Scolaris Content Display Scolaris Content Display

Inhibidores selectivos de la recaptación de serotonina (ISRS) para la recuperación del accidente cerebrovascular

Contraer todo Desplegar todo

Referencias

Acler 2009 {published data only}

Acler M, Avesani A, Fiaschi A, Manganotti P. Serotonergic modulation of brain excitability and motor recovery in patients affected by stroke. A double blind placebo RCT. International Journal of Stroke 2008;3:336. CENTRAL
Acler M, Robol E, Fiaschi A, Manganotti P. A double blind placebo RCT to investigate the effects of serotonergic modulation on brain excitability and motor recovery in stroke patients. Journal of Neurology 2009;256(7):1152-8. CENTRAL

AFFINITY 2020 {published data only}

ACTRN12611000774921. Assessment of fluoxetine in stroke recovery (AFFINITY) trial, 2011. www.anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12611000774921 (first received 22 July 2011). CENTRAL
ACTRN12619000986178. The assessment of fluoxetine in stroke recovery imaging and blood biomarkers sub-study AFFINITY BM. www.anzctr.org.au/Trial/Registration/TrialReview.aspx ?ACTRN=12619000986178 (first received 11 July 2019). CENTRAL
AFFINITY Trial Collaboration. Safety and efficacy of fluoxetine on functional outcome after acute stroke (AFFINITY): a randomised, double-blind, placebo-controlled trial. Lancet Neurology 2020;19(8):651-60. CENTRAL
Hankey G. Assessment oF FluoxetINe In sTroke recoverY (AFFINITY). www.affinitytrial.org/ (accessed 19th February 2019). CENTRAL

Almeida 2006 {published data only}

Almeida OP, Waterreus A, Hankey GJ. Preventing depression after stroke: results from a randomized placebo-controlled trial. Journal of Clinical Psychiatry 2006;67(7):1104-9. CENTRAL

Andersen 1994 {published data only}

Andersen G, Vestergaard K, Lauritzen L. Effective treatment of post-stroke depression with selective serotonin reuptake inhibitors. Journal of Neurology 1994;241:S42. CENTRAL
Andersen G, Vestergaard K, Lauritzen L. Effective treatment of post-stroke depression with the selective serotonin reuptake inhibitor citalopram. Stroke 1994;25(6):1099-104. CENTRAL
Andersen G, Vestergaard K, Lauritzen L. Post-stroke depression treated with citalopram. Acta Neurologica Scandinavica 1994;89 Suppl 155:20. CENTRAL
Andersen G, Vestergaard K, Lauritzen L. Post-stroke depression treated with citalopram - a selective serotonin reuptake inhibitor. Canadian Journal of Neurological Sciences 1993;20:S115. CENTRAL
Andersen G, Vestergaard K, Lauritzen L. Post-stroke depression treated with citalopram a selective serotonin reuptake inhibitor. In: 7th Scandinavian Meeting on Cerebrovascular Disease. 1993:54. CENTRAL
Andersen G, Vestergaard K, Lauritzen LU. Effective treatment of depression following apoplexy with citalopram. Ugeskrift for Laeger 1995;157(14):2000-3. CENTRAL
Flicker C, Andersen G, Bayer L. A placebo controlled study of citalopram treatment for post-stroke depression. In: 11th Annual Meeting of the American Association for Geriatric Psychiatry. 1998. CENTRAL

Andersen 2013 {published data only}

Andersen G. The efficacy of citalopram treatment in acute stroke (TALOS). www.clinicaltrials.gov/ct2/show/NCT01937182 (first received 9 September 2013). CENTRAL
Damsbo AG, Mortensen JK, Kraglund KL, Johnsen SP, Andersen G, Blauenfeldt RA. Prestroke physical activity and poststroke cognitive performance. Cerebrovascular Diseases 2020;49(6):632-8. CENTRAL
Kraglund KL, Mortensen JK, Damsbo AG, Modrau B, Simonsen SA, Iversen, HK, et al. Neuroregeneration and vascular protection by citalopram in acute ischemic stroke (TALOS). Stroke 2018;49(11):2568-76. CENTRAL
Kraglund KL, Mortensen JK, Damsbo AG, Modrau B, Simonsen SA, Iversen HK, et al. Neuroregeneration and vascular protection by citalopramin in acute ischemic stroke (TALOS). Stroke 2018;49(11):2568-76. CENTRAL
Kraglund KL, Mortensen JK, Grove EL, Johnsen SP, Andersen G. TALOS: a multicenter, randomised, double blind, placebo controlled trial to test the effects of citalopram in patients with acute stroke; protocol. International Journal of Stroke 2015;10:985-87. CENTRAL
Kraglund KL, Mortensen JK, Johnsen SP, Andersen G, Grove EL. Antiplatelet effects of citalopram in patients with ischaemic stroke: a randomized, placebo-controlled, double-blind study. Scientific Reports 2019;9(1):200482019. CENTRAL

Asadollahi 2018 {published data only}

Asadollahi M, Ramezani M, Khanmoradi Z, Karimialavijeh E. The efficacy comparison of citalopram, fluoxetine, and placebo on motor recovery after ischemic stroke: a double-blind placebo-controlled randomized controlled trial. Clinical Rehabilitation 2018;32(8):1069-75. CENTRAL

Bembenek 2020 {published data only}

Bembenek JP, Niewada M, Klysz B, Mazur A, Kurczych K, Gluszkiewicz M, et al. Fluoxetine for stroke recovery improvement - the doubleblind, randomised placebo-controlled FOCUS-Poland trial. Neurologia i Neurochirurgia Polska 2020;54(6):544-51. CENTRAL
Fluoxetine or control under supervision: Poland. Polish Office for Registration of Medicinal Products, Medical Devices and Biocidal Products - EduraCT No: 2014-001335-372014. CENTRAL

Birchenall 2019 {published and unpublished data}

Birchenall J, Térémetz M, Roca P, Lamy JC, Oppenheim C, Maier MA, et al. Individual recovery profiles of manual dexterity, and relation to corticospinal lesion load and excitability after stroke - a longitudinal pilot study. Neurophysiologie Clinique 2019;49(2):149-64. CENTRAL
NCT02063425. Evaluation by transcranial magnetic stimulation of the benefit of fluoxetine on motor recovery after stroke (EFLUSTIM). clinicaltrials.gov/ct2/show/NCT02063425 (first received 14 February 2014). CENTRAL

Brown 1998 {published data only}

Brown KW, Sloan RL, Pentland B. Fluoxetine as a treatment for post-stroke emotionalism. Acta Psychiatrica Scandinavica 1998;98(6):455-8. CENTRAL

Burns 1999 {published data only}

Burns A, Russell E, Stratton-Powell H, Tyrell P, O'Neill P, Baldwin R. Sertraline in stroke-associated lability of mood. International Journal of Geriatric Psychiatry 1999;14(8):681-5. CENTRAL

Cao 2020 {published data only}

Cao JX, Liu L, Sun YT, Zeng QH, Wang Y, Chen JC. Effects of the prophylactic use of escitalopram on the prognosis and the plasma copeptin level in patients with acute cerebral infarction. Brazilian Journal of Medical and Biological Research 2020;53(11):e8930. CENTRAL
Cao JX, Liu L, Sun YT, Zeng QH, Yang ZD, Chen JC. Escitalopram improves neural functional prognosis and endothelial dysfunction in patients with acute cerebral infarction. Restorative Neurology and Neuroscience 2020;38(5):385-93. CENTRAL

Chen 2001 {published data only}

Chen WY, Liu FY, Yang AP. Study of effect of integrative Chinese herbs with fluoxetine on rehabilitation of neurological impairment in patients with post-stroke depression. Journal of Chengdu University of Traditional Chinese Medicine 2001;24(4):20-3. CENTRAL

Chen 2002 {published data only}

Chen W, Wang G-F, Chen X-H, Sheng Y-L, Zhu H. Effects of paroxetine on function recovery in patients with post-stroke depression. Chinese Journal of Clinical Rehabilitation 2002;6(13):2014-5. CENTRAL

Chen 2005a {published data only}

Chen KN. Changes of neurotransmitter in patients with post-stroke depression observed with encephalofluctuography technology. Chinese Journal of Clinical Rehabilitation 2005;9(16):118-9. CENTRAL

Chen 2005b {published data only}

Chen T, Li J, Han M. A study on paroxetine in the treatment for post-stroke depression. Jiangxi Medicine 2005;40(7):382-4. CENTRAL

Chen 2015 {published data only}

Chen Z, Xu X, Wu Y, Huang L. Clinical efficacy of drug intervention in patients with acute depression after stroke. International Journal of Psychiatry 2015;42(5):97-100. CENTRAL

Cheng 2003 {published data only}

Cheng F, Shao G, Bao S. Study of effect on neurologic function rehabilitation in patient with post-stroke depression. Chinese Journal of Clinical Rehabilitation 2003;7(1):108-9. CENTRAL

Chollet 2011 {published data only}

Chollet F, Tardy J, Albucher J-F, Thalamas E, Berard E, Lamy C, et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurology 2011;10(2):123-30. CENTRAL

Dam 1996 {published data only}

Dam M, Tonin P, De Boni A, Pizzolato G, Casson S, Ermani M, et al. Effects of fluoxetine and maprotiline on functional recovery in poststroke hemiplegic patients undergoing rehabilitation therapy. Stroke 1996;27(7):1211-4. CENTRAL

Dike 2019 {published data only}

Dike F. Pharmacological enhancement of motor function recovery in patients with ischaemic stroke: a trial of fluoxetine. www.pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=967 (last accessed 16 February 2021). CENTRAL
Dike FO, Ekeh BC, Ogun AS, Ekrikpo EU, Walshak P, Ogunniy AA. Pharmacological enhancement of motor function recovery in patients with ischaemic stroke: a trial of fluoxetine. Journal of Neurology and Stroke 2019;9(1):47-51. CENTRAL

EFFECTS 2020 {published data only}NCT02683213

EFFECTS Trial Collaboration. Safety and efficacy of fluoxetine on functional recovery after acute stroke (EFFECTS): a randomised, double-blind, placebo-controlled trial. Lancet Neurology 2020;19(8):661-9. CENTRAL
ISRCTN13020412. Establishing the effect(s) and safety of fluoxetine initiated in the acute phase of stroke. www.isrctn.com/ISRCTN13020412 (first received 19 December 2014). CENTRAL
Lundström E, Isaksson E, Näsman P, Wester P, Mårtensson B, Norrving B. Correction to: Update on the EFFECTS study of fluoxetine for stroke recovery: a randomised controlled trial in Sweden. Trials 2020;21(388):s13063-020-04327-w. CENTRAL
Lundström E, Isaksson E, Näsman P et al. Update on the EFFECTS study of fluoxetine for stroke recovery: a randomised controlled trial in Sweden. Trials 2020;21(233):s13063-020-4124-7. CENTRAL
Lundstrom E, Isaksson E, Nasman P, Wester P, Martensson B, Norrving B, et al. Safety and efficacy of fluoxetine on functional recovery after acute stroke (EFFECTS): a randomised, double-blind, placebo-controlled trial. Lancet Neurology 2020;19(8):661‐9. CENTRAL

Feng 2004 {published data only}

Feng B-L, Wang Q-C, Zheng-Yuan LI. Influence of Jieyu Huoxue decoction on rehabilitation of patients with depression after cerebral infarction. Journal of Chinese Integrated Medicine 2004;2(3):182-4. CENTRAL

FOCUS 2019 {published data only}

Anonymous. The FOCUS (Fluoxetine or Control under Supervision) trial results: effects of a 6 month course of fluoxetine on the Stroke Impact Scale, mood and fatigue in patients with stroke. www.ed.ac.uk/clinical-brain-sciences/research/completed-studies-trials/focus-stroke-trial (accessed 12 02 2021). CENTRAL
Cook R, Thomas V, Martin R. Does fluoxetine improve recovery after stroke? BMJ 2019;364:1029. CENTRAL
Dennis M, Forbes J, Graham C, Hackett M, Hankey GJ, House A, et al. Fluoxetine to improve functional outcomes in patients after acute stroke: the FOCUS RCT. Health Technology Assessment 2020;24(22):1-94. CENTRAL
Dennis M, Forbes J, Graham C, Hackett ML, Hankey GJ, House A, et al. Fluoxetine and fractures after stroke: exploratory analyses from the FOCUS Trial. Stroke 2019;50(11):3280-2. CENTRAL
FOCUS Trial Collaboration. Effects of fluoxetine on functional outcomes after acute stroke (FOCUS): a pragmatic, double-blind, randomised, controlled trial. Lancet 2019;393(10168):265-74. CENTRAL
Graham C, Lewis S, Forbes J, Mead G, Hackett ML, Hankey GJ, et al. The FOCUS, AFFINITY and EFFECTS trials studying the effect(s) of fluoxetine in patients with a recent stroke: statistical and health economic analysis plan for the trials and for the individual patient data meta-analysis. Trials 2017;18(1):627. CENTRAL
Lundstrom E, Dennis M, Mead G. The effects of fluoxetine on fracture risk after stroke: Further analyses from the focus trial. European Stroke Journal 2019;4 suppl 1:7142019. CENTRAL
Mead G, Dennis M. Feasibility of reporting results of large randomised controlled trials to participants: experience from the Fluoxetine or Control under Supervision (FOCUS) trial. BMJ Open 2020;10(11):e0404922020. CENTRAL
Mead G. Fluoxetine Or Control Under Supervision (FOCUS) trial: to establish the effect(s) of routine administration of Fluoxetine in patients with a recent stroke. www.isrctn.com/ISRCTN83290762 (first received 23 May 2012). CENTRAL
Mead GE, Dennis MS, Innes K, MacLeod M, Sandercock PA, House A, et al. A multicentre randomised trial to establish the effect(s) of routine administration of fluoxetine in patients with a recent stroke (Fluoxetine Or Control Under Supervision, FOCUS). In: 21st European Stroke Conference. 2012:Abst OAID 26. CENTRAL
Mead GE, Graham C, Billot L, Näsman P, Lundström E, Lewis S, on behalf of the FOCUS , AFFINITY and EFFECTS trialists. Update to the FOCUS, AFFINITY and EFFECTS trials studying the effect(s) of fluoxetine in patients with a recent stroke: statistical analysis plan for the trials and for the individual patient data meta-analysis. Trials 2020;21(1):971. CENTRAL
Mead GE, Legg L, Tilney R, Hsieh CF, Wu S, Lundström E, et al. Fluoxetine for stroke recovery: meta-analysis of randomized controlled trials. International Journal of Stroke 2020;15(4):365-76. CENTRAL

Fruehwald 2003 {published data only}

Fruehwald S, Gatterbauer E, Rehak P, Baumhackl U. Early fluoxetine treatment of post-stroke depression: a three-month double-blind placebo-controlled study with an open-label long-term follow up. Journal of Neurology 2003;250(3):347-51. CENTRAL

Gao 2017 {published data only}

Gao J, Lin M, Zhao J, Bi S, Ni Z, Shang X. Different interventions for post-ischaemic stroke depression in different time periods: a single-blind randomized controlled trial with stratification by time after stroke. Clinical Rehabilitation 2017;31(7):71-81. CENTRAL

GlaxoSmithKline 1998 {published data only}

GlaxoSmithKline. An 8-week double-blind placebo controlled parallel group study to assess the efficacy and tolerability of paroxetine in patients suffering from depression following stroke. GSK Clinical Study Register www.gsk clinicalstudyregister.com (accessed 31 August 2012). CENTRAL

Gong 2020 {published data only}

Gong L, Yang X, Feng Y, Fei Z, Wang M, Qin B, et al. The efficacy of integrative anti-depressive therapy on motor recovery after ischemic stroke: a randomized clinical trial. European Journal of Integrative Medicine 2020;35:101102. CENTRAL

Guo 2009 {published data only}

Guo R-Y, Su L, Wang CX. Effects of Linggui Bafa on the therapeutic effect and quality of life in patients of post-stroke depression. Chinese Acupuncture & Moxibustion 2009;29(10):785-90. CENTRAL

He 2004 {published data only}

He P. Randomized controlled observation on the effect of early application of fluoxetine in preventing depression after stroke. Chinese Journal of Clinical Rehabilitation 2004;8(28):6016-7. CENTRAL

He 2005 {published data only}

He Y, Wang X, Xiao C. Prospective study of effects of paroxetine with mental intervention on depression and anxiety after stroke. Nervous Diseases and Mental Health 2005;5(1):6-13. CENTRAL
Wang X, He Y, Xiao C-L. A clinical trial of paroxetine and psychotherapy in patients with post-stroke depression and anxiety. Chinese Mental Health Journal 2005;19:564-6. CENTRAL

He 2016 {published data only}

ChiCTR-TRC-12002078. Multi-center randomized clinical study of antidepressant treatment (fluoxetine) on secondary prevention of ischemic stroke. www.chictr.org.cn/showprojen.aspx?proj=7471 (first received 3 April 2012). CENTRAL
Guo Y, He Y, Tang B, Ma K, Cai Z, Zeng S, et al. Effect of using fluoxetine at different time windows on neurological functional prognosis after ischaemic stroke. Restorative Neurology and Neuroscience 2016;34:177-87. CENTRAL
He Y, Cai Z, Zeng S, Chen S, Tang B, Liang Y, et al. Effect of fluoxetine on three-year recurrence in acute ischemic stroke: a randomized controlled clinical study. Clinical Neurology and Neurosurgery 2018;168:1-6. CENTRAL
He Y-T, Tang B-S, Cai Z-L, Zeng S-L, Jiang X, Guo Yi. Effects of fluoxetine on neural functional prognosis after ischemic stroke: a randomized controlled study in China. Journal of Stroke and Cerebrovascular Diseases 2016;25(4):761-70. CENTRAL

Hu 2002 {published data only}

Hu Y, Suo A, Xiang L. The comparative study of the effectiveness of fluoxetine on the stroke patients with depressive symptoms. Shanghai Archives of Psychiatry 2002;14:149-50. CENTRAL

Hu 2018 {published data only}

Hu H, Wang M. Effects of escitalopram on post-stroke depression and cognitive function of patients with neurological function. Hebei Medicine 2018;24(2):325-8. CENTRAL

Huang 2002 {published data only}

Huang X-H. The clinical correlation study and the effect of fluoxetine intervention on poststroke depression. Chinese Journal of Clinical Rehabilitation 2002;6(15):2296-7. CENTRAL

Jia 2005 {published data only}

Jia W. Effect of early intervention on recovery of motor function and recurrent stroke in patients with post-stroke depression. Chinese Journal of Clinical Rehabilitation 2005;9(12):4-5. CENTRAL

Kim 2017 {published data only}

Kim JS, Lee E-J, Chang D-ll, Park J-H, Ahn SH, Cha J-K, et al. Efficacy of early administration of escitalopram on depressive and emotional symptoms and neurological dysfunction after stroke: a multicentre, double-blind, randomised, placebo-controlled study. Lancet Psychiatry 2017;4(1):33-41. CENTRAL
Lee EJ, Kim JS, Chang DI, Park JH, Ahn SH, Cha JK, et al. Differences in therapeutic responses and factors affecting post-stroke depression at a later stage according to baseline depression. Journal of Stroke 2018;20(2):258-67. CENTRAL
Lee EJ, Kim JS, Chang DI, Park JH, Ahn SH, Cha JK, et al. Post-stroke depressive symptoms: varying responses to escitalopram by individual symptoms and lesion location. Journal of Geriatric Psychiatry and Neurology 2020;10:891988720957108. CENTRAL
Lee EJ, Oh MS, Kim JS, Chang DI, Park JH, Cha JK, EMOTION investigators. Serotonin transporter gene polymorphisms may be associated with poststroke neurological recovery after escitalopram use. Journal of Neurology, Neurosurgery and Psychiatry 2018;89(3):271-6. CENTRAL
NCT01278498. The preventative effect of escitalopram on depression and related emotional disorders in acute stroke patients, 2011. clinicaltrials.gov/ct2/show/NCT01278498 (first received 19 January 2011). CENTRAL [NCT01278498]

Kong 2007 {published data only}

Kong Y. Fluoxetine for poststroke depression: a randomized placebo controlled clinical trial. Neural Regeneration Research 2007;2(3):162-5. CENTRAL

Lai 2006 {published data only}

Lai J. The effect of using paroxetine to treat post stroke depression. Journal of Guangdong Medical College 2006;24(6):585-6. CENTRAL

Li 2004a {published data only}

Li J, He Q-Y, Han M-F. Recent effect of fluoxetine in improving neurologic impairment and preventing post-stroke depression in the early stage. Chinese Journal of Clinical Rehabilitation 2004;8(7):1208-9. CENTRAL

Li 2004b {published data only}

Li C-M, Jiang X-D, Liao G, Lei J-M, Lan S, Ni F-W. Effect of antidepressant drugs in early period on the recovery of post-stroke depression. Chinese Journal of Clinical Rehabilitation 2004;8(19):3713-5. CENTRAL

Li 2005 {published data only}

Li Y, Wang X, Qian FS. Related factors of post-stroke depression and effect of paroxetine. Shandong Archives of Psychiatry 2005;18(4):209-10. CENTRAL

Li 2006 {published data only}

Li W-Q, Li D-X. The efficacy of citalopram for post-stroke depression and its effects on stroke rehabilitation. International Journal of Cerebrovascular Diseases 2006;14(4):275-8. CENTRAL

Li 2007 {published data only}

Li Z. Clinical efficacy of paroxetine in the treatment of post-stroke depression. Modern Journal of Integrated Traditional Chinese and Western Medicine 2007;16(34):5103-4. CENTRAL

Li 2008 {published data only}

Li L-T, Wang S-H, Ge H-Y, Chen J, Yue S-W, Yu M. The beneficial effects of the herbal medicine free and easy wanderer plus (FEWP) and fluoxetine on post-stroke depression. Journal of Alternative and Complementary Medicine 2008;14(7):841-6. CENTRAL

Li 2017 {published data only}

Li J, Wang J, Li Y, Zhang W, Zhao J. Effects of escitalopram oxalate on post-stroke depression, cognition and neurological function. Journal of Hebei Medical University 2017;38(5):589-92. CENTRAL

Liu 2006 {published data only}

Liu Y, Xu R. Effect of citalopram treatment on post-stroke depression and neurological functional rehabilitation. Chinese Journal of Rehabilitation 2006;21(3):174-5. CENTRAL

Marquez Romero 2013 {published data only}

Marquez-Romero JM, Arauz A, Ruiz-Sandoval JL, Cruz-Estrada Ede L, Huerta-Franco MR, Aguayo-Leytte G, et al. Fluoxetine for motor recovery after acute intracerebral hemorrhage (FMRICH): study protocol for a randomized, double-blind, placebo-controlled, multicenter trial. Trials 2013;14:77. CENTRAL
Marquez-Romero JM, Reyes-Martínez M, Huerta-Franco MR, Ruiz-Franco A, Silos H, Arauz A. Fluoxetine for motor recovery after acute intracerebral hemorrhage, the FMRICH trial. Clinical Neurology and Neurosurgery 2020;190:105656. CENTRAL
Marquez-Romero JM, Reyes-Martínez M, Huerta-Franco MR, Ruiz-Franco A, Silos H, Arauz A. Fluoxetine for motor recovery after acute intracerebral hemorrhage, the FMRICH Trial. Correspondence from Marquez-Romero2018. CENTRAL
Marquez-Romero JM. Fluoxetine for motor recovery after acute intracerebral hemorrhage (FMRICH). www.clinicaltrials.gov/ct2/show/NCT01737541 (first received 29 November 2012). CENTRAL

Meara 1998 {published data only}

Meara RJ, Thalanany M, Balonwu V, Hobson P. The treatment of depression after stroke with the selective serotonin reuptake inhibitor sertraline. Cerebrovascular Diseases 1998;8 Suppl 4:90. CENTRAL

Miao 2004 {published data only}

Miao S-Y, Shi Y-J. Related factors of post-stroke depression and therapeutical effect of citalopram. Chinese Journal of Clinical Rehabilitation 2004;8(19):3718-9. CENTRAL

Murray 2005 {published data only}

Murray V, Von Arbin M, Bartfai A, Berggren A-L, Landtblom A-M, Lumdmark J, et al. Double-blind comparison of sertraline and placebo in stroke patients with minor depression and less severe major depression. Journal of Clinical Psychiatry 2005;66(6):708-16. CENTRAL

NCT00177424 {published data only}

NCT00177424. Setraline for prevention post stroke depression and improving rehabilitation outcomes. clinicaltrials.gov/ct2/show/NCT00177424 (first received 15 September 2005). CENTRAL

NCT01674868 {published data only}

NCT01674868. Fluoxetine for motor, aphasia, and neglect recovery after ischemic stroke (FLAN). www.clinicaltrials.gov/ct2/show/NCT01674868 (first received 29 August 2012). CENTRAL

NCT02737930 {published data only}

NCT02737930. Fluoxetine for visual recovery after ischaemic stroke (FLUORESCE). www.clinicaltrials.gov/ct2/show/NCT02737930 (first received 14 April 2016). CENTRAL

Pan 2018 {published data only}

Pan X-L, Chen H-F, Cheng X, Hu C-C, Wang J-W, Fu Y-M, et al. Effects of paroxetine on motor and cognitive function recovery in patients with non-depressed ischemic stroke: an open randomized controlled study. Brain Impairment;doi: 10.1017/BrImp.2018.6. CENTRAL

Pariente 2001 {published data only}

Guiraud-Chaumeil B, Pariente J, Albucher J-F, Loubinoux I, Chollet F. Rehabilitation after stroke [Recuperation neurologique post-ischemique]. Bulletin de l'Académie Nationale de Médecine 2002;6:1015-24. CENTRAL
Pariente J, Loubinoux I, Carel C, Albucher JF, Leger A, Manelfe C, et al. Fluoxetine modulates motor performance and cerebral activation of patients recovering from stroke. Annals of Neurology 2001;50(6):718-29. CENTRAL

Rasmussen 2003 {published data only}

Rasmussen A, Lunde M, Poulsen D, Sørensen K, Qvitzau S, Bech P. A double-blind placebo controlled study of sertraline in the prevention of depression in stroke patients. European Neuropyschopharmacology 2002;12:231. CENTRAL
Rasmussen A, Lunde M, Poulsen DL, Sørensen K, Qvitzau S, Bech P. A double-blind, placebo-controlled study of sertraline in the prevention of depression in stroke patients. Psychosomatics 2003;44(3):216-22. CENTRAL
Rasmussen A. Depression and stroke. Nordic Journal of Psychiatry 2001;55(4):288. CENTRAL
Rasmussen A. Prophylactic treatment for post-stroke depression and comorbidity. Journal of Psychosomatic Research 2000;48(3):66. CENTRAL

Razazian 2014 {published data only}

Razazian N, Esmaeili O, Almasi A. Effect of fluoxetine on motor improvement in ischemic stroke patients: a double blind clinical trial study. Zahedan Journal of Research in Medical Sciences 2016;18(7):e75492016. CENTRAL
Razazian N. A survey for assessment of effectiveness of fluoxetine on motor improvement in ischemic stroke patients. www.en.irct.ir/trial/8797 (first received 9 January 2014). CENTRAL

Restifo 2001 {published data only}

Restifo DA, Lo Prest R, Lanza S, Giuffrida S, D'Aleo G, Rifici Di Bella C, et al. Motor cortex reorganization induced by fluoxetine in poststroke hemiplegic patients undergoing rehabilitation therapy: a study with transcranial magnetic stimulation. Neurorehabilitation and Neural Repair 2001;15(4):284. CENTRAL

Robinson 2000a {published data only}

Jorge RE, Robinson RG, Arndt S, Starkstein S. Mortality and post-stroke depression: a placebo controlled trial of antidepressants. American Journal of Psychiatry 2003;160:1823-9. CENTRAL
Narushima K, Kosier JT, Robinson RG. Preventing post-stroke depression: a 12 week double blind randomised treatment trial and 21 month follow-up. Journal of Nervous and Mental Diseases 2002;190:296-303. CENTRAL
Robinson RG, Schultz SK, Castillo C, Kopel T, Kosier JT, Newman RM, et al. Nortriptyline versus fluoxetine in the treatment of depression and in short-term recovery after stroke: a placebo-controlled, double-blind study. American Journal of Psychiatry 2000;157(3):351-9. CENTRAL

Robinson 2000b {published data only}

Jorge RE, Robinson RG, Arndt S, Starkstein S. Mortality and post-stroke depression: a placebo controlled trial of antidepressants. American Journal of Psychiatry 2003;160:1823-9. CENTRAL
Narushima K, Robinson RG. Preventing post-stroke depression: a 12 week double blind randomised treatment trial and 21 month follow-up. Journal of Nervous and Mental Diseases 2002;190:296-303. CENTRAL
Robinson RG, Schultz SK, Castillo C, Kopel T, Kosier JT, Newman RM, et al. Nortriptyline versus fluoxetine in the treatment of depression and in short-term recovery after stroke: a placebo-controlled, double-blind study. American Journal of Psychiatry 2000;157(3):351-9. CENTRAL

Robinson 2008 {published data only}

Jorge RE, Acion L, Moser D, Adams HP, Robinson RG. Escitalopram and enhancement of cognitive recovery following stroke. Archives of General Psychiatry 2010;67(2):187-96. CENTRAL
Robinson RG, Arndt S. Incomplete financial disclosure in a study of escitalopram and problem solving therapy for prevention of post-stroke depression. JAMA 2009;301:1023-4. CENTRAL
Robinson RG, Jorge RE, Moser DJ, Acion L, Solodkin A, Small SL, et al. Escitalopram and problem-solving therapy for prevention of poststroke depression. JAMA 2008;299(20):2391-400. CENTRAL

Savadi Oskouie 2012 {published data only}

Savadi Oskouie D, Sharifipour E, Sadeghi Bazargani H, Hashemilar M, Nikanfar M, Ghazanfari Amlashi S, et al. Efficacy of citalopram on acute ischemic stroke outcome: a randomized clinical trial. Neurorehabilitation and Neural Repair 2017;31(7):638-47. CENTRAL
Savadi Oskouie D. Evaluation of the effect of citalopram on three months functional prognosis of acute ischemic stroke patients: a randomized clinical trial. www.en.irct.ir/trial/1766 (first received 22 April 2012). CENTRAL

Shah 2016 {published data only}

Shah IA, Asimi RP, Kawoos Y, Wani MA, Wani MA, Dar MA. Effect of fluoxetine on motor recovery after acute haemorrhagic stroke: a randomized trial. Journal of Neurology and Neurophysiology2016;7(2). CENTRAL [DOI: 10.4172/2155-9562.1000364]

Song 2006 {published data only}

Song J-G. Effects of fluoxetine hydrochloride on depressive symptoms and P300 after cerebral stroke. Chinese Journal of Clinical Rehabilitation 2006;10(14):160-2. CENTRAL

Wang 2003 {published data only}

Wang X, Tan Z, Wu Z, Gao J, Feng M. The effects of anti-depression therapy on post-stroke depression and neurologic rehabilitation in the elderly patients. Chinese Journal of Geriatrics 2003;22(5):270-3. CENTRAL

Wang 2009 {published data only}

Wang X. The efficacy of paroxetine in the treatment of post-stroke depression in 55 cases. Chinese Community Physician (Medical Professional Half-monthly) 2009;11(7):11. CENTRAL

Wen 2006 {published data only}

Wen Z-X. The influence of post-stroke prophylactic anti-depression treatment on nerve functional rehabilitation. Acta Academiae Medicinae Qingdao Universitatis 2006;42(3):253-4. CENTRAL

Wiart 2000 {published data only}

Wiart L, Petit H, Joseph PA, Mazaux JM, Barat M. Fluoxetine in early poststroke depression: a double-blind placebo-controlled study. Stroke 2000;31(8):1829-32. CENTRAL

Xie 2005 {published data only}

Xie R, Liu J, Quan H. A prospective random clinical contrast study of treatment with sertraline in elderly patients with post-stroke depression. Chinese Journal of Clinical Neuroscience 2005;13(3):294-7. CENTRAL

Xu 2001 {published data only}

Xu J, Tan J, Ou L. A study on treatment of fluoxetine to depression in early recovery stage of cerebral infarction. Chinese Journal of Rehabilitation Medicine 2001;16(5):281-3. CENTRAL

Xu 2006 {published data only}

Xu J, Wang J, Liu J. Preventive effects of antidepressants on post-stroke depression. Chinese Mental Health Journal 2006;20(3):186-8. CENTRAL

Yang 2002 {published data only}

Yang J, Zhao Y, Bai S. Controlled study on antidepressant treatment of patients with post-stroke depression. Chinese Mental Health Journal 2002;16(12):871-2. CENTRAL

