Dementia Neuropsy

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 121

Volume 16 • Number 3 Suppl.

1
September 2022
São Paulo • Brazil

OFFICIAL JOURNAL OF THE SCIENTIFIC DEPARTMENT OF COGNITIVE NEUROLOGY AND AGING OF THE BRAZILIAN ACADEMY OF NEUROLOGY

Consensus

1 Subjective cognitive decline, mild cognitive impairment,


and dementia - syndromic approach

21 Diagnosis of Alzheimer’s disease

36 Diagnosis of frontotemporal dementia

49 Diagnosis of vascular cognitive impairment

69 Diagnosis and management of Parkinson’s disease dementia


and dementia with Lewy bodies

83 Treatment of dementia

96 Management in severe dementia


Volume 16 • Number 03
September 2022
São Paulo • Brazil

OFFICIAL JOURNAL OF THE SCIENTIFIC DEPARTMENT OF COGNITIVE NEUROLOGY AND AGING OF THE BRAZILIAN ACADEMY OF NEUROLOGY

EDITORIALEditors
BOARD
Ricardo Nitrini Sonia Maria Dozzi Brucki
Editor-in-Chief
University of São Paulo, São Paulo SP, Brazil University of São Paulo, São Paulo SP, Brazil

Ricardo Nitrini Associate Editors Sonia Maria Dozzi Brucki


Universidade de São Paulo, São Paulo, São Paulo, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil
Eliane Correa Miotto Mônica Sanches Yassuda
University of São Paulo, São Paulo SP, Brazil University of São Paulo, São Paulo SP, Brazil
Co-Editors
Section Editors
Eliane Correa Miotto Mônica Sanches Yassuda
Universidade deHistory
São Paulo,Note
São Paulo, São Paulo, Brazil Neuroimaging through
Universidade clinical
de São Paulo, Sãocases
Paulo, São Paulo, Brazil
http://lattes.cnpq.br/6923820631915917
Eliasz Engelhardt Marcio Luiz Figueredo Balthazar http://lattes.cnpq.br/0754786947169577
Leandro Tavares Lucato
https://orcid.org/0000-0003-2711-1627 https://orcid.org/0000-0002-9182-2450
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil University of Campinas, Campinas SP, Brazil University of São Paulo, São Paulo SP, Brazil

Advisory Editorial Board José Luiz Consultive Board


Sá Cavalcanti Renato Anghinah
Alexandre Castro-Caldas Jerusa Federal
Smid University of Rio de Janeiro, Rio de Janeiro RJ, Brazil UniversityVicente
Orestes of São Paulo, São Paulo SP, Brazil
Forlenza
André
Universidade Católica Palmini
Portuguesa, Lisboa, Portugal Márcia
Universidade Lorena
de São Paulo, Fagundes
São Paulo, São Paulo,Chaves
Brazil Wilson Jacob
Universidade Filho
de São Paulo, São Paulo, São Paulo, Brazil
Catholic University
André Luis Fernandes of Rio Grande do Sul, Porto Alegre RS, Brazil
Palmini João Carlos
FederalBarbosa
University Machado
of Rio Grande do Sul, Porto Alegre RS, Brazil Patricio
UniversityFuentes
of São Paulo, São Paulo SP, Brazil
Claudia Sellitto Porto Paulo
Universidade Caramelli
Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil Universidad de Chile, Santiago, Chile
Andrea Camaz Deslandes
University of São Paulo, São Paulo SP, Brazil John R.Federal
HodgesUniversity of Minas Gerais, Belo Horizonte MG, Brazil Paulo Caramelli
Universidade Federal
Jersondo Rio Laks
de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil University of New South Wales, Sydney, Australia
Paulo Henrique Ferreira Bertolucci
Andrew J. Lees
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil John C.Federal
MorrisUniversity of São Paulo, São Paulo SP, Brazil Paulo Henrique Ferreira Bertolucci
University College London, London, United Kingdom Washington University School of Medicine, Saint Louis, United States of America
Benito Pereira Damasceno
Paulo Roberto de Brito Marques
Editorial
Universidade de Campinas, Board
Campinas, São Paulo, Brazil Unidade deJamary Oliveira
Neurociência Filho e Cognitiva, Rio de Janeiro, Rio de
Comportamental Moyses Chaves
Universidade Estadual de Pernambuco, Recife, Pernambuco, Brazil
Breno Satler de Oliveira Diniz Janeiro, Brazil
Federal University of Bahia, Salvador BA, Brazil Federal University of Goiás, Goiânia GO, Brazil
Alexandre
University of Texas Health ScienceCastro-Caldas
Center at Houston, Houston, TX, United José Luiz Sá Calvalcanti
Jerusa Smid Orestes Vicente Forlenza
States of AmericaPortuguese Catholic University, Lisbon, Portugal
University of São Paulo, São Paulo SP, Brazil University of São Paulo, São Paulo SP, Brazil
Cláudia da Costa LeiteCamaz Deslandes Kenichi Meguro Ricardo F. Allegri
Andrea
Universidade de São Paulo, São Paulo, São Paulo, Brazil João Carlos
Tohoku University, Barbosa Machado
Sendai, Japan PatriciodeFuentes
Universidad Buenos Aires, Buenos Aires, Argentina
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil Faculty of Medical Sciences of Minas Gerais, Belo Horizonte MG, Brazil University of Chile, Santiago, Chile
Claudia Sellitto Porto Leila Maria Cardao Chimelli Ricardo de Oliveira Souza
Andrew Lees John
Universidade R. doHodges
Federal Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil PaulodeR.
Unidade de BritoComportamental
Neurociência Marques de Cognitiva, Rio de Janeiro, Rio
University
Dora Selma Fix VenturaCollege London, London, UK Leonardo Ferreira
University Caixeta
of New South Wales, Sydney, Australia deState
Janeiro, Brazil of Pernambuco, Recife PE, Brazil
University
Benito
Universidade de São Paulo, Pereira Damasceno
São Paulo, São Paulo, Brazil John
Universidade C. deMorris
Federal Goiás, Goiânia, GO, Brazil Sandra Weintraub
Ricardo F. Allegri
Facundo F. Manes Leonel Tadao Takada Northwestern University, Chicago, United States of America
Instituto de Neurología, Buenos Aires, Argentina Universidade de São Paulo, São Paulo, São Paulo, Brazil Sérgio Luís Blay
Francisco de Assis Carvalho do Vale Márcia Lorena Fagundes Chaves Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil Sergio Teixeira Ferreira
Francisco Javier Lopera Restrepo Marcia Radanovic Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
University of Antioquia, Medelín, Colombia Universidade de São Paulo, São Paulo, São Paulo, Brazil
Tânia Marcourakis
Giacomo Rizzolatti Maria Alice de Mattos Pimenta Parente Universidade de São Paulo, São Paulo, São Paulo, Brazil
Università degli Studi di Parma, Parma, Italy Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
Tales Alexandre Aversi-Ferreira
Helenice Charchat-Fichman Maria Rita dos Santos e Passos Bueno Universidade Federal de Goiás, Catalão, GO, Brazil
Universidade Estácio de Sá, Rio de Janeiro, Rio de Janeiro, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil
Henrique Cerqueira Guimarães Mayana Zatz Thomas H. Bak
Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil University of Edinburgh, Edinburgh, United Kingdom
Howard Chertkow Michael D. Geschwind Vilma Regina Martins
McGill University, Montreal, Quebec, Canada University of California, San Francisco, United States of America Instituto de Pesquisa Ludwig, São Paulo, São Paulo, Brazil
Jamary Oliveira Filho M.-Marsel Mesulam Wilson Jacob Filho
Universidade Federal da Bahia, Salvador, Bahia, Brazil Northwestern University, Chicago, United States of America
Jerson Laks Moyses de Paul a Rodrigues Chaves Yves Joanette
Universidade Federal de Goiás, Goiânia, GO, Brazil University of Montreal, Quebec, Canada

Denise Ieiri de Moraes


Executive Editor Editorial Assistant
fabiana.montanari@abneuro.org.br dementia@abneuro.org.br
Associate editors
• Behavioral disorders in dementia Karolina Gouveia César Freitas
Analuiza Camozzatto de Pádua Universidade de Taubaté, Taubaté, São Paulo, Brazil
Universidade Federal de Ciências de Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil Nilton Santos Custodio Capuñay
Peruvian Institute of Neurosciences, Lima, Peru
Antonio Lucio Teixeira Junior
University of Houston, Texas, United States of America
• Clinical Trials
Florindo Stella Pablo Miguel Bagnati
Universidade Estadual Paulista, Rio Claro, São Paulo, Brazil Universidad de Buenos Aires, Buenos Aires, Argentina

• Biomarkers in dementia diagnosis • Genetics


Ezequiel Surace Leonel Tadao Takada
University of Buenos Aires, Buenos Aires, Argentina Universidade de São Paulo, São Paulo, Brazil
Breno Satler de Oliveira Diniz Mauricio Arcos-Burgos
University of Texas Health Science Center at Houston, Houston, TX, United States of America Universidad de Antioquia, Medellín, Colombia
Maria Niures Pimentel dos Santos Matioli
Centro Universitário Lusíada, Santos, São Paulo, Brazil • Neuroimaging
Artur Martins Novaes Coutinho
• Mild cognitive impairment Universidade de São Paulo, São Paulo, São Paulo, Brazil
Renata Kochhann Douglas Mendes Nunes
Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil

• Caregiver: stress and orientation • Neuroimaging through Clinical cases


Marcia Cristina Nascimento Dourado Marcio Luiz Figueredo Balthazar
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil Universidade de Campinas, Campinas, São Paulo, Brazil
Thaís Bento Lima da Silva Leandro Tavares Lucato
Universidade de São Paulo, São Paulo, São Paulo, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil

• Cognition and Aging • Language and Aphasia


Lilian Schafirovits Morillo Marcela Lima Silagi de Siqueira
Universidade de São Paulo, São Paulo, São Paulo, Brazil Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil

Maira Tonidandel Barbosa Mirna Li Hosogi


Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil Universidade de São Paulo, São Paulo, São Paulo, Brazil

• Dementia costs • Neuropathology


Ceres Eloah de Lucena Ferretti Lea Tenenholz Grinberg
Universidade de São Paulo, São Paulo, SP Brazil University of California, San Francisco, United States of America
Roberta Diehl-Rodriguez
• Lewy Body Dementia/ Parkinson Disease/Atypical parkinsonism Universidade de São Paulo, São Paulo, São Paulo, Brazil
Jacy Bezerra Parmera
Universidade de São Paulo, São Paulo, São Paulo, Brazil • Developmental Neuropsychology
Rubens Gisbert Cury Claudia Berlin de Mello
Universidade de São Paulo, São Paulo, São Paulo, Brazil Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil

Vitor Tumas Maria Joana Mader Joaquim


Universidade de São Paulo, Ribeirão Preto, S]ao Paulo, Brazil Universidade Federal do Paraná, Curitiba, Paraná, Brazil
Vitor Geraldi Haase
• Frontotemporal Dementia Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
Leonardo Cruz de Souza
Universidade Federal de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil • Neuropsychology: diagnostic tests
Valéria Santoro Bahia Mario Amore Cecchini
University of Edinburgh, Edinburgh, United Kingdom
Universidade de São Paulo, São Paulo, São Paulo, Brazil
Valeska Marinho Rodrigues • History Note
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil, Eliasz Engelhardt
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
• Vascular Dementia
Claudia Kimie Suemoto • Rehabilitation
Universidade de São Paulo, São Paulo São Paulo, Brazil Cláudia Maia Memória
Universidade de São Paulo, São Paulo, São Paulo, Brazil
Lucas Porcello Schilling
Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
• Subjective cognitive decline
• Diagnosis in indigenous and low education populations Adalberto Studart-Neto
Universidade de São Paulo, São Paulo São Paulo, Brazil
Elisa de Paula França Resende
Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil Wyllians Jose Vendramini Borelli
Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
Juliana Nery de Souza-Talarico
Universidade de São Paulo, São Paulo, São Paulo, Brazil Eduardo Sturzeneker Trés
Universidade de São Paulo, São Paulo, São Paulo, Brazil
Maíra Okada de Oliveira
Universidade de São Paulo, São Paulo, São Paulo, Brazil
• Translation and Transcultural Validation
• Epidemiology Marcia Maria Pires Camargo Novelli
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
Alessandro Ferrari Jacinto
Universidade Estadual Paulista, Botucatu, São Paulo, Brazil • Social Media and Visual Abstract Editors
Cleusa Pinheiro Ferri Raphael Machado de Castilhos
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
Déborah Oliveira Ricardo Pires Alvim
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
Volume 16 • Number 3 Suppl.1
September 2022
São Paulo • Brazil

OFFICIAL JOURNAL OF THE SCIENTIFIC DEPARTMENT OF COGNITIVE NEUROLOGY AND AGING OF THE BRAZILIAN ACADEMY OF NEUROLOGY

EDITORIALEditors
BOARD
Ricardo Nitrini Sonia Maria Dozzi Brucki
University of São Paulo, São Paulo SP, Brazil University of São Paulo, São Paulo SP, Brazil

Jerusa Smid Breno José Alencar Pires Barbosa


Associate Editors Lucas Porcello Schilling
Universidade de São Paulo, São Paulo SP, Brazil Universidade Federal de Pernambuco, Recife PE, Brazil Pontifícia Universidade do Rio Grande do Sul, Porto Alegre RS, Brazil
Eliane Correa Miotto Mônica Sanches Yassuda
University of São Paulo, São Paulo SP, Brazil University of São Paulo, São Paulo SP, Brazil
CONSULTIVE BOARD
Section Editors
History Note Neuroimaging through clinical cases
Adalberto Studart-Neto Eliasz Engelhardt Marcio
Jerson LaksLuiz Figueredo Balthazar Leandro Tavares
Mônica Lucato
Sanches Yassuda
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil
Benito Pereira Damasceno José University
IbiapinaofSiqueira
Campinas, Campinas SP, Brazil
Neto University of São Paulo, São Paulo SP, Brazil
Norberto Anízio Ferreira Frota
Ceres Eloah de Lucena Ferretti Karolina Gouveia César-Freitas Orestes Vicente Forlenza
Advisory Editorial Board José Luiz Sá Cavalcanti Renato Anghinah
Claudia Kimie Suemoto Leonardo Cruz de Souza
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil
Paulo Caramelli
University of São Paulo, São Paulo SP, Brazil
AndréCamargos
Einstein Francisco Palmini MárciaLeonel TadaoFagundes
Lorena Takada Chaves Wilson Jacob FilhoPaulo Henrique Ferreira Bertolucci
Catholic University of Rio Grande do Sul, Porto Alegre RS, Brazil
Eliasz Engelhardt FederalLilian Schafirovits
University of Rio GrandeMorillo
do Sul, Porto Alegre RS, Brazil University of São Paulo, SãoRaphael Machado Castilhos
Paulo SP, Brazil
Claudia
Elisa de Paula França Sellitto
ResendePorto Paulo Caramelli
Maira Tonidandel Barbosa Renata Kochhann
University of São Paulo, São Paulo SP, Brazil Federal University of Minas Gerais, Belo Horizonte MG, Brazil
Felipe Kenji Sudo Marcela Lima Silagi Ricardo Nitrini
Jerson Laks Paulo Henrique Ferreira Bertolucci
Fernanda Tovar-Moll
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil Marcia Cristina Nascimento Dourado
Federal University of São Paulo, São Paulo SP, Brazil
Rodrigo Rizek Schultz
Florindo Stella Márcia Lorena Fagundes Chaves Sonia Maria Dozzi Brucki
Francisco Assis Carvalho
Editorial Vale
Board JamaryMárcia Radanovic
Oliveira Filho Moyses Chaves Thais Helena Machado
Gilberto Sousa Alves FederalMárcio
UniversityLuiz Figueiredo
of Bahia, Balthazar
Salvador BA, Brazil Federal University of Goiás,Valéria Santoro
Goiânia GO, Brazil Bahia
Alexandre
Helen Bedinoto DurganteCastro-Caldas JerusaMarcusSmidVinicius Della Coletta Orestes Vicente Forlenza Valeska Marinho
Portuguese Catholic University, Lisbon, Portugal University of São Paulo, São Paulo SP, Brazil University of São Paulo, SãoVictor
Paulo SP, Brazil
Henrique Ballalai Ferraz Maria Teresa Carthery-Goulart Calil da Silveira
Andrea Camaz Deslandes JoãoMariana
Carlos Spitz
Barbosa Machado Patricio Fuentes Vitor Tumas
Ivan Aprahamian
Federal University of Rio de Janeiro, Rio de Janeiro RJ, Brazil Faculty of Medical Sciences of Minas Gerais, Belo Horizonte MG, Brazil University of Chile, Santiago, Chile
Jacy BezerraAndrew
ParmeraLees JohnMirna Lie Hosogi
R. Hodges Wyllians Vendramini Borelli
Paulo R. de Brito Marques
University College London, London, UK University of New South Wales, Sydney, Australia State University of Pernambuco, Recife PE, Brazil
Consensus
CONTENTS
1 Subjective cognitive decline, mild cognitive impairment, and dementia - syndromic approach: recommendations
of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology
Jerusa Smid, Adalberto Studart-Neto, Karolina Gouveia César-Freitas, Marcia Cristina Nascimento Dourado,
Renata Kochhann, Breno José Alencar Pires Barbosa, Lucas Porcello Schilling, Márcio Luiz Figueiredo Balthazar,
Norberto Anízio Ferreira Frota, Leonardo Cruz de Souza, Paulo Caramelli, Paulo Henrique Ferreira Bertolucci,
Márcia Lorena Fagundes Chaves, Sonia Maria Dozzi Brucki, Ricardo Nitrini, Elisa de Paula França Resende, Francisco Assis Carvalho Vale

21 Diagnosis of Alzheimer’s disease: recommendations of the Scientific Department of Cognitive Neurology and
Aging of the Brazilian Academy of Neurology
Lucas Porcello Schilling, Marcio Luiz Figueredo Balthazar, Márcia Radanovic,
Orestes Vicente Forlenza, Marcela Lima Silagi, Jerusa Smid, Breno José Alencar Pires Barbosa, Norberto Anízio
Ferreira Frota, Leonardo Cruz de Souza, Francisco Assis Carvalho Vale, Paulo Caramelli, Paulo Henrique Ferreira
Bertolucci, Márcia Lorena Fagundes Chaves,Sonia Maria Dozzi Brucki, Benito Pereira Damasceno, Ricardo Nitrini

36 Diagnosis of frontotemporal dementia: recommendations of the Scientific Department of Cognitive Neurology and
Aging of the Brazilian Academy of Neurology
Leonardo Cruz de Souza, Mirna Lie Hosogi, Thais Helena Machado, Maria Teresa Carthery-Goulart, Mônica Sanches
Yassuda, Jerusa Smid,Breno José Alencar Pires Barbosa, Lucas Porcello Schilling, Marcio Luiz Figueredo Balthazar,
Norberto Anízio Ferreira Frota, Francisco Assis Carvalho Vale, Paulo Caramelli, Paulo Henrique Ferreira Bertolucci,
Márcia Lorena Fagundes Chaves, Sonia Maria Dozzi Brucki, Ricardo Nitrini,Valéria Santoro Bahia, Leonel Tadao Takada

