Acessibilidade / Reportar erro

Treatment of dementia: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology

ABSTRACT

There is currently no cure for neurodegenerative or vascular dementias, but some pharmacological and non-pharmacological interventions may contribute to alleviate symptoms, slow disease progression and improve quality of life. Current treatment approaches are based on etiology, symptom profile and stage of dementia. This manuscript presents recommendations on pharmacological and non-pharmacological treatments of dementia due to Alzheimer’s disease, vascular cognitive impairment, frontotemporal dementia, Parkinson’s disease dementia, and dementia with Lewy bodies.

Keywords:
Dementia; Drug Therapy; Behavior; Cognition

RESUMO

Atualmente não há tratamento curativo para as demências neurodegenerativas ou para a demência vascular, mas algumas intervenções farmacológicas e não farmacológicas podem contribuir para aliviar os sintomas, retardar a progressão da doença e melhorar a qualidade de vida. As abordagens terapêuticas atuais são baseadas na etiologia, no perfil dos sintomas e no estágio da demência. Neste artigo apresentamos recomendações sobre os tratamentos farmacológicos e não farmacológicos da demência devida à doença de Alzheimer, comprometimento cognitivo vascular, demência frontotemporal, demência da doença de Parkinson e demência com corpos de Lewy.

Palavras-chave:
Demência; Tratamento Farmacológico; Comportamento; Cognição

INTRODUCTION

Research estimates that by 2050 about 152 million people will have dementia, or one new case every three seconds, mostly from low- and middle-income countries (LMIC). Currently, about two thirds of people diagnosed with dementia (PwD) live in LMIC11. Mukadam N, Sommerlad A, Huntley J, Livingston G. Population attributable fractions for risk factors for dementia in low-income and middle-income countries: an analysis using cross-sectional survey data. Lancet Glob Health. 2019;7(5):e596-603. doi:10.1016/S2214-109X(19)30074-9.
https://doi.org/10.1016/S2214-109X(19)30...
, added to a high estimate of underdiagnosed cases-approximately 800.000 people in Brazil-according to recent data22. Nakamura AE, Opaleye D, Tani G, Ferri CP. Dementia underdiagnosis in Brazil. Lancet. 2015;385(9966):418-9. doi:10.1016/S0140-6736(15)60153-2.
https://doi.org/10.1016/S0140-6736(15)60...
.

Dementia is one of the leading causes of disability in older adults, with significant impacts on the autonomy and quality of life (QoL) of PwD and their families. Neurodegenerative diseases and cerebrovascular disorders are the main causes of dementia, with Alzheimer’s disease (AD) accounting for more than half of the cases33. Nitrini R, Bottino CM, Albala C, Custodio Capuñay NS, Ketzoian C, Llibre Rodriguez JJ, et al. Prevalence of dementia in Latin America: a collaborative study of population-based cohorts. Int Psychogeriatr. 2009;21(4):622-30. doi:10.1017/S1041610209009430.
https://doi.org/10.1017/S104161020900943...
. In Brazil, AD ranked among the top ten causes of deaths in 201944. Institute of Health Metrics [homepage na Internet]. Brazil. 2018 [citado em 22 ago 2022]. Disponível em: http://www.healthdata.org/brazil
http://www.healthdata.org/brazil...
. Other degenerative causes include frontotemporal dementia (FTD), Parkinson’s disease (PD) dementia (PDD) and dementia with Lewy bodies (DLB).

Although we currently lack a cure for neurodegenerative or vascular dementias, some pharmacological and non-pharmacological interventions can contribute to alleviate symptoms, slow disease progression and improve QoL55. Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. Treatment of Alzheimer's Disease: recommendations and suggestions of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. doi:10.1590/S0004-282X2005000600035.
https://doi.org/10.1590/S0004-282X200500...
. Therapeutic approaches are based on etiology, symptom profile and stage of dementia. Thus, this manuscript presents updated information and recommendations on current available treatments in Brazil for dementia due to AD, vascular cognitive impairment (VCI), FTD, PDD and DLB.

TREATMENT OF DEMENTIA DUE TO AD

Pharmacological treatment

Degeneration of basal forebrain cholinergic neurons precedes clinical manifestations in AD, thus implying an early deficit in cholinergic activity in mild to moderate stages of dementia due to AD66. Bartus RT. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp Neurol. 2000;163(2):495-529. doi:10.1006/exnr.2000.7397.
https://doi.org/10.1006/exnr.2000.7397...
. Cholinesterase inhibitors (ChEI) donepezil, galantamine, and rivastigmine inhibit the ChE enzyme and increase acetylcholine availability in the brain. These drugs are approved for treating mild and moderate AD dementia, but have also shown positive effects in severe stages77. Small G, Bullock R. Defining optimal treatment with cholinesterase inhibitors in Alzheimer's disease. Alzheimers Dement. 2011;7(2):177-84. doi:10.1016/j.jalz.2010.03.016.
https://doi.org/10.1016/j.jalz.2010.03.0...
. For moderate-to-severe AD dementia, studies recommend combining ChEI with the glutamatergic NMDA receptor antagonist memantine.

Despite modest effects, use of ChEI and memantine have shown improvement in disease symptoms or slower progression when compared with placebo. Clinical benefits can be seen in cognitive, behavioral, and functional domains55. Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. Treatment of Alzheimer's Disease: recommendations and suggestions of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. doi:10.1590/S0004-282X2005000600035.
https://doi.org/10.1590/S0004-282X200500...
),(77. Small G, Bullock R. Defining optimal treatment with cholinesterase inhibitors in Alzheimer's disease. Alzheimers Dement. 2011;7(2):177-84. doi:10.1016/j.jalz.2010.03.016.
https://doi.org/10.1016/j.jalz.2010.03.0...
. Despite conflicting evidence, research shows that all ChEI may reduce AD mortality and galantamine may reduce the risk of conversion to severe dementia88. Xu H, Garcia-Ptacek S, Jönsson L, Wilmo A, Nordström P, Eriksdotter M. Long-term effects of cholinesterase inhibitors on cognitive decline and mortality. Neurology. 2021;96(e):2220-30. doi:10.1212/WNL.0000000000011832.
https://doi.org/10.1212/WNL.000000000001...
.

When prescribing these drugs, physicians must clarify to PwD and caregivers that the expected clinical effects are usually modest, sometimes characterized only by stabilization or slower worsening. Therapy should begin with the lowest dosage to avoid adverse events (AEs) and be increased at least every four weeks for ChEI and one week for memantine. Regarding ChEI, usage should reach the therapeutic doses, i.e., 5-10mg for donepezil, 16-24 mg for galantamine, 6-12mg for oral rivastigmine and 9.5mg for rivastigmine patch (the 13.3mg patch is also available and can be prescribed for selected cases). Ideally, the highest dose within the therapeutic range of each drug should be targeted, given the greater potential for clinical benefits. Figure 1 illustrates the dose regimen for drug therapy of AD dementia.

Figure 1
Overview of pharmacological treatment for AD dementia.

1In case of improvement or stabilization, the highest dose of rivastigmine patch can be postponed; 2memantine is indicated for treating moderate or severe AD dementia.


Common AEs consist of nausea, vomiting, diarrhea, headache and insomnia, and are less likely to occur if medication is taken after a meal. Donepezil should be administered at bedtime; in case of insomnia or nightmares, it can be taken in the morning. Less common, but more serious AEs are bradyarrhythmia and syncope. Treatment effects and duration vary from person to person, and any positive effect may be lost over time. No research has confirmed the superiority of one ChEI over the others, despite some specificities on the mechanisms of action and frequency/types of AEs. Switching to another ChEI is encouraged, particularly when no clinical benefit is observed after a few months of follow-up or in case of AEs that do not resolve. With progression to moderate dementia, the association with memantine is indicated55. Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. Treatment of Alzheimer's Disease: recommendations and suggestions of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. doi:10.1590/S0004-282X2005000600035.
https://doi.org/10.1590/S0004-282X200500...
),(99. Ministério da Saúde (BR). Secretaria de Atenção à Saúde. Portaria Conjunta nº 13, de 28 de novembro de 2017. Aprova o Protocolo Clínico e Diretrizes Terapêuticas da Doença de Alzheimer. Diário Oficial da União. 8 dez 2017;1:201.),(1010. Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, et al. Early diagnosis and treatment of Alzheimer's disease: New definitions and challenges. Braz J Psychiatry. 2020;42(4):431-41. doi:10.1590/1516-4446-2019-0735.
https://doi.org/10.1590/1516-4446-2019-0...
. Memantine is usually well tolerated; the most common AEs are drowsiness, dizziness and headache55. Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. Treatment of Alzheimer's Disease: recommendations and suggestions of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. doi:10.1590/S0004-282X2005000600035.
https://doi.org/10.1590/S0004-282X200500...
),(77. Small G, Bullock R. Defining optimal treatment with cholinesterase inhibitors in Alzheimer's disease. Alzheimers Dement. 2011;7(2):177-84. doi:10.1016/j.jalz.2010.03.016.
https://doi.org/10.1016/j.jalz.2010.03.0...
.

Practical issues for prescribing and discontinuing ChEI and memantine

  • ChEI increase vagal tone and are contraindicated in patients with baseline bradycardia or known cardiac conduction system disorder (i.e., atrioventricular block > 1st degree). Caution is required when any of the three ChEI are used in combination with drugs that induce bradycardia or alter atrioventricular conduction (e.g., beta blockers or calcium channel blockers);

  • Caution is required when prescribing ChEI to individuals with chronic obstructive pulmonary disease or asthma;

  • Discontinuation or switching (in the case of ChEI) shall be considered when there is no clinical benefit or in case of relevant AEs;

  • Avoid abrupt discontinuation of ChEI or memantine;

  • In the case of AEs with higher doses, dose reduction may be considered, noting whether there is a decrease in AEs as well as changes in cognitive, functional, or neuropsychiatric symptoms;

  • Discontinue medication in the absence of clinical benefits or if AEs persist.

Pharmacological treatment of neuropsychiatric symptoms

Neuropsychiatric symptoms may precede the cognitive symptoms in AD, becoming more common at the second half of the mild dementia stage, increasing along the moderate stage and usually decreasing at severe stage.

Clinical trials specifically addressing neuropsychiatric symptoms in AD have been disappointing. The benefits of ChEI tend to favor specific neuropsychiatric symptoms, such as depression, anxiety, dysphoria, and apathy1111. Vale FAC, Corrêa Neto Y, Bertolucci PHF, Machado JCB, Silva DJ, Allam N, et al. Treatment of Alzheimer's disease in Brazil: II. Behavioral and psychological symptoms of dementia. Dement Neuropsychol. 2011;5(3):189-97. doi:10.1590/S1980-57642011DN05030006.
https://doi.org/10.1590/S1980-57642011DN...
. For depression, which may be present in more than half of AD patients, there is some evidence for the indication of sertraline or mirtazapine as the best antidepressant options1212. He Y, Li H, Huang J, Huang S, Bai Y, Li Y, et al. Efficacy of antidepressant drugs in the treatment of depression in Alzheimer disease patients: A systematic review and network meta-analysis. J Psychopharmacol. 2021;35(8):901-9. doi:10.1177/02698811211030181.
https://doi.org/10.1177/0269881121103018...
. A recent clinical trial, however, observed a trend to increased mortality with mirtazapine used to treat agitation in AD dementia1313. Banerjee S, High J, Stirling S, Shepstone L, Swart AM, Telling T, et al. Study of mirtazapine for agitated behaviours in dementia (SYMBAD): a randomised, double-blind, placebo-controlled trial. Lancet. 2021;398(10310):1487-97. doi:10.1016/S0140-6736(21)01210-1.
https://doi.org/10.1016/S0140-6736(21)01...
.

Aggressiveness is a common neuropsychiatric symptom which may contribute to agitation and increased caregiver burden. Treatment encompasses both pharmacological and non-pharmacological interventions (see next session). Specific AD drugs (i.e., ChEI and memantine) might have modest but significant effects on these symptoms and even if there is a good initial effect, it may be lost after some time. Given the absence of approved pharmacological treatment for neuropsychiatric symptoms in AD dementia, a sequence of drugs may be considered for treatment of agitation and aggressiveness1414. Davies SJ, Burhan AM, Kim D, Gerretsen P, Graff-Guerrero A, Woo VL, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol. 2018;32(5):509-23. doi:10.1177/0269881117744996.
https://doi.org/10.1177/0269881117744996...
. According to this algorithm1414. Davies SJ, Burhan AM, Kim D, Gerretsen P, Graff-Guerrero A, Woo VL, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol. 2018;32(5):509-23. doi:10.1177/0269881117744996.
https://doi.org/10.1177/0269881117744996...
, first-line treatment would be atypical antipsychotics, the first choice being risperidone, followed by aripiprazole or quetiapine. If these drugs fail or are offset by AEs, carbamazepine may be an option. In case of a new failure the next step would be citalopram, followed by gabapentin and prazosin. Citalopram is a good therapeutic option for agitation. Benzodiazepines must be avoided in dementia due to AD.

Another common neuropsychiatric symptom is sleep disorder. The first step to a successful treatment of sleep disorder in AD is to ask why and how it occurs. A sleep disorder may be the consequence of a sleep wake cycle inversion, and in this case sleep hygiene and non-pharmacological interventions may be helpful. For pain or another discomfort, the cause should be addressed; issues related to sundowning, fragmented sleep or difficulty in starting sleep should be addressed. The sleep may be interrupted by delusions and hallucinations, when an antipsychotic, particularly with sedative effects (e.g., quetiapine) may be useful. There is no evidence that hypnotics or mirtazapine have positive effects. Trazodone might improve sleep, although the effects are usually modest1515. Camargos EF, Louzada LL, Quintas JL, Naves JO, Louzada FM, Nóbrega OT. Trazodone improves sleep parameters in Alzheimer disease patients: a randomized, double-blind, and placebo-controlled study. Am J Geriatr Psychiatry. 2014;22(12):1565-74. doi:10.1016/j.jagp.2013.12.174.
https://doi.org/10.1016/j.jagp.2013.12.1...
. A recent Brazilian study has shown that short-term use (i.e., 14 days) of zopiclone or zolpidem may be clinically helpful in treating insomnia in AD dementia1616. Louzada LL, Machado FV, Quintas JL, Ribeiro GA, Silva MV, Mendonça-Silva DL, et al. The efficacy and safety of zolpidem and zopiclone to treat insomnia in Alzheimer's disease: a randomized, triple-blind, placebo-controlled trial. Neuropsychopharmacology. 2022;47(2):570-9. doi:10.1038/s41386-021-01191-3.
https://doi.org/10.1038/s41386-021-01191...
.