Yang 2011 {published data only}

Yang J. Therapeutic effect of paroxetine on patients with early poststroke depression and the serum interleukins. Chinese Journal of Cerebrovascular Diseases 2011;8(5):235-8. CENTRAL

Ye 2004 {published data only}

Ye L-X, Wang H, Wang Y-D, Zhong L, Liang D-S, Guo Y. Effect of anti-depressive therapy on the rehabilitation of psychological and neurological function after stroke. Chinese Journal of Clinical Rehabilitation 2004;8(31):6826-8. CENTRAL
Ye LX. Effect of paxil and berhomine on post-stroke anxiety-depression and neurological recovery. Chinese Journal of Clinical Rehabilitation 2006;10(6):153-5. CENTRAL

Zhao 2011 {published data only}

Zhao P, Wang J-P. Effects of antidepressants on neurofunctional recovery of post-stroke patients with aphasia. Journal of Dalian Medical University 2011;33(1):55-7. CENTRAL

Zhou 2008 {published data only}

Zhou Z-l, Liang L-Z, Yan Y-X. Preventive effects of fluoxetine on post-stroke depression. Chinese Journal of Modern Applied Pharmacy 2008;25(3):263-4. CENTRAL

ACTRN12619000573156 {published data only}

ACTRN12619000573156. FRAMBOISE: Fluoxetine, Recovery And Motor BiOmarkers In StrokE. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=377306 (first received 11 April 2019). CENTRAL

Andersen 1993 {published data only}

Andersen G, Vestergaard K, Riis JO. Citalopram for post-stroke pathological crying. Lancet 1993;342(8875):837-9. CENTRAL

Andersen 2012 {published data only}

Andersen G. Efficacy of escitalopram treatment in acute stroke and the role of specific genotypes in stroke. www.clinicaltrialsregister.eu/ctr-search/search?query=2011-005541-12 (first received 27 February 2012). CENTRAL
Andersen G. Escitalopram Treatment In Acute Stroke (ESTIAS). www.clinicaltrials.gov/ct2/show/NCT01561092 (first received 22 March 2012). CENTRAL

Anderson 2002 {published data only}

Anderson C, Hackett M, Carter K, Ni MC. Maximising outcome by overcoming depression in stroke (MOODS) trial. Cerebrovascular Diseases 2002 2002;13 Suppl 3:79. CENTRAL

Anonymous 2012a {published data only}

Anonymous 2012. Stroke and depression. Johns Hopkins Medical Letter: Health After 50 2012;23(11):3. CENTRAL

Anonymous 2012b {published data only}

Anonymous. Drug & Device News. Medical Malpractice Law & Strategy, New York Law Journal Newsletters 2012;30(2):5. CENTRAL

Berends 2009 {published data only}

Berends HI, Nijlant JM, Putten MJ, Movig KL, IJzerman MJ. Single dose of fluoxetine increases muscle activation in chronic stroke patients. Clinical Neuropharmacology 2009;32(1):1-5. CENTRAL [also known as Flu 2006]

Bonin Pinto 2019 {published data only}

Bonin Pinto C, Morales-Quezada L, de Toledo Piza PV, Zeng D, Saleh Vélez FG, Ferreira IS, et al. Combining fluoxetine and rTMS in poststroke motor recovery: a placebo-controlled double-blind randomized phase 2 clinical trial. Neurorehabilitation and Neural Repair 2019;33:643-55. CENTRAL [DOI: 10.1177/1545968319860483.]
NCT02208466. Effects rTMS combined with fluoxetine on motor recovery in stroke patients. www.clinicaltrials.gov/ct2/show/NCT02208466 (first received 5 August 2014). CENTRAL

Chen 2019 {published data only}

Chen Ma, Ping L. Effectiveness of paroxetine in the treatment of poststroke depression. Medicine 2018;97(29):1-4. CENTRAL

Choi Kwon 2008 {published data only}

Choi-Kwon S, Choi J, Kwon SU, Kang DW, Kim JS. Fluoxetine improves the quality of life in patients with poststroke emotional disturbances. Cerebrovascular Diseases 2008;26(3):266-71. CENTRAL

Finkenzeller 2009 {published data only}

Finkenzeller W, Zobel I, Rietz S, Schramm E, Berger M. Interpersonal psychotherapy and pharmacotherapy for post-stroke depression. Feasibility and effectiveness. Der Nervenarzt 2009;80(7):805-12. CENTRAL

Foster 2019 {published data only}

Foster E, Yaroslavtseva O, Chundamala J, Leibovitch F, Eng JJ, Ali F, et al. FLOW Trial: fluoxetine to open the critical period time window to improve motor recovery after stroke. International Journal of Stroke 2019;14 Suppl 3:36. CENTRAL

Gourab 2015 {published data only}

Gourab K, Schmit BD, Hornby GT. Increased lower limb spasticity but not strength or function following a single-dose serotonin reuptake inhibitor in chronic stroke. Archives of Physical Medicine and Rehabilitation 2015;96(12):2112-9. CENTRAL

Graffagnino 2002 {published data only}

Graffagnino C. Poststroke depression and functional recovery (SADBRAIN). Duke University Medical Centre2002. CENTRAL

Ji 2000 {published data only}

Ji QM, Xie LP. Efficacy of fluoxetine in the treatment of 20 patients with depression after stroke. Herald of Medicine 2000;19(4):329. CENTRAL

Kitago 2020 {published data only}

Dose-finding study of selective serotonin reuptake inhibitors to enhance neuroplasticity [Dose finding study but the intervention is a combination of an SSRI and paired associative stimulation, so we can exclude]. Source not provided from searches2020. CENTRAL

Li 2002 {published data only}

Li F, Gu DX, Ceng SH, Xu JW. Effect of paroxetine on prognosis of patients with post cerebral infarction depression. Chinese Journal of New Drugs and Clinical Remedies. 2002;21(1):11-3. CENTRAL

Liang 2003 {published data only}

Liang Z, Shuliang T. Clinical efficacy of fluoxetine in treatment of patients with depression after acute stroke. Chinese Journal of Clinical Rehabilitation 2003;7(13):1924-5. CENTRAL

Liu 2004 {published data only}

Liu L-X. Recent effect of drug intervention on post-stroke anxiety. Chinese Journal of Clinical Rehabilitation 2004;8(30):6600-1. CENTRAL

Liu 2020 {published data only}

Liu W, Ding W. Study on the efficacy and mechanism of paroxetine hydrochloride combined with repetitive transcranial magnetic stimulation in the treatment of post-stroke depression. International Journal of Clinical and Experimental Medicine 2020;13(10):7881-8. CENTRAL

Mosarrezaii 2018 {published data only}

Mosarrezaii A, Salmasi BA, Taghavi SA. Studying effect of fluoxetine on improvement of motor performance in patients with ischemic stroke. Journal of Research in Medical and Dental Sciences 2018;6(3):118-22. CENTRAL

NCT01963832 {published data only}

NCT01963832. RCT of a neuroplasticity agent and CI therapy for severe arm paresis after stroke. clinicaltrials.gov/ct2/show/NCT01963832 (first received 16 October 2013). CENTRAL

Robinson 2011 {published data only}

Robinson RG, Mikami K, Jang M, Jorge RE. Prevention of anxiety disorder after stroke. Journal of Neuropsychiatry and Clinical Neurosciences 2011;23(2):18. CENTRAL

Sitzer 2002 {published data only}

Sitzer M, Huff W, Steckel R. Prevention of poststroke depression after acute ischemic stroke using the selective serotonin reuptake inhibitor sertraline (PreDis-study). Stroke 2002;33(2):651-2. CENTRAL

Sun 2015 {published data only}

Sun YT, Bao YH, Wang SL, Chu JM, Li LP. Efficacy observation on the treatment of post-stroke depression by acupuncture at the acupoints based on ziwuliuzhu and Prozac. Chinese Acupuncture and Moxibustion 2015;355(2):119-22. CENTRAL

Vogel 2020 {published data only}

Vogel AC, Okeng'o K, Chiwanga F, Ismail SS, Buma D, Pothier L. MAMBO: Measuring Ambulation, Motor, and Behavioral Outcomes with post-stroke fluoxetine in Tanzania: protocol of a phase II clinical trial. Journal of the Neurological Sciences 2020;408:116563. CENTRAL [PMID: DOI:10.1016/j.jns.2019.116563]

Xu 2007 {published data only}

Xu B, Zhou WY, Zhang SJ. Observation of effect of Wulung capsule in treating post-stroke depression. Chinese Journal of Integrated Traditional and Western Medicine 2007;27(7):640-2. CENTRAL

Zhou 2003 {published data only}

Zhou B. Effects of fluoxetine on neurofunctional recovery of non depressed patients after stroke. Chinese Journal of Clinical Rehabilitation 2003;7(3):374-5. CENTRAL

Referencias de los estudios en espera de evaluación

Guo 2016 {published data only}

Guo Y, He Y, Tang B, Ma K, Cai Z, Zeng S, et al. Effect of using fluoxetine at different time windows on neurological functional prognosis after ischemic stroke. Restorative Neurology and Neuroscience 2016;34(2):177–87. CENTRAL
Guo Y. Effect of using fluoxetine at different time windows after ischemic stroke on neurological functional prognosis: a randomized controlled trial. chictr.org.cn/showprojen.aspx?proj=12925 (first received 27 December 2015). CENTRAL

He 2018 {published data only}

ChiCTR-TRC-12002078. Multi-center randomized clinical study of antidepressant treatment (fluoxetine) on secondary prevention of ischemic stroke. www.chictr.org.cn/showprojen.aspx?proj=7471 (first received 3 April 2012). CENTRAL
He Y, Cai Z, Zeng S, Chen S, Tang B, Liang Y, et al. Effect of fluoxetine on three-year recurrence in acute ischemic stroke: a randomized controlled clinical study. Clinical Neurology and Neurosurgery 2018;168:1-6. CENTRAL

Jurcau 2016 {published data only}

Jurcau A, Simion A. Improved post-ischaemic stroke recovery over 1 year with escitalopram for 3 months. European Journal of Neurology 2016;23:614. CENTRAL

NCT00967408 {published data only}

NCT00967408. Effects of clinical and functional outcome of escitalopram in adult stroke patients. clinicaltrials.gov/ct2/show/NCT00967408 (first received 27 August 2009). CENTRAL [NCT00967408]

ChiCTR1800019467 {published data only}

ChiCTR1800019467. The effect and mechanism of fluoxetine on the automatic regulation of cerebral blood flow for ischemic stroke. https://www.chictr.org.cn/showprojen.aspx?proj=32801. CENTRAL

CTRI/2018/12/016568 {published data only}

CTRI/2018/12/016568. An interventional study to look at efficacy of fluoxetine in patients with post-stroke anxiety. http://ctri.nic.in CTRI/2018/12/016568 (first received 10 December 2018). CENTRAL

EudraCT 2005‐005266‐37 {published data only}2005‐005266‐37

EudraCT 2005-005266-37. Influence of escitalopram on the incidence of depression and dementia following acute middle cerebral artery territory infarction. A randomised placebo-controlled double blind study. www.clinicaltrialsregister.eu/ctr-search/search?query=2005-005266-37 (first received 7 April 2006). CENTRAL

IRCT201112228490N1 {published data only}

IRCT201112228490N1. Effect of fluoxetine on functional recovery of patients with cerebrovascular accident following middle cerebral artery trunk obstruction: a randomized clinical trial. www.en.irct.ir/trial/8954 (first received 25 January 2012). CENTRAL

IRCT2012101011062N1 {published data only}

IRCT2012101011062N1. A study of sertraline effect on quality of life in stroke inpatients. www.en.irct.ir/trial/11413 (first received 28 November 2012). CENTRAL

IRCT2017041720258N37 {published data only}

IRCT2017041720258N37. Evaluation of fluoxetine and standard treatment efficacy on change to side effect of stroke of ischemic strokes in both hemispheres in anterior circulation. www.irct.ir/trial/17976 (first received 11 October 2017). CENTRAL

IRCT20210307050617N1 {published data only}

IRCT20210307050617N1. The efficacy comparison of fluoxetine and citalopram on motor recovery after ischemic stroke: single-blind placebo-controlled randomized clinical trial. https://en.irct.ir/trial/54896 (first received 27 March 2021). CENTRAL

NCT02386475 {published data only}

NCT02386475. Effect of serotonin and levodopa in ischemic stroke. www.clinicaltrials.gov/ct2/show/NCT02386475 (first received 12 March 2015). CENTRAL

NCT02767999 {published data only}

NCT02767999. Serotonin Selective Reuptake Inhibitor (SSRI) effects on cerebral connectivity in acute ischemic stroke (RECONISE). www.clinicaltrials.gov/ct2/show/NCT02767999 (first received 11 May 2016). CENTRAL

NCT02865642 {published data only}

NCT02865642. Cortical ischemic stroke and serotonin (CISS). www.clinicaltrials.gov/ct2/show/NCT02865642 (first received 12 August 2016). CENTRAL

NCT03448159 {published data only}

NCT03448159. Fluoxetine Opens Window to improve motor recovery after stroke (FLOW). www.clinicaltrials.gov/ct2/show/NCT03448159 (first received 27 February 2018). CENTRAL

NCT03826875 {published data only}

NCT03826875. Depression in hemorrhagic stroke. www.clinicaltrials.gov/ct2/show/NCT03826875 (first received 1 February 2019). CENTRAL

TCTR20181216001 {published data only}

TCTR20181216001. Randomized controlled trial of fluoxetine or placebo on quality of life after acute ischemic stroke. www.thaiclinicaltrials.org/show/TCTR20181216001 (first received 16 December 2018). CENTRAL

Allida 2019

Allida S, Patel K, House A, Hackett ML. Pharmaceutical interventions for emotionalism after stroke. Cochrane Database of Systematic Reviews 2019, Issue 3. Art. No: CD003690. [DOI: 10.1002/14651858.CD003690.pub4]

Deeks 2021

Deeks JJ, Higgins JP, Altman DG, editors. Chapter 10 : Analysing data and performing metaanalyses. In: Higgins JP , Thomas J, Chandler J, Cumpston M, Li T, Page MJ, et al( editor s) . ) , Cochrane Handbook for Systematic Reviews of Interventions version 6.2 (updated February 2021) Cochrane 2021. Available at www.training.cochrane.org/handbook.

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997;315(7109):629-34.

GBD 2015

GBD 2015. Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic analysis for the Global Burden of Disease Study. Lancet 2016;388(10053):1545–602.

GRADE 2013

Schünemann H, Brożek J, Guyatt G, Oxman A, editor(s). Handbook for grading the quality of evidence and the strength of recommendations using the GRADE approach (updated October 2013). GRADE Working Group, 2013. Available from gdt.guidelinedevelopment.org/app/handbook/handbook.html.

GRADEproGDT 2015 [Computer program]

McMaster University (developed by Evidence Prime, Inc.)GRADEpro Guideline Development Tool [Software]. McMaster University (developed by Evidence Prime, Inc.), 2015.

Hatano 1976

Hatano S. Experience from a multicentre stroke register: a preliminary report. Bulletin of the World Health Organization 1976;54(5):541-53.

Higgins 2017

Higgins JP, Altman DG, Sterne JA, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Churchill R, Chandler J, Cumpston MS, editor(s), Cochrane Handbook for Systematic Reviews of Interventions version 5.2.0 (updated June 2017), The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Johnson 2016

Johnson W, Onuma O, Owolabi M, Sachdev S. Stroke: a global response is needed. Bulletin of the World Health Organization 2016;94(9):634-634A.

Lang 2004

Lang UE, Jockers-Scherübl MC, Hellweg R. State of the art of the neurotrophin hypothesis in psychiatric disorders: implications and limitations. Journal of Neural Transmission 2004;111(3):387-411.

Legg 2019

Legg LA, Tilney R, Hsieh C, Wu S, Lundström E, Rudberg A, et al. Selective serotonin reuptake inhibitors for stroke recovery. Cochrane Database of Systematic Reviews 2019, Issue 11. Art. No: CD009286. [DOI: 10.1002/14651858.CD009286.pub3]

Li 2020

Li, X, Zhang C. Comparative efficacy of nine antidepressants in treating Chinese patients with post-stroke depression: a network meta-analysis. Journal of Affective Disorders 2020;266:540-8.

Lim 2009

Lim CM, Kim SW, Park JY, Kim C, Yoon SH, Lee JK. Fluoxetine affords robust neuroprotection in the postischemic brain via its anti-inflammatory effect. Journal of Neuroscience Research 2009;87(4):1037-45.

Loubinoux 1999

Loubinoux I, Boulanouar K, Ranjeva JP, Carel C, Berry I, Rascol O, et al. Cerebral functional magnetic resonance imaging activation modulated by a single dose of the monoamine neurotransmission enhancers fluoxetine and fenozolone during hand sensorimotor tasks. Journal of Cerebral Blood Flow and Metabolism 1999;19(12):1365-75.

Mantel 1959

Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. Journal of the National Cancer Institute 1959;22(4):719-48.

Mead 2020

Mead GE, Graham C, Billot L, Näsman P, Lundström E, Lewis S, for FOCUS, AFFINITY and EFFECTS trialists. Update to the FOCUS, AFFINITY and EFFECTS trials studying the effect(s) of fluoxetine in patients with a recent stroke: statistical analysis plan for the trials and for the individual patient data meta-analysis. Trials 2020;21(1):971. [DOI: https://doi.org/10.1186/s13063-020-04875-1]

Ming 2005

Ming GL, Song H. Adult neurogenesis in the mammalian central nervous system. Annual Review of Neuroscience 2005;28:223-50.

Molnar 2008

Molnar FJ, Hutton B, Fergusson D. Does analysis using “last observation carried forward” introduce bias in dementia research? CMAJ 2008;179(8):751–3.

NHS 2021

NHS. Side effects-selective serotonin reuptake inhibitors (SSRI). www.nhs.uk (accessed 10 August 2021).

Pälvimäki 1994

Pälvimäki E-P, Laakso A, Kuoppamäki M, Syvälahti E, Hietala J. Up-regulation of beta l-adrenergic receptors in rat brain after chronic citalopram and fluoxetine treatments. Psychopharmacology 1994;115(4):543-6.

Pinto 2017

Pinto CB, Saleh Velez FG, Lopes F, de Toledo Piza PV, Dipietro L, Wang QM, et al. SSRI and motor recovery in stroke: reestablishment of inhibitory neural network tonus. Frontiers in Neuroscience 2017;11:637. [DOI: 10.3389/fnins.2017.00637]

Review Manager 2014 [Computer program]

The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.4. The Cochrane Collaboration, 2020.

Santarelli 2003

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, et al. Requirement of hippocampal neurogenesis antidepressant treatments and animal models of depressive-like behaviour. Behavioural Pharmacology 2003;301:805-9.

Schmidt 2007

Schmidt HD, Duman RS. The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behaviour. Behavioural Pharmacology 2007;18(5-6):391-418.

Schünemann 2017

Schünemann HJ, Oxman AD, Higgins JP, Vist GE, Glasziou P, Akl E, Guyatt GH on behalf of the Cochrane GRADEing Methods Group and the Cochrane Statistical Methods Group. Chapter 11: Completing ‘Summary of findings’ tables and grading the confidence in or quality of the evidence. In: Higgins JP, Churchill R, Chandler J, Cumpston MS, editor(s), Cochrane Handbook for Systematic Reviews of Interventions version 5.2.0 (updated June 2017). The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Shin 2009

Shin TK, Kang MS, Lee HY, Seo MS, Kim SG, Kim CD, et al. Fluoxetine and sertraline attenuate postischemic brain injury in mice. Korean Journal of Physiology and Pharmacology 2009;13(3):257-63.

Sterne 2017

Sterne JA, Egger M, Moher D, Boutron I, editor(s). Chapter 10: Addressing reporting biases. In: Higgins JP, Churchill R, Chandler J, Cumpston MS, editor(s), Cochrane Handbook for Systematic Reviews of Interventions version 5.2.0 (updated June 2017). The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Taupin 2006

Taupin P. Adult neurogenesis and neuroplasticity. Restorative Neurology and Neuroscience 2006;24(1):9-15.

Wan 2014

Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Medical Research Methodology 2014;14:135.

Wiltrout 2007

Wiltrout C, Lang B, Yan Y, Dempsey RJ, Vemuganti R. Repairing brain after stroke: a review on post-ischaemic neurogenesis. Neurochemistry International 2007;50(7-8):1028-41.

Xie 2019

Xie Y, Liu PP, Lian YJ, Liu H-B, Kang J-S. The effect of selective serotonin reuptake inhibitors on cognitive function in patients with Alzheimer’s disease and vascular dementia: focusing on fluoxetine with long follow-up periods. Signal Transduction and Targeted Therapy 2019;4:30. [DOI: 10.1038/s41392-019-0064-7]

Yi 2010

Yi ZM, Liu F, Zhai SD. Fluoxetine for the prophylaxis of post-stroke depression in patients with stroke: a meta-analysis. International Journal of Clinical Practice 2010;64(9):1310-7.

Zittel 2008

Zittel S, Weiller C, Liepert J. Citalopram improves dexterity in chronic stroke patients. Neurorehabilitation and Neural Repair 2008;22(3):311-4. [DOI: 10.1177/1545968307312173]

Referencias de otras versiones publicadas de esta revisión

Legg 22019

Legg LA, Tilney R, Hsieh CF, Wu S, Lundström E, Rudberg AS, et al. Selective serotonin reuptake inhibitors (SSRIs) for stroke recovery. Cochrane Database of Systematic Reviews 2019, Issue 11. Art. No: CD009286. [DOI: 10.1002/14651858.CD009286.pub3]

Mead 2011

Mead GE, Hankey GJ, Kutlubaev MA, Lee R, Bailey M, Hackett ML. Selective serotonin reuptake inhibitors (SSRIs) for stroke. Cochrane Database of Systematic Reviews 2011, Issue 10. Art. No: CD009286. [DOI: 10.1002/14651858.CD009286]

Mead 2012

Mead GE, Hsieh CF, Lee R, Kutlubaev MA, Claxton A, Hankey GJ, et al. Selective serotonin reuptake inhibitors (SSRIs) for stroke recovery. Cochrane Database of Systematic Reviews 2012, Issue 11. Art. No: CD009286. [DOI: 10.1002/14651858.CD009286.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Acler 2009

Study characteristics

Methods

Study type: interventional (clinical trial)

Intervention model: parallel assignment

Primary purpose: treatment

Participants

20 participants

Location: Italy

Setting: inpatient

Inclusion criteria: first‐ever ischaemic stroke, CT or MRI documenting a single monohemispheric lesion, age below 80 years, within 3 months of onset

Exclusion criteria: major affective disorders, alcohol abuse and dementia leading to un‐cooperative behaviour, pacemakers, metal in the head, concomitant neuropathies, systemic vasculopathies, major affective disorders

Treatment: 10 people, mean age 68 ± 7 years, 6 men

Control: 10 people, mean age 65 ± 7 years, 6 men

Interventions

Citalopram 10 mg daily

Placebo: identical pill daily

Duration of treatment: at least 4 months

Duration of follow‐up: not stated

Outcomes

Motor cortex excitability

NIHSS

Lindmark Scale

BI

HDRS

BDI

No data on death, GI upset, bleeds or seizures

Funding source

Source of funding not stated; unclear whether or not a drug company was involved in the study

Notes

Dates of study not stated. Any conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Stated blinded, placebo was 'an identical pill'

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Stated blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is not stated whether data from all recruited participants are reported

Selective reporting (reporting bias)

High risk

Side effects were not reported although they were assessed

Other bias

Unclear risk

AFFINITY 2020

Study characteristics

Methods

Multicentre

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: quadruple (participant, care provider, investigator, outcomes assessor)

Primary purpose: treatment

Participants

1280 participants

Country: Australia (n = 532), New Zealand (n = 42), and Vietnam (n = 706)

Setting: inpatient

At randomisation number allocated: N = 1280: fluoxetine (n = 642); placebo (n = 638)

% male: fluoxetine (64%); placebo (62%)

Age: mean age: fluoxetine = 63.5± 12.5; placebo = 64.6 ± 12.2

Subtype of stroke

  • Total anterior circulation infarct: fluoxetine (9%); placebo (9%)

  • Partial anterior circulation infarct: fluoxetine (49%); placebo (52%)

  • Lacunar infarct: fluoxetine (21%); placebo (105%)

  • Posterior circulation infarct: fluoxetine (114%); placebo (103%)

  • Uncertain: fluoxetine (2%); placebo (1%)

Inclusion criteria

  • Age > 18 years

  • Clinical diagnosis of stroke 2 to 15 days previously

  • Brain imaging consistent with ischaemic or haemorrhagic stroke (including normal CT brain scan)

  • Persisting measurable focal neurological deficits causing a functional deficit at the time of randomisation

Exclusion criteria:

  • History of epileptic seizures

  • History of bipolar disorder

  • History of drug overdose or attempted suicide

  • Ongoing treatment with any selective serotonin reuptake inhibitor

  • Allergy or contra‐indication to fluoxetine

  • Use of medications that may interact seriously with fluoxetine

  • Not available for follow‐up over the next 365 days e.g. no fixed home address

  • Life‐threatening illness (e.g. advanced cancer) that is likely to reduce 365‐day survival

  • Pregnant, breast‐feeding or of child‐bearing potential and not using contraception

  • Enrolled in another interventional clinical research trial involving an investigational product (medicine) or device

Interventions

Fluoxetine 20 mg once daily or matching placebo capsules for 6 months

Outcomes

Primary outcome

  • Functional outcome as measured by the mRS at 180 days after randomisation

Secondary outcomes at 180 and 365 days after randomisation

  • Survival

  • Mood (PHQ‐9)

  • Cognitive function (TICSm)

  • Communication (SIS)

  • Motor function (SIS)

  • Overall health status (SIS)

  • Health‐Related Quality of Life (HRQoL) (EuroQol)

  • Functional recovery (smRSq) at the 365‐day assessments

  • New diagnosis of depression requiring treatment with antidepressants

  • Fatigue (vitality domain of the SF‐36)

  • Serious adverse events at any time during follow‐up including new stroke, acute coronary syndrome, epileptic seizures, fall, new fractures or death

Funding source

The AFFINITY trial was funded by the Australian NHMRC Project Grant 1059094. The minimisation algorithm was provided by The Stroke Research Group, Division of Clinical Neuroscience, University of Edinburgh, Edinburgh, UK

Notes

ACTRN12611000774921Recruitment January 11, 2013, and June 30, 2019. GJH has received grants from the NHMRC of Australia, Vetenskapsrådet (The Swedish Research Council), and UK National Institute for Health Research Technology, during the conduct of the study; and personal fees from the American Heart Association, outside of the submitted work. MLH, CE‐B, LB, and TL have received grants from the NHMRC of Australia during the conduct of the study. CSA has received grants from the NHMRC of Australia, and grants and personal fees from Takeda, outside of the submitted work. All other members of the writing group declare no competing interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: " The patient’s clinician entered the patient’s baseline data into a secure, password‐protected, centralised, web‐based randomisation system that checked the data for completeness and consistency and generated a unique study identification number and treatment pack number corresponding to fluoxetine or placebo in a 1:1 ratio."

Allocation concealment (selection bias)

Low risk

Quote: " All patients, carers, investigators, and outcome assessors were masked to the allocated treatment by use of placebo capsules that were visually identical to the fluoxetine capsules even when broken open. Success of masking was not assessed."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "All patients, carers, investigators, and outcome assessors were masked to the allocated treatment by use of placebo capsules that were visually identical to the fluoxetine capsules even when broken open. Success of masking was not assessed."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "All patients, carers, investigators, and outcome assessors were masked to the allocated treatment by use of placebo capsules that were visually identical to the fluoxetine capsules even when broken open. Success of masking was not assessed."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Less than 5% dropped out or died and there is no compelling evidence of a difference between the 2 groups

Selective reporting (reporting bias)

Low risk

All outcomes in protocol were reported

Other bias

Low risk

The study appears to be free of other sources of bias

Almeida 2006

Study characteristics

Methods

Parallel design

Analysis: ITT (last observation carried forward), withdrawn owing to becoming depressed, AE, treating practitioner started antidepressant, medical advice, no reason given, not contactable ‐ numbers not included

Participants

Location: Australia

Setting: inpatient

Treatment: 55 people, mean ± SD age 68 ± 13 years, 67% men

Control: 56 people, mean ± SD age 67 ± 13 years, 62% men

Stroke criteria: acute ischaemic or haemorrhagic stroke, diagnosis by clinical signs (ICD‐10) and CT (100% imaged, 10/111 CT scan did not show acute ischaemia); stroke on average < 2 weeks prior to randomisation

Not depressed (HADS‐D had to be over 7)

Other entry criteria: not stated

Comparability of treatment groups: more participants in treatment group with previous heart attack and stroke, also higher levels of hypertension

Exclusion criteria: severe communication difficulties, unstable medical condition, severe cognitive impairment and depression (MMSE < 10), taking antidepressants within 4 weeks of stroke, contraindication to sertraline, previous reaction to sertraline, could not speak English

Interventions

Treatment: sertraline 50 mg daily (night)

Control: matched placebo

Duration: treatment continued for 24 weeks

Duration of follow‐up (post‐treatment to study end): 28 weeks

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS, proportion depressed

Change in MMSE scores

mRS

Death

Leaving the trial early

Check list of possible AEs read out to participant by a research nurse

Funding source

Funded by an unrestricted grant from Rotary Health Research Fund

Notes

Recruitment June 2004 to June 2006

Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Low risk

Centralised

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Stated in paper, matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated in paper

Incomplete outcome data (attrition bias)
All outcomes

High risk

Performed last observation carried forward

Selective reporting (reporting bias)

Low risk

Trial protocol published on www.strokecentre.org/trials . This weblink is no longer available so we have been unable to check whether all the outcomes were reported. We have contacted the author to check this‐who confirm that all endpoints were reported

Other bias

Low risk

No other obvious biases

Andersen 1994

Study characteristics

Methods

Parallel design

Analysis (ITT) last observation carried forward and per protocol: death (1 treatment, 1 control) withdrawn owing to AE (6 treatment, 1 control), all excluded from analysis

Participants

Location: Denmark

Setting: mixed

Treatment: 33 people, mean ± SD age 68 ± 4 years, 36% men

Control: 33 people, mean ± SD age 66 ± 9 years, 66% men

Stroke criteria: ischaemic stroke and PICH; diagnosis via clinical signs and CT (100%); stroke 2 to 52 weeks prior to randomisation (average time 12 weeks)

Depression criteria: HDRS score > 12 (score transformed to appropriate DSM‐III‐R criteria)

Other entry criteria: none stated

Comparability of treatment groups: balanced

Exclusion criteria: depression within last year, receiving current treatment for depression, severe dementia or communication problems, degenerative or expansive neurological disease, decreased consciousness

Interventions

Experimental: citalopram 10 mg in participants > 66 years, 20 mg in participants < 67 years daily; dose doubled if no response to treatment within 3 weeks

Comparator: matched placebo

Duration: treatment continued for 6 weeks

Duration of follow‐up (post‐treatment to study end): 0

Note that although the protocol on www.strokecentre.org/trials states that mood scores were measured up to 1 year post‐stroke, this probably refers to the time since stroke at the time of randomisation

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS

Melancholia scale

Proportion no longer meeting entry criteria (< 13 on HDRS)

50% reduction in HDRS score

Additional: leaving the study early

Death

AEs (unwanted drug effects were registered and evaluated at the same intervals using a side effect scale)

Unable to use: BI, Social Activities Index, MMSE (data not presented)

Funding source

Funded by Lundbeck Foundation, Medical Research Foundation for North Jutland County, The Aalgorg Diocese Research Foundation, Consultant Otorhinolaryngologist Kopp's Foundation and Stine and Martinus Sorensen's Foundation. Lundbeck Pharma A/S provided the citalopram and placebo; thus we have classified this as 'unclear risk'.

