesis. European Journal of Pharmacology 2005;526(1‐3):125‐39.

De Deurwaerdère 1998

De Deurwaerdère P, Stinus L, Spampinato U. Opposite change of in vivo dopamine release in the rat nucleus accumbens and striatum that follows electrical stimulation of dorsal raphe nucleus: role of 5‐HT3 receptors. Journal of Neuroscience 1998;18(16):6528‐38.

Debette 2002

Debette S, Kozlowski O, Steinling M, Rousseaux M. Levodopa and bromocriptine in hypoxic brain injury. Journal of Neurology 2002;249(12):1678‐82.

Declercq 2013

Declercq T, Petrovic M, Azermai M, Vander Stichele R, De Sutter AIM, Van Driel ML, et al. Withdrawal versus continuation of chronic antipsychotic drugs for behavioural and psychological symptoms in older people with dementia. Cochrane Database of Systematic Reviews 2013, Issue 4. [DOI: 10.1002/14651858.CD007726.pub2]

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG, editor(s) on behalf of the Cochrane Statistical Methods Group. Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration. Available from www.cochrane‐handbook.org2011.

Diesfeldt 2013

Diesfeldt HF. Interpreting change scores of the Behavioural Rating Scale for Geriatric Inpatients (GIP). Tijdschrift voor Gerontologie en Geriatrie 2013;44(4):166‐74. [PUBMED: 23921986]

Doody 2013

Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. New England Journal of Medicine 2013;369(4):341‐50. [PUBMED: 23883379]

Egger 1997

Egger M, Davey‐Smith G, Schneider M, Minder CSO. Bias in meta‐analysis detected by a simple, graphical test. The British Medical Journal 1997;315(7109):629‐34.

Ellison 1986

Ellison DW, Beal MF, Mazurek MF, Bird ED, Martin JB. A postmortem study of amino acid neurotransmitters in Alzheimer's disease. Annals of Neurology 1986;20(5):616‐21.

Feldman 2001

Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E. A 24‐week, randomized, double‐blind study of donepezil in moderate to severe Alzheimer's disease. Neurology 2001;57(4):613‐20. [PUBMED: 11524468]

Feldman 2005

Feldman H, Gauthier S, Hecker J, Vellas B, Xu Y, Ieni JR, et al. Efficacy and safety of donepezil in patients with more severe Alzheimer's disease: a subgroup analysis from a randomized, placebo‐controlled trial. International Journal of Geriatric Psychiatry 2005;20(6):559‐69. [PUBMED: 15920715]

Folstein 1975

Folstein MF, Folstein SE, McHugh PR. "Mini‐mental state". A practical method for grading the cognitive state of patients for the clinician. Journal of Psychiatric Research 1975;12(3):189‐98. [PUBMED: 1202204]

Förstl 1993

Förstl H, Burns A, Levy R, Cairns N, Luthert P, Lantos P. Neuropathological correlates of behavioural disturbance in confirmed Alzheimer's disease. British Journal of Psychiatry 1993;163:364‐8.

Garcia‐Alloza 2005

Garcia‐Alloza M, Gil‐Bea FJ, Diez‐Ariza M, Chen CP, Francis PT, Lasheras B, et al. Cholinergic‐serotonergic imbalance contributes to cognitive and behavioral symptoms in Alzheimer's disease. Neuropsychologia 2005;43(3):442‐9.

Gauthier 2002

Gauthier S, Feldman H, Hecker J, Vellas B, Ames D, Subbiah P, et al. Efficacy of donepezil on behavioral symptoms in patients with moderate to severe Alzheimer's disease. International Psychogeriatrics 2002;14(4):389‐404. [PUBMED: 12670060]

General Assembly of the World Medical Association

General Assembly of the World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. The Journal of the American College of Dentists 2014;81(3):14‐8. [PUBMED: 25951678]

Gerrard 2007

Gerrard P, Malcolm R. Mechanisms of modafinil: A review of current research. Neuropsychiatric Disease and Treatment 2007;3(3):349‐64.

Gitlin 2012

Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. The Journal of the American Medical Association 2012;308(19):2020‐9.

Grace 2011

Grace J. Frontal systems behavior scale. Encyclopedia of Clinical Neuropsychology 2011:1090‐3.

Guyatt 2008

Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‐Ytter Y, Alonso‐Coello P, et al. GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. The British Medical Journal 2008;336(7650):924‐6.

Harrison 2016

Harrison F, Aerts L, Brodaty H. Apathy in dementia: systematic review of recent evidence on pharmacological treatments. Current Psychiatry Reports 2016;18(11):103. [PUBMED: 27726067]

Heres 2006

Heres S, Davis J, Maino K, Jetzinger E, Kissling W, Leucht S. Why olanzapine beats risperidone, risperidone beats quetiapine, and quetiapine beats olanzapine: an exploratory analysis of head‐to‐head comparison studies of second‐generation antipsychotics. American Journal of Psychiatry 2006;163(2):185‐94. [PUBMED: 16449469]

Herrmann 2004a

Herrmann N, Lanctôt KL, Khan LR. The role of norepinephrine in the behavioral and psychological symptoms of dementia. Journal of Neuropsychiatry and Clinical Neurosciences 2004;16(3):261‐76.

Herrmann 2004b

Herrmann N, Lanctôt K, Eryavec G, Van Reekum R, Khan LR. Growth hormone response to clonidine predicts aggression in Alzheimer's disease. Psychoneuroendocrinology 2004;29(9):1192‐7.

Herrmann 2013

Herrmann N, Lanctôt KL, Hogan DB. Pharmacological recommendations for the symptomatic treatment of dementia: the Canadian consensus conference on the diagnosis and treatment of dementia 2012. Alzheimer's Research & Therapy 2013;5(Suppl 1):S5.

Higgins 2011

Higgins JPT, Altman DG, Sterne, JAC, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Hoehn‐Saric 1990

Hoehn‐Saric R, Lipsey JR, McLeod DR. Apathy and indifference in patients on fluvoxamine and fluoxetine. Journal of Clinical Psychopharmacology 1990;10(5):343‐5.

Hurt 2008

Hurt C, Bhattacharyya S, Burns A, Camus V, Liperoti R, Marriott A, et al. Patient and caregiver perspectives of quality of life in dementia. An investigation of the relationship to behavioural and psychological symptoms in dementia. Dementia and Geriatric Cognitive Disorders 2008;26(2):138‐46.

Kales 2015

Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. The British Medical Journal 2015;350:h369.

Kang 2012

Kang JY, Lee JS, Kang H, Lee HW, Kim YK, Jeon HJ, et al. Regional cerebral blood flow abnormalities associated with apathy and depression in Alzheimer's disease. Alzheimer Disease and Associated Disorders 2012;26(3):217‐24.

Kapasi 2016

Kapasi A, Schneider JA. Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons. Biochimica et Biophysica Acta 2016;1862(5):878‐86.

Katz 1963

Katz S, Ford AB, Moskowitz RW, Jackson BA, Jaffe MW. Studies of illness in the aged. The index of ADL: A standardized measure of biological and psychosocial function. JAMA 1963;185:914‐9. [PUBMED: 14044222]

Kaufer 1998a

Kaufer D, Cummings JL, Christine D. Differential neuropsychiatric symptom responses to tacrine in Alzheimer's disease: relationship to dementia severity. Journal of Neuropsychiatry and Clinical Neurosciences 1998;10(1):55‐63.

Kaufer 1998b

Kaufer D. Beyond the cholinergic hypothesis: the effect of metrifonate and other cholinesterase inhibitors on neuropsychiatric symptoms in Alzheimer's disease. Dementia and Geriatric Cognitive Disorders 1998;9 Suppl 2:8‐14.

Lanari 2006

Lanari A, Amenta F, Silvestrelli G, Tomassoni D, Parnetti L. Neurotransmitter deficits in behavioural and psychological symptoms of Alzheimer's disease. Mechanisms of Ageing and Development 2006;127(2):158‐65.

Lanctôt 2001

Lanctôt KL, Herrmann N, Mazzotta P. Role of serotonin in the behavioral and psychological symptoms of dementia. Journal of Neuropsychiatry and Clinical Neurosciences 2001;13(1):5‐21.

Lanctôt 2007a

Lanctôt KL, Herrmann N, Rothenburg L, Eryavec G. Behavioral correlates of GABAergic disruption in Alzheimer's disease. International Psychogeriatrics 2007;19(1):151‐8.

Lanctôt 2007b

Lanctôt KL, Moosa S, Herrmann N, Leibovitch FS, Rothenburg L, Cotter A, et al. A SPECT study of apathy in Alzheimer's disease. Dementia and Geriatric Cognitive Disorders 2007;24(1):65‐72.

Landes 2001

Landes AM, Sperry SD, Strauss ME, Geldmacher DS. Apathy in Alzheimer’s disease. Journal of the American Geriatrics Society 2001;49(12):1700–7.

Lawton 1969

Lawton MP, Brody EM. Assessment of older people: self‐maintaining and instrumental activities of daily living. Gerontologist 1969;9(3):179‐86. [PUBMED: 5349366]

Leonpacher 2016

Leonpacher AK, Peters ME, Drye LT, Makino KM, Newell JA, Devanand DP, et al. Effects of citalopram on neuropsychiatric symptoms in Alzheimer'sdDementia: evidence from the CitAD Study. American Journal of Psychiatry 2016;173(5):473‐80. [PUBMED: 27032628]

Levy 1998

Levy ML, Cummings JL, Fairbanks LA, Masterman D, Miller BL, Craig AH, et al. Apathy is not depression. Journal of Neuropsychiatry and Clinical Neurosciences 1998;10(3):314‐9.

Lexchin 2003

Lexchin J, Bero LA, Djulbegovic B, Clark O. Pharmaceutical industry sponsorship and research outcome and quality: systematic review. The British Medical Journal (Clinical research ed.) 2003;326(7400):1167‐70. [PUBMED: 12775614]

Lonergan 2009

Lonergan E, Luxenberg J. Valproate preparations for agitation in dementia. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD003945.pub3; PUBMED: 19588348]

Lowe 1988

Lowe SL, Francis PT, Procter AW, Palmer AM, Davison AN, Bowen DM. Gamma‐aminobutyric acid concentration in brain tissue at two stages of Alzheimer's disease. Brain 1988;111(Pt 4):785‐99.

Marin 1991

Marin RS, Biedrzycki RC, Firinciogullari S. Reliability and validity of the apathy evaluation scale. Psychiatry Research 1991;38(2):143‐62. [PUBMED: 1754629]

Marin 1994

Marin RS, Firinciogullari S, Biedrzycki RC. Group differences in the relationship between apathy and depression. Journal of Nervous and Mental Disease 1994;182(4):235‐9.

Marin 1995

Marin RS, Fogel BS, Hawkins J, Duffy J, Krupp B. Apathy: a treatable syndrome. Journal of Neuropsychiatry and Clinical Neurosciences 1995;7(1):23‐30.

McKhann 1984

McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease: report of the NINCDS‐ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's disease. Neurology 1984;34(7):939‐44.

McKhann 2011

McKhann GM, Knopman DS, Chertkow H, Hyman, BT, Jack CR, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging‐Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer's & Dementia 2011;7(3):263‐9.

Mitchell 2011

Mitchell RA, Herrmann N, Lanctôt KL. The role of dopamine in symptoms and treatment of apathy in Alzheimer's disease. CNS Neuroscience & Therapeutics 2011;17(5):411‐27.

Mulin 2011

Mulin E, Leone E, Dujardin K, Delliaux M, Leentjens A, Nobili F, et al. Diagnostic criteria for apathy in clinical practice. International Journal of Geriatric Psychiatry 2011;26(2):158‐65. [PUBMED: 20690145]

O'Regan 2015

O'Regan J, Lanctôt KL, Mazereeuw G, Herrmann N. Cholinesterase inhibitor discontinuation in patients with Alzheimer's disease: a meta‐analysis of randomized controlled trials. Journal of Clinical Psychiatry 2015;76(11):e1424‐31. [PUBMED: 26646039]

Overall 1962

Overall JE, Gorham DR. The Brief Psychiatric Rating Scale. Psychological Reports 1962;19:799‐812.

Overshott 2004

Overshott R, Byrne J, Burns A. Nonpharmacological and pharmacological interventions for symptoms in Alzheimer's disease. Expert Review of Neurotherapeutics 2004;4(5):809‐21.

Padala 2007

Padala PR, Burke WJ, Bhatia SC. Modafinil therapy for apathy in an elderly patient. Annals of Pharmacotherapy 2007;41(2):346‐9.

Politis 2004

Politis AM, Mayer LS, Passa M, Maillis A, Lyketsos CG. Validity and reliability of the newly translated Hellenic Neuropsychiatric Inventory (H‐NPI) applied to Greek outpatients with Alzheimer's disease: a study of disturbing behaviors among referrals to a memory clinic. International Journal of Geriatric Psychiatry 2004;19(3):203‐8. [PUBMED: 15027034]

Porsteinsson 2014

Porsteinsson AP, Drye LT, Pollock BG, Devanand DP, Frangakis C, Ismail Z, et al. CitAD Research Group. Effect of citalopram on agitation in Alzheimer disease: the CitAD randomized clinical trial. The Journal of the American Medical Association 2014;311(7):682‐91.

Prince 2013

Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimer's & Dementia 2013;9(1):63‐75.e2.

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.

Robert 2009

Robert P, Onyike CU, Leentjens AF, Dujardin K, Aalten P, Starkstein S, et al. Proposed diagnostic criteria for apathy in Alzheimer's disease and other neuropsychiatric disorders. European Psychiatry 2009;24(2):98‐104.

Roccaforte 1990

Roccaforte WH, Burke WJ. Use of psychostimulants for the elderly. Hospital & Community Psychiatry 1990;41(12):1330‐3.

Rockwood 2001

Rockwood K, Mintzer K, Truyen L, Wessel T, Wilkinson D. Effects of a flexible galantamine dose in Alzheimer's disease: a randomized, controlled trial.. Journal of Neurology, Neurosurgery and Psychiatry 2001;71(5):589‐95.