Delusions and hallucinations may be present in up to one third of AD patients and indicate poorer prognosis, with increased functional impairment and higher mortality. Treatment of these neuropsychiatric symptoms includes avoiding typical antipsychotics, due to AEs and cognitive worsening. There is evidence of a modest effect of ChEI, but often there is a need of using antipsychotics, and in this case the evidence is stronger for risperidone and olanzapine, while for quetiapine the evidence is scanter. Studies with brexpiprazole are ongoing, but evidence of efficacy and safety has not been definitively established to date1717. Caraci F, Santagati M, Caruso G, Cannavò D, Leggio GM, Salomone S, et al. New antipsychotic drugs for the treatment of agitation and psychosis in Alzheimer's disease: focus on brexpiprazole and pimavanserin. F1000Res. 2020;9:F1000 Faculty Rev-686. doi:10.12688/f1000research.22662.1.
https://doi.org/10.12688/f1000research.2...
. Although some studies suggest that cannabinoids may have potential positive effects in neurodegenerative diseases, there is no current evidence to support their clinical use in dementia1818. Brucki SMD, Adoni T, Almeida CMO, Andrade DC, Anghinah R, Barbosa LM, et al. Cannabinoids in Neurology - Position paper from Scientific Departments from Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2021;79(4):354-69. doi:10.1590/0004-282X-ANP-2020-0432.
https://doi.org/10.1590/0004-282X-ANP-20...
.

Pharmacological treatment for AD-related conditions

AD may have associated complications. Parkinsonism may be present in 9% to 70%, depending on the disease stage. There are differences with typical PD, with tremor being mild or absent, and usually presenting symmetrical bradykinesia and rigidity from the start. Levodopa may be tried, but response is less likely than in PD.

Between 10% and 20% of AD patients will have at least one seizure. Newer anticonvulsants (e.g., lamotrigine or levetiracetam) are first-line treatments for epilepsy in PwD1919. Frederiksen KS, Cooper C, Frisoni GB, Frölich L, Georges J, Kramberger MG, et al. A European Academy of Neurology guideline on medical management issues in dementia. Eur J Neurol. 2020;27(10):1805-20. doi:10.1111/ene.14412.
https://doi.org/10.1111/ene.14412...
.

Non-pharmacological treatments for AD: cognitive symptoms

Cognitive stimulation therapy (CST)

Recommended treatment according to the National Institute for Health and Clinical Excellence of the United Kingdom2020. York Health Economics Consortium. NICE: Overview of Systematic Reviews of Non-pharmacological Interventions for Dementia [Internet]. Final Report. Heslington: University of York; 2017 [citado em 22 ago 2022]. Disponível em: https://www.nice.org.uk/guidance/ng97/evidence/appendix-o-yhec-report-pdf-174697045530
https://www.nice.org.uk/guidance/ng97/ev...
to stimulate language, memory, executive functions; approved by the Alzheimer’s Disease International and used in many countries2121. Aguirre E, Spector A, Orrell M. Guidelines for adapting cognitive stimulation therapy to other cultures. Clin Interv Aging. 2014;9:1003-7. doi:10.2147/CIA.S61849.
https://doi.org/10.2147/CIA.S61849...
. CST was shown to improve cognition, QoL, neuropsychiatric symptoms in PwD from LMIC2222. Meyer C, O'Keefe F. Non-pharmacological interventions for people with dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. doi:10.1177/1471301218813234.
https://doi.org/10.1177/1471301218813234...
),(2323. Stoner CR, Lakshminarayanan M, Durgante H, Spector A. Psychosocial interventions for dementia in low- and middle-income countries (LMICs): a systematic review of effectiveness and implementation readiness. Aging Ment Health. 2021;25(3):408-19. doi:10.1080/13607863.2019.1695742.
https://doi.org/10.1080/13607863.2019.16...
. CST and its implementation manual have been adapted for use in Brazil2424. Bertrand E, Naylor R, Laks J, Marinho V, Spector A, Mograbi DC. Cognitive stimulation therapy for Brazilian people with dementia: examination of implementation' issues and cultural adaptation. Aging Ment Health. 2019;23(10):1400-4. doi:10.1080/13607863.2018.1488944.
https://doi.org/10.1080/13607863.2018.14...
and the intervention was found to be feasible and beneficial to improve mood in PwD in a recent clinical trial2525. Marinho V, Bertrand E, Naylor R, Bomilcar I, Laks J, Spector A, et al. Cognitive stimulation therapy for people with dementia in Brazil (CST-Brasil): Results from a single blind randomized controlled trial. Int J Geriatr Psychiatry. 2021;36(2):286-93. doi:10.1002/gps.5421.
https://doi.org/10.1002/gps.5421...
.

Multicomponent/multidisciplinary cognitive rehabilitation program (MCRP)

MCRP includes cognitive rehabilitation, stimulation and computer-assisted therapy, physical training, physiotherapy, reading and games. This program may decrease depressive symptoms and improve QoL indicators of PwD/caregivers. However, MCRP effects on cognition are questionable2323. Stoner CR, Lakshminarayanan M, Durgante H, Spector A. Psychosocial interventions for dementia in low- and middle-income countries (LMICs): a systematic review of effectiveness and implementation readiness. Aging Ment Health. 2021;25(3):408-19. doi:10.1080/13607863.2019.1695742.
https://doi.org/10.1080/13607863.2019.16...
),(2626. Madureira BG, Pereira MG, Avelino PR, Costa HS, Menezes KKP. Efeitos de programas de reabilitação multidisciplinar no tratamento de pacientes com doença de Alzheimer: uma revisão sistemática. Cad Saude Coletiva. 2018;26(2):222-32. doi:10.1590/1414-462x201800020446.
https://doi.org/10.1590/1414-462x2018000...
.

Reality orientation (RO)

Neurosensory stimulation to reorient daily activities may improve cognition and adherence to pharmacological treatment in mild-moderate AD dementia2727. Cammisuli DM, Danti S, Bosinelli F, Cipriani G. Non-pharmacological interventions for people with Alzheimer's disease: A critical review of the scientific literature from the last ten years. Eur Geriatr Med. 2016,7(1):57-64. doi:10.1016/j.eurger.2016.01.002.
https://doi.org/10.1016/j.eurger.2016.01...
),(2828. Carrion C, Folkvord F, Anastasiadou D, Aymerich M. Cognitive therapy for dementia patients: a systematic review. Dement Geriatr Cogn Disord. 2018;46(1-2):1-26. doi:10.1159/000490851.
https://doi.org/10.1159/000490851...
. Results for neuropsychiatric symptoms are inconclusive2929. Chiu H-Y, Chen P-Y, Chen Y-T, Huang H-C. Reality orientation therapy benefits cognition in older people with dementia: A meta-analysis. Int J Nurs Stud. 2018;86:20-8. doi:10.1016/j.ijnurstu.2018.06.008.
https://doi.org/10.1016/j.ijnurstu.2018....
.

Non-pharmacological treatments for AD: neuropsychiatric symptoms

To increase the chance of success with a non-pharmacological treatment it is essential to adopt an individualized approach that considers:

  • When and how the neuropsychiatric symptom started? (Was there a trigger?)

  • How did it evolve? (Are there associated symptoms?)

  • How did it end?

  • What are the associated worsening and improving factors?

Psychosocial/psychoeducational interventions

These are group or person-centered approaches to inform the management and care of dementia, including emotional and social stimulation of PwD/caregivers. Face-to-face or technology-based multicomponent psychoeducational and Cognitive-Behavioral Therapy approaches (cognitive reframing, self-monitoring, mood management techniques, mindfulness-based training) may improve neuropsychiatric symptoms (agitation, restlessness, anxiety, sleep disturbances), delay the progression of cognitive impairment, and benefit caregiver confidence, self-efficacy, burden, depression, stress levels, and QoL of the dyad3030. Alves GS, Casali ME, Veras AB, Carrilho CG, Bruno Costa E, Rodrigues VM, et al. A systematic review of home-setting psychoeducation interventions for behavioral changes in dementia: some lessons for the COVID-19 Pandemic and post-pandemic assistance. Front Psychiatry. 2020;11:577871. doi:10.3389/fpsyt.2020.577871.
https://doi.org/10.3389/fpsyt.2020.57787...
)- (3333. Shim M, Tilley JL, Im S, Price K, Gonzalez A. A systematic review of mindfulness-based interventions for patients with mild cognitive impairment or dementia and caregivers. J Geriatr Psychiatry Neurol. 2021;34(6):528-54. doi:10.1177/0891988720957104.
https://doi.org/10.1177/0891988720957104...
. Positive Psychology practices are recommended for mental health promotion of PwD/caregivers according to the Alzheimer’s Association Psychosocial Measurement Workgroup3333. Shim M, Tilley JL, Im S, Price K, Gonzalez A. A systematic review of mindfulness-based interventions for patients with mild cognitive impairment or dementia and caregivers. J Geriatr Psychiatry Neurol. 2021;34(6):528-54. doi:10.1177/0891988720957104.
https://doi.org/10.1177/0891988720957104...
and the European INTERDEM Social Health Taskforce3434. Gaugler JE, Bain LJ, Mitchell L, Finlay J, Fazio S, Jutkowitz E & Alzheimer's Association Psychosocial Measurement Workgroup. Reconsidering frameworks of Alzheimer's dementia when assessing psychosocial outcomes. Alzheimers Dement. 2019;5:388-97. https://doi.org/10.1016/j.trci.2019.02.008.
https://doi.org/https://doi.org/10.1016/...
),(3535. Dröes RM, Chattat R, Diaz A, Gove D, Graff M, Murphy K. Social health and dementia: a European consensus on the operationalization of the concept and directions for research and practice. Aging Ment Health. 2017;21(1):4-17. doi:10.1080/13607863.2016.1254596.
https://doi.org/10.1080/13607863.2016.12...
.

Reminiscence therapy (RT)

RT is a group-based intervention to recall autobiographical memories. It is used for mild-moderate dementia with modest evidence to improve depressive symptoms, mood, cognition2222. Meyer C, O'Keefe F. Non-pharmacological interventions for people with dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. doi:10.1177/1471301218813234.
https://doi.org/10.1177/1471301218813234...
, and well-being3636. Oksnebjerg L, Diaz-Ponce A, Gove D, Moniz-Cook E, Mountain G, Chattat R, et al. Towards capturing meaningful outcomes for people with dementia in psychosocial intervention research: a pan-European consultation. Health Expect. 2018;21(6):1056-65. doi:10.1111/hex.12799.
https://doi.org/10.1111/hex.12799...
. Lacking theoretical basis and methodological systematization make RT effects questionable3737. Arroyo-Anlló EM, Díaz-Marta JP, Chamorro Sánchez J. Técnicas de rehabilitación neuropsicológica en demencias: hacia la ciber-rehabilitación neuropsicológica. Pensam Psicol. 2012;10(1):107-27..

Music therapy (MT)/dancing

Significant positive effects were found of MT on memory3838. Moreira SV, Reis Justi FR, Moreira M. Can musical intervention improve memory in Alzheimer's patients? Evidence from a systematic review. Dement Neuropsychol. 2018;12(2):133-42. doi:10.1590/1980-57642018dn12-020005.
https://doi.org/10.1590/1980-57642018dn1...
, emotional/mood2222. Meyer C, O'Keefe F. Non-pharmacological interventions for people with dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. doi:10.1177/1471301218813234.
https://doi.org/10.1177/1471301218813234...
and neuropsychiatric symptoms2727. Cammisuli DM, Danti S, Bosinelli F, Cipriani G. Non-pharmacological interventions for people with Alzheimer's disease: A critical review of the scientific literature from the last ten years. Eur Geriatr Med. 2016,7(1):57-64. doi:10.1016/j.eurger.2016.01.002.
https://doi.org/10.1016/j.eurger.2016.01...
. Dancing may also lead to beneficial effects on cognition and psychical health2020. York Health Economics Consortium. NICE: Overview of Systematic Reviews of Non-pharmacological Interventions for Dementia [Internet]. Final Report. Heslington: University of York; 2017 [citado em 22 ago 2022]. Disponível em: https://www.nice.org.uk/guidance/ng97/evidence/appendix-o-yhec-report-pdf-174697045530
https://www.nice.org.uk/guidance/ng97/ev...
.