Notes

Recruitment 1 February 1991 to 29 February 1992. Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Blocks of 4 used

Allocation concealment (selection bias)

Low risk

Centralised opaque envelopes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Those who were blinded were not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Although there were dropouts, analysis performed both per protocol and using last observation carried forward

Selective reporting (reporting bias)

High risk

Trial published on www.strokecentre.org/trials

The primary outcome was reported. We have been unable to access this record on 28 September 2021, the paper also describes the social activities index in the list of outcomes but this was not reported in the results so we have changed this to high risk of bias for this 2021 update

Other bias

Unclear risk

Andersen 2013

Study characteristics

Methods

Multicentre

Study type: interventional (clinical trial)

Intervention model: parallel assignment

Primary purpose: treatment

Participants

642 participants

Country: Denmark

Setting: inpatient

At randomisation number allocated: citalopram n = 319; placebo n = 323

% male at baseline: citalopram n = 199/319 (62%); placebo n = 222/323 (69%)

Age at baseline: mean age, citalopram 68 ± 13 (n = 319); placebo 68 ± 13 (n = 323)

Subtype of stroke at baseline: not available

Severity of stroke at baseline: NIHSS, citalopram 5.3 ± 5.6; placebo 4.8 ± 4.8

Time since stroke onset: mean time from last known 'well' to first treatment 1.7 days (median 1, IQR 0 to 6)

Inclusion criteria:

  • First ever ischaemic stroke

  • Age > 18 years

Exclusion Criteria

  • Haemorrhagic stroke

  • Dementia or other neurodegenerative disease

  • Antidepressant medical treatment within 6 months of admission

  • Acute need for antidepressant treatment

  • Drug abuse or other conditions that may indicate non‐compliant behaviour

  • Liver failure (increased liver enzyme levels up to or more than 2 times upper limit)

  • Renal failure (eGFR below 30 mL/min per 1.73 m2)

  • Hyponatremia (S‐potassium below 130 mmol/L

  • Actively bleeding ulcer

  • Fatal stroke or other severe co‐morbidity that markedly decreases expected life span

  • Prolonged corrected QT‐interval (QTc above 480 ms)

  • Ongoing treatment with drugs known to prolong the QTc interval

Interventions

Experimental: citalopram 20 mg (10 mg if aged ≥ 65 years or having reduced liver/kidney function) or placebo once daily for 6 months

Comparator: ½ to 2 tablets with no intrinsic drug activity per day for 6 months

Outcomes

Primary outcomes

  • Vascular death, TIA/stroke and myocardial infarction within 6 months

  • Functional status at 6 months (mRS)

Secondary outcomes within or at 6 months

  • Vascular death

  • Death of any cause

  • TIA/stroke

  • Bleeding

  • Myocardial infarction

  • Disability/dependence (mRS and BI)

  • Physical activity (PASE)

  • Cognitive and organic cerebral impairment (MMSE and the Symbol Digit Modalities Test)

  • Fatigue (Multidimensional Fatigue Inventory)

  • Post‐stroke depression (Major Depression Inventory test (MDI), Global depression scale (self and clinician and Hamilton Depression Scale ‐ 6 item (HAM‐D6))

  • Pathological crying (Pathological Crying Scale)

  • Lesion size (FLAIR positive lesion size on MRI 24 hours after treatment with Alteplase)

Funding source

TrygFonden, the Danish Council for Independent Research, the Regional Medicine Fund, and the Aarhus University Research Foundation

Notes

Dates study conducted: September 2013 to December 2016

Declarations of Interest: Dr Kraglund received speaker honoraria from Bristol‐Meyers Squibb and Pfizer. Dr Iversen received speaker honoraria from Boehringer Ingelheim, Bristol‐Myers Squibb, Bayer, AstraZeneca, and Pfizer and has previously participated in advisory board meetings for Boehringer Ingelheim, Bristol‐Myers Squibb, Pfizer, Bayer, AstraZeneca, and Amgen. Dr Grove has received speaker honoraria from AstraZeneca, Baxter, Bayer, Boehringer Ingelheim, Bristol‐Myers Squibb, MSD, and Pfizer and has previously participated in advisory board meetings for AstraZeneca, Bayer, Boehringer Ingelheim, and Bristol‐Myers Squibb. Dr Andersen reports other from MSD, personal fees from AstraZeneca, outside the submitted work

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A computer‐generated randomization code was used to randomize patients in blocks of 10."

Allocation concealment (selection bias)

Low risk

Quote: "Citalopram was commercially available (Sandoz, Denmark) and production of the placebo and randomization was prepared by a pharmacy independently of the investigators (Glostrup Pharmacy, Denmark). The tablets were indistinguishable and were supplied in numbered containers."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participant, care provider, and investigator assured and unlikely that the blinding could have been broken

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessor assured and unlikely that the blinding could have been broken

Incomplete outcome data (attrition bias)
All outcomes

High risk

Attrition/exclusions reported with reasons provided (including did not start on study medication, consent withdrawn, side effects, indication for open label, other reasons (no detail provided)). At < 31 days of study medication, twice as many participants in the citalopram group withdrew consent (n = 29/318 (9%)) compared to the placebo group (n = 14/320 (4%)). However, at < 31 days twice as many participants in the placebo group (n = 12/318(4%)) compared to the citalopram group (n = 6/320 (2%) were switched to open label. Attrition/exclusions: 51/319 (16%) in the citalopram group and 39/319 (11%) in the placebo group.

The investigators use LOCF in their intention‐to‐treat analysis. LOCF assumes that missing values are missing completely at random and ignores improvements or deteriorations in the participants condition since dropout and therefore stops improvements or declines in outcome measures. LOCF introduces risk of false or biased conclusions (Molnar 2008)

Selective reporting (reporting bias)

Low risk

The study protocol is available and all of the study's prespecified (primary and secondary) outcomes that are of interest to the review have been reported in the prespecified way

Other bias

Low risk

The study appears to be free of other sources of bias

Asadollahi 2018

Study characteristics

Methods

RCT

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: double (participant, outcomes assessor)

Primary purpose: treatment

Participants

90 participants

Country: Iran

Setting: inpatient

At randomisation number allocated: N = 90: citalopram (n = 30); fluoxetine (n = 30); placebo (n = 30)

% male: citalopram (60%) fluoxetine (50%); placebo (56.6%)

Age: mean age: citalopram = 61.7 ± 9.6; fluoxetine = 60.2 ± 8.52; placebo = 58.7 ± 8.56

Inclusion criteria

  • > 18 years of age

  • suffering from hemiparesis or hemiplegia as a result of a first‐time acute Ischaemic stroke within the past 24 hours

  • an initial Fugl‐Meyer Motor Scale score of under 55

Exclusion criteria

  • NIHSS score < 5

  • Prior disabilities including aphasia, cognitive disorders and motor disorders due to stroke, or any other neurodegenerative disease

  • Pregnancy or breastfeeding

  • Currently taking antidepressants

  • Contraindications of therapy, including renal insufficiency (glomerular filtration rate < 30mL/min), abnormal liver function tests, hyponatremia, and a long QT interval on an electrocardiogram

  • Any significant adverse effects (agitation, hypertension, or other signs of serotonin syndrome) after initiation of treatment

Interventions

Participants were randomly allocated to 1 of 3 groups: Group A received 20 mg orally of fluoxetine daily, Group B received 20 mg orally of citalopram daily, and Group C received a placebo orally. The duration of the therapy was 90 days. In addition to the medications, all of the participants received physiotherapy

Outcomes

FMMS

Funding source

No financial support received

Notes

IRCT20141116019971N3. Recruitment January 2015 to January 2016. Authors declared no conflicts of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A computer‐generated schedule was used by investigators to assign the patients into one of three groups by block randomization: Group A—20mg P.O. daily of citalopram, Group B—20mg P.O. daily of fluoxetine, and Group C—a placebo (microcrystalline cellulose)."

Allocation concealment (selection bias)

Low risk

Computer‐generated schedule confirmed with authors

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "All of the drugs for each group of subjects were over‐encapsulated by a pharmacist."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Three evaluators were used in our study, namely, neurology residents who were blind to the interventions."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

5/90 discontinued medication due to moving away‐but this would not have introduced because the reasons for missing data are unlikely to be related to the true outcome (Quote: "The main reason for participants leaving the study was noncompliance in terms of taking their drugs regularly (10 subjects stopped taking the treatment but completed their follow‐up), while five participants intentionally failed to attend follow‐up after two months because they were resident in other cities distant from the location of the study.")

Selective reporting (reporting bias)

Low risk

There was just one outcome measure planned (see reference on Iranian clinical trials register) and this was reported

Other bias

Low risk

The study appears to be free of other sources of bias

Bembenek 2020

Study characteristics

Methods

Study type: interventional (clinical trial)

Actual enrolment 61

Allocation: randomised

Intervention model: parallel assignment

Masking: double (participant, care provider)

Primary purpose: treatment

Participants

61 participants

Country: Poland

Setting: inpatient

At randomisation number allocated: N = 61: fluoxetine (n = 30); placebo (n = 31)

% male: fluoxetine (73.3%); placebo (58.1%)

Age: mean age: fluoxetine = 66.6 ± 12.6; placebo = 66.35 ± 12.46

Subtype of stroke

  • Total anterior circulation infarct: fluoxetine (40.0%); placebo (38.7%)

  • Partial anterior circulation infarct: fluoxetine (23.3%); placebo (35.5%)

  • Lacunar infarct: fluoxetine (6.7%); placebo (0.0%)

  • Posterior circulation infarct: fluoxetine (20.0%); placebo (16.1%)

  • Uncertain: fluoxetine (2%); placebo (2%)

Severity of stroke: NIHSS, Median (IQR) fluoxetine (5 (3 to 8)); placebo (6 (4 to 8))

Inclusion criteria

  • Age ≥ 18 years

  • Ischaemic or haemorrhagic stroke confirmed by neuroimaging

  • Within 2 to 15 days from the stroke onset

  • Evidence of neurological deficit at randomisation

Exclusion criteria

  • Subarachnoid haemorrhage (unless secondary to intracerebral bleeding)

  • High probability that the patient would not be available during follow‐up (e.g. another life‐threatening illness)

  • Pregnant or breast‐feeding or of child bearing age not taking contraception

  • History of epileptic seizures

  • Attempted suicide or self‐harm

  • Allergy or contra indication to fluoxetine

  • Taken a monoamine oxidase inhibitor in last 5 weeks

  • Current or recent depression requiring treatment with selective serotonin reuptake inhibitor

  • Already participating in a CTIMP

  • Current use of drugs that cause significant interactions with fluoxetine: history of epileptic seizures; history of allergy to fluoxetine; suicide attempt or self‐ harm; hepatic impairment (ALT > 3 above the upper normal limit) and renal impairment (creatinine > 180 micromol/L)

Interventions

Fluoxetine 20 mg daily (1 capsule) for 6 months (180 capsules) vs placebo

Outcomes

The primary outcomes at 6 months

  • mRS

Secondary endpoints

  • SIS at 6 months

  • NIHSS at baseline, 6 and 12 months

  • Brunnstrom scale at 6 month follow‐up

  • Medical Research Council scale ay 6 month follow‐up

  • BI at 6 and 12 month follow‐up

  • EuroQol 5D‐5L

  • MHI‐5

  • Overall recovery on a VAS

  • Diagnosis of new depression

  • Compliance with drug intake

  • Treatment effects and the occurrence of possible adverse reactions are assessed up to 12 months

Funding source

POLPHARMA S.A. manufactured and donated fluoxetine and placebo for this study. Anna Członkowska, Jan Bembenek, and Katarzyna Kurczych were supported by statutory activity of the Institute of Psychiatry and Neurology, Warsaw, Poland

Notes

Recruitment 19 December 2014 and 13 March 201 8. Authors declared no conflicts of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Patients were randomly assigned in a 1:1 ratio to receive either fluoxetine or a placebo, by use of a computer‐based permuted block randomisation

Allocation concealment (selection bias)

Low risk

Allocation was concealed (further information obtained from the study author Jan Bembenek to confirm this)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Clinicians involved in randomisation and outcome assessments, the patients, and their families, were all masked so as to be unaware of treatment allocation. The placebo capsules were visually identical to the fluoxetine capsules, even when broken open."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Clinicians involved in randomisation and outcome assessments, the patients, and their families, were all masked so as to be unaware of treatment allocation"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

5 withdrew consent (8%) before 6 months but the withdrawals are balanced between groups (3 in the fluoxetine group and 2 in the control group)

From personal communication with the author: a further 2 patients dropped out between 6 and 12 months

Selective reporting (reporting bias)

Low risk

Trial was registered and all outcomes were reported

Other bias

Low risk

The study appears to be free of other sources of bias

Birchenall 2019

Study characteristics

Methods

Study type: interventional (clinical trial)

Intervention model: parallel assignment

Primary Purpose: treatment

Participants

6 participants

Country: France

Setting: inpatient

At randomisation number allocated: 6 although unclear as to which group

% male: not available

Age: not available

Subtype of stroke: not available

Severity of stroke: not available

Time since stroke onset: not available

Inclusion criteria

  • Age 18 to 80 years

  • Social security affiliation

  • Day 3 to day 15 after stroke or brain haemorrhage

  • Hemiparesia with upper limb motor deficit (Fugl‐Meyer score ‐ hand ≤ 10)

  • Informed consent

Exclusion criteria

  • NIHSS > 20

  • Depression (criteria DSM5‐R) with MADRS score > 19

  • History of recurrent bipolar or depressive disorders

  • History of behavior or suicidal idea

  • Family history of extension of the interval QT or congenital long interval QT

  • History of clinical stroke

  • Aphasia preventing correct evaluation of motor and depression scales.

  • Patients treated by antidepressant drugs, IMAO, and neuroleptics in the past month

  • Benzodiazepines within 48 hours preceding inclusion.

  • Intolerance or allergy to fluoxetine (Sandoz® 20 mg pill)

  • Severe swallowing disorders preventing oral administration of the treatment

  • Planned carotid surgery

  • Pregnant or breast‐feeding woman

  • Hepatic failure (TGO and TGP > 2N); severe renal failure (creatinine > 180 micromol/L)

  • Concomitant severe disease not allowing follow‐up

  • Participation to another therapeutic study

  • Contraindication to MRI and TMS

Withdrawal criteria: not stated

Interventions

Experimental: fluoxetine; 1 pill of 20 mg/day, during 3 months

Comparator: placebo of fluoxetine; 1 pill of 20 mg/day, during 3 months

Outcomes

Primary outcome

  • Slope of the curve of recruitment of the MEPs at 3 months

Secondary outcomes recorded at 3 and 6 months

  • Slope of recruitment of the MEPs (effect of a first dose of fluoxetine on the slope of recruitment of the MEPs)

  • Slope of recruitment of the MEPs (persistence of fluoxetine effect on the slope of recruitment of the MEPs to month 6)

  • Index finger force control in paretic hand

  • Index finger force control in non‐paretic hand

Funding source

Not stated

Notes

No published data, unpublished data say 6 patients, none of whom died, so we have used this information

Dates study conducted: February 2014 to August 2015

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information available

Allocation concealment (selection bias)

Unclear risk

No information available

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information available

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information available

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information available

Selective reporting (reporting bias)

Unclear risk

No information available

Other bias

Unclear risk

No information available

Brown 1998

Study characteristics

Methods

Parallel design

Analysis: per protocol: 1 withdrawn (treatment), excluded from analysis

Participants

Diagnosis: stroke, time from stroke to randomisation not reported

Randomised 10 to treatment and 10 to control

Treatment: 9 completed treatment, mean ± SD age 61.4 ± 8.6 years, 55% men

Control: 10 people completed placebo, mean ± SD age 63.7 ± 5.4 years, 60% men

Emotionalism criteria: emotionalism of at least 4 weeks' duration assessed during semi‐structured interview using a modified Lawson and MacLeod rating scale, in addition to frequency of outbursts

Exclusion criteria: cognitive impairment, dysphasia, major depressive disorder

Interventions

Treatment: fluoxetine 20 mg daily

Control: matched placebo

Duration: 10 days

Duration of follow‐up: (end of treatment to end of study) 0

Outcomes

Used leaving the study early

Unable to use data from HDRS, Lawson and MacLeod Scale, self‐rating scales (mean and SD not presented)

Also reported emotional outbursts; we have not used these in our analyses

AEs: not presented

Funding source

Funder not stated

Notes

Dates of study not stated; conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation method not stated

Allocation concealment (selection bias)

Unclear risk

Randomised by independent statistician

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

States blinding of patients and nursing staff, matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

States blinding of rating clinicians

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 withdrawn (5% of participants); we categorised this as low risk

Selective reporting (reporting bias)

Unclear risk

Insufficient information to make judgement

Other bias

Low risk

No other obvious biases, baseline balanced

Burns 1999

Study characteristics

Methods

Parallel design

Analysis: ITT: 2 withdrawn and 1 death (treatment), 1 death (placebo), last value carried forward

Participants

Diagnosis: stroke.

Months from stroke: median (range) 10.5 months (1 ± 156) in sertraline group and 5.5 months (1.5 ± 48) in the control group

Treatment: 14 people

Control: 14 people

Exclusion criteria: less than 1 month since stroke, depression or dementia using the DSM III‐R criteria

Interventions

Treatment: sertraline 50 mg daily

Control: matched placebo

Duration: treatment continued for 8 weeks

Duration of follow‐up: 2 weeks off treatment. All scores became non‐significant (though data not reported so could not be used in the analysis)

Outcomes

Able to use

  • improved score on lability scale

  • improved score on clinician's interview based impression of change

  • diminished tearfulness

  • leaving the study early

  • death

  • AEs

Method of collecting AEs was not stated

Unable to use: MADRS, BI, MMSE (data not presented)

Funding source

Funded by an unrestricted personal grant from Pfizer, the manufacturers of sertraline

Notes

Dates of study not stated, conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Blocks of 4 using list produced by medical statistics department

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matched placebo, both active drug and placebo were packed in gelatine capsules with an identical appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Run‐out was single‐blind, treatment was double‐blind, but unclear whether outcome assessors were blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Analysis: ITT, LOCF
4/28 did not complete the study (14%)

Selective reporting (reporting bias)

Low risk

Trial details published on www.strokecentre.org/trials, although unable to use data from MADRS

Given that the main aim was to explore effect on emotionalism, this is unlikely to have biased results

Other bias

Unclear risk

Placebo group younger, uncertain influence on bias

Statistical analysis was carried out independently by the Applied Statistics Research Unit in Canterbury

Cao 2020

Study characteristics

Methods

RCT

Study type: interventional (clinical trial)

Primary purpose: prevention

Participants

100 participants

January 2013 to April 2016

Country: China

Setting: inpatient

At randomisation number allocated: N = 99: escitalopram (n = 52); usual care (n = 47)

% male: escitalopram (%); usual care (%)

Inclusion criteria

  • First ever acute anterior circulation cerebral infarction

  • Met the American Heart Association/American Stroke Association diagnostic criteria for stroke

  • Hospitalised within 1 week of stroke onset

  • Stroke confirmed cranial MRI

Exclusion criteria

  • Recurrent stroke, haemorrhagic stroke and posterior circulation stroke

  • Pre‐stroke depression, history of depression, or receiving mood stabilisers, antipsychotics, or any antidepressant before enrolment

  • Depression caused by other organic brain diseases, or depression caused by psychoactive substances and non‐addictive substances

  • Consciousness disorder, aphasia, and dementia

  • HAMD of ≥ 17

  • NIHSS score of ≥ 20

  • History of cancer and psychosis

  • Chronic obstructive pulmonary disease, heart failure, pulmonary, hepatic, or renal failure, or other severe chronic diseases

  • Serious suicidal tendencies

  • Laboratory and accessory examinations revealing coagulation dysfunction

  • Patients who refuse to participate or cooperate

Interventions

Experimental: prophylactic escitalopram in addition to the basic therapies. Started with 5 mg and gradually titrated to 10 mg/d, oral administration in the morning for 90 days

Comparator: usual care. Secondary prevention of cerebral infarction, brain protection therapy and rehabilitation, without any antidepressants. People with difficulty sleeping could receive Zolpidem or benzodiazepines for a short period of time

Outcomes

Primary and secondary outcome measures not stated

  • 17‐item HAMD

  • NIHSS

  • MMSE

  • BI

Funding source

Notes

No trial registration information

Dates study conducted: All patients were hospitalised patients treated for acute ischaemic stroke from January 2013 to April 2016

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table

Allocation concealment (selection bias)

Unclear risk

Insufficient information available to permit a judgement of ‘low risk’ or ‘high risk’. This is usually the case if the method of concealment is not described or not described in sufficient detail to allow a definite judgement

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Control group did not receive a placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Treatment evaluation conducted by a physician blind to patient’s clinical data, but it does not state whether the physician was blind to treatment allocation. So the judgement was that the risk of bias was unclear

Incomplete outcome data (attrition bias)
All outcomes

High risk

There are two publications‐the numbers initially included in the two papers do not match; also two patients out of 49 randomised to escitalopram dropped out. Given the inconsistency in the numbers reported, we judge that there is the potential for high risk of bias

Selective reporting (reporting bias)

Unclear risk

No protocol is available

Other bias

Unclear risk

The authors did not reply to questions and so we have graded this as unclear

Chen 2001

Study characteristics

Methods

Randomised trial

Aim: to observe effects of integrative Chinese herb YuLeShu and fluoxetine on the depressive symptoms and rehabilitation of neurological impairment in patients with post‐stroke depression

Participants

Country: China

Setting: not described

Participants: internal carotid system cerebral infarction or haemorrhage within previous 2 months

Fluoxetine: 19 people, mean age 61.71 ± 8.13 years, 8 men

Control: 18 people, mean age 62.85 ± 7.32 years, 7 men

Depression: diagnosis of depression according to DSM‐IV

Inclusion criteria: HDRS ≥ 20 but < 35 and/or Zung SDS ≥ 41

Exclusion criteria: HDRS > 35, previous depression, aphasia, severe cardiac, pulmonary, hepatic and renal diseases, previous stroke

Interventions

3 groups: fluoxetine plus usual care versus YuLeShu plus usual care versus usual care. We are using the fluoxetine plus usual care versus usual care alone in the comparison

Outcomes

HDRS

Zung SDS

BI

Scandinavian Neurological Stroke Scale (also known as CSS)

Stated no side effects, but not clear which side effects were sought, or how they were sought. They were reported at 4, 8 and 12 weeks after treatment

Funding source

Funded by a local scientific academic fund, drug company not involved

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "using a computer", but method not described. Insufficient information about the sequence generation process available to permit a judgement of ‘low risk’ or ‘high risk'

Allocation concealment (selection bias)

Unclear risk

The method of concealment is not described or not described in sufficient detail to allow a definite judgement of ‘low risk’ or ‘high risk’

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information about blinding of outcome assessment available to permit a judgement of ‘low risk’ or ‘high risk’

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 2 people dropped out of the fluoxetine group, 1 dropped out of the YuLeShu group and 2 dropped out of the control group

Selective reporting (reporting bias)

Unclear risk

Protocol not published. Insufficient information available to permit a judgement of ‘low risk’ or ‘high risk’

Other bias

Unclear risk

Reported that of the people who completed the tests, there were no differences in baseline

No comment on whether there were differences in baseline for the entire group

Chen 2002

Study characteristics

Methods

Parallel group (3 groups: doxepin, paroxetine, placebo; we used the paroxetine and placebo data in our review)

Aim: treat depression and determine effect on neurological function

Participants

Country: China

Setting: unclear

Stroke diagnosis: diagnostic criteria of the 4th National Meeting of the Cerebrovascular Diseases proved by CT or MRI

Time since stroke: not known

Depression diagnosis: Classification and Diagnosis of Psychosis in China (2nd edition)

Treatment: 24 people, age and gender not given

Control: 24 people, age and gender not given

Exclusion: pre‐stroke mental disease, cognition disorder (MMSE < 24), marked deterioration in depression during treatment (HAMD > 24) or suicide mood, intolerance to drug

Interventions

Treatment: paroxetine 20 mg 3 times per day

Control: placebo guvitamine once per day

Duration of treatment: 8 weeks

Duration of follow‐up (post‐treatment to study end): unclear: follow‐up is performed 'after treatment' so we assume this is at 8 weeks (so post‐treatment to study end = 0)

Outcomes

HAMD

BI

CSS

Death/side effects/leaving the trial early

Method of reporting side effects not stated

Funding source

Funder not stated, unclear if there was drug company involvement

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about the sequence generation process available to permit a judgement of ‘low risk’ or ‘high risk’

Allocation concealment (selection bias)

Unclear risk

The method of concealment is not described to allow a definite judgement of ‘low risk’ or ‘high risk’

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information available to permit a judgement of ‘low risk’ or ‘high risk’

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information about blinding of outcome assessment available to permit a judgement of ‘low risk’ or ‘high risk’

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 4 in placebo and 0 in paroxetine

Selective reporting (reporting bias)

Unclear risk

Insufficient information available to permit a judgement of ‘low risk’ or ‘high risk’

Other bias

Unclear risk

Demographic data not provided, so we cannot determine whether the baseline was balanced

Chen 2005a

Study characteristics

Methods

To observe the changes of neurotransmitter in people with post‐stroke depression by using encephalofluctuography technology, and observe the effect of antidepression treatment on the activity of neurotransmitter

Participants

48 participants with post‐stroke depression

Interventions

Treatment: 24 people received citalopram 20 mg plus usual care, or fluoxetine if side effects such as nausea, emesis

Control: 24 people usual care alone

Outcomes

Encephalofluctuography technology

Level of sympathin and 5‐hydroxytryptamine at 4 weeks and 3 months after treatment started

Funding source

Not stated

Notes

No data from our endpoints of interest, so data not included in a meta‐analysis

Recruitment March 2001 to December 2001

Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Randomly divided" but method not stated. Insufficient information about the sequence generation process available to permit a judgement of ‘low risk’ or ‘high risk’

Allocation concealment (selection bias)

Unclear risk

The method of concealment is not described or not described in sufficient detail to allow a definite judgement of ‘low risk’ or ‘high risk’

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

Unclear

Other bias

Unclear risk

Unclear

Chen 2005b

Study characteristics

Methods

Parallel group

Analysis: according to allocated treatment group

Participants

Country: China

Setting: inpatient

Stroke criteria: first ever stroke, onset time ≤ 7 days, haemorrhagic and ischaemic, clinical diagnosis plus confirmation by imaging (though not clear whether a stroke lesion had to be present), at least 1 limb with muscle power grade 3 or less, BI ≤ 50, no consciousness disturbance

Mood criteria: HAMD > 16

Treatment: 40 people, mean age 63.5 years, 29 men

Control: 38 people, mean age 65.8 years, 25 men

No difference in baseline depression and BI between treatment and control group

Excluded: severe cardiac, hepatic and renal organic diseases, psychiatric disorders

Interventions

Treatment: paroxetine 20 mg daily plus routine stroke medication, nerve nutritional agents, acupuncture and rehabilitation

Control: routine stroke medication, nerve nutritional agents, acupuncture and rehabilitation

Duration of treatment: 12 weeks

Duration of follow‐up (post‐treatment to study end): 0 weeks

Outcomes

HAMD

BI

Death

Number completing the trial

AEs not reported

Funding source

No description of funding

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: none

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

No obvious risks, baseline similar

Chen 2015

Study characteristics

Methods

To investigate the effect of early antidepressant intervention on the acute depression and rehabilitation of neurological function after stroke

Participants

96 acute depression patients after stroke were selected and randomly divided into study group and control group. Stroke diagnosed according to the diagnostic criteria of stroke by the 4th Congress of Chinese Cerebrovascular Diseases, which requires both clinical and imaging criteria to be met. HAMD ≥ 17. Excluded patients with mental disorders. Range of disease: 2‐9 months

Interventions

The study group orally took the antidepressant ( citalopram) and the control group took placebo for 8 weeks

Outcomes

Depression (HAMD), neurological function (NIHSS), ADL (BI). Substance P and neuropeptide Y

Funding source

No information regarding funding

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table

Allocation concealment (selection bias)

Unclear risk

Information not provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Information not provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

information not provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

information not provided

Selective reporting (reporting bias)

Unclear risk

Information not provided

Other bias

Unclear risk

No information provided e.g. on source of funding

Cheng 2003

Study characteristics

Methods

Parallel design

Aim: to treat depression and augment rehabilitation

Analysis: according to allocated treatment group

Participants

Location: China

Setting: inpatient

Treatment: 25 people

Control: 32 people

Whole group (including non‐depression group, depression control group and depression treatment group): 132 (mean age 62 ± 12 years, 79 men)

Stroke: ischaemic stroke or PICH, clinical diagnosis plus confirmation on brain imaging (not clear that a stroke lesion had to be present), clear consciousness

Depression diagnosis (at 2 weeks after stroke onset): psychiatric interview, DSM IV criteria

Excluded: major psychological trauma history in previous 1 year, severe mental retardation, severe impairment of lingual expression or comprehension, major complicated medical event in previous 1 year

Interventions

Treatment: fluoxetine 20 mg daily

Control: no fluoxetine

Duration of treatment: 6 months

Duration of follow‐up (post‐treatment to study end): 6 months

Outcomes

SSS

ADL

HAMD

Zung SDS

Zung SAS

No deaths, none left trial early

No data on AEs

Funding source

No description of funding

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

59 participants were diagnosed to have depression by symptoms but only 57 were included in the results table

Selective reporting (reporting bias)

High risk

No protocol, no report of the results of the self‐rating anxiety scale

Other bias

Unclear risk

No clear description of differences between the treatment and control group

Chollet 2011

Study characteristics

Methods

Randomised parallel‐group trial

Participants

Location: France

Setting: stroke units

Inclusion criteria: aged 18 to 85 years with FMMS of 55 or less, acute ischaemic stroke with hemiparesis or hemiplegia, 5 to 10 days after stroke onset, unclear if there had to be a visible lesion on brain imaging

Treatment: 59 people, mean ± SD age 66.4 ± 11.7 years; 63% men

Control: 59 people, mean ± SD age 62.9 ± 13.4 years; 59% men

Comparability of treatment groups: total FMMS score fluoxetine 17.1 compared with 13.4 in placebo
Previous stroke more common in the fluoxetine group; fluoxetine group had more diabetes

Exclusions: clinical depression or treatment with antidepressants, MADRS > 19, aphasia severe enough to mask detection/assessment of depression, pregnancy, patient on neuroleptics/benzodiazepines, owing to undergo carotid endarterectomy, other major diseases that would prevent follow‐up

Interventions

Treatment: fluoxetine 20 mg daily for 90 days

Control: identical capsules to active drug

Duration of treatment: 90 days

Duration of follow‐up (treatment end to study end): 0 days

Outcomes

Primary outcome: the mean change of FMMS score between inclusion (day 0) and day 90 after the start of the study drug

Secondary endpoints were NIHSS, mRS and MADRS measured at days 0, 30 and 90

Funding source

Funded by French national programme for clinical research: the sponsor had no involvement in study design, data collection, data analysis, data interpretation or writing the report

Notes

Recruitment 14 March 2005 to 9 June 2009. Authors state "no conflicts of interest"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Balanced by centre with an allocation based on a block size of 4 generated with a computer random‐number generator

Allocation concealment (selection bias)

Low risk

Sequentially‐numbered opaque envelopes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical capsules for control arm

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All study site investigators and all investigators were masked to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 2 participants died (1 in each group) and 3 dropped out; not stated how missing outcome data were dealt with

Selective reporting (reporting bias)

Low risk

Trial protocol published on www.strokecentre.org/trials, all outcomes were reported

Other bias

Unclear risk

Note difference in baseline; particularly for the total FMMS score (17.1 in fluoxetine and 13.4 in the placebo): it is not clear what effect this had on results, so we have classified this as 'unclear risk'

Dam 1996

Study characteristics

Methods

Parallel design

Analysis: per protocol: withdrawn because of AEs (2 treatment), all excluded from analysis

Participants

Location: Italy

Setting: unclear

Treatment: 18 people, mean ± SD age 68 ± 9 years, 44% men

Control: 17 people, mean ± SD age 68 ± 5.5 years, 44% men

Stroke criteria: ischaemic, unilateral MCA territory stroke, diagnosis via clinical signs and CT (100%), stroke 1 to 6 months prior to randomisation (average time 3 months)

Other inclusion criteria: unable to walk

Comparability of treatment groups: balanced

Exclusion: history of major affective disorders; alcohol abuse; or a history or evidence or both of severe heart, lung, kidney or liver diseases or mental deterioration

Interventions

Treatment: fluoxetine 20 mg daily

Control: matched placebo

Duration: treatment continued on average 74 ± 6 days, duration not reported for control group

Duration of follow‐up (treatment end to study end): 0

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS

Additional: graded neurological scale (HSS), BI

Leaving the study early

Death

AEs including seizures ‐ unclear if these were reported systematically

Funding source

Funding source not stated

Notes

Dates of recruitment and conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

No description

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Examining neurologists blind to treatment".