49 Diagnosis of vascular cognitive impairment: recommendations of the scientific department of cognitive neurology and
aging of the Brazilian Academy of Neurology
Breno José Alencar Pires Barbosa, José Ibiapina Siqueira Neto, Gilberto Sousa Alves, Felipe Kenji Sudo, Claudia Kimie Suemoto,
Fernanda Tovar-Moll, Jerusa Smid, Lucas Porcello Schilling, Marcio Luiz Figueredo Balthazar, Norberto Anízio Ferreira Frota,
Leonardo Cruz de Souza, Francisco Assis Carvalho Vale, Paulo Caramelli, Paulo Henrique Ferreira Bertolucci,
Sonia Maria Dozzi Brucki, Ricardo Nitrini, Eliasz Engelhardt, Márcia Lorena Fagundes Chaves

69 Diagnosis and management of Parkinson’s disease dementia and dementia with Lewy bodies: recommendations
of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology
Jacy Bezerra Parmera, Vitor Tumas, Henrique Ballalai Ferraz, Mariana Spitz, Maira Tonidandel Barbosa, Jerusa Smid,
Breno José Alencar Pires Barbosa, Lucas Porcello Schilling, Márcio Luiz Figueiredo Balthazar, Leonardo Cruz de Souza,
Francisco Assis Carvalho Vale, Paulo Caramelli, Paulo Henrique Ferreira Bertolucci, Márcia Lorena Fagundes Chaves,
Sonia Maria Dozzi Brucki, Ricardo Nitrini, Raphael Machado Castilhos, Norberto Anízio Ferreira Frota

83 Treatment of dementia: recommendations of the Scientific Department of Cognitive


Neurology and Aging of the Brazilian Academy of Neurology
Paulo Caramelli, Valeska Marinho, Jerson Laks, Marcus Vinicius Della Coletta, Florindo Stella, Einstein Francisco Camargos,
Jerusa Smid, Breno José Alencar Pires Barbosa, Lucas Porcello Schilling, Marcio Luiz Figueredo Balthazar,
Norberto Anízio Ferreira Frota, Leonardo Cruz de Souza, Francisco Assis Carvalho Vale, Márcia Lorena Fagundes Chaves,
Sonia Maria Dozzi Brucki, Ricardo Nitrini,Helen Bedinoto Durgante, Paulo Henrique Ferreira Bertolucci

96 Management in severe dementia: recommendations of the Scientific Department of Cognitive Neurology and
Aging of the Brazilian Academy of Neurology
Sonia Maria Dozzi Brucki, Ivan Aprahamian, Wyllians Vendramini Borelli, Victor Calil da Silveira, Ceres Eloah de Lucena Ferretti,
Jerusa Smid, Breno José Alencar Pires Barbosa, Lucas Porcello Schilling,Márcio Luiz Figueiredo Balthazar, Norberto Anízio Ferreira Frota,
Leonardo Cruz de Souza, Francisco Assis Carvalho Vale, Paulo Caramelli, Paulo Henrique Ferreira Bertolucci,
Márcia Lorena Fagundes Chaves, Ricardo Nitrini, Rodrigo Rizek Schultz, Lilian Schafirovits Morillo
Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S101EN

Subjective cognitive decline,


mild cognitive impairment,
and dementia - syndromic approach:
recommendations of the Scientific
Department of Cognitive Neurology
and Aging of the Brazilian Academy of Neurology
Jerusa Smid1 , Adalberto Studart-Neto1 , Karolina Gouveia César-Freitas1 ,
Marcia Cristina Nascimento Dourado2 ,Renata Kochhann3 , Breno José Alencar Pires Barbosa1,4,5 ,
Lucas Porcello Schilling6,7,8 , Márcio Luiz Figueiredo Balthazar9 , Norberto Anízio Ferreira Frota10,11 ,
Leonardo Cruz de Souza12 , Paulo Caramelli12 , Paulo Henrique Ferreira Bertolucci13 ,
Márcia Lorena Fagundes Chaves14,15 , Sonia Maria Dozzi Brucki1 , Ricardo Nitrini1 ,
Elisa de Paula França Resende16 , Francisco Assis Carvalho Vale17

1
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
2
Universidade Federal do Rio de Janeiro,Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil.
3
Hospital Moinhos de Vento, Porto Alegre RS, Brazil.
4
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
5
Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.
6
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
7
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
8
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
9
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil.
10
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
11
Universidade de Fortaleza, Fortaleza CE, Brazil.
12
Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
13
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil.
14
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
15
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina,Departamento de Medicina Interna, Porto Alegre RS, Brazil.
16
Universidade Federal de Minas Gerais, Hospital das Clínicas, Belo Horizonte MG, Brazil.
17
Universidade Federal de São Carlos, Departamento de Medicina, Centro de Ciências Biológicas da Saúde, São Carlos SP, Brazil.
Correspondence: Jerusa Smid; Email: jsmid77@hotmail.com.
Conflict of interest: JS: Participation as speaker in symposia sponsored by Roche laboratory; LPS: Participation in advisory boards for Biogen. Participation as
speaker in symposia sponsored by Aché, Apsen, and Biogen laboratories; MLFB: Participation in advisory boards for Biogen. Development of material for continuous
medical education and participation as speaker in symposia sponsored by EMS and Torrent laboratories; PC: Participation as principal investigator in clinical trials
for the Novo Nordisk and Roche laboratories. Participation in advisory boards for the Aché, Biogen, EMS, Nutricia, and Roche laboratories. Development of material
for continuous medical education and participation as speaker in symposia sponsored by the Aché, Nutricia, Libbs, Roche, Sandoz, Torrent, and Zodiac laboratories;
PHFB: Participation in advisory boards for the Biogen and Novo Nordisk laboratories. Supervision of training activities for Biogen, Janssen-Cilag, and Novo Nordisk
laboratories and for Quintiles. Participation as speaker in symposia sponsored by the Apsen, Nutricia, Roche, and Sandoz laboratories; EPFR: funding by the
Alzheimer’s Association, the World Federation of Neurology, and the National Institute of Health (grant number R21AG069252); LCS: Participation in advisory board
for Biogen. Participation as speaker in symposia sponsored by Biogen; RN: Participation in advisory board for Biogen; KGCF, MCND, RK, ASN, BJAPB, NAFF, MLFC,
SMDB, FACV: There is no conflict of interest to declare.
Received on July 18, 2021; Received in its final form on January 03, 2022; Accepted on April 27, 2022.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   1


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

ABSTRACT. This consensus, performed by the Brazilian Academy of Neurology (BAN) will approach practically how to evaluate patients with cognitive complaints
and how to clinically and etiologically diagnose the three clinical syndromes associated with the different stages of cognitive decline: subjective cognitive
decline (SCD), mild cognitive impairment (MCI), and dementia. This BAN consensus discusses SCD diagnosis for the first time, updates MCI and dementia
diagnoses, recommends the adequate cognitive tests and the relevant etiological work-up and care of patients with cognitive decline at different levels of
care within the Brazilian Unified Health System. We also review the main assessment instruments used in Brazil and Latin America.
Keywords: Dementia; Cognitive Dysfunction; Neuropsychological Tests.

DECLÍNIO COGNITIVO SUBJETIVO, COMPROMETIMENTO COGNITIVO LEVE E DEMÊNCIA: DIAGNÓSTICO SINDRÔMICO: RECOMENDAÇÕES DO DEPARTAMENTO
CIENTÍFICO DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. Este consenso realizado pela Academia Brasileira de Neurologia (ABN) abordará de maneira prática como avaliar pacientes com queixas cognitivas
e como realizar o diagnóstico clínico e etiológico das três síndromes clínicas associadas aos estágios de declínio cognitivo: declínio cognitivo subjetivo (DCS),
comprometimento cognitivo leve (CCL) e demência. O diagnóstico de DCS é discutido pela primeira vez em consenso da ABN e as atualizações para
o diagnóstico de CCL e demência são abordadas, bem como a recomendação para o uso de testes cognitivos apropriados, investigação etiológica pertinente
e cuidados aos pacientes com declínio cognitivo nos diferentes níveis de atenção do Sistema Único de Saúde. Foi realizada pesquisa dos principais
instrumentos de avaliação utilizados em nosso meio e na América Latina.
Palavras-chave: Demência; Disfunção Cognitiva; Testes Neuropsicológicos.

INTRODUCTION APPROACH TO PATIENTS WITH COGNITIVE AND

W hen we evaluate persons with cognitive complaints


(self-reported or referred by an informant),
the first question we must answer is whether they
BEHAVIORAL COMPLAINTS

Medical history
have dementia. Dementia is defined as a syndrome Clinical evaluation begins with patients’ medical history.
characterized by cognitive and/or behavioral decline in The medical history should also be taken with a family
which symptoms interfere with activities of daily living member or other close informants because anosognosia
(ADL) causing functional impairment when compared (the inability to the recognize cognitive impairment)
with previous functionality and that is not explained is relatively common. Interviews with patients and their
by delirium or major psychiatric1. informants should be carried out separately whenever
Mild cognitive impairment (MCI) is a condition in possible to address all complaints without embarrassing
which the individual has a cognitive impairment that does patients and their informant. Physicians should ask specific
not interfere with their autonomy in ADL performance. questions to identify which cognitive domains are affected,
They may experience slight difficulties performing complex whether patients show behavioral symptoms, and how
tasks that used to be trivial but still can maintain their their cognitive decline impacts patients’ functionality.
Investigable cognitive domains by medical history include
independence with minimal assistance2. More recently,
memory, attention, visual-spatial functions, praxis,
research has described a new syndrome for a group
executive functions, and language (Table 1).
of individuals who have cognitive complaints (mainly
During medical history, physicians should also
memory) but who, when tested, show normal performance
assess conditions that can cause a false impression
in neuropsychological tests. This situation is called
of cognitive impairment, such as untreated hearing
subjective cognitive decline (SCD)3.
or visual impairment. Furthermore, symptoms that may
Therefore, dementia, MCI, and SCD are mainly suggest delirium, such as fluctuation of the arousal and
clinical diagnoses. Physicians should be able to diagnose attention levels should be observed. Clinicians should
cognitive-behavioral syndromes during clinical ask patients about their use of medications, including
appointments after taking patients’ medical history and over-the-counter drugs since many medications with
performing cognition and functionality assessment. anticholinergic or sedative effects can cause or worsen
Laboratory testing and neuroimaging are helpful to cognitive impairment. Moreover, clinicians should enquire
define the etiology of the syndrome. Figure 1 illustrates about patients’ sleep pattern in search for symptoms
the continuum of cognitive decline. This study aims of obstructive apnea, REM sleep behavior disorder,
1) to propose a standardized assessment of patients and daytime sleepiness. Dysautonomia symptoms such
with cognitive complaints; 2) to present the diagnostic as urinary or, bowel dysfunction, erectile dysfunction
criteria for dementia, MCI, and SCD; and 3) to propose and postural hypotension should also be investigated.
a flowchart for investigating cognitive decline within Motor symptoms, such as changes in gait and balance,
the Brazilian Unified Health System (SUS). and sensory symptoms should be investigated.

2   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Self-perception of cognitive decline

Normal aging
Cognitive performance

Independence in activities of daily living

Pathological aging

Normal Subjective Cognitive Decline Mild Cognitive Impairment Dementia

Age (years)
Figure 1. Continuum of cognitive decline in normal and pathological aging.

Table 1. Targeted medical history of patients with cognitive and behavioral complaints
Does the patient get more confused or disoriented sometimes?
Attention Is the patient easily distracted?
Is the patient missing objects?

Does the patient have more difficulties remembering recent facts than remote ones?
Has the patient shown temporal disorientation?
Memory Is the patient more repetitive and asking the same questions many times?
Does the patient need more annotations to remember appointments?
Has the patient forgotten appointments?

Does the patient have difficulties finding words?


Is the patient having difficulties naming objects?
Cognitive Language
Is the patient having difficulties articulating words?
symptoms Is the patient having difficulties understanding what you say to them?

Has the patient got lost in previously known routes?


Spatial orientation Is the patient having difficulties learning new routes?
Is the patient having difficulties locating themselves inside the house?

Is the patient having difficulties using utensils?


Praxies
Is the patient having difficulties dressing themselves?

Is the patient having difficulties planning or organizing activities and travels?


Is the patient having difficulties multitasking?
Executive functions
Is the patient having difficulties solving daily problems?
Is the patient having difficulties making decisions?

Is the patient having difficulties managing their own finances?


Instrumental activities of Is the patient having difficulties shopping?
daily living (IADLs) Is the patient having problems at work?
Functionality Is the patient having cooking problems?

Basic activities of daily Is the patient capable of doing the activities below independently?
living (BADLs) Dressing/Bathing/Hygiene/Feeding/Transference/Continence
Continue...

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   3


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Table 1. Continuation.
Does the patient look sad, dejected or cry easily?
Does the patient not see pleasure in life or says they have no future?
Humor
Is the patient more irritable or impatient?
Is the patient isolating themself, not getting along with others?

Is the patient worried about planned events?


Anxiety Is the patient unable to relax or is excessively tense?
Does the patient worry excessively about even trivial things?

Does the patient have no interest in the world around them?


Apathy Does the patient have more difficulty engaging in conversations or tasks?
Is the patient more indifferent?

Does the patient act impulsively, without thinking?


Has the patient been saying things that should not be said in public?
Disinhibition
Has the patient acted in an embarrassing way?
Did the patient suffer personality changes? Is the patient more socially isolated?

Is the patient uncooperative?


Behavior and Does the patient not allow to be helped?
neuropsychiatric Agitation Is the patient verbally or physically aggressive?
symptoms Does the patient repeatedly move objects around them?
Does the patient have ritualistic or compulsive behavior?

Does the patient believe in things that are not real?


Does the patient think someone is trying to harm or rob him?
Delusions
Does the patient claim that their relatives are not who they say they are?
Does the patient claim that the house they live in is not theirs?

Does the patient report hearing voices or act as if they heard voices that are not heard by others?
Does the patient speak to themself?
Hallucinations
Does the patient see people or animals that are not seen by others?
Does the patient behave as if they saw something that others do not see?

Did the patient have any change in eating habits?


Appetite
Has the patient changed food preference (e.g., they started to prefer sweets)?

Does the patient have difficulty initiating or maintaining sleep?


Does the patient speak or move in sleep as if they were awake?
Sleep
Does the patient often have vivid dreams or nightmares?
Does the patient snore, wake up fatigued, or have daytime sleepiness?

Cognitive screening tests Other cognitive screening tests that have been
Cognitive screening tests consist of brief structured validated in Brazil are: The Blessed Information-Memory-
instruments which enable physicians to globally assess Concentration Test 7, Cognitive Abilities Screening
individuals’ cognition. There are several standardized Instrument – Short (CASI-S) 8, the Addenbrooke’s
instruments, of which the Mini Mental State Examination Cognitive Examination revised version (ACE-R, which
(MMSE) is the best known and most used4,5. MMSE is also includes the MMSE)9,10, the Montreal Cognitive
simple and easy to apply; the whole assessment takes Assessment (MoCA) 11,12 and the CERAD ’s words
from about five to seven minutes. It evaluates temporal list13. Among those, MoCA stands out as it evaluates
and spatial orientation, memory, attention, calculation, executive function with the trails and clock drawing;
language, and constructive skills. MMSE has many test the visual-constructive abilities with the cube
advantages. It is easy to apply, general practitioners copying and clock drawing test; the episodic memory
know it well, and it can possibly stage disease progression. with the five words delayed recall; attention with the
Healthy older adults keep similar MMSE scores over digit span, serials A’s and serial subtractions; language
time, whereas patients with Alzheimer’s disease (AD), with naming, repetition, and phonemic verbal fluency;
for example, lose an average of two to four points per year6. abstraction; and orientation. Therefore, the MoCA

4   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

assesses a broader number of cognitive domains than collaborators developed the Brief Cognitive Screening
the MMSE, especially the executive functions. Battery (BCSB)22-24. This test consists of naming and
The MMSE and the MoCA scores are strongly learning a list of 10 drawings to be freely recalled five
affected by the level of education. We suggest using the min later, after an interference activity consisting of
cutoff points in Table 2 considering the several studies semantic verbal fluency (animals/1 min) and the clock
conducted in different Brazilian populations5,12,14-21. drawing test22-24.It is an easy-to-apply instrument useful
Considering the challenge of having a test to assess for diagnosing episodic memory impairment in patients
populations with lower educational level, Nitrini and with low and high educational levels25.

Table 2. Tests suggested for cognitive screening and their respective cutoff scores.