Physical-related/lifestyle-related interventions

Repetitive transcranial magnetic stimulation (rTMS) and acupuncture seem to improve cognitive functions, whereas acupuncture, exercise and light therapy presented potential to enhance functional performance2222. Meyer C, O'Keefe F. Non-pharmacological interventions for people with dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. doi:10.1177/1471301218813234.
https://doi.org/10.1177/1471301218813234...
and cognition3939. Jia RX, Liang JH, Xu Y, Wang YQ. Effects of physical activity and exercise on the cognitive function of patients with Alzheimer disease: a meta-analysis. BMC Geriatr. 2019;19(1):181. doi:10.1186/s12877-019-1175-2.
https://doi.org/10.1186/s12877-019-1175-...
),(4040. Wang LY, Pei J, Zhan YJ, Cai YW. Overview of meta-analyses of five non-pharmacological interventions for Alzheimer's Disease. Front Aging Neurosci. 2020;12:594432. doi:10.3389/fnagi.2020.594432.
https://doi.org/10.3389/fnagi.2020.59443...
. Long-term exercise improves blood flow, hippocampal volume, neurogenesis, also neuropsychiatric symptoms4141. Meng Q, Lin MS, Tzeng IS. Relationship between exercise and Alzheimer's disease: a narrative literature review. Front Neurosci. 2020;14:131. doi:10.3389/fnins.2020.00131.
https://doi.org/10.3389/fnins.2020.00131...
. Occupational therapy (error-reduction techniques) (4242. Smallfield S, Heckenlaible C. Effectiveness of occupational therapy interventions to enhance occupational performance for adults with Alzheimer's Disease and related major neurocognitive disorders: a systematic review. Am J Occup Ther. 2017;71(5):7105180010p1-p9. doi:10.5014/ajot.2017.024752.
https://doi.org/10.5014/ajot.2017.024752...
and physiotherapy4343. Marques CLS, Borgato MH, Moura Neto E, Bazan R, Luvizutto GJ. Physical therapy in patients with Alzheimer's disease: a systematic review of randomized controlled clinical trials. Fisioter Pesqui. 2019;26(3):311-21. doi:10.1590/1809-2950/18037226032019.
https://doi.org/10.1590/1809-2950/180372...
),(4444. Zhu XC, Yu Y, Wang HF, Jiang T, Cao L, Wang C, et al. Physiotherapy intervention in Alzheimer's disease: systematic review and meta-analysis. J Alzheimers Dis. 2015;44(1):163-74. doi:10.3233/JAD-141377.
https://doi.org/10.3233/JAD-141377...
tend to enhance functioning and delay physical decline; results are inconclusive for improvement in cognition. Early interventions targeting lifestyle modifiable risk factors (arterial hypertension, diabetes, obesity, physical inactivity, high alcohol consumption, smoking, social isolation), clinical conditions (hearing loss and depression), and poor nutrient intake, may work as protective measures for reducing AD risk11. Mukadam N, Sommerlad A, Huntley J, Livingston G. Population attributable fractions for risk factors for dementia in low-income and middle-income countries: an analysis using cross-sectional survey data. Lancet Glob Health. 2019;7(5):e596-603. doi:10.1016/S2214-109X(19)30074-9.
https://doi.org/10.1016/S2214-109X(19)30...
),(1010. Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, et al. Early diagnosis and treatment of Alzheimer's disease: New definitions and challenges. Braz J Psychiatry. 2020;42(4):431-41. doi:10.1590/1516-4446-2019-0735.
https://doi.org/10.1590/1516-4446-2019-0...
),(4545. Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, et al. World-Wide FINGERS Network: A global approach to risk reduction and prevention of dementia. Alzheimers Dement. 2020;16(7):1078-94. doi:10.1002/alz.12123.
https://doi.org/10.1002/alz.12123...
),(4646. Moreira SC, Jansen AK, Silva FM. Dietary interventions and cognition of Alzheimer's disease patients: a systematic review of randomized controlled trial. Dement Neuropsychol. 2020;14(3):258-82. doi:10.1590/1980-57642020dn14-030008.
https://doi.org/10.1590/1980-57642020dn1...
.

TREATMENT OF VASCULAR COGNITIVE IMPAIRMENT

The general principles of approach and treatment for VCI are the same as for dementia due to AD. They encompass the treatment of comorbidities, including neuropsychiatric symptoms, providing information and support to PwD and caregivers, and maintaining independence.

Primary and secondary prevention

The multidisciplinary team should focus primarily on controlling the main risk factors for new cerebrovascular events and cognitive impairment in VCI. Although we recognize risk factors for post-stroke dementia, such as low education and diabetes mellitus4747. Pendlebury ST, Rothwell PM. Prevalence, incidence, and factors associated with pre-stroke and post-stroke dementia: a systematic review and meta-analysis. Lancet Neurol. 2009;8(11):1006-18. doi:10.1016/S1474-4422(09)70236-4.
https://doi.org/10.1016/S1474-4422(09)70...
, factors for stroke itself have long been known and controllable, including smoking, arterial hypertension, diabetes mellitus and dyslipidemia (notably high LDL-cholesterol levels) (4848. Boehme AK, Esenwa C, Elkind MSV. Stroke risk factors, genetics, and prevention. Circ Res. 2017;120(3):472-95. doi:10.1161/CIRCRESAHA.116.308398.
https://doi.org/10.1161/CIRCRESAHA.116.3...
.

However, in recent decades several studies have proposed measuring the weight of these risk factors on the appearance of new strokes, on post-stroke cognition and functionality, often with conflicting results. Based on these observations we recommend:

  • Blood pressure control: There is strong evidence that increased blood pressure levels are associated with stroke4848. Boehme AK, Esenwa C, Elkind MSV. Stroke risk factors, genetics, and prevention. Circ Res. 2017;120(3):472-95. doi:10.1161/CIRCRESAHA.116.308398.
    https://doi.org/10.1161/CIRCRESAHA.116.3...
    . There are still no studies that have confirmed the impact of blood pressure control on worsening cognitive impairment. Intensive blood pressure control (systolic pressure of 120 versus 140 mmHg) does not show a reduction in the incidence of dementia (all etiologies). In frail older adults, with symptoms of orthostatic hypotension and increased risk of falls, individualize blood pressure targets to avoid symptoms of hypotension;

  • Glycemic control: There is strong evidence that increased blood glucose levels are associated with stroke4848. Boehme AK, Esenwa C, Elkind MSV. Stroke risk factors, genetics, and prevention. Circ Res. 2017;120(3):472-95. doi:10.1161/CIRCRESAHA.116.308398.
    https://doi.org/10.1161/CIRCRESAHA.116.3...
    . However, there is no evidence that strict glycemic control in older adults prevents cardiovascular events. Studies have shown that tight blood glucose control in older adults was associated with greater frailty and mortality, with no benefit in the evolution of dementia4949. ADVANCE Collaborative Group; Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358(24):2560-72. doi:10.1056/NEJMoa0802987.
    https://doi.org/10.1056/NEJMoa0802987...
    ),(5050. Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, et al. Effects of intensive glucose lowering on brain structure and function in people with type 2 diabetes (ACCORD MIND): a randomised open-label substudy. Lancet Neurol. 2011;10(11):969-77. doi:10.1016/S1474-4422(11)70188-0.
    https://doi.org/10.1016/S1474-4422(11)70...
    . Acceptable fasting glycemic values in individuals with VCI should be around up to 150 mg/dL and postprandial < 180 mg/dL, as well as glycated hemoglobin targets should be less stringent (e.g., < 8%)5151. Costa e Forti A, Pires AC, Pittito BA, Gerchman F, Oliveira JEP, Zajdenverg L, et al. Diretrizes da Sociedade Brasileira de Diabetes: 2019-2020. São Paulo: Clannad; 2019.;

  • Antiplatelet agents: There is strong evidence to support using these agents for secondary prevention of non-embolic stroke, either acetylsalicylic acid (81 to 100mg/day) or clopidogrel (75mg/day) (5252. Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, et al. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack. Stroke. 2014;45(7):2160-236. doi:10.1161/STR.0000000000000024.
    https://doi.org/10.1161/STR.000000000000...
    . In cases of VCI without evidence of previous stroke, therapy should be individualized5353. Smith EE, Saposnik G, Biessels GJ, Doubal FN, Fornage M, Gorelick PB, et al. Prevention of stroke in patients with silent cerebrovascular disease: a scientific statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2017;48(2):e44-71. doi:10.1161/STR.0000000000000116.
    https://doi.org/10.1161/STR.000000000000...
    , especially in individuals with significant risk of falls. In recent years, no studies have been published providing support for the benefits of antiaggregants in the evolution of VCI5454. Davis KAS, Bishara D, Molokhia M, Mueller C, Perera G, Stewart RJ. Aspirin in people with dementia, long-term benefits, and harms: a systematic review. Eur J Clin Pharmacol. 2021;77(7):943-54. doi:10.1007/s00228-021-03089-x.
    https://doi.org/10.1007/s00228-021-03089...
    ;

  • Statins: Statins are of interest in the secondary prevention of cerebrovascular disease due to both their lipid-lowering and pleiotropic effects on vascular function, combining to inhibit atherosclerosis. There is good evidence that statins given late in life to people at risk for vascular disease do not prevent cognitive decline or dementia5555. McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev. 2014;(7):CD007514. doi:10.1002/14651858.CD007514.pub3.
    https://doi.org/10.1002/14651858.CD00751...
    . For individuals with VCI and a history of ischemic (non-embolic) stroke, statin use should follow secondary prevention recommendations with individual risk analysis. The use of statins in older adults and subjects with vascular risk factors is not recommended for the sole purpose of primary prevention or treatment of dementia;

  • Multidisciplinary interventions: In a systematic review of 15 prospective studies with over 30,000 individuals without dementia, involvement in at least one physical activity of minimal to moderate intensity was associated with a 35% reduction in the relative risk of cognitive decline in one to 12 years of follow-up5656. Sofi F, Valecchi D, Bacci D, Abbate R, Gensini GF, Casini A, et al. Physical activity and risk of cognitive decline: a meta-analysis of prospective studies. J Int Med. 2011;269(1):107-17. doi:10.1111/j.1365-2796.2010.02281.x.
    https://doi.org/10.1111/j.1365-2796.2010...
    . In another study with 639 older adults without disability and with cerebral white matter changes, physical activity reduced the risk of cognitive and functional deficits, regardless of the severity of white matter changes or other risk factors5757. Verdelho A, Madureira S, Ferro JM, Baezner H, Blahak C, Poggesi A, et al. Physical activity prevents progression for cognitive impairment and vascular dementia: results from the LADIS (Leukoaraiosis and Disability) study. Stroke. 2012;43(12):3331-5. doi:10.1161/STROKEAHA.112.661793.
    https://doi.org/10.1161/STROKEAHA.112.66...
    . Hence, practice of physical activity at mild to moderate intensity should be strongly recommended to all individuals at the primary care level as a means of primary and secondary prevention of VCI. On the other hand, evidence that physical rehabilitation reduces post-stroke cognitive loss is still insufficient5858. Gillespie DC, Bowen A, Chung CS, Cockburn J, Knapp P, Pollock A. Rehabilitation for post-stroke cognitive impairment: an overview of recommendations arising from systematic reviews of current evidence. Clin Rehabil. 2015;29(2):120-8. doi:10.1177/0269215514538982.
    https://doi.org/10.1177/0269215514538982...
    ;

  • Diet and supplements: A balanced diet, rich in vegetables and fruits, impacts many organ systems. One recent systematic review with metanalysis has shown that high adherence to the Mediterranean diet reduced the risk of global cognitive decline in non-demented older adults5959. Coelho-Júnior HJ, Trichopoulou A, Panza F. Cross-sectional and longitudinal associations between adherence to Mediterranean diet with physical performance and cognitive function in older adults: A systematic review and meta-analysis. Ageing Res Rev. 2021;70:101395. doi:10.1016/j.arr.2021.101395.
    https://doi.org/10.1016/j.arr.2021.10139...
    . The use of vitamin and mineral supplements should only be indicated in cases with insufficiency. It is not recommended to use vitamin supplements for the prevention or treatment of VCI.

Pharmacological treatment

ChEI and memantine

The pharmacological treatment commonly prescribed in VCI, particularly vascular dementia, is mainly based on ChEI and memantine. These medications are routinely used in clinical practice to treat cognitive decline, functionality changes and neuropsychiatric symptoms. However, randomized controlled clinical trials have shown limited efficacy of these drugs.

Randomized, placebo-controlled studies demonstrate little efficacy of ChEI in pure vascular dementia6060. Brucki SMD, Ferraz AC, Freitas GR, Massaro AR, Radanovic M, Schultz RR. Treatment of vascular dementia. Recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol. 2011;5(4):275-87. doi:10.1590/S1980-57642011DN05040005.
https://doi.org/10.1590/S1980-57642011DN...
),(6161. Smith EE, Barber P, Field TS, Ganesh A, Hachinski V, Hogan DB, et al. Canadian Consensus Conference on Diagnosis and Treatment of Dementia (CCCDTD) 5: Guidelines for management of vascular cognitive impairment. Alzheimers Dement (N Y). 2020;6(1):e12056. doi:10.1002/trc2.12056.
https://doi.org/10.1002/trc2.12056...
. There is a statistically significant but clinically slight cognitive benefit for cognitive functions, mainly processing speed and executive functions6262. Dichgans M, Markus HS, Salloway S, Verkkoniemi A, Moline M, Wang Q, et al. Donepezil in patients with subcortical vascular cognitive impairment: a randomised double-blind trial in CADASIL. Lancet Neurol. 2008;7(4):310-8. doi:10.1016/S1474-4422(08)70046-2.
https://doi.org/10.1016/S1474-4422(08)70...
. In general, there is a slight attenuation of cognitive decline compared to placebo6161. Smith EE, Barber P, Field TS, Ganesh A, Hachinski V, Hogan DB, et al. Canadian Consensus Conference on Diagnosis and Treatment of Dementia (CCCDTD) 5: Guidelines for management of vascular cognitive impairment. Alzheimers Dement (N Y). 2020;6(1):e12056. doi:10.1002/trc2.12056.
https://doi.org/10.1002/trc2.12056...
. Some studies report favorable results regarding functional performance, but the gain is not consistent. Improvement of neuropsychiatric symptoms, when present, is also limited. On the other hand, agitation has been observed as an AE of ChEI and memantine6363. Black S, Román GC, Geldmacher DS, Salloway S, Hecker J, Burns A, et al. Efficacy and tolerability of donepezil in vascular dementia: positive results of a 24-week, multicenter, international, randomized, placebo-controlled clinical trial. Stroke. 2003;34(10):2323-30. doi:10.1161/01.STR.0000091396.95360.E1.
https://doi.org/10.1161/01.STR.000009139...
),(6464. Orgogozo JM, Rigaud AS, Sto¨ffler A, Mo¨bius HJ, Forette F. Efficacy and safety of memantine in patients with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke. 2002;33(7):1834-9. doi:10.1161/01.str.0000020094.08790.49.
https://doi.org/10.1161/01.str.000002009...
.