Rosenberg 2016

Rosenberg PB, Lanctôt KL, Herrmann N, Mintzer JE, Porsteinsson AP, Sun X, et al. Changes in neuropsychiatric inventory associated with semagacestat treatment of Alzheimer's disease. Journal of Alzheimer's Disease : JAD 2016;54(1):373‐81. [PUBMED: 27567808]

Rossor 1982

Rossor MN, Garrett NJ, Johnson AL, Mountjoy CQ, Roth M, Iversen LL. A post‐mortem study of the cholinergic and GABA systems in senile dementia. Brain 1982;105(Pt 2):313‐30.

Schneider 1997

Schneider LS, Olin JT, Doody RS, Clark CM, Morris JC, Reisberg B, et al. Validity and reliability of the Alzheimer's Disease Cooperative Study‐Clinical Global Impression of Change. The Alzheimer's Disease Cooperative Study. Alzheimer Disease and Associated Disorders 1997;11 Suppl 2:S22‐32. [PUBMED: 9236949]

Schneider 2007

Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain pathologies account for most dementia cases in community‐dwelling older persons. Neurology 2007;69(24):2197‐204.

Schünemann 2011

Schünemann H, Hill S, Guyatt G, Akl EA, Ahmed F. The GRADE approach and Bradford Hill's criteria for causation. Journal of Epidemiology and Community Health 2011;65(5):392‐5.

Seitz 2011

Seitz DP, Adunuri N, Gill SS, Gruneir A, Herrmann N, Rochon P. Antidepressants for agitation and psychosis in dementia. Cochrane Database of Systematic Reviews 2011, Issue 2. [DOI: 10.1002/14651858.CD008191.pub2; PUBMED: 21328305]

Sepehry 2017

Sepehry AA, Sarai M, Hsiung GR. Pharmacological therapy for apathy in Alzheimer's disease: a systematic review and meta‐analysis. Canadian Journal of Neurological Sciences. 2017;44(3):267‐75. [PUBMED: 28148339]

Starkstein 1997

Starkstein SE, Sabe L, Vázquez S, Di Lorenzo G, Martínez A, Petracca G, et al. Neuropsychological, psychiatric, and cerebral perfusion correlates of leukoaraiosis in Alzheimer's disease. Journal of Neurology, Neurosurgery, and Psychiatry 1997;63(1):66‐73.

Starkstein 2009

Starkstein SE, Mizrahi R, Capizzano AA, Acion L, Brockman S, Power BD. Neuroimaging correlates of apathy and depression in Alzheimer's disease. Journal of Neuropsychiatry and Clinical Neurosciences 2009;21(3):259‐65.

Steinberg 2008

Steinberg M, Shao H, Zandi P, Lyketsos CG, Welsh‐Bohmer KA, Norton MC, et al. Point and 5‐year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. International Journal of Geriatric Psychiatry 2008;23(2):170‐7.

Storga 1996

Storga D, Vrecko K, Birkmayer JG, Reibnegger G. Monoaminergic neurotransmitters, their precursors and metabolites in brains of Alzheimer patients. Neuroscience Letters 1996;203(1):29‐32.

Tagariello 2009

Tagariello P, Girardi P, Amore M. Depression and apathy in dementia: same syndrome or different constructs? A critical review. Archives of Gerontology and Geriatrics 2009;49(2):246‐9.

Tariot 2000

Tariot PN, Solomon PR, Morris JC, Kershaw P, Lilienfeld S, Ding C. A 5‐month, randomized, placebo‐controlled trial of galantamine in AD. The Galantamine USA‐10 Study Group. Neurology 2000;54(12):2269‐76.

Tekin 2001

Tekin S, Mega MS, Masterman DM, Chow T, Garakian J, Vinters HV, et al. Orbitofrontal and anterior cingulate cortex neurofibrillary tangle burden is associated with agitation in Alzheimer disease. Annals of Neurology 2001;49(3):355‐61.

Van der Linde 2016

Van der Linde RM, Dening T, Stephan BCM, Prina AM, Evans E, Brayne C. Longitudinal course of behavioural and psychological symptoms of dementia: systematic review. British Journal of Psychiatry 2016;209(5):366‐77.

Vartiainen 1995

Vartiainen H, Tiihonen J, Putkonen A, Koponen H, Virkkunen M, Hakola P, et al. Citalopram, a selective serotonin reuptake inhibitor, in the treatment of aggression in schizophrenia. Acta Psychiatrica Scandinavica 1995;91(5):348‐51.

Vialta‐Franch 2013

Vilalta‐Franch J, Calvó‐Perxas L, Garre‐Olmo J, Turró‐Garriga O, López‐Pousa S. Apathy syndrome in Alzheimer's disease epidemiology: prevalence, incidence, persistence, and risk and mortality factors. Journal of Alzheimer's Disease 2013;33(2):535‐43.

Volkow 2002

Volkow ND, Fowler JS, Wang G, Ding Y, Gatley SJ. Mechnism of action of methylphenidate: Insights from PET imaging studies. Journal of Attention Disorders 2002;6(1):S31‐S43.

Walsh 1997

Walsh SL, Cunningham KA. Serotonergic mechanisms involved in the discriminative stimulus, reinforcing and subjective effects of cocaine. Psychopharmacology (Berlin) 1997;130(1):41‐58.

WHO 1992

World Health Organization. The ICD‐10 Classification of Mental and Behavioural Disorders: clinical descriptions and diagnostic guidelines. 1992. apps.who.int/iris/handle/10665/37958#sthash.Vaxjp53U.dpuf (accessed 05 February 2016).

World Alzheimer Report 2015

Prince M, Wimo A, Guerchet M, Ali G, Wu Y, Prina M. World Alzheimer Report 2015: The Global Impact of Dementia. www.worldalzreport2015.org/downloads/world‐alzheimer‐report‐2015.pdf (accessed 05 February 2016).

Referencias de otras versiones publicadas de esta revisión

Ruthirakuhan 2016

Ruthirakuhan MT, Herrmann N, Abraham EH, Lanctôt KL. Pharmacological interventions for apathy in Alzheimer's disease (Protocol). Cochrane Database of Systematic Reviews 2016, Issue 5. [DOI: 10.1002/14651858.CD012197]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Jump to:

CitAD trial

Methods

Multicenter, randomized, double‐blind, placebo‐controlled trial

Participants

Participant information obtained from Porsteinsson 2014:

  • diagnosis of “probable” AD according to NINCDS‐ADRDA criteria

  • MMSE score between 5 – 28 inclusive

  • clinically significant agitation from which a physician determined that medication was appropriate (NPI agitation/aggression occurring “frequently” with “moderate” or “marked” severity)

  • must have a caregiver who spends at least several hours a week with the participant and required to supervise medications and participate in outcome assessments

  • stable dose of ChEI and memantine within a month preceding randomization

  • withdrawal of psychotropic medications other than predefined rescue medications required

Interventions

Participants were randomized to receive:

  • citalopram (target dose of 30 mg/day with planned titration over 3 weeks from a starting dose of 10 mg/day) (N = 94)

  • placebo (N = 92)

During the first 3 weeks after randomization, clinicians could adjust the medication dosage according to response and tolerability. Caregivers received a standardized practical psychosocial intervention of 3 components: provision of educational materials, 24‐hour availability of crisis management services, and a 20‐ to 30‐minute counseling session at each scheduled study visit

Duration: 9‐week treatment phase

Enrollment: 186 participants randomized

Outcomes

Outcomes were obtained from Leonpacher 2016:
Primary:

  • NRS

  • modified CGIC

Secondary:

  • NPI total, individual and distress scores

  • CMAI (short form)

  • ADCS‐ADL

  • use of rescue lorazepam

Notes

We have selected Leonpacher 2016 to be the primary paper. However, additional information for this study was obtained from clinicaltrials.gov (NCT00898807) and Porsteinsson 2014. Study dates: July 2009 ‐ September 2013. Eight sites in the USA and Canada were included. Specific site locations not disclosed. Coordinating site: Johns Hopkins University, Baltimore, MD, USA. Funding provided by the National Institute on Aging and NIMH grant R01AG031348, and in part by HIH grant P50‐AG05142

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Patients were randomized to receive…”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “Patients were randomized to receive citalopram at a target dosage of 30 mg/day… or matching placebo.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “…there was no difference in adherence between the citalopram and placebo groups and that side effects were generally modest and consistent with those known to be associated with SSRIs (gastrointestinal complaints, respiratory tract infections and falls). The adverse effects of cognitive worsening (of unknown clinical significance) and QT prolongation, however, raise concerns about 30 mg/day dosage used…”

Comment: Study withdrawals and reason for withdrawals have been reported in Porsteinsson 2014 (Safety and Adherence section and Table 3) and appear to be balanced between groups

Selective reporting (reporting bias)

High risk

The authors provided medians and interquartile ranges for each NPI item at baseline (Table 1) and at 9 weeks (Table 2), but did not report means ± SD scores. The authors of this meta‐analysis computed these values themselves. However, as CitAD trial disclosed that these values were not normally distributed, there may be a selective reporting bias

Other bias

Low risk

No other identified biases

De Deyn 2004

Methods

Multicenter, double‐blind, randomized, placebo‐controlled trial

Participants

  • ≥ 40 years old

  • reside in LTC homes or continuing‐care hospitals and were expected to continue patient status for 6 months following enrollment

  • diagnosis of “possible” or “probable” AD according to NINCDS‐ADRDA and DSM‐IV‐TR criteria

  • clinically significant psychotic symptoms (delusions or hallucinations) due to AD. Must be 1) at least moderate in severity at study entry and randomization; 2) present at least once a week for the month preceding study entry; 3) requires pharmacological intervention, in the opinion of the investigator

  • MMSE score ≥ 5 at study entry and randomization

  • stable dose of ChEI prior to study entry

Interventions

Following placebo lead‐in phase (up to 14 days) participants were randomized to receive either:

  • olanzapine (1.0 mg/day) (N = 129)

  • olanzapine (2.5 mg/day) (N = 134)

  • olanzapine (5.0 mg/day; 2.5 mg/day for first week, titrated to final dose by 2.5 mg/week increments) (N = 125)

  • olanzapine (7.5 mg/day; 2.5 mg/day for first week, titrated to final dose by 2.5 mg/week increments) (N = 132)

  • placebo (N = 129)

Duration: 10‐week treatment phase (+ maximum 14‐day placebo lead in)

Enrollment: 652 patients randomized, however 649 included in analysis.

Outcomes

Primary:

  • NPI‐NH psychosis total (sum of hallucinations and delusions)

  • CGI‐C

  • CGI‐S

Secondary:

  • NPI‐NH total and individual scores

  • BPRS

  • MMSE

  • SIB

Safety:

  • *SAS

  • AIMS

  • POMA

  • Other safety assessments: spontaneously reported TEAEs, changes in vital signs, electrocardiography and measurements of laboratory hematologies and chemistries

Notes

Study dates not reported. 61 sites in Europe, Australia, Israel, Lebanon, and South Africa were included. Specific site locations not disclosed. Corresponding author’s institution: Lily Research Laboratories, Indianapolis, IN, USA. Contract/grant sponsor: Eli Lilly and Company.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Patients randomly assigned to receive…”

Comment: Probably done

Allocation concealment (selection bias)

Unclear risk

This information has not been made available

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “The proportion of patients completing the 10‐week treatment period was not significantly different among the five treatment groups (Table 1)…”

Comment: Study withdrawals and reason for withdrawals have been reported in Table 1 and appear to be balanced between groups

Selective reporting (reporting bias)

Low risk

The authors reported means ± standard deviations on each of the 12‐item NPI‐NH scores (Table 2), BPRS (total, negative and positive) scores, and CGI scores (Table 3)

Other bias

Low risk

No other identified biases

Frakey 2012

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

  • diagnosis of mild‐to‐moderate stage “probable” AD according to NINCDS‐ADRDA criteria

  • clinically elevated symptoms of apathy based on FrSBe scale

  • stable dose of ChEI for 30 days

Interventions

Participants were urn‐randomized into either:

  • modafinil (100 mg/day in the morning for the first week and 200 mg/day in the morning for remaining 7 weeks) (N = 11)

  • placebo (N = 11)

Duration: 8 weeks

Enrolment: 23 participants randomized, 1 participant excluded from analysis due to AE‐related drop‐out

Outcomes

Primary:

  • FrSBe apathy score

Other:

  • ADLQ

  • DAFS

  • ZBI

Notes

Study dates: July 2005 ‐ September 2007. Study site: Butler Hospital, Providence, RI, USA. Salary support for the corresponding author provided by a National Research Service Award from the National Institute of Mental Health. Cephalon provided study medication, placebo, and $40,000 USD through an unrestricted investigator‐initiated grant

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “A randomized, double‐blind, placebo‐controlled design was employed.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “Participants were urn‐randomized into either the experimental group (modafinil) or the control group (placebo) using apathy severity (mild, moderate, or severe), dementia severity (mild or moderate), presence of antidepressant medication, and presence of memantine as randomization factors.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."“Both the physician (S.S.) and the clinician (L.L.F.) who performed the assessments were blind to the medication status of the participants.”

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “One experimental group participant was withdrawn from the study after 2 weeks due to an increase in motor tics.”

Comment: Though no participant flow diagram was provided in text, participant withdrawal information and reasoning described in text

Selective reporting (reporting bias)

Low risk

Quote: “The mean scores and SDs for our 2 groups for each of the outcome measures are presented in Table 2.”