Cognitive Test Cognitive domains Suggested cut-off scores for the Brazilian population

According to education level:


Temporal and spatial orientation
Illiterate: ≤19
Verbal episodic memory
Mini Mental State 1-4 years: ≤24
Attention and calculation
Examination (MMSE)5 5-8 years: ≤26
Language
9-11 years: ≤27
Visual constructive skills
≥ 12 years: ≤28

Figure Memory Subtest:


Incidental memory: ≤4
Immediate memory: ≤6
Learning: ≤6
Visual and verbal episodic memory Delayed recall: ≤5
Brief Cognitive Screening
Executive functions Recognition: ≤7
Battery (BCSB)24
Visual constructive skills
Semantic Verbal Fluency (animals) by education level26
Illiterate: ≤8
1-7 years: ≤11
≥ 8 years: ≤12

For Dementia (by education level):


Illiterate: ≤8
1-4 years: ≤15
Executive functions
5-8 years: ≤16
Visual constructive skills
9-11 years: ≤19
Verbal episodic memory
Montreal Cognitive ≥ 12 years: ≤21
Attention
Assessment (MoCA)12 For cognitive impairment no dementia [CIND] (by education level):
Language
Illiterate: ≤11
Abstraction
1-4 years: ≤17
Temporal and spatial orientation
5-8 years: ≤19
9-11 years: ≤19
≥ 12 years: ≤21

The authors recommend using the MMSE and semantic fluency27. Eventually, dementia specialists may
the BCSB for cognitive screening (supplementary include other cognitive tests to assess specific cognitive
materials). Clinicians may instead use MoCA in domains, such as language, praxis, visuospatial
patients with high educational level (>12 completed skills, episodic memory, attention, and executive
years of study) and MCI or mild dementia or due to functions assessment batteries. Clinicians may require
the need of better executive function assessment. neuropsychological tests when cognitive screening
Table 2 shows our suggested cut-off scores for MMSE5, tests are insufficiently sensitive to detect cognitive
MoCA12, figure delayed recall from the BCSB24-26, and the impairment, especially in individuals with MCI.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   5


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Neurological examination The first three domains are assessed with patients and
In addition to cognitive assessment, the neurological informants and the last three domains, with informants
physical exam of patients with cognitive complaints only. The CDR global score ranges between 0 (normal),
must evaluate neurological signs that may point 0.5 (MCI), 1 (mild dementia), 2 (moderate dementia),
to a possible etiology. Some findings may have and 3 (severe dementia).
particular relevance, such as brisk reflexes, signs of A brief cognitive assessment using the MMSE and
parkinsonism, gait changes, and ocular motricity. dementia staging by the CDR are needed to request
Primitive reflexes are often seen in patients with frontal the Brazilian Unified Health System (SUS) to provide
involvement (hence the term “frontalization” signs) and the symptomatic pharmacological treatment for
severe dementia. Primitive reflexes include the grasp dementia due to AD. SUS provides acetylcholinesterase
(handgrip), rooting (searching), snout (lips protrusion), inhibitors for mild to moderate AD and memantine
and palmomental ones. to moderate to severe AD 38. This consensus will
comprehensively discuss dementia treatment39.
Functional evaluation
Daily function evaluation is a core feature in assessing
patients with cognitive decline because the level SUBJECTIVE COGNITIVE DECLINE (SCD)
of functional impairment will determine patients’
syndromes. Most instruments used for functional Concept and diagnostic criteria
assessment, which are validated for Brazilian Subjective cognitive decline (SCD) is defined as self-
populations, are based on informants’ responses28. perceived cognitive decline with neither objective
In Brazil, one of the most used instruments is Pfeffer impairment on cognitive tests nor functional
Functional Activities Questionnaire (FAQ), which impairment on ADLs 40,41. The literature has used
includes 10 questions especially focused on instrumental several expressions to define self-perceived cognitive
activities (supplementary material)29. It consists of impairment, such as “subjective memory complaint,”
a simple questionnaire that is easy to understand and “subjective cognitive impairment,” and “subjective
quick to apply (average of seven minutes). FAQ scores cognitive complaint”3,42. In 2014, an international
range from 0 to 30 and scores above four points indicate working group (the Subjective Cognitive Decline
functional impairment. This test avoids the influence Initiative, SCD-I) proposed diagnostic criteria for SCD
of age or education level30,31. Another scale which also focused on standardizing terminology for research in
has a Portuguese version and proved to be useful for preclinical AD (Table 3). This group recommends to use
diagnosing dementia in a Brazilian study is the Bayer the term “cognitive,” instead of “memory,” because the
activities of daily living scale (B-ADL)32. The Informant first symptoms may not be limited only to amnestic and
Questionnaire on Cognitive Decline in the Elderly “decline” instead of a “complaint,” as it refers to the idea
(IQCODE) is a structured interview administered
of progressive deterioration and not only an isolated
to informants which combines questions related
and non-progressive complaint.
to cognitive functioning and functional performance.
It was translated and adapted for Brazilian populations
and can be helpful to screen dementia in individuals
Table 3. Approaches to Classifying SCD among a sample of Subjective
with varying educational level 33,34. The Katz scale,
Cognitive Decline Initiative (SCD-I) Working Group studies3,42.
which has been translated and adapted for Brazilian
populations, can be used to assess basic activities Criteria 1 and 2 must be present:
1. Self-experienced persistent decline in cognitive capacity in comparison
of daily living (BADLs)28,35.
with a previously normal status, unrelated to an acute event.

Cognitive impairment staging 2. Normal performance on standardized cognitive tests, which are
used to classify mild cognitive impairment (MCI) (adjusted for age,
When a person is diagnosed with cognitive decline, gender, and education level).
clinicians need to stage this condition. The Clinical
Dementia Rating (CDR) scale has been used worldwide Exclusion criteria:
1. Diagnosis of mild cognitive impairment or dementia.
for staging cognitive decline and has been validated for
Brazil36. CDR rates six domains: memory, orientation, 2. Cannot be explained by a psychiatric* or neurologic disease
judgment and problem solving, community affairs, (apart from AD), medical disorder, medication, or substance use.
home and hobbies, and personal care37. Clinicians can *Symptoms of depression or anxiety that fail to meet criteria for a psychiatric disorder are
use a semi-structured interview to assist with scoring. not considered exclusion criteria.

6   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Epidemiology exercise and control of cardiovascular risk factors.


A Mayo Clinic study showed an SCD prevalence between Patients with SCD should be screened for psychiatric
12.3% and 57% among cognitively healthy participants disorders, such as depression, sleep disorders like
aged between 70 and 95 years. Overall, 24% of the total sleep apnea, as well as for the use of anticholinergic
population reported memory concerns43. Similarly, medications and other clinical disorders associated
a Brazilian study, conducted in the municipality with cognitive impairment, such as hypothyroidism.
of Tremembé, reported an SCD prevalence of 27.6% among
all participants over 60 years old, which represented 45.2%
of individuals with normal cognitive tests44. Longitudinal MILD COGNITIVE IMPAIRMENT (MCI)
epidemiological studies have associated SCD with an
increased risk for progression to MCI and dementia3,43,45,46. Concept and diagnostic criteria
In a meta-analysis of 29 studies, the annual conversion MCI consists of an intermediate clinical condition
rate from SCD to MCI and dementia was around 6.7% between normal aging and dementia, which SCD
and 2.3%, respectively, compared with a conversion to may precede. The MCI criteria initially defined by
dementia of only 1% among older adults without SCD. Petersen emphasized the presence of complaints
Among studies which conducted follow-ups for longer and memory impairment with performance on tests
than four years, the evolution to MCI and dementia usually 1.5 standard deviations below the mean for age
reached 26.7% and 14.1%, respectively47. Furthermore, and education, during basic activities of daily living53.
older adults with SCD showed a higher prevalence of The literature has described the risk of progression
positive AD biomarkers48-51. to AD as 10 to 12% per year53. Thereafter, research
has recognized MCI as a more heterogeneous and
SCD etiologies comprehensive entity regarding its clinical presentation,
Many people with SCD remain stable or even improve. etiology, and prognosis, considering deficits in other
This is because not only neurodegenerative diseases cognitive domains besides memory 54,55. The MCI
(such as AD) are among the causes of SCD. Several definition includes the following criteria: (1) cognitive
other conditions can be associated with SCD, such as complaints reported by the patient and/or informant;
normal aging, personality traits, psychiatric disorders (2) report of cognitive decline during the past year;
(especially depression and anxiety), sleep disorders (3) changes in cognition (memory and/or other
(like obstructive sleep apnea syndrome), and the use domains) evinced at the clinical objective evaluation
of psychoactive medications with anticholinergic when compared with normal adults of the same age
or GABAergic effects42,52. and educational level; (4) no difficulty with daily
Therefore, several questions arise: How to identify activities, preserved general cognitive functioning;
which individuals within this heterogeneous group and (5) absence of dementia56,57.
will progress to MCI or dementia? What are the SCD Over the years, these clinical criteria received
characteristics which increase the risk for progression? updates since the MCI definition initially proposed in
In other words, how can we determine whether 1999 (known as the Mayo Clinic criteria53) had gaps,
an individual with SCD has an underlying preclinical AD? such as involvement of other cognitive domains,
Therefore, some authors have proposed the term the possibility that cognitive complaints could come
“SCD plus.” Patients with SCD plus have a higher from informants, and acceptance of minimally impaired
probability of showing a neurodegenerative pathology. complex instrumental functions. These updates were
Individuals at increased risk of conversion to Alzheimer- included in the 2004 Key Symposium consensus 55.
type dementia are older adults (≥ 60 years) with From then on, MCI includes four subtypes: single-domain
progressive amnestic cognitive complaints within the amnestic MCI, multiple-domain amnestic MCI,
last five years who worry about this decline and has single-domain non-amnestic MCI, and multiple-domain
family members confirming this decline3,40. non-amnestic MCI (Figure 2). The several MCI subtypes
It is important to identify people with SCD at are due to possible degenerative, vascular, metabolic,
higher risk of developing AD mainly in the context of and psychiatric etiologies, among others55.
therapeutic clinical trials. In clinical practice, patients In 2011, the National Institute on Aging and
with SCD must be followed up, especially those who Alzheimer’s Association (NIA-AA) again updated the
meet the SCD plus criteria. Although there are no consensus for the clinical diagnosis of MCI, establishing
pharmacological treatments, non-pharmacological diagnostic criteria for the symptomatic stage of AD
interventions, should be encouraged such as aerobic pre-dementia58, thus creating the AD-due MCI terminology

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   7


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

for cases with the clinical characteristics, as per Table 4. of the Diagnostic and Statistical Manual of Mental
Moreover, it included the presence of biomarkers into the Disorders, Fifth Edition (DSM-5), which recognized it as
criteria for research, specialized centers, and clinical trials, a mild neurocognitive disorder59. In 2022, the 11th edition
allowing for different degrees of AD etiology probability58. of the International Statistical Classification of Diseases
Finally, in 2013, the MCI consensus underwent yet and Related Health Problems (ICD 11) proposed the term
another terminology increments during the establishment mild neurocognitive disorder60.

Cognitive complaint

Not normal for age


No dementia
Cognitive decline
Essentially normal functional activities

MCI

Yes Memory impaired? No

Amnestic MCI Non-amnestic MCI

Yes Memory impairment only? No Yes Single impaired non-memory cognitive domain? No

Amnestic MCI Amnestic MCI Non-amnestic MCI Non-amnestic MCI


Single domain Multiple domain Single domain Multiple domain

Etiology

FTD DLB
Degenerative AD AD
AD AD

Vascular VCI VCI

Psychiatric Depression Depression

MCI: Mild Cognitive Impairment; AD: Alzheimer’s disease; VCI: vascular cognitive impairment; FTD: frontotemporal dementia; DLB: dementia with Lewy bodies.
Figure 2. Diagnostic classification of MCI after the Key Symposium criteria.

Table 4. Diagnostic criteria for Mild Cognitive Impairment due to Alzheimer’s disease by the National Institute on Aging and Alzheimer’s Association (NIA-AA)58.
Clinical and cognitive characteristics
Cognitive worries reflecting a change in cognition reported by patients, informants or clinicians (i.e., historical or observed evidence
of decline over time)

Objective evidence of impairment in one or more cognitive domains, typically including memory (i.e., formal, or bedside testing
to establish level of cognitive function in multiple domains)

Preservation of independence in functional abilities. They may show slight problems performing complex tasks, such as taking longer
to complete the task or making some mistakes
Not demented

Etiology of MCI consistent with AD pathophysiological process


Rule out vascular, traumatic, medical causes of cognitive decline, where possible
Provide evidence of longitudinal decline in cognition, when feasible
Report history consistent with AD genetic factors
MCI: Mild Cognitive Impairment; AD: Alzheimer’s disease.

8   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

For the clinical diagnosis, in addition to a detailed burden of this disease; MCI prevalence and incidence
medical history, it is important to question about the rates ranged, respectively, from 3 to 42% and from 21.5
presence of systemic diseases, use of medications and to 71.3 per 1000 person-years, and the prevalence of
have the evidence of objective cognitive assessment with cognitive impairment with no dementia (CIND) ranged
a mild decline in relation to the individuals’ previous from 5.1 to 35.9%69.
cognitive level in one or more cognitive domains (complex We find varying terminology in the few population
attention, executive function, learning and memory, studies conducted in Brazil. Overall, two studies,
language, perceptuomotor, or social cognition), besides conducted in the municipality of Porto Alegre
not meeting the diagnostic criteria for dementia58. (Rio Grande do Sul State), found an MCI prevalence
Therefore, cognitive deficits in MCI do not interfere with of 6.1%70 and a 13.2 per 1000 person-years incidence71,
the ability to be independent in ADLs, thus complex whereas the study conducted in the municipality of
instrumental activities of daily life are preserved, Tremembé, in São Paulo State, found a CIND prevalence
such as paying bills or controlling medications, of 19.5% for the population aged 60 years and older72.
but there may be need for more effort or compensatory
strategies 58. Independence in instrumental ADLs
distinguishes MCI from dementia. Nevertheless, DEMENTIA: A SYNDROMIC APPROACH
when compared to normal aging, individuals with MCI
perform more poorly59. Concept and diagnostic criteria
The criteria for dementia proposed in 2011 by the
Epidemiology NIA-AA and the Brazilian Academy of Neurology pare
Epidemiological studies of MCI, although scarce and shown in Table 51,71. In 2014, the American Psychiatric
heterogeneous regarding their adopted criteria (used Association through the DSM-5, proposed the use of
tests and educational level of the studied population), the term major neurocognitive disorder and expanded
are important since longitudinal studies have shown the diagnosis of dementia to situations where there is
that people with MCI are at increased risk of developing only one cognitive domain affected58.
dementia, up to five times as high annually than the For the most recent edition of the International
general population62-65. The estimated prevalence of MCI Statistical Classification of Diseases and Related Health
in most international population studies ranges from 10 Problems (ICD-11), published in 2019 and which
to 22% in people aged 65 years or older62,64-68. A meta- will come into force in 2022, there is still a need for
analysis found a wide range of definitions and concepts cognitive decline in at least two cognitive domains for
to estimate MCI prevalence and incidence, representing the diagnosis of dementia , not allowing the diagnosis
a challenge for researchers’ understanding of the of cognitive decline in a single domain72,73.

Table 5. Diagnostic criteria for dementia according to the National Institute on Aging and Alzheimer’s Association (NIA-AA) and the Brazilian Academy of Neurology1,73.
1. Dementia is diagnosed when there are cognitive or behavioral (neuropsychiatric) symptoms that:
1.1. Interfere with the ability to work or to carry out usual activities
1.2. Represent a decline from previous levels of functioning and performance;
1.3. Cannot be explained by delirium or major psychiatric disorder.
2. Cognitive impairment is detected and diagnosed by a combination of:
2.1. Medical history with the patient and a reliable informant; and
2.2. An objective cognitive assessment, either a “bedside” mental status examination or neuropsychological testing. Neuropsychological testing should
be performed when medical history and bedside mental status examination cannot provide a confident diagnosis.
3. Cognitive or behavioral impairment involves a minimum of two of the following domains:
3.1. Memory: impaired ability to acquire and remember new information — symptoms include repetitive questions or conversations, misplacing personal
belongings, forgetting events or appointments, getting lost on a familiar route;
3.2. Executive functions: impaired reasoning and carrying out complex tasks and judging — symptoms include poor understanding of safety risks,
inability to manage finances, poor decision-making ability, inability to plan complex or sequential activities;
3.3. Visuospatial abilities: symptoms include inability to recognize faces or common objects or to find objects in the visual field, inability to handle utensils
and dressing oneself for reasons other than visual or motor deficiency.
3.4. Language (speaking, reading, writing): symptoms include difficulty in finding common words while speaking, hesitations; speech, spelling, and
writing errors and exchange of words or phonemes, unexplicable by a sensory or motor deficit.
3.5. Behavior or personality: symptoms include uncharacteristic mood fluctuations such as agitation, impaired motivation, initiative, apathy, loss of drive,
social withdrawal, decreased interest in previous activities, loss of empathy, compulsive or obsessive behaviors, and socially unacceptable behaviors.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   9


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Epidemiology reduce the prevalence of dementia in 2050 by 8.7


The prevalence of dementia in Brazil ranges from or 16.2% in Brazil77.
5.1 to 17.5%, var ying according to the studied
reg ion a nd st udy desig n75 . T he preva lence of Etiologies of MCI and dementia
dementia in the world population aged ≥60 years The literature divides the etiologies of dementias into
is 5 to 7% in most regions, with the highest prevalence primary (or neurodegenerative) and secondary ones
in Latin America, at 8.5%76 . In a Brazilian study, (Figure 3). The altered accumulation of misfolded proteins
(misfolded proteins diseases) in the central nervous
the prevalence of dementia is even higher, reaching
system (CNS)78 pathologically characterizes degenerative
23% in illiterate individuals aged 60 years or older
dementias. AD, Lewy body (LBD), and frontotemporal
living at the municipality of Tremembé72. dementias (FTD) are the most frequent ty pes
In Brazil, a study suggests that 32.3% of dementia of primary dementias. Other various neurological
cases are due to seven modifiable risk factors: diabetes diseases, usually associated with movement disorders
mellitus, systemic arterial hypertension, middle-age (such as parkinsonism or chorea), can develop dementia
obesity, physical inactivity, depression, smoking, throughout its evolution, such as Parkinson’s disease,
and poor education77. Reducing the prevalence of each progressive supranuclear palsy, corticobasal syndrome,
risk factor by 10% or 20% per decade could potentially and Huntington’s disease.

Cognitive complaint brought by the patient and/or companion

Demonstration of cognitive impairment in assessment

Yes No
Subjective Cognitve Decline
Functional decline
No Yes
Mild cognitive impairment Dementia

Degenerative dementias Non-degenerative dementias

Amnestic predominance Alzheimer’s Disease (AD)

Disexecutive predominance Frontal variant of AD


Dementia with Lewy bodies With evidence of Without evidence
structural lesion of structural lesion
Behavioral Predominance Frontotemporal dementia

Aphasic predominance Primary progressive Aphasias


Encefalites Vascular dementia HAND
Posterior Cortical Atrophy (AD) Normal Pressure Hydrocephalus Neurosyphilis
Visuospatial predominance
Dementia with Lewy bodies Chronic Traumatic Encephalopathy Thyroid diseases
CNS Neoplasms Uremic
Parkinson’s Disease Dementia Autoimmune encephalitis Hypermonemia
Dementia with Lewy bodies Infectious Encephalitis Drugs
With Parkinsonism
Progressive Supranuclear Palsy B12 deficiency
Corticobasal Syndrome Thiamine Deficiency

Rapidly Progressive Creutzfeldt-Jakob Disease

Figure 3. Main etiological differential diagnoses of dementia.

10   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Epidemiological and clinical studies found that Historically, neurodegenerative dementias were
AD dementia is the most prevalent cause of dementia described based on their clinical phenotypes (e.g.,
in Brazil, followed by vascular dementia (VD) due to AD is described as amnestic dementia in most cases).
cerebrovascular disease 75. A study conducted with However, in the last two decades, studies searching
clinicopathological analysis also found the same an early diagnosis based on biomarkers , that allow
ordering of causes related to dementia 79. Another
the identification of pathological proteins in vivo
study from a Brazilian brain bank with 480 participants
found that 50% of its sample met the neuropathological have been increasing 77 . Interestingly, the same
criteria for AD, 35%, VD; 18%, LBD; 17% received other pathological protein can lead to different phenotypes,
diagnoses (e.g., frontotemporal lobar degeneration); while the same phenotype can be associated with
and 20% showed mixed pathology (most were AD different proteins80. Figure 4 illustrates this broad
associated with VD)80. spectrum of proteinopathies and clinical phenotypes.