Comment: Unclear if this refers to outcome assessors or the neurologist caring for the participant. However, placebo was 'matched' so this is low risk as the treating physician and the participants would have been blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

See above

Incomplete outcome data (attrition bias)
All outcomes

High risk

2/35 dropouts (5.7%), per‐protocol analysis

Selective reporting (reporting bias)

Low risk

Trial available, including results on www.strokecentre.org/trials: all specified outcome measures were reported

Other bias

Low risk

Baseline characteristics similar in the 2 groups

Dike 2019

Study characteristics

Methods

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: single (investigator)

Primary purpose: treatment

Participants

60 participants

Country: Nigeria

Setting: inpatient

At randomisation number allocated: N = 1500: fluoxetine (n = 750 ); placebo (n = 750 )

% male: fluoxetine (53.3%); placebo (43.3%)

Age: mean age: fluoxetine = 59 ± 11; placebo = 62 ± 9

Inclusion criteria

  • 18 to 85 years of age

  • Ischaemic stroke, unilateral, supra‐tentorial confirmed by neuroimaging

  • Presentation within first 14 days of stroke onset

  • NIHSS score ≤ 16

  • Hemiparesis or hemiplegia

  • FMMS ≤ 55

  • Informed consent

Exclusion criteria

  • Haemorrhagic stroke on CT

  • Glasgow coma score < 8

  • NIHSS score > 16

  • Cardiovascular/metabolic/respiratory instability: hypertensive emergency or hypotension/acidosis or alkalosis/RR > 24 cycles per minute

  • Previous central/peripheral nerve injury

  • Current use of a medication likely to have an adverse interaction with fluoxetine

  • Concurrent medications interacting with SSRI

  • Substantial premorbid disability

  • Depression (MADRS score > 19)

  • Current use of antidepressant medication

  • Pregnancy

Interventions

Experimental: 20 mg fluoxetine for 30 days plus standard treatment

Comparator: standard treatment

Outcomes

Primary outcome

  • Changes in FMMS at day 14 and day 30

Secondary outcomes

  • NIHSS at day 30

  • mRS at day 30

Funding source

N o funding

Notes

PACTR201412000967245. Recruitment between January 2015 and May 2016. All authors declare no conflicts of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Block randomization was used in the assignment of study participants. Permuted blocks of six (6) for two groups were drawn up. A random selection of possibilities was done using a list of random numbers generated with STATA 12."

Allocation concealment (selection bias)

Low risk

Quote: "Allocation concealment was done using sequentially numbered envelopes."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "This was a single‐blind randomized controlled trial that compared motor recovery between 2 groups of stroke patients: those on fluoxetine 20mg daily + standard therapy; and the control group who received standard therapy only." Thus the participants would have known their treatment allocation

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

22/60 lost to follow‐up (presumably including the 7 in the intervention group and the 8 in the control group who died). 6 in fluoxetine group stopped treatment but were still included in the analysis. It is unclear what effect this would have had, hence the judgement about unclear risk of bias

Selective reporting (reporting bias)

Low risk

All the prespecified outcomes were reported

Other bias

Unclear risk

Insufficient information to to assess whether an important risk of bias exists

EFFECTS 2020

Study characteristics

Methods

Multicentre RCT

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: quadruple (participant, care provider, investigator, outcomes assessor)

Primary purpose: treatment

Participants

1500 participants

Country: Sweden

Setting: inpatient

At randomisation number allocated: N = 1500: fluoxetine (n = 750 ); placebo (n = 750 )

% male: fluoxetine (62%); placebo (62%)

Age: mean age: fluoxetine = 70.6 ± 11.3; placebo = 71.0± 10.5

Subtype of stroke

  • Total anterior circulation infarct: fluoxetine (27%); placebo (29%)

  • Partial anterior circulation infarct: fluoxetine (46%); placebo (44%)

  • Lacunar infarct: fluoxetine (15%); placebo (16%)

  • Posterior circulation infarct: fluoxetine (10%); placebo (9%)

  • Uncertain: fluoxetine (x2%); placebo (2%)

Inclusion criteria

  • Age ≥ 18

  • Informed consent can only be obtained from a patient who according to the trial investigator is mentally capable of decision‐making and who, after having received information and got answers to their questions, wants to participate in the trial

  • Brain imaging is compatible with intracerebral haemorrhage or ischaemic stroke

  • Randomisation can be performed between 2 and 15 days after stroke onset and by the research group at the person's local/emergency hospital

  • Persisting focal neurological deficit is present at the time of randomisation severe enough to warrant treatment from the physicians and the patient's and relative's perspective

Exclusion criteria

  • Subarachnoidal haemorrhage (except where secondary to a primary intracerebral haemorrhage)

  • Unlikely to be available for follow‐up for the next 12 months e.g. no fixed home address

  • Unable to speak Swedish and no close family member available to help with follow‐up forms

  • Other life‐threatening illness (e.g. advanced cancer) that will make 12‐month survival unlikely

  • History of epileptic seizures

  • History of allergy or contraindications to fluoxetine

  • Pregnant or breastfeeding

  • Previous drug overdose or attempted suicide

  • Already enrolled into a CTIMP

  • Current or recent (within the last month) depression requiring treatment with an SSRI antidepressant

  • Current use of medications which have serious interactions with fluoxetine

  • Use of any MAOI during the last 5 weeks

Interventions

Fluoxetine (20 mg once daily) for 6 months with oral capsules

Outcomes

Outcomes collected at 6 months and 12 months

Primary outcome

  • mRS

Secondary outcomes

  • Death from all causes

  • HRQoL (EQ5D‐5L)

  • Depression and anxiety (MHI‐5)

  • Level of fatigue (vitality subscale of the Health Questionnaire)

  • Recovery from stroke (SIS)

  • New diagnosis of depression since randomisation

  • Adverse events (including participant‐completed diary)

  • Health and social care utilisation

  • Adherence to trial medication

  • Motor function (NIHSS)

  • Aphasia (NIHSS), aphasia (Norsk Grunntest for Afasi)

  • Depression (MADRS + DSM‐IV/DSM‐V)

  • Cognitive function (MoCA)

Funding source

The Swedish Research Council, the Swedish Heart‐Lung Foundation, the Swedish Brain Foundation, the Swedish Society of Medicine, King Gustav V and Queen Victoria’s Foundation of Freemasons, and the Swedish Stroke Association (STROKE‐Riksförbundet)

Notes

clinicaltrials.gov/ct2/show/NCT02683213. Recruitment between Oct 20, 2014, and June 28, 2019

BN has received honoraria for data monitoring committee work in the SOCRATES and THALES trials (AstraZeneca) and the NAVIGATE‐ESUS trial (Bayer). HW has received grants from the Swedish Medical Research Council (Vetenskapsrådet) during the conduct of the study; the grant was for the study that is presented inthe submitted manuscript. GJH has received grants from the National Health and Medical Research Council (NHMRC) of Australia, Vetenskapsrådet, and UK National Institute for Health Research Technology, during the conduct of the study; and personal fees from American Heart Association, outside of the submitted work. MD reports that the University of Edinburgh received some funding from the grants for EFFECTS (Vetenskapsrådet) in relation to its provision of a randomisation system. MLH has received grants from the NHMRC of Australia, outside of the submitted work. All other authors declare no competing interests

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A physician or nurse entered patients’ baseline data into a secure web‐based randomisation system. The system checked the data for completeness and consistency and allocated each patient an identification number and a treatment number. Patients were randomly assigned in a 1:1 ratio to either oral fluoxetine 20 mg once daily or placebo for 6 months."

Allocation concealment (selection bias)

Low risk

Quote: "secure web‐based randomisation system"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients, their families, health‐care personnel, investigators, outcomes assessors, and staff in the coordinating centre (Karolinska Institutet, Department of Clinical Sciences Danderyd Hospital, Stockholm, Sweden), and the pharmacy were masked to treatment allocation. The placebo capsules were visually identical to the fluoxetine capsules, even when broken open. The success of the masking procedure was not assessed. An emergency unmasking system was available but was designed so that the coordinating centre and staff doing follow‐up continued to be masked throughout the study."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Patients, their families, health‐care personnel, investigators, outcomes assessors, and staff in the coordinating centre (Karolinska Institutet, Department of Clinical Sciences Danderyd Hospital, Stockholm, Sweden), and the pharmacy were masked to treatment allocation"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 11 dropped out; 2 crossed over; the rest were included in the analysis

Selective reporting (reporting bias)

Low risk

All outcomes that were prespecified were reported

Other bias

Low risk

The study appears to be free of other sources of bias

Feng 2004

Study characteristics

Methods

Aim: to study the influence of Jieyu Huoxue decoction on rehabilitation of patients with depression after cerebral infarction

Participants

Country: China

4 groups: fluoxetine plus usual care, Jieyu Huoxue decoction plus usual care, usual care in people with depression, usual care in people with no depression

We are using data from 'fluoxetine plus usual care' versus 'usual care in people with depression'

Setting: mixed inpatient and outpatient

Stroke criteria: ischaemic stroke within 1 month of stroke onset, clinical diagnosis plus confirmation by imaging. Did not state whether a visible lesion was needed to make a diagnosis

Depression: psychiatric interview using DSM IV, Zung SDS ≥ 41

Included those with no previous psychiatric history

54 participants with post‐stroke depression were randomised

18 received fluoxetine plus usual care, 18 received usual care only and 18 received Jieyu Huoxue decoction

Of the 54 participants with depression randomised, mean age: 71.5 ± 6.7 years, 24 men

Excluded: previous stroke, previous depression, and severe cardiac, pulmonary, hepatic and renal diseases

Interventions

Treatment: fluoxetine 20 mg daily plus usual stroke care

Control: usual stroke care

Duration of treatment: 60 days

Duration of follow‐up (post‐treatment to study end): 0 weeks

Outcomes

Zung SDS

ADL: although score not referenced, so not used in analysis

MESSS

Reported side effects in fluoxetine group but not in the control group

Unclear how side effects were collected

Funding source

Funding source not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not stated

Allocation concealment (selection bias)

Unclear risk

No description

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

8 participants dropped out (2 in fluoxetine group, 2 in the depression control group, 1 in the Jieyu Huoxue decoction, 3 in no‐depression control)

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline balanced

FOCUS 2019

Study characteristics

Methods

Multicentre RCT

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

3127 participants

Country: UK

Setting: inpatient

At randomisation number allocated: N = 3127: fluoxetine (n = 1564); placebo (n = 1563)

% male: fluoxetine (62%); placebo (61%)

Age: mean age: fluoxetine = 71·2 ± 12.4; placebo = 71·5 ± 12.1

Subtype of stroke

  • Total anterior circulation infarct: fluoxetine (20%); placebo (20%)

  • Partial anterior circulation infarct: fluoxetine (36%); placebo (35%)

  • Lacunar infarct: fluoxetine (20%); placebo (18%)

  • Posterior circulation infarct: fluoxetine (12%); placebo (15%)

  • Uncertain: fluoxetine (2%); placebo (2%)

Severity of stroke: NIHSS, Median (IQR) fluoxetine (6 (3 to 11)); placebo (6 (3 to 11))

Time since stroke onset: mean days: fluoxetine 6.9 ± 3.6; placebo 7.0 ± 3.6

Inclusion criteria

  • Age > 18 years

  • Brain imaging consistent with intracerebral haemorrhage or ischaemic stroke

  • Randomisation can be performed between 2 and 15 days after stroke onset

  • Persisting focal neurological deficit is present at the time of randomisation

Exclusion criteria

  • SAH

  • Unlikely to be available for follow up at 12 months

  • Patient and/or carer unable to understand spoken or written English

  • Other life‐threatening illness

  • Pregnant or breast‐feeding or of child bearing age not taking contraception

  • History of epileptic seizures

  • Attempted suicide or self‐harm

  • Allergy or contra indication to fluoxetine

  • Taken a monoamine oxidase inhibitor in last 5 weeks

  • Current or recent depression requiring treatment with selective serotonin reuptake inhibitor

  • Already participating in a CTIMP

Interventions

Experimental: 20 mg orally once daily for 6 months

Comparator: matching placebo orally once daily for 6 months

Outcomes

Primary outcome

  • mRS at 6 months

Secondary outcome measures

  • Deaths from all causes at 6 and 12 months

  • mRS at 12 months

  • SIS

  • Euroquol 5D‐5L

  • MHI‐5

  • Vitality subscale of SF36 (as an assessment of fatigue)

  • Diagnosis of depression

  • Other AEs

  • Adherence to the trial medication

  • Health and social care resources used during follow‐up

Funding source

MHRA approval granted. Start‐up phase funded by The Stroke Association. Main phase funded by NIHR

Notes

ISRCTN83290762. Recruitment 10 September 2012 to 31 March 2017. Authors declared no conflicts of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomly assigned in a 1:1 ratio to receive fluoxetine or placebo, by use of a centralised randomization system."

Allocation concealment (selection bias)

Low risk

Quote: "Patients were randomly assigned in a 1:1 ratio to receive fluoxetine or placebo, by use of a centralised randomization system."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients, their families, and the health‐care team including the pharmacist, staff in the coordinating centre, and anyone involved in outcome assessments were all masked to treatment allocation by use of a placebo capsule that was visually identical to the fluoxetine capsules even when broken open."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Patients, their families, and the health‐care team including the pharmacist, staff in the coordinating centre, and anyone involved in outcome assessments were all masked to treatment allocation by use of a placebo capsule that was visually identical to the fluoxetine capsules even when broken open."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

For the primary outcome of mRS at 6 months data were available for fluoxetine n = 1553/1564 (99.3%) and placebo n = 1553/1563 (99.3%)

Selective reporting (reporting bias)

Low risk

The study protocol is available and all of the study's prespecified (primary and secondary) outcomes that are of interest in the review have been reported

Other bias

Low risk

The study appears to be free of other sources of bias

Fruehwald 2003

Study characteristics

Methods

Parallel design

Analysis: per protocol

Withdrawals: death (1 treatment), withdrawn owing to AEs (1 treatment, 2 control), all excluded from analysis

Participants

Location: Austria

Setting: inpatients

Treatment: 28 people, mean ± SD age 65 ± 14 years, 46% men

Control: 26 people, mean ± SD age 64 ± 14 years, 71% men

Stroke criteria: ischaemic stroke and PICH; diagnosis via clinical signs and CT (100%); stroke on average 11 days prior to randomisation

Depression criteria: psychiatric interviews, HDRS score > 15

Other entry criteria: not stated

Comparability of treatment groups: non‐significant trend towards more women and right‐sided strokes in treatment group

Exclusion criteria: MMSE < 20, more than mild communication deficit, diseases of the central nervous system and previous neurodegenerative or expansive neurological disorders

Interventions

Treatment: fluoxetine 20 mg daily, dose escalation at 4 weeks if HDRS score > 13

Control: matched placebo

Duration of treatment: 12 weeks

Duration of follow‐up (end of treatment to study end): 15 months

Outcomes

Depression: change in scores from baseline to end of treatment of HDRS, BDI, and CGI (item 1)

Proportion of responders (< 13 HDRS)

Additional: SSS

Death

AEs (selected data)

Unable to use: RS, BI, MMSE (data not presented at follow‐up)

AEs data on dizziness, nausea and cephalalgia (data not presented by group)

Funding source

The medication was supplied by Lannacher Heilmittel, Lannach, Austria

Notes

Recruitment 1 June 1998 to 31 Decmeber 1998. Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computerised randomisation, using random permutated block design

Allocation concealment (selection bias)

Low risk

Centralised concealment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

States blinded, used matching placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

States blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

4/54, per protocol analysis

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline balanced

All participants were randomly assigned to either fluoxetine or placebo treatment by the drug company independently of the research teams and the study centres

Gao 2017

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

274 participants

Country: China

Setting: outpatient

At randomisation number allocated: N = 274, citalopram (n = 91); placebo (n = 91); cognitive behavioural therapy (n = 92)

% male: 51.8%

Age: mean age, citalopram 66.0 ± 7.3 (n = 91); placebo 67.2 ± 9.6 (n = 91); cognitive behavioural therapy 64.9 ± 8.0 (n = 92)

Subtype of stroke: not available

Severity of stroke: not available

Time since stroke onset: acute ischaemic stroke within the previous 7 days

Inclusion criteria

  • Age > 18

  • First ever ischaemic stroke meeting World Health Organization (WHO) diagnostic criteria confirmed by MRI

  • No history of depression

  • No antidepressant use prior to the study

Exclusion criteria

  • No consent

  • Premorbid stroke related impairment

  • BI < 10

Interventions

Experimental: citalopram 20 mg per day for a minimum of 3 months + general discussions

Comparator 1: placebo + general discussions

Comparator 2: placebo + cognitive behavioural therapy

Outcomes

  • Depressive symptoms (17‐item HAMD), Bech‐Rafaelsen Melancholia Scale (MES)) at 3 months

  • Drug side‐effects (Udvalg for Kliniske Undersogelser side‐effect scale at 2, 4, and 6 weeks, and 3 months

  • Performance in ADL (BI) at 3 months

  • Functional impairment (FIM scale) at 3 months

Funding source

Natural Science Foundation of China [81100243, 81171131, 81272564, 81272795, 81100893, 81172197, and 81372484], the Natural Science Foundation of Liaoning Province in China [No. L2013296], and Liaoning Science and Technology Plan Projects [No. 2011225020]

Notes

This trial was particular in that recruitment happened at 4 different time points: at 0 months, 3 months, 6 months, and 9 months from discharge. Inclusion criteria required that participants suffered from post‐stroke depression. Participants were invited to complete the BDI and those with a score > 10 were included, provided other criteria were met

Group 'placebo + general discussions' and 'citalopram + general discussions were included. No significant differences observed in the 2 included groups

Dates study conducted: participants enrolled between October 2011 and June 2013

Declarations of interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization into one of three intervention groups was undertaken by an independent researcher using computer‐generated random number sequences ..."

Allocation concealment (selection bias)

Low risk

Quote: "... that were prepared in advance and placed in consecutively numbered, sealed, opaque envelopes."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Study described as "single blind"

Quote: "The researcher successively opened the envelopes corresponding to different time periods and determined the intervention by patient number."

Quote: "The study therapists acted as clinical evaluators."

Quote: "The study therapists were asked not to divulge any treatment information to their patients."

Comment: Care providers, investigator and outcome assessors were all aware of allocation

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "The study therapists acted as clinical evaluators."

Quote: "The study therapists were asked not to divulge any treatment information to their patients."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "in Group A [placebo + general discussions], one patient violated protocol in the second time period, one could no longer be reached, and one left the study owing to stroke recurrence in the third time period; in Group B [citalopram + general discussions], persistent side‐effects from the drugs led five patients to leave the study (two owing to orthostatic dizziness, one owing to palpitation, and two owing to constipation)"

Comment: Attrition reported for each intervention group and reasons given

Group A (placebo + general discussions) 3/91 = 3% attrition

Group B (citalopram + general discussions) 5/91 = 5% attrition

Overall = 4% attrition

Selective reporting (reporting bias)

Unclear risk

There is no study protocol available. Therefore insufficient information to judge high or low

Other bias

Low risk

The study appears to be free from other sources of bias

GlaxoSmithKline 1998

Study characteristics

Methods

Parallel group

Analysis: according to treatment group

Participants

Location: not stated

Setting: not stated

Stroke criteria: "documented diagnosis of stroke within 12 months prior to screening"

Mood: MADRS score > 17

Treatment: 112 people, age 64.3 ± 11.4 years, 61 men

Control: 117 people, 65.6 ± 10.5 years, 64 men

Excluded: concurrent psychiatric disorders, concurrent psychotropic pharmacotherapy, patients who posed a suicidal risk, patients with substance abuse/dependence, concurrent psychotropic pharmacotherapy, MMSE < 24, participating in another clinical trial, serious medical condition or clinically significant finding on screening or baseline evaluation that would preclude the administration of paroxetine and an intolerance to paroxetine

Interventions

Treatment: paroxetine 20 to 50 mg daily

Control: placebo (not stated whether matching)

Duration of treatment: 8 weeks

Duration of follow‐up (treatment to study end): 0 weeks

Outcomes

Change from baseline to endpoint in MADRS

Proportion of participants scoring < 8 on the MADRS total score at the endpoint (we used this in our analysis)

Changes from baseline to endpoint on the BI

Change from baseline to endpoint on RS score

Change from baseline to endpoint on the Clinical Global Improvement Severity of Illness Score (CGI‐S Proportion of responders based on CGI‐Global Improvement (CGI‐G) score (score of < 4) at endpoint

GI side effects reported, but unclear whether these are 'events' or 'participants', so we cannot use these data. It is not clear how the side effects were collected

Withdrawal from study

Funding source

Source of funding not stated, but we assume it was funded by GlaxoSmithKline

Notes

Study period 29 August 1998 to 15 October 1999. Conflicts of interest not stated. Study number PAR625. Date updated: 11 March 2005

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation method not described

Allocation concealment (selection bias)

Unclear risk

Allocation concealment: not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Blinding not described, used placebo but not stated whether identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Blinding not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

20 in each group dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Insufficient information to make clear judgement

Gong 2020

Study characteristics

Methods

RCT

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: double (participant, outcomes assessor)

Primary purpose: treatment

The study was designed as a double‐blind, randomized clinical trial in order to obtain evidence for using Shu‐Gan‐JieYu capsule as an integrative, anti‐depressive treatment for motor recovery in post‐stroke patients

Participants

254 participants

Country: China

Setting: inpatient

At randomisation number allocated: N = 254:

Group A (Shu‐Gan‐Jie‐Yu only (n = 64), Group B (fluoxetine n = 64), Group C (Shu‐Gan‐Jie‐Yu and fluoxetine (n = 64)), and Group D (placebo (n = 62))

% male: Group A (Shu‐Gan‐Jie‐Yu only (n = 68.5%), Group B (fluoxetine n = 64.2%), Group C (Shu‐Gan‐Jie‐Yu and fluoxetine (n = 67.3%)), and Group D (placebo (n = 61.0%))

Age: mean age:

Group A (Shu‐Gan‐Jie‐Yu only (58.36 ± 15.28), Group B (fluoxetine 56.68 ± 17.59), Group C (Shu‐Gan‐Jie‐Yu and fluoxetine (55.95 ± 19.66), and Group D (placebo (57.79 ± 17.54)

Inclusion criteria

  • Age ≥ 18

  • suffering from hemiparesis or hemiplegia as a result of a first‐time acute ischemic stroke within the past 24 hours

  • initial FMMS score lower than 55

Exclusion criteria

  • NIHSS score less than 5

  • Prior disabilities, including aphasia, cognitive disorders, and motor disorders due to stroke, or any other neurodegenerative disease

  • Pregnant or breastfeeding

  • Currently taking antidepressants

  • Recurrent suffering from acute stroke

  • Met the recombinant tissue plasminogen activator (rtPA) treatment and treated by thrombolysis

  • Contraindications for therapy, including renal insufficiency (glomerular filtration rate < 30 mL/min), abnormal liver function tests, hyponatremia, or a long QT interval on an electrocardiogram

Interventions

Group A (Shu‐Gan‐Jie‐Yu only), Group B (fluoxetine), Group C (Shu‐Gan‐Jie‐Yu and fluoxetine), and Group D (placebo)

Outcomes

mRS: categorised as (0&1), (2&3), (> 4)

Fugl‐Meyer: continuous

Funding source

Supported by a grant from The National Natural Science Foundation of China (81373619) and the Plateau Discipline of Neurology of Integrated Traditional Chinese and Western Medicine in Pudong New Area (PDZY‐2018‐0606)

Notes

No trila registration number. Recruitment from July 2015 to December 2018. The authors declare that they have no conflicts of interest

Please note: final per protocol number stated as 222, but Table 1 baseline only includes 219; Table 2 mRS n=291; Table 3 30 & 90 days n=222

Denominators change for each outcome – even different for mRS 90 day and Fugel‐Meyer 90 day

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Simple randomization method … computer‐generated schedule to assign the patients who met the criteria into one of four groups by block randomization, in a ratio of 1:1:1:1, without stratification"

Allocation concealment (selection bias)

Unclear risk

The method of allocation concealment is not described to allow a definite judgement

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "All capsules looked identical ... in order to guarantee the consistency of capsule appearance. All of the patients were told the effects of the drugs ... could improve motor recovery even if they did not have a depressive disorder."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Three neurology residents who were blinded to the interventions served as eva‐ luators in the study. The evaluators obtained the patients’ FMMS and mRS scores at enrollment and follow‐up on day 30 and day 90 after the start of the intervention. They also recorded any adverse events observed during the study."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Final per protocol number stated as 222, but Table 1 baseline only includes 219; Table 2 mRS n = 291; Table 3 30 and 90 days n = 222. Denominators change for each outcome – and are different for mRS 90 day and Fugel‐Meyer 90 day

Selective reporting (reporting bias)

Unclear risk

No published protocol to cross check for selective reporting

Other bias

High risk

Non‐standard analysis of outcomes

Guo 2009

Study characteristics

Methods

Parallel group, 3‐arm trial, comparing sertraline plus routine care versus routine care versus acupuncture plus routine care. We are using the sertraline plus routine care versus routine care in this review

Aim: to treat depression

Analysis: according to allocated treatment

Participants

Country: China

Setting: unknown

Stroke criteria: first ever stroke, clinical diagnosis plus relevant lesion on imaging, age ≥ 60 years old

Depression criteria: HAMD score ≥ 8, no depression prior to stroke

Treatment: 40 people, mean age 67.6 ± 12.43 years, 23 men

Control: 40 people, mean age 64.5 ± 12.07 years, 22 men

Exclusions: psychiatric disorders or family psychiatric disorders, severe cognitive impairment, global aphasia, sensory aphasia, apraxia, severe cardiac, hepatic, renal, lung or other severe somatic disorder, consciousness disturbance, severe deafness, family or patient unable to comply

Interventions

Treatment: sertraline 50 mg daily plus stroke care (acute, secondary prevention, rehabilitation and psychotherapy)

Control: stroke care (acute, secondary prevention, rehabilitation and psychotherapy)

Duration of treatment: 6 weeks

Duration of follow‐up: (treatment end to study end): 6 months

Outcomes

HAMD

NIHSS

FIM (reported cognition and mobility scores only)

SF‐36

AEs not reported

Funding source

Funded by a local scientific academic fund

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random‐number table

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Outcome assessor blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts, analysed by allocated treatment

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

No obvious risk, balance baseline

He 2004

Study characteristics

Methods

Parallel group

Analysis: according to treatment allocation

Participants

Location: China

Setting: inpatient

Inclusion criteria: all pathological types of stroke, clinical diagnosis plus confirmation by imaging (did not state that a visible lesion was needed to make the diagnosis), first ever stroke

Depression diagnosis: 'HAMD scores'. Translation of paper: did not have to have depression at recruitment

Treatment: 36 people, mean age 70.8 ± 6.7 years, 25 men

Control: 35 people, mean age 70.4 ± 6.8 years, 23 men

Exclusion: psychiatric disorders, dysphasia, consciousness disturbance, agnosia, severe dementia

Interventions

Treatment: fluoxetine 20 mg daily plus usual stroke care

Control: usual stroke care

Duration of treatment: 8 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

SSS

No description of how side effects were collected

Funding source

Funded by local scientific academic fund

Notes

Reported that there were no AEs, so we have assumed no seizures or GI side effects

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Outcome assessors blind"

Incomplete outcome data (attrition bias)
All outcomes

High risk

13 dropped out after randomisation

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

Balanced baseline, no obvious risks

He 2005

Study characteristics

Methods

Parallel design. 3 groups: paroxetine, paroxetine plus psychotherapy, control. We are using paroxetine and control data in this review

Analysis: according to treatment group

Participants

Location: China

Setting: inpatient

Stroke criteria: first ever stroke; ischaemic and haemorrhagic, timing: "acute", clinical diagnosis plus confirmation by imaging (though not clear whether a stroke lesion had to be present or not)

Mood criteria: meets ICD‐10 organic depression and organic anxiety diagnostic criteria on psychiatric interview, HAMD score ≥ 17 and HAMA score ≥ 14

Treatment: 27 people, mean age 62.4 ± 6.1 years, 14 men

Control: 27 people, mean age 63.2 ± 5.7 years, 16 men

Exclusion: previous psychiatric disorder, antidepressants and "nerve block agents" in recent 3 months, severe cognitive impairment, aphasia, severe cardiac, hepatic and renal function impairment, allergy to paroxetine, severe suicidal behaviour

Interventions

Treatment: paroxetine 20 mg plus routine stroke treatment

Control: routine stroke treatment

Duration of treatment: 6 weeks

Duration of follow‐up: end of treatment to study end: 0

Outcomes

SSS

BI

HAMD

HAMA

TESS

Also reported GI upset and dizziness. They did not list any seizures in the list of AEs, so we are assuming no seizures in either groups

Unclear how side effects were collected

Funding source

Funded by a local scientific academic fund

Notes

The authors mentioned using the SDS and the SAS for evaluation, but they did not report the results of SDS and SAS

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts, analysed according to treatment group

Selective reporting (reporting bias)

High risk

No protocol, the authors mentioned using the SDS and the SAS for evaluation but they did not report the results

Other bias

Low risk

Balanced baseline

He 2016

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: prevention

Participants

374 participants

Country: China

Setting: inpatient

At randomisation numbers allocated: N = 300

Experimental group 1: fluoxetine immediately after enrolment n = 100; comparator group 1: fluoxetine 7 days after enrolment n = 100; comparator group 2: no fluoxetine n = 100

% male: unclear

Age: experimental, unclear; comparator 1, unclear; comparator 2, unclear

Subtype of stroke: unclear

Severity of stroke NIHSS score at baseline: unclear

Experimental: unclear

Comparator 1: unclear

Comparator 2: unclear

Time from stroke onset: within 1 week after onset of cerebral infarction

Inclusion criteria

  • ICD‐10 diagnostic criteria for acute cerebral infarction

  • Age 18 to 80 years

  • First onset of stroke within 1 week

  • NIHSS score > 2

  • Stroke related impairment

  • Informed consent by patients or legal representative

Exclusion criteria

  • Coma

  • Haemorrhagic stroke

  • Previous neurological impairment

  • Use of antidepressants over previous 3 months

  • Use of benzodiapines over previous 2 weeks

  • Self‐harm, suicidal ideation or need for antidepressants

  • Abnormal liver enzymes or creatinine levels

  • Gastrointestinal disorders affect drug absorption seriously

  • Life‐threatening illness (e.g. malignancy)

  • Allergic

  • Mental health disorders

  • Pregnant or breast feeding

  • Allergic

  • Enrolled in another interventional clinical research trial within previous 3 months

  • Scheduled endovascular intervention

Withdrawal criteria

  • Unblinding

  • Serious adverse reactions e.g. anaphylactic shock

  • Need for immediate stroke‐related surgery

  • Complications

  • Antidepressant use

  • Self‐harm, suicidal intention, urgent need for antidepressants

  • Withdrawal from the study

Interventions

Experimental: 20 mg of fluoxetine a day for 90 days and conventional therapy

Comparator: conventional therapy

Outcomes

Primary outcome at days 15, 90 and 180

  • NIHSS score

Secondary outcome at days 90 and 180

  • BI score

Funding source

This study was funded by Science and Technology Department of Guangdong, China (grant number: 2011B031800130), Science and Technology Innovation Committee of Shenzhen, China (grant number: 201101020), and Health and Family Planning Committee of Shenzhen, China (grant number: 201501009). It was registered on the Chinese Clinical Trial Registry (number: ChiCTR‐TRC‐12002078)

Notes

Dates study conducted: unclear. Either from June 2011 to December 2012 (ChiCTR‐TRC‐12002078) or from December 2015 to June 2016 (ChiCTR‐IPR ‐ 15007658)

Declarations of Interest: none reported

Trial registration detail (ChiCTR‐TRC‐12002078) does not match but rather matches ChiCTR‐IPR ‐ 15007658.

Baseline demographic and clinical characteristics for each group not presented, but rather the baseline demographic and clinical characteristics for those completing the trial (i.e. a subset of all those randomised at baseline) are presented

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Table of random numbers"

Allocation concealment (selection bias)

Unclear risk

Insufficient information on method of allocation concealment to judge high or low

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of high or low

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The evaluator was banned from participation in the treatment or from querying of the randomisation data."