Comment: Table 2 provides descriptive statistics for independent and dependent variables assessed in the study, providing means ± standard deviations for each outcome

Other bias

Low risk

No other identified biases

Herrmann 2005

Methods

Data pooled from 3 multicenter, double‐blind, placebo‐controlled, parallel‐group studies

Participants

Similar criteria between the 3 studies:

  • diagnosis of mild‐moderate “probable” AD according to NINCDS‐ADRDA criteria

  • ≥ 6‐month history of gradual onset, progressive cognitive decline

Rockwood 2001:

  • sMMSE score between 11 and 24

  • ADAS‐Cog‐11 score ≥ 12

Tariot 2000:

  • sMMSE score between 10 and 22

  • ADAS‐Cog‐11 score ≥ 18

Data obtained by authors from Janssen‐Ortho Inc.:

  • sMMSE score between 10 and 24

  • ADAS‐Cog‐11 score ≥ 18

Interventions

Rockwood 2001:

Participants were randomized to either:
‐ Galantamine (IR) (N = 261)
‐ Placebo (N = 125)

Duration: 3‐month treatment phase (+1 month placebo run‐in)

Enrollment: 368 participants randomized

Tariot 2000:

Participants were randomized to either:

  • Galantamine (IR) 8 mg/day (N = 140)

  • Galantamine (IR) 16 mg/day (N = 279)

  • Galantamine 24 mg/day (N = 273)

  • Placebo (N = 286)

Duration: 5‐month treatment phase (+1 month placebo run‐in)

Enrollment: 978 participants randomized

Data obtained by authors from Janssen‐Ortho Inc.

Participants were randomized to either:

  • Galantamine IR (N = 327)

  • Galantamine CR (N = 320)

  • Placebo (N = 324)

Duration: 6‐month treatment phase (+1 month placebo run)

Enrollment: 971 participants randomized

Outcomes

Primary outcome for pooled analysis:

  • NPI individual, cluster, and total scores

Primary outcome for included studies include changes from baseline in:

  • ADAS‐cog‐11

  • CIBIC‐plus

Behavioral outcome for included studies include changes from baseline in:

  • NPI individual, cluster, and total scores

Notes

Study dates note reported. Sites of studies included in analyses include the USA, Canada, Great Britain, South Africa, Australia, and New Zealand. Specific site locations not disclosed. Corresponding author’s institution: Sunnybrook Health Sciences Centre, Toronto, Canada.This research was supported by a grant from the American Health Assistance Foundation‐Alzheimer's Disease Research Program (#A2003‐236) and by the Dean's Fund of the University of Toronto. All studies were supported by Jannssen.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Data were pooled from 2033 subjects...who had participated in one of three randomized, double‐blind placebo‐controlled trials".
Comment: Probably done

Allocation concealment (selection bias)

Unclear risk

This information has not been made available

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

2 of the studies included in this pooled analysis (Tariot 2000; Rockwood 2001) have each included a figure on participant flow. However, as the data from Janssen‐Ortho Inc. has not been made available, we are unsure about the risk of bias

Selective reporting (reporting bias)

Low risk

The authors reported on means ± standard deviations on each of the 12‐item NPI scores, at baseline and over treatment duration

Other bias

Low risk

No other identified biases

Herrmann 2007

Methods

Randomized, double‐blind, placebo‐controlled cross‐over trial

Participants

  • > 55 years old

  • residents from 2 long‐term facilities associated with university‐affiliated general hospitals

  • diagnosis of primary degenerative dementia according to DSM‐IV and “probable” AD of at least 1 year’s duration according to NINCDS‐ADRDA criteria

  • MMSE score of < 15

  • significant BPSD as indicated by ≥ 8 on NPI

Interventions

Participants underwent a placebo washout of all psychotropic drugs based on a minimum of 5 half‐lives of the drug used

Participants were then randomized to receive:

  • valproate liquid suspension (initiated at 125 mg orally, twice/day with forced titration to 500 mg orally, twice/day over the first 2 weeks. Thereafter, dose could be increased to a maximum of 1 500 mg/day or decreased based on tolerability)

  • placebo

Loxapine 2.5 mg maximum 4 doses/week was available as a rescue medication

Duration: 14 weeks (6‐week treatment phases of valproate or placebo + 2‐week placebo washout and tapering)

Enrollment: 14 patients randomized

Outcomes

Primary:

  • NPI‐NH agitation subscore

Secondary:

  • NPI‐NH total and individual scores

  • CMAI

Notes

Study dates not reported. Study site: 1) Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; 2) North York General Hospital, Toronto, Canada. Additional data and information was provided by Herrmann and Lanctôt upon request. Funding provided by the Alzheimer’s Society of Canada (grant No. 01‐07).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Patients were randomized to receive…”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “Patients were randomized to receive valproate liquid suspension or an identical placebo…”

Comment: Probably done.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “Twelve patients experienced at least one adverse event during valproate treatment, compared to 8 patients during placebo treatment. Although this difference is not statistically significant, the mean number of adverse events experienced with valproate treatment (4.15 +/‐ 3.67) was significantly greater than with placebo (1.23 +/‐ 1.69) treatment (Z = ‐2.82, p = 0.005).”

Comment: Withdrawal numbers and adverse events per group are provided in the Results section and are not balanced between groups

Selective reporting (reporting bias)

Low risk

Mean change scores ± SD scores are provided in Table 1 for NPI‐agitation subcategory, total NPI, total CMAI and MMSE scores. NPI‐apathy mean change scores ± SD scores were calculated with data provided by authors upon request

Other bias

Low risk

No other identified biases

Herrmann 2008

Methods

Randomized, double‐blind, placebo‐controlled, cross‐over trial

Participants

  • ≥ 55 years old

  • diagnosis of “possible” or “probable” AD according to NINCDS‐ADRDA criteria

  • MMSE score ≥ 10 (mild‐to‐moderate cognitive impairment)

  • NPI Apathy subscale score ≥ 1

  • stable dose of ChEI for 3 months and not receiving any other psychotropic medications, including antidepressants or antipsychotics

Interventions

Participants took a dextroamphetamine (D‐amph) challenge test (10 mg D‐amph orally). Following up to a 1‐week washout phase, participants were randomized to receive either:

‐ methylphenidate (initiated at 5 mg orally twice/day for 3 days and increased to 10 mg orally twice/day for 11 days)

‐ placebo

Duration: 5 weeks (2‐week treatment phases with a 1‐week placebo washout between phases) + at least 1 week washout after the D‐amph challenge test

Enrollment: 13 participants randomized

Outcomes

Primary:

  • AES

  • CGI

Other:

  • NPI total and individual apathy score

  • MMSE

  • measurements of acute subjective response to D‐amph: ARCI, POMS, CPT

Notes

Study dates: October 2003 ‐ October 2006. Study site: Sunnybrook Health Sciences Centre, Toronto, Canada. This research was supported by a grant from the American Health Assistance Foundation‐Alzheimer’s Disease Research Program (#A2003‐236) and by the Dean’s Fund of the University of Toronto.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “We conducted a double‐blind, randomized, placebo‐controlled crossover trial...”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “Patients were randomized to receive methylphenidate or an identical placebo...”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind…”

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “Eleven of the 13 patients completed the study, with both dropouts occurring during the methylphenidate treatment phase. One dropout completed all placebo assessments and the baseline methylphenidate assessment, which was carried forward for the efficacy analysis. The second patient dropped out after 8 days of methylphenidate treatment but completed a retrieved dropout assessment. This patient did not participate in the placebo phase. Results are therefore available for 13 patients treated with methylphenidate and 12 patients treated with placebo.”

Comment: Though no participant flow diagram was provided in text, withdrawal information and reasoning described in text

Selective reporting (reporting bias)

Low risk

Table 1 provides means ± standard deviations for treatment change scores (end of treatment‐ baseline) for AES total, NPI apathy, NPI total and MMSE for participants during the methylphenidate and placebo treatment phases

Other bias

Low risk

No other identified biases

Herrmann 2016

Methods

Multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group study

Participants

  • ≥ 55 years old

  • institutionalized people in LTC

  • diagnosis of “probable” AD according to NINCDS‐ADRDA criteria

  • primary degenerative dementia according to DSM‐V criteria

  • sMMSE score ≤ 15

  • treated with donepezil, galantamine or rivastigmine (oral) for ≥ 2 years, with a stable dose for ≥ 3 months prior to study entry

  • people receiving a concomitant psychotropic had to be on a stable dose for ≥ 1 month prior to study entry

Interventions

Participants were randomized to either:

  • ChEI continuation (N = 21)

  • placebo (N = 19)

Duration: 8 weeks

Enrolment: 40 participants randomized

Outcomes

Primary:

  • CGI‐C

Secondary:

  • safety

  • number of individual and total AEs

  • efficacy and tolerability

  • AES

  • NPI‐NH score

  • CMAI

  • sMMSE

  • SIB

  • ADCS‐ADL‐sev

  • QUALID

Notes

Study dates: July 2010 ‐ September 2015. Study sites: 1) Sunnybrook Health Sciences Centre, Toronto, Canada; 2) North York General Hospital, Toronto, Canada. Additional data provided by Herrmann et al. This study was funded by the Alzheimer's Society of Canada (#:12‐74) and the Coleman Fund (internal funding)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “...patients were randomized with a 1:1 balanced by ChEI to continue receiving their ChEI (continuation)...or to receive an identical‐looking placebo substitution.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “Randomization was completed independently by the pharmacy...in permuted blocks using a computer‐generated code.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: 'Patients, family members, nurses, clinicians, outcome assessors, and investigators were unaware of treatment group assignments or block size'.
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The reasons for withdrawal were provided in the participant flow diagram (figure 1). Number of early terminations and time to early termination were balanced between groups

Selective reporting (reporting bias)

Low risk

Mean and ± SD of the baseline, endpoint and change scores of all primary and secondary outcome measures were reported in table 3

Other bias

Unclear risk

This was a discontinuation study completed in people who had been receiving long‐term ChEI treatment

Kaufer 1998

Methods

Multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group study

Participants

  • diagnosis of “probable” AD according to NINCDS‐ADRDA criteria

  • sMMSE score between 10 and 26

  • modified HIS ≤ 4

Interventions

Participants were randomized to either:

  • metrifonate (initiated for a 2‐week period with a 2.0 mg/kg single daily dose, followed by a single daily maintenance dose of 0.65 mg/kg; 30 ‐ 60 mg total) (N = 260)

  • placebo (N = 133)

Duration: 26 weeks

Enrollment: 408 patients randomized, 393 were included in the analysis as they were a part of the valid intention‐to‐treat population.

Outcomes

Primary:

  • NPI total and individual scores

Notes

Study dates not reported. 25 sites in the USA. Specific site locations not disclosed. Corresponding author’s institution: University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. No funding support reported in paper.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: 'Subjects were randomized to either placebo...or metrifonate...treatment groups'.
Comment: Probably done

Allocation concealment (selection bias)

Unclear risk

This information has not been made available

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: '...double‐blind...'
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: '...double‐blind...'
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

This information has not been made available

Selective reporting (reporting bias)

High risk

LSM changes in NPI total and NPI‐subitem scores were provided in table 1. As LSM are adjusted for covariates, there is a risk of bias. Additionally, no standard error or standard deviation was reported in this paper. As a result, the authors of this meta analysis computed a standard deviation based on other studies (Raskind 1999) investigating the use of metrifonate in people with AD

Other bias

Low risk

No other identified biases

Lanctôt 2002

Methods

Multicentre, double‐blind, randomized, placebo‐controlled cross‐over study

Participants

  • ≥ 55 years old

  • diagnosis of primary degenerative dementia criteria according to DSM‐IV, and “probable” AD (≥ 1 year's duration) according to NINCDS‐ADRDA criteria

  • significant behavioral problems (NPI ≥ 8)

  • MMSE score < 24

  • independent clinical decision to receive psychotropic medication for behavioral disorder

Interventions

  • approximately 1‐week placebo run‐in, during which all psychotropic and antiparkinsonian medications were discontinued

  • fenfluramine challenge as described in Lanctôt 2002, following placebo run‐in

Participants were then randomized to:

  • sertraline (50 mg/day for 3 days, followed by 100 mg/day for 25 days)

  • placebo (1 orally/day for 3 days, followed by 2 orally/day for 25 days)

  • 1 week placebo washout before crossing over to alternate study treatment

Duration: 9‐week treatment phase (4 weeks of first treatment phase + 1 week placebo washout, and cross over to 4 weeks of second treatment phase) + approximately 1‐week placebo run‐in

Enrollment: 22 participants randomized

Outcomes

The following are primary outcomes:

Behavior:

  • NPI total and individual scores

  • CMAI

  • BEHAVE‐AD

  • CSDD

Function:

  • FAST

Cognition:

  • MMSE

Notes

Study dates not provided. Study sites: 1) Sunnybrook Health Sciences Centre, Toronto, Canada; 2) Baycrest Centre for Geriatric Care, Toronto, Canada; 3) North York General Hospital, Toronto, Canada. Additional data requested, and provided by Dr. Krista Lanctôt.This study was funded by Physicians' Services Incorporated Foundation (96‐06), Alzheimer Society of Canada Research Program, and Kunin Lunenfeld Applied Research Unite of Baycrest Centre for Geriatric Care

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...administered in a randomized, double‐blind trial."
Comment: Probably done

Allocation concealment (selection bias)

Unclear risk

This information has not been made available

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double blind..."
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double blind..."
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Of those 26, three patients dropped out before randomization and one
dropped out because the caregiver withdrew consent. Of the remaining 22 patients, one was withdrawn due to severe antipsychotic withdrawal dyskinesia and a fall shortly after randomization (not included in efficacy analyses). Therefore, there were 21 patients who completed the entire study."