FTD: Frontotemporal dementia; ALS: Amyotrophic lateral sclerosis; MND: Motor neuron disease; svPPA: Semantic variant of primary progressive aphasia; nfvPPA: Nonfluent (or agrammatic)
variant of primary progressive aphasia; CBS: Corticobasal syndrome; PSP: Progressive supranuclear palsy; AD: Alzheimer’s disease; lvPPA: Logopenic variant of primary progressive aphasia;
DLB: Dementia with Lewy bodies; PD: Parkinson’s disease; MSA: Multiple systems atrophy.
Figure 4. Spectrum of proteinopathies causing neurodegenerative dementias and their respective clinical phenotypes.
Pathological proteins: TDP-43, FUS, Tau, beta-amyloid and alpha-synuclein.

Secondary causes are potentially treatable and which must be carried out early to avoid cognitive
should be investigated for early treatment. The most sequelae in cases of potentially reversible etiology.
frequent cause of secondary dementia is VD, which Depression is one of the main differential diagnoses
is the second leading cause of dementia in Brazil. of dementia. Major depressive disorder courses
The investigation of risk factors and mechanisms with cognitive decline, executive dysfunction,
of cerebral vascular involvement is essential for and attention deficit83. The relation between dementia
adequate treatment of VD patients78. Vitamin B12 and depression is complex because depressive
symptoms can be the initial manifestation of dementia.
deficiency, neurosyphilis, and normal pressure
Therefore, depression is a risk factor for dementia
hydrocephalus are causes of secondary dementia that
and severe depression can cause potentially reversible
should be initially ruled out. dementia84. Depression is one of the 12 modifiable
Rarer causes, shown as rapidly progressive conditions risk factors responsible for 40% of dementia cases
(evolving to dementia within one to two years of the worldwide85. Accordingly, every patient with depressive
onset of the first symptoms, usually within weeks symptoms should receive adequate treatment for
to months) are prion, autoimmune, and infectious depression. However, if cognitive deficits remain after
dementias, among others 82 . Rapidly progressive optimized treatment, clinical practice should suspect
dementia (RPD) requires extensive investigation, of a degenerative etiology.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   11


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Cognitive decline associated with HIV infection, Etiological investigation of MCI and dementia
called HAND (HIV-associated neurocognitive disorder), The current recommendation for investigating MCI
shows an increasing prevalence after the use of highly and dementia in the Brazilian population involves
effective antiretroviral treatment for people living with a neuroimaging study (computed tomography
HIV86. HAND prevalence in Brazil ranges from 52.4% to scan - CT - or, ideally, brain magnetic resonance
73.6%, with a higher percentage of asymptomatic cases, imaging - MRI) and laboratory tests which investigate
named asymptomatic neurocognitive impairment87,88. non-neurodegenerative etiologies. These tests include
Research should investigate the use of psychoactive complete blood count, serum concentrations of
medications, especially benzodiazepines, antipsychotics, creatinine, TSH, albumin, liver enzymes, vitamin B12,
non-benzodiazepine hypnotics, and anticholinergic ionized calcium, serological reactions for syphilis;
medications, as possible etiologies of dementia. Table 6 and HIV serology for those with atypical clinical
shows some anticholinergic medications associated presentations or suggestive symptoms 90. Clinical
with cognitive decline89. practice should also request cerebrospinal fluid (CSF)
examination for patients with early-onset dementia
(before 65 years of age), atypical presentations,
Table 6. List of drugs with centrally acting strong anticholinergic properties*. and suspected inflammatory, prion, or infectious
Antiarrhythmic Antimuscarinics (urinary incontinence) diseases 90 . Clinicians should always assess RPD
Disopyramide Darifenacin with brain MRIs (with diffusion-weighted imaging),
Fesoterodine electroencephalogram, CSF, and broader laboratory
Antidepressants
Flavoxate
Amitriptyline investigation, depending on the clinical hypotheses.
Oxybutynin
Amoxapine
Solifenacin
This MRI recommendation is especially due to the
Clomipramine possibility of finding atrophy in the hippocampal region,
Tolterodine
Desipramine which is impossible by CT scan in the early stages of the
Trospium
Doxepin (>6mg)
disease. The finding of atrophy in the mesial temporal
Imipramine
Nortriptyline regions is suggestive of the diagnosis of MCI due to AD,
Paroxetine however, it is not exclusive to this pathology, since
Protriptyline a considerable proportion of cases of amnestic MCI with
Trimipramine hippocampal atrophy may have different causes other
Antiemetics Antiparkinsonian agents than AD, denominated SNAP (suspected non-Alzheimer
Prochlorperazine Biperiden pathophysiology)91. Another advantage of brain MRI
Promethazine Trihexyphenidyl is its better identification of cerebrovascular disease,
Benztropine especially of small vessel disease.
Antihistamines (first generation)
Brompheniramine Antipsychotics The clinical indications for biomarkers include
Carbinoxamine Chlorpromazine 1) identification of individuals with MCI and mild
Chlorpheniramine Clozapine dementia due to AD pathology and 2) diagnostic
Clemastine Loxapine uncertainty about the dementia etiology. Biomarkers
Cyproheptadine Olanzapine
Dexbrompheniramine available for clinical use can be divided into specific AD
Perphenazine
Dexchlorpheniramine Thioridazine pathology or neurodegeneration biomarkers. Specific
Dimenhydrinate Trifluoperazine AD pathology biomarkers include 1) measurement
Diphenhydramine (oral) of the beta-amyloid peptide and phosphorylated tau
Doxylamine Antispasmodics
Atropine (excludes ophthalmic)
(phospho-tau) in the CSF and 2) positron emission
Hydroxyzine
Meclizine Belladonna alkaloids computed tomography (PET) with amyloid peptide
Clidinium-chlordiazepoxide Scopolamine (excludes ophthalmic) marker and PET with marker for tau protein.
Dicyclomine Until now, PET-amyloid is offered in few centers
Skeletal muscle relaxants
Homatropine (excludes ophthalmic) in Brazil and PET-tau is still unavailable in Brazil.
Cyclobenzaprine
Hyoscyamine Neurodegeneration biomarkers consist of PET with
Orphenadrine
Methscopolamine
18F-fluorodeoxyglucose (PET-FDG), measurement
Propantheline
Promethazine of total tau protein in the CSF, and light chain
Pyrilamine neurofilament. More recently, plasma biomarkers
Triprolidine have been developed but they still lack validation for
* Adapted from Beers Criteria89. clinical use (tau-181, tau-217)92-94.

12   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Confirmation of the AD in vivo requires amyloid Primar y care physicians must receive back
and tau pathology. CSF measurement or PET-amyloid patients referred to secondary or tertiary care
uptake can confirm the amyloid pathology, whereas for long-term follow-up of AD and VD dementia.
measuring CSF phospho-tau or PET-tau uptake Primar y care physicians should request a new
can conf ir m the Tau patholo g y. PE T-F DG is specialist evaluation in case of any doubt regarding
a neurodegeneration biomarker whose metabolic the follow-up of the case.
pattern may suggest the pathology of degenerative
dementias with good sensitivity and specificity Secondary healthcare
(e.g., the pattern of hypometabolism in the areas Physicians in secondary care emergencies must
of temporoparietal association, posterior cingulate, be able to identify cases of RPD, because patients
and precuneus has a sensitivity and specificity >90% often seek emergenc y units due to the rapid
for AD diagnosis)95. Furthermore, PET FDG is used progression of symptoms. W hen recognizing
in the diagnostic criteria of LBD, primary progressive a case of RPD, physicians must urgently conduct
aphasias, and FTD. an initial investigation and, preferably, refer patients
to tertiary care for extensive work-up.
Specialists (neurologists, geriatricians, and
PROPOSAL FOR THE MANAGEMENT OF PATIENTS psychiatrists) who receive patients referred for
WITH COGNITIVE DECLINE AT DIFFERENT dementia, whose secondary etiologies were excluded
LEVELS OF CARE IN THE BRAZILIAN PUBLIC at the primary level, should confirm the diagnosis
HEALTH SERVICE - SUS and establish treatment. For cases in which there is
no greater complexity in managing neuropsychiatric
Primary healthcare
Identification of modifiable risk factors for dementia symptoms, recommendations on how to treat
prevention should be done at primary health care. dementia should be explained to the primary care
Recently, a study described 12 important modifiable physician who will follow up the patient.
risk factors to prevent or delay the onset of 40% of Secondary care should request biomarker tests
dementia in the world and 56% in low- and middle- when necessary. Physicians should request biomarker
income countries84. They are diagnosed at different tests in atypical cases (initial non-amnestic clinical
stages of life, with the possibility of involvement by presentation or early-onset dementia) and interpreting
the health system in all of them. Risk factors include: them requires trained secondary care professionals.
1) age up to 45 years: low educational level; 2) age from If it is impossible to assess biomarkers or the
45 to 65 years old: systemic arterial hypertension, obesity, professional is unfamiliar with result interpretation,
hearing loss, traumatic brain injury, and alcohol abuse; patients must be referred to tertiary centers.
and 3) age above 65 years old: smoking, depression, In cases of early-onset dementia in which the
sedentary lifestyle, diabetes, social isolation, and air specialist demands further etiological investigation,
pollution. Clinical practice must consider preventive
patients should be referred to tertiary services
and educational actions to identify, promote treatment,
for diagnosis elucidation.
and combat these different factors84.
Primary care physicians should actively seek out
Tertiary healthcare
complaints of cognitive decline in patients, especially
Cases which maybe referred to tertiary care include
after 60 years of age. Clinicians should perform
screening tests for cognitive and functional decline RPD, early-onset dementia, suspected genetic form
in patients with cognitive complaints (or reported of cognitive decline, cases of unclear diagnosis,
by caregivers). In case of a clinical diagnosis of MCI cases of difficult management of neuropsychiatric
or dementia, physicians should request etiological symptoms, and cases for biomarker investigation
work-up. When ruling out potentially reversible causes, which secondary care is unable to assess.
patients should be referred to secondary healthcare, In cases of clinical stability or advanced stage
to neurology, geriatrics, or psychiatry specialists, of dementia, patients can be counter-referred
with a hypothesis of degenerative or vascular etiology. to primary or secondary care. Figure 5 summarizes
The RPD cases must be promptly referred to secondary the competences of each level of healthcare in caring
or tertiary care, as a matter of urgency. for patients with cognitive decline.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   13


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

Primary Healthcare

Perform complementary
Perform clinical
Identify modifiable Ask about cognitive Apply cognitive exams (neuroimaging To receive patients
diagnosis of cognitive
risk factors for complaints, especially and functional and laboratory tests) with AD or VD against
syndrome (SCD, MCI
dementia prevention. in older adults. screening tests. to exclude potentially referenced for follow-up.
or dementia).
reversible causes.

Referral to Secondary Healthcare in the following situations:


1. Degenerative or vascular dementia.
2. If the hypothesis of MCI due to AD (amnestic MCI).
3. RPD (urgently).
4. When there are a diagnostic doubts about the cause of the dementia.

Secondary Healthcare

Evaluate patients
Against referring less Carry out an initial Receive patients
with a hypothesis Initiate treatment Request biomarkers
complex AD and DV investigation in with AD or DV
of degenerative for degenerative in atypical cases.
cases to physician of patients with against referenced
dementia for or vascular dementia.
Primary Healthcare. RPD urgently. for follow-up.
diagnostic confirmation.

Referral to Tertiary Healthcare in the following situations


1. Biomarkers are not accessible and are needed.
2. Early-onset dementia or atypical dementias requiring further etiological investigation.
3. RPD urgently after initial investigation.

Tertiary Healthcare

Evaluation of patients with:


1. Diagnostic doubt about the etiology of the dementia
2. RPD
3. Early onset dementias
4. Suspected genetic form of dementia

When there is clinical stability or in an advanced stage of dementia, patients can be counter-referred to primary or secondary healthcare

AD: Alzheimer’s Disease, VD: Vascular Dementia, MCI: Mild cognitive impairment; SCD: Subjective cognitive decline; RPD: Rapidly progressive dementia.
Figure 5. Hierarchy of care for patients with cognitive syndromes according to the levels of care in the Brazilian Unified Health System (SUS).

ACKNOWLEDGEMENTS
PC, LCS and RN are funded by CNPq, Brazil (bolsa de RK, EPFR, FACV: drafting of the manuscript; BJAPB,
produtividade em pesquisa). FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC, PHFB, RN,
Authors’ contributions. JS, ASN, KGCF, MCND, RK, EPFR, SMDB: critical revision of the manuscript for important
FACV, BJAPB, LPS: concept; JS, ASN, KGCF, MCND, intellectual content.

REFERENCES
1. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, mer’s disease. Alzheimers Dement. 2011;7(3):263-9. doi:10.1016/j.
Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s jalz.2011.03.005.
disease: recommendations from the National Institute on Aging-Al- 2. Petersen RC. Clinical practice. Mild cognitive impairment. N Engl J Med.
zheimer’s Association workgroups on diagnostic guidelines for Alzhei- 2011;364(23):2227-34. doi:10.1056/NEJMcp0910237.

14   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

3. Jessen F, Amariglio RE, Van Boxtel M, Breteler M, Ceccaldi M, Chételat G, of education. Dement Neuropsychol. 2007;1:32-6. doi:10.1590/
et al. A conceptual framework for research on subjective cognitive decline S1980-57642008DN10100006.
in preclinical Alzheimer’s disease. Alzheimers Dement. 2014;10(6):844-52. 24. Nitrini R, Bucki SMD, Yassuda MS, Fichman HC, Caramelli P. The Figure
doi:10.1016/j.jalz.2014.01.001. Memory Test: diagnosis of memory impairment in populations with
4. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical heterogeneous educational background. Dement Neuropsychol.
method for grading the cognitive state of patients for the clinician. 2021;15(2):173-85. doi:10.1590/1980-57642021dn15-020004.
J Psychiatr Res. 1975;12(3):189-98. doi:10.1016/0022-3956(75)90026-6. 25. Nitrini R, Caramelli P, Herrera Júnior E, Porto CS, Charchat-Fichman H,
5. Brucki SMD, Nitrini R, Caramelli P, Bertolucci PHF, Okamoto IH. Sugestões Carthery MT, et al. Performance of illiterate and literate nondemented
para o uso do mini-exame do estado mental no Brasil. Arq Neuropsiquiatr. elderly subjects in two tests of long-term memory. J Int Neuropsychol
2003;61(3B):777-81. doi:10.1590/S0004-282X2003000500014. Soc. 2004;10(4):634-8. doi:10.1017/S1355617704104062.
6. Barocco F, Spallazzi M, Concari L, Gardini S, Pelosi A, Caffarra P. 26. Yassuda MS, Silva HS, Lima-Silva TB, Cachioni M, Falcão DVS, Lopes A, et al.
The Progression of Alzheimer’s Disease: Are Fast Decliners Really Normative data for the Brief Cognitive Screening Battery stratified by age
Fast? A Four-Year Follow-Up. J Alzheimers Dis. 2017;57(3):775-86. and education. Dement Neuropsychol. 2017;11(1):48-53. doi:10.1590/
doi:10.3233/JAD-161264. 1980-57642016dn11-010008.
7. Viana GS, Rouquayrol MZ, Bruin VM, Albuquerque JJ. Use of the 27. Caramelli P, Carthery-Goulart MT, Porto CS, Charchat-Fichman H,
Information, Memory and Concentration (IMC) Test in the epidemiological Nitrini R. Category fluency as a screening test for Alzheimer disease in
study of senile dementia in Fortaleza,Ceará (Brazil). Cad Saude Publica. illiterate and literate patients. Alzheimer Dis Assoc Disord. 2007;21(1):65-7.
1991;7(3):396-408. doi:10.1590/s0102-311x1991000300008. doi:10.1097/WAD.0b013e31802f244f.
8. Damasceno A, Delicio AM, Mazo DFC, Zullo JFD, Scherer P, Ng RTY, et al. 28. Chaves MLF, Godinho CC, Porto CS, Mansur L, Carthery-Goulart MT,
Validation of the Brazilian version of mini-test CASI-S. Arq Neuropsiquiatr. Yassuda MS, et al. Cognitive, functional and behavioral assessment:
2005;63(2B):416-21. doi:10.1590/s0004-282x2005000300010. Alzheimer’s disease. Dement Neuropsychol. 2011;5(3):153-66.
9. Carvalho VA, Caramelli P. Brazilian adaptation of the Addenbrooke’s doi:10.1590/S1980-57642011DN05030003.
Cognitive Examination-Revised (ACE-R). Dement Neuropsychol. 29. Pfeffer RI, Kurosaki TT, Harrah CH Jr, Chance JM, Filos S. Measurement
2007;1(2):212-6. doi:10.1590/S1980-57642008DN10200015 of functional activities in older adults in the community. J Gerontol.
10. César KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. Addenbrooke’s 1982;37(3):323-9. doi:10.1093/geronj/37.3.323.
cognitive examination-revised: Normative and accuracy data for seniors 30. Sanchez MADS, Correa PCR, Lourenço RA. Cross-cultural
with heterogeneous educational level in Brazil. Int Psychogeriatrics. Adaptation of the “Functional Activities Questionnaire – FAQ” for
2017;29(8):1345-53. doi:10.1017/S1041610217000734. use in Brazil. Dement Neuropsychol. 2011;5(4):322-7. doi:10.1590/
11. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, S1980-57642011DN05040010.
Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief screening 31. Assis LO, Paula JJ, Assis MG, Moraes EN, Malloy-Diniz LF.
tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53(4):695-9. Psychometric properties of the Brazilian version of Pfeffer’s Functional
doi:10.1111/j.1532-5415.2005.53221.x. Activities Questionnaire. Front Aging Neurosci. 2014;6:255. doi:10.3389/
12. Cesar KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. MoCA Test: fnagi.2014.00255.
normative and diagnostic accuracy data for seniors with heterogeneous 32. Bustamante SEZ, Bottino CMC, Lopes MA, Azevedo D, Hototian SR,
educational levels in Brazil. Arq Neuropsiquiatr. 2019;77(11):775-81. Litvoc J, et al. Combined instruments on the evaluation of dementia in
doi:10.1590/0004-282X20190130. the elderly: preliminary results. Arq Neuropsiquiatr. 2003;61(3A):601-6.
13. Bertolucci PH, Okamoto IH, Brucki SM, Siviero MO, Toniolo Neto J, doi:10.1590/s0004-282x2003000400014.
Ramos LR. Applicability of the CERAD neuropsychological battery to 33. Sanchez MAS, Lourenço RA. Informant Questionnaire on Cognitive
Brazilian elderly. Arq Neuropsiquiatr. 2001;59(3-A):532-6. doi:10.1590/ Decline in the Elderly (IQCODE): cross-cultural adaptation for use
s0004-282x2001000400009. in Brazil. Cad Saude Publica. 2009;25(7):1455-65. doi:10.1590/
14. Bertolucci PH, Brucki SM, Campacci SR, Juliano Y. The Mini-Mental State S0102-311X2009000700003.
Examination in a general population: impact of educational status. Arq 34. Perroco TR, Bustamante SEZ, Moreno MPQ, Hototian SR, Lopes MA,
Neuropsiquiatr. 1994;52(1):1-7. Azevedo D, et al. Performance of Brazilian long and short IQCODE
15. Almeida OP. Mini mental state examination and the diagnosis of dementia on the screening of dementia in elderly people with low education.
in Brazil. Arq Neuropsiquiatr. 1998;56(3B):605-12. doi:10.1590/ Int Psychogeriatr. 2009;21(3):531-8. doi:10.1017/S1041610209008849.
S0004-282X1998000400014. 35. Lino VTS, Pereira SRM, Camacho LAB, Ribeiro Filho ST, Buksman S.
16. Laks J, Batista EMR, Guilherme ERL, Contino ALB, Faria MEV, Cross-cultural adaptation of the Independence in Activities of Daily Living
Figueira I, et al. Mini-mental state examination in community- Index (Katz Index). Cad Saude Publica. 2008;24(1):103-12. doi:10.1590/
dwelling elderly: preliminary data from Santo Antônio de Pádua, s0102-311x2008000100010.
Rio de Janeiro, Brazil. Arq Neuropsiquiatr. 2003;61(3B):782-5. doi:10.1590/ 36. Chaves MLF, Camozzato AL, Godinho C, Kochhann R, Schuh A,
S0004-282X2003000500015. Almeida VL, et al. Validity of the clinical dementia rating scale for the
17. Castro-Costa E, Fuzikawa C, Uchoa E, Firmo JOA, Lima-Costa MF. detection and staging of dementia in Brazilian patients. Alzheimer Dis
Norms for the mini-mental state examination: adjustment of the cut-off Assoc Disord. 2007;21(3):210-7. doi:10.1097/WAD.0b013e31811ff2b4.
point in population-based studies (evidences from the Bambuí health 37. Morris JC. The Clinical Dementia Rating (CDR): current version and scoring
aging study). Arq Neuropsiquiatr. 2008;66(3A):524-8. doi:10.1590/ rules. Neurology. 1993;43(11):2412-4. doi:10.1212/wnl.43.11.2412-a.
s0004-282x2008000400016. 38. Ministério da Saúde (BR). Portaria nº 1.298, de 21 de novembro de
18. Kochhann R, Varela JS, Lisboa CSM, Chaves MLF. The Mini Mental State 2013. Aprova o Protocolo Clínico e Diretrizes Terapêuticas da Doença
Examination: Review of cutoff points adjusted for schooling in a large de Alzheimer. Diário Oficial da União. 2013 Out 24.
Southern Brazilian sample. Dement Neuropsychol. 2010;4(1):35-41. 39. Caramelli P, Marinho V, Laks J, Coletta MVD, Stella F, Camargos EF et al.
doi:10.1590/S1980-57642010DN40100006. Treatment of Dementia. Arq Neuropsiquiatr. 2022; submitted.
19. Memória CM, Yassuda MS, Nakano EY, Forlenza OV. Brief screening 40. Jessen F, Amariglio RE, Buckley RF, Flier WM, Han Y, Molinuevo JL,
for mild cognitive impairment: validation of the Brazilian version of the et al. The characterisation of subjective cognitive decline. Lancet Neurol.
Montreal cognitive assessment. Int J Geriatr Psychiatry. 2013;28(1):34-40. 2020;19(3):271-8. doi:10.1016/S1474-4422(19)30368-0.
doi:10.1002/gps.3787. 41. Rabin LA, Smart CM, Amariglio RE. Subjective Cognitive Decline in
20. Apolinario D, Santos MF, Sassaki E, Pegoraro F, Pedrini AVA, Cestari B, Preclinical Alzheimer’s Disease. Annu Rev Clin Psychol. 2017;13:369-96.
et al. Normative data for the Montreal Cognitive Assessment (MoCA) and doi:10.1146/annurev-clinpsy-032816-045136.
the Memory Index Score (MoCA-MIS) in Brazil: Adjusting the nonlinear 42. Molinuevo JL, Rabin LA, Amariglio R, Buckley R, Dubois B, Ellis KA, et al.
effects of education with fractional polynomials. Int J Geriatr Psychiatry. Implementation of subjective cognitive decline criteria in research studies.
2018;33(7):893-9. doi:10.1002/gps.4866. Alzheimers Dement. 2017;13(3):296-311. doi:10.1016/j.jalz.2016.09.012.
21. Lourenço RA, Veras RP. Mini-Mental State Examination: psychometric 43. Harten AC, Mielke MM, Swenson-Dravis DM, Hagen CE, Edwards KK,
characteristics in elderly outpatients. Rev Saude Publica. 2006;40(4):712-9. Roberts RO, et al. Subjective cognitive decline and risk of MCI: The Mayo
doi:10.1590/S0034-89102006000500023. Clinic Study of Aging. Neurology. 2018;91(4):e300-12. doi:10.1212/
22. Nitrini R, Lefèvre BH, Mathias SC, Caramelli P, Carrilho PE, Sauaia N, WNL.0000000000005863.
et al. Neuropsychological tests of simple application for diagnosing 44. César-Freitas KG, Suemoto CK, Power MC, Brucki SMD, Nitrini R. Incidence
dementia. Arq Neuropsiquiatr. 1994;52(4):457-65. doi:10.1590/ of dementia in a Brazilian population: The Tremembé Epidemiologic
s0004-282x1994000400001. Study. Alzheimers Dement. 2022;18(4):581-90. doi:10.1002/alz.12423.
23. Nitrini R, Caramelli P, Porto CS, Charchat-Fichman H, Formigoni AP, 45. Reisberg B, Shulman MB, Torossian C, Leng L, Zhu W. Outcome over seven
Carthery-Goulart MT, et al. Brief cognitive battery in the diagnosis years of healthy adults with and without subjective cognitive impairment.
of mild Alzheimer ’ s disease in subjects with medium and high levels Alzheimers Dement. 2010;6(1):11-24. doi:10.1016/j.jalz.2009.10.002.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   15