Incomplete outcome data (attrition bias)
All outcomes

High risk

For the primary outcome of NIHSS score at 15, 90 and 180 days there was 8/187 (4%) lost to follow‐up in the experimental group; 16/187 (15%) in the comparator group. Twice as many participants in the comparator group (16/187(9%)) compared to the fluoxetine group (8/187 (4%)) were lost to follow‐up. Attrition and exclusions were not fully reported

> 5% lost to follow‐up

Selective reporting (reporting bias)

High risk

The trial registration number/protocol does not match the study design presented, but rather matches ChiCTR‐IPR ‐ 15007658

Other bias

High risk

The baseline data presented in table 1: comparison of data at baseline between control group and the treatment group are not true baseline characteristics (i.e. at randomisation). The data presented in table 1 are the baseline characteristics of all those completing the trial which is a subgroup of all participants randomised. We cannot tell if there is whether there was any baseline imbalance in important demographic or clinical characteristics

Hu 2002

Study characteristics

Methods

Parallel design

Aim: to study effect of antidepressants on depressive symptoms and nervous function

Participants

Country: China

Setting: inpatient

Stroke criteria: all pathological stroke types, clinical diagnosis plus confirmation by imaging (though unclear whether a relevant lesion had to be visible), onset of stroke 0.5 to 2 months, no obvious aphasia

Depression: according to CCMD‐II‐R

Treatment: 42 people, mean age 61.4 ± 3.6 years, 32 men

Control: 30 people, mean age 60 ± 4.8 years, 23 men

Interventions

Treatment: fluoxetine 20 mg daily

Control: no other antidepressant

Duration of treatment: 8 weeks

Duration of follow‐up (end of treatment to study end): 0

Outcomes

HAMD

MESSS

However, these data were not usable, as they were reported as proportions above or below "decrement levels"

Reported side effects but unclear how this was done

None left the trial early

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Balanced baseline, no other obvious risks

Hu 2018

Study characteristics

Methods

Aim: to explore the effect of escitalopram on the neural function and cognitive function of patients with post‐stroke depression

RCT

Participants

82 stroke patients selected who were in hospital from October 2016 to October 2017. Met diagnostic criteria for stroke. Both haemorrhagic and ischaemic stroke included. Patients also met the diagnostic criteria of depression in Chinese classification of mental disorders; Patients did not take other antidepressants before; HAMD Score > 18

Exclusions: 1) patients with transient cerebral ischaemia, secondary cerebral ischaemia and cerebral infarction; 2) patients with haematological diseases, coagulation dysfunction, arteriovenous malformations; 3) patients with severe organ diseases, respiratory failure; 4) patients taking other antidepressants recently; 5) patients with severe depression and cognitive impairment; 6) patients allergic to the study drugs, and dropped out of the study

Interventions

Both groups were given usual medical care, the treatment group was given escitalopram oxalate tablets (H20103327, Shandong Jingwei Pharmaceutical Co., Ltd. 10mg*7 tablets) 10mg, once per day, for 12 weeks

Outcomes

HAMD score
NIHSS score
MMSE score
FIM score

Funding source

Scientific research program of Shandong Provincial Department of Health (Funding number: 2012ws1783)

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table method

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol available to check

Other bias

Unclear risk

Not described

Huang 2002

Study characteristics

Methods

Parallel design

Aim: efficacy and tolerance of fluoxetine in early post‐stroke depression

Analysis: according to treatment group

Participants

Country: China

Setting: inpatient

Stroke criteria: first ever stroke, with single unilateral lesion, clinical diagnosis with imaging consistent with stroke, both ischaemic and haemorrhagic, recruited 2 weeks after stroke onset

Depression criteria: CCMD II‐R depression diagnosis

Treatment: 40 people, age and gender not stated

Control: 40 people, age and gender not stated

Participants in the treatment and control groups were selected from a group of 168 first‐ever acute stroke patients with average age of 62 ± 8.1 years, 76 men

Interventions

Treatment: fluoxetine 20 mg daily

Control: placebo

Duration of treatment: 4 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

CSS

Did not report death

Unclear how AEs were reported: no obvious AEs were found, but they did not specifically report seizures

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not stated

Allocation concealment (selection bias)

Unclear risk

Method not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Placebo used, but unclear if identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts, analysed according to treatment group

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

No description of the differences between treatment and control group in baseline characteristics

Jia 2005

Study characteristics

Methods

Parallel design

Aim: to determine the effect of early intervention for post stroke depression on movement after 3 months of stroke

Participants

Country: China

Setting: inpatient

Inclusion: aged 40 to 75 years, all pathological types of stroke, clinical diagnosis plus confirmation by imaging (did not state whether a relevant lesion had to be present to make a diagnosis), able to give informed consent

Depression diagnosis: Zung SDS > 41 for screening for depression, HDRS for evaluation of the depression severity level

Treatment: 92 people randomised, 90 accepted allocation, mean age 55.6 ± 6.5 years, 60 men

Control: 92 people randomised, 90 accepted allocation, mean age 55.1 ± 6.8, 55 men

Excluded: organic psychiatric disorders such as Alzheimer's disease or degenerative disease, functional disorders such as schizophrenia and affective disorders

Interventions

Treatment: either fluoxetine or sertraline (given sertraline if also had anxiety) plus routine stroke care

Control: routine stroke care

Duration of treatment: 3 months

Duration of follow‐up: 3 years but the authors did not describe the extent of neurological function damage and HAMD scores in the third year

Outcomes

HAMD

Extent of neurological damage

Recurrent stroke

Death

Did not report AEs

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts: 6 in treatment group (2 refused allocation), 4 in control group (2 refused allocation)

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Balanced baseline

Kim 2017

Study characteristics

Methods

Multicentre

Study type: interventional (clinical trial)

Intervention model: parallel assignment

Primary purpose: prevention

Participants

478 participants

Country: South Korea

Setting: inpatient. at neurology departments in 17 university hospitals throughout South Korea

At randomisation number allocated: N = 478, escitalopram (n = 241); placebo (n = 237)

% male at baseline: unclear

Age at baseline: unclear

Subtype of stroke at baseline: unclear

Severity of stroke at baseline: unclear

Time since stroke onset: acute ischaemic stroke or intracerebral haemorrhage within the previous 21 days

Inclusion criteria

  • Age > 20 years

  • Patients with acute stroke (ischaemic stroke or cerebral haemorrhage) confirmed by neuroimaging within 21 days after stroke onset

  • Patients with haemorrhagic transformation of infarcted tissue will not be included, but if investigators judge the risk of bleeding is small (i.e. reduced amount of blood in follow‐up neuroimaging) those patients can be enrolled

  • Patients with mRS ≥ 2 on screening

  • Patients without definite history of depression

  • Patients who fulfil the following criteria in the K‐MADRS test: The combined score of the 9th question (pessimistic thoughts) and the 10th question (suicidal idea) ≤ 7 The score of the 10th question < 6

  • Patients without serious communication problem

  • Consent

Exclusion criteria

  • MRS 0 or 1 on screening

  • History of depression or have taken antidepressants

  • Diagnosis of bipolar disorder or other psychiatric disorders

  • Severe dementia or aphasia and unable to communicate

  • Taken migraine medication on screening or expected to take migraine medication frequently due to severe migraine

  • Suicidal ideation on screening test or those who express their wish to be treated for depression

  • Depression requiring treatment diagnosed by physician

  • SSRI medication required for other reasons

  • Taken antiepileptic drugs on screening

  • History of traumatic brain injury, brain tumour, or other brain disease (except stroke) within 30 days prior to screening

  • Uncommon causes of stroke (e.g. subarachnoid haemorrhage, venous thrombosis, arteriovenous malformation, or Moyamoya disease)

  • Bleeding diathesis, haemophilia, or thrombocytopenia

  • Severe concomitant illness (e.g. liver disease, renal disease, malignancy)

  • Patients with abnormal blood tests, renal insufficiency, heart failure

  • Pregnant or breastfeeding

  • Participating in another clinical (interventional) trial

Withdrawal criteria: not stated

Interventions

Experimental: escitalopram: first week 5 mg, 2nd week ~ 12 week: 10 mg

Comparator: "sugar pill". First week 5 mg, 2nd week ~ 12 week: 10 mg

Outcomes

Primary outcomes collected at 3 months

  • Occurrence rate of depression (MADRS score ≥ 16)

Secondary outcomes

  • Prevention of depression at 3 months

  • Prevention of emotional incontinence (modified Kim's criteria) at 3 and 6 months

  • Prevention of anger proneness (modified Spielberger trait anger scale) at 3 and 6 months

  • Recovery of neurologic dysfunction (NIHSS, mRS, BI, motor function test from Hemispheric Stroke Scale at 3 months)

  • Improvement of cognitive function (MoCA) at 3 and 6 months

  • Improvement of quality of life (Stroke Specific Quality of Life scale) at 3 and 6 months

  • Improvement of caregiver burden (Sense of Competence Questionnaire scores) at 3 and 6 months

Funding source

Dong‐A Pharmaceutical Company, grants from the Ministry for Health, Welfare, and Family Affairs, South Korea

Notes

NCT01278498

Baseline demographic and clinical characteristics for each group not presented, but rather the baseline demographic and clinical characteristics for those completing the trial (i.e., a subset of all those randomised at baseline) are presented

Dates study conducted: January 2011 to December 2015

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Eligible patients were enrolled by investigators at each centre, and randomly assigned in a 1:1 ratio using a web‐based system to the escitalopram group or the placebo group after being assigned a subject number. Randomisation was done with random permuted blocks of sizes four to six, and was stratified by centre. The placebo was identical in appearance to escitalopram"

Allocation concealment (selection bias)

Low risk

Quote: "Eligible patients were enrolled by investigators at each centre, and randomly assigned in a 1:1 ratio using a web‐based system to the escitalopram group or the placebo group after being assigned a subject number. Randomisation was done with random permuted blocks of sizes four to six, and was stratified by centre"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The placebo was identical in appearance to escitalopram"

Quote: "The individual treatment code was stored separately by the main medical statistician (E‐JL) and two designated statisticians. All investigators including interviewers and assessors of the outcome, participants, and care providers were masked to treatment assignment throughout the study. The code could be unblinded only with the approval of the steering committee."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "All investigators including interviewers and assessors of the outcome, participants, and care providers were masked to treatment assignment throughout the study. The code could be unblinded only with the approval of the steering committee."

Incomplete outcome data (attrition bias)
All outcomes

High risk

The following participants were excluded from the 'full analysis set' post‐randomisation from both escitalopram group and placebo group:

  • did not take at least 1 dose of study medication (escitalopram = 4, placebo = 6)

  • did not undergo at least 1 assessment of the primary endpoint (escitalopram = 27, placebo = 36)

Reasons for attrition were reported (withdrew consent, violated protocol, considered for treatment for depression, death). Numbers were similar in both groups

At 12 weeks, escitalopram group 67/241 (28%) attrition and placebo 73/237(31%) attrition

Attrition much greater than 5%

It is not clear how missing data were imputed for the intention‐to‐treat analysis; Quote: "we used latest available records for analysis."

Selective reporting (reporting bias)

Low risk

The study protocol is available and all the study's prespecified (primary outcomes and secondary outcomes) that are of interest in the review have been reported in the prespecified way.

Other bias

High risk

The baseline data presented in table 1: comparison of data at baseline between control group and the treatment group are not true baseline characteristics (i.e. at randomisation). The data presented in table 1 are the baseline characteristics of all those completing the trial which is a subgroup of all participants randomised. We cannot tell if there is whether there was any baseline imbalance in important demographic or clinical characteristics

Kong 2007

Study characteristics

Methods

Parallel

Aim: to study whether fluoxetine could prevent post‐stroke depression and improve neurological function

Participants

Country: China

Setting: inpatient

Stroke: met diagnostic criteria of various cerebrovascular diseases formulated in the 4th National Cerebrovascular Disease conference and confirmed as stroke by CT or MRI, all hemiplegic, within 7 days of onset

HAMD score of no depression

Treatment: 48 people, mean age 64 ± 7 years, 60% men

Control: 42 people, mean age 62 ± 7 years, 57% men

Exclusion: major depression, current antidepressants, allergy to fluoxetine, substance abuse, bipolar disorder, schizophrenia, MMSE ≤ 23/30, substance abuse, obvious liver and renal deficit

Interventions

Treatment: fluoxetine 20 mg daily

Control: matching placebo capsules

Duration of treatment: 8 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

HAMD

BI

NIHSS

Reported "somatic side effects and hyponatraemia" but not death or other side effects

Authors state that "side effect rating was assessed at each visit" but unclear how this was done

Funding source

Source of funding not stated. Fluoxetine and placebo were supplied by Lilly Pharmaceutical Company

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated table of random digits

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical capsules, participants blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

States that researchers were blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

17/90 dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Balanced baseline

Lai 2006

Study characteristics

Methods

Parallel design

Analysis: analysed according to allocated treatment groups

Participants

Location China

Setting: inpatients

Treatment: 40 people

Control: 40 people

Total: mean age 60 ± 14 years, 43 men

Stroke criteria: unclear stroke types, clinical diagnosis plus brain imaging (though not clear that stroke lesion had to be present), acute stroke

Depression criteria: HAMD at least 7, or Zung SDS > 53, but no clear description about using which scale for inclusion criteria

Other entry criteria: none stated

Comparability of treatment groups: unclear

Exclusion criteria: unclear

Interventions

Treatment: paroxetine 20 mg daily

Control: placebo

Duration: treatment continued for 2 months

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

Depression: HAMD, Zung SDS (abnormal if the score is > 53)

Additional: Zung SAS (abnormal is the score is > 50)

Death

The author described that they recorded AEs but they did not report any AEs

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not stated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Placebo used, not stated if matching

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant dropped out

Selective reporting (reporting bias)

High risk

No protocol, stated that they would evaluate side effects but these were not reported

Other bias

Unclear risk

Demographic details at baseline not described

Li 2004a

Study characteristics

Methods

Parallel group

Aim: to study effects of fluoxetine on neurological impairment and post‐stroke depression

Participants

Location: China

Setting: inpatient

Stroke: inclusion: all pathological types, clinical diagnosis plus confirmation by imaging that relevant lesion visible, CSS 16 to 30

Depression criteria: HAMD scores ≥ 17 and DSM IV diagnostic criteria

Treatment: 33 people, mean age 60.33 years, 24 men

Control: 34 people, mean age 60.44 years, 23 men

Excluded severe psychiatric disorders, severe cardiac, pulmonary, hepatic and renal disease

Interventions

Treatment: fluoxetine 20 mg daily plus routine acute stroke care

Control: routine acute stroke care

Duration of treatment: 4 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

CSS

Depression incidence

Laboratory monitoring parameters

AEs (method of reporting not stated)

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer random numbers

Allocation concealment (selection bias)

Low risk

Opaque sealed envelopes

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

Balanced baseline

Li 2004b

Study characteristics

Methods

Parallel design

Aim: to treat depression

Participants

Country: China

Setting: inpatient

Stroke criteria: ischaemic stroke, clinical diagnosis plus imaging confirmation (though not clear that a relevant lesion had to be seen), stroke onset time ≤ 7 days

Depression criteria: HAMD score ≥ 8

Treatment: 37 people, age 48 to 87 years, 17 men

Control: 36 people, age 53 to 82 years, 15 men

Exclusion: previous depression or psychiatric interview, dementia (according to MMSE scores), aphasia, severe cardiac, pulmonary, hepatic, renal function impairment, consciousness disturbance

Interventions

Treatment: fluoxetine 20 mg daily plus usual stroke care

Control: usual stroke care

Duration: 8 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

CSS (cannot use as reported as a categorical variable)

MMSE (reported as a dichotomous variable)

BI (reported as a dichotomous variable)

Data for continuous variables not provided

Death reported

Side effects in treatment group only reported, not control group. Method of reporting side effects not stated

Funding source

Source of funding not stated

Notes

Note that the sum of numbers in each category of HAMD at 8 weeks in the control group adds up to 30, not 32

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Method not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 6 in treatment and 4 in control group. Total dropouts = 10/73 (14%)

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline balanced

Li 2005

Study characteristics

Methods

Parallel design

Improvement of post‐stroke depression and augmentation of rehabilitation

Participants

Country: China

Setting: inpatient

Stroke criteria: all stroke, clinical diagnosis plus confirmation on imaging (though not clear whether a relevant lesion had to be present)

Depression according to CCMD‐II‐R

Treatment: 74 participants

Control: 74 participants

Participaients in the treatment and control groups were selected from a group of 368 stroke patients with an average age of 57 ± 11.8 years, age range 33 to 84 years, 240 men

Excluded: previous psychiatric disorders, severe dementia, aphasia, consciousness disturbance

Interventions

Treatment: paroxetine 20 mg daily plus routine stroke treatment

Control: routine stroke treatment

Duration of treatment: 4 weeks

Duration of follow‐up (end of treatment to study end): 0

Outcomes

HAMD

SSS

Deaths

Side effects not recorded

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation method not described

Allocation concealment (selection bias)

Unclear risk

No description

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated whether blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analysed according to allocated treatment group, no participant dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

No description of differences between treatment and control group

Li 2006

Study characteristics

Methods

Parallel group

Participants

All pathological types of stroke, CT or MRI needed for diagnosis

Inclusion criteria: depression diagnosed by Chinese Classification of Mental Disorders 3 and HAMD ≥ 18, no previous organic brain disorder, and no previous psychiatric history, clear consciousness, no comprehension problems, normal language, first acute stroke, first episode of depression

Treatment: 52 people, mean ± SD age 61.12 ± 10.25, 32 men

Control: 53 people, mean ± SD age 60.89 ± 9.12, 35 men

Interventions

Treatment: citalopram 20 mg daily plus usual care

Control: usual care

Duration of treatment: 12 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

HDRS (also known as HAMD)

BI

CSS

MMSE

Side effects reported according to the participant's complaints and observation, no description of who recorded AEs; and reported only for the treatment group

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

No description

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

High risk

2 dropouts in treatment group, 4 in control group. 1 in treatment group died, and 2 in the control group died (i.e. > 5%)

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

Baseline balanced

Li 2007

Study characteristics

Methods

Aim: to investigate the efficacy and safety of paroxetine in the treatment of post‐stroke depression. Patients randomly divided into two groups

Participants

Stroke diagnosis: 4th Congress of Chinese Cerebrovascular Diseases, with brain CT or MRI to confirm the diagnosis.

30 cases of ischemic stroke and 14 cases of hemorrhagic stroke in the treatment group, and 31 cases of ischemic stroke and 11 cases of hemorrhagic stroke in the control group.

Patients with prior depression, anxiety and schizophrenia were excluded

HAMD Score ≥ 18

Interventions

Both groups were given usual medical care, the treatment group was give paroxetine tablets (Zhejiang Huahai Pharmaceutical Co., Ltd.,), 20mg, once per day orally in the morning for 8 weeks; the control group was given the placebo, dose not reported, once per day orally in the morning for 8 weeks

Outcomes

Depression score at treatment of 2‐week, 4‐week and 8‐week

Neurological deficiency score at treatment of 2‐week, 4‐week and 8‐week

Funding source

Unclear

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information provided

Selective reporting (reporting bias)

Unclear risk

No information provided

Other bias

Unclear risk

No information provided

Li 2008

Study characteristics

Methods

Parallel trial, 3 (fluoxetine versus "free and easy wandering" versus placebo), we are using the fluoxetine versus placebo comparison in this review

Participants

Country: China

Setting: unclear

Stroke criteria: by neuroimaging, ischaemic or PICH

Depression diagnosis: "each patient was evaluated by a psychiatrist", HAMD > 20 included

Fluoxetine group: 60 people, mean age 69.2 ± 3.5 years, men 41.6%

Control: 30 people, mean age 67.8 ± 3.9 years, men 56.7%

Excluded psychiatric illness other than depression, antidepressants within previous 2 weeks, MMSE < 23, severe aphasia

Interventions

Treatment: fluoxetine 20 to 40 mg daily

Control: placebo

Duration of treatment: 8 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

BI

Description of why participants left the trial early

AEs (reported by participant or observed/elicited by physician at each visit)

Funding source

Funded by the Natural Science Foundation of Shandong Province, People's Republic of China. None of authors had financial ties with the companies producing the medications in this study

Notes

Note twice as many in fluoxetine as in control group

Study conducted between March 2006 to September 2007. None of the authors or departments involved in the study had financial ties with the companies producing the medications used in this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Paper states blinded, used placebo (though unclear if matching, thus unclear (had a matching placebo been used then it would have been low)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

4/90 dropped out (< 5%)

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

Balanced baseline

Li 2017

Study characteristics

Methods

Aim: to investigate the effect of escitalopram oxalate in the early treatment of post‐stroke depression, cognition and neurological function

Participants

The diagnosis of ischaemic stroke was based on the Chinese guidelines for the diagnosis and treatment of acute ischaemic stroke (2010), and the diagnosis of haemorrhagic stroke was based on the Chinese diagnostic criteria for adult spontaneous cerebral haemorrhage (2010). Patient met the Chinese diagnostic criteria for mental disorders and depression, with HAMD ≥ 17 points. MMSE was used to screen the patients with post‐stroke cognitive impairment

The patients with unstable vital signs, severe cognitive impairment, severe depression, severe aphasia and unconsciousness were excluded

Interventions

Both groups received routine medicine and rehabilitation exercise. The treatment group was additionally treated with escitalopram oxalate tablets (Lexpro, produced by Lingbei pharmaceutical factory of Denmark, Xi'an Janssen Pharmaceutical sub package), 10 mg orally after breakfast every day for 24 weeks; the control group was additionally treated with placebo, 1 tablet each time, orally after breakfast for 24 weeks

Outcomes

HAMD, MMSE, NIHSS and FIM were used to evaluate depression, cognitive function, neurological deficit and the patients' ability to live independently respectively, by 2 psychiatrists and 2 neurologists who were uninformed of the treatment. Above scales were respectively before treatment, 4 weeks and 24 weeks after treatment

Funding source

Unclear

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information

Allocation concealment (selection bias)

Unclear risk

No information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Outcome assessed by 2 neurologist and 2 pyschiatrists unaware of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No information

Other bias

Unclear risk

No information

Liu 2006

Study characteristics

Methods

Parallel design

Aim: to study effect of citalopram on post‐stroke depression and neurological functional rehabilitation

Participants

Country: China

Setting: inpatient

Stroke criteria: stroke during "recovery phase" at 6 to 9 months, NIHSS score ≥ 13, HAMD score ≥ 17

60 people randomised, of whom 38 were men, mean age 60.7 ± 8.6 years. Demographics for treatment and control groups were not provided

Treatment: 30 people, age and gender not stated

Control: 30 people, age and gender not stated

Exclusion criteria: previous psychiatric disorder, dementia, aphasia, consciousness disturbance

Interventions

Treatment: citalopram 20 mg daily plus routine stroke care

Control: routine stroke care

Duration of treatment: 6 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

NIHSS

BI

Death

Funding source

Source of funding not stated

Notes

AEs not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline balance reported by authors

Marquez Romero 2013

Study characteristics

Methods

Multicentre

Study type: interventional (clinical trial)

Primary purpose: supportive care

Participants

32 participants

Country: Mexico

Setting: inpatient

At randomisation number allocated: N = 32: fluoxetine (n = 15); placebo (n = 17)

% men: 50%

Age: mean age 55.1 ± 12.2

Subtype of stroke: not available

Severity of stroke: NIHSS, Median (IQR): fluoxetine (12 (5)); placebo (14 (5))

Time since stroke onset: within 10 days

Inclusion criteria

  • Age > 18 years

  • Patients who had an acute intracerebral haemorrhage within the past 10 days causing hemiparesis or hemiplegia

  • FMMS scores of ≤ 55

  • Written informed consent

Exclusion criteria

  • NIHSS score > 20

  • Premorbid disability, evidenced by residual motor deficit from a previous stroke

  • Comprehension deficit or severe aphasia

  • Previous diagnosis of depression or one of the following: HADS score ≥ 11 points; taking antidepressant drugs 2 weeks before inclusion

  • Use of neuroleptic drugs or benzodiazepines 2 weeks before inclusion

  • Other life‐threatening illnesses

Withdrawal criteria

  • Detection of eligibility violations

  • Poor compliance (< 90%) or noncompliance

  • Use of any medication or treatment during the trial that could affect the study results

  • Occurrence of a serious adverse event:

    • participant has an acute reaction (allergy, shock) to the investigational product

    • participant develops depression, evidenced by HADS score ≥ 11 points at visit

    • participant withdraws consent or is uncooperative

Interventions

Experimental: fluoxetine 20 mg orally once daily for 90 days

Comparator: matching placebo orally once daily for 90 days

Outcomes

Primary outcome

  • FMMS score (baseline and 90 days): change from baseline in FMMS score at 90 days

Secondary outcomes

  • BI (baseline and 90 days): change from baseline in BI at 90 days

  • mRS (baseline and 90 days): change from baseline in mRS at 90 days

  • NIHSS (baseline and 90 days): change from baseline in NIHSS at 90 days

Funding source

Psicofarma S.A. de C.V.

Notes

NCT01737541

Terminated (study recruitment was suspended due to lack of funding)

Dates study conducted: November 2012 to August 2014

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A pharmaceutical laboratory (Psicofarma™ S.A. de C.V.) will be responsible for the manufacture and randomization of the investigational product, which will be achieved using a web‐based randomization program. This program will be set to assign participants equally to each site at a ratio of 1:1."

Allocation concealment (selection bias)

Low risk

Quote: "Each of the sites will be assigned 22 participants. The manufacturer will then deliver the pre‐randomized bottles containing the investigational product to each recruiting center. Study subjects who satisfy the eligibility criteria at each recruiting center will receive the investigational product corresponding to a consecutive number assigned according to their entrance to the study."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Fluoxetine and placebo tablets will be identical in form, color, odor and packaging."

"Both the investigator and the subject will be blinded to the assignment of the study drugs. The manufacturer of the tablets will label the investigational drugs by the randomization code number. The labeled experimental products will be provided to the recruiting centers by the manufacturer. An envelope containing all randomization codes will be delivered to the principal investigator and will be kept sealed until the conclusion of the trial."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and unlikely that blinding could have been broken

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Aimed to recruit 44 per group (total of 88) 35 in each group + 20% to allow for predicted 20% loss to follow‐up

Actual enrolment N = 32. Quote: "Two patients (one in each group) did not take any medication returning the unopened bottles at visit 1 and had to be excluded from analysis.". We judged that this is unlikely to have influenced bias, as there is one missing from each group and this represents a similar proportion in each group

Comment: report includes data from 30 participants (14 participants in the fluoxetine group and 16 in the placebo group)

Selective reporting (reporting bias)

Low risk

The study protocol is available and all of the study's prespecified (primary and secondary) outcomes that are of interest in the review have been reported

Other bias

Low risk

The study appears to be free of other sources of bias

Meara 1998

Study characteristics

Methods

Parallel design
Analysis: unclear

Participants

Location: Wales, UK

Setting: inpatient

Treatment: unclear

Control: unclear

Stroke criteria: ischaemic stroke > 11 weeks prior to randomisation

Depression criteria: GDS (15‐item) score > 4

Other entry criteria: not stated

Exclusion criteria: moderate to severe dementia, severe aphasia, communication difficulties, poorly controlled epilepsy

Interventions

Treatment: sertraline 50 mg daily, dose escalation to 100 mg for non‐responders at 2 weeks

Control: matched placebo

Duration: treatment continued for 6 weeks

Outcomes

Depression: change in scores from baseline to end of treatment on GDS

Unable to use GDS, BI, MMSE, FAI, FAST

Leaving trial early

Death

AEs

Funding source

Source of funding not stated

Notes

Contacted author for more details but no response

We could not use the data in our meta‐analysis

Dates of study not stated. Conflicts not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind reported, those who were blind not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Double‐blind reported, those who were blind not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

Insufficient data to make a judgement

Other bias

Unclear risk

Insufficient data to make a judgement

Miao 2004

Study characteristics

Methods

Parallel group

9 not allocated (5 in treatment group refused allocation, 4 in the control group refused allocation)

Participants

Country: China

Setting: mixed inpatient and outpatient

All stroke pathological types, clinical diagnosis plus confirmation by imaging that a relevant lesion was visible, 2 to 8 months after stroke, clear consciousness, no comprehension problem, 1 lesion in 1 hemisphere, normal language comprehension

Mood: depression after stroke onset, HAMD score ≥ 20

Participants: 90 randomised, 34 in each group at treatment end

Treatment: 34 people, age 58.16 ± 8.49 years, 19 men

Control: 34 people, age 62.45 ± 8.24 years, 18 men

Exclusion criteria: other organic brain disorders and other aetiologies‐related depression

Interventions

Treatment: citalopram 20 mg daily plus usual stroke care

Control: usual stroke care

Duration of treatment: 6 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

HAMD

SDS

Efficacy

Death

AEs (only in the citalopram group)

Method of recording AEs was not stated

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Simple random sampling"

Comment: no further description given

Allocation concealment (selection bias)

Unclear risk

Allocation not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding described

Incomplete outcome data (attrition bias)
All outcomes

High risk

9 not allocated after randomisation, 13 dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline balanced

Murray 2005

Study characteristics

Methods

Parallel design

Analysis: ITT (last observation carried forward) and per‐protocol: death (2 control), no efficacy (16 treatment, 22 control), withdrawn owing to AE (8 treatment, 5 control), withdrew consent (1 control), all excluded from analysis

Participants

Location: Sweden

Setting: mixed

Treatment: 62 people, mean ± SD age 71 ± 10 years, 52% men

Control: 61 people, mean ± SD age 71 ± 10 years, 44% men

Stroke criteria: all subtypes, diagnosis by WHO criteria and CT (100%); stroke 3 to 367 days prior to randomisation (average time 128 days)

Depression criteria: psychiatric interview (DSM‐IV, major and minor) and MADRS > 9

Other entry criteria: > 17 years of age, stroke within the previous 12 months

Comparability of treatment groups: significant trend towards more left‐hemisphere lesion strokes in treatment group

Exclusion criteria: under 18 years of age, severely impaired communication, apparent difficulties adhering to study protocol, acute myocardial infarction, other psychiatric illnesses other than depression, significant risk of suicide, antidepressants during the month after randomisation, current use of psychotropic medication or opiate analgesic drugs

Participants with < 20% reduction in MADRS score at 6 weeks were excluded

Interventions

Treatment: sertraline 50 mg daily; possible dose escalation to 100 mg after 4 weeks

Control: matching placebo

Duration of treatment: 26 weeks

Duration of follow‐up: (treatment end to study end): 0

Outcomes

Depression: change in scores from baseline to end of treatment on MADRS

Additional: leaving the study early

Death

Unable to use: Scandinavian Supervision Stroke Scale, BI, Stroke Unit Mental Status, Examination social performance, treatment costs, mortality, relative's situation, neuropsychological performance, neurological recovery (data not presented)

AEs (selected data presented) using a modified version of the Udvalg for Kliniske Undersogelser side effect rating scale

Funding source

Funded by an unrestricted grant, study drug and placebo from Pfizer AG Sweden and grants from the AFA Insurances and Marianne and Marcus Wallenberg Foundation

Notes

Recruitment September 1998 to January 2001. Conflicts stated; some of the authors have received grants from pharmaceutical companies

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation

Allocation concealment (selection bias)

Low risk

Centralised randomisation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

States blinding and used matching placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

States blinding

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

123 enrolled. 69 completed. Last observation carried forward

Selective reporting (reporting bias)

High risk

No protocol, paper stated that ADL data and SSS data were collected, but these were not reported

Other bias

Unclear risk

Balanced baseline except that more participants had left hemisphere brain lesion in sertraline group than in placebo group (statistically significant)

NCT00177424

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: prevention

Participants

Number of participants: unclear

Country: USA

Setting: inpatient

At randomisation number allocated: unclear

% male: unclear

Age: unclear

Subtype of stroke: unclear

Severity of stroke: unclear

Time since stroke onset: unclear

Inclusion criteria

  • Age > 40 years old

  • Ischaemic stroke within 3 months of study entry

  • Admitted to a University of Pittsburgh Medical Center hospital for acute inpatient treatment or rehabilitation of stroke

  • English‐speaking

  • Women willing to use an effective form of birth control throughout the study

Exclusion criteria

  • Major depressive episode (DSM‐IV‐TR criteria)

  • History of any bipolar disorder

  • Psychotic or history of a psychotic disorder

  • Alcohol or substance abuse or dependence (DMS‐IV TR criteria) within 3 months of study entry

  • Current treatment with antidepressant medication for any reason (e.g. anxiety disorder, neuropathic pain)

  • Primary haemorrhagic stroke

  • Language impairment severe enough to prevent assessment

  • CNS disease other than prior stroke or psychiatric illness (e.g. head trauma, multiple sclerosis, HIV with CNS involvement)

  • Pulse < 50 or > 100 beats per minute

  • Significant hyponatraemia (Na < 130 meq)

  • Current hypothyroid state

  • Medically unstable including symptoms of delirium

  • History of sensitivity to sertraline

  • Pregnant or breastfeeding

Interventions

Experimental: sertraline 12.5 mg/d for 3 days, increased to 25 mg/d for 4 days, then 50 mg/day for 7 days, then increased to 75 mg/day. Target dose = 75 mg per day for the remainder of participation in the study

Comparator: matched placebo

Outcomes

Primary outcome collected at 12 months

  • Major depression at 12 months

Secondary outcomes collected at 12 months:

  • Severity of depressive symptoms post‐stroke as measured by the HDRS

  • Level of disability as measured by the FIM

Funding source

None stated

Notes

Terminated (recruitment goals could not be met). Last update 27 June 27 2014

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information to permit judgement

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgement

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to permit judgement

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement

Other bias

Unclear risk

Insufficient information to permit judgement

NCT01674868

Study characteristics

Methods

Study type: interventional (clinical trial)

Intervention model: parallel assignment

Primary purpose: treatment

Participants

0 participants (aimed to recruit 25 participants)

Country: USA

Setting: inpatient

At randomisation number allocated: 0

% male: not available

Age: not available

Subtype of stroke: not available

Severity of stroke: not available

Time since stroke onset: not available

Inclusion criteria

  • Ischaemic infarction within 15 days

  • Admission NIHSS item 5 score ≥ 2

  • Able to give informed consent, with surrogate consent acceptable

Exclusion criteria

  • Pre‐stroke mRS score equal or ≥ 3

  • Pregnant or lactating

  • Taking an SSRI on admission

  • Taking a medication likely to have adverse interaction with an SSRI

  • Unable to return for follow‐up testing days 90, 180

  • Concurrent medical condition likely to worsen patient's functional status over next 6 months

  • Unable to competently participate in testing for 45 minutes to 2 hours with rest breaks

  • for MRI substudy: contraindication to MRI

Interventions

Experimental: fluoxetine 20 mg daily for 90 days starting day 5 to10 after stroke

Comparator: placebo participants will take 1 placebo pill daily for 90 days

Outcomes

Primary outcome measure

  • FMMS (baseline to 90 days, baseline to 180 days)

Secondary outcome measures

  • Western Aphasia Battery (baseline to 90 days)

  • Behavioral Inattention Test (baseline to 90 days, baseline to 180 days)

  • FIM (baseline to discharge)

  • Fatigue Severity Scale (baseline to 90 days, baseline to 180 days)

  • BDI (baseline to 90 days, baseline to 180 days)

  • Western Aphasia Battery (baseline to 180 days)

  • mRS (baseline to 90 days, baseline to 180 days)

Funding source

Not stated

Notes

NCT01674868

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Dates study conducted: April 2013 to December 2015 (estimated completion date)

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Allocation concealment (selection bias)

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Selective reporting (reporting bias)

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

Other bias

Unclear risk

Withdrawn: unable to find patients meeting inclusion/exclusion criteria

NCT02737930

Study characteristics

Methods

RCT

Participants

Acute stroke

18 Years to 85 years

MRI‐confirmed acute ischaemic stroke resulting in an isolated homonymous visual field loss

17 enrolled up to August 2020. No results published

Interventions

Fluoxetine 20mg for 90 days or matching placebo

Outcomes

Improvement in size of visual field deficit (degrees) (primary outcome)

Secondary outcomes: improvement in size of visual field deficit (square degrees), Improvement in parametric mean deviation

Functional field score

Visual Function Questionnaire‐25 score

Patient Health Questionnaire‐9 score

mRS score

Post‐stroke changes in cortical visual representation as measured by functional MRI

Post‐stroke changes in retinal nerve fibre layer thickness

Funding source

Bogachan Sahin

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No published data

Allocation concealment (selection bias)

Unclear risk

No published data

Pan 2018

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

170 participants

Country: China

Setting: inpatient

At randomisation number allocated: 170, paroxetine (n = 85); usual care (n = 85)

% male: paroxetine (71.8); usual care (unclear)

Age: mean age paroxetine = 65.6 ± 7.56; placebo = unclear

Subtype of stroke: not stated.