Comment: Although no participant flow diagram was included, attrition was described in detail in the paper

Selective reporting (reporting bias)

Low risk

Lanctôt 2002 reported on treatment responders in this paper. Dr. Lanctôt provided relevant data for this meta‐analysis. We were able to extract the mean ± standard deviations, and frequencies for all relevant outcome measures

Other bias

Low risk

No other identified biases

Morris 1998

Methods

Multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group study

Participants

  • diagnosis of “probable” AD according to the NINCDS‐ADRDA criteria

  • sMMSE score between 10 and 26

  • modified Ischemia Scale scores < 4

  • body weight ≥ 45 and ≤ 94 kg (98 to 207 pounds)

  • caregiver with whom participant is in contact at least 4 times a week

Interventions

Participants were randomized to either:

  • metrifonate (loading dose, based on weight, of 100 to 180 mg (2.0 mg/kg) for 2 weeks followed by maintenance dose, based on weight, of 30 to 60 mg (0.65 mg/kg) for 24 weeks) (N = 273)

  • placebo (N = 135)

Duration: 26‐week double‐blind period (+ 2‐week screening period at beginning of the study + follow‐up visit at 8 weeks post‐treatment; 36 weeks total)

Enrolment: 408 participants randomized

Outcomes

Primary:

  • ADAS‐Cog

  • CIBIC‐plus

Secondary:

  • NPI total and individual scores

  • DAD

  • GDS

  • ADAS‐Noncog

  • MMSE

  • CIBIS‐plus

Notes

Study dates not reported. 24 sites in the USA included. Specific site locations not disclosed. Corresponding author’s institution: Washington University, St. Louis, MO, USA. The data in this report were collected from protocol D95‐018, sponsored by Bayer Corporation. For the purposes of this meta‐analysis, the authors have collected information from the double‐blind phase only.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "At the time of enrolment, the 408 patients in this study were randomized to the placebo (N=135) or the metrifonate (N=237) group..."
Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: "…according to a randomization code with blocks of six generated by computer at Bayer Corporation…"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: “...double‐blind...” “Only the statistician…had access to the randomization code…”
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: “...double‐blind...” “…investigators were masked as to random code assignment…”
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The reasons for withdrawal were provided in the participant flow diagram (figure 1)

Selective reporting (reporting bias)

High risk

LSM changes in NPI total and NPI‐subitem scores were extracted from figure 4. As LSM are adjusted for covariates, there is a risk of bias. Additionally, no standard error or standard deviation was reported in this paper. As a result, the authors of this meta‐analysis computed a standard deviation based on other studies (Raskind 1999) investigating the use of metrifonate in people with AD

Other bias

Low risk

No other identified biases

MSAD trial

Methods

Multi‐center, randomized, double‐blind, placebo controlled, parallel‐group study

Participants

  • diagnosis of "clinically probably" or "clinically possible" AD according to NINCDS‐ADRDA criteria

  • sMMSE score between 5 and 17

  • FAST score ≤ 6 at baseline

  • CT or MRI scan within the past 24 months consistent with AD pathology

Interventions

Participants were randomized to either:

  • donepezil (5 mg/day for the first 28 days and 10 mg/day thereafter per the clinician's judgement) (N = 144)

  • placebo (N = 146)

Duration: 24 weeks

Enrolment: 290 participants randomized

Outcomes

Outcomes were obtained from Gauthier 2002:
Primary:

  • NPI total and individual scores

Notes

We have selected Gauthier 2002 as the primary paper, but this group (MSAD investigators) have also reported on the study in two other published papers (Feldman 2001; Feldman 2005). Study dates not reported. 32 sites including 22 in Canada, 6 in Australia, and 4 in France. Specific site locations not disclosed. Corresponding author’s: McGill Centre for Studies in Aging, Montreal, Canada. The results of this study are supported by Pfizer, Inc. (New York, NY) and Eisai, Inc. (Teaneck, NJ).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...patients...were randomized to receive...".

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote from Feldman et al "At baseline, eligible patients were randomized in a 50/50 split using a computerized randomization schedule..."

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Quote from Feldman et al "Blinding was established with identical film‐coated tablets within a blister packaged card."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Study withdrawals and reason for withdrawals have been reported in Figure 1 in Feldman 2001, and appear to be balanced between groups

Selective reporting (reporting bias)

High risk

"The outcome measure of interest was 12‐item Neuropsychiatric Inventory (NPI)." The authors reported on baseline least‐square means (LSM) ± standard errors on each of the 12‐item NPI scores. As LSM are adjusted for covariates, there is a risk of bias. They also included LSM change scores in figure 1. However, they did not include standard errors of these mean change scores. As such, we approximate standard deviation of change scores by calculating the standard deviation from the standard error of baseline scores

Other bias

Low risk

No other identified biases

Padala 2017

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

  • Diagnosis of dementia of the Alzheimer type (DSM‐IV TR criteria)

  • MMSE score between 18 and 29

  • AES score > 40

  • Ability to provide informed consent by patient or caregiver

  • Stable dose of antidepressants for 2 months prior to study enrollment

  • Stable dose of ChEI and memantine for 4 months prior to study enrollment

Interventions

Participants were randomized to receive either:

  • methylphenidate (5 mg twice/day, titrated to 10 mg twice/day at 2 weeks)

  • placebo

  • psychosocial intervention provided for each group

Duration: 12‐week treatment period (+ 2 week discontinuation phase)

Enrollment: 60 participants randomized initially

Outcomes

Primary:

  • AES

Secondary:

  • MMSE

  • CGI

  • ADL

  • IADL

  • ZBD

Notes

Study dates: August 2007 ‐ June 2010. Study site: VA Medical Center, Omaha, NE, USA. This study was funded by a VA Merit Review Entry Program grant to Dr. Prasad Padala

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “…randomized double‐blind, placebo‐controlled trial...”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: "...randomized to methylphenidate (N=30) or placebo (N=30) groups using a random block design developed by a statistician using sealed envelopes."
Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: “…double‐blind…”

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “One subject dropped off from the placebo group.”

Comment: Although no participant flow diagram was provided in text, withdrawal information and reasoning described in text

Selective reporting (reporting bias)

Low risk

The authors reported mean ± standard deviations for apathy scores in each intervention arm

Other bias

Low risk

No other identified biases

Raskind 1999

Methods

Multicenter, randomized, double‐blind, placebo controlled, parallel‐group study

Participants

  • diagnosis of “probable” AD according to NINCDS‐ADRDA criteria

  • MMSE score between 10 and 26

  • modified HIS < 4

  • weighed between 43 and 98 kg (95‐215 lbs)

  • present caregiver with whom patient was in contact with at least 4 times a week

Interventions

Participants were randomized to either:

  • metrifonate (50 mg/daily) (N = 177)

  • placebo (N = 87)

Duration: 26 weeks (+ 8‐week post‐treatment follow‐up visit)

Enrollment: 264 patients randomized

Outcomes

Primary:

  • ADAS‐Cog

  • MMSE

  • NPI total and individual scores

  • ADAS‐Noncog

  • DAD

  • CIBIC‐Plus

  • CIBIS‐Plus and GDS

Secondary:

  • safety

Notes

Study dates not reported. Multicenter study in the USA. Specific number and site locations not disclosed. Corresponding author’s institution: Northwest Mental Illness Research, Education and Clinical Center, Washington, DC, USA.The data in this report were collected from protocol D96‐010, sponsored by Bayer Corporation. For the purposes of this meta‐analysis, we have collected information from the double‐blind phase only.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: '...The 264 patients enrolled in this study were randomly assigned to the placebo...or the metrifonate group...'Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: '...patients...were randomly assigned...according to a computer‐generated randomization code..."
Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: '...double‐blind...'.
Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: 'The investigators were blinded as to random code assignment'
Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The reasons for withdrawal were provided in text (page 322)

Selective reporting (reporting bias)

High risk

LSM changes in NPI total and NPI‐subitem scores were provided in figure 2. As LSM are adjusted for covariates, there is a risk of bias. SD was computed from provided SE of change scores

Other bias

Low risk

No other identified biases

Rosenberg 2013

Methods

Multicenter, randomized, double‐blind, placebo‐controlled trial

Participants

  • diagnosis of “possible” or “probable” AD according to NINCDS‐ADRDA criteria

  • clinical stability as judged by the local investigator

  • MMSE score ≥ 10

  • clinically significant apathy for at least 4 weeks (NPI apathy frequency of “often” or greater and an apathy severity of “moderate” or “marked”)

  • stable dose for the prior 3 months if treated with SSRIs or SNRI

Interventions

Participants were randomized to receive either:

  • Methylphenidate: 1x twice/day orally (10 mg/day) for first 3 days, tapered up to 2 x orally twice/day (20 mg/day) (N = 29)

  • Placebo (N = 31)

  • Psychosocial intervention provided for each group

Duration: 6 weeks

Enrollment: 60 participants randomized

Outcomes

Primary:

  • AES

  • ADCS‐CGI‐C

Secondary:

  • NPI apathy score

  • MMSE

  • safety: AEs, electrolyte panels and electrocardiogram results

Notes

Study dates: June 2010 ‐ August 2012. Study sites: 1) Johns Hopkins University Baltimore, MD, USA; 2) Medical University of South Carolina, Charleston, SC, USA; 3) Sunnybrook Health Sciences Centre,

Toronto, Ontario, Canada. Funding was provided by the National Institute on Aging (R01 AG033032‐01 and 1 K08 AG029157‐01A1).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “…randomized, double‐blind, placebo‐controlled multicenter trial...”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “The randomization scheme, stratified by clinical center with permuted length blocks, assigned participants to methylphenidate or placebo in a 1:1 ration. The coordinating center generated the treatment assignment schedule using a documented, auditable SAS program.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: “Study drug was supplied as identical‐appearing capsules containing either 5 mg methylphenidate or lactose (placebo).”

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The time to early termination did not differ significantly by group".
Comment. Probably done. Additionally, participant flow, CONSORT diagram is provided in figure 1

Selective reporting (reporting bias)

Low risk

Table 2 provides means ± standard errors for measures for the scores, change, and treatment effects of apathy (AES, ADCS‐CGI‐C and NPI) at 6 weeks of methylphenidate and placebo groups

Other bias

Low risk

No other identified biases

Ruths 2008

Methods

Multicenter, randomized, double‐blind, placebo‐controlled trial

Participants

  • ≥ 65 years old

  • long‐term (≥ 3 months) nursing home residents

  • diagnosis of dementia according to the ICD‐10 criteria

  • long‐term (≥ 3 months) stable dose of either risperidone, olanzapine, or haloperidol for BPSD

Interventions

Participants were randomly assigned to either:

  • placebo (antipsychotic drug discontinuation) (intervention group (IG)) (N = 27) or

  • continued antipsychotic drug treatment at current dose (no discontinuation) (reference group, RG) (N = 28)

Duration: 4‐week intervention

Enrollment: 55 participants randomized

Outcomes

Primary:

  • successful antipsychotic discontinuation, i.e. IG patients still off antipsychotic at study completion

Secondary:

  • NPI‐NH total scores, individual scores, and factor scores, as well as proportion improved/worsened behavior

Notes

Study dates not reported. 13 sites in Bergen and Oslo, Norway. Specific site locations not disclosed. Corresponding author’s institution: University of Kalfarveien, Bergen, Norway. Funding: No funding information provided.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Participants were consecutively assigned to antipsychotic drug discontinuation (intervention group, IG) or no discontinuation (reference group, RG) by means of computer generated, random, permuted blocks of four.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “All study medications were provided by an independent pharmacy… to maintain blindness.”,“…patients received identically looking capsules…”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The study was a multicenter double‐blind, controlled four week intervention."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "…double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “Seven patients completed the study prematurely (IG, n = 4; RG, n = 3; X2 = 0.20, p = 0.70), due to un‐blinding for randomization code (IG, n = 1; RG, n = 2), behavioural deterioration (IG, n = 2), restless legs (IG, n = 1) or delirium (RG, n = 1).”

Comment: Study withdrawals and reason for withdrawals are described and appear to be balanced between groups

Selective reporting (reporting bias)

Low risk

The authors reported the number of IG participants still on antipsychotics at study completion. Mean NPI total score difference were provided from baseline to Week 4. Changes in behavioral symptoms between groups are presented in Table 2. Means ± standard deviations of differences in change in BPSD between groups are presented in Table 3 for NPI total and factor scores. Additional information regarding NPI‐subscores were requested by the authors, and were provided by Dr. Sabine Ruths

Other bias

Unclear risk

This is a discontinuation study

Semgacestat trial

Methods

Multinational, randomized, double‐blind, placebo‐controlled trial

Participants

Participant information obtained from Doody 2013:

  • ≥ 55 years

  • diagnosis of mild‐to‐moderate AD according to NINCDS‐ADRDA criteria

  • MMSE score between 6 – 26 inclusive

  • lack of significant clinical depression (GDS score ≤ 6)

  • have a family member, caregiver, or other knowledgeable informant to provide information about symptoms

  • stable dose of ChEI and memantine allowed

Interventions

Participants were randomized to receive:

  • semagacestat (100 mg/day) (N = 507)

  • semagacestat (140 mg/day) (N = 529)

  • placebo (N = 501)

Duration: 76‐week treatment phase

Enrolment: 1537 patients randomized

Outcomes

Outcomes were obtained from Rosenberg 2016:
Primary:

  • NPI total and individual scores

Other:

  • ADAS‐Cog

  • MMSE

Notes

Study dates: March 2008 ‐ May 2011. Multicenter study in the USA. Specific number and site locations not disclosed. Although we have chosen Rosenberg 2016 as the primary paper, additional information regarding participant population was obtained from Doody 2013. Funding provided by Eli Lilly and the University of California at San Diego (the latter as a fiduciary for the Alzheimer’s Disease Cooperative Study), a clinical trials consortium established by the National Institute on Aging.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “participants were randomly assigned to receive…”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

This information has not been made available

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “Adverse events were more common in the two semagacestat groups than in the placebo group…The percentage of patients who discontinued the study drug because of adverse events was higher with semagacestat than with placebo (26% with 100 mg and 30% with 140 mg vs. 11% with placebo, P>0.001 for both comparisons).” This quote is from Doody 2013

Comment: Withdrawal percentages by group are provided in Doody 2013 and are not balanced between groups. AEs are presented in Table 3

Selective reporting (reporting bias)

Low risk

Change in apathy subscores are presented by treatment group in Figure C on p. 377 of Rosenberg 2016, but means ± SD scores needed to be computed by the authors of this meta‐analysis

Other bias

Low risk

No other identified biases

Sival 2002

Methods

Randomized, double‐blind, placebo‐controlled cross‐over trial

Participants

  • patients were admitted to the psychogeriatric short‐stay ward in a psychiatric teaching hospital

  • diagnosis of senile dementia according to DSM‐IV and NINCDS‐ADRDA criteria

  • clinically significant aggressive behavior according to Patel’s criteria (an overt act, involving the delivery of noxious stimuli to ‐ but not necessarily aimed at ‐ another object, organism or self, which is clearly not accidental)

  • score of ≥ 3 on at least 1 of the items of the SDAS‐9

Interventions

Participants were randomized to receive:

  • sodium valproate (2 x 6 ml of a 40 mg/ml suspension; 240 mg twice/day)

  • placebo

Duration: 8 weeks (1‐week baseline + 3‐week placebo period + 1‐week washout + 3‐week treatment phase with sodium valproate). Extension of the baseline period was allowed once for 1 week in participants who did not show a score ≥ 3 on one of the items of the SDAS‐9

Enrollment: 42 participants randomized

Outcomes

Primary:

  • SDAS‐9

  • CGI

Secondary:

  • GIP‐scales (apathy)

  • Nurse observation

Other:

  • MMSE

  • adverse events (especially symptoms of drowsiness, nausea, vomiting, diarrhea, confusion, disturbance in speech, disturbance of coordination, occurrence or enhancement of tremor, itching and loss of hair)

Notes

Study dates not reported. Study site: Parnassia Psycho Medical Center, The Hague, The Netherlands. Funding provided by a grant from the Van Helten Foundation, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands (grant number SHV94/AANV/5), a grant from the National Fund for Mental Health, Utrecht, The Netherlands (grant number 4145), and a grant from the Stichting tot Steun VCVGZ, Bennekom, The Netherlands (grant number ST07064BB. VE)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “The sequence of the treatment periods was assigned at random. The code was not accessible for the investigators.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: “During the ‘treatment period with placebo’ and during the wash‐out period a placebo suspension was given, identical to the active medication in appearance, quantity, smell and taste.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “…there were three drop‐outs. One patient had high fever of unknown origin during treatment with placebo, one patient was hit by a stroke during treatment with placebo, and one patient broke his hip during the wash‐out period. None of the dropouts could be associated with the intake of sodium valproate.”