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

46. Rönnlund M, Sundström A, Adolfsson R, Nilsson L-G. Subjective memory 68. Petersen RC, Roberts RO, Knopman DS, Geda YE, Cha RH, Pankratz
impairment in older adults predicts future dementia independent of baseline VS, et al. Prevalence of mild cognitive impairment is higher in men. The
memory performance: Evidence from the Betula prospective cohort study. Mayo Clinic Study of Aging. Neurology. 2010;75(10):889-97. doi:10.1212/
Alzheimers Dement. 2015;11(11):1385-92. doi:10.1016/j.jalz.2014.11.006. WNL.0b013e3181f11d85.
47. Mitchell AJ, Beaumont H, Ferguson D, Yadegarfar M, Stubbs B. 69. Ward A, Arrighi HM, Michels S, Cedarbaum JM. Mild cognitive impairment:
Risk of dementia and mild cognitive impairment in older people with disparity of incidence and prevalence estimates. Alzheimers Dement.
subjective memory complaints: meta-analysis. Acta Psychiatr Scand. 2012;8(1):14-21. doi:10.1016/j.jalz.2011.01.002.
2014;130(6):439-51. doi:10.1111/acps.12336. 70. Godinho C, Camozzato AL, Onyszko D, Chaves ML. Estimation of the
48. Snitz BE, Lopez OL, McDade E, Becker JT, Cohen AD, Price JC, et al. risk of conversion of mild cognitive impairment of Alzheimer type to
Amyloid-β Imaging in Older Adults Presenting to a Memory Clinic Alzheimer’s disease in a south Brazilian population-based elderly cohort:
with Subjective Cognitive Decline: A Pilot Study. J Alzheimers Dis. the PALA study. Int Psychogeriatr. 2012;24(4):674-81. doi:10.1017/
2015;48(Suppl 1):S151-9. doi:10.3233/JAD-150113. S1041610211002043.
49. Perrotin A, Mormino EC, Madison CM, Hayenga AO, Jagust WJ. Subjective 71. Chaves ML, Camozzato AL, Godinho C, Piazenski I, Kaye J.
cognition and amyloid deposition imaging: a Pittsburgh Compound Incidence of mild cognitive impairment and Alzheimer disease in
B positron emission tomography study in normal elderly individuals. Southern Brazil. J Geriatr Psychiatry Neurol. 2009;22(3):181-7.
Arch Neurol. 2012;69(2):223-9. doi:10.1001/archneurol.2011.666. doi:10.1177/0891988709332942.
50. Amariglio RE, Mormino EC, Pietras AC, Marshall GA, Vannini P, Johnson KA, 72. César KG, Brucki SMD, Takada LT, Nascimento LFC, Gomes CMS,
et al. Subjective cognitive concerns, amyloid-β, and neurodegeneration Almeida MCS, et al. Prevalence of Cognitive Impairment Without
in clinically normal elderly. Neurology. 2015;85(1):56-62. doi:10.1212/ Dementia and Dementia in Tremembé, Brazil. Alzheimer Dis Assoc Disord.
WNL.0000000000001712. 2016;30(3):264-71. doi:10.1097/WAD.0000000000000122.
51. Buckley RF, Maruff P, Ames D, Bourgeat P, Martins RN, Masters CL, et al. 73. Frota NAF, Nitrini R, Damasceno BP, Forlenza O, Dias-Tosta E,
Subjective memory decline predicts greater rates of clinical progression in Silva AB, et al. Critérios para o diagnóstico de doença de Alzheimer.
preclinical Alzheimer’s disease. Alzheimers Dement. 2016;12(7):796-804. Dement Neuropsychol. 2011;5(Suppl 1):5-10.
doi:10.1016/j.jalz.2015.12.013. 74. World Health Organization. International Classification of Diseases
52. Studart A, Nitrini R. Subjective cognitive decline: The first clinical manifestation 10th Revision (ICD-10) . Version 2019. Geneva: WHO; 2019 [Accessed
of Alzheimer’s disease? Dement Neuropsychol. 2016;10(3):170-7. 20 dez 2021]. Available at: https://icd.who.int/browse10/2019/en#/VI
doi:10.1590/S1980-5764-2016DN1003002. 75. Boff MS, Sekyia FS, Bottino CMC. Prevalence of dementia among
53. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. brazilian population: systematic review. Rev Med. 2015;94(3):154-61.
Mild cognitive impairment: clinical characterization and outcome. doi:10.11606/issn.1679-9836.v.94i3p154-161.
Arch Neurol. 1999;56(3):303-8. doi:10.1001/archneur.56.3.303. 76. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global
54. Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern prevalence of dementia: a systematic review and metaanalysis. Alzheimers
Med. 2004;256(3):183-94. doi:10.1111/j.1365-2796.2004.01388.x.
Dement. 2013;9(1):63-75.e2. doi:10.1016/j.jalz.2012.11.007.
55. Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund LO, et al.
77. Oliveira D, Jun Otuyama L, Mabunda D, Mandlate F, Gonçalves-Pereira M,
Mild cognitive impairment--beyond controversies, towards a consensus:
Xavier M, et al. Reducing the Number of People with Dementia Through
report of the International Working Group on Mild Cognitive Impairment. J
Primary Prevention in Mozambique, Brazil, and Portugal: An Analysis
Intern Med. 2004;256(3):240-6. doi:10.1111/j.1365-2796.2004.01380.x.
of Population-Based Data. J Alzheimers Dis. 2019;70(s1):S283-91.
56. Morris JC. Revised criteria for mild cognitive impairment may
doi:10.3233/JAD-180636.
compromise the diagnosis of Alzheimer disease dementia. Arch Neurol.
78. Allegri RF. Moving from neurodegenerative dementias, to cognitive
2012;69(6):700-8. doi:10.1001/archneurol.2011.3152.
proteinopathies, replacing “where” by “what”… Dement Neuropsychol.
57. Portet F, Ousset PJ, Visser PJ, Frisoni GB, Nobili F, Scheltens P, et al.
2020;14(3):237-42. doi:10.1590/1980-57642020dn14-030005.
Mild cognitive impairment (MCI) in medical practice: a critical review of the
79. Grinberg LT, Nitrini R, Suemoto CK, Ferretti-Rebustini REL, Leite REP,
concept and new diagnostic procedure. Report of the MCI Working Group
Farfel JM, et al. Prevalence of dementia subtypes in a developing country:
of the European Consortium on Alzheimer’s Disease. J Neurol Neurosurg
a clinicopathological study. Clinics. 2013;68(8):1140-5. doi:10.6061/
Psychiatry. 2006;77(6):714-8. doi:10.1136/jnnp.2005.085332.
clinics/2013(08)13.
58. Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, et al.
The diagnosis of mild cognitive impairment due to Alzheimer’s disease: 80. Suemoto CK, Ferretti-Rebustini REL, Rodriguez RD, Leite REP,
recommendations from the National Institute on Aging-Alzheimer’s Soterio L, Brucki SMD, et al. Neuropathological diagnoses and clinical
Association workgroups on diagnostic guidelines for Alzheimer’s disease. correlates in older adults in Brazil: A cross-sectional study. PLoS Med.
Alzheimers Dement. 2011;7(3):270-9. doi:10.1016/j.jalz.2011.03.008. 2017;14(3):e1002267. doi:10.1371/journal.pmed.1002267.
59. American Psychiatric Association. Diagnostic and statistical manual of 81. Elahi FM, Miller BL. A clinicopathological approach to the diagnosis of
mental disorders. 5th ed. Washington (DC): APA, 2013. dementia. Nat Rev Neurol. 2017;13(8):457-76. doi:10.1038/nrneurol.2017.96.
60. World Health Organization. International Classification of Diseases 82. Geschwind MD. Rapidly Progressive Dementia. Continuum (Minneap Minn).
11th Revision (ICD-11). Version: 2019 April. Geneva: WHO; 2019 [Accessed 2016;22(2 Dementia):510-37. doi:10.1212/CON.0000000000000319.
20 dez 2021]. Available at: https://icd.who.int/browse11/l-m/en 83. Dias NS, Barbosa IG, Kuang W, Teixeira AL. Depressive disorders in the
61. Gold DA. An examination of instrumental activities of daily living elderly and dementia: An update. Dement Neuropsychol. 2020;14(1):1-6.
assessment in older adults and mild cognitive impairment. J Clin Exp doi:10.1590/1980-57642020dn14-010001.
Neuropsychol. 2012;34(1):11-34. doi:10.1080/13803395.2011.614598. 84. B e n n e t t S , T h o m a s A J . D e p re s s i o n a n d d e m e n t i a : c a u s e ,
62. Busse A, Hensel A, Gühne U, Angermeyer MC, Riedel-Heller SG. Mild consequence or coincidence? Maturitas. 2014;79(2):184-90.
cognitive impairment: Long-term course of four clinical subtypes. Neurology. doi:10.1016/j.maturitas.2014.05.009.
2006;67(12):2176-85. doi:10.1212/01.wnl.0000249117.23318.e1. 85. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S,
63. Farias ST, Mungas D, Reed BR, Harvey D, DeCarli C. Progression of mild et al. Dementia prevention, intervention, and care: 2020 report of the
cognitive impairment to dementia in clinic- vs community-based cohorts. Lancet Commission. Lancet. 2020;396(10248):413-46. doi:10.1016/
Arch Neurol. 2009;66(9):1151-7. doi:10.1001/archneurol.2009.106. S0140-6736(20)30367-6.
64. Manly JJ, Tang M, Schupf N, Stern Y, Vonsattel JPG, Mayeux R. Frequency 86. Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, et al. Updated
and course of mild cognitive impairment in a multiethnic community. research nosology for HIV-associated neurocognitive disorders. Neurology.
Ann Neurol. 2008;63(4):494-506. doi:10.1002/ana.21326. 2007;69(18):1789-99. doi:10.1212/01.WNL.0000287431.88658.8b.
65. Plassman BL, Langa KM, Fisher GG, Heeringa SG, Weir DR, Ofstedal MB, 87. Rodrigues RA, Oliveira RL, Grinsztejn B, Silva MTT. Validity of
et al. Prevalence of cognitive impairment without dementia in the United the International HIV dementia scale in Brazil. Arq Neuropsiquiatr.
States. Ann Intern Med. 2008;148(6):427-34. doi:10.7326/0003-4819- 2013;71(6):376-9. doi:10.1590/0004-282X20130042
148-6-200803180-00005. 88. Gascón MRP, Vidal JE, Mazzaro YM, Smid J, Marcusso RMN, Capitão CG,
66. Di Carlo A, Lamassa M, Baldereschi M, Inzitari M, Scafato E, Farchi G, et al. Neuropsychological Assessment of 412 HIV-Infected Individuals in
et al. CIND and MCI in the Italian elderly: frequency, vascular risk factors, São Paulo, Brazil. AIDS Patient Care STDS. 2018;32(1):1-8. doi:10.1089/
progression to dementia. Neurology. 2007;68(22):1909-16. doi:10.1212/ apc.2017.0202
01.wnl.0000263132.99055.0d. 89. 2019 American Geriatrics Society Beers Criteria® Update Expert Panel.
67. Overton M, Pihlsgard M, Elmstahl S. Prevalence and Incidence of Mild American Geriatrics Society 2019 Updated AGS Beers Criteria® for
Cognitive Impairment across Subtypes, Age, and Sex. Dement Geriatr Potentially Inappropriate Medication Use in Older Adults. J Am Geriatr
Cogn Disord. 2019;47(4-6):219-32. doi:10.1159/000499763. Soc. 2019;67(4):674-94. doi:10.1111/jgs.15767.

16   Syndromic approach to SCD, MCI, and dementia.   Smid J, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):1-17

90. Caramelli P, Teixeira AL, Buchpiguel CA, Lee HW, Livramento JA, 93. Loeffler T, Schilcher I, Flunkert S, Hutter-Paier B. Neurofilament-Light Chain as
Fernandez LL, et al. Diagnosis of Alzheimer’s disease in Brazil: Biomarker of Neurodegenerative and Rare Diseases With High Translational
Supplementary exams. Dement Neuropsychol. 2011;5(3):167-77. Value. Front Neurosci. 2020;14:579. doi:10.3389/fnins.2020.00579.
doi:10.1590/S1980-57642011DN05030004. 94. Palmqvist S, Janelidze S, Quiroz YT, Zetterberg H, Lopera F,
91. Petersen RC, Aisen P, Boeve BF, Geda YE, Ivnik RJ, Knopman DS, et al. Stomrud E, et al. Discriminative Accuracy of Plasma Phospho-tau217
Mild cognitive impairment due to Alzheimer disease in the community. for Alzheimer Disease vs Other Neurodegenerative Disorders. JAMA.
Ann Neurol. 2013;74(2):199-208. doi:10.1002/ana.23931. 2020;324(8):772-81. doi:10.1001/jama.2020.
92. Thijssen EH, La Joie R, Wolf A, Strom A, Wang P, Iaccarino L, et al. 95. Lesman-Segev OH, La Joie R, Iaccarino L, Lobach I, Rosen HJ, Seo SW,
Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease et al. Diagnostic Accuracy of Amyloid versus 18F-Fluorodeoxyglucose
and frontotemporal lobar degeneration. Nat Med. 2020;26(3):387-97. Positron Emission Tomography in Autopsy-Confirmed Dementia.
doi:10.1038/s41591-020-0762-2. Ann Neurol. 2021;89(2):389-401. doi:10.1002/ana.25968.