Favorable results of ChEI seem more evident in mixed dementia (vascular + AD) compared to ‘pure’ vascular dementia6565. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359(9314):1283-90. doi:10.1016/S0140-6736(02)08267-3.
https://doi.org/10.1016/S0140-6736(02)08...
. Galantamine is approved in Brazil for treatment of mild to moderate AD dementia with concomitant significant cerebrovascular disease and has shown positive effects on cognitive speed and QoL of individuals with mixed dementia in a controlled clinical trial in the country6666. Caramelli P, Laks J, Palmini AL, Nitrini R, Chaves ML, Forlenza OV, et al. Effects of galantamine and galantamine combined with nimodipine on cognitive speed and quality of life in mixed dementia: a 24-week, randomized, placebo-controlled exploratory trial (the REMIX study). Arq Neuropsiquiatr. 2014;72(6):411-7. doi:10.1590/0004-282x20140055.
https://doi.org/10.1590/0004-282x2014005...
. Possible benefits of ChEI in mixed dementia, although limited, are attributed to the reduction of cholinergic depletion caused by concomitant AD6060. Brucki SMD, Ferraz AC, Freitas GR, Massaro AR, Radanovic M, Schultz RR. Treatment of vascular dementia. Recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol. 2011;5(4):275-87. doi:10.1590/S1980-57642011DN05040005.
https://doi.org/10.1590/S1980-57642011DN...
),(6565. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359(9314):1283-90. doi:10.1016/S0140-6736(02)08267-3.
https://doi.org/10.1016/S0140-6736(02)08...
),(6666. Caramelli P, Laks J, Palmini AL, Nitrini R, Chaves ML, Forlenza OV, et al. Effects of galantamine and galantamine combined with nimodipine on cognitive speed and quality of life in mixed dementia: a 24-week, randomized, placebo-controlled exploratory trial (the REMIX study). Arq Neuropsiquiatr. 2014;72(6):411-7. doi:10.1590/0004-282x20140055.
https://doi.org/10.1590/0004-282x2014005...
. There is insufficient clarity regarding the differences in efficacy of ChEI, as well as memantine, between multiple infarcts and subcortical vascular dementia6464. Orgogozo JM, Rigaud AS, Sto¨ffler A, Mo¨bius HJ, Forette F. Efficacy and safety of memantine in patients with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke. 2002;33(7):1834-9. doi:10.1161/01.str.0000020094.08790.49.
https://doi.org/10.1161/01.str.000002009...
),(6767. Ballard C, Sauter M, Scheltens P, He Y, Barkhof F, Straaten EC, et al. Efficacy, safety and tolerability of rivastigmine capsules in patients with probable vascular dementia: the VantagE study. Curr Med Res Opin. 2008;24(9):2561-74. doi:10.1185/03007990802328142.
https://doi.org/10.1185/0300799080232814...
)-(6969. Moretti R, Torre P, Antonello RM, Cazzato G, Pizzolato G. Different responses to rivastigmine in subcortical vascular dementia and multi-infarct dementia. Am J Alzheimers Dis Other Demen. 2008;23(2):167-76. doi:10.1177/1533317507312558.
https://doi.org/10.1177/1533317507312558...
. Initiation of ChEI treatment in patients with severe dementia is not recommended.

Among the AEs of ChEI already described above, bradycardia and arterial hypotension are the main conditions associated that require special attention in patients with vascular dementia6262. Dichgans M, Markus HS, Salloway S, Verkkoniemi A, Moline M, Wang Q, et al. Donepezil in patients with subcortical vascular cognitive impairment: a randomised double-blind trial in CADASIL. Lancet Neurol. 2008;7(4):310-8. doi:10.1016/S1474-4422(08)70046-2.
https://doi.org/10.1016/S1474-4422(08)70...
),(6565. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359(9314):1283-90. doi:10.1016/S0140-6736(02)08267-3.
https://doi.org/10.1016/S0140-6736(02)08...
),(6767. Ballard C, Sauter M, Scheltens P, He Y, Barkhof F, Straaten EC, et al. Efficacy, safety and tolerability of rivastigmine capsules in patients with probable vascular dementia: the VantagE study. Curr Med Res Opin. 2008;24(9):2561-74. doi:10.1185/03007990802328142.
https://doi.org/10.1185/0300799080232814...
)-(7171. Narasimhalu K, Effendy S, Sim C, Lee JM, Chen I, Hia SB, et al. A randomized controlled trial of rivastigmine in patients with cognitive impairment no dementia because of cerebrovascular disease. Acta Neurol Scand. 2010;121(4):217-24. doi:10.1111/j.1600-0404.2009.01263.x.
https://doi.org/10.1111/j.1600-0404.2009...
.

In current clinical practice, memantine is commonly added to ChEI in subjects with moderate or severe vascular dementia. However, the benefits of memantine are very limited or controversial for controlling cognitive and neuropsychiatric symptoms, as well as for slowing cognitive or functional decline6161. Smith EE, Barber P, Field TS, Ganesh A, Hachinski V, Hogan DB, et al. Canadian Consensus Conference on Diagnosis and Treatment of Dementia (CCCDTD) 5: Guidelines for management of vascular cognitive impairment. Alzheimers Dement (N Y). 2020;6(1):e12056. doi:10.1002/trc2.12056.
https://doi.org/10.1002/trc2.12056...
. For this reason, the drug has not been approved for treatment of vascular dementia. The few randomized, placebo-controlled studies report that memantine is well-tolerated and that drowsiness has been the AE of major concern7272. Wilcock G, Möbius HJ, Stöffler A; MMM 500 group. A double-blind, placebo-controlled multicentre study of memantine in mild to moderate vascular dementia (MMM500). Int Clin Psychopharmacol. 2002;17(6): 297-305. doi:10.1097/00004850-200211000-00005.
https://doi.org/10.1097/00004850-2002110...
.

Treatment of neuropsychiatric symptoms in VCI

Overall, the neuropsychiatric symptoms in VCI are like those observed in AD, and the treatment is similar1414. Davies SJ, Burhan AM, Kim D, Gerretsen P, Graff-Guerrero A, Woo VL, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol. 2018;32(5):509-23. doi:10.1177/0269881117744996.
https://doi.org/10.1177/0269881117744996...
),(7373. Reus VI, Fochtmann LJ, Eyler AE, Hilty DM, Horvitz-Lennon M, Jibson MD, et al. The American Psychiatric Association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173(5):543-6. doi:10.1176/appi.ajp.2015.173501.
https://doi.org/10.1176/appi.ajp.2015.17...
)-(7575. Oliveira LF, Camargos EF, Martini LLL, Machado FV, Novaes MRCG. Use of psychotropic agents to treat agitation and aggression in Brazilian patients with Alzheimer's disease: A naturalistic and multicenter study. Psychiatry Res. 2021;295:113591. doi:10.1016/j.psychres.2020.113591.
https://doi.org/10.1016/j.psychres.2020....
. The non-pharmacological interventions include occupational therapy, cognitive stimulation, art therapy, involvement in social interaction activities, physical activity, and psychological support for PwD and caregivers.

Regarding pharmacological interventions, general recommendations follow the commentaries made above (AD section), namely, to avoid benzodiazepines, to prescribe antipsychotics in low doses and for strictly necessary periods, antidepressants for depression, anxiety, panic and mild agitation, and anticonvulsants when indicated.

The pharmacological treatment of patients with vascular dementia should be regularly monitored for cardiovascular signs and symptoms.

TREATMENT OF FRONTOTEMPORAL DEMENTIA (BEHAVIORAL AND LANGUAGE VARIANTS)

Frontotemporal dementia (FTD) encompasses different clinical syndromes associated with progressive behavioral/personality deterioration or language disorders, and is considered the second most frequent cause of young-onset dementia7676. Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal dementia. Int Rev of Psychiatry. 2013;25(2):130-7. doi:10.3109/09540261.2013.776523.
https://doi.org/10.3109/09540261.2013.77...
.

Treatment of behavioral variant frontotemporal dementia

The clinical syndrome associated with behavioral and personality changes called behavioral variant (bvFTD) is the most common clinical presentation and specific diagnostic criteria are available for clinical use7777. Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain. 2011;134(Pt 9):2456-77. doi:10.1093/brain/awr179.
https://doi.org/10.1093/brain/awr179...
.

There are no approved treatments for bvFTD, either disease-modifying drugs or symptomatic treatments to ameliorate disturbing symptoms. Only a few small studies have shown benefits of symptomatic pharmacological approaches7878. Trieu C, Gossink F, Stek ML, Scheltens P, Pijnenburg YAL, Dols A. Effectiveness of pharmacological interventions for symptoms of behavioral variant frontotemporal dementia: a systematic review. Cogn Behav Neurol. 2020;33(1):1-15. doi:10.1097/WNN.0000000000000217.
https://doi.org/10.1097/WNN.000000000000...
.

Considering that there are no specific treatments, care should focus on managing behavioral symptoms and disability, and reducing caregiver burden that is high in bvFTD. In a recent Brazilian study including 20 bvFTD and 30 AD individuals, Lima-Silva et al. found out that bvFTD subjects presented higher levels of neuropsychiatric symptoms and that their caregivers experienced higher levels of distress than AD caregivers. Moreover, bvFTD caregiver’s distress and burden were related to cognitive and functional impairment7979. Lima-Silva TB, Bahia VS, Carvalho VA, Guimarães HC, Caramelli P, Balthazar ML, et al. Neuropsychiatric symptoms, caregiver burden and distress in behavioral-variant frontotemporal dementia and Alzheimer's disease. Dement Geriatr Cogn Disord. 2015;40(5-6):268-75. doi:10.1159/000437351.
https://doi.org/10.1159/000437351...
.

Multidisciplinary management may be needed throughout the disease process, but this has not been specifically established for bvFTD treatment. A multidisciplinary team may offer a more comprehensive care management and seems to be an essential tool for dealing with other degenerative diseases8080. Wylie MA, Shnall A, Onyike CU, Huey ED. Management of frontotemporal dementia in mental health and multidisciplinary settings. Int Rev Psychiatry. 2013;25(2):230-6. doi:10.3109/09540261.2013.776949.
https://doi.org/10.3109/09540261.2013.77...
. However, the needs in bvFTD may be more specific and studies regarding their management should be conducted. Social, financial, psychological counseling may be required to address specific issues and more knowledge on those topics should be gathered to help services’ organization. For instance, the emergence of swallowing problems during dementia progression may require specialized evaluation. Problems with inappropriate eating speed, passivity, coughing, and choking beginning in the mild stages and following the worsening cognitive and behavioral declines were recently reported and add information about specific treatment needs8181. Marin SMC, Mansur LL, Oliveira FF, Marin LF, Wajman JR, Bahia VS, et al. Swallowing in behavioral variant frontotemporal dementia. Arq Neuropsiquiatr. 2021;79(1):8-14. doi:10.1590/0004-282X20200060.
https://doi.org/10.1590/0004-282X2020006...
.

Regarding pharmacological approaches, a recent systematic review gathered information aiming to identify pharmacological interventions that could be used to treat the more troublesome behaviors in bvFTD. Trieu et al. (7878. Trieu C, Gossink F, Stek ML, Scheltens P, Pijnenburg YAL, Dols A. Effectiveness of pharmacological interventions for symptoms of behavioral variant frontotemporal dementia: a systematic review. Cogn Behav Neurol. 2020;33(1):1-15. doi:10.1097/WNN.0000000000000217.
https://doi.org/10.1097/WNN.000000000000...
found 23 studies (11 randomized controlled trials, eight open-label studies, one proof-of-concept study, and three case series) involving a sample of 573 individuals. Sixteen out of the 23 studies used pharmacological interventions, resulting in benefits, as measured by the Neuropsychiatric Inventory, for trazodone, citalopram, rivastigmine, paroxetine, ameliorating symptoms such as disinhibition, hyperorality, and depression.

Two reviews8282. Portugal MG, Marinho V, Laks J. Pharmacological treatment of frontotemporal lobar degeneration: systematic review. Braz J Psychiatry. 2011:33(1):81-90. doi:10.1590/s1516-44462011000100016.
https://doi.org/10.1590/s1516-4446201100...
),(8383. Gambogi LB, Guimarães HC, Souza LC, Caramelli P. Treatment of the behavioral variant of frontotemporal dementia: a narrative review. Dement Neuropsychol. 2021;15(3):331-8. doi:10.1590/1980-57642021dn15-030004.
https://doi.org/10.1590/1980-57642021dn1...
published by Brazilian authors report that most studies have small sample sizes, short duration of treatment, and non-uniform measures while evaluating efficacy and tolerability.

Portugal et al. (8282. Portugal MG, Marinho V, Laks J. Pharmacological treatment of frontotemporal lobar degeneration: systematic review. Braz J Psychiatry. 2011:33(1):81-90. doi:10.1590/s1516-44462011000100016.
https://doi.org/10.1590/s1516-4446201100...
conducted a systematic review and reported better results for drugs with serotonergic action, such as selective serotonin reuptake inhibitors or SSRIs (paroxetine, citalopram, fluvoxamine) and trazodone to treat behavioral symptoms, but not cognition. Authors suggest that using SSRIs as the first-line treatment seems to be the best practice until better data on evidence-based strategies to rely upon are available.

In a narrative review aiming to guide clinical practice, Gambogi et al. (8383. Gambogi LB, Guimarães HC, Souza LC, Caramelli P. Treatment of the behavioral variant of frontotemporal dementia: a narrative review. Dement Neuropsychol. 2021;15(3):331-8. doi:10.1590/1980-57642021dn15-030004.
https://doi.org/10.1590/1980-57642021dn1...
conducted a literature search on bvFTD treatment. According to drug class, ChEI and memantine do not have significant therapeutic actions. Antidepressants (SSRIs and trazodone) improved behavioral symptoms with better tolerability; antipsychotics reduced agitation (quetiapine, risperidone, aripiprazole), improper behaviors (risperidone and aripiprazole), and scores on the Neuropsychiatric Inventory (olanzapine) in uncontrolled cases series and clinical reports. However, mortality issues related to antipsychotic use in dementia, cardiovascular risks, and motor AEs limit their use in bvFTD. Psychostimulants for apathy and anticonvulsants for abnormal behaviors were investigated, but must be used with caution due to AEs. In a symptom-focused approach, authors suggest SSRIs or trazodone as the first option for disinhibition, compulsive/perseverative behaviors, stereotyped behaviors, and hyperorality. Psychostimulants may be an option for apathy and antipsychotics should be used with care and reserved to manage challenging and hazardous behaviors.

In summary, several low-quality reports are focusing on symptomatic treatment for bvFTD and no specific treatment proved to be effective to date. ChEI and memantine should not be used as they have not shown any significant therapeutic action. SSRIs or trazodone seem the best practice to manage behavioral problems as a first step, and the use of second-generation antipsychotics should be reserved for the management of psychotic manifestations, troublesome or hazardous behaviors.