Comment: Withdrawal numbers and reasons for withdrawal are provided in the Results section

Selective reporting (reporting bias)

Low risk

Mean change scores ± SD scores are presented in Table 2, demonstrating the effects of sodium valproate compared to placebo on aggressive behavior and other types of disturbed behavior (including the GIP apathetic behavior subscore)

Other bias

Low risk

No other identified biases

Sultzer 2008

Methods

Multicenter, randomized, double‐blind, placebo‐controlled trial

Participants

  • diagnosis of AD according to DSM‐IV criteria or “probable” AD according to NINCDS‐ADRDA criteria

  • MMSE score between 5 and 26

  • delusions, hallucinations, agitation or aggression had occurred nearly every day over the previous week or intermittently over 4 weeks

  • symptoms must be rated at least “moderate” in severity on the BPRS conceptual disorganization, suspiciousness, or hallucinatory behavior item, or had occurred at least weekly with "moderate” severity or greater on the delusion, hallucination, agitation or aberrant motor behavior subscale item on the NPI

  • if taking a ChEI, must be on a stable dose

Interventions

Participants were randomized initially (2:2:2:3 ratio) to either:

  • olanzapine (2.5 mg or 5 mg) (N = 100)

  • quetiapine (25 mg or 50 mg) (N = 94)

  • risperidone (0.5 mg or 1 mg) (N = 85)

  • placebo (N = 142)

Treating physician selected the number of low‐ or high‐dose capsules for initial treatment and could adjust the dosage, as indicated clinically, over 36 weeks of trial. At any time after the first 2 weeks of treatment, the clinician could discontinue the initially‐assigned (Phase 1) medication based on their clinical judgment. Phase 1 would end and the participant could enter Phase 2 and be assigned randomly to masked treatment with an atypical antipsychotic medication not assigned to them in Phase 1 or with citalopram. Participants could also go directly to an open‐choice treatment

Duration: Phase 1: 12 weeks; Phase 2: 24 weeks (36 weeks total)

Enrolment: 421 participants randomized

Outcomes

Psychiatric and behavioral symptoms:

  • NPI total scores

  • BPRS total and individual scores

  • CSDD

  • CGIC

Cognitive skills, functional abilities, care needs and quality of life:

  • ADAS‐Cog

  • MMSE

  • ADCS‐ADL

  • DS

  • CAS

  • ADRQL

Notes

For the purposes of this meta‐analysis, data from Phase 1 of the study were used. Phase 2 was not included as there was no placebo control. Study dates: March 2001 ‐ October 2004. 42 sites included. Site locations not disclosed. Principal Investigator institutions: University of Southern Carolina, Columbia, SC, USA; University of Rochester, Rochester, NY, USA. Funding was provided by the National Institute of Mental Health (N01 MH9001) and in part by the Department of Veterans Affairs. Astra‐Zeneca Pharmaceuticals, Forest Pharmaceuticals, Janssen Pharmaceutical and Eli Lilly provided medications for the study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “This protocol is fundamentally a randomized‐treatment assignment…”

Comment: Probably done. This quote is from Schneider 2003, where the CATIE‐AD research design and methods were originally described

Allocation concealment (selection bias)

Low risk

Quote: “Medication has been prepared into identically appearing ‘low strength’ capsules containing risperidone 0.5 mg, olanzapine 2.5 mg, quetiapine 25mg, citalopram 10 mg or placebo, or ‘higher strength’ capsules containing 1 mg,5 mg, 50 mg, 20 mg or placebo, respectively, in order to preserve the blind.”

Comment: Probably done. This quote is from Schneider 2003

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double blind..."

Comment: Probably done. This quote is from Schneider 2003

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done. This quote is from Schneider 2003

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote:“There were no significant overall differences among treatment groups with regard to the time to discontinuation of treatment for any reason”

“There were no significant differences among the groups with regards to the proportion of patients who had at least one serious adverse event and the proportion who had any adverse event”

Comment: Time to treatment discontinuation and adverse events are reported in Tables 2 and 3 and appear to be balanced between groups. These quotes are from Schneider 2006, where the CATIE‐AD time to treatment discontinuation and adverse events data are originally described

Selective reporting (reporting bias)

Low risk

Quote: “Two sets of clinical outcomes were measured: 1) psychiatric and behavioural symptoms… 2) cognition, functional skills, care needs and quality of life”

Comment: Baselines scores on clinical measures provided, along with mean change ± standard deviation scores for clinical symptoms from baseline to last observation in Phase 1. Mean change ± standard deviations on clinical symptom measures between baseline and treatment week 12 are provided for cognitive measures.

Other bias

Low risk

No other identified biases

Tariot 2001

Methods

Multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group study

Participants

  • diagnosis of "probably" or "possible" AD according to NINCDS‐ADRDA criteria

  • sMMSE score between 5 and 26

  • Score of 3 or 4 on at least 1 symptom on the NPI‐NH

Interventions

Participants were randomized to either:

  • donepezil (5 mg/day for the first 28 days and 10 mg/day thereafter per the clinician's judgement) (N = 103)

  • placebo (N = 105)

Duration: 24 weeks

Enrolment: 208 participants randomized

Outcomes

Primary:

  • NPI total and individual scores

Secondary:

  • MMSE

  • CDR‐Sum of the Boxes

  • Physical Self‐Maintenance Scale

Notes

Study dates not reported. 27 sites across the USA. Specific site locations not disclosed. Corresponding author’s institution: Departments of Psychiatry, Medicine and Neurology, University of Rochester Medical Center, Monroe Community Hospital, Rochester, NY, USA. The results of this study are supported by Pfizer, Inc. (New York, NY) and Eisai, Inc. (Teaneck, NJ).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...patients...were randomized..."

Comment: Probably done

Allocation concealment (selection bias)

Low risk

Quote: "...randomized in blocks of four, using a computerized randomization schedule..."

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Blinding was achieved using identical appearing film‐coated tablets of donepezil and placebo..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Study withdrawals and reason for withdrawals have been reported in the first paragraph of the Results section of Tariot 2001

Selective reporting (reporting bias)

High risk

LSM changes in NPI‐subitem scores were provided in figure 1. As LSM are adjusted for covariates, there is a risk of bias. SD was computed from provided SE of change scores

Other bias

Low risk

No other identified biases

Tariot 2011

Methods

Multicenter, randomized, double‐blind, placebo‐controlled, flexible‐dose trial

Participants

  • > 54 years old

  • community residents

  • diagnosis of “possible” or “probable” AD according to NINCDS‐ADRDA criteria

  • weight > 39 kg

  • MMSE score between 12 ‐ 20

  • NPI delusions, hallucinations and agitation/aggression < 1

Interventions

Participants were assigned to either:

  • valproate (1 tablet daily for 1 week, with a weekly titration continued until target dose of 10 ‐ 12 mg/kg of body weight per day) (N = 153)

  • placebo (N = 160)

Dose reduction was permitted if clinically warranted, and the target dose could be resumed if appropriate. Adherence of 80% was required

Duration: 24‐month double‐blind treatment phase + 2‐month single‐blind placebo treatment period

Enrolment: 313 participants randomized

Outcomes

Primary:

  • NPI score ≥ 3 on 1 or more items assessing delusions, hallucinations, and agitation/aggression persisting for 2 weeks

  • study physician’s judgment that the new agitation and/or psychosis was clinically significant on the basis of an evaluation to rule out situational disturbances or delirium; or completion of the study.

Secondary:

  • ADAS‐cog

  • CMAI

  • ADCS‐ADL

  • CDR‐SOB

  • ADCS‐CGIC

Other:

  • NPI total and individual score

  • QOL‐AD

  • MMSE

Safety and tolerability:

  • vital signs (weight, systolic and diastolic blood pressure, and temperature)

  • adherence

  • day/night time drowsiness

Volumetric Magnetic Resonance Image:

  • A subset of participants had MRI scans to assess effects of valproate vs placebo on whole‐brain volume, ventricular volume, and hippocampal volume

Notes

Study dates: October 2003 ‐ December 2009. 46 sites included. Site locations not disclosed. Study Director site: University of Rochester Medical Center, Rochester, NY, USA. Data for this study were obtained from the University of California, San Diego Alzheimer's Disease Cooperative Study Legacy Database. Funding provided by National Institute on Aging (U01AG010483). Additional support provided by a research grant and material support from Abbott Laboratories (NCT00071721).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Patients were assigned to 1 of 2 treatment groups in permuted blocks of 4, according to a randomization list created and maintained by the ADCS Data Core.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

“The trial used a 125‐mg enteric‐coated extended‐release divalproex sodium formulation or identical‐appearing placebo…”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "...double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “In the valproate group, 61.40% discontinued treatment prematurely, and in the placebo group, 60.6% did so; reasons are shown in Figure 1.”

Comment: Study withdrawals and reason for withdrawals have been reported in Figure 1 and all AEs experienced in each group are outlined in Table 3

Selective reporting (reporting bias)

Low risk

The authors reported means ± SD scores of each outcome over time for each study visit in Table 3. Additional data were provided upon request

Other bias

Low risk

No other identified biases

Trzepacz 2013

Methods

Multicenter, randomized, double‐blind, placebo‐controlled trial.

Participants

  • ≥ 60 years old community dwellings

  • reliable caregiver with frequent contact with participant

  • men or non‐fertile women

  • diagnosis of “probable” AD according to NINCDS‐ADRDA criteria and DSM‐IV‐TR criteria

  • clinically significant, persistent agitation/aggression that was disruptive to daily living or put themselves or others in harm's way for at least 3 days a week for at least 5 weeks prior to study entry

  • MMSE score between 6 to 26

  • NPI‐10 total score ≥ 10 at screening and randomization visits

  • NPI‐4‐A/A subscore ≥ 4 at screening and randomization visits

  • CT or MRI brain scan within 2 years that is consistent with AD

  • modified HIS scores ≤ 4

  • patients could not meet DSM‐IV‐TR criteria for delirium and/or have Delirium‐Rating‐Scale‐Revised‐98 score of ≥ 18

  • discontinuation from concomitant psychotropic medications and medically stable

  • stable doses of 4 antidepressant medications (sertraline, citalopram, escitalopram, fluoxetine) and AChEIs and memantine

Interventions

Participants were randomly assigned to either:

  • mibampator (3 mg/day) (N = 63)

  • placebo (N = 69)

After 1‐week of treatment twice a day with either 3 mg mibampator or placebo, a one‐time dose reduction to 1 mg twice daily due to intolerability was permitted, which remained their dose for the remainder of the study

Duration: 12‐week double‐blind treatment phase (+ 3 ‐ 28 day screening period and 1‐week single‐blind washout)

Enrolment: 132 participants randomized

Outcomes

Primary:

  • NPI‐4 A/A

Secondary:

  • CMAI

  • CSDD

  • FrSBe total and individual scores

  • CGI‐S‐AA

  • CGI‐S‐GF

  • ADAS‐Cog14

  • ADCS‐ADL

  • safety: TEAEs, laboratory test changes, vital signs changes, electrocardiograms

Notes

Study dates: February 2009 ‐ June 2011. Multicentre study in USA. Specific number and site location details are not disclosed. Research supported by Eli Lilly and Company

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “The interactive voice response system (IVRS) was used to assign blisterpacks containing double‐blind study drug to each patient.”

Comment: Probably done

Allocation concealment (selection bias)

Low risk

“The interactive voice response system (IVRS) was used to assign blisterpacks containing double‐blind study drug to each patient.”

Comment: Probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "…double‐blind..."

Comment: Probably done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "…double‐blind..."

Comment: Probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “There were no significant differences between groups for SAEs, discontinuation due to AEs, TEAEs or TEAEs possibly related to study drug as deemed by the investigator...”

Comment: Authors describe all SAEs and reasons for discontinuation reasons due to AE for each treatment group in the ‘Safety Evaluation’ section p. 8

Selective reporting (reporting bias)

High risk

The authors used MMRM analysis to assess the primary outcome, NPI‐4‐A/A least square mean change from baseline after treatment, (Figure 2), as well as secondary efficacy measures including FrSBe total and subscale least square means change from baseline after treatment (Figure 3.