Smid J, et al.   Syndromic approach to SCD, MCI, and dementia.   17


Dement Neuropsychol 2022 September;16(3 Suppl. 1):18-20

https://doi.org/10.1590/1980-5764-DN-2022-S101EN

Supplementary Material I
BRIEF COGNITIVE SCREENING BATTERY1

Available at:
https://www.demneuropsy.com.br/imageBank/suplementar/Brief_Cognitive_Screening_Battery.pdf

REFERENCES
1. Nitrini R, Brucki SMD, Yassuda MS, Fichman HC, Caramelli P. The Figure Memory Test: diagnosis of memory impairment in
populations with heterogeneous educational background. Dement Neuropsychol. 2021 Apr-Jun;15(2):173-185. doi: 10.1590/
1980-57642021dn15-020004.

Supplementary Material II
FUNCTIONAL ACTIVITIES QUESTIONNAIRE3

This test, created by Pfeffer et al.1, was modified to be applied in people with heterogeneous
educational attainment by Amaducci et al2. in 1991 for a worldwide epidemiological study
organized by the WHO and then by Nitrini et al., in 1997 3 for an epidemiological
study in Brazil4 so it could be answered directly by the informant. However, if necessary,
the examiner can apply the questionnaire. Answers always follow the same criteria. Ask the
informant to circle the correct answer.

Patient Name:
Date: / /
Informant(s): (degree of kinship or function):

1) Do they handle their own money?


0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

2) Can they buy clothes, food, objects for their home by themselves?
0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

18  
Dement Neuropsychol 2022 September;16(3 Suppl. 1):18-20

3) Can they heat water to make coffee and turn off the stove?
0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

4) Can they prepare a meal?


0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

5) Can they keep up with current events or community or neighborhood ones?


0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

6) Can they pay attention, understand, and discuss a radio or television show or newspaper and magazine news?
0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

7) Can they remember commitments, family events, and holidays?


0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

8) Can they handle their own medications?


0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

9) Can they walk around the neighborhood and find their way back home?
0 = Normal 0 = Never did, but could do it now
1 = Does, with difficulty 1 = Never did it and now would have difficulty
2 = Needs help
3 = Unable

  19
Dement Neuropsychol 2022 September;16(3 Suppl. 1):18-20

10) Can they be safely left at home alone?


0 = Normal 0 = Never stayed, but could stay now
1 = Yes, but with precautions 1 = Never stayed and now would have difficulty
2 = Yes, for short periods
3 = No

Score (0 to 30) = ____

Scores higher than 5 indicate functional impairment

REFERENCES
1. Pfeffer RI, Kurosaki TT, Harrah CH, Chance JM, Filos S. Measurement 3. Herrera E Jr, Caramelli P, Nitrini R. Estudo epidemiológico populacional
of functional activities in older adults in the community. J Gerontol. de demência na cidade de Catanduva, estado de São Paulo, Brasil.
1982;37(3):323-9. doi:10.1093/geronj/37.3.323. Rev Psiquiatr Clín. 1997;25(2),70-3.
2. Amaducci L, Baldereschi M, Amato MP, Lippi A, Nencini P, Maggi S, 4. Herrera E Jr, Caramelli P, Silveira AS, Nitrini R. Epidemiologic
Litvak J. The World Health Organization cross-national research program survey of dementia in a community-dwelling Brazilian population.
on age-associated dementias. Aging (Milano). 1991;3(1):89-96. A lzheimer Dis Assoc Disord. 20 02;16(2):103-8. doi:10.1097/
doi:10.1007/BF03323983. 00002093-200204000-00007.

20  
Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S102EN

Diagnosis of Alzheimer’s disease:


recommendations of the Scientific Department
of Cognitive Neurology and Aging of
the Brazilian Academy of Neurology
Lucas Porcello Schilling1,2,3 , Marcio Luiz Figueredo Balthazar4 , Márcia Radanovic5 ,
Orestes Vicente Forlenza5,6 , Marcela Lima Silagi7,8 , Jerusa Smid8 , Breno José Alencar Pires Barbosa8,9,10 ,
Norberto Anízio Ferreira Frota11,12 , Leonardo Cruz de Souza13 , Francisco Assis Carvalho Vale14 ,
Paulo Caramelli13 , Paulo Henrique Ferreira Bertolucci15 , Márcia Lorena Fagundes Chaves16,17 ,
Sonia Maria Dozzi Brucki8 , Benito Pereira Damasceno4 , Ricardo Nitrini8

ABSTRACT. This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian
Academy of Neurology on the diagnostic criteria for Alzheimer’s disease (AD) in Brazil. The authors conducted a literature review
regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care
levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive,
and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers
is also discussed for both clinical diagnosis (in specific conditions) and research.
Keywords: Alzheimer Disease; Dementia; Diagnosis.

1
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
2
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
3
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
4
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil.
5
Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil.
6
Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brazil.
7
Universidade Federal de São Paulo, Departamento de Fonoaudiologia, São Paulo SP, Brazil.
8
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
9
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
10
Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.
11
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
12
Universidade de Fortaleza, Fortaleza CE, Brazil.
13
Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
14
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brazil.
15
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil.
16
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
17
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brazil.
Corresponding Author: Lucas Porcello Schilling; Email: lucas.schilling@pucrs.br.
Conflict of interest: LPS: Participation in advisory boards for Biogen. Participation as speaker in symposia sponsored by Aché, Apsen, and Biogen laboratories;
MLFB: Participation in advisory boards for Biogen. Development of material for continuous medical education and participation as speaker in symposia sponsored
by EMS and Torrent laboratories; JS: Participation as speaker in symposia sponsored by Roche; PC: Participation as principal investigator in clinical trials for Novo
Nordisk and Roche laboratories. Participation in advisory boards for Aché, Biogen, EMS, Nutricia, and Roche laboratories. Development of material for continuous
medical education and participation as speaker in symposia sponsored by Aché, Nutricia, Libbs, Roche, Sandoz, Torrent, and Zodiac laboratories; PHFB: Participation
in advisory boards for Biogen and Novo Nordisk laboratories. Supervision of training activities for Biogen, Janssen-Cilag, and Novo Nordisk laboratories and for
Quintiles. Participation as speaker in symposia sponsored by Apsen, Nutricia, Roche, and Sandoz laboratories; RN: Participation in advisory board for Biogen;
LCS: Participation in advisory board for Biogen. Participation as speaker in symposia sponsored by Biogen. MR, OVF, MLS, BJAPB, NAFF, FACV, MLFC, SMDB, BPD:
There is no conflict of interest to declare.
Received on August 13, 2021; Received in its final version on November 22, 2021; Accepted on April 27, 2022.

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   21


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

DIAGNÓSTICO DA DOENÇA DE ALZHEIMER: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO


DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. Este artigo apresenta o consenso realizado pelo Departamento Científico de Neurologia Cognitiva e do Envelhecimento da Academia Brasileira
de Neurologia sobre os critérios diagnósticos da Doença de Alzheimer (DA) no Brasil. Foi realizada uma revisão da literatura e dos critérios clínicos
e de pesquisa para DA, sendo propostos protocolos para o diagnóstico de DA em níveis de atenção primária, secundária e terciária. Dentro deste cenário
clínico, são apresentados os critérios diagnósticos para DA típica e atípica, além de instrumentos de avaliação clínica, cognitiva e funcional; bem como
propedêutica complementar com exames laboratoriais e de neuroimagem. A utilização de biomarcadores é também apresentada, tanto para o diagnóstico
clínico em situações específicas quanto para pesquisa.
Palavras-chave: Doença de Alzheimer; Demência; Diagnóstico.

INTRODUCTION Pathophysiology
The main hypothesis in AD pathophysiology establishes
Epidemiology and relevance that the degenerative process is triggered by a hyper-

T he continuous aging of the world population


increases the prevalence and incidence of chronic
and neurodegenerative diseases. Currently, dementia
production and/or decreased clearance and consequent
accumulation of amyloid-beta peptide (Aβ) and tau
protein in the affected brain tissues, accompanied by ho-
affects an estimated 50 million people worldwide and meostatic changes that lead to a collapse of the neuronal
has 10 million new diagnoses per year, of which about cytoskeleton. The APP is usually cleaved by the enzyme
60% are due to Alzheimer’s disease (AD). Projections α-secretase (ADAM10), generating soluble peptides
indicate an estimated 150 million people with dementia (APPs); in AD, an alternative and sequential cleavage of
due to AD by 20501. In Brazil, an estimated 1.7 million APP occurs by secretases β (BACE-1) and y, generating
people have dementia, with a prevalence of approximately insoluble Aβ peptides that aggregate and deposit in
1,036/100,000 inhabitants2. the extracellular space, triggering several pathological
events that cause neuronal death and formation of se-
Risk factors nile or neuritic plaques (NPs). Neurofibrillary tangles
Risk factors for AD can be divided into environmental (NFTs) are intracellular deposits composed of hyper-
and genetic. Environmental factors are more related phosphorylated tau protein. The tau protein maintains
to the sporadic form of the disease (late-onset or se- the integrity of intraneuronal microtubules, a function
nile AD), whose main risk factor is aging3. Other risk that is lost after the hyperphosphorylation process.
factors include low schooling level, systemic arterial The initial clinical symptoms of amnestic AD
hypertension, diabetes, obesity, sedentary lifestyle, are related to the increased density of NFTs in
head trauma, depression, smoking, hearing loss, and so- the hippocampal formation, nucleus basalis of
cial isolation4, which can all be prevented and modified. Meynert, and paralimbic regions (fusiform gyrus and
From the genetic point of view, mutations inferior and middle temporal gyri), corresponding to
responsible for the autosomal dominant forms stages III and IV of Braak’s neuropathological staging7.
of AD stand out. Unlike the multifactorial etiology of In preclinical stages of AD (Braak stages I and II), NFTs
late-onset sporadic AD, autosomal dominant forms occur almost exclusively in limbic system structures
(which are relatively rare) have early onset, occurring (entorhinal cortex, subiculum, and hippocampus,
before 65 years (presenile AD), and are strongly in addition to the amygdala, nucleus basalis of Meynert,
associated with mutations of the amyloid precursor and temporopolar cortex). In mild AD (Braak stage V),
protein (APP), presenilin 1, or presenilin 2 genes, which the density of NFTs increases in the limbic system
are identified in 70% of cases, and with a dominant, and they emerge at the associative neocortical regions
autosomal inheritance pattern5. of the middle and superior temporal gyri (related
Though late-onset forms of AD are rarely associated to language symptoms) as well as at the prefrontal,
with dominant inheritance, they can be related retrosplenial, and posterior parietal cortices (related
to genetic risk factors such as the ε4 allele of the to executive dysfunction and spatial disorientation).
apolipoprotein E (APOE) gene, which increases the NPs also deposit in these areas, progressively increasing
risk for AD development and anticipates its onset in the density of NFTs and NPs in the whole neocortex,
a few years. Homozygosis for the APOEε4 allele increases including unimodal (visual, auditory, and somestesic)
this risk in five times compared to heterozygotes6. and multimodal association areas in the temporoparietal

22   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

and dorsolateral frontal junction. Severe AD corresponds appear as subjective memory impairment (SCI) followed
to Braak advanced stage VI, in which all cortical by mild cognitive impairment (MCI) – usually of the
association areas and the basal ganglia are affected multiple-domain amnestic type (also impairing the
by NFTs and NPs, with relative sparing of the motor language and executive functions) –, later evolving to
and sensory cortices. full-blown dementia, when they begin to interfere in the
Other pathophysiological mechanisms in AD activities of daily living and in the patient’s autonomy14.
include synaptic dysfunction, neurotransmitters
(mainly acetylcholine) and neurotrophin depletion, Logopenic variant of primary progressive aphasia (lvPPA)
mitochondrial dysfunction and deficits in insulin
The first and predominant symptoms in this variant are
signaling pathways, increase in oxidative stress and
language alterations with non-fluent speech, pauses due
inflammation, vascular changes 8, and cholesterol
metabolism9. Recent studies suggest that the interaction to word-finding difficulty, and errors (also phonological)
between different pathophysiological processes, when repeating long sentences and in spontaneous
such as white matter involvement associated with Aβ speech, but preserving semantics, syntax (grammar),
accumulation10 and soluble Aβ oligomers interaction comprehension of single words, and motor production
with other proteins (such as α-synuclein and tau), of speech15,16.
destabilize microtubules, mitochondrial and synaptic
dysfunction, and neurodegeneration11,12. Posterior cortical atrophy (PCA) or posterior variant
PCA is a rare form of AD which usually starts between
the ages of 50 and 60 years. Its occipitotemporal variant
CLINICAL CHARACTERIZATION presents with impairment of the visual identification
of objects, faces, or symbols; its biparietal variant,
Clinical characterization more common, is characterized by visual-spatial dys-
AD usually manifests in the typical amnestic presenta- function, topographical disorientation, poor hand-eye
tion, with a predominant difficulty of episodic memory
coordination, limb apraxia, visual neglect, and at clinical
associated with degenerative lesions of medial temporal
evaluation, elements of Balint’s syndrome (optic atax-
structures. This pattern occurs in about 85% of cases.
ia, oculomotor apraxia, and simultanagnosia) and/or
Atypical and less frequent presentations may begin
mainly with impairment in language, visual-spatial, Gerstmann syndrome (acalculia, agraphia, left-right
executive, or complex motor functions. The most com- disorientation, and finger agnosia)17,18.
mon atypical (usually presenile) forms are the logopenic In the early stages of the disease, episodic memory,
variant of primary progressive aphasia (lvPPA) and language, and executive functions are still relatively
the visual-spatial-apraxic of posterior cortical atro- preserved. PCA presents with AD neuropathology
phy (PCA), whereas the least common forms are the in 62 to 100% of cases19.
corticobasal syndrome (CBS) and the behavioral and
dysexecutive variant (ADbdv). Behavioral and dysexecutive variant (ADbdv)
Due to these non-amnestic variants, memory The AD dysexecutive variant (ADdv) affects mainly
impairment is no longer mandatory for AD diagnosis planning, working memory, and multi-tasking, with loss
according to the most recent diagnostic criteria of inhibitory control and alternating attention, de-
from the Brazilian Academy of Neurology 13 based pression, anxiety and neuropsychiatric symptons.
on the National Institute on Aging-Alzheimer’s In turn, behavioural symptoms are rare.
Association (NIA-AA)14. To help identify these initial
Though the behavioral variant of AD is similar to
forms of AD presentation, we will detail their clinical
the behavioral variant of frontotemporal dementia
characteristics, evolution stages, and corresponding
neuropathological substrates. (bvFTD), it presents greater deficits in memory,
apathy, delusional ideas, and hallucinations, with less
AD amnestic presentation disinhibition, compulsive or persevering behavior,
The typical AD presentation starts with difficulties affective indifference, or personality change 20-23.
remembering messages and recent news and repeat- This presentation of AD is rare, occurring in about
ing the same questions, comments, and narratives. 2% of large samples of AD patients and with 7-20%
Initially mild and intermittent, the symptoms first of patients clinically diagnosed as FTD19,24,25.

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   23


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

Corticobasal syndrome (CBS) fecal incontinence, parkinsonism, myoclonus, epileptic


CBS manifests with remarkably asymmetric or unilater- seizures (in up to 20% of cases)30,31, and gait difficulties.
al signs and symptoms of stiffness, dystonia, my- Later, patients have difficulty sitting and swallowing.
oclonus, bradykinesia, and tremor. It is associated The average survival time is five to 12 years after
with gait alteration, asymmetric apraxia, alien hand the onset of symptoms, but with significant variability
phenomenon, sensory hemineglect, and visual-spa- among patients32.
tial deficits, besides the more typical symptoms of
episodic and visual-spatial memory deficits and apha- Diagnosis
sia19,23. AD causes 15-50% of CBS cases, degenerating
cortical structures and basal ganglia, including the Clinical diagnosis of AD
substantia nigra, and manifesting clinically mainly The clinical diagnosis of AD dementia is based on
as motor symptoms. a thorough evaluation, especially of the patient’s
affected cognitive domains and functional impair-
Clinical stages of dementia ment, as described in the diagnostic criteria and
neuropsychological assesment section 13,14 . AD is
Mild dementia a progressive pathological process with different clinical
The mild dementia stage is characterized by progres- stages, and dementia occurs when pathological changes
sive worsening of amnestic symptoms associated with have already spread33.
other cognitive disorders, such as impaired working Understanding this cognitive continuum is
memory, attentional control (difficulty multitasking), essential for an appropriate clinical evaluation
language alterations (anomia), executive dysfunction of the patient and, with complementar y tests
(struggles with planning, problem-solving), and tem-
(including biomarkers), an accurate diagnosis in
poral-spatial disorientation26.
atypical or early-onset cases. Biomarkers also allow
Neuropsychiatric symptoms occur in all stages (in up
identifying patients and indicating possible future
to 80% of cases) and worsen as dementia progresses,
specific treatments for AD34.
especially apathy, depression, anxiety27,28, and a lack of
awareness regarding cognitive deficits (anosognosia),
Diagnosis of dementia due to Alzheimer’s disease (modified from
which occurs in up to 50% of patients19,29.
McKhann et al., 201113 and Frota et al., 201114)
• Probable Alzheimer’s disease dementia
Moderate dementia
If the patient meets the criteria for diagnosis
In the moderate dementia stage, patients become
more dependent on others to perform instrumen- of dementia35 and has the following characteristics:
tal activities of daily living (although still capable I. Insidious onset (months or years);
of self-care) and have greater difficulty remembering II. Clear history or observation of cognitive worsening;
the name of relatives, remote events, or more signif- III. Initial and more prominent cognitive deficits
icant recent events. Other cognitive symptoms may in one of the following categories:
worsen, such as temporal and spatial disorientation, • Amnestic presentation (there must be another
development of transcortical sensory aphasia, ideo- affected domain).
motor apraxia, dyscalculia, visual agnosia, and neu- • Non-amnestic presentation (there must be
ropsychiatric symptoms such as delusions (typically another domain affected).
of betrayal or theft), hallucinations, and agitation, • Language (memory of words).
with or without aggressiveness. • Visual-spatial (spatial cognition or agnosia
for objects or faces, simultanagnosia,
Severe dementia and alexia).
In the severe dementia stage, patients are entirely • Executive functions (alterations of reasoning,
dependent on a caregiver, with memory reduced judgment, and problem-solving).
to fragments of information, temporal and personal IV. Tomography or, preferably, magnetic resonance
disorientation (maintaining only self-awareness), imaging of the brain should be performed
and speech restricted to a few unintelligible words. to exclude other diagnostic possibilities or
In more advanced stages, they can present urinary and comorbidities, especially cerebrovascular disease;