Severity of stroke: NIHSS, Median (IQR): paroxetine 8 (6 – 10); usual care (unclear)

Time since stroke onset: within 1 week

Inclusion criteria

  • Age between 50 and 80 years old

  • Diagnostic criteria met (Fourth National Cerebrovascular Disease Conference) and confirmation by MRI

  • Ability to participate in assessments within 1 week of stroke onset

  • FMMS score of < 55 points

  • MoCA score of < 26 points

Exclusion criteria

  • NIHSS score > 20 points

  • Aphasia

  • History of pre‐stroke depression and taken antidepressants or benzodiazepines

  • HAMD score > 7 points

  • Receipt of thrombolytic therapy

  • Complications such as infection, bed sores, or heart failure that might affect rehabilitation

Withdrawal criteria: not stated

Interventions

Experimental: orally administrated paroxetine at dosages of 10 mg/day during week 1 and 20 mg/day thereafter, for a total treatment duration of 3 months

Comparator: usual care

Outcomes

Outcomes were collected at 15, 90 and 180 days

  • Movement assessed using FMMS

  • Cognitive impairment assessed using the MoCA

  • Depression assessed using HAMD

Funding source

No grant funding from any grant funding agency, commercial or not‐for‐profit organisations

Notes

There is no study protocol/trial register reference

Baseline sociodemographic and clinical characteristics are provided only for those who completed the study

The authors state that one of the inclusion criteria is MOCA score of < 26 points. In the Results section they state that there were "72 cases of cognitive impairment" (i.e. a MoCA score of < 26 points) in the comparator group and 82 in the experimental group at days 15, 90 and 180. This suggests that either that the inclusion criteria were not strictly adhered to or if 100% of participants had a MoCA score of < 26 points at baseline then 10/82 participants in the comparator group and 3/85 in the experimental group have improved on the MoCA between days 0 and 15

Dates study conducted: participants recruited between January 2012 and June 2014

Declarations of interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Random number table"

Allocation concealment (selection bias)

Unclear risk

Insufficient information to judge high or low

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to judge high or low

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "All scale evaluators were trained and tested by the main investigator and were blind to the group assignment."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All data available for all participants in the experimental group (n = 85/85) and data available for (n = 82/85) participants in the comparison group for the Fugl–Meyer Motor Scale and the HAMD score

For the MoCA (see 'Other bias' below)

< 5% overall loss to follow‐up

Selective reporting (reporting bias)

Unclear risk

There is no study protocol/trial register reference, so insufficient information to judge high or low

Other bias

High risk

The authors state that one of the inclusion criteria is Montreal Cognitive Assessment (MoCA) score of < 26 points. In the Results section they state that there were "72 cases of cognitive impairment" (i.e., a MoCA score of < 26 points) in the comparator group and 82 in the experimental group at days 15, 90 and 180. This suggests that either that the inclusion criteria were not strictly adhered to or, if 100% of participants had a MoCA score of < 26 points at baseline then 10/82 participants in the comparator group and 3/85 in the experimental group have improved on the MoCA between days 0 and 15. The results 'Comparison of MoCA scores' and table 3 suggests otherwise

Pariente 2001

Study characteristics

Methods

Prospective double‐blind cross‐over placebo‐controlled study of 8 people with pure motor hemiparesis

Participants

Lacunar ischaemic stroke, assessed by brain CT

Quote: "Early phase of recovery"

Interventions

Single dose of fluoxetine

Outcomes

fMRI (raw data provided)

Finger tapping (presented as a graph, no raw data)

NIHSS, motricity index, BI, trunk control test, Ashworth Scale, somatosensory scale (no data)

Funding source

Source of funding not stated

Notes

We could not use these data in our meta‐analyses. The authors reported that fluoxetine led to hyperactivation in the ipsi‐lesional (i.e. on the same side as the stroke lesion) primary motor cortex during a motor task; moreover, fluoxetine significantly improved motor skills of the affected side

Dates of recruitment not given. Conflicts not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation code kept at the centre and broken at the end of the study

Allocation concealment (selection bias)

Low risk

Randomisation code kept at the centre and broken at the end of the study

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, placebo given

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data on fMRI appears complete

Selective reporting (reporting bias)

Unclear risk

Data on clinical outcomes were not reported

Other bias

Low risk

Balanced baseline

Rasmussen 2003

Study characteristics

Methods

Parallel design

Analysis: ITT (last observation carried forward) and per‐protocol: details of those excluded from analyses (35 treatment, 35 control) unclear

Participants

Location: Denmark

Setting: unclear

Treatment: 70 people, mean ± SD age 72 ± 9, 50% men

Control: 67 people, mean ± SD age 68 ± 11, 51% men

Stroke criteria: ischaemic and PICH; diagnosis by clinical signs and symptoms; stroke 0 to 4 weeks prior to randomisation

Other entry criteria: not stated

Comparability of treatment groups: participants in treatment group older on average

Interventions

Treatment: sertraline 50 mg daily; at any time after 2 weeks dose could be increased in 50 mg increments up to 150 mg daily; average dose 62.9 mg daily

Control: matched placebo

Duration of treatment: 12 months

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS

Proportion scoring > 2 on the CGI or > 16 on the GDS at end of treatment

Additional: leaving the study early. Did not report death

Unable to use: HDRS, GDS, aphasia severity rating scale, European Stroke Scale, MMSE, Cambridge Cognitive Examination, SF‐36, BI (data not presented)

AEs (detailed data not presented) evaluated by using the Udvalg for Kliniske Undersogelser Side Effect Rating Scale

Did not report death

Funding source

Funding from Pfizer A/S, Gert Jorgensen legat and the Brain Cause. It is unclear whether the drug companies had input into the design and analysis of the study

Notes

Recruitment January 1996 to May 1998. Conflicts not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not stated

Allocation concealment (selection bias)

Unclear risk

Method not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

No data on patients who dropped out. Used ITT analysis and last observation carried forward

Selective reporting (reporting bias)

Low risk

Trial details published on www.strokecentre.org/trials

Other bias

Unclear risk

Those given sertraline were slightly older (by 4 years) but this is unlikely to introduce bias

There was no significant difference between groups

Razazian 2014

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

172 participants

Country: Iran (Islamic Republic of)

Setting: inpatient

At randomisation number allocated: fluoxetine n = 86; placebo n = 86

% male: unclear

Age: fluoxetine group = unclear; placebo = unclear

Subtype of stroke: not available

Severity of stroke: not available

Time since stroke onset: not available

Inclusion criteria

  • Middle cerebral artery stroke (documented with imaging)

  • Hemiplegia, monoplegia or paresis

  • No coma

  • Consent

  • Suitable for discharge

  • Not admitted to Intensive care unit

Exclusion criteria

  • Death from any cause during study

  • Irregular use of drugs

  • Irregular return for re‐examinations

  • Seizures

  • Severe diarrhoea, vomiting,

  • Severe insomnia

  • Metabolic disorder

  • History of psychiatric disorder or severe depression prior to stroke

  • SAH, lobar ICH, brain tumour or stroke in other vascular territories

  • Use of any MAOI, selegiline, cyproheptadine

Withdrawal criteria: not stated

Interventions

Experimental: fluoxetine, 20 mg once a day for 90 days

Comparator: placebo fluoxetine for 90 days

All participants received 30 sessions of routine physiotherapy during the rehabilitation period

Outcomes

Primary outcomes collected at day 45 and day 90

  • Motor deficit (BI)

  • Psychiatric disorder (HDRS)

Funding source

Kermanshah University of Medical Sciences

Notes

IRCT201312088323N7

Baseline demographic and clinical characteristics for each group not presented, but rather the baseline demographic and clinical characteristics for those completing the trial (i.e. a subset of all those randomised at baseline) are presented

Dates study conducted: participants recruited between June 2013 and September 2014

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "random permuted blocks"

Comment: insufficient information about the block randomisation to permit judgement

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgement of high or low

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "placebo that was identical to the active drug in appearance and packaging"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of high or low

Incomplete outcome data (attrition bias)
All outcomes

High risk

13% attrition at 90 days. 13% (n = 11/86) from the experimental group and 13% (n = 11/86) from the comparator group were excluded form the full set analysis at 90 days follow‐up. Reasons for attrition reported

Selective reporting (reporting bias)

Low risk

Protocol available and all the study's prespecified outcomes that are of interest to the review have been reported in a prespecified way

Other bias

High risk

The baseline data presented in table 1: patients demographic characteristics and risk factors and not true baseline characteristics (i.e. at randomisation). The data presented in table 1 are the characteristics of the full analysis set which is a subgroup of all participants randomised. We cannot tell if there is whether there was any baseline imbalance in important demographic or clinical characteristics

Restifo 2001

Study characteristics

Methods

Double‐blind study

Participants

10 participants with disabling hemiplegia owing to hemispheric ischaemic stroke in territory of left MCA

Interventions

Treatment: fluoxetine 20 mg daily for 3 months plus usual care (including Bobath rehabilitation)

Control: usual care including Bobath rehabilitation

Outcomes

Transmagnetic stimulation to establish motor reorganisation

The authors reported that fluoxetine might modulate the primary motor cortex reorganisation

Funding source

Source of funding not stated

Notes

Abstract only, full paper could not be found by our searches. Dates of study and conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Random allocation"; method not described

Allocation concealment (selection bias)

Unclear risk

Quote: "Random allocation"; method not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

A placebo was used, not clear if it was matching

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unclear from abstract

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear from abstract

Selective reporting (reporting bias)

Unclear risk

Unclear from abstract

Other bias

Unclear risk

Unclear from abstract

Robinson 2000a

Study characteristics

Methods

Parallel design

Comparison of fluoxetine, nortriptyline and placebo. We are using the fluoxetine and placebo data

Analysis: per protocol, number excluded from analyses varied

Data provided for depressed and non‐depressed separately. We are labelling the depressed group as Robinson 2000a (this trial), and the non‐depressed group as Robinson 2000b

Participants

Location: USA and Argentina

Setting: mixed

Treatment: 23 people with depression, mean ± SD age 65 ± 14 years; 17 men

Control: 17 people with depression, mean ± SD age 73 ± 10 years; 9 men

Stroke criteria: all subtypes, diagnosis by clinical signs and CT (100%), stroke within 6 months of recruitment, 18 to 85 years of age

Stroke on average 16 weeks (fluoxetine) and 6 weeks (placebo) prior to randomisation

Exclusion criteria: other significant medical illness, severe comprehension deficit, prior history of head injury, prior history of other brain disease (with the exception of stroke), participants on antidepressants (other than fluoxetine) were allowed to stop their antidepressant for a 2‐week washout period

Interventions

Treatment: fluoxetine 10 mg daily (3 weeks), 20 mg daily (3 weeks), 30 mg daily (3 weeks), 40 mg daily (3 weeks)

Control: matched placebo

Duration: treatment continued for 12 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS

Additional: MMSE, JHFI

Death

AEs (method of reporting these was not stated)

Funding source

Funded by NIMH grants and grants from the Raul Carrea Institute of Neurological Research and Fundacion Perez Companc. Eli Lilly and company supplied the fluoxetine and placebo

Notes

Note difference in time since stroke between treatment groups

Dates of recruitment not stated. Conflicts not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random‐number table

Allocation concealment (selection bias)

Low risk

Concealment held by independent person

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Per‐protocol and ITT analyses

Selective reporting (reporting bias)

Low risk

Protocol published www.strokecentre.org/trials

Other bias

Unclear risk

Imbalance in treatment groups for time since stroke and gender

Robinson 2000b

Study characteristics

Methods

Parallel design

Comparison of fluoxetine, nortriptyline and placebo. We are using the fluoxetine and placebo data

Analysis: per protocol, number excluded from analyses varies

Data provided for depressed and non‐depressed separately. We are labelling the depressed group as Robinson 2000a, and the non‐depressed group as Robinson 2000b (this trial)

Participants

Location: USA and Argentina

Setting: mixed

Treatment: 17 non‐depressed people, mean ± SD age 66 ± 13 years, 15 men

Control: 16 non‐depressed people, mean ± SD age 67 9 years, 12 men

Stroke criteria: all subtypes, diagnosis by clinical signs and CT (100%), stroke within 6 months of recruitment, aged 18 to 85 years of age

Stroke on average 8 weeks (treatment) and 5 weeks (control) prior to randomisation

Comparability of treatment groups: unclear

Exclusion criteria: other significant medical illness, severe comprehension deficit, prior history of head injury, prior history of other brain disease (with the exception of stroke), participants on antidepressants (other than fluoxetine) were allowed to stop their antidepressant for a 2‐week washout period

Interventions

Treatment: fluoxetine 10 mg daily (3 weeks), 20 mg daily (3 weeks), 30 mg daily (3 weeks), 40 mg daily (3 weeks)

Control: matched placebo

Duration: treatment continued for 12 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

Depression: change in scores from baseline to end of treatment on HDRS

Additional: MMSE, JHFI

Death

AEs (method of reporting these was not stated)

Funding source

Funded by NIMH grants and grants from the Raul Carrea Institute of Neurological Research and Fundacion Perez Companc. Eli Lilly and company supplied the fluoxetine and placebo

Notes

Note difference in time since stroke between groups

Dates of recruitment not stated. Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random‐number table

Allocation concealment (selection bias)

Low risk

Concealment held by independent person

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT and per‐protocol

Selective reporting (reporting bias)

Low risk

Trial on www.strokecentre.org/trials

Other bias

Unclear risk

Note imbalance in time since stroke and in gender

Robinson 2008

Study characteristics

Methods

Parallel group, 3‐arm (escitalopram, placebo, problem‐solving therapy group). We are using the escitalopram versus placebo arm in this review

Analysis: ITT

Participants

Country: USA

Setting: mixed: neurology department and newspaper advertisements

Stroke criteria: ischaemic or haemorrhagic stroke not because of complications of intracranial aneurysm or intracranial vascular malformation; within 3 months of index stroke

Mood: excluded if DSM IV for major or minor depression or HAMD > 17

Treatment (escitalopram): 59 people, mean ± SD age 61.2 ± 13.7, 38 men

Control (matched placebo): 58 people, mean ± SD age 63.9 ± 11.1, 37 men

Exclusion: acute coronary syndrome, neurodegenerative disorders, DSM IV criteria for alcohol or substance abuse

Interventions

Treatment: escitalopram 5 mg to 10 mg (depending on age ‐ lower dose given to > 65 years old)

Control: matched placebo

Duration of treatment: 12 months

Duration of follow‐up (treatment end to study end): 0

Outcomes

Diagnosis of depression

HAMD (dichotomised)

FIM (though no raw data provided in the paper for meta‐analysis)

Social functioning examination

Repeatable Battery for Neuropyschological Status

The Iowa subset provided detailed information about cognition

Participants, family members and primary care physicians were asked about AEs at 3 monthly intervals or sooner if an individual reported an AE using a standardised checklist

Funding source

The initial report states that "This work was supported solely by National Institute of Mental Health Grant RO1MH‐65134. All the study medications were purchased using NIMH grant funds." In a subsequent letter to the Journal, the authors disclosed honoraria and expenses from pharmaceutical companies, and that 1 of the authors owned Pfizer stock. However, the authors stated that the design and analysis of any of the expenses of the study were supported by monies, materials or any intellectual input from Forest Laboratories

Notes

The escitalopram group had significantly more diabetes than the placebo group

Financial disclosures: see above

Recruitment: 9 July 2003 to 1 October 2007

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised blocks of 3, 6, and 9

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Outcome assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analyses, all participants used in analysis

Dropouts: 5 in placebo and 7 drop‐outs in escitalopram

Selective reporting (reporting bias)

Low risk

All specified outcome data reported. Trial published on www.strokecentre.org/trials

Other bias

Unclear risk

There was more diabetes in the escitalopram group than placebo group

Savadi Oskouie 2012

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

144 participants

Country: Islamic Republic of Iran

Setting: inpatient

At randomisation number allocated: N = 144; citalopram (n = 72); placebo (n = 72)

% male at baseline: citalopram n = unclear; placebo n = unclear

Age at baseline: citalopram (n = unclear); placebo (n = unclear)

Subtype of stroke at baseline: unclear

Severity of stroke at baseline: unclear

Time since stroke onset: within 7 days

Inclusion criteria

  • Acute ischaemic stroke

  • No previous use of citalopram or other antidepressants in the month prior to stroke onset

  • Pre‐stroke NIHSS < 20

  • No depression MADRS > 18

Exclusion criteria

  • Request of patients to leave the study

  • Previous chronic disease likely to interfere with assessment of effects of citalopram including: chronic infections, liver or kidney failure, cancer

  • Previous stroke‐related disability

  • Pregnancy or breastfeeding or any conditions that makes follow‐up impossible

  • Severe loss of consciousness

  • Thrombolytic therapy

  • Endarterectomy

  • Depression (MADRS > 18)

Withdrawal criteria: not stated

Interventions

Experimental: oral citalopram 20 mg once daily

Comparator: placebo

Outcomes

Primary outcome

  • 50% reduction in NIHSS score at 3 months compared to baseline

Secondary outcome

  • mRS score at 3 months

  • 50% reduction in NIHSS (motor) score at 3 months compared to baseline

  • 50% reduction in NIHSS (language) score at 3 months compared to baseline

  • Mortality

Funding source

Neurosciences Research Center (NSRC) of Tabriz University of Medical Sciences

Notes

IRCT201203192150N2

Baseline demographic and clinical characteristics for each group not presented, rather the baseline demographic and clinical characteristics for those completing the trial (i.e. a subset of all those randomised at baseline) are presented

Dates study conducted: May 2012 to January 2014

Declarations of interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A total of 144 patients were randomized through an allocation sequence based on 2 blocks with size of 72, generated with a computer random number generator."

Allocation concealment (selection bias)

Low risk

Quote: "Allocation was concealed using the sequentially numbered black envelopes."

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not explicitly stated that key study personnel and care providers were blinded, although implied by:

Quote: "The blinding code remained confidential until the end of the study."

Quote: "placebo of the same shape and full packaging during the first day after hospital admission."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not explicitly stated that outcome assessors were blinded, although perhaps implied by the fact:

Quote: "The blinding code remained confidential until the end of the study."

Incomplete outcome data (attrition bias)
All outcomes

High risk

Primary outcome data were available for 58 (81%) of the citalopram group and 50 (69%) of the placebo group. Reasons for attrition are reported but there are differences between groups: number of participants in the placebo group (n = 11) dead compared to the citalopram group (n = 4). 3 times the number of participants in the placebo group were depressed (n = 6) compared to the citalopram group (n = 2). Did not want to continue (placebo group (n = 5), citalopram group (n = 8)

Intention‐to‐treat analyses were carried out (suppl table) assuming that 1. those lost to follow‐up had a poor outcome (i.e. did not improve their NIHSS scores from baseline) and 2. those participants in the placebo group who did not want to continue had a good outcome

Overall loss of > 5%

Selective reporting (reporting bias)

Low risk

The study protocol is available and all the study's prespecified (primary outcomes and secondary outcomes) that are of interest in the review have been reported in the prespecified way

Other bias

High risk

The baseline data presented in table 1: comparison of demographic and baseline variables and not true baseline characteristics (i.e. at randomisation). The data presented in table 1 are the characteristics of the full analysis set at 3 months which is a subgroup of all participants randomised. We cannot tell if there is whether there were any baseline imbalance in important demographic or clinical characteristics. However, given that approximately 3 times the number of participants in the placebo group (n = 11) died compared to the citalopram (n = 4) and 3 times the number of participants in the placebo group were depressed (n = 6) compared to the citalopram group (n = 2), this suggests that there may have been important group differences in clinical characteristics at baseline

Shah 2016

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: supportive care

Participants

89 participants

Country: India

Setting: inpatient

At randomisation number allocated: N = 89: fluoxetine (n = 45); placebo (n = 44)

% male: unclear

Age: unclear

Subtype of stroke: unclear

Severity of stroke: unclear

Time since stroke onset: within 5 to 10 days

Inclusion criteria

  • Age 18 to 80 years old

  • Patients who had an acute ICH within the past 5 t0 10 days causing hemiparesis or hemiplegia

  • FMMS scores of 55 or less

Exclusion criteria

  • NIHSS score > 20

  • Diagnosis of depression MADRS score > 19 points

  • Premorbid disability, evidenced by residual motor deficit from a previous stroke

  • Use of neuroleptic drugs or benzodiazepines 4 weeks before inclusion

  • Other life‐threatening illnesses that would prevent follow‐up

  • Pregnancy

Withdrawal criteria: not stated

Interventions

Experimental: fluoxetine 20 mg orally once daily for 90 days

Comparator: matching placebo orally once daily for 90 days

Outcomes

Primary outcome

  • FMMS score (baseline and 90 days): change from baseline in FMMS score at 90 days

Funding source

Not stated

Notes

Baseline demographic and clinical characteristics for each group not presented, rather the baseline demographic and clinical characteristics for those completing the trial (i.e. a subset of all those randomised at baseline) are presented

Dates study conducted: January 2014 to January 2015

Declarations of Interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about the sequence generation process to permit judgement of high or low

Allocation concealment (selection bias)

Unclear risk

Insufficient information about the method of allocation concealment to permit judgement of high or low

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Patients, attendants, study staff and investigators were masked to treatment allocation." However, "matching was done on a 1:1 basis for age, sex, severity of stroke", which suggests that some key study personnel were not blinded and this non‐blinding is likely to introduce bias

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of high or low

Incomplete outcome data (attrition bias)
All outcomes

High risk

3/45 (7%) participants in the fluoxetine and 2/44 (5%) in the placebo group were lost to follow‐up. Reasons for attrition/exclusion not reported. 6% lost to follow‐up

Selective reporting (reporting bias)

Unclear risk

No study protocol available. Insufficient information to permit judgement of high or low

Other bias

High risk

The use of matching suggests a matched case control design rather than an RCT design

We cannot tell whether there was any baseline imbalance in important demographic or clinical characteristics

Song 2006

Study characteristics

Methods

Aim: to evaluate changes in depression and cognitive impairment in people with post‐stroke depression treated with fluoxetine

Parallel trial

Participants

Country: China

Setting: inpatient

Stroke diagnosed by clinical criteria and "proved on CT" (though not clear if lesion had to be visible)

Depression: diagnosed in accordance with the CCMD‐II‐R

Treatment: 41 people, mean age 51 ± 7 years, 25 men), time since stroke: 3.5 days

Control: 41 people, mean age 50 ± 8 years, 24 men), time since stroke: 3.7 days

Excluded: previous mental disorders, previous "neurological disorder", if other psychiatric drugs had been taken, these had to be stopped for 1 week before fluoxetine was administered

Interventions

Treatment: fluoxetine 20 mg daily

Control: placebo (although not stated whether this was identical to fluoxetine)

Duration of treatment: 6 weeks

Duration of follow‐up (treatment end to study end): 0

Side effects not reported

Outcomes

SDS

P300 (an event‐related potential)

Although the stated aim was to assess cognitive impairment, it is not clear how this was measured

Funding source

Source of funding not stated

Notes

Recruitment: December 1999 to June 2003. Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Placebo, but not clear whether identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

Unclear

Other bias

Unclear risk

Balanced baseline

Wang 2003

Study characteristics

Methods

Parallel design

3‐arm trial: routine care, fluoxetine plus routine care, amitriptyline plus routine care. We are using the routine care and fluoxetine plus routine care in this analysis

Aim: to observe effects of antidepressant therapy on post‐stroke and neurological rehabilitation in the elderly

Participants

Country: China

Setting: inpatient

Stroke criteria: ischaemic stroke, clinical diagnosis plus confirmation by imaging (although not clear whether a stroke lesion had to be present)

Depression diagnosed according to CCMD‐II‐R diagnostic criteria, HAMD ≥ 18

Treatment: 64 people, mean age 75.6 ± 19.7 years, 39 men

Control: 56 people, mean age 74.9 ± 20.8 years, 29 men

Excluded: psychiatric disorder history, severe cardiac, pulmonary, hepatic and renal diseases

Interventions

Treatment: fluoxetine 20 mg to 80 mg daily (start at 20 mg/day, increase dosage at 3 weeks if poor therapeutic effect and no AE), plus usual stroke care

Control: usual stroke care

Duration of treatment: 12 to 24 weeks

Duration of follow‐up (treatment end to study end): 6 to 9 months

Outcomes

HAMD

Neurological function impairment score

BI

AEs not recorded

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

13 dropped out of fluoxetine group, and 9 dropped out of control group

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Baseline appeared balanced but no statistical comparison between groups

Wang 2009

Study characteristics

Methods

Aim: to investigate the efficacy of paroxetine in the treatment of post‐stroke depression

Parallel RCT design. Placebo‐same appearance as paroxetine

Participants

Diagnosis of stroke: all patients underwent CT or MRI examination, and the diagnosis was in accordance with the Chinese diagnostic criteria formulated by the Second National Conference of Cerebrovascular Disease in 1986;

Depression diagnosis: HAMD ≥ 18, SDS (self‐rating depression scale) ≥ 50

Excluded: patients with medical history of mental illness, aphasia, epilepsy, glaucoma and drug allergy before stroke or used other antipsychotics or antidepressants in the past 2 weeks

Interventions

The treatment group was given paroxetine 20mg/day in the morning, which could be increased to 40 mg/day according to the condition, for a total of 2 months; the control group was given placebo with the same appearance. No other antipsychotics were used during the study period and benzodiazepines were given to patients with severe insomnia

Outcomes

HAMD

AEs

Funding source

Unclear

Notes

The data in Table 2 were different to the data in the Results section. Possibly the data of treatment group were mistakenly listed for the control group. Thus there is a high risk of incorrect data reporting

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information provided

Selective reporting (reporting bias)

Unclear risk

No information provided

Other bias

Unclear risk

No information provided

Wen 2006

Study characteristics

Methods

Parallel trial

Aim: to explore effects of prophylactic anti‐depression therapy on nerve functional rehabilitation after stroke

Analysis: according to treatment group

Participants

Country: China

Setting: inpatient

Stroke criteria: acute stroke of all pathological subtypes, clinical diagnosis plus confirmation by imaging (although not clear whether a stroke lesion had to be present)

Treatment: 42 people, mean age 56.8 years, men 19

Control: 42 people, mean age 57.2 years, men 16

Excluded those with primary psychiatric impairment and premorbid mood disorders, pre‐existing neurological disease causing confusion, severe systematic diseases and pulmonary, hepatic and renal failure

Interventions

Treatment: fluoxetine 20 mg daily plus routine stroke care

Control: routine stroke care

Duration of treatment: 8 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

HAMD

MESSS

AEs (method of obtaining data not stated)

Death

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analysed according to treatment group, no dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Balanced baseline

Wiart 2000

Study characteristics

Methods

Purpose: to treat early depression

Parallel design

Analysis: ITT (last observation carried forward), withdrawn owing to AE (1 treatment), protocol violation (1 treatment)

Participants

Location: France

Setting: unclear

Treatment: 16 people, mean ± SD age 66 ± 7 years, 65% men

Control: 15 people, mean ± SD age 66 ± 12 years, 40% men

Stroke criteria: ischaemic stroke and PICH, diagnosis by clinical signs and CT (100%); stroke on average 47 ± 22 days (treatment group) and 48 ± 20 days (control group)

Depression criteria: psychiatric interview (ICD‐10 criteria) and MADRS score > 19

Other entry criteria: all antidepressant or neuroleptic drugs stopped 10 days prior to enrolment

Comparability of treatment groups: balanced

Exclusion criteria: severe psychiatric problems which required hospitalisation, severe aphasia, previous stroke, severe cognitive impairment, chronic alcoholism, chronic associated handicapping pathology, contraindication to fluoxetine

Interventions

Treatment: fluoxetine 20 mg daily

Control: matched placebo

Duration of treatment: 45 days

Duration of follow‐up (treatment end to study end): 0

Outcomes

Depression: change in scores from baseline to end of treatment of MADRS, 50% reduction in MADRS score

Additional: FIMs

MMSE

Motricity Index

Leaving the study early

Death

AEs ("evaluated qualitatively and quantitatively". Complete blood count, liver test and renal function test were carried out at each assessment visit)

Funding source

Lilly France Laboratory provided methodological and financial support

Notes

Dates of recruitment not stated. Conflicts of interest not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of randomisation not stated

Allocation concealment (selection bias)

Unclear risk

Method not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Identical white capsules" given

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Method of blinding not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Used last observation carried forward; 2/31 (both in fluoxetine group) dropped out. This is > 5%

Selective reporting (reporting bias)

Low risk

Trial published on www.strokecentre.org/trials. The primary outcome was reported

Other bias

Unclear risk

Baseline balanced

Xie 2005

Study characteristics

Methods

Aim: to study the effect of treatment with sertraline in elderly patients with post‐stroke depression

Parallel study

Participants

Country: China

Setting: unclear

Recruitedquote: "clinically stable stroke patients with post‐stroke depression"

No other inclusion and exclusion criteria given

Mood: Zung SDS score ≥ 40 or GDS score 5 to 10

Treatment: 65 people, mean age 69.8 years, 29 men

Control: 65 people, mean age 70.7 years, 27 men

Time since stroke: mean 87.8 days, range 48 to 142 days

Interventions

Treatment: sertraline 50 mg/day plus usual stroke care

Control: usual stroke care

Duration of treatment: 12 weeks

Duration of follow‐up: 0

Outcomes

Zung SDS, GDS, ADL score

AEs were not reported

Funding source

Local scientific academic fund funded the study

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

No clear description between treatment and control

Xu 2001

Study characteristics

Methods

Parallel

Aim: to study the effect of fluoxetine on depression in early recovery stage of cerebral infarction