Other bias

Low risk

No other identified biases

AChEI: anti‐cholinesterase inhibitor; AD: Alzheimer’s disease; ADAS‐Cog: Alzheimer’s Disease Assessment Scale‐Cognitive subscale; ADAS‐Cog11: Alzheimer’s Disease Assessment Scale‐ 11‐item cognitive subscale; ADAS‐Cog14: Alzheimer’s Disease Assessment Scale‐ 14‐item cognitive; ADAS‐Noncog: Alzheimer’s Disease Assessment Scale‐Noncognitive subscale; ADCS: Alzheimer’s Disease Cooperative Study; ADCS‐ADL: Alzheimer’s Disease Cooperative Study – Activities of Daily Living; ADCS‐ADL‐sev: Alzheimer’s Disease Cooperative Study‐Activities of Daily Living severity scale; ADCS‐CGI‐C: Alzheimer’s Disease Cooperative Study – Clinical Global Impression of Change; ADLQ: Activities of Daily Living Questionnaire; ADRQL: Alzheimer’s Disease Related Quality of Life; AE: adverse events; AES: Apathy Evaluation Scale; AIMS: Abnormal Involuntary Movement Scale; ARCI: Addiction Research Centre Inventory; BEHAVE‐AD: Behavioural Pathology in Alzheimer’s Disease Rating Scale; BID: twice daily; BPRS: Brief Psychiatric Rating Scale; BPSD: behavioral and psychological symptoms of dementia; CAS: Caregiver Activity Scale; CATIE‐AD: Clinical Antipsychotic Trials of Intervention Effectiveness‐Alzheimer’s Disease trial; CDR‐SOB: Clinical Dementia Rating sum of boxes; CGI: Clinical Global Impression scale; CGI‐C: Clinical Global Impression‐Change scale; CGI‐S: Clinical Global Impression‐Severity scale; CGI‐S‐AA: Clinical Global Impression‐Severity of Symptoms of Agitation/Aggression; CGI‐S‐GF: Clinical Global Impression‐Severity of Symptoms of Global Functioning; ChEI: Cholinesterase inhibitors; CIBIC‐plus: Clinician’s Interview‐Based Impression of Change plus Caregiver Input; CMAI: Cohen‐Mansfield Agitation Inventory; CPT: Conners’ Continuous Performance Task; CSDD: Cornell Scale for Depression in Dementia; CT: Computerized Tomography; DAD: Disability Assessment in Dementia; DAFS: Direct Assessment of Functional Status; D‐amph: dextroamphetamine challenge; DS: Dependence Scale; 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; FAST: Functional Assessment Staging of Alzheimer’s Disease; FrSBe: Frontal Systems Behaviour Scale; GDS: Global Deterioration Scale; HIS: Hachinski Ischemic Score; IADL: Instrumental Activities of Daily Living; ICD‐10: International Classification of Diseases‐ Tenth Revision; IG: intervention group; IR: immediate release; IVRS: interactive voice response system; kg: kilogram; lbs: pounds; L.L.F.: study neuropsychologist; LSM: least‐square means; LTC: long‐term care; mg: milligram; MMRM: mixed‐effects model repeated measures analysis; MMSE: Mini‐Mental State Exam; MRI: Magnetic Resonance Imaging; MSAD: moderate‐to‐severe AD; N: number; NINCDS‐ADRDA: National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association; NJ: New Jersey; NPI: Neuropsychiatric Inventory; NPI‐4 A/A: Neuropsychiatric Inventory‐4 domain subscale, which combines the following domains: agitation/aggression, aberrant motor behavior, irritability/emotional lability and disinhibition; NPI‐10: Neuropsychiatric Inventory‐10 domains; NPI‐NH: Neuropsychiatric Inventory‐Nursing Home Version; NRS: Neurobehavioural Rating Scale; NY: New York; PO: by mouth, in Latin per os; POMA: Performance‐Oriented Mobility Assessment‐II; POMS: Profile of Mood States; QOL‐AD: Quality of Life‐AD; QUALID: Quality of Life in Late‐Stage Dementia; RG: reference group; SAEs: serious adverse events; *SAS: Simpson‐Angus Scale; SAS: SAS version 9.2 (SAS Institute Inc, Cary, North Carolina); SDAS: Social Dysfunction and Aggression Scale; SD: standard deviation; SE: standard error; SIB: Severe Impairment Battery; sMMSE: Standardized Mini‐Mental State Exam; SNRI: serotonin and norepinephrine reuptake inhibitor; S.S.: study neurologist; SSRI: selective serotonin reuptake inhibitor; TEAEs: treatment emergent adverse events; ZBI: Zarit Burden Interview

Characteristics of excluded studies [ordered by study ID]

Jump to:

Study

Reason for exclusion

Ballard 2004

Apathy not reported

Ballard 2005

Apathy not investigated nor reported

Ballard 2008

Apathy not investigated nor reported

Banerjee 2011

Apathy not reported

Breder 2004

Apathy not investigated nor reported

Bridges‐Parlet 1997

Apathy not investigated nor reported

Brodaty 2003

Apathy not investigated nor reported

Burns 1999

Apathy not investigated nor reported

Cohen‐Mansfield 1999

Apathy not reported

De Deyn 1999

Apathy not investigated nor reported

De Deyn 2005

Apathy not reported

De Vasconcelos 2007

Apathy not investigated nor reported

Deberdt 2005

Apathy not reported

Devanand 2011

Apathy not reported

Devanand 2012

Apathy not reported

Findlay 1989

Apathy not reported

Holmes 2004

Apathy not reported

Howard 2012

Apathy not reported

Johannsen 2006

Apathy not reported

Katz 1999

Apathy not investigated nor reported

Lyketsos 2003

Apathy not reported

Magai 2000

Apathy not investigated nor reported

Mintzer 2006

Apathy not investigated nor reported

Nyth 1992

Apathy not investigated nor reported

Petracca 1996

Apathy not investigated nor reported

Petracca 2001

Apathy not investigated nor reported

Raskind 2000

Apathy not investigated nor reported

Reifler 1989

Apathy not investigated nor reported

Rosenberg 2010

Apathy not reported

Rosler 1999

Apathy not investigated nor reported

Roth 1996

Apathy not investigated nor reported

Satterlee 1995

Apathy not investigated nor reported

Schneider 2006

Apathy not reported

Seltzer 2004

Apathy not reported

Street 2000

Apathy not reported

Streim 2004

Apathy not reported

Tariot 2004a

Apathy not investigated nor reported

Tariot 2004b

Apathy not investigated or reported

Van Reekum 2002

Apathy not reported

Wilcock 2000

Apathy not investigated nor reported

Winblad 2001

Apathy not reported

Data and analyses

Open in table viewer
Comparison 1. Methylphenidate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the AES Show forest plot

3

145

Mean Difference (IV, Random, 95% CI)

‐4.99 [‐9.55, ‐0.43]

Analysis 1.1

Comparison 1 Methylphenidate, Outcome 1 Change in apathy from baseline as measured by the AES.

Comparison 1 Methylphenidate, Outcome 1 Change in apathy from baseline as measured by the AES.

1.1 < 12 weeks

2

85

Mean Difference (IV, Random, 95% CI)

‐2.62 [‐4.80, ‐0.44]

1.2 ≥ 12 weeks

1

60

Mean Difference (IV, Random, 95% CI)

‐9.90 [‐13.50, ‐6.30]

2 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐3.85, 3.69]

Analysis 1.2

Comparison 1 Methylphenidate, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 1 Methylphenidate, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore.

3 Adverse Events Show forest plot

3

145

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.67, 2.42]

Analysis 1.3

Comparison 1 Methylphenidate, Outcome 3 Adverse Events.

Comparison 1 Methylphenidate, Outcome 3 Adverse Events.

3.1 < 12 weeks

2

85

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.44, 3.72]

3.2 ≥ 12 weeks

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.73, 2.86]

4 Change in NPS from baseline as measured by the NPI Show forest plot

1

25

Mean Difference (IV, Fixed, 95% CI)

0.16 [‐7.89, 8.21]

Analysis 1.4

Comparison 1 Methylphenidate, Outcome 4 Change in NPS from baseline as measured by the NPI.

Comparison 1 Methylphenidate, Outcome 4 Change in NPS from baseline as measured by the NPI.

5 Change in cognition from baseline as measured by the MMSE Show forest plot

3

145

Mean Difference (IV, Fixed, 95% CI)

1.98 [1.06, 2.91]

Analysis 1.5

Comparison 1 Methylphenidate, Outcome 5 Change in cognition from baseline as measured by the MMSE.

Comparison 1 Methylphenidate, Outcome 5 Change in cognition from baseline as measured by the MMSE.

5.1 < 12 weeks study duration

2

85

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐0.49, 2.49]

5.2 ≥ 12 weeks study duration

1

60

Mean Difference (IV, Fixed, 95% CI)

2.6 [1.43, 3.77]

6 Change in functional permance from baseline as measured by the ADL Show forest plot

1

60

Mean Difference (IV, Fixed, 95% CI)

0.5 [‐0.39, 1.39]

Analysis 1.6

Comparison 1 Methylphenidate, Outcome 6 Change in functional permance from baseline as measured by the ADL.

Comparison 1 Methylphenidate, Outcome 6 Change in functional permance from baseline as measured by the ADL.

7 Change in functional performance from baseline as measured by the IADL Show forest plot

1

60

Mean Difference (IV, Fixed, 95% CI)

2.3 [0.74, 3.86]

Analysis 1.7

Comparison 1 Methylphenidate, Outcome 7 Change in functional performance from baseline as measured by the IADL.

Comparison 1 Methylphenidate, Outcome 7 Change in functional performance from baseline as measured by the IADL.

8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC Show forest plot

2

85

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.16, 2.11]

Analysis 1.8

Comparison 1 Methylphenidate, Outcome 8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC.

Comparison 1 Methylphenidate, Outcome 8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC.

9 Dropouts due to adverse events Show forest plot

3

145

Risk Ratio (M‐H, Fixed, 95% CI)

2.18 [0.64, 7.45]

Analysis 1.9

Comparison 1 Methylphenidate, Outcome 9 Dropouts due to adverse events.

Comparison 1 Methylphenidate, Outcome 9 Dropouts due to adverse events.

Open in table viewer
Comparison 2. Modafinil

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale Show forest plot

1

22

Mean Difference (IV, Fixed, 95% CI)

0.27 [‐3.51, 4.05]

Analysis 2.1

Comparison 2 Modafinil, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale.

Comparison 2 Modafinil, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale.

2 Change in functional performance from baseline as measured by the ADL‐Q Show forest plot

1

22

Mean Difference (IV, Fixed, 95% CI)

‐0.54 [‐1.40, 0.32]

Analysis 2.2

Comparison 2 Modafinil, Outcome 2 Change in functional performance from baseline as measured by the ADL‐Q.

Comparison 2 Modafinil, Outcome 2 Change in functional performance from baseline as measured by the ADL‐Q.

Open in table viewer
Comparison 3. Cholinesterase inhibitors

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs) Show forest plot

6

3598

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐0.80, ‐0.00]

Analysis 3.1

Comparison 3 Cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).

Comparison 3 Cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).

1.1 Licensed ChEIs (and ≤ 24 weeks study duration)

3

2531

Mean Difference (IV, Random, 95% CI)

‐0.21 [‐0.85, 0.43]

1.2 Unlicensed ChEIs ( and > 24 weeks study duration)

3

1067

Mean Difference (IV, Random, 95% CI)

‐0.63 [‐0.98, ‐0.29]

2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity) Show forest plot

6

3598

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐0.80, ‐0.00]

Analysis 3.2

Comparison 3 Cholinesterase inhibitors, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity).

Comparison 3 Cholinesterase inhibitors, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity).

2.1 Moderate AD (MMSE ≥ 18)

4

3100

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.79, ‐0.07]

2.2 Severe AD (MMSE < 18)

2

498

Mean Difference (IV, Random, 95% CI)

‐0.36 [‐1.82, 1.10]

Open in table viewer
Comparison 4. Discontinuation of cholinesterase inhibitors

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

40

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.88, 3.10]

Analysis 4.1

Comparison 4 Discontinuation of cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 4 Discontinuation of cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Open in table viewer
Comparison 5. Atypical antipsychotics

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score Show forest plot

2

1070

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

0.14 [0.00, 0.28]

Analysis 5.1

Comparison 5 Atypical antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score.

Comparison 5 Atypical antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score.

Open in table viewer
Comparison 6. Discontinuation of antipsychotics

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.24 [‐0.51, 0.03]

Analysis 6.1

Comparison 6 Discontinuation of antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 6 Discontinuation of antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Open in table viewer
Comparison 7. Antidepressants

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

2

126

Mean Difference (IV, Fixed, 95% CI)

‐1.24 [‐1.44, ‐1.04]

Analysis 7.1

Comparison 7 Antidepressants, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 7 Antidepressants, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Open in table viewer
Comparison 8. Mibampator

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the FrSBe‐apathy T score Show forest plot

1

132

Mean Difference (IV, Fixed, 95% CI)

‐1.2 [‐1.94, ‐0.46]

Analysis 8.1

Comparison 8 Mibampator, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy T score.

Comparison 8 Mibampator, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy T score.

Open in table viewer
Comparison 9. Valproate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore Show forest plot

3

257

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

0.02 [‐0.23, 0.26]

Analysis 9.1

Comparison 9 Valproate, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore.

Comparison 9 Valproate, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore.

Open in table viewer
Comparison 10. Semagacestat

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

939

Mean Difference (IV, Fixed, 95% CI)

0.20 [0.15, 0.25]

Analysis 10.1

Comparison 10 Semagacestat, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 10 Semagacestat, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

2Study flow diagram.
Figures and Tables -
Figure 1

2Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 2

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

Figure 3   CaptionRisk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 3

Figure 3   CaptionRisk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Forest plot of comparison: 7 Methylphenidate, outcome: 7.1 Apathy (AES only).
Figures and Tables -
Figure 4

Forest plot of comparison: 7 Methylphenidate, outcome: 7.1 Apathy (AES only).

Forest plot of comparison: 7 Methylphenidate, outcome: 7.3 Adverse Events.
Figures and Tables -
Figure 5

Forest plot of comparison: 7 Methylphenidate, outcome: 7.3 Adverse Events.