24   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

V. The diagnosis of probable AD dementia should III. Visual-spatial or praxic skills: Does the patient
not be applied when there are: have difficulty orienting themselves spatially
• Evidence of significant cerebrovascular (outside and indoors), dressing, combing,
disease defined by a history of stroke shaving, using everyday objects, recognizing
temporally related to the onset or worsening familiar faces? Have they lost dexterity in tasks
of cognitive impairment; presence of multiple in which they used to do well?
or extensive brain infarctions; or extensive IV. Language: Does the patient have difficulty
lesions in the white matter evidenced finding words in conversations or naming
by neuroimaging; or objects and people? Or in understanding
• Central features of dementia with Lewy words or sentences, explaining situations and
bodies (visual hallucinations, parkinsonism, making themselves understood, presenting poor
REM sleep behavior disorder, and cognitive vocabulary and reduced speech fluency?
fluctuation); or
• Prominent features of the behavioral Neuropsychological assessment
variant of FTD (hyperorality, hypersexuality, According to specific studies on the subject 36,37 ,
perseveration); or
the diagnosis of AD in its initial stage (or MCI)
• Prominent characteristics of PPA manifesting
has greater reliability when using two tests for
as semantic variant (with fluent speech,
each of the four cognitive domains most affected
anomia, and semantic memory difficulties)
by the disease and greater sensitivity when de-
or as non-fluent variant (with agrammatism
fining deficit score as >1 standard deviation (SD),
and/or marked speech apraxia); or
and not >1.5 or >2SD), relative to normative values.
• Evidence of concomitant neurological or
Thus, in addition to conducting a global cognitive test
non-neurological active disease, or medication
(MMSE, Mini-Mental State Examination; or MoCA,
use that may substantially affect cognition.
Montreal Cognitive Assessment), the evaluation must
Anamnesis include episodic memory, language, executive func-
tions, and visual-spatial functions, with two subtests
A detailed anamnesis focused on the most common
for each cognitive domain.
cognitive and neuropsychiatric alterations of AD al-
The main instruments recommended for cognitive
lows diagnosing the disease more safely, establishing
assessment in AD in Brazil are presented below.
its subtype based on its initial presentation and stage,
and differentiating it from other neurodegenerative Given the country’s socio-cultural and educational
diseases. Interrogation of patients and their relative/ heterogeneity, it is advisable to use instruments with
caregiver should cover (1) neuropsychiatric disorders cutoff scores adjustable by level of education to avoid
such as depression, anxiety, apathy, delusional ideas, false-positive results in the diagnostic process 38.
hallucinations, and aberrant or uninhibited motor be- The instruments are subdivided into cognitive
haviors, which are socially inappropriate; and (2) cogni- screening tests, specific tests for evaluating different
tive difficulties in the following domains, most affected cognitive domains, and instruments for assessing
by the disease: functionality (Table 1).
I. Episodic memory: Does the patient forget More recently, the sum of boxes (CDR-Sum
recent facts and dates, items to purchase, of Boxes – CDR-SB) have replaced the Clinical Dementia
appointments, or places where he/she keeps Rating (CDR) global scores. The first allows detecting
objects? Or does he/she keep repeating the same smaller differences within and between subsequent
questions or comments? global scores as well as within and between stages
II. Executive functions: Does the patient have of the disease, helping differentiate MCI from initial
difficulty staying focused, making decisions, dementia, as seen in O’Bryant et al.87, who described the
planning activities, solving everyday problems, following ranges of CDR-SB corresponding to CDR-GS
shopping, and dealing with small amounts scores: 0.5 to 4.0 for the CDR-GS of 0.5; 4.5 to 9.0
of money? Do they present loss of motivation for CDR-GS of 1; 9.5 to 15.5 for CDR-GS of 2; and 16.0
and initiative? Do they have impaired judgment? to 18.0 for CDR-GS of 3.

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   25


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

Table 1. Main instruments for cognitive assessment in AD.


Type of instrument Main tests and normative studies

Screening tests

Mini-Mental State Examination (MMSE)39,40, Montreal Cognitive Assessment (MoCA)41,42, Cognitive Abilities
Brief tests
Screening Instrument – Short Version (CASI-S)43,44, Brief Cognitive Screening Battery (BBRC)45,46

Addenbrooke’s Cognitive Examination – revised version (ACE-R)47,48, Cambridge Cognitive Examination


Multi-functional batteries (CAMCOG)49-51, Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-COG)52,53, Consortium
to Establish a Registry for Alzheimer’s Disease (CERAD)54,55, Mattis Dementia Rating Scale (MDRS)56,57

Evaluation of single cognitive domains

Verbal episodic memory Rey Auditory Verbal Learning Test (RAVLT)58,59, CERAD Battery Word List Learning Subtest

Subtest “figure recognition” (BBRC), Subtest “geometric figure recall” (CERAD Battery),
Nonverbal memory
Rey-Osterrieth Complex Figure60,61

Language Verbal Fluency Test (phonemic and semantic)62,63, Boston Naming Test (BNT)64,65

Attention control and executive function Digit Span Task in forward and inverse order66,67, Clock Drawing Test68,69, Verbal Fluency Test

Visual-spatial /
CERAD / MoCA Figure Copy Subtest, Clock Drawing Test
visual-constructive abilities

Functional assessment

Functional Activities Questionnaire (FAQ)70,71, Informant Questionnaire on Cognitive Decline in the Elderly
(IQCODE)72,73, Direct Assessment of Functional Status-Revised (DAFS-R)74,75, Disability Assessment
Instrumental activities of daily living
for Dementia (DAD)76,77, Activities of Daily Living Questionnaire (ADLQ)78,79, Bayer Activities of Daily
Living Scale (B-ADL)80,81, AD8 Dementia Screening Interview82

Basic activities of daily living Katz scale83,84, Functional Activities Questionnaire (FAQ)70,72

Dementia staging Clinical Dementia Rating scale (CDR)85,86


Note: The global CDR scores (CDR-GS: 0, 0.5, 1, 2, or 3) have the limitation of being based on the scores of the item Memory, considering the other items as secondary, and thus
underestimating relevant information from instrumental activities that may be primarily and early affected.

Laboratory tests in AD clinical propaedeutics to rule out secondary lesions and to identify patterns
Several clinical conditions may cause cognitive impair- of brain atrophy specific to the disease. MRI provides
ment, such as hypothyroidism, hypovitaminosis, and better anatomical resolution and different acquisition
neurosyphilis. An initial medical evaluation should techniques that are more useful than CT for differen-
conduct a basic laboratory evaluation to rule out the tial diagnoses with other dementias, such as those of
main secondary causes of cognitive decline. It should vascular or prion pathology.
also seek to identify systemic diseases and comorbid- As a neurodegenerative disease, AD invariably
ities that could worsen the neurological condition, occurs with cerebral atrophy. The most common pattern
such as dyslipidemia and diabetes88-91. of volumetric alteration is atrophy of mesial temporal
The list of recommended laboratory tests should structures (MTS), in structures such as the hippocampus
include hematological, renal, hepatic, lipid, and metabolic and entorhinal cortex, which correlates with the clinical
profiles (serum sodium, potassium, and calcium), findings of episodic memory deficit. However, atrophy
fasting glucose, folic acid dosage, vitamin B12, TSH, can also affect different regions, especially in atypical
free T4, syphilis serology, and especially in atypical cases presenile presentations, such as linguistic, dysexecutive
or in case of clinical suspicion, HIV testing92. and/or behavioral (frontal), and visual-spatial variants,
among others, which will be discussed later93.
Structural neuroimaging Computed tomography (CT) is a useful, more available,
Brain evaluation by structural neuroimaging such as and lower-cost effective study that can be used even in
computed tomography (CT) or magnetic resonance im- primary health care. Similarly to MRI, CT can rule out
aging (MRI) is essential for properly diagnosing AD, both structural lesions such as subdural hematoma, tumors,

26   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

and hydrocephalus94. It can also evaluate hippocampal differentiate AD from normal older adults, although
atrophy, especially via coronal plane reconstruction. other dementias may also present hippocampal atrophy,
Although MRI has a higher anatomical resolution such as vascular dementia or dementia with Lewy
than CT, the Medial Temporal Atrophy Scale (MTA or bodies. The Scheltens scale assesses the width of the
Scheltens scale) can also be used in CT (Figure 1)95. choroidal fissure and temporal horn as well as the height
The MTA is sensitive to diagnose AD and specific to of the hippocampus(Figure 2)94,96.

Score Width of the choroidal fissure Length of the temporal horn Hippocampus height
0 Normal Normal Normal
1 ↑ Normal Normal
2 ↑↑ ↑ ↓
3 ↑↑↑ ↑↑ ↓↓
4 ↑↑↑ ↑↑↑ ↓↓↓
Figure 1. Application of the MTA scale on MRI (above) and CT (below). Under 75 years old, ≥2 is abnormal; over 75 years old, ≥3 is abnormal95.

Figure 2. Structures evaluated in the MTA scale. A: temporal horn; B: choroidal fissure; C: hippocampus.

As aforementioned, MRI presents better anatomical and specificity of 80 to 85% in differentiating individuals
resolution and allows performing other imaging with AD from cognitively normal individuals 97 .
techniques. Visual inspection of MTS using scales such Some radiological centers and software available on
as the MTA is still the most widespread and available the Internet measure hippocampal volume by MRI,
method in the clinical diagnosis of AD, with sensitivity which may increase sensitivity/specificity for diagnosis.

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   27


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

Although this information is essential, especially for Biomarker-assisted diagnosis


follow-up, it still lacks standardization for the Brazilian
population considering age and sex. Regarding MTS Biomarkers in cerebrospinal fluid
in AD, the subregions of the hippocampus, such as CA1 The cerebrospinal fluid (CSF) biomarkers used for AD di-
and subiculum, can be quantified, but without widespread agnosis are the 42-amino acid Aβ peptide (Aβ1-42) and
clinical application98. A recent review of MCI due to the tau protein in its total composition and as phos-
AD showed low sensitivity/specificity (73 and 71%, phorylated residue at threonine 181 (T-tau and P-tau,
respectively) of MTS measures to differentiate patients respectively). The “AD pathological signature” in CSF
whose condition evolved or not to dementia99. consists of a pattern determined by reduced Aβ1-42
Other MRI techniques do not yet have a consolidated concentration and increased concentrations of T-tau
role in clinical practice for AD diagnosis. As an example, and P-tau102-104.
although the MRI proton magnetic resonance spectroscopy In recent years, the use of CSF AD biomarkers
technique – which assesses brain metabolites such as for diagnosis has significantly advanced. Ideally,
N-acetylaspartate (Naa), creatine (Cr), and myo-inositol concentrations of Aβ1-42 should be normalized
(mI) – shows differences between groups of patients with in relation to those of Aβ1-40, which do not vary
and without AD, it still has many limitations and technique significantly among dementias. The ratio Aβ1-42/Aβ1-40
heterogeneity as to be applied in clinical practice as can also better predict the PET measurement of amyloid
a marker of the disease. The main findings of the technique load than the concentration of Aβ1-42 alone105-107.
are a decrease in NAA and NAA/Cr ratio and an increase There is a correspondence between the pattern of
in mI and mI/Cr ratio100. CSF biomarkers and the pathophysiological changes
underlying AD. The reduction of Aβ1-42 and the
Several other techniques, such as diffusion tensor
increase of P-tau in CSF indicate cerebral amyloidosis
imaging, texture analysis, MRI infusion, and functional
and tauopathy, mechanisms that form, respectively,
connectivity, are still restricted for research.
NPs and NFTs. The increase in T-tau signals the ongoing
neurodegenerative process, usually represented by structural
Structural neuroimaging in atypical AD
changes (atrophy) and regional metabolic impairment.
A common feature of atypical presentations of AD, is the
relative preservation of MTS in relation to atrophy
Molecular neuroimaging biomarkers
of other brain regions. Each variant has its imaging
Pathophysiological processes related to AD can be
characteristics, which generally correlate with clinical
alternatively inferred in vivo by molecular imaging
symptoms. Some neuroimaging characteristics of methods based on positron emission tomography (PET)
the three most common atypical presentations will by injecting different radiotracers.
be briefly described: visual-spatial variant (part of the The progressive degenerative process of AD causes
PCA spectrum); linguistic variant, most commonly cerebral hypometabolism, which can be assessed
lvPPA; and ADbdv. using [18F]Fluorodeoxyglucose (FDG) as a tracer.
• PCA: predominance of parietal and posterior Patients with AD present hypometabolism patterns
temporal atrophy. The Koedam’s parietal atrophy involving the posterior cingulate, precuneus,
scale ranges from 0 to 3 and assesses the temporoparietal, and medial temporal cortices108-111.
integrity of the precuneus and dilatation of the Several molecular agents can evaluate the cerebral
posterior cingulate, parieto-occipital, and parietal accumulation of Aβ from peptide Aβ affinity, such as the
lobe sulci. It may be helpful in the diagnosis [11C]Pittsburgh compound-B (PiB), the [18F]Flutemetamol,
of PCA, and scores ≥ 2 can be considered the [18F]Florbetaben, and [18F]Florbetapir 112,113.
abnormal, according to the author’s proposal101; PiB presents a greater limitation to clinical use since it
• lvPPA: asymmetric atrophy of temporoparietal has carbon in its molecular structure, with a half-life of
structures, predominantly in the left hemisphere only 20 minutes. Compared to the measurement of Aβ
(dominant for language)16; in CSF, molecular imaging by PET has the advantage
• ADbdv: is the most heterogeneous presentation of topographically identifying Aβ accumulation in
in terms of image. It presents greater atrophy the brain, involving the precuneus and bilateral
in the dorsolateral prefrontal cortex compared fronto-temporo-parietal cortices113.
to typical AD but may also present a pattern of PET-specific radiotracers, such as [18F]Flortaucipir,
temporoparietal atrophy20. can also assess the accumulation of tau protein,

28   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

an essential pathological characteristic of AD 114. the underlying presence of one or more pathogenic
Tau accumulation observed in molecular neuroimaging processes characteristic of AD, where: “A” is the infor-
studies has significant clinical correlations: a greater mation obtained by biomarkers indicative of cerebral
accumulation of tau is related to the increased severity Aβ accumulation (i.e., Aβ1-42 reduction in the CSF or
of cognitive decline115-117 and, in atypical AD, the regions positivity in molecular imaging methods of amyloid
with higher radiotracer retention are associated detection); “T” is the positivity of biomarkers indicative
with symptoms related to these regions, such as of tau hyperphosphorylation (increased P-Tau in CSF or
occipital lobes in PCA, and to the pattern of glycolytic positivity in molecular imaging methods with tau-PET);
hypometabolism118. Aβ biomarkers have good sensitivity and “N” indicates the occurrence of neurodegeneration
to identify cases of incipient AD 119 whereas P-Tau by the increase of T-Tau levels in the CSF, hippocampal
markers have greater specificity to diagnose AD120,121. and/or temporoparietal atrophy in MRI, or loss of re-
Currently, biomarkers are used mainly in research. gional cerebral metabolic integrity, according to glucose
From the clinical point of view, biomarkers should uptake profile by [18F]FDG-PET122,123. According to
be used to assess conditions considered atypical, specific topographic patterns, such measurements are
either for an initial non-amnestic clinical presentation further related to the presence of degenerative process
or in patients with early-onset dementia, which will at an early stage, corresponding to the degree of cerebral
be further addressed in the next section. atrophy and cognitive decline115,124.
According to the 2018 NIA/AA recommendations,
When to request CSF biomarkers in AD? which included the AT(N) classification, the AD
The indications for the CSF examination , in general, diagnostic spectrum requires evidence of accumulation
did not change since the last Brazilian Academy of Aβ peptide and AD is defined by the combination of
of Neurology consensus92. CSF examination is thus the positivity of this biomarker (A+) and biomarkers
indicated in the investigation of presenile dementia indicative of phosphorylated tau protein (T+). Positivity
(before 65 years) and in cases with atypical clinical pre- of biomarkers for cerebral amyloidosis (A+) and
sentation or course, communicating hydrocephalus, neurodegeneration (e.g., increase in CSF total tau)
and any evidence or suspicion of inflammatory, infec- (N+) in the absence of P-tau increase (T-) markers
tious, or prion disease of the central nervous system. concomitantly reflects AD pathological alteration and
I f the entire diagnostic process involving suspected non-AD pathological process. In short:
anamnesis, cognitive assessment, general examination, A -: not in the AD spectrum
and neuroimaging studies, the etiology of the dementia A+: AD spectrum
syndrome remains doubtful, CSF biomarkers may A+/T+: AD
be helpful. This uncertainty commonly arises in the A+/T-/N+: AD + suspicion of another non-AD
differential diagnosis of atypical AD presentations with pathological process
other dementias, such as the behavioral variant of FTD NIA/AA authors122 advise that these recommendations
or agrammatic and semantic PPA. apply only to research (observational and interventional)
CSF biomarkers can also be used in the workup and should not be understood as guidelines or diagnostic
for cognitive disorders to predict dementia in criteria for clinical practice, considering that: (1) they do
oligosymptomatic cases, where the identification of the not consider clinical symptoms, signs, and functional
AD ‘pathological signature’ in MCI allows inferring the impairment; and (2) 40% of cognitively normal older
underlying etiology of the disease with high accuracy. adults may present AD biomarkers and neuropathological
The development of potentially disease-modifying alterations125, of which about 20% will never develop
drugs, such as anti-Aβ monoclonal antibodies, dementia, even in their nineties126.
will promote CSF biomarker assessment, which is
essential for indicating these medications. Pathological diagnosis of AD
The pathological diagnosis of AD is based on the pres-
Biological diagnosis of AD through biomarkers ence of cortical atrophy, especially in the hippocam-
Diagnosis assisted by biomarkers allowed formulating pus and frontoparietal regions (associative areas),
recommendations for the biological diagnosis of AD with marked neuronal loss and extracellular NPs and
by the A/T(N) classification122. According to this pro- intraneuronal NFTs, which are the histopathological
posal, suspected cases of AD can be classified according markers of AD that establish its definitive diagno-
to the positivity of biomarkers that allow inferring sis. These markers are initially formed in the limbic

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   29


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

system (hippocampus and entorhinal cortex), pro- Diagnosis of AD at different levels of health care
gressing to the association cortex, subcortical nuclei, In Brazil, economic realities and access to health ser-
and brainstem structures. Other neuropathological vices are quite heterogeneous. This requires adapting
findings include neuronal loss in the pyramidal the instruments and diagnostic approaches within
layers of the cerebral cortex and synaptic degener- the levels of care established in the Guidelines of the
ation affecting associative limbic and cortical areas, Unified Health System (Sistema Único de Saúde – SUS)
starting from the hippocampus, with relative preser- (Primary, Secondary, and Tertiary Care).
vation of the primary areas (motor, somatosensory, Table 2 shows the suggested protocols for the
and visual). evaluation and diagnosis of AD at each level of care:

Table 2. Suggested protocol for the diagnosis of AD at each level of health care.
Diagnosis in Primary Care

Ask the patient and their relative/caregiver about cognitive, neuropsychiatric, and behavioral symptoms. The interviews
Anamnesis
should be preferably conducted separately.