Treatment of primary progressive aphasias

Primary progressive aphasia (PPA) refers to clinical syndromes characterized by an insidious and progressive loss of language abilities associated with neurodegenerative diseases. The three variants differ in anatomic, phenotypic presentation, and underlying pathology8484. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, Cappa SF, et al. Classification of primary progressive aphasia and its variants. Neurology. 2011;76(11):1006-14. doi:10.1212/WNL.0b013e31821103e6.
https://doi.org/10.1212/WNL.0b013e318211...
.

The non-fluent/agrammatic variant PPA (nfvPPA) is associated with atrophy of left frontal lobe, insula, and supplementary motor areas, and progressive, effortful, non-fluent speech, with syntactic deficits, grammatical errors, and omissions. The semantic variant PPA (svPPA) is associated with left anterior temporal lobe atrophy and fluent speech with deficits in lexical retrieval, naming, and word comprehension. The logopenic variant PPA (lvPPA) presents AD as the pathological substrate in most cases and is associated with left posterior temporal/parietal atrophy, and slow speech with long-word finding pauses, anomia, and repetition deficits8484. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, Cappa SF, et al. Classification of primary progressive aphasia and its variants. Neurology. 2011;76(11):1006-14. doi:10.1212/WNL.0b013e31821103e6.
https://doi.org/10.1212/WNL.0b013e318211...
.

Up to this moment, there are no disease-modifying agents approved for these language syndromes associated with neurodegenerative diseases. Symptomatic treatments aiming to improve language disorders have been shown to enhance oral and written naming abilities8585. Cotelli M, Manenti R, Ferrari C, Gobbi E, Macis A, Cappa SF. Effectiveness of language training and non-invasive brain stimulation on oral and written naming performance in Primary Progressive Aphasia: A meta-analysis and systematic review. Neurosci Biobehav Rev. 2020;108:498-525. doi:10.1016/j.neubiorev.2019.12.003.
https://doi.org/10.1016/j.neubiorev.2019...
. Language training therapies improve naming accuracy for trained items at short- and long-term follow-up and are considered the standard treatment for PPA8585. Cotelli M, Manenti R, Ferrari C, Gobbi E, Macis A, Cappa SF. Effectiveness of language training and non-invasive brain stimulation on oral and written naming performance in Primary Progressive Aphasia: A meta-analysis and systematic review. Neurosci Biobehav Rev. 2020;108:498-525. doi:10.1016/j.neubiorev.2019.12.003.
https://doi.org/10.1016/j.neubiorev.2019...
),(8686. Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive brain stimulation (tDCS or TMS) paired with language therapy in the treatment of primary progressive aphasia: an exploratory meta-analysis. Brain Sci. 2020;10(9):597. doi:10.3390/brainsci10090597.
https://doi.org/10.3390/brainsci10090597...
.

There are very few studies on PPA treatment in Brazil and Latin-America, with small sample sizes, demonstrating the lack of local information about language therapies in PPA8787. Beber BC, Brandão L, Chaves MLF. A warning to the Brazilian Speech-Language Pathology and Audiology community about the importance of scientific and clinical activities in primary progressive aphasia. CoDAS. 2015;27(5):505-8. doi:10.1590/2317-1782/20152015081.
https://doi.org/10.1590/2317-1782/201520...
. Notwithstanding, an important piece of information was gathered by a Brazilian systematic review organized by Carthery-Goulart et al. (8888. Carthery-Goulart MT, Silveira AC, Machado TH, Mansur LL, Parente MAMP, Senaha MLH, et al. Nonpharmacological interventions for cognitive impairments following primary progressive aphasia: A systematic review of the literature. Dement Neuropsychol. 2013;7(1):122-31. doi:10.1590/S1980-57642013DN70100018.
https://doi.org/10.1590/S1980-57642013DN...
. The literature on evidence-based strategies of language rehabilitation in PPA was summarized and the language behavioral treatments classified into impairment-directed and functional interventions. The results suggest, as a practical option, the recommendations for the use of impairment-directed therapies aimed at naming and lexical retrieval in svPPA. In relation to nfvPPA and lvPPA, the small number of studies limits conclusions about the best therapeutic option for such variants8888. Carthery-Goulart MT, Silveira AC, Machado TH, Mansur LL, Parente MAMP, Senaha MLH, et al. Nonpharmacological interventions for cognitive impairments following primary progressive aphasia: A systematic review of the literature. Dement Neuropsychol. 2013;7(1):122-31. doi:10.1590/S1980-57642013DN70100018.
https://doi.org/10.1590/S1980-57642013DN...
.

While impairment-directed intervention targets the remediation or slowing the progression of specific language and speech impairments, the functional interventions focus on communication, including environmental modifications. The functional interventions key components rely on building communication strategies and practicing the strategies with a communication partner. However, the frequency and dosage of such interventions using rigorous outcomes is an unmet need up to this moment8989. Volkmer A, Spector A, Meitanis V, Warren JD, Beeke S. Effects of functional communication interventions for people with primary progressive aphasia and their caregivers: A systematic review. Aging Ment Health. 2020;24(9):1381-93. doi:10.1080/13607863.2019.1617246.
https://doi.org/10.1080/13607863.2019.16...
.

The maintenance of therapy gains, in the absence of continuous training, is more evident in the short-term than long-term in all PPA variants and is influenced by treatment length and session frequency. Generalization to untreated items varies following each PPA subtype and occurs more often in nfvPPA and lvPPA than in svPPA9090. Cadório I, Lousada M, Martins P, Figueiredo D. Generalization and maintenance of treatment gains in primary progressive aphasia (PPA): A systematic review. Int J Lang Commun Disord. 2017;52(5):543-60. doi:10.1111/1460-6984.12310.
https://doi.org/10.1111/1460-6984.12310...
.

Language therapies usually focus on enhancing abilities in trained activities or tasks and such training may limit benefits to the practiced domains8686. Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive brain stimulation (tDCS or TMS) paired with language therapy in the treatment of primary progressive aphasia: an exploratory meta-analysis. Brain Sci. 2020;10(9):597. doi:10.3390/brainsci10090597.
https://doi.org/10.3390/brainsci10090597...
),(8888. Carthery-Goulart MT, Silveira AC, Machado TH, Mansur LL, Parente MAMP, Senaha MLH, et al. Nonpharmacological interventions for cognitive impairments following primary progressive aphasia: A systematic review of the literature. Dement Neuropsychol. 2013;7(1):122-31. doi:10.1590/S1980-57642013DN70100018.
https://doi.org/10.1590/S1980-57642013DN...
. Recent reviews have shown a potential benefit for non-invasive brain stimulation combined with language therapies in improving oral or written naming accuracy for trained and untrained items8585. Cotelli M, Manenti R, Ferrari C, Gobbi E, Macis A, Cappa SF. Effectiveness of language training and non-invasive brain stimulation on oral and written naming performance in Primary Progressive Aphasia: A meta-analysis and systematic review. Neurosci Biobehav Rev. 2020;108:498-525. doi:10.1016/j.neubiorev.2019.12.003.
https://doi.org/10.1016/j.neubiorev.2019...
),(8686. Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive brain stimulation (tDCS or TMS) paired with language therapy in the treatment of primary progressive aphasia: an exploratory meta-analysis. Brain Sci. 2020;10(9):597. doi:10.3390/brainsci10090597.
https://doi.org/10.3390/brainsci10090597...
.

As a practical orientation cognitive-linguistic interventions targeting impairment and aiming at remediation and symptoms’ improvement seem the best practice. In this sense, a recent Brazilian study investigated the short- and mid-term effects of four training programs directed to language and speech deficits in 18 PPA individuals (different subtypes) carried out during four months (two weekly sessions). All cases improved performance on trained items during the active phase of treatment. Statistically significant clinical benefits were observed in 13 individuals, while for five, the results were maintained. However, generalization to untrained items or to other tasks were observed only in two individuals9191. Machado TH, Carthery-Goulart MT, Campanha AC, Caramelli P. Cognitive intervention strategies directed to speech and language deficits in primary progressive aphasia: practice-based evidence from 18 cases. Brain Sci. 2021;11(10):1268. doi:10.3390/brainsci11101268.
https://doi.org/10.3390/brainsci11101268...
.

TREATMENT OF PARKINSON’S DISEASE DEMENTIA AND DEMENTIA WITH LEWY BODIES

PD and DLB are neurodegenerative disorders that share some common features such as dopaminergic depletion and movement disorders9292. Aarsland D. Cognitive impairment in Parkinson's disease and dementia with Lewy bodies. Parkinsonism Relat Disord. 2016;22(Suppl 1):S144-8. doi:10.1016/j.parkreldis.2015.09.034.
https://doi.org/10.1016/j.parkreldis.201...
. Lewy body inclusions throughout the cortex ensue the typical triad of the dementia syndrome in DLB: dementia, complex visual hallucinations and parkinsonism. PDD, in turn, starts with a dysexecutive syndrome9393. Burn DJ. Cortical Lewy body disease and Parkinson's disease dementia. Curr Opin Neurol. 2006;19(6):572-9. doi:10.1097/01.wco.0000247607.34697.a2.
https://doi.org/10.1097/01.wco.000024760...
),(9494. Outeiro TF, Koss DJ, Erskine D, Walker L, Kurzawa-Akanbi M, Burn D, et al. Dementia with Lewy bodies: an update and outlook. Mol Neurodegener. 2019;14(1):5. doi:10.1186/s13024-019-0306-8.
https://doi.org/10.1186/s13024-019-0306-...
.

DLB may show parkinsonism later in the course of the disease, whereas complex visual hallucinations appear early, together with delirium without other clinical causes. PD first symptoms, on the other hand, encompass asymmetric bradykinesia, tremor and muscle rigidity9595. Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018;125(4):615-50. doi:10.1007/s00702-017-1821-9.
https://doi.org/10.1007/s00702-017-1821-...
. Memory complaints are not the first symptoms in DLB or in PDD, as usually observed in dementia due to AD and VCI. PDD typically presents with a frontal dysexecutive syndrome9595. Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018;125(4):615-50. doi:10.1007/s00702-017-1821-9.
https://doi.org/10.1007/s00702-017-1821-...
),(9696. Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C, Mizuno Y, et al. Clinical diagnostic criteria for dementia associated with Parkinson's disease. Mov Disord. 2007;22(12):1689-707. doi:10.1002/mds.21507.
https://doi.org/10.1002/mds.21507...
.

The identification of the disease which is causing dementia is important to identify drugs that should and should not be prescribed when neuropsychiatric symptoms occur. Antipsychotics should be avoided in DLB, as there is a hypersensitivity to neuroleptic malignant syndrome and because they may aggravate parkinsonism, regardless of the dose prescribed. When psychosis exists in PDD, levodopa and other antiparkinsonian drugs should be re-evaluated, before prescribing any antipsychotics to deal with the behavioral and mood symptoms9797. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. doi:10.1212/WNL.0000000000004058.
https://doi.org/10.1212/WNL.000000000000...
),(9898. Broadstock M, Ballard C, Corbett A. Latest treatment options for Alzheimer's disease, Parkinson's disease dementia and dementia with Lewy bodies. Expert Opin Pharmacother. 2014;15(13):1797-810. doi:10.1517/14656566.2014.936848.
https://doi.org/10.1517/14656566.2014.93...
.

Cognitive and neuropsychiatric symptoms in PDD and DLB are treated with rivastigmine, the only approved ChEI for these indications9898. Broadstock M, Ballard C, Corbett A. Latest treatment options for Alzheimer's disease, Parkinson's disease dementia and dementia with Lewy bodies. Expert Opin Pharmacother. 2014;15(13):1797-810. doi:10.1517/14656566.2014.936848.
https://doi.org/10.1517/14656566.2014.93...
),(9999. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
https://doi.org/10.1176/appi.ajp.2015.14...
. Memantine appears to be associated with some improvement in behavioral aspects, but studies have not reported benefits for cognition in these individuals9898. Broadstock M, Ballard C, Corbett A. Latest treatment options for Alzheimer's disease, Parkinson's disease dementia and dementia with Lewy bodies. Expert Opin Pharmacother. 2014;15(13):1797-810. doi:10.1517/14656566.2014.936848.
https://doi.org/10.1517/14656566.2014.93...
),(9999. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
https://doi.org/10.1176/appi.ajp.2015.14...
. Memantine has not been approved for treatment of DLB or PDD.

As for treatment of neuropsychiatric symptoms and syndromes, stimuli regulation should be tried before any medication is applied. Excessive stimulation, or the lack of proper stimulation, can worsen orientation and behavior. Room light, physical exercise, cognitive stimulation, calm environment and maintenance of clinical status can help control behavioral problems before medication comes into place100100. Connors MH, Quinto L, McKeith I, Brodaty H, Allan L, Bamford C, et al. Non-pharmacological interventions for Lewy body dementia: a systematic review. Psychol Med. 2018;48(11):1749-58. doi:10.1017/S0033291717003257.
https://doi.org/10.1017/S003329171700325...
. Even if medication is needed, all these measures should be enforced.

The clinical diagnosis, differentiating PDD from DLB, can be a challenge, even for the specialist, since the two conditions seem to be different spectra of the same pathological family, being mainly differentiated by the clinical course.

The cortical and subcortical distribution of Lewy bodies will lead to different clinical presentations. Although it seems somewhat theoretical, this distinction is essential in some details of the medication management inherent to each situation. Diagnostic criteria for PDD and DLB have been published before9696. Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C, Mizuno Y, et al. Clinical diagnostic criteria for dementia associated with Parkinson's disease. Mov Disord. 2007;22(12):1689-707. doi:10.1002/mds.21507.
https://doi.org/10.1002/mds.21507...
),(9797. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. doi:10.1212/WNL.0000000000004058.
https://doi.org/10.1212/WNL.000000000000...
.