Forest plot of comparison: 3 Cholinesterase Inhibitors, outcome: 3.1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).
Figures and Tables -
Figure 6

Forest plot of comparison: 3 Cholinesterase Inhibitors, outcome: 3.1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).

Comparison 1 Methylphenidate, Outcome 1 Change in apathy from baseline as measured by the AES.
Figures and Tables -
Analysis 1.1

Comparison 1 Methylphenidate, Outcome 1 Change in apathy from baseline as measured by the AES.

Comparison 1 Methylphenidate, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore.
Figures and Tables -
Analysis 1.2

Comparison 1 Methylphenidate, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 1 Methylphenidate, Outcome 3 Adverse Events.
Figures and Tables -
Analysis 1.3

Comparison 1 Methylphenidate, Outcome 3 Adverse Events.

Comparison 1 Methylphenidate, Outcome 4 Change in NPS from baseline as measured by the NPI.
Figures and Tables -
Analysis 1.4

Comparison 1 Methylphenidate, Outcome 4 Change in NPS from baseline as measured by the NPI.

Comparison 1 Methylphenidate, Outcome 5 Change in cognition from baseline as measured by the MMSE.
Figures and Tables -
Analysis 1.5

Comparison 1 Methylphenidate, Outcome 5 Change in cognition from baseline as measured by the MMSE.

Comparison 1 Methylphenidate, Outcome 6 Change in functional permance from baseline as measured by the ADL.
Figures and Tables -
Analysis 1.6

Comparison 1 Methylphenidate, Outcome 6 Change in functional permance from baseline as measured by the ADL.

Comparison 1 Methylphenidate, Outcome 7 Change in functional performance from baseline as measured by the IADL.
Figures and Tables -
Analysis 1.7

Comparison 1 Methylphenidate, Outcome 7 Change in functional performance from baseline as measured by the IADL.

Comparison 1 Methylphenidate, Outcome 8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC.
Figures and Tables -
Analysis 1.8

Comparison 1 Methylphenidate, Outcome 8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC.

Comparison 1 Methylphenidate, Outcome 9 Dropouts due to adverse events.
Figures and Tables -
Analysis 1.9

Comparison 1 Methylphenidate, Outcome 9 Dropouts due to adverse events.

Comparison 2 Modafinil, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale.
Figures and Tables -
Analysis 2.1

Comparison 2 Modafinil, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale.

Comparison 2 Modafinil, Outcome 2 Change in functional performance from baseline as measured by the ADL‐Q.
Figures and Tables -
Analysis 2.2

Comparison 2 Modafinil, Outcome 2 Change in functional performance from baseline as measured by the ADL‐Q.

Comparison 3 Cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).
Figures and Tables -
Analysis 3.1

Comparison 3 Cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs).

Comparison 3 Cholinesterase inhibitors, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity).
Figures and Tables -
Analysis 3.2

Comparison 3 Cholinesterase inhibitors, Outcome 2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity).

Comparison 4 Discontinuation of cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.
Figures and Tables -
Analysis 4.1

Comparison 4 Discontinuation of cholinesterase inhibitors, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 5 Atypical antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score.
Figures and Tables -
Analysis 5.1

Comparison 5 Atypical antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score.

Comparison 6 Discontinuation of antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.
Figures and Tables -
Analysis 6.1

Comparison 6 Discontinuation of antipsychotics, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 7 Antidepressants, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.
Figures and Tables -
Analysis 7.1

Comparison 7 Antidepressants, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Comparison 8 Mibampator, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy T score.
Figures and Tables -
Analysis 8.1

Comparison 8 Mibampator, Outcome 1 Change in apathy from baseline as measured by the FrSBe‐apathy T score.

Comparison 9 Valproate, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore.
Figures and Tables -
Analysis 9.1

Comparison 9 Valproate, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore.

Comparison 10 Semagacestat, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.
Figures and Tables -
Analysis 10.1

Comparison 10 Semagacestat, Outcome 1 Change in apathy from baseline as measured by the NPI‐apathy subscore.

Summary of findings for the main comparison. Methylphenidate compared to placebo for apathy in Alzheimer's disease

Methylphenidate compared to placebo for apathy in Alzheimer's disease

Patient or population: Apathy in people with mild‐to‐moderate Alzheimer's disease
Setting: Multicenter, USA and Canada
Intervention: methylphenidate
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo

Risk with Methylphenidate

Change in apathy (AES score)
assessed with: AES
Scale from: 0 to 42
follow‐up: range 2 weeks to 12 weeks

The mean change from baseline in apathy was ‐4.2 to 0.6

MD 4.99 lower
(9.55 lower to 0.43 lower)

145
(3 RCTs)

⊕⊕⊝⊝
LOW 1, 2

AES: Limited data on clinically meaningful changes

Change in apathy (NPI‐apathy subscale score)
assessed with: NPI‐apathy subscale
Scale from: 0 to 12
follow‐up: 2 weeks to 6 weeks

The mean change from baseline in apathy ‐2.6 to ‐1.69

MD 0.08 lower
(3.85 lower to 3.69 higher)

85
(2 RCTs)

⊕⊕⊝⊝
LOW 1, 2

1‐ to 2‐point change suggested to be clinically significant in people with a clinically significant apathy (Rosenberg 2013)

Adverse events
assessed with: Number of participants reporting ≥ 1 adverse event
follow‐up: 2 weeks to 12 weeks

Study population

RR 1.28
(0.67 to 2.42)

145
(3 RCTs)

⊕⊕⊝⊝
LOW 1, 2

534 per 1000

684 per 1000
(358 to 1,000)

Change in NPS
assessed with: NPI
Scale from: 0 to 144
follow‐up: 2 weeks

The mean change from baseline in NPS was ‐2.08

MD 0.16 higher
(7.89 lower to 8.21 higher)

25
(1 RCT)

⊕⊕⊝⊝
LOW 1

4‐point change suggested to be clinically significant

Change in cognition
assessed with: MMSE
Scale from: 0 to 30
follow‐up: 2 weeks to 12 weeks

The mean change from baseline in cognition was ‐1.08 to ‐0.3

MD 1.79 higher
(0.53 higher to 3.05 higher)

145
(3 RCTs)

⊕⊕⊕⊝
MODERATE 1

MMSE: 2‐ to 4‐point change suggested to be clinically significant

Change in functional performance assessed with: ADL scale
Scale from: 0 to 6
follow‐up: 12 weeks

The mean change from baseline in functional performance was 0.4

MD 0.50 higher
(0.39 lower to 1.39 higher)

60
(1 RCT)

⊕⊕⊕⊝
MODERATE 3

Limited data on clinically meaningful changes

Change in functional performance assessed with: IADL scale
Scale from: 0 to 8 for women, and 0 to 5 for men, to avoid potential for gender bias
follow‐up: 12 weeks

The mean change from baseline in functional performance was ‐0.6

MD 2.30 higher
(0.74 higher to 3.86 higher)

60
(1 RCT)

⊕⊕⊕⊝
MODERATE 3

Limited data on clinically meaningful changes

Change in global disease severity
assessed with: ADCS‐CGIC or CGIC
follow‐up: 2 weeks to 6 weeks

Study population

RR 0.56
(0.15 to 2.10)

85
(2 RCTs)

⊕⊕⊕⊝
MODERATE 1

116 per 1000

65 per 1000
(17 to 244)

Dropouts
assessed with: Number of participants who dropped out prior to study completion.
follow‐up: 2 weeks to 12 weeks

Study population

RR 2.10
(0.60 to 7.38)

145
(3 RCTs)

⊕⊕⊝⊝
LOW 4

41 per 1000

86 per 1000
(25 to 303)

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

AD: Alzheimer's disease; AEs: Adverse Events; MMSE: Mini‐Mental Status Examination; MD: Mean Difference; NPS: Neuropsychiatric Symptom, SMD: Standardized Mean Difference, CI: Confidence interval; RR: Risk ratio; OR: Odds ratio

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

1Quality downgraded one level due imprecision (wide 95% confidence interval).
2Quality downgraded one level due to inconsistency (substantial heterogeneity was present).
3Quality downgraded one level due to imprecision (only one study, with a relatively small sample size).
4Quality downgraded two levels due to very serious imprecision (very wide 95% confidence interval).

Figures and Tables -
Summary of findings for the main comparison. Methylphenidate compared to placebo for apathy in Alzheimer's disease
Summary of findings 2. Modafinil compared to placebo for apathy in Alzheimer's disease

Modafinil compared to placebo for apathy in Alzheimer's disease

Patient or population: Apathy in people with mild‐to‐moderate Alzheimer's disease
Setting: Single site, USA
Intervention: modafinil
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo

Risk with Modafinil

Change in apathy
assessed with: FrSBe‐apathy subscale (T‐score converted from raw score)
Scale from: 14 to 70 (raw score)
follow‐up: mean 8 weeks

The mean change from baseline in apathy was ‐6.82

MD 0.27 higher
(3.51 lower to 4.05 higher)

22
(1 RCT)

⊕⊕⊝⊝
LOW 1

Limited data on clinically meaningful changes on the FrSBe apathy score

Adverse Events ‐ reported, but not analyzed in this review

Change in NPS ‐ not investigated

Change in cognition ‐ not investigated

Change in functional performance
assessed with: ADLQ
Scale from: 0 to 84
follow‐up: mean 8 weeks

The mean change from baseline in functional performance was 0

MD 0.54 lower
(1.40 lower to 0.32 higher)

22
(1 RCT)

⊕⊕⊝⊝
LOW 1

Limited data on clinically meaningful changes

Change in global disease severity ‐ not investigated

Dropouts ‐ reported, but not analyzed in this review

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

CI: Confidence interval; RR: Risk ratio; MD: mean difference

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

1Quality downgraded two levels due to small sample size and imprecision (wide 95% confidence interval).

Figures and Tables -
Summary of findings 2. Modafinil compared to placebo for apathy in Alzheimer's disease
Table 1. Study and participant characteristics

STUDY DURATION

N OF PARTICIPANTS

Diagnosis

MEAN AGE (YRS)

MEAN MMSE (SD)

MEAN BL NPI‐APATHY (SD) score

COUNTRY

NUMBER OF SITES

TREATMENT GROUPS

METHYLPHENIDATE

Herrmann 2008

2 weeks Cross‐over design: 2 treatment phases of 2 weeks with a 1‐week washout between phases

13 total

Possible or probable AD (NINCDS‐ADRDA), and apathy (NPI‐apathy subscale ≥ 1)

77.9 (7.8)

19.9 (4.7)

5.9 (3)

Canada

3

Group 1: Methylphenidate (10 mg twice a day)
Group 2: Placebo

Rosenberg 2013

6 weeks

60 participants
Group 1: 29
Group 2: 31

Possible or probable AD (NINCDS‐ADRDA), and clinically significant apathy for at least 4 weeks

76 (8)

20 (5)

Group 1: 7 (2)
Group 2: 7 (2)

USA, Canada

3

Group 1: Methylphenidate (target: 20 mg daily)
Group 2: Placebo

Padala 2017

12 weeks

60 participants
Group 1: 30
Group 2: 30

Dementia of the AD type (DSM‐IV‐TR), and presence of apathy (AES > 40)

76.6 (7.9)

23.8 (2.5)

Not reported

(AES only)

USA

1

Group 1: Methylphenidate (target: 20 mg daily)
Group 2: Placebo

MODAFINIL

Frakey 2012

8 weeks

Group 1: 11
Group 2: 11

Possible or probable AD (NINCDS‐ADRDA criteria) and clinically significant apathy (FrSBe Tscore ≥ 65)

Group 1: 75.3 (8.3)
Group 2: 29.4 (7.6)

Not disclosed

Not reported

USA

1

Group 1: Modafinil (200 mg daily)
Group 2: Placebo

CHOLINESTERASE INHIBITORS

Tariot 2001

24 weeks

Group 1: 103

Group 2: 105

Possible or probable AD with cerebrovascular disease (but not vascular dementia) (NINCDS‐ADRDA criteria)

Group 1: 85.4
Group 2:
85.9

Group 1: 14.4 (5.4)
Group 2: 14.4 (5.8)

Not reported

USA

27

Group 1: Donepezil ‐ 5 mg/day for 28 days. 10 mg/day after 28 days based on tolerability.
Group 2: placebo

MSAD trial

24 weeks

Group 1: 144
Group 2: 146

AD (DSM‐IV and NINCDS‐ADRDA criteria)

moderate‐severe AD

73.6

Group 1: 11.7 (0.35)
Group 2: 12.0 (0.34) **

Group 1:
3.48 (0.29)
Group 2:
3.48
(0.28)

Canada, Australia, France

32

Group 1: Donepezil ‐ 5 mg/day for 28 days. 10 mg/day after 28 days based on tolerability.
Group 2: placebo

Herrmann 2005

Range: 3 ‐ 6 months (12 ‐ 24 weeks)

Group 1: 1347
Group 2: 686

Probable AD (NINCDS‐ADRDA criteria)

mild‐moderate AD

76

18

Group 1:
2.34 (3.2)
Group 2:
2.32 (3.3)

USA, Canada, Great Britain, South Africa, Australia, and New Zealand

Multicenter, but number not disclosed

Group 1: Galantamine
Group 2: Placebo

Kaufer 1998

26 weeks

Group 1: 273
Group 2: 135

Probable AD (NINCDS‐ADRDA criteria)

mild‐moderate AD

Not reported

Not reported

Not reported

USA

25

Group 1: Metrifonate (2 weeks – 2.0 mg/kg, followed by 0.65 mg/kg)
Group 2: Placebo

Morris 1998

26 weeks

Group 1: 273
Group 2: 135

Probable AD (NINCDS‐ADRDA criteria)

mild‐moderate AD

Group 1: 73.5 (8.1)
Group 2: 73.7 (7.3)

Group 1: 18.8 (5)
Group 2: 19.4 (4.3)

Not reported

USA

24

Group 1: Metrifonate (2 weeks – 2.0 mg/kg, followed by 0.65 mg/kg)
Group 2: Placebo

Raskind 1999

26 weeks

Group 1: 177
Group 2: 87

Probable AD (NINCDS‐ADRDA criteria)

mild‐moderate AD

Group 1: 74.6 (8.3)
Group 2 : 74.5 (7.5)