Participants

Country: China

Setting: outpatient in rehabilitation clinic

Stroke: first acute cerebral infarction, no description of the diagnostic criteria and the need for imaging confirmation, excluded large cerebral infarction or lacunar infarction (clinical condition too severe or too mild); onset to recruitment time mean 30 days

Zung SDS ≥ 40

Treatment: 32 people

Control: 31 people (no details of participant characteristics)

Excluded if previous antidepressants

Interventions

Treatment: fluoxetine 20 mg daily plus usual stroke care

Control: usual stroke care

Duration of treatment: 8 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

Zung SDS

ADL (BI)

Neural function deficient

Death

AEs not reported

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

10/62 dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

No clear description of stroke criteria and imaging

Xu 2006

Study characteristics

Methods

Parallel group

Aim: to test whether early prophylactic antidepressant treatment by paroxetine has any beneficial influence on the rate of post‐stroke depression and rehabilitation

Participants

Country: China

Setting: inpatient

Stroke criteria: stroke onset time ≤ 3 days, age ≤ 75 years old, no previous psychiatric disorders, no obvious cognitive impairment or aphasia

Depression diagnosis was not mentioned as an inclusion criteria, so we assumed that patients did not have to have depression to enter the trial

Treatment: 32 people, mean age 65 ± 12 years, 17 men

Control: 32 people, mean age 63 ± 11 years, 16 men

Exclusion: no severe hepatic or renal impairment, DSM IV depression not stated as an inclusion, but none met criteria for depression initially

Interventions

Treatment: paroxetine 20 mg daily

Control: placebo

Duration of treatment: 12 weeks

Duration of follow‐up (treatment end to study end): 0

Outcomes

MESSS

ADL

Post‐stroke diagnosis incidence of depression according to DSM IV

AEs not recorded

Funding source

Study funded by local scientific academic fund

Notes

The number of participants in Table 1 (p187) were wrong (paroxetine/placebo: N = 32/32 should be N = 28/29)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Sequence numbers"

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Placebo used, but unclear if it was matched

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

7/64 (11%) participants dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

Baseline balance

Yang 2002

Study characteristics

Methods

Parallel group

Aim: to study effects of antidepressant in treatment of people with post‐stroke depression

Participants

Country: China

Setting: inpatients and outpatients

Stroke criteria: recovery phase of stroke (2 to 6 months after ischaemic stroke, and 1.5 to 6 months after haemorrhagic stroke). We included this in the 3 to 6 month group. Clinical diagnosis of stroke (not stated whether confirmation by imaging was needed)

Depression: HAMD > 7

Treatment: 64 people, mean age 64 ± 3 years, 40 men

Control: 57 people, mean age 63 ± 5 years, 32 men

Interventions

Treatment: paroxetine 20 mg daily plus stroke treatment and rehabilitation

Control: stroke treatment and rehabilitation

Duration of treatment: 4 months

Duration of follow‐up: 0

Outcomes

Death

They collected data on HAMD and CSS but did not report these data

ADL score: did not state which one, so not used

AEs not reported

Funding source

Source of funding not reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

11/121 (9%) dropouts

Selective reporting (reporting bias)

High risk

No protocol. The paper stated that ADL data and depression data were collected, but these data were not reported

Other bias

Low risk

No baseline differences between groups, no other obvious source of bias

Yang 2011

Study characteristics

Methods

Aim: to treat early post‐stroke depression

Participants

Country: China

Setting: inpatient

Stroke: all pathological types, clinical diagnosis plus confirmation of lesion on imaging, no previous psychiatric and psychological disorders, age < 75 years old, stroke onset time < 72 hours, NIHSS score: 4 to 19

Mood: HAMD ≥ 8

Treatment: 20 people, mean age 64 ± 8 years, 8 men

Control: 22 people, mean age 64 ± 10 years, 13 men

Note inconsistency between abstract (20 in treatment and 22 in control, but in tables of results, there are 22 in treatment and 20 in control). We have used the data from the abstract

Excluded: functional psychiatric disorder, functional depression, psychoactive substance and addictive substance induced psychiatric disorders, infectious disease, severe cognitive impairment to affect communication, severe aphasia to affect communication, severe cardiac, pulmonary, hepatic and renal function impairment, previous organic brain disease such as brain tumour, or symptomatic stroke, encephalitis

Interventions

Treatment: paroxetine 20 mg daily plus usual stroke care

Control: usual stroke care

Duration of treatment: at least 3 months

Duration of follow‐up: 0

Outcomes

HAMD score, IL‐1β and IL‐6 level

Death

AEs not reported

Funding source

Source of funding: local scientific academic fund

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Case sequence" randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Low risk

No difference in baseline

Ye 2004

Study characteristics

Methods

Aim: to investigate whether antidepressive therapy is needed for people with post‐stroke depression or not, and the effect of different antidepressive drugs on the rehabilitation of psychological and neurological function after stroke

3 groups: paroxetine, imipramine and control. We are using the paroxetine versus control arm in this review

Participants

Country: China

Setting: inpatient

Stroke: all pathological subtypes, clinical diagnosis plus confirmation by imaging (did not state whether a visible lesion was needed to make the diagnosis), no positive psychiatric disorders or family history, clear consciousness, no comprehension problem

Mood: inclusion criteria: HAMD score > 21, HAMA scale > 14

Treatment: 30 people, age 58.04 ± 8.28 years, 22 men

Control: 30 people, age 59.21 ± 9.52 years, 17 men

Exclusion criteria: severe cardiac, hepatic and renal diseases, multiple infarcts or haemorrhage

Interventions

Treatment: paroxetine 20 mg/day plus acute stroke routine care and rehabilitation

Control: acute stroke routine care plus rehabilitation

Duration of treatment: 12 weeks

Duration of follow‐up (end of treatment to end of study): 0

Outcomes

Chinese Neurological Impairment Scale, modified BI, HAMD, HAMA, Therapeutic Effect for Depression and Neurologic Function

Death, GI upset

Method of recording side effects not stated

Funding source

Source of funding not stated

Notes

Inconsistent description about the number of recruitment and randomisation between abstract (N = 90) and result part (N = 93) of the text. The number for final analysis is consistent in the text

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Used "number table", but unclear if this was a random number table

Allocation concealment (selection bias)

Low risk

The study designer was not involved in assessment and treatment, the assessors did not know the allocation

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The participants were blinded. Not clear if those delivering the treatment were blind, but no placebo used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Only 1 dropped out in paroxetine group

Selective reporting (reporting bias)

Unclear risk

No protocol

Other bias

Unclear risk

Different numbers reported to have been recruited and randomised, baseline similar

Zhao 2011

Study characteristics

Methods

Study type: interventional (clinical trial)

Primary purpose: treatment

Participants

Country: People's Republic of China

Setting: inpatient

At randomisation number allocated: N = 82: fluoxetine (n = 41); placebo (n = 41)

% male: 58.5

Age: mean age 65 ± 12

Subtype of stroke: Ischaemic stroke: 61/82 (74%); haemorrhagic stroke: 21/82 (26%)

Severity of stroke: MESSS: fluoxetine 23.2 ± 6.2 (n = 37); placebo 22.8 ± 5.8 (n = 34)

Time since stroke onset: within 10 days

Inclusion criteria

  • Consistent with the Diagnostic Criteria for Cerebrovascular Disease formulated by the Fourth National Conference of Chinese Medical Association in 1995, and prove with brain CT or MRI

  • Obvious aphasia and unable to communicate normally after language function evaluation

  • Age 75 years old or less

  • Without previous psychiatric illness

  • No severe cognitive impairment

Exclusion criteria: none

Withdrawal criteria: not stated

Interventions

Experimental:fluoxetine 20 mg daily for 12 weeks

Comparator: no fluoxetine

Outcomes

Outcomes collected at 2nd, 4th and 12 week of treatment and 12 weeks after the end of treatment

  • Severity of stroke (MESSS)

  • Performance in ADLs

Funding source

Not available

Notes

Dates study conducted: 2008 to 2010

Declarations of interest: none reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The participants were randomised into 2 groups (with fluoxetine or without fluoxetine) according to the sequence number and a block randomisation table

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgement of high or low

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of high or low

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of high or low

Incomplete outcome data (attrition bias)
All outcomes

High risk

Attrition rate of fluoxetine group vs control group was 4/41 (9.8%) vs 7/41 (17.1%)

> 5% loss to follow‐up

Selective reporting (reporting bias)

Unclear risk

No trial protocol available. Insufficient information to permit judgement of high or low

Other bias

Low risk

The study appears to be free from other sources of bias

Zhou 2008

Study characteristics

Methods

Aim: to study effect of early paroxetine on post‐stroke depression and rehabilitation

Parallel design

Analysis: according to treatment groups

Participants

Country: China

Setting: inpatient

Stroke criteria: all stroke, clinical diagnosis plus confirmation by imaging (though not clear if a relevant stroke lesion had to be visible), stroke onset time ≤ 7 days, no obvious cognitive impairment, no obvious aphasia

HAMD score < 8

Treatment: 36 people, mean age 63 ± 9.3 years, 16 men

Control: 40 people, mean age 61 ± 9.6 years, 19 men

Excluded: previous psychiatric disorders, severe hepatic and renal impairment, taking agents with obvious interaction with fluoxetine in recent 1 month

Interventions

Treatment: fluoxetine 20 mg daily plus acute stroke routine medication

Control: acute stroke routine medication

Duration of treatment: 8 weeks

Duration of follow‐up: 0

Outcomes

No raw data provided for any of the following outcomes: diagnosis of depression (CCMD‐3, HAMD, ADL, MESSS)

Reported no deaths in either group. Unclear how data on side effects were collected

Funding source

Source of funding not stated

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts, analysed according to allocated treatment group

Selective reporting (reporting bias)

High risk

No protocol, no raw data provided for several of the outcomes

Other bias

Low risk

Baseline similar

ADL: activities of daily living
AE: adverse event
ALT: Alanine aminotransferase test
BDI: Beck Depression Inventory
BI: Barthel Index
CCMD‐II‐R: Chinese Classification of Mental Disorders, second edition, revised
CCMD‐3: Chinese Classification of Mental Disorders‐3
CGI: Clinical Global Impressions Scale
CSS: Chinese Stroke Scale
CT: computerised tomography
CTIMP: Clinical Trial of an Investigational Medical Product
DSM: Diagnostic and Statistical Manual of Mental Disorders
EEG: electroencephalogram
eFGR: estimated glomerular filtration rate
FAI: Frenchay Activities Index
FAST: Frenchay Aphasia Screening Test
FIM: Functional Independence Measure
FMMS: Fugl‐Meyer Motor Scale
fMRI: functional magnetic resonance imaging
GDS: Geriatric Depression Scale
GI: gastrointestinal
HADS: Hospital Anxiety and Depression Scale
HAMA: Hamilton Anxiety scales
HAMD/HDRS: Hamilton Depression Rating Scale
HSS: Hemispheric Stroke Scale
ICD: International Classification of Diseases
ICH: intracerebral haemorrhage
IL: interleukin
ITT: intention‐to‐treat
IQR: interquartile range
JHFI: Johns Hopkins Functioning Inventory
LOCF: last‐observation‐carried‐forward
MADRS: Montgomery‐Åsberg Depression Rating Scale
MAOI: mono‐amino‐oxidase inhibitor
MCA: middle cerebral artery
MEP: motor evoked potentials
MESSS: Modified Edinburgh‐Scandinavian Stroke Scale
MHI‐5: Mental Health Inventory
MMSE: Mini‐Mental State Examination
MoCA: Montreal Cognitive Assessment
MRI: magnetic resonance imaging
mRS: modified Rankin score
NIHSS: National Institutes of Health Stroke Scale
PASE: Physical Activity Scale for the Elderly
PICH: primary intracerebral haemorrhage
RCT: randomised controlled trial
RS: Rankin score
SAH: subarachnoid haemorrhage
SAS: Zung Self‐rating Anxiety Scale
SD: standard deviation
SDS: Zung Self‐rating Depression Scale
SF‐36: Short Form‐36
SIS: Stroke Impact Scale
SSRI: selective serotonin reuptake inhibitors
SSS: Scandinavian Stroke Scale
TESS: Treatment Emergent Symptom Scale
TIA: transient ischaemic attack
VAS: visual analogue scale
WHO: World Health Organization

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

ACTRN12619000573156

Trial abandoned prior to initiating recruitment due to COVID‐19 pandemic

Andersen 1993

Cross‐over design: double‐blind placebo‐controlled cross‐over protocol as follows: 7 days initial baseline registration, 21 days citalopram or placebo (randomised), 7 days wash‐out, 7 days baseline registration, and cross‐over to second 21‐day treatment period

Andersen 2012

The trial never started

Anderson 2002

The trial never started

Anonymous 2012a

Unable to find publication after extensive searching

Anonymous 2012b

Unable to find publication after extensive searching

Berends 2009

Mean time from stroke onset to fluoxetine was 39.1 months

Bonin Pinto 2019

Participants were recruited within 2 years (not 1 year) of stroke

Chen 2019

Tandospirone + escitalopram (combination therapy) and escitalopram (monotherapy) in people with vascular depression

Choi Kwon 2008

Participants more than 1 year post‐stroke

Finkenzeller 2009

SSRI plus active intervention (psychotherapy) versus active treatment (psychotherapy) alone. This trial had been included in the original 2012 review but due to the potential interaction between the SSRI and psychotherapy we decided to exclude it in this update

Foster 2019

2 arms: 1 with SSRI plus exercise and 1 arm with placebo plus exercise

Gourab 2015

Time of stroke onset > 12 months

Graffagnino 2002

Previously listed in 'Studies awaiting classification' (Mead 2012). Unable to access any full publication and we received no response from the author. Given the insufficient information to assess eligibility and, owing to the length of time since the study abstract (2002) was published, we have now excluded this study. CRSREF: 3340767

Ji 2000

SSRI plus active intervention versus active treatment alone

Kitago 2020

Combined intervention

Li 2002

There is no random component in the sequence generation process

Liang 2003

There is no random component in the sequence generation process. This had been included in the 2012 review but on review of the methodology the review authors decided to exclude this for the update

Liu 2004

SSRI plus active intervention versus active treatment alone

Liu 2020

Wrong comparator

Mosarrezaii 2018

Qu0te: "Patients received numbered cards according to the order of hospitalization. The recipients of the cards with odd and even numbers constituted the case and control group, respectively." Allocation concealment procedure was inadequately concealed

NCT01963832

Study withdrawn (not funded)

Robinson 2011

Ineligible outcomes: prevention of generalised anxiety disorder

Sitzer 2002

Previously listed in 'Studies awaiting classification' (Mead 2012). Unable to access any full publication and we received no response from the author. Given the insufficient information to assess eligibility and, owing to the length of time since the study abstract (2002) was published, we have now excluded this study

Sun 2015

Mean time since onset 19.2 ± 3.5 months. No placebo or usual‐care control group (Prozac, acupuncture, and prozac plus acupuncture)

Vogel 2020

Open‐label single‐group study

Xu 2007

This had been included in the 2012 review but it compares fluoxetine plus wulung capsule versus wulung capsule alone. Wulung capsule is an active comparator so we have therefore excluded this trial for this update

Zhou 2003

There is no random component in the sequence generation process. This trial had been included in the 2012 version of the review but for this update we excluded it

SSRI: selective serotonin reuptake inhibitor

Characteristics of studies awaiting classification [ordered by study ID]

Guo 2016

Methods

Study type: interventional (clinical trial)

Actual enrolment: 300

Allocation: randomised

Intervention model: parallel assignment

Masking: single (outcomes assessor)

Primary purpose: prevention

Participants

Country: China

Setting: inpatient

At randomisation numbers allocated: N = 300

Experimental group 1: fluoxetine immediately after enrolment n = 100
Comparator group 1: fluoxetine 7 days after enrolment n = 100
Comparator group 2: no fluoxetine n = 100

% male: unclear

Age: Experimental, unclear; Comparator 1, unclear; Comparator 2, unclear

Subtype of stroke: unclear

Severity of stroke NIHSS score at baseline: unclear

Experimental: unclear

Comparator 1: unclear

Comparator 2: unclear

Time from stroke onset: within 1 week after onset of cerebral infarction

Inclusion criteria

  • ICD‐10 diagnostic criteria for acute cerebral infarction

  • Age 18 to 80 years

  • First onset of stroke within 1 week

  • NIHSS > 2

  • Stroke‐related impairment

  • Informed consent by participants or legal representative

Exclusion criteria

  • Coma

  • Haemorrhagic stroke

  • Previous neurological impairment

  • Use of antidepressants over previous 3 months

  • Use of benzodiazepines over previous 2 weeks

  • Self‐harm, suicidal ideation or need for antidepressants

  • Abnormal liver enzymes or creatinine levels

  • Gastrointestinal disorders affect drug absorption seriously

  • Life‐threatening illness (e.g. malignancy)

  • Allergic

  • Mental health disorders

  • Pregnant or breast feeding

  • Allergic

  • Enrolled in another interventional clinical research trial within previous 3 months

Withdrawal criteria

  • Unblinding

  • Serious adverse reactions e.g. anaphylactic shock

  • Need for immediate stroke‐related surgery

  • Complications

  • Antidepressant use

  • Self‐harm, suicidal intention, urgent need for antidepressants

  • Withdrawal from the study

Interventions

Experimental: 20 mg of fluoxetine per day for 90 days given immediately after enrolment and conventional therapy of cerebral infarction

Comparator 1: 20 mg of fluoxetine a day for 90 days given 7 days after enrolment and conventional therapy of cerebral infarction

Comparator 2: no fluoxetine and conventional therapy of cerebral infarction

Outcomes

Primary outcome at days 15, 90, and 180

  • NIHSS score

Secondary outcome at days 90 and 180

  • BI score

Notes

ChiCTR‐TRC‐15007658

[email protected]

Baseline demographic and clinical characteristics for each group not presented, but rather the baseline demographic and clinical characteristics for those completing the trial (i.e. a subset of all those randomised at baseline) are presented

He 2018

Methods

Study type: interventional (clinical trial)

Actual enrolment: 404

Allocation: randomised

Intervention model: parallel assignment

Masking: single (outcomes assessor)

Primary purpose: prevention

Participants

Country: China

Setting: inpatient

At randomisation numbers allocated: N = 404

Experimental group: fluoxetine n = 202

Comparator group n = 202

% male: 70.5%

Age: Experimental: 61.14 ± 10.48; Comparator 62.72 ± 11.86

Subtype of stroke: unclear

Severity of stroke NIHSS score at baseline:

Experimental: Median 6 (IQR 4, 8)

Comparator: Median 5 (IQR 3, 8)

Time since stroke onset: mean days, fluoxetine 4.28 ± 1.89; placebo 4.08 ± 2.15

Inclusion criteria

  • ICD‐10 diagnostic criteria for acute cerebral infarction

  • Age 18 to 80 years

  • within 1 week of stroke onset

  • Written informed consent by participants or legal representatives

Exclusion criteria

  • Coma

  • History of stroke

  • Pregnant or breast feeding

  • Self‐injury, suicidal intention or depression and need for antidepressants

  • History of peptic ulcer or gastritis

  • Life‐threatening illness (e.g. cardiac insufficiency, malignancy)

  • Use of antidepressants over previous 3 months

  • Use of benzodiazepines over previous 2 weeks

  • Allergic

  • Enrolled in another interventional clinical research trial within previous 3 months

Withdrawal criteria

  • Violation of randomisation or blinding rules during the follow‐up

  • Serious adverse reactions, such as anaphylactic shock

  • Serious infections or medical complications.

  • Antidepressants use

  • Self‐injury, suicidal intention or depression and need for antidepressants

  • Withdrawal from the study (participant or legal relatives)

Interventions

Experimental: 20 mg of fluoxetine a day for 90 days and conventional therapy

Comparator: conventional therapy

Outcomes

  • Recurrence rate of cerebral infarction within 3 years

  • Improvement of NIHSS, hypertension, diabetes, hyperlipids at day 90

Notes

ChiCTR‐TRC‐12002078

[email protected]

Jurcau 2016

Methods

Study type: interventional (clinical trial)

Actual enrolment: 89

Allocation: randomised

Intervention model: parallel assignment

Masking: unclear

Primary purpose: treatment

Participants

Country: Romania

Setting: inpatient

At randomisation numbers allocated: N = 89

Experimental group: escitalopram = 43

Comparator group: ?secondary preventive treatment = 46

% male: unclear

Age: unclear

Subtype of stroke: unclear

Severity of stroke: unclear

Time since stroke onset: unclear

Inclusion criteria: unclear

Exclusion criteria: unclear

withdrawal criteria: unclear

Interventions

Experimental: escitalopram 10 mg/day for 12 weeks

Comparator: ?secondary preventive treatment = 46

Outcomes

Outcomes collected at 3, 6 and 12 months post‐stroke

  • NIHSS

  • BI

  • MMSE

  • BDI

  • HAM‐D17

Notes

Does not appear to be a clinical trial register number

NCT00967408

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 200 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: quadruple (participant, care provider, investigator, outcomes assessor)

Primary purpose: treatment

Participants

Country: Italy

Setting: inpatient

Inclusion criteria

  • > 18 years

  • First ischaemic or haemorrhagic stroke

Exclusion criteria

  • Unstable medical conditions

  • Unable to understand study aims and procedures

  • Severe aphasia

  • Other progressive neurological disease

  • Previous or concomitant psychiatric illness

  • Not willing to participate

Interventions

Experimental: escitalopram and rehabilitation. Escitalopram given 5 mg once a day for the first week, 10 mg once a day from the second to fourth week, and 20 mg daily until the 6th month

Comparator: placebo and rehabilitation

Outcomes

Primary outcome collected at 2 and 6 months

  • FIM

Secondary outcomes collected at 2 and 6 months:

  • MMSE

  • Trunk Control Test

  • Canadian Stroke Scale

  • Motricity Index

  • Token test

  • The Bells Test

  • Stroop Test

  • Wisconsin Card Sorting test

  • Verbal Fluency

  • Raven's Matrices Test

  • Trail Making A‐B Test

  • Center for Epidemiological Studies Depression Scale

Notes

clinicaltrials.gov/ct2/show/NCT00967408

Contacted author Prof Cisari; response received; data being analysed

BDI: Beck Depression Inventory
BI: Bathel Index
FIM: Functional Independence Measure
HAM‐D17: Hamilton Depression Scale
ICD‐10: International Statistical Classification of Diseases, 10th revision
IQR: interquartile range
MMSE: Mini‐Mental State Examination
NIHSS: National Institutes of Health Stroke Scale

Characteristics of ongoing studies [ordered by study ID]

ChiCTR1800019467

Study name

The effect and mechanism of fluoxetine on the automatic regulation of cerebral blood flow for ischemic stroke

Methods

Study type: interventional (clinical trial)

Estimated enrolment:

Allocation: randomised

Intervention model: parallel assignment

Masking: unclear

Primary purpose: treatment

Participants

China

Inclusion criteria

  • Age 30 to 80 years

  • First‐time acute (in the past 72 hours) ischaemic stroke

  • Diagnosis of stroke confirmed by imaging

  • Consent

Exclusion criteria

  • Haemorrhagic stroke

  • Coma

  • Massive cerebral infarction

  • Poor coordination of transcranial doppler ultrasonography

  • Currently participating or have participated in other clinical trials within 3 months

Interventions

Experimental: 20 mg of fluoxetine daily for 90 days

Comparator: conventional therapy

Outcomes

Baseline, 30 days after treatment, 90 days after treatment, 180 days after treatment

Automatic regulation of cerebral blood flow

Starting date

Contact information

Notes

CTRI/2018/12/016568

Study name

An interventional study to look at efficacy of fluoxetine in patients with post‐stroke anxiety

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 60 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: triple (participant, investigator and outcome assessor)

Primary purpose: treatment

Participants

Country: India

Setting: inpatient

Inclusion criteria

  • Age 18 years to 99 years

  • Ischaemic stroke, haemorrhagic stroke and TIA

  • Able to understand English or Hindi

Exclusion criteria

  • Known psychiatric disorder

  • Moderate to severe depression (HAM‐D > 13)

  • Aphasia

  • Severe cognitive deficit

  • Refusal to participate in study

Interventions

Experimental: 20 mg of fluoxetine orally, daily for 12 weeks

Comparator: standard care

Outcomes

Primary and secondary outcomes at 12 weeks

Primary outcome

  • Improvement in HAM‐A

Secondary outcomes

  • Frequency of anxiety and depression after stroke

  • Activities of daily living measured by BI

  • Improvement in QoL as measured by 36 item short form questionnaire (SF‐36)

Starting date

December 2018

Contact information

Dr Deepti Vibha, [email protected]

Notes

EudraCT 2005‐005266‐37

Study name

Influence of escitalopram on the incidence of depression and dementia following acute middle cerebral artery territory infarction. A randomised, placebo‐controlled, double‐blind study

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 60 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: double (detail unclear)

Primary purpose: prevention

Participants

Country: Germany

Setting: inpatient

Inclusion criteria

  • Acute MCA territory infarction

  • Aiming to recruit 60 over 3 years

  • Within 7 days of stroke onset

  • Prepared to and considered able to follow the whole trial period

Exclusion criteria

  • Dementia

  • Recurrent major depression

  • Major stroke

  • Alcohol and drug dependency

  • Pregnancy, breastfeeding

  • Participating in other trials of medicinal products

  • Impaired liver/kidney disease

  • Life expectancy less than 6 months

Aiming to recruit 60 participants

Interventions

Experimental: escitalopram

Comparator: placebo

Outcomes

Depression (MADRS) after 180 days

Incidence of dementia after 180 days (Clinical Dementia Rating scale)

Severity of dementia

Zarit Burden Interview

Incidence of depression (Depression Visual Analogue Scale)

Severity of post‐stroke depression

Quality of life (SF‐36)

Bayer Activities of Daily Living score

NPI

Starting date

MHRA approval 7 April 2006; start date not known

Contact information

Not available. National Competent Authority is Germany‐BFarm

Sponsor Name: Central Institute for Mental Health, Mannheim, Division of Gerontopsychiatry

Notes

Details available on EudraCT website

www.clinicaltrialsregister.eu/ctr‐search/trial/2005‐005266‐37/DE

IRCT201112228490N1

Study name

Effect of fluoxetine on functional recovery of patients with cerebrovascular accident following middle cerebral artery trunk obstruction: a randomised clinical trial

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 60 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: unclear

Primary purpose: treatment

Participants

Iran (Islamic Republic of Iran)

Inclusion criteria

  • Age 55 to 85 years

  • Informed consent

  • Unilateral occlusion of middle cerebral artery trunk

  • Resident in Rasht

  • Admission NIHSS < 20

  • No history of alcohol abuse

  • No history of insomnia

  • No epilepsy

  • "No history of cerebral haemorrhage and heart of cerebral stroke" [sic]

  • No history of systemic diseases of other organs, including liver failure and kidney

  • No cardiac pace maker, severe neuropathy, systemic vascular disease or major affective disorders

  • No concomitant stroke in an area other than the stroke of the middle cerebral artery

Exclusion criteria

  • Dissatisfaction of patient during the study

  • Occurrence of serious adverse drug affects at any time during drug administration

  • Alcohol abuse during the study period

  • Occurrence of post‐stroke depression, concomitant use of the MAOIs or serotonergic drugs such as tricyclic antidepressants and SSRI

Interventions

Intervention: fluoxetine, 15 mg oral pill for the first month and 20 mg for the next 2 months

Comparator: placebo, 15 mg oral pill for the first month and 20 mg for the next 2 months

Outcomes

Primary outcomes collected at discharge, 1 and 3 months

  • Disability (mRS)

  • Activities of Daily Living (BI)

  • Functional recovery (NIHSS)

  • Depression (BDI questionnaire)

Secondary outcomes collected at discharge

  • Cerebral blood flow changes of middle cerebral artery (TCD)

Starting date

5 April 2012

Contact information

Dr Babak Bakhshayesh Eghbali

Poorsina hospital, Guilan University of Medical Sciences

[email protected]

Notes

IRCT201112228490N1

Contacted 7 February 2019

IRCT2012101011062N1

Study name

A study of sertraline effect on quality of life in stroke inpatients

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 80 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: unclear

Primary purpose: prevention

Participants

Iran (Islamic Republic of Iran)

Inclusion criteria

  • 55 years old to 75 years old

  • First‐ever stroke

Exclusion criteria

  • History of stroke

  • Renal failure

  • Hepatic failure

  • Cardiac failure

  • Substance related disorders

Aiming to recruit 80 participants

Interventions

Experimental: 3 weeks after stroke sertraline 50 mg a day for 12 months versus

Comparator: 3 weeks after stroke a placebo tablet every day

Outcomes

Primary outcomes collected at 3 months, 6 months, 9 months

  • Quality of life (NHP)

Secondary outcomes collected at 3 months, 6 months, 9 months

  • Depression (BDI)

Starting date

28 November 2012. Contacted author for an update on 4 May 2021 but no response received

Contact information

Reza Pirzeh, Tabriz University of Medical Sciences, Iran (Islamic Republic of)

[email protected]

Notes

IRCT2012101011062N1

IRCT2017041720258N37

Study name

Evaluation of fluoxetine and standard treatment efficacy on change to side effect of stroke of ischaemic strokes in both hemispheres in anterior circulation

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 60 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: unclear

Primary purpose: treatment

Participants

Islamic Republic of Iran

Inclusion criteria

  • Age 40 to 70 years

  • No previous history ischaemic stroke

  • Diagnosis of stroke confirmed by imaging

  • Within 2 to 7 days of stroke onset

Exclusion criteria

  • Not available during study period

  • History of side effect of fluoxetine and other antipsychotic drugs

  • Pregnant or breast feeding

  • Depression in the previous month and treatment with antipsychotic drugs

  • Use of any MAOI in the last 5 months

Aiming to recruit 60 participants

Interventions

Experimental: 20 milligram fluoxetine and ‎standard treatment (antiplatelets, anticoagulant and statin)

Comparator: placebo and ‎standard treatment (antiplatelets, anticoagulant and statin)

Outcomes

Primary outcome collected at 1 and 2 months

  • Change to side effect of stroke (NIHSS questionnaire)

Starting date

11 October 2017. Trials register states it's complete but no results are available (email to author on 4 May 2021, no reply)

Contact information

Fariba Farokhi, Arak University of Medical Sciences Iran (Islamic Republic of Iran)

[email protected]

Notes

www.irct.ir/trial/17976

IRCT20210307050617N1

Study name

The efficacy comparison of fluoxetine and citalopram on motor recovery after ischemic stroke: single‐blind placebo‐controlled randomized clinical trial

Methods

3 arm RCT‐ citalopram, fluoxetine or placebo

Participants

Ischaemic stroke, at least 18 years old, hemiparesis or hemiplegia after the first ischaemic stoke in 24 hours. A score greater than 2 NIHSS on the motor items

Interventions

Fluoxetine 20 mg daily for 3 months or citalopram 10 mg for first 10 days then 20 mg daily. Placebo‐starch given for 3 months

Outcomes

Rate of improvement in motor function

Starting date

22 August 2020

Contact information

[email protected]

Notes

NCT02386475

Study name

Effect of serotonin and levodopa in ischemic stroke (SELEIS)

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 240 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: single (outcomes assessor)

Primary purpose: treatment

Participants

Country: Spain

Setting: inpatient

Inclusion criteria:

  • Age > 18 years

  • NIHSS 5 to 20 points

  • mRS < 3 prior to stroke

  • Participants without prior cognitive impairment or depressive syndrome

  • Assigned treatment initiated within the first 5 days of stroke

Exclusion criteria:

  • Aphasia

  • Prior myocardial or cerebral haemorrhage

  • TIA

  • History of cognitive impairment or prior depressive syndrome

  • mRS 3 or higher

  • Life‐threatening illness that is likely to reduce 1‐year survival

  • Use of levodopa, an antidepressant or neuroleptic

Aiming to recruit 240 participants.