Lewy body diseases spectrum

Due to the inherent characteristics of PDD and DLB, several clinical-pathological correlation studies include the two conditions as different spectra of the same pathological process. In the present consensus, we consider the two diseases from this viewpoint, i.e., different involvements of a common pathological substrate. In PDD, the accumulation of Lewy bodies is mainly concentrated in subcortical areas, while in DLB the pathological process affects cortical areas (Figure 2) (101101. Dugger BN, Boeve BF, Murray ME, Parisi JE, Fujishiro H, Dickson DW, et al. Rapid eye movement sleep behavior disorder and subtypes in autopsy-confirmed dementia with Lewy bodies. Mov Disord. 2012;27(1):72-8. doi:10.1002/mds.24003.
https://doi.org/10.1002/mds.24003...
)-(103103. Hughes AJ, Daniel SE, Blankson S, Lees AJ. A clinicopathological study of 100 cases of Parkinson's disease. J Neurol Neurosurg Psychiatry. 1992;55(3):181-4. doi:10.1136/jnnp.55.3.181.
https://doi.org/10.1136/jnnp.55.3.181...
.

Figure 2
Topographic distribution pattern of Lewy bodies in dementia with Lewy bodies (DLB), Parkinson’s disease (PD) and dementia associated with Parkinson’s disease (PDD).

Management of DLB

There is no specific disease-modifying treatment for DLB. Non-pharmacological and pharmacological interventions can be used in symptomatic management. There are no specific non-pharmacological interventions in DLB100100. Connors MH, Quinto L, McKeith I, Brodaty H, Allan L, Bamford C, et al. Non-pharmacological interventions for Lewy body dementia: a systematic review. Psychol Med. 2018;48(11):1749-58. doi:10.1017/S0033291717003257.
https://doi.org/10.1017/S003329171700325...
),(104104. Rongve A, Aarsland D. Management of Parkinson's disease dementia: practical considerations. Drugs Aging. 2006;23(10):807-22. doi:10.2165/00002512-200623100-00004.
https://doi.org/10.2165/00002512-2006231...
.

Motor symptoms

Levodopa is the drug of choice to treat motor symptoms. Slow titration is important to achieve some motor response with minimal psychiatric AEs. Response to levodopa in DLB can be limited, especially in the later stages, due to the predominance of levodopa non-responsive symptoms. Dopamine agonists are not recommended due to the risk of worsening hallucinations. Anticholinergics are contraindicated9797. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. doi:10.1212/WNL.0000000000004058.
https://doi.org/10.1212/WNL.000000000000...
)-(9999. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
https://doi.org/10.1176/appi.ajp.2015.14...
),(105105. Caviness JN, Lue L, Adler CH, Walker DG. Parkinson's disease dementia and potential therapeutic strategies. CNS Neurosci Ther. 2011;17(1):32-44. doi:10.1111/j.1755-5949.2010.00216.x.
https://doi.org/10.1111/j.1755-5949.2010...
),(106106. Gauthier S. Pharmacotherapy of Parkinson disease dementia and Lewy body dementia. Front Neurol Neurosci. 2009;24:135-9. doi:10.1159/000197892.
https://doi.org/10.1159/000197892...
.

Neuropsychiatric symptoms

The use of antipsychotics must be done very judiciously, due to the high sensitivity of these patients, with important paradoxical reactions107107. Garcia-Ptacek S, Kramberger MG. Parkinson disease and dementia. J Geriatr Psychiatry Neurol. 2016;29(5):261-70. doi:10.1177/0891988716654985.
https://doi.org/10.1177/0891988716654985...
. Typical antipsychotics are contraindicated. Atypical antipsychotics, specifically quetiapine and clozapine, can be used, always under close supervision, to detect AEs9797. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. doi:10.1212/WNL.0000000000004058.
https://doi.org/10.1212/WNL.000000000000...
),(9999. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
https://doi.org/10.1176/appi.ajp.2015.14...
),(108108. Sezgin M, Bilgic B, Tinaz S, Emre M. Parkinson's disease dementia and Lewy body disease. Semin Neurol. 2019;39(2):274-82. doi:10.1055/s-0039-1678579.
https://doi.org/10.1055/s-0039-1678579...
. Of special note, clozapine may cause neutropenia in about 3% of cases, which demands weekly monitoring of blood cell count within the first 18 weeks of treatment.

As for ChEI (mainly rivastigmine), besides the cognitive benefits several studies have shown positive effects on neuropsychiatric symptoms, particularly visual hallucinations, delusions, and apathy. It is essential to be aware that the withdrawal of ChEI, for any reason, may be associated with cognitive deterioration9999. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
https://doi.org/10.1176/appi.ajp.2015.14...
),(109109. Taylor JP, McKeith IG, Burn DJ, Boeve BF, Weintraub D, Bamford C, et al. New evidence on the management of Lewy body dementia. Lancet Neurol. 2020;19(2):157-69. doi:10.1016/S1474-4422(19)30153-X.
https://doi.org/10.1016/S1474-4422(19)30...
)-(111111. Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase inhibitors for dementia with Lewy bodies, Parkinson's disease dementia and cognitive impairment in Parkinson's disease. Cochrane Database Syst Rev. 2012;(3):CD006504. doi:10.1002/14651858.CD006504.pub2.
https://doi.org/10.1002/14651858.CD00650...
.

CONCLUSIONS

Although current treatments for neurodegenerative and vascular dementias are only symptomatic, several evidenced-based therapies are available and may provide benefits to Pwd and to their caregivers. The clinician must combine the best pharmacological options with the best available non-pharmacological interventions, taking into consideration the underlying etiology, symptoms profile and severity of the dementia syndrome, as well as the clinical and personal history of each individual.

It is important to inform the PwD and their families about the potential expected benefits of the overall treatment, as well as the possible AEs of the prescribed drugs. Regular follow-up is critical (ideally every three to four months in the beginning of treatment and around every six months later on) to evaluate the clinical effects and whether adjustments (e.g., drug dosing, additional pharmacological and non-pharmacological interventions) are needed.

Given that the modest effects of the available treatments and the lack of approved disease-specific drug therapies, prevention of dementia is a key principle. Several modifiable risk factors have been identified and lifestyle and clinical interventions (e.g., adequate control of hypertension and diabetes, regular physical activity, diet programs, cognitive stimulation activities) shall be recommended for middle-age and older adults to reduce the risk of cognitive impairment and dementia, thus contributing to mitigate the personal, socioeconomic and public health impacts of these devastating disorders. (4545. Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, et al. World-Wide FINGERS Network: A global approach to risk reduction and prevention of dementia. Alzheimers Dement. 2020;16(7):1078-94. doi:10.1002/alz.12123.
https://doi.org/10.1002/alz.12123...

ACKNOWLEDGEMENTS

PC, JL, LCS and RN are funded by CNPq, Brazil (bolsa de produtividade em pesquisa).