Group 1: 18.7 (4.76)
Group 2: 18.7 (4.97)

Not reported

USA (additional sites are not disclosed)

Multicenter, but number not disclosed

Group 1:
50 mg, OD
Group 2: placebo

CHOLINESTERASE DISCONTINUATION

Herrmann 2016

8 weeks

Group 1: 21
Group 2: 19

Probable AD (NINCDS‐ADRDA criteria)

moderate‐severe AD

89.3

Group 1: 8.1(5.2)
Group 2: 10 (5.1)

Group 1:
3.29 (4.0)
Group 2:
2.16 (4.0)

Canada

2

Group 1: Donepezil,
rivastigmine,
galantamine (oral only)
Group 2: Placebo

ATYPICAL ANTIPSYCHOTICS

De Deyn 2004

10 weeks

Group 1: 132
Group 2: 125
Group 3: 134
Group 4: 129
Group 5: 129

Possible or probable AD (NINCDS‐ADRDA criteria and DSM‐IV‐TR), and clinically significant psychotic symptoms

76.6 (10.4)

13.7 (5.1)

Group 1: 3.2 (3.9)
Group 2: 3.2 (3.7)
Group 3: 3.4 (3.9)
Group 4: 3.4 (3.7)
Group 5: 3.0 (3.5)

Europe, Australia, Israel, Lebanon, and South Africa

61

Group 1: 7.5 mg OLZ
Group 2: 5 mg OLZ
Group 3: 2.5 mg OLZ
Group 4: 1.0 mg OLZ
Group 5: Placebo

Sultzer 2008

Up to 36 weeks (12 weeks of treatment) data available

Group 1: 100
Group 2: 94
Group 3: 85
Group 4: 142

Dementia of the AD type (DSM‐IV) or probable AD (NINCDS‐ADRDA) and daily delusions, hallucinations, agitation, or aggression over 4 weeks prior to study entry

77.9 (7.5)

Group 1: 15 (5.4)
Group 2: 14.9 (6.1)
Group 3: 15.7 (6.1)
Group 4: 14.7 (5.8)

Not reported

USA

42

Group 1: OLZ
Group 2: QUE
Group 3: RIS
Group 4: Placebo

ANTIPSYCHOTIC DISCONTUATION

Ruths 2008

4 weeks

Group 1: 28
Group 2: 27

Dementia diagnosis according to ICD‐10

83.4 (6.9)

Not provided

Group 1:
1.4 (1.5)Group 2: 1.9 (1.5)

Norway

9

Group 1: Antipsychotics (haloperidol, risperidone, or olanzapine)
Group 2:
Placebo

ANTIDEPRESSANTS

Lanctôt 2002

4 weeks: Cross‐over design: 2 treatment phases of 4 weeks with a 1‐week washout between phases

22 total

Primary degenerative dementia (DSM‐IV) and probable AD (NINCDS‐ADRDA), and significant behavioral problems (NPI ≥ 8)

82 (6)

4.1 (4.7)

Group 1:
1.27 (3.5)
Group 2:
1.45 (3.6)

Canada

3

Group 1: Sertraline (100 mg daily)
Group 2: Placebo

CitAD trial

9 weeks

Group 1: 94
Group 2: 92

Probable AD (NINCDS‐ADRDA), and significant behavioral problems (NPI ≥ 8), and clinically significant agitation on the NPI > 3

Group 1: 78 (9)
Group 2: 79 (8)

Group 1: 17 (6.2)
Group 2: 14.4 (6.9)

Group 1:
6 (0.9)
Group 2:
6 (0.9)

USA

6

Group 1: Citalopram (30 mg daily)
Group 2: Placebo

MIBAMPATOR

Trzepacz 2013

12 weeks

Group 1: 63
Group 2: 69

Probable AD (NINCDS‐ADRDA) (DSM‐IV‐TR), and clinically significant agitation/aggression

Group 1: 77.2 (8.2)
Group 2: 77.7 (7.6)

Group 1: 16.0 (6.1) Group 2: 18 (5.3)

Not reported

USA

Multicenter, but number not disclosed

Group 1: Mibampator (target dose: 3 mg daily)
Group 2: Placebo

VALPROATE

Herrmann 2007

6 weeks: Cross‐over design: 2 treatment phases of 6 weeks with a 2‐week washout between phases

Group 1: 14
Group 2: 13

Probable AD (NINCDS‐ADRDA), primary degenerative dementia (DSM‐IV)

85.6 (4.5)

4.5 (4.6)

Group 1: 2.4 (3.8)
Group 2:
3.0(4.3)

Canada

2

Group 1: Valproate (mean dose: 1134.6 (400.1) mg daily)
Group 2: Placebo

Sival 2002

3 weeks: Cross‐over design: 2 treatment phases of 3 weeks with a 1‐week washout between phases

Group 1: 42
Group 2: 42

Senile dementia (NINCDS‐ADRDA)(DSM‐IV)

80.4 (6.8)

11.4 (5)

Not reported

Netherlands

1

Group 1: Valproate (2 x 240 mg)
Group 2: Placebo

Tariot 2011

24 months (+ 2‐month single‐blind placebo phase)

Group 1: 153
Group 2: 160

Possible or probable AD (NINCDS‐ADRDA)

Group 1: 74.9
Group 2: 76.6

Group 1: 16.9 (3.0)
Group 2: 16.9 (2.9)

Group 1:
1.1 (2.7)
Group 2:
1.2 (2.9)

USA

46

Group 1: Valproate (flexible‐dose) (mean modal dose: 250 mg daily)
Group 2: Placebo

SEMAGACESTAT

Semgacestat trial

76 weeks

Group 1: 463
Group 2: 472
Group 3: 473

Mild‐moderate AD (NINCDS‐ADRDA)

Group 1: 72.7 (7.9)
Group 2: 73 (8.5)
Group 3: 73.3 (8)

Group 1: 20.9 (3.5)
Group 2: 20.8 (3.5)
Group 3: 20.9 (3.6)

Not reported

USA

91

Group 1: LY100
Group 2: LY140
Group 3: Placebo

AD: Alzheimer's disease, BL: baseline, DSM: Diagnostic and Statistical Manual of Mental Disoders, FrSBe: Frontal Systems Behavior Scale, ICD: International Classification of Diseases, LY: LY450319 (Eli Lillyand Company study drug), NINCDS‐ADRDA: National Institute of Neurological and Communicative disorders and the Alzheimer's Disease and Related Disorders Association, NPI: Neuropsychiatric Inventory, OLZ: olanzapine, QUE: quetiapine, RIS: risperidone, SD: standard deviation.

Figures and Tables -
Table 1. Study and participant characteristics
Table 2. Outcome Measures and Assessments

Study

Apathy

AE reported

NPS

Cognition

Function

Global Change

Dropouts due to AEs reported

METHYLPHENIDATE

Herrmann 2008

AES‐Informant

NPI‐apathy subscale

Yes

NPI‐total

MMSE

N/A

CGI‐C

Yes

Rosenberg 2013

AES‐Informant

NPI‐apathy subscale

Yes

NPI‐total (not reported)

MMSE

N/A

ADCS‐CGIC

Yes

Padala 2017

AES‐Clinician

Yes

N/A

MMSE

ADL
IADL

N/A

Yes

MODAFINIL

Frakey 2012

FrSBe‐apathy subscale

Yes

N/A

N/A

ADLQ

N/A

Yes

CHOLINESTERASE INHIBITORS

Tariot 2001

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

MSAD trial

NPI‐apathy subscale

Herrmann 2005

NPI‐apathy subscale

Kaufer 1998

NPI‐apathy subscale

Morris 1998

NPI‐apathy subscale

Raskind 1999

NPI‐apathy subscale

CHOLINESTERASE DISCONTINUATION

Herrmann 2016

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison.

ATYPICAL ANTIPSYCHOTICS

De Deyn 2004

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

Sultzer 2008

BPRS‐withdrawn depression factor score

ANTIPSYCHOTIC DISCONTINUATION

Ruths 2008

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

ANTIDEPRESSANTS

Lanctôt 2002

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

CitAD trial

NPI‐apathy subscale

MIBAMPATOR

Trzepacz 2013

FrSBe‐apathy T score

These outcomes were not investigated for this drug comparison

VALPROATE

Herrmann 2007

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

Sival 2002

GIP‐apathetic behavior subscore

These outcomes were not investigated for this drug comparison

Tariot 2011

NPI‐apathy subscale

These outcomes were not investigated for this drug comparison

SEMAGACESTAT

Semgacestat trial

NPI‐apathy

These outcomes were not investigated for this drug comparison

ADCS‐CGIC: Alzheimer's Diserase Cooperative Study ‐ Clinical Global Impression of Change, ADL: Activities of Daily Living scale, ADLQ: Lawton and Brody Fucntional Assessment, AES: Apathy Evaluation Scale, CGI‐C: Clinical Global Impression of Change, FrSBe: Frontal Systems Behavior Scale, GIP: The Behavior Observation Scale for Intramural Psychogeriatric Patients, IADL: Instrumental Activities of Daily Living Scale, MMSE: Mini‐Mental State Examination, NPI: Neuropsychiatric Inventory.

Figures and Tables -
Table 2. Outcome Measures and Assessments
Comparison 1. Methylphenidate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the AES Show forest plot

3

145

Mean Difference (IV, Random, 95% CI)

‐4.99 [‐9.55, ‐0.43]

1.1 < 12 weeks

2

85

Mean Difference (IV, Random, 95% CI)

‐2.62 [‐4.80, ‐0.44]

1.2 ≥ 12 weeks

1

60

Mean Difference (IV, Random, 95% CI)

‐9.90 [‐13.50, ‐6.30]

2 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐3.85, 3.69]

3 Adverse Events Show forest plot

3

145

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.67, 2.42]

3.1 < 12 weeks

2

85

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.44, 3.72]

3.2 ≥ 12 weeks

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.73, 2.86]

4 Change in NPS from baseline as measured by the NPI Show forest plot

1

25

Mean Difference (IV, Fixed, 95% CI)

0.16 [‐7.89, 8.21]

5 Change in cognition from baseline as measured by the MMSE Show forest plot

3

145

Mean Difference (IV, Fixed, 95% CI)

1.98 [1.06, 2.91]

5.1 < 12 weeks study duration

2

85

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐0.49, 2.49]

5.2 ≥ 12 weeks study duration

1

60

Mean Difference (IV, Fixed, 95% CI)

2.6 [1.43, 3.77]

6 Change in functional permance from baseline as measured by the ADL Show forest plot

1

60

Mean Difference (IV, Fixed, 95% CI)

0.5 [‐0.39, 1.39]

7 Change in functional performance from baseline as measured by the IADL Show forest plot

1

60

Mean Difference (IV, Fixed, 95% CI)

2.3 [0.74, 3.86]

8 Change in global disease severity from baseline as measured by the CGIC and the ADCS‐CGIC Show forest plot

2

85

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.16, 2.11]

9 Dropouts due to adverse events Show forest plot

3

145

Risk Ratio (M‐H, Fixed, 95% CI)

2.18 [0.64, 7.45]

Figures and Tables -
Comparison 1. Methylphenidate
Comparison 2. Modafinil

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the FrSBe‐apathy subscale Show forest plot

1

22

Mean Difference (IV, Fixed, 95% CI)

0.27 [‐3.51, 4.05]

2 Change in functional performance from baseline as measured by the ADL‐Q Show forest plot

1

22

Mean Difference (IV, Fixed, 95% CI)

‐0.54 [‐1.40, 0.32]

Figures and Tables -
Comparison 2. Modafinil
Comparison 3. Cholinesterase inhibitors

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with licensed versus unlicensed ChEIs) Show forest plot

6

3598

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐0.80, ‐0.00]

1.1 Licensed ChEIs (and ≤ 24 weeks study duration)

3

2531

Mean Difference (IV, Random, 95% CI)

‐0.21 [‐0.85, 0.43]

1.2 Unlicensed ChEIs ( and > 24 weeks study duration)

3

1067

Mean Difference (IV, Random, 95% CI)

‐0.63 [‐0.98, ‐0.29]

2 Change in apathy from baseline as measured by the NPI‐apathy subscore (subgroup analysis with disease severity) Show forest plot

6

3598

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐0.80, ‐0.00]

2.1 Moderate AD (MMSE ≥ 18)

4

3100

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.79, ‐0.07]

2.2 Severe AD (MMSE < 18)

2

498

Mean Difference (IV, Random, 95% CI)

‐0.36 [‐1.82, 1.10]

Figures and Tables -
Comparison 3. Cholinesterase inhibitors
Comparison 4. Discontinuation of cholinesterase inhibitors

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

40

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.88, 3.10]

Figures and Tables -
Comparison 4. Discontinuation of cholinesterase inhibitors
Comparison 5. Atypical antipsychotics

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore and the BPRS withdrawn depression factor score Show forest plot

2

1070

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

0.14 [0.00, 0.28]

Figures and Tables -
Comparison 5. Atypical antipsychotics
Comparison 6. Discontinuation of antipsychotics

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.24 [‐0.51, 0.03]

Figures and Tables -
Comparison 6. Discontinuation of antipsychotics
Comparison 7. Antidepressants

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

2

126

Mean Difference (IV, Fixed, 95% CI)

‐1.24 [‐1.44, ‐1.04]

Figures and Tables -
Comparison 7. Antidepressants
Comparison 8. Mibampator

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the FrSBe‐apathy T score Show forest plot

1

132

Mean Difference (IV, Fixed, 95% CI)

‐1.2 [‐1.94, ‐0.46]

Figures and Tables -
Comparison 8. Mibampator
Comparison 9. Valproate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore and GIP‐apathy subscore Show forest plot

3

257

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

0.02 [‐0.23, 0.26]

Figures and Tables -
Comparison 9. Valproate
Comparison 10. Semagacestat

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in apathy from baseline as measured by the NPI‐apathy subscore Show forest plot

1

939

Mean Difference (IV, Fixed, 95% CI)

0.20 [0.15, 0.25]

Figures and Tables -
Comparison 10. Semagacestat