Interventions

Placebo comparator: placebo

Active comparator: citalopram 20 mg

Active comparator: sinemet plus 100 mg

Sinemet plus + citalopram group

Outcomes

Rankin Scale at 12 months

Starting date

1 January 2015. Study completed 31 October 2019. No results available‐contacted author on 4 May 2021

Contact information

Dolores Cocho mailto:dcocho%40fhag.es?subject=NCT02386475, SELEIS, Effect of Serotonin and Levodopa in Ischemic Stroke

Notes

NCT02386475

NCT02767999

Study name

Resting state MRI connectivity in acute ischemic stroke: serotonin selective reuptake Inhibitor (SSRI) in enhancing motor recovery: a placebo controlled study

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 60 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: double (participant, investigator)

Primary purpose: treatment

Participants

Country: France

Setting: inpatient

Inclusion criteria

  • Age 18 years to 85 years

  • First‐ever ischaemic stroke

  • Cortical or subcortical stroke

  • NIHSS > 12 or motor NIHSS > 6 at inclusion

  • MRI‐proved ischaemic stroke

Exclusion criteria

  • Pregnant or breast‐feeding

  • Alcoholism

  • Ongoing SSRI treatment or interruption < 1 month

  • Allergic reaction after SSRI administration

  • MRI contraindication

  • NIHSS > 22

  • Severe aphasia

  • Coma

Interventions

Experimental: 20 mg of fluoxetine capsule a day from day 0 to day 90 and have fMRI

Comparator: cellulose placebo a day from day 0 to day 90 and have fMRI

Outcomes

Primary outcome at 90 days

  • Intracerebral connectivity in the motor network between fluoxetine and placebo group

Secondary outcome at 90 days

  • Intracerebral connectivity in the motor network between good responder participants (defined by 8 points gain on the NIHSS, assessed between day 0 and day 30 and between day 0 and day 90, or 2 points gain on the mRS assessed between day 0 and day 30 and between day 0 and day 90)

  • Intracerebral connectivity in the motor network between non‐responder participants

Starting date

January 2016

Contact information

Virginie Sattler, Dr: sattler.v@chu‐toulouse.fr

François Chollet, MD PhD: chollet.f@chu‐toulouse.fr

Notes

NCT02767999

NCT02865642

Study name

Cortical Ischemic Stroke and Serotonin (CISS)

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 90 participants

Allocation: randomised

Intervention model: parallel assignment

Masking: quadruple (participant, care provider, investigator, outcomes assessor)

Primary purpose: supportive care

Participants

Country: Switzerland

Setting: inpatient

Inclusion criteria

  • First‐ever stroke

  • Clinically significant contralesional hand plegia or paresis as a main symptom

  • Involvement of the pre‐and/or post‐central gyri confirmed on DWI and FLAIR scans

  • Written informed consent

Exclusion criteria

  • Aphasia or cognitive deficits severe enough to preclude understanding of study purposes

  • Prior cerebrovascular events

  • Significant stenosis (70% to 99% according to NASCET) or occlusion of the carotid and intracranial arteries on MRA

  • Purely subcortical stroke

  • Known brain lesion (tumour, old cerebral haemorrhage)

  • Other medical conditions interfering with task performance or SSRI‐treatment, specifically: prolonged corrected QT interval (QTc) on electrocardiogram, ongoing drug/alcohol abuse

  • Simultaneous intake of medications which can lead to prolonged QTc syndrome known or suspected hypersensitivity to one of the ingredients of Cipralex® or placebo

  • Simultaneous administration of antidepressants

  • Conditions interfering with MRI (e.g. patients with a cardiac pacemaker or cochlear implant)

  • Pregnant or breastfeeding

  • Women in childbearing age without sufficient birth control (at least 2 contraceptive methods)

Interventions

Experimental: escitalopram 5 mg/day at the baseline visit (day 14 (±7) post‐stroke) for 7 days followed by a weekly dosage increase of 5 mg/day till target dose of escitalopram 20 mg/day. Participants remain on escitalopram 20 mg/day until visit 3 (day 90 (±14) post‐stroke) followed by dosage reduction of escitalopram 10 mg/day for 1 week

Comparator: placebo 5 mg/day at the baseline visit (day 14 (±7) post‐stroke) for 7 days followed by a weekly dosage of 5 mg/day until target dose of placebo 20 mg/day. Participants remain on placebo 20 mg/day until visit 3 (day 90 (±14) post‐stroke) followed by placebo 10 mg/day for 1 week

Outcomes

Primary outcome

  • Effect of escitalopram on sensorimotor network at month 9 (task‐related fMRI (act‐fMRI))

Secondary outcomes

  • Imaging patterns of rs‐fMRI at month 3 and month 9

  • Imaging patterns of act‐fMRI at month 3 and month 9

  • JTT monthly from baseline to month 9

  • Mean cortical volume changes at month 3 and month 9

  • Serum concentration of escitalopram at month 3

  • Genetic polymorphisms in genes at month 3

Other outcomes

  • Glutamate/glutamine concentration at month 3 and month 9

  • rTMS at month 3 and month 9

  • Number of adverse events due to study medication monthly until month 9

Starting date

August 2016

Contact information

Manuela Pastore‐Wapp manuela.pastore‐[email protected]

Notes

NCT02865642

NCT03448159

Study name

FLuoxetine Opens Window to improve motor recovery after stroke (FLOW)

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 176 participants

Allocation: randomised

Intervention model: parallel assignment

Intervention model description

  • Intervention group (trial drug (fluoxetine) and exercise intervention)

  • Placebo group (placebo and exercise intervention)

Masking: quadruple (participant, care provider, investigator, outcomes assessor)

Primary purpose: treatment

Participants

Country: Canada

Setting: inpatient

Inclusion criteria

  • Age > 25 years

  • Between 60 to 210 days post‐stroke at enrolment

  • Lower limb FMMS < 30

Exclusion criteria

  • Subarachnoid haemorrhage

  • Pre‐morbid mRS > 2

  • Substantial premorbid disability or pre‐existing deficit or language comprehension deficit that could interfere with assessments

  • Diagnosis of major depressive disorder/anxiety disorder requiring antidepressant use within 6 weeks of enrolment

  • Taking neuroleptic drugs, benzodiazepines, MAOIs within 30 days of enrolment

  • Unstable serious medical condition (e.g. terminal cancer, renal or liver failure, congestive heart failure)

  • Resting blood pressure exceeding 180/100 mmHg

  • Requires more than a one‐person assist for transfer

  • Planned surgery that would affect participation in the trial

  • Participating in another exercise programme more than 1 day a week

  • Pregnant

  • Ongoing history of illicit drug use or alcohol abuse or both

  • Unwilling or unable to comply with trial requirements

  • Unable to understand English

Interventions

Experimental: fluoxetine hydrochloride (Prozac): 10 mg Prozac per day for 3 to 5 weeks and then 20 mg for 12 weeks (the duration of the exercise intervention)

Comparator: an over‐encapsulated placebo (identical 'sugar pill'): 10 mg 'sugar pill' a day for 3 to 5 weeks and then 20 mg for 12 weeks (the duration of the exercise intervention)

Outcomes

Primary outcomes at 12 weeks

  • Fugl‐Meyer Lower Extremity Score at 12 weeks

Secondary outcomes at 12 weeks and 6 months

  • Ambulatory function measured using 6‐Minute Walk Test/10 Metre Walk Test

  • Lower limb strength measured using knee strength

  • Balance measured using Berg Balance Assessment

  • Grip Strength

  • Waist‐to‐Hip Ratio

  • Body Mass Index

  • SIS

  • Fugl‐Meyer Lower Extremity Score at 6 months

  • Fugl‐Meyer Upper Extremity Score

  • PHQ‐9

  • Simple and Choice Reaction Time Test

  • Trail Making Test ‐ A & B

  • Montreal Cognitive Assessment

  • Fasting Blood Draws

Starting date

1 November 2018

Contact information

Farrell Leibovitch

mailto:farrell%40canadianstroke.ca?subject=NCT03448159, 17‐5134.0, Fluoxetine Opens Window to Improve Motor Recovery After Stroke

Notes

NCT03448159

NCT03826875

Study name

Depression in haemorrhagic stroke

Methods

Study type: interventional (clinical trial)

Estimated enrolment: 224 participants

Allocation: randomised

Intervention model: parallel assignment

Intervention model description: double‐blinded placebo‐controlled randomised trial

Masking: triple (participant, care provider, investigator)

Primary purpose: prevention

Participants

Country: USA

Setting: inpatient

Inclusion criteria

  • Age 18 to 85 years

  • Subarachnoid haemorrhage from a ruptured cerebral aneurysm

  • Consent

Exclusion criteria

  • Non‐English speaking

  • Taking therapy for depression or related mental health diagnoses before admission

  • Medical contraindications to fluoxetine therapy

  • Pregnancy or considering getting pregnant during the trial period at the time of consent.

  • Active psychosis

  • Incarcerated or in police custody

  • Comorbidity or a score > 26 on the MoCA

Interventions

Experimental: fluoxetine 20 mg/day for a period of 1 year

Comparator: placebo 20 mg/day for a period of 1 year

Outcomes

Primary outcomes at 1 year

  • Depression measured using HAM‐D

  • Depression measured using PHQ‐9

Secondary outcomes at 1 year:

  • Anxiety measured using Hamilton Rating Scale for Anxiety

  • Fatigue measured using Fatigue Severity Scale

  • Healthcare utilization measured using Self‐Report Health Service Utilization and Medication Use

  • Social support measured using Multidimensional Scale of Perceived Social Support (MSPSS)

Starting date

1 March 2019

Contact information

Cory M Kelly: [email protected]

Notes

NCT03826875

TCTR20181216001

Study name

Randomized controlled trial of fluoxetine or placebo on quality of life after acute ischemic stroke

Methods

Study type: interventional (clinical trial)

Allocation: randomised

Intervention model: parallel assignment

Masking: double blind (no detail)

Primary purpose: efficacy

Participants

Country: Thailand

Setting: inpatient

Inclusion criteria

  • Age > 18

  • Acute ischaemic stroke patient

  • Consent

Exclusion criteria

  • History of or current psychiatric condition ‐ hemorrhagic complication

  • EQ‐5D‐5L score > 0.9

  • Language barrier

Interventions

Experimental: fluoxetine 20 mg once daily for 90 days

Comparator: matching placebo once daily for 90 days

Outcomes

Primary outcomes at 3 months

  • Quality of life measured by EQ‐5D‐5L

Secondary outcomes at 3 months

  • Post stroke depression measured by Thai HADS

  • Disability measured by the Modified Rankin Score

Starting date

1 January 2019

Contact information

Sirikanya Lorwatanapongsa: [email protected]

Notes

TCTR20181216001

BDI: Beck Depression Inventory;
BDNF: brain‐derived neurotrophic factor;
BI: Barthel Index;
CTIMP: Clinical Trial of an Investigational Medicinal Product;
DWI: diffusion‐weighted imaging;
FLAIR: fluid‐attenuated inversion recovery;
FMMS: Fugl Meyr Motor Score;
fMRI: functional magnetic resonance imaging;
HAM‐A:Hamilton Anxiety Rating Scale;
HAM‐D: Hamilton Depression Rating Scale;
JTT: Jebsen Taylor Test;
MADRS: Montgomery‐Åsberg Depression Rating Scale;
MAOI: mono‐amino‐oxidase inhibitor;
MCA: middle cerebral artery;
MHRA: Medicines and Healthcare products Regulatory Agency
MRA: magnetic resonance angiography;
MRI: magnetic resonance imaging;
mRS: modified Rankin score;
NHP: Nottingham Health Profile;
NIHSS: National Institute of Health Stroke Scale
NPI: Neuropsychiatric Inventory Scale;
PHQ‐9: Patient Health Questionnaire;
QoL: quality of life;
RCT: randomised controlled trial;
rTMS: repetitive transcranial magnetic stimulation;
SF‐36: Short Form‐36;
smRSq: simplified modified Rankin Scale questionnaire;
SSRI: selective serotonin reuptake inhibitor;
TCD: transcranial Doppler;
TIA: transient ischaemic attack;

Data and analyses

Open in table viewer
Comparison 1. SSRI versus control at end of treatment, by SSRI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Disability (primary outcome). Studies at low risk of bias Show forest plot

5

5436

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

‐0.00 [‐0.05, 0.05]

Analysis 1.1

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 1: Disability (primary outcome). Studies at low risk of bias

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 1: Disability (primary outcome). Studies at low risk of bias

1.1.1 Fluoxetine

5

5436

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

‐0.00 [‐0.05, 0.05]

1.2 Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias Show forest plot

5

5926

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.93, 1.03]

Analysis 1.2

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 2: Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 2: Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias

1.3 Neurological deficit score (studies at low risk of bias) Show forest plot

1

30

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

‐0.39 [‐1.12, 0.33]

Analysis 1.3

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 3: Neurological deficit score (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 3: Neurological deficit score (studies at low risk of bias)

1.3.1 Fluoxetine

1

30

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

‐0.39 [‐1.12, 0.33]

1.4 Motor deficits (studies at low risk of bias) Show forest plot

6

5518

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

0.03 [‐0.02, 0.08]

Analysis 1.4

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 4: Motor deficits (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 4: Motor deficits (studies at low risk of bias)

1.4.1 Fluoxetine

6

5518

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

0.03 [‐0.02, 0.08]

1.5 Depression, continuous data (studies at low risk of bias) Show forest plot

4

5356

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

‐0.14 [‐0.19, ‐0.08]

Analysis 1.5

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 5: Depression, continuous data (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 5: Depression, continuous data (studies at low risk of bias)

1.5.1 Fluoxetine

4

5356

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

‐0.14 [‐0.19, ‐0.08]

1.6 Depression, dichotomous data (studies at low risk of bias) Show forest plot

3

5907

Risk Ratio (M‐H, Fixed, 95% CI)

0.75 [0.65, 0.86]

Analysis 1.6

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 6: Depression, dichotomous data (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 6: Depression, dichotomous data (studies at low risk of bias)

1.6.1 Fluoxetine

3

5907

Risk Ratio (M‐H, Fixed, 95% CI)

0.75 [0.65, 0.86]

1.7 Death (trials at low risk of bias) Show forest plot

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.82, 1.24]

Analysis 1.7

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 7: Death (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 7: Death (trials at low risk of bias)

1.7.1 Fluoxetine

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.82, 1.24]

1.8 Seizures (studies at low risk of bias) Show forest plot

6

6080

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.00, 1.98]

Analysis 1.8

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 8: Seizures (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 8: Seizures (studies at low risk of bias)

1.8.1 Fluoxetine

6

6080

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.00, 1.98]

1.9 Gastrointestinal side effects (studies at low risk of bias) Show forest plot

1

30

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.33, 8.83]

Analysis 1.9

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 9: Gastrointestinal side effects (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 9: Gastrointestinal side effects (studies at low risk of bias)

1.9.1 Fluoxetine

1

30

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.33, 8.83]

1.10 Bleeding (studies at low risk of bias) Show forest plot

6

6088

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.69, 1.70]

Analysis 1.10

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 10: Bleeding (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 10: Bleeding (studies at low risk of bias)

1.10.1 Fluoxetine (except for Asadollahi 2018)

6

6088

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.69, 1.70]

1.11 Fractures (studies at low risk of only) Show forest plot

6

6080

Odds Ratio (M‐H, Fixed, 95% CI)

2.35 [1.62, 3.41]

Analysis 1.11

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 11: Fractures (studies at low risk of only)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 11: Fractures (studies at low risk of only)

1.12 Cognition (trials at low risk of bias) Show forest plot

4

5373

Mean Difference (IV, Fixed, 95% CI)

‐1.22 [‐2.37, ‐0.07]

Analysis 1.12

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 12: Cognition (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 12: Cognition (trials at low risk of bias)

1.13 Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias) Show forest plot

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.40]

Analysis 1.13

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 13: Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 13: Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias)

1.13.1 Fluoxetine

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.40]

1.14 Fatigue at end of treatment (studies at low risk of bias only) Show forest plot

4

5524

Mean Difference (IV, Fixed, 95% CI)

‐0.06 [‐1.24, 1.11]

Analysis 1.14

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 14: Fatigue at end of treatment (studies at low risk of bias only)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 14: Fatigue at end of treatment (studies at low risk of bias only)

1.15 Quality of life at end of treatment (studies at low risk of bias) Show forest plot

3

5482

Mean Difference (IV, Fixed, 95% CI)

0.00 [‐0.02, 0.02]

Analysis 1.15

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 15: Quality of life at end of treatment (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 15: Quality of life at end of treatment (studies at low risk of bias)

1.16 Disability (all studies regardless of risk of bias) Show forest plot

32

7667

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

‐0.18 [‐0.23, ‐0.14]

Analysis 1.16

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 16: Disability (all studies regardless of risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 16: Disability (all studies regardless of risk of bias)

1.16.1 Fluoxetine

19

6590

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

‐0.09 [‐0.13, ‐0.04]

1.16.2 Sertraline

1

130

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

‐1.38 [‐1.76, ‐0.99]

1.16.3 Paroxetine

5

293

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

‐1.29 [‐1.55, ‐1.03]

1.16.4 Citalopram

5

446

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

‐0.68 [‐0.88, ‐0.48]

1.16.5 Escitalopram

2

208

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

‐0.67 [‐1.00, ‐0.34]

1.17 Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias) Show forest plot

8

6792

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.93, 1.01]

Analysis 1.17

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 17: Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 17: Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias)

1.17.1 Fluoxetine

6

6039

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.94, 1.03]

1.17.2 Sertraline

1

111

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.97, 1.04]

1.17.3 Citalopram

1

642

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.82, 0.98]

Open in table viewer
Comparison 2. SSRI versus control at end of follow up, by SSRI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Disability (studies at low risk of bias only) Show forest plot

2

2591

Mean Difference (IV, Fixed, 95% CI)

‐0.24 [‐2.59, 2.11]

Analysis 2.1

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 1: Disability (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 1: Disability (studies at low risk of bias only)

2.2 Independent on modified rankin score (0‐2) (studies at low risk of bias only) Show forest plot

2

3137

Odds Ratio (M‐H, Fixed, 95% CI)

1.03 [0.89, 1.19]

Analysis 2.2

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 2: Independent on modified rankin score (0‐2) (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 2: Independent on modified rankin score (0‐2) (studies at low risk of bias only)

2.3 Depression, continuous data (studies at low risk of bias only) Show forest plot

2

2684

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.36, 0.44]

Analysis 2.3

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 3: Depression, continuous data (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 3: Depression, continuous data (studies at low risk of bias only)

2.4 Depression, dichotomous (studies at low risk of bias only) Show forest plot

1

3083

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.73, 1.04]

Analysis 2.4

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 4: Depression, dichotomous (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 4: Depression, dichotomous (studies at low risk of bias only)

2.5 Motor deficits (studies at low risk of bias only) Show forest plot

2

2688

Mean Difference (IV, Fixed, 95% CI)

‐0.77 [‐3.00, 1.46]

Analysis 2.5

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 5: Motor deficits (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 5: Motor deficits (studies at low risk of bias only)

2.6 Cognition (studies at low risk of bias only) Show forest plot

2

2689

Mean Difference (IV, Fixed, 95% CI)

‐0.35 [‐2.32, 1.62]

Analysis 2.6

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 6: Cognition (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 6: Cognition (studies at low risk of bias only)

2.7 Death (studies at low risk of bias only) Show forest plot

2

3144

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.77, 1.12]

Analysis 2.7

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 7: Death (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 7: Death (studies at low risk of bias only)

2.8 Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias) Show forest plot

2

3188

Odds Ratio (M‐H, Fixed, 95% CI)

0.90 [0.54, 1.51]

Analysis 2.8

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 8: Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 8: Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias)

2.9 Disability, all studies irrespective of risk of bias Show forest plot

2

2691

Mean Difference (IV, Fixed, 95% CI)

‐0.25 [‐2.56, 2.06]

Analysis 2.9

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 9: Disability, all studies irrespective of risk of bias

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 9: Disability, all studies irrespective of risk of bias

2.10 Independent on mRS (0‐2) all studies irrespective of risk of bias Show forest plot

2

3134

Odds Ratio (M‐H, Fixed, 95% CI)

1.03 [0.89, 1.19]

Analysis 2.10

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 10: Independent on mRS (0‐2) all studies irrespective of risk of bias

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 10: Independent on mRS (0‐2) all studies irrespective of risk of bias

PRISMA flow diagram for this update

Figuras y tablas -
Figure 1

PRISMA flow diagram for this update

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

Figuras y tablas -
Figure 2

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

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

Figuras y tablas -
Figure 3

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

Funnel plot, all studies irrespective or risk of bias, for disability at end of treatment.

Figuras y tablas -
Figure 4

Funnel plot, all studies irrespective or risk of bias, for disability at end of treatment.

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 1: Disability (primary outcome). Studies at low risk of bias

Figuras y tablas -
Analysis 1.1

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 1: Disability (primary outcome). Studies at low risk of bias

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 2: Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias

Figuras y tablas -
Analysis 1.2

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 2: Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 3: Neurological deficit score (studies at low risk of bias)

Figuras y tablas -
Analysis 1.3

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 3: Neurological deficit score (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 4: Motor deficits (studies at low risk of bias)

Figuras y tablas -
Analysis 1.4

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 4: Motor deficits (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 5: Depression, continuous data (studies at low risk of bias)

Figuras y tablas -
Analysis 1.5

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 5: Depression, continuous data (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 6: Depression, dichotomous data (studies at low risk of bias)

Figuras y tablas -
Analysis 1.6

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 6: Depression, dichotomous data (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 7: Death (trials at low risk of bias)

Figuras y tablas -
Analysis 1.7

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 7: Death (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 8: Seizures (studies at low risk of bias)

Figuras y tablas -
Analysis 1.8

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 8: Seizures (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 9: Gastrointestinal side effects (studies at low risk of bias)

Figuras y tablas -
Analysis 1.9

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 9: Gastrointestinal side effects (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 10: Bleeding (studies at low risk of bias)

Figuras y tablas -
Analysis 1.10

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 10: Bleeding (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 11: Fractures (studies at low risk of only)

Figuras y tablas -
Analysis 1.11

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 11: Fractures (studies at low risk of only)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 12: Cognition (trials at low risk of bias)

Figuras y tablas -
Analysis 1.12

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 12: Cognition (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 13: Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias)

Figuras y tablas -
Analysis 1.13

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 13: Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 14: Fatigue at end of treatment (studies at low risk of bias only)

Figuras y tablas -
Analysis 1.14

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 14: Fatigue at end of treatment (studies at low risk of bias only)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 15: Quality of life at end of treatment (studies at low risk of bias)

Figuras y tablas -
Analysis 1.15

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 15: Quality of life at end of treatment (studies at low risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 16: Disability (all studies regardless of risk of bias)

Figuras y tablas -
Analysis 1.16

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 16: Disability (all studies regardless of risk of bias)

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 17: Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias)

Figuras y tablas -
Analysis 1.17

Comparison 1: SSRI versus control at end of treatment, by SSRI, Outcome 17: Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 1: Disability (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.1

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 1: Disability (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 2: Independent on modified rankin score (0‐2) (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.2

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 2: Independent on modified rankin score (0‐2) (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 3: Depression, continuous data (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.3

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 3: Depression, continuous data (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 4: Depression, dichotomous (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.4

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 4: Depression, dichotomous (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 5: Motor deficits (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.5

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 5: Motor deficits (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 6: Cognition (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.6

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 6: Cognition (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 7: Death (studies at low risk of bias only)

Figuras y tablas -
Analysis 2.7

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 7: Death (studies at low risk of bias only)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 8: Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias)

Figuras y tablas -
Analysis 2.8

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 8: Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias)

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 9: Disability, all studies irrespective of risk of bias

Figuras y tablas -
Analysis 2.9

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 9: Disability, all studies irrespective of risk of bias

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 10: Independent on mRS (0‐2) all studies irrespective of risk of bias

Figuras y tablas -
Analysis 2.10

Comparison 2: SSRI versus control at end of follow up, by SSRI, Outcome 10: Independent on mRS (0‐2) all studies irrespective of risk of bias

Summary of findings 1. Fluoxetine versus control at end of treatment, for stroke recovery, using data from high quality trials only

Fluoxetine versus control at end of treatment, by SSRI, for stroke recovery*

Patient or population: people with stroke recovery
Settings: hospital
Intervention: fluoxetine versus control at end of treatment

*Summary of findings table based on studies with low risk of bias.

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Fluoxetine versus control at end of treatment

Disability (primary analysis)

 

SMD 0.0 (‐0.05, 0.05)

 

5436 
(5 studies)

⊕⊕⊕⊕
High

Independent on modified Rankin score (mRS 0 to 2) (primary analysis)

Study population

RR 0.98
(0.93 to 1.03)

 

 

5926 
(5 studies)

⊕⊕⊕⊕
High

1541/2971 (i.e. 52 per hundred)

1498/2955

(i.e. 51 per hundred)

Neurological deficit score

 

SMD ‐0.39 (95% CI (‐1.12 to 0.33)

 

30 participants, one study

⊕⊕⊝⊝

Lowa

This is a small effect (based on the 'rule‐of‐thumb' method for interpreting SMD)

Depression (continuous data)

 

SMD ‐0.14 (‐0.19 to ‐0.08)

 

5356
(4 studies)

⊕⊕⊕⊕
High

This is a small effect (based on the 'rule‐of‐thumb' method for interpreting SMD)

Death

Study population

RR 1.01
(0.82 to 1.24)

6090
(6 studies)

⊕⊕⊕⊝
Moderate

168/3029 (i.e. 55 per thousand)

170/3061

(i.e. 56 per thousand)

Number of seizures

Study population

RR 1.40
(1.00 to 1.98)

6080 
(6 studies)

⊕⊕⊕⊝

Moderatec

54/3024 (i.e. 18 per thousand)

76/3056 (i.e. 25 per thousand)

Bone fractures

Study population

RR 2.35 
(1.62 to 3.41)

6080
(6 studies)

⊕⊕⊕ ⊕ 
High

40/3024

(i.e. 13 per thousand)

93/3056

(i.e. 30 per thousand)

*The basis for the assumed risk (e.g. the mean control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; SMD: standardised mean difference

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aNeurological deficit from only one trial of 30 people so we have downgraded for imprecision (GRADE 2013).

bDeath downgraded for imprecision.

cSeizures downgraded for imprecision.

Note that because we included only the low risk of bias studies in our review, none of the evidence was downgraded because of study quality.

A range of different outcome scales were used for disability (including Barthel Index and daily activities subscale of the Stroke Impact Scale), and depression (including emotional role function of the Stroke Impact Scale).

Figuras y tablas -
Summary of findings 1. Fluoxetine versus control at end of treatment, for stroke recovery, using data from high quality trials only
Table 1. Sensitivity analysis for co‐primary outcomes depending on the method of analysis

mRS (RR and 95% CI)

Disability (SMD and 95% CI)

Fixed‐effect

0.98 (0.93, 1.03)

‐0.0 (‐0.05, 0.05)

Random‐effects

0.98 (0.92, 1.04)

‐0.00 (‐0.05, 0.05)

CI: confidence interval
mRS: modified Rankin Scale
RR: risk ratio
SMD: standardised mean difference

Figuras y tablas -
Table 1. Sensitivity analysis for co‐primary outcomes depending on the method of analysis
Comparison 1. SSRI versus control at end of treatment, by SSRI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Disability (primary outcome). Studies at low risk of bias Show forest plot

5

5436

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

‐0.00 [‐0.05, 0.05]

1.1.1 Fluoxetine

5

5436

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

‐0.00 [‐0.05, 0.05]

1.2 Independent on modified Rankin score (mRS 0 to 2) (primary outcome). Studies at low risk of bias Show forest plot

5

5926

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.93, 1.03]

1.3 Neurological deficit score (studies at low risk of bias) Show forest plot

1

30

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

‐0.39 [‐1.12, 0.33]

1.3.1 Fluoxetine

1

30

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

‐0.39 [‐1.12, 0.33]

1.4 Motor deficits (studies at low risk of bias) Show forest plot

6

5518

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

0.03 [‐0.02, 0.08]

1.4.1 Fluoxetine

6

5518

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

0.03 [‐0.02, 0.08]

1.5 Depression, continuous data (studies at low risk of bias) Show forest plot

4

5356

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

‐0.14 [‐0.19, ‐0.08]

1.5.1 Fluoxetine

4

5356

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

‐0.14 [‐0.19, ‐0.08]

1.6 Depression, dichotomous data (studies at low risk of bias) Show forest plot

3

5907

Risk Ratio (M‐H, Fixed, 95% CI)

0.75 [0.65, 0.86]

1.6.1 Fluoxetine

3

5907

Risk Ratio (M‐H, Fixed, 95% CI)

0.75 [0.65, 0.86]

1.7 Death (trials at low risk of bias) Show forest plot

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.82, 1.24]

1.7.1 Fluoxetine

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.82, 1.24]

1.8 Seizures (studies at low risk of bias) Show forest plot

6

6080

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.00, 1.98]

1.8.1 Fluoxetine

6

6080

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.00, 1.98]

1.9 Gastrointestinal side effects (studies at low risk of bias) Show forest plot

1

30

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.33, 8.83]

1.9.1 Fluoxetine

1

30

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.33, 8.83]

1.10 Bleeding (studies at low risk of bias) Show forest plot

6

6088

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.69, 1.70]

1.10.1 Fluoxetine (except for Asadollahi 2018)

6

6088

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.69, 1.70]

1.11 Fractures (studies at low risk of only) Show forest plot

6

6080

Odds Ratio (M‐H, Fixed, 95% CI)

2.35 [1.62, 3.41]

1.12 Cognition (trials at low risk of bias) Show forest plot

4

5373

Mean Difference (IV, Fixed, 95% CI)

‐1.22 [‐2.37, ‐0.07]

1.13 Leaving the study before the end of scheduled follow‐up for reasons other than death (trials at low risk of bias) Show forest plot

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.40]

1.13.1 Fluoxetine

6

6090

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.40]

1.14 Fatigue at end of treatment (studies at low risk of bias only) Show forest plot

4

5524

Mean Difference (IV, Fixed, 95% CI)

‐0.06 [‐1.24, 1.11]

1.15 Quality of life at end of treatment (studies at low risk of bias) Show forest plot

3

5482

Mean Difference (IV, Fixed, 95% CI)

0.00 [‐0.02, 0.02]

1.16 Disability (all studies regardless of risk of bias) Show forest plot

32

7667

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

‐0.18 [‐0.23, ‐0.14]

1.16.1 Fluoxetine

19

6590

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

‐0.09 [‐0.13, ‐0.04]

1.16.2 Sertraline

1

130

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

‐1.38 [‐1.76, ‐0.99]

1.16.3 Paroxetine

5

293

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

‐1.29 [‐1.55, ‐1.03]

1.16.4 Citalopram

5

446

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

‐0.68 [‐0.88, ‐0.48]

1.16.5 Escitalopram

2

208

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

‐0.67 [‐1.00, ‐0.34]

1.17 Independent on modified Rankin score (mRS 0 to 2) (all studies regardless of risk of bias) Show forest plot

8

6792

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.93, 1.01]

1.17.1 Fluoxetine

6

6039

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.94, 1.03]

1.17.2 Sertraline

1

111

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.97, 1.04]

1.17.3 Citalopram

1

642

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.82, 0.98]

Figuras y tablas -
Comparison 1. SSRI versus control at end of treatment, by SSRI
Comparison 2. SSRI versus control at end of follow up, by SSRI

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Disability (studies at low risk of bias only) Show forest plot

2

2591

Mean Difference (IV, Fixed, 95% CI)

‐0.24 [‐2.59, 2.11]

2.2 Independent on modified rankin score (0‐2) (studies at low risk of bias only) Show forest plot

2

3137

Odds Ratio (M‐H, Fixed, 95% CI)

1.03 [0.89, 1.19]

2.3 Depression, continuous data (studies at low risk of bias only) Show forest plot

2

2684

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.36, 0.44]

2.4 Depression, dichotomous (studies at low risk of bias only) Show forest plot

1

3083

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.73, 1.04]

2.5 Motor deficits (studies at low risk of bias only) Show forest plot

2

2688

Mean Difference (IV, Fixed, 95% CI)

‐0.77 [‐3.00, 1.46]

2.6 Cognition (studies at low risk of bias only) Show forest plot

2

2689

Mean Difference (IV, Fixed, 95% CI)

‐0.35 [‐2.32, 1.62]

2.7 Death (studies at low risk of bias only) Show forest plot

2

3144

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.77, 1.12]

2.8 Leaving the trial before the end of follow‐up, for reasons other than death ( studies at low risk of bias) Show forest plot

2

3188

Odds Ratio (M‐H, Fixed, 95% CI)

0.90 [0.54, 1.51]

2.9 Disability, all studies irrespective of risk of bias Show forest plot

2

2691

Mean Difference (IV, Fixed, 95% CI)

‐0.25 [‐2.56, 2.06]

2.10 Independent on mRS (0‐2) all studies irrespective of risk of bias Show forest plot

2

3134

Odds Ratio (M‐H, Fixed, 95% CI)

1.03 [0.89, 1.19]

Figuras y tablas -
Comparison 2. SSRI versus control at end of follow up, by SSRI