Referências

  • 1
    Mukadam N, Sommerlad A, Huntley J, Livingston G. Population attributable fractions for risk factors for dementia in low-income and middle-income countries: an analysis using cross-sectional survey data. Lancet Glob Health. 2019;7(5):e596-603. doi:10.1016/S2214-109X(19)30074-9.
    » https://doi.org/10.1016/S2214-109X(19)30074-9
  • 2
    Nakamura AE, Opaleye D, Tani G, Ferri CP. Dementia underdiagnosis in Brazil. Lancet. 2015;385(9966):418-9. doi:10.1016/S0140-6736(15)60153-2.
    » https://doi.org/10.1016/S0140-6736(15)60153-2
  • 3
    Nitrini R, Bottino CM, Albala C, Custodio Capuñay NS, Ketzoian C, Llibre Rodriguez JJ, et al. Prevalence of dementia in Latin America: a collaborative study of population-based cohorts. Int Psychogeriatr. 2009;21(4):622-30. doi:10.1017/S1041610209009430.
    » https://doi.org/10.1017/S1041610209009430
  • 4
    Institute of Health Metrics [homepage na Internet]. Brazil. 2018 [citado em 22 ago 2022]. Disponível em: http://www.healthdata.org/brazil
    » http://www.healthdata.org/brazil
  • 5
    Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. Treatment of Alzheimer's Disease: recommendations and suggestions of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. doi:10.1590/S0004-282X2005000600035.
    » https://doi.org/10.1590/S0004-282X2005000600035
  • 6
    Bartus RT. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp Neurol. 2000;163(2):495-529. doi:10.1006/exnr.2000.7397.
    » https://doi.org/10.1006/exnr.2000.7397
  • 7
    Small G, Bullock R. Defining optimal treatment with cholinesterase inhibitors in Alzheimer's disease. Alzheimers Dement. 2011;7(2):177-84. doi:10.1016/j.jalz.2010.03.016.
    » https://doi.org/10.1016/j.jalz.2010.03.016
  • 8
    Xu H, Garcia-Ptacek S, Jönsson L, Wilmo A, Nordström P, Eriksdotter M. Long-term effects of cholinesterase inhibitors on cognitive decline and mortality. Neurology. 2021;96(e):2220-30. doi:10.1212/WNL.0000000000011832.
    » https://doi.org/10.1212/WNL.0000000000011832
  • 9
    Ministério da Saúde (BR). Secretaria de Atenção à Saúde. Portaria Conjunta nº 13, de 28 de novembro de 2017. Aprova o Protocolo Clínico e Diretrizes Terapêuticas da Doença de Alzheimer. Diário Oficial da União. 8 dez 2017;1:201.
  • 10
    Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, et al. Early diagnosis and treatment of Alzheimer's disease: New definitions and challenges. Braz J Psychiatry. 2020;42(4):431-41. doi:10.1590/1516-4446-2019-0735.
    » https://doi.org/10.1590/1516-4446-2019-0735
  • 11
    Vale FAC, Corrêa Neto Y, Bertolucci PHF, Machado JCB, Silva DJ, Allam N, et al. Treatment of Alzheimer's disease in Brazil: II. Behavioral and psychological symptoms of dementia. Dement Neuropsychol. 2011;5(3):189-97. doi:10.1590/S1980-57642011DN05030006.
    » https://doi.org/10.1590/S1980-57642011DN05030006
  • 12
    He Y, Li H, Huang J, Huang S, Bai Y, Li Y, et al. Efficacy of antidepressant drugs in the treatment of depression in Alzheimer disease patients: A systematic review and network meta-analysis. J Psychopharmacol. 2021;35(8):901-9. doi:10.1177/02698811211030181.
    » https://doi.org/10.1177/02698811211030181
  • 13
    Banerjee S, High J, Stirling S, Shepstone L, Swart AM, Telling T, et al. Study of mirtazapine for agitated behaviours in dementia (SYMBAD): a randomised, double-blind, placebo-controlled trial. Lancet. 2021;398(10310):1487-97. doi:10.1016/S0140-6736(21)01210-1.
    » https://doi.org/10.1016/S0140-6736(21)01210-1
  • 14
    Davies SJ, Burhan AM, Kim D, Gerretsen P, Graff-Guerrero A, Woo VL, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol. 2018;32(5):509-23. doi:10.1177/0269881117744996.
    » https://doi.org/10.1177/0269881117744996
  • 15
    Camargos EF, Louzada LL, Quintas JL, Naves JO, Louzada FM, Nóbrega OT. Trazodone improves sleep parameters in Alzheimer disease patients: a randomized, double-blind, and placebo-controlled study. Am J Geriatr Psychiatry. 2014;22(12):1565-74. doi:10.1016/j.jagp.2013.12.174.
    » https://doi.org/10.1016/j.jagp.2013.12.174
  • 16
    Louzada LL, Machado FV, Quintas JL, Ribeiro GA, Silva MV, Mendonça-Silva DL, et al. The efficacy and safety of zolpidem and zopiclone to treat insomnia in Alzheimer's disease: a randomized, triple-blind, placebo-controlled trial. Neuropsychopharmacology. 2022;47(2):570-9. doi:10.1038/s41386-021-01191-3.
    » https://doi.org/10.1038/s41386-021-01191-3
  • 17
    Caraci F, Santagati M, Caruso G, Cannavò D, Leggio GM, Salomone S, et al. New antipsychotic drugs for the treatment of agitation and psychosis in Alzheimer's disease: focus on brexpiprazole and pimavanserin. F1000Res. 2020;9:F1000 Faculty Rev-686. doi:10.12688/f1000research.22662.1.
    » https://doi.org/10.12688/f1000research.22662.1
  • 18
    Brucki SMD, Adoni T, Almeida CMO, Andrade DC, Anghinah R, Barbosa LM, et al. Cannabinoids in Neurology - Position paper from Scientific Departments from Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2021;79(4):354-69. doi:10.1590/0004-282X-ANP-2020-0432.
    » https://doi.org/10.1590/0004-282X-ANP-2020-0432
  • 19
    Frederiksen KS, Cooper C, Frisoni GB, Frölich L, Georges J, Kramberger MG, et al. A European Academy of Neurology guideline on medical management issues in dementia. Eur J Neurol. 2020;27(10):1805-20. doi:10.1111/ene.14412.
    » https://doi.org/10.1111/ene.14412
  • 20
    York Health Economics Consortium. NICE: Overview of Systematic Reviews of Non-pharmacological Interventions for Dementia [Internet]. Final Report. Heslington: University of York; 2017 [citado em 22 ago 2022]. Disponível em: https://www.nice.org.uk/guidance/ng97/evidence/appendix-o-yhec-report-pdf-174697045530
    » https://www.nice.org.uk/guidance/ng97/evidence/appendix-o-yhec-report-pdf-174697045530
  • 21
    Aguirre E, Spector A, Orrell M. Guidelines for adapting cognitive stimulation therapy to other cultures. Clin Interv Aging. 2014;9:1003-7. doi:10.2147/CIA.S61849.
    » https://doi.org/10.2147/CIA.S61849
  • 22
    Meyer C, O'Keefe F. Non-pharmacological interventions for people with dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. doi:10.1177/1471301218813234.
    » https://doi.org/10.1177/1471301218813234
  • 23
    Stoner CR, Lakshminarayanan M, Durgante H, Spector A. Psychosocial interventions for dementia in low- and middle-income countries (LMICs): a systematic review of effectiveness and implementation readiness. Aging Ment Health. 2021;25(3):408-19. doi:10.1080/13607863.2019.1695742.
    » https://doi.org/10.1080/13607863.2019.1695742
  • 24
    Bertrand E, Naylor R, Laks J, Marinho V, Spector A, Mograbi DC. Cognitive stimulation therapy for Brazilian people with dementia: examination of implementation' issues and cultural adaptation. Aging Ment Health. 2019;23(10):1400-4. doi:10.1080/13607863.2018.1488944.
    » https://doi.org/10.1080/13607863.2018.1488944
  • 25
    Marinho V, Bertrand E, Naylor R, Bomilcar I, Laks J, Spector A, et al. Cognitive stimulation therapy for people with dementia in Brazil (CST-Brasil): Results from a single blind randomized controlled trial. Int J Geriatr Psychiatry. 2021;36(2):286-93. doi:10.1002/gps.5421.
    » https://doi.org/10.1002/gps.5421
  • 26
    Madureira BG, Pereira MG, Avelino PR, Costa HS, Menezes KKP. Efeitos de programas de reabilitação multidisciplinar no tratamento de pacientes com doença de Alzheimer: uma revisão sistemática. Cad Saude Coletiva. 2018;26(2):222-32. doi:10.1590/1414-462x201800020446.
    » https://doi.org/10.1590/1414-462x201800020446
  • 27
    Cammisuli DM, Danti S, Bosinelli F, Cipriani G. Non-pharmacological interventions for people with Alzheimer's disease: A critical review of the scientific literature from the last ten years. Eur Geriatr Med. 2016,7(1):57-64. doi:10.1016/j.eurger.2016.01.002.
    » https://doi.org/10.1016/j.eurger.2016.01.002
  • 28
    Carrion C, Folkvord F, Anastasiadou D, Aymerich M. Cognitive therapy for dementia patients: a systematic review. Dement Geriatr Cogn Disord. 2018;46(1-2):1-26. doi:10.1159/000490851.
    » https://doi.org/10.1159/000490851
  • 29
    Chiu H-Y, Chen P-Y, Chen Y-T, Huang H-C. Reality orientation therapy benefits cognition in older people with dementia: A meta-analysis. Int J Nurs Stud. 2018;86:20-8. doi:10.1016/j.ijnurstu.2018.06.008.
    » https://doi.org/10.1016/j.ijnurstu.2018.06.008
  • 30
    Alves GS, Casali ME, Veras AB, Carrilho CG, Bruno Costa E, Rodrigues VM, et al. A systematic review of home-setting psychoeducation interventions for behavioral changes in dementia: some lessons for the COVID-19 Pandemic and post-pandemic assistance. Front Psychiatry. 2020;11:577871. doi:10.3389/fpsyt.2020.577871.
    » https://doi.org/10.3389/fpsyt.2020.577871
  • 31
    Duan Y, Lu L, Chen J, Wu C, Liang J, Zheng Y, et al. Psychosocial interventions for Alzheimer's disease cognitive symptoms: a Bayesian network meta-analysis. BMC Geriatr. 2018;18(1):175. doi:10.1186/s12877-018-0864-6.
    » https://doi.org/10.1186/s12877-018-0864-6
  • 32
    Piersol CV, Canton K, Connor SE, Giller I, Lipman S, Sager S. Effectiveness of interventions for caregivers of people with Alzheimer's Disease and related major neurocognitive disorders: a systematic review. Am J Occup Ther. 2017;71(5):7105180020p1-p10. doi:10.5014/ajot.2017.027581.
    » https://doi.org/10.5014/ajot.2017.027581
  • 33
    Shim M, Tilley JL, Im S, Price K, Gonzalez A. A systematic review of mindfulness-based interventions for patients with mild cognitive impairment or dementia and caregivers. J Geriatr Psychiatry Neurol. 2021;34(6):528-54. doi:10.1177/0891988720957104.
    » https://doi.org/10.1177/0891988720957104
  • 34
    Gaugler JE, Bain LJ, Mitchell L, Finlay J, Fazio S, Jutkowitz E & Alzheimer's Association Psychosocial Measurement Workgroup. Reconsidering frameworks of Alzheimer's dementia when assessing psychosocial outcomes. Alzheimers Dement. 2019;5:388-97. https://doi.org/10.1016/j.trci.2019.02.008.
    » https://doi.org/https://doi.org/10.1016/j.trci.2019.02.008
  • 35
    Dröes RM, Chattat R, Diaz A, Gove D, Graff M, Murphy K. Social health and dementia: a European consensus on the operationalization of the concept and directions for research and practice. Aging Ment Health. 2017;21(1):4-17. doi:10.1080/13607863.2016.1254596.
    » https://doi.org/10.1080/13607863.2016.1254596
  • 36
    Oksnebjerg L, Diaz-Ponce A, Gove D, Moniz-Cook E, Mountain G, Chattat R, et al. Towards capturing meaningful outcomes for people with dementia in psychosocial intervention research: a pan-European consultation. Health Expect. 2018;21(6):1056-65. doi:10.1111/hex.12799.
    » https://doi.org/10.1111/hex.12799
  • 37
    Arroyo-Anlló EM, Díaz-Marta JP, Chamorro Sánchez J. Técnicas de rehabilitación neuropsicológica en demencias: hacia la ciber-rehabilitación neuropsicológica. Pensam Psicol. 2012;10(1):107-27.
  • 38
    Moreira SV, Reis Justi FR, Moreira M. Can musical intervention improve memory in Alzheimer's patients? Evidence from a systematic review. Dement Neuropsychol. 2018;12(2):133-42. doi:10.1590/1980-57642018dn12-020005.
    » https://doi.org/10.1590/1980-57642018dn12-020005
  • 39
    Jia RX, Liang JH, Xu Y, Wang YQ. Effects of physical activity and exercise on the cognitive function of patients with Alzheimer disease: a meta-analysis. BMC Geriatr. 2019;19(1):181. doi:10.1186/s12877-019-1175-2.
    » https://doi.org/10.1186/s12877-019-1175-2
  • 40
    Wang LY, Pei J, Zhan YJ, Cai YW. Overview of meta-analyses of five non-pharmacological interventions for Alzheimer's Disease. Front Aging Neurosci. 2020;12:594432. doi:10.3389/fnagi.2020.594432.
    » https://doi.org/10.3389/fnagi.2020.594432
  • 41
    Meng Q, Lin MS, Tzeng IS. Relationship between exercise and Alzheimer's disease: a narrative literature review. Front Neurosci. 2020;14:131. doi:10.3389/fnins.2020.00131.
    » https://doi.org/10.3389/fnins.2020.00131
  • 42
    Smallfield S, Heckenlaible C. Effectiveness of occupational therapy interventions to enhance occupational performance for adults with Alzheimer's Disease and related major neurocognitive disorders: a systematic review. Am J Occup Ther. 2017;71(5):7105180010p1-p9. doi:10.5014/ajot.2017.024752.
    » https://doi.org/10.5014/ajot.2017.024752
  • 43
    Marques CLS, Borgato MH, Moura Neto E, Bazan R, Luvizutto GJ. Physical therapy in patients with Alzheimer's disease: a systematic review of randomized controlled clinical trials. Fisioter Pesqui. 2019;26(3):311-21. doi:10.1590/1809-2950/18037226032019.
    » https://doi.org/10.1590/1809-2950/18037226032019
  • 44
    Zhu XC, Yu Y, Wang HF, Jiang T, Cao L, Wang C, et al. Physiotherapy intervention in Alzheimer's disease: systematic review and meta-analysis. J Alzheimers Dis. 2015;44(1):163-74. doi:10.3233/JAD-141377.
    » https://doi.org/10.3233/JAD-141377
  • 45
    Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, et al. World-Wide FINGERS Network: A global approach to risk reduction and prevention of dementia. Alzheimers Dement. 2020;16(7):1078-94. doi:10.1002/alz.12123.
    » https://doi.org/10.1002/alz.12123
  • 46
    Moreira SC, Jansen AK, Silva FM. Dietary interventions and cognition of Alzheimer's disease patients: a systematic review of randomized controlled trial. Dement Neuropsychol. 2020;14(3):258-82. doi:10.1590/1980-57642020dn14-030008.
    » https://doi.org/10.1590/1980-57642020dn14-030008
  • 47
    Pendlebury ST, Rothwell PM. Prevalence, incidence, and factors associated with pre-stroke and post-stroke dementia: a systematic review and meta-analysis. Lancet Neurol. 2009;8(11):1006-18. doi:10.1016/S1474-4422(09)70236-4.
    » https://doi.org/10.1016/S1474-4422(09)70236-4
  • 48
    Boehme AK, Esenwa C, Elkind MSV. Stroke risk factors, genetics, and prevention. Circ Res. 2017;120(3):472-95. doi:10.1161/CIRCRESAHA.116.308398.
    » https://doi.org/10.1161/CIRCRESAHA.116.308398
  • 49
    ADVANCE Collaborative Group; Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358(24):2560-72. doi:10.1056/NEJMoa0802987.
    » https://doi.org/10.1056/NEJMoa0802987
  • 50
    Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, et al. Effects of intensive glucose lowering on brain structure and function in people with type 2 diabetes (ACCORD MIND): a randomised open-label substudy. Lancet Neurol. 2011;10(11):969-77. doi:10.1016/S1474-4422(11)70188-0.
    » https://doi.org/10.1016/S1474-4422(11)70188-0
  • 51
    Costa e Forti A, Pires AC, Pittito BA, Gerchman F, Oliveira JEP, Zajdenverg L, et al. Diretrizes da Sociedade Brasileira de Diabetes: 2019-2020. São Paulo: Clannad; 2019.
  • 52
    Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, et al. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack. Stroke. 2014;45(7):2160-236. doi:10.1161/STR.0000000000000024.
    » https://doi.org/10.1161/STR.0000000000000024
  • 53
    Smith EE, Saposnik G, Biessels GJ, Doubal FN, Fornage M, Gorelick PB, et al. Prevention of stroke in patients with silent cerebrovascular disease: a scientific statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2017;48(2):e44-71. doi:10.1161/STR.0000000000000116.
    » https://doi.org/10.1161/STR.0000000000000116
  • 54
    Davis KAS, Bishara D, Molokhia M, Mueller C, Perera G, Stewart RJ. Aspirin in people with dementia, long-term benefits, and harms: a systematic review. Eur J Clin Pharmacol. 2021;77(7):943-54. doi:10.1007/s00228-021-03089-x.
    » https://doi.org/10.1007/s00228-021-03089-x
  • 55
    McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev. 2014;(7):CD007514. doi:10.1002/14651858.CD007514.pub3.
    » https://doi.org/10.1002/14651858.CD007514.pub3
  • 56
    Sofi F, Valecchi D, Bacci D, Abbate R, Gensini GF, Casini A, et al. Physical activity and risk of cognitive decline: a meta-analysis of prospective studies. J Int Med. 2011;269(1):107-17. doi:10.1111/j.1365-2796.2010.02281.x.
    » https://doi.org/10.1111/j.1365-2796.2010.02281.x
  • 57
    Verdelho A, Madureira S, Ferro JM, Baezner H, Blahak C, Poggesi A, et al. Physical activity prevents progression for cognitive impairment and vascular dementia: results from the LADIS (Leukoaraiosis and Disability) study. Stroke. 2012;43(12):3331-5. doi:10.1161/STROKEAHA.112.661793.
    » https://doi.org/10.1161/STROKEAHA.112.661793
  • 58
    Gillespie DC, Bowen A, Chung CS, Cockburn J, Knapp P, Pollock A. Rehabilitation for post-stroke cognitive impairment: an overview of recommendations arising from systematic reviews of current evidence. Clin Rehabil. 2015;29(2):120-8. doi:10.1177/0269215514538982.
    » https://doi.org/10.1177/0269215514538982
  • 59
    Coelho-Júnior HJ, Trichopoulou A, Panza F. Cross-sectional and longitudinal associations between adherence to Mediterranean diet with physical performance and cognitive function in older adults: A systematic review and meta-analysis. Ageing Res Rev. 2021;70:101395. doi:10.1016/j.arr.2021.101395.
    » https://doi.org/10.1016/j.arr.2021.101395
  • 60
    Brucki SMD, Ferraz AC, Freitas GR, Massaro AR, Radanovic M, Schultz RR. Treatment of vascular dementia. Recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol. 2011;5(4):275-87. doi:10.1590/S1980-57642011DN05040005.
    » https://doi.org/10.1590/S1980-57642011DN05040005
  • 61
    Smith EE, Barber P, Field TS, Ganesh A, Hachinski V, Hogan DB, et al. Canadian Consensus Conference on Diagnosis and Treatment of Dementia (CCCDTD) 5: Guidelines for management of vascular cognitive impairment. Alzheimers Dement (N Y). 2020;6(1):e12056. doi:10.1002/trc2.12056.
    » https://doi.org/10.1002/trc2.12056
  • 62
    Dichgans M, Markus HS, Salloway S, Verkkoniemi A, Moline M, Wang Q, et al. Donepezil in patients with subcortical vascular cognitive impairment: a randomised double-blind trial in CADASIL. Lancet Neurol. 2008;7(4):310-8. doi:10.1016/S1474-4422(08)70046-2.
    » https://doi.org/10.1016/S1474-4422(08)70046-2
  • 63
    Black S, Román GC, Geldmacher DS, Salloway S, Hecker J, Burns A, et al. Efficacy and tolerability of donepezil in vascular dementia: positive results of a 24-week, multicenter, international, randomized, placebo-controlled clinical trial. Stroke. 2003;34(10):2323-30. doi:10.1161/01.STR.0000091396.95360.E1.
    » https://doi.org/10.1161/01.STR.0000091396.95360.E1
  • 64
    Orgogozo JM, Rigaud AS, Sto¨ffler A, Mo¨bius HJ, Forette F. Efficacy and safety of memantine in patients with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke. 2002;33(7):1834-9. doi:10.1161/01.str.0000020094.08790.49.
    » https://doi.org/10.1161/01.str.0000020094.08790.49
  • 65
    Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359(9314):1283-90. doi:10.1016/S0140-6736(02)08267-3.
    » https://doi.org/10.1016/S0140-6736(02)08267-3
  • 66
    Caramelli P, Laks J, Palmini AL, Nitrini R, Chaves ML, Forlenza OV, et al. Effects of galantamine and galantamine combined with nimodipine on cognitive speed and quality of life in mixed dementia: a 24-week, randomized, placebo-controlled exploratory trial (the REMIX study). Arq Neuropsiquiatr. 2014;72(6):411-7. doi:10.1590/0004-282x20140055.
    » https://doi.org/10.1590/0004-282x20140055
  • 67
    Ballard C, Sauter M, Scheltens P, He Y, Barkhof F, Straaten EC, et al. Efficacy, safety and tolerability of rivastigmine capsules in patients with probable vascular dementia: the VantagE study. Curr Med Res Opin. 2008;24(9):2561-74. doi:10.1185/03007990802328142.
    » https://doi.org/10.1185/03007990802328142
  • 68
    Román GC, Salloway S, Black SE, Royall DR, Decarli C, Weiner MW, et al. Randomized, placebo-controlled, clinical trial of donepezil in vascular dementia: differential effects by hippocampal size. Stroke. 2010;41(6):1213-21. doi:10.1161/STROKEAHA.109.570077.
    » https://doi.org/10.1161/STROKEAHA.109.570077
  • 69
    Moretti R, Torre P, Antonello RM, Cazzato G, Pizzolato G. Different responses to rivastigmine in subcortical vascular dementia and multi-infarct dementia. Am J Alzheimers Dis Other Demen. 2008;23(2):167-76. doi:10.1177/1533317507312558.
    » https://doi.org/10.1177/1533317507312558
  • 70
    Auchus A, Brashear HR, Salloway S, Korczyn A, De Deyn PP, Gassmann-Mayer C; GAL-INT-26 Study Group. Galantamine treatment of vascular dementia: a randomized trial. Neurology. 2007;69(5):448-58. doi:10.1212/01.wnl.0000266625.31615.f6.
    » https://doi.org/10.1212/01.wnl.0000266625.31615.f6
  • 71
    Narasimhalu K, Effendy S, Sim C, Lee JM, Chen I, Hia SB, et al. A randomized controlled trial of rivastigmine in patients with cognitive impairment no dementia because of cerebrovascular disease. Acta Neurol Scand. 2010;121(4):217-24. doi:10.1111/j.1600-0404.2009.01263.x.
    » https://doi.org/10.1111/j.1600-0404.2009.01263.x
  • 72
    Wilcock G, Möbius HJ, Stöffler A; MMM 500 group. A double-blind, placebo-controlled multicentre study of memantine in mild to moderate vascular dementia (MMM500). Int Clin Psychopharmacol. 2002;17(6): 297-305. doi:10.1097/00004850-200211000-00005.
    » https://doi.org/10.1097/00004850-200211000-00005
  • 73
    Reus VI, Fochtmann LJ, Eyler AE, Hilty DM, Horvitz-Lennon M, Jibson MD, et al. The American Psychiatric Association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173(5):543-6. doi:10.1176/appi.ajp.2015.173501.
    » https://doi.org/10.1176/appi.ajp.2015.173501
  • 74
    Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer's disease: an international Delphi consensus. Int Psychogeriatr. 2019;31(1):83-90. doi:10.1017/S1041610218000534.
    » https://doi.org/10.1017/S1041610218000534
  • 75
    Oliveira LF, Camargos EF, Martini LLL, Machado FV, Novaes MRCG. Use of psychotropic agents to treat agitation and aggression in Brazilian patients with Alzheimer's disease: A naturalistic and multicenter study. Psychiatry Res. 2021;295:113591. doi:10.1016/j.psychres.2020.113591.
    » https://doi.org/10.1016/j.psychres.2020.113591
  • 76
    Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal dementia. Int Rev of Psychiatry. 2013;25(2):130-7. doi:10.3109/09540261.2013.776523.
    » https://doi.org/10.3109/09540261.2013.776523
  • 77
    Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain. 2011;134(Pt 9):2456-77. doi:10.1093/brain/awr179.
    » https://doi.org/10.1093/brain/awr179
  • 78
    Trieu C, Gossink F, Stek ML, Scheltens P, Pijnenburg YAL, Dols A. Effectiveness of pharmacological interventions for symptoms of behavioral variant frontotemporal dementia: a systematic review. Cogn Behav Neurol. 2020;33(1):1-15. doi:10.1097/WNN.0000000000000217.
    » https://doi.org/10.1097/WNN.0000000000000217
  • 79
    Lima-Silva TB, Bahia VS, Carvalho VA, Guimarães HC, Caramelli P, Balthazar ML, et al. Neuropsychiatric symptoms, caregiver burden and distress in behavioral-variant frontotemporal dementia and Alzheimer's disease. Dement Geriatr Cogn Disord. 2015;40(5-6):268-75. doi:10.1159/000437351.
    » https://doi.org/10.1159/000437351
  • 80
    Wylie MA, Shnall A, Onyike CU, Huey ED. Management of frontotemporal dementia in mental health and multidisciplinary settings. Int Rev Psychiatry. 2013;25(2):230-6. doi:10.3109/09540261.2013.776949.
    » https://doi.org/10.3109/09540261.2013.776949
  • 81
    Marin SMC, Mansur LL, Oliveira FF, Marin LF, Wajman JR, Bahia VS, et al. Swallowing in behavioral variant frontotemporal dementia. Arq Neuropsiquiatr. 2021;79(1):8-14. doi:10.1590/0004-282X20200060.
    » https://doi.org/10.1590/0004-282X20200060
  • 82
    Portugal MG, Marinho V, Laks J. Pharmacological treatment of frontotemporal lobar degeneration: systematic review. Braz J Psychiatry. 2011:33(1):81-90. doi:10.1590/s1516-44462011000100016.
    » https://doi.org/10.1590/s1516-44462011000100016
  • 83
    Gambogi LB, Guimarães HC, Souza LC, Caramelli P. Treatment of the behavioral variant of frontotemporal dementia: a narrative review. Dement Neuropsychol. 2021;15(3):331-8. doi:10.1590/1980-57642021dn15-030004.
    » https://doi.org/10.1590/1980-57642021dn15-030004
  • 84
    Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, Cappa SF, et al. Classification of primary progressive aphasia and its variants. Neurology. 2011;76(11):1006-14. doi:10.1212/WNL.0b013e31821103e6.
    » https://doi.org/10.1212/WNL.0b013e31821103e6
  • 85
    Cotelli M, Manenti R, Ferrari C, Gobbi E, Macis A, Cappa SF. Effectiveness of language training and non-invasive brain stimulation on oral and written naming performance in Primary Progressive Aphasia: A meta-analysis and systematic review. Neurosci Biobehav Rev. 2020;108:498-525. doi:10.1016/j.neubiorev.2019.12.003.
    » https://doi.org/10.1016/j.neubiorev.2019.12.003
  • 86
    Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive brain stimulation (tDCS or TMS) paired with language therapy in the treatment of primary progressive aphasia: an exploratory meta-analysis. Brain Sci. 2020;10(9):597. doi:10.3390/brainsci10090597.
    » https://doi.org/10.3390/brainsci10090597
  • 87
    Beber BC, Brandão L, Chaves MLF. A warning to the Brazilian Speech-Language Pathology and Audiology community about the importance of scientific and clinical activities in primary progressive aphasia. CoDAS. 2015;27(5):505-8. doi:10.1590/2317-1782/20152015081.
    » https://doi.org/10.1590/2317-1782/20152015081
  • 88
    Carthery-Goulart MT, Silveira AC, Machado TH, Mansur LL, Parente MAMP, Senaha MLH, et al. Nonpharmacological interventions for cognitive impairments following primary progressive aphasia: A systematic review of the literature. Dement Neuropsychol. 2013;7(1):122-31. doi:10.1590/S1980-57642013DN70100018.
    » https://doi.org/10.1590/S1980-57642013DN70100018
  • 89
    Volkmer A, Spector A, Meitanis V, Warren JD, Beeke S. Effects of functional communication interventions for people with primary progressive aphasia and their caregivers: A systematic review. Aging Ment Health. 2020;24(9):1381-93. doi:10.1080/13607863.2019.1617246.
    » https://doi.org/10.1080/13607863.2019.1617246
  • 90
    Cadório I, Lousada M, Martins P, Figueiredo D. Generalization and maintenance of treatment gains in primary progressive aphasia (PPA): A systematic review. Int J Lang Commun Disord. 2017;52(5):543-60. doi:10.1111/1460-6984.12310.
    » https://doi.org/10.1111/1460-6984.12310
  • 91
    Machado TH, Carthery-Goulart MT, Campanha AC, Caramelli P. Cognitive intervention strategies directed to speech and language deficits in primary progressive aphasia: practice-based evidence from 18 cases. Brain Sci. 2021;11(10):1268. doi:10.3390/brainsci11101268.
    » https://doi.org/10.3390/brainsci11101268
  • 92
    Aarsland D. Cognitive impairment in Parkinson's disease and dementia with Lewy bodies. Parkinsonism Relat Disord. 2016;22(Suppl 1):S144-8. doi:10.1016/j.parkreldis.2015.09.034.
    » https://doi.org/10.1016/j.parkreldis.2015.09.034
  • 93
    Burn DJ. Cortical Lewy body disease and Parkinson's disease dementia. Curr Opin Neurol. 2006;19(6):572-9. doi:10.1097/01.wco.0000247607.34697.a2.
    » https://doi.org/10.1097/01.wco.0000247607.34697.a2
  • 94
    Outeiro TF, Koss DJ, Erskine D, Walker L, Kurzawa-Akanbi M, Burn D, et al. Dementia with Lewy bodies: an update and outlook. Mol Neurodegener. 2019;14(1):5. doi:10.1186/s13024-019-0306-8.
    » https://doi.org/10.1186/s13024-019-0306-8
  • 95
    Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018;125(4):615-50. doi:10.1007/s00702-017-1821-9.
    » https://doi.org/10.1007/s00702-017-1821-9
  • 96
    Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C, Mizuno Y, et al. Clinical diagnostic criteria for dementia associated with Parkinson's disease. Mov Disord. 2007;22(12):1689-707. doi:10.1002/mds.21507.
    » https://doi.org/10.1002/mds.21507
  • 97
    McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. doi:10.1212/WNL.0000000000004058.
    » https://doi.org/10.1212/WNL.0000000000004058
  • 98
    Broadstock M, Ballard C, Corbett A. Latest treatment options for Alzheimer's disease, Parkinson's disease dementia and dementia with Lewy bodies. Expert Opin Pharmacother. 2014;15(13):1797-810. doi:10.1517/14656566.2014.936848.
    » https://doi.org/10.1517/14656566.2014.936848
  • 99
    Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Pharmacological management of Lewy body dementia: a systematic review and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/appi.ajp.2015.14121582.
    » https://doi.org/10.1176/appi.ajp.2015.14121582
  • 100
    Connors MH, Quinto L, McKeith I, Brodaty H, Allan L, Bamford C, et al. Non-pharmacological interventions for Lewy body dementia: a systematic review. Psychol Med. 2018;48(11):1749-58. doi:10.1017/S0033291717003257.
    » https://doi.org/10.1017/S0033291717003257
  • 101
    Dugger BN, Boeve BF, Murray ME, Parisi JE, Fujishiro H, Dickson DW, et al. Rapid eye movement sleep behavior disorder and subtypes in autopsy-confirmed dementia with Lewy bodies. Mov Disord. 2012;27(1):72-8. doi:10.1002/mds.24003.
    » https://doi.org/10.1002/mds.24003
  • 102
    Ohma E, Ikuta F. Parkinson's disease: distribution of Lewy bodies and monoamine neuron system. Acta Neuropathol. 1976;34(4):311-9. doi:10.1007/BF00696560.
    » https://doi.org/10.1007/BF00696560
  • 103
    Hughes AJ, Daniel SE, Blankson S, Lees AJ. A clinicopathological study of 100 cases of Parkinson's disease. J Neurol Neurosurg Psychiatry. 1992;55(3):181-4. doi:10.1136/jnnp.55.3.181.
    » https://doi.org/10.1136/jnnp.55.3.181
  • 104
    Rongve A, Aarsland D. Management of Parkinson's disease dementia: practical considerations. Drugs Aging. 2006;23(10):807-22. doi:10.2165/00002512-200623100-00004.
    » https://doi.org/10.2165/00002512-200623100-00004
  • 105
    Caviness JN, Lue L, Adler CH, Walker DG. Parkinson's disease dementia and potential therapeutic strategies. CNS Neurosci Ther. 2011;17(1):32-44. doi:10.1111/j.1755-5949.2010.00216.x.
    » https://doi.org/10.1111/j.1755-5949.2010.00216.x
  • 106
    Gauthier S. Pharmacotherapy of Parkinson disease dementia and Lewy body dementia. Front Neurol Neurosci. 2009;24:135-9. doi:10.1159/000197892.
    » https://doi.org/10.1159/000197892
  • 107
    Garcia-Ptacek S, Kramberger MG. Parkinson disease and dementia. J Geriatr Psychiatry Neurol. 2016;29(5):261-70. doi:10.1177/0891988716654985.
    » https://doi.org/10.1177/0891988716654985
  • 108
    Sezgin M, Bilgic B, Tinaz S, Emre M. Parkinson's disease dementia and Lewy body disease. Semin Neurol. 2019;39(2):274-82. doi:10.1055/s-0039-1678579.
    » https://doi.org/10.1055/s-0039-1678579
  • 109
    Taylor JP, McKeith IG, Burn DJ, Boeve BF, Weintraub D, Bamford C, et al. New evidence on the management of Lewy body dementia. Lancet Neurol. 2020;19(2):157-69. doi:10.1016/S1474-4422(19)30153-X.
    » https://doi.org/10.1016/S1474-4422(19)30153-X
  • 110
    Velayudhan L, Ffytche D, Ballard C, Aarsland D. New therapeutic strategies for Lewy body dementias. Curr Neurol Neurosci Rep. 2017;17(9):68. doi:10.1007/s11910-017-0778-2.
    » https://doi.org/10.1007/s11910-017-0778-2
  • 111
    Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase inhibitors for dementia with Lewy bodies, Parkinson's disease dementia and cognitive impairment in Parkinson's disease. Cochrane Database Syst Rev. 2012;(3):CD006504. doi:10.1002/14651858.CD006504.pub2.
    » https://doi.org/10.1002/14651858.CD006504.pub2

Publication Dates

  • Publication in this collection
    28 Nov 2022
  • Date of issue
    Sept 2022

History

  • Received
    20 Feb 2022
  • Reviewed
    27 Apr 2022
  • Accepted
    27 Apr 2022
Academia Brasileira de Neurologia, Departamento de Neurologia Cognitiva e Envelhecimento R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices, Torre Norte, São Paulo, SP, Brazil, CEP 04101-000, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revistadementia@abneuro.org.br | demneuropsy@uol.com.br