Perform general physical examination looking for signs of systemic diseases and a complete neurological examination
Clinical examination
attentive to focal signs.

Laboratory tests Perform laboratory tests to detect causes of secondary dementias and comorbidities that may contribute to the clinical picture.

Applying a brief cognitive screening test, such as MoCA, BBRC, or CASI-S is suggested. BBRC stands out due to its easy
application and high sensitivity, even for individuals with low schooling levels. Moreover, applying brief highly sensitive tasks,
Cognitive assessment such as semantic verbal fluency (animals) and a word-learning test, which are highly accurate to detect dysfunction of the
hippocampal system (amnesia), is recommended. Data on functionality can be addressed in the anamnesis or specific brief
questionnaires, such as the Functional Activities Questionnaire (FAQ).

A brain CT is essential to rule out other causes of dementia (such as tumors, hydrocephalus, or cerebral infarctions)
Structural neuroimaging
and to identify, within the limitations of the method, patterns of atrophy compatible with AD.

Diagnosis in Secondary Care

Anamnesis Ask the patient and their relative/caregiver about cognitive, neuropsychiatric, and behavioral symptoms.

Perform general physical examination looking for signs of systemic diseases and a complete neurological examination
Clinical examination
attentive to focal signs.

Perform laboratory tests to detect causes of secondary dementias and comorbidities that may contribute to the clinical
Laboratory tests
picture. If chronic meningitis is suspected, a lumbar puncture should be performed for CSF analysis.

Using a brief test of cognitive screening or multifunctional battery of medium coverage is suggested, with at least one task
Cognitive assessment
to examine each cognitive domain; complement by applying a functional assessment tool, as described above.

Brain CT or MRI (preferably) is essential to rule out other causes of dementia and further investigate mesial structures,
Structural neuroimaging
with visual scales or manual or automated volumetry.

Using biomarkers is indicated in cases of diagnostic doubt between AD and other neurodegenerative dementias not in
the amyloid spectrum, such as FTD, to correctly follow the guidelines for the use of AD approved medications, such as
cholinesterase inhibitors and memantine. Other situations are referred to in biomarkers sections. Importantly, the future
Biomarker assessment
emergence and availability of potentially disease-modifying drugs will require evaluating all patients with mild AD as potential
candidates for these treatments. Biomarkers can be requested from the secondary level of health care if they are reserved
for selected cases and requested and interpreted by trained professionals.

Diagnosis in Tertiary Care

Anamnesis Conduct a detailed initial interview with the patient and their relative/caregiver.

Perform a general physical examination looking for signs of systemic diseases and a complete neurological examination
Clinical examination
attentive to focal signs.

Perform laboratory tests to detect causes of secondary dementias and comorbidities that may contribute to the clinical
Laboratory tests picture and other tests if suspecting a relevant systemic disease. If chronic meningitis is suspected, a lumbar puncture
should be performed for CSF analysis.
Continue...

30   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

Table 2. Continuation.
A comprehensive neuropsychological evaluation using a cognitive screening test is suggested for applying instruments
Cognitive assessment
to further examine all cognitive domains and to assess functionality and neuropsychiatric disorders.

Brain MRI is essential to rule out other causes of dementia and further investigate the mesial temporal and other brain
Structural neuroimaging
regions using visual scales or manual or automated volumetry.

FDG-PET or single-photon emission computerized tomography (SPECT) studies may show regional alterations in brain
Functional neuroimaging metabolism or changes in blood flow in cases of incipient and/or mild dementia, even in the absence of structural changes
in neuroimaging.

Using biomarkers is indicated in cases of diagnostic doubt between AD and other neurodegenerative dementias not in
the amyloid spectrum, such as FTD, to correctly follow the guidelines for the use of AD approved medications, such as
Biomarker assessment cholinesterase inhibitors and memantine. Other situations are referred to in biomarkers sections. Importantly, the future
emergence and availability of potentially disease-modifying drugs will require evaluating all patients with mild AD as potential
candidates for these treatments.

FUTURE PROSPECTS instrumentation in AD diagnosis. Moreover, de-


termining plasma levels of Aβ, tau protein, and
Diagnosis of preclinical AD neurofilament light chain (NFL) –another neuronal
The pathogenic process of AD begins many years before cytoskeletal protein – with ultrasensitive methods
the first clinical manifestations of the disease, and the could provide reliable estimates of cerebral amyloi-
analysis of biomarkers indicates the presence of asymp- dogenesis and neurodegeneration in early stages of
tomatic individuals127. Preclinical AD is therefore a long AD121,132,133. Plasma amyloid levels present a reliable
and silent stage of the disease that precedes the first correlation, measured by the relationship between
cognitive alterations which will later lead to the diag- peptides Aβ1-42 and Aβ1-40, and future positivity in am-
nosis of mild cognitive impairment (MCI) due to AD. It yloid PET134. Phosphorylated tau levels are present in
corresponds to a window of opportunity to implement other degenerative disorders but have been reported
interventions for delaying (or, ideally, interrupting) as elevated in plasma in individuals with AD, with the
the pathogenic process of AD128. The existence of inter- 181
P-tau form showing greater specificity 133,135 .
ventions modifying the pathogenesis of AD, associated The presence of NFL in CSF indicates nonspecific neu-
with the possibility of identifying the disease in the ronal damage. Recent studies have shown a positive
asymptomatic phase will represent an effective way to correlation between plasma and NFL cerebrospinal
establish the prevention of dementia. fluid levels, but only regarding neurodegeneration136.
The diagnostic criteria for preclinical AD, restricted A recent meta-analysis showed that NFL levels in
to the research context, were proposed to identify both CSF and plasma have high diagnostic sensitivity
individuals at risk of AD in the asymptomatic phase129-131. for AD and other neurodegenerative dementias137.
Three evolutionary stages inherent to preclinical Other approaches using genomics, transcriptomics,
AD were proposed: the first shows isolated evidence metabolomics, lipidomics, and proteomics have been
of cerebral amyloidogenesis according to the positivity applied to generate different biomarkers for AD.
of Aβ biomarkers; the second shows evidence of ongoing One study showed that altered microRNAs resulting
neurodegenerative process according to CSF and/or from the failure of the synaptic function are potential
brain imaging biomarkers; and the third, indicates very plasma biomarkers for AD138. A Brazilian study showed
subtle cognitive or behavioral alterations, insufficient decreased levels of ADAM10 PPA-secretases in platelets,
for the diagnosis of MCI129. decreased PSEN1 levels in platelets and leukocytes,
and lower bace1 (β-secretase) levels in leukocytes139.
Peripheral biomarkers of AD
Limitations for using these methods to better diag- Implications of early diagnosis for
nose AD include the low availability and high cost of disease-modifying therapies
molecular image obtainment by PET and the need The biomarker-based classification system proposed in
to perform lumbar puncture to obtain CSF samples. 2018 indicates a broader concept of the pathological
Developing new biomarkers in peripheral blood, process in AD. However, clinical trials still face many
with good diagnostic accuracy and predictive sensi- challenges. Despite a growing understanding that
tivity, would thus significantly advance laboratory clinical evaluation alone is limited for evaluating an

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   31


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

intervention outcome, cognitive improvement mea- ACKNOWLEDGEMENTS


sures are still the main outcomes in all clinical trials. PC, LCS and RN are funded by CNPq, Brazil (bolsa de
Identifying the best molecular targets or a combination produtividade em pesquisa).
of them by developing better protocols to assess the
results of interventions using biochemical, physiolog- Authors’ contributions. LPS, MLFB, MR, OVF, MLS, BPD, RN,
ical, and neuropsychological measures as outcomes is JS, BJAPB: concept; LPS, MLFB, MR, OVF, MLS, BPD, RN:
essential to identify individuals in preclinical stages of drafting of the manuscript; BJAPB, FACV, JS, LCS, LPS,
AD and facilitate early therapeutic interventions. This MLFB, MLFC, NAFF, PC, PHFB, RN, SMDB: critical revision
is the premise of most efforts to find new therapies. of the manuscript for important intellectual content.

REFERENCES
1. Gauthier S, Rosa-Neto P, Morais JA, Webster C. World Alzheimer Report 17. Crutch SJ, Schott JM, Rabinovici GD, Murray M, Snowden JS, van der
2021: Journey through the diagnosis of dementia. London: Alzheimer’s Flier WM, et al. Consensus classification of posterior cortical atrophy.
Disease International (UK); 2021. 313 p. Alzheimers Dement. 2017;13(8):870-84. doi:10.1016/j.jalz.2017.01.014
2. Melo SC, Champs APS, Goulart RF, Malta DC, Passos VMA. Dementias 18. Fumagalli GG, Basilico P, Arighi A, Mercurio M, Scarioni M, Carandini T,
in Brazil: increasing burden in the 2000–2016 period. Estimates et al. Parieto-occipital sulcus widening differentiates posterior cortical
from the Global Burden of Disease Study 2016. Arq Neuropsiquiatr. atrophy from typical Alzheimer disease. Neuroimage Clin. 2020;28:102453.
2020 ;78(12):762-71. doi:10.1590/0004-282X20200059 doi:10.1016/j.nicl.2020.102453
3. Robinson M, Lee BY, Hane FT. Recent progress in Alzheimer’s disease 19. Alladi S, Xuereb J, Bak T, Nestor P, Knibb J, Patterson K, et al. Focal
research, part 2: genetics and epidemiology. J Alzheimers Dis. cortical presentations of Alzheimer’s disease. Brain. 2007;130(10):2636-45.
2017;57(2):317-30. doi:10.3233/JAD-161149 doi:10.1093/brain/awm213
4. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, 20. Ossenkoppele R, Pijnenburg YA, Perry DC, Cohn-Sheehy BI,
et al. Dementia prevention, intervention, and care: 2020 report of the Scheltens NM, Vogel JW, et al. The behavioural/dysexecutive variant of
Alzheimer’s disease: clinical, neuroimaging and pathological features.
Lancet Commission. Lancet. 2020;396(10248):413-46. doi:10.1016/
Brain. 2015;138(9):2732-49. doi:10.1093/brain/awv191
S0140-6736(20)30367-6
21. Ossenkoppele R, Singleton EH, Groot C, Dijkstra AA, Eikelboom WS,
5. Kim JH. Genetics of Alzheimer’s disease. Dement Neurocogn Disord.
Seeley WW, et al. Research criteria for the behavioral variant of Alzheimer
2018;17(4):131-36. doi:10.12779/dnd.2018.17.4.131
disease: a systematic review and meta-analysis. JAMA Neurol.
6. Berkowitz CL, Mosconi L, Rahman A, Scheyer O, Hristov H, Isaacson RS.
2022;79(1):48-60.
Clinical application of APOE in Alzheimer’s prevention: a precision medicine 22. Townley RA, Graff-Radford J, Mantyh WG, Botha H, Polsinelli AJ,
approach. J Prev Alzheimers Dis. 2018;5(4):245-52. doi:10.14283/jpad.2018.35 Przybelski SA, et al. Progressive dysexecutive syndrome due to Alzheimer’s
7. Braak H, Zetterberg H, Del Tredici K, Blennow K. Intraneuronal tau disease: a description of 55 cases and comparison to other phenotypes.
aggregation precedes diffuse plaque deposition, but Amyloid-β changes Brain Commun. 2020;2(1):1-19. doi:10.1093/braincomms/fcaa068
occur before increases of tau in cerebrospinal fluid. Acta Neuropathol. 23. Graff-Radford J, Yong KXX, Apostolova LG, Bouwman FH, Carrillo M,
2013;126(5):631-41. doi:10.1007/s00401-013-1139-0 Dickerson BC, et al. New insights into atypical Alzheimer’s disease in
8. Brundel M, Bresser J, van Dillen JJ, Kappelle LJ, Biessels GJ. Cerebral the era of biomarkers. Lancet Neurol. 2021;20(3):222-34. doi:10.1016/
microinfarcts: a systematic review of neuropathological studies. J Cereb S1474-4422(20)30440-3
Blood Flow Metab. 2012;32(3):425-36. doi:10.1038/jcbfm.2011.200 24. Snowden JS, Stopford CL, Julien CL, Thompson JC, Davidson Y,
9. Sáiz-Vazquez O, Puente-Martínez A, Ubillos-Landa S, Pacheco-Bonrostro J, Gibbons L, et al. Cognitive phenotypes in Alzheimer’s disease and genetic
Santabárbara J. Cholesterol and Alzheimer’s disease risk: a meta-meta- risk. Cortex. 2007;43(7):835-45. doi:10.1016/s0010-9452(08)70683-x
analysis. Brain Sci. 2020;10(6):386. doi:10.3390/brainsci10060386 25. Mendez MF, Joshi A, Tassniyom K, Teng E, Shapira JS. Clinicopathologic
10. Schilling LP, Pascoal TA, Zimmer ER, Mathotaarachchi S, Shin M, Rieder differences among patients with behavioral variant frontotemporal dementia.
CRM, et al. Regional Amyloid-β load and white matter abnormalities Neurology. 2013;80(6):561-8. doi:10.1212/WNL.0b013e3182815547
contribute to hypometabolism in Alzheimer’s dementia. Mol Neurobiol. 26. Reisberg B, Ferris SH, Leon MJ, Crook T. The Global Deterioration Scale
2019;56(7):4916-24. doi:10.1007/s12035-018-1405-1 for assessment of primary degenerative dementia. Am J Psychiatry.
11. Guerrero-Muñoz MJ, Gerson J, Castillo-Carranza DL. Tau oligomers: 1982;139(9):1136-9. doi:10.1176/ajp.139.9.1136
the toxic player at synapses in Alzheimer’s disease. Front Cell Neurosci. 27. Tatsch MF, Bottino CMC, Azevedo D, Hototian SR, Moscoso MA,
2015;9:464. doi:10.3389/fncel.2015.00464 Folquitto JC, et al. Neuropsychiatric symptoms in Alzheimer disease and
12. Sengupta P, Satpute-Krishnan P, Seo AY, Burnette DT, Patterson GH, cognitively impaired, nondemented elderly from a community-based sample
Lippincott-Schwartz J. ER trapping reveals Golgi enzymes continually in Brazil: prevalence and relationship with dementia severity. Am J Geriatr
revisit the ER through a recycling pathway that controls Golgi organization. Psychiatry. 2006;14(5):438-45. doi:10.1097/01.JGP.0000218218.47279.db
Proc Natl Acad Sci U S A. 2015;112(49):E6752-E61. doi:10.1073/ 28. Gauthier S, Cummings J, Ballard C, Brodaty H, Grossberg G, Robert P, et al.
Management of behavioral problems in Alzheimer’s disease. Int Psychogeriatr.
pnas.1520957112
2010;22(3):346-72. doi:10.1017/S1041610209991505
13. Frota NAF, Nitrini R, Damasceno BP, Forlenza OV, Dias-Tosta E, Silva AB,
29. Lacerda IB, Santos RL, Belfort T, Neto JPS, Dourado MCN. Patterns
et al. Criteria for the diagnosis of Alzheimer’s disease. Recommendations
of discrepancies in different objects of awareness in mild and
os the Scientific Department of Cognitive Neurology and Aging of the
moderate Alzheimer’s disease. Aging Ment Health. 2020;24(5):789-96.
Brazilian Academy of Neurology. Dement Neuropsychol. 2011;5(3):146-52.
doi:10.1080/13607863.2018.1544219
doi:10.1590/S1980-57642011DN05030002 30. Palop JJ, Mucke L. Epilepsy and cognitive impairments in Alzheimer’s
14. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, disease. Arch Neurol. 2009;66(4):435-40. doi:10.1001/archneurol.2009.15
Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: 31. Xu Y, Lavrencic L, Radford K, Booth A, Yoshimura S, Anstey KJ, et al.
recommendations from the National Institute on Aging-Alzheimer’s Systematic review of coexistent epileptic seizures and Alzheimer’s disease:
Association workgroups on diagnostic guidelines for Alzheimer’s disease. incidence and prevalence. J Am Geriatr Soc. 2021;69(7):2011-20.
Alzheimers Dement. 2011;7(3):263-9. doi:10.1016/j.jalz.2011.03.005 doi:10.1111/jgs.17101
15. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, 32. Vermunt L, Sikkes SAM, van den Hout A, Handels R, Bos I,
Cappa SF, et al. Classification of primary progressive aphasia and its van der Flier WM, et al. Duration of preclinical, prodromal, and dementia
variants. Neurology. 2011;76(11):1006-14. stages of Alzheimer’s disease in relation to age, sex, and APOE genotype.
16. Montembeault M, Brambati SM, Gorno-Tempini ML, Migliaccio R. Clinical, Alzheimers Dement. 2019;15(7):888-98. doi:10.1016/j.jalz.2019.04.001
anatomical, and pathological features in the three variants of primary 33. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM,
progressive aphasia: a review. Front Neurol. 2018;9:692. doi:10.3389/ et al. Toward defining the preclinical stages of Alzheimer’s disease:
fneur.2018.00692 recommendations from the National Institute on Aging-Alzheimer’s Association

32   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Part I. Clinical and neuropsychological assessment of Alzheimer’s disease.
Dement. 2011;7(3):280-92. doi:10.1016/j.jalz.2011.03.003 Neurology. 1989;39(9):1159-65. doi:10.1212/wnl.39.9.1159
34. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, et al. 55. Bertolucci PHF, Okamoto IH, Brucki SMD, Siviero MO, Toniolo J Neto,
The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Ramos LR. Applicability of the CERAD neuropsychological battery to
Nature. 2016;537(7618):50-6. doi:10.1038/nature19323 Brazilian elderly. Arq Neuropsiquiatr. 2001;59(3A):532-36. doi:10.1590/
35. S m i d J , C é s a r- F r e i t a s K G , D o u r a d o M C N , K o c h h a n n R , S0004-282X2001000400009
Studart-Neto A, Barbosa BJAPB, et al. Subjective cognitive decline, 56. Vitaliano PP, Breen AR, Russo J, Albert M, Vitiello MV, Prinz PN. The
mild cognitive impairment, and dementia – syndromic approach. Dement clinical utility of the dementia rating scale for assessing Alzheimer patients.
Neuropsychol. 2022;16(S1). J Chronic Dis. 1984;37(9-10):743-53. doi:10.1016/0021-9681(84)90043-2
36. Jak AJ, Bondi MW, Delano-Wood L, Wierenga C, Corey-Bloom J, 57. Porto CS, Fichman HC, Caramelli P, Bahia VS, Nitrini R. Brazilian
Salmon DP, et al. Quantification of five neuropsychological approaches version of the Mattis dementia rating scale: diagnosis of mild dementia
to defining mild cognitive impairment. Am J Geriatr Psychiatry. in Alzheimer’s disease. Arq Neuropsiquiatr. 2003;61(2B):339-45.
2009;17(5):368-75. doi:10.1097/JGP.0b013e31819431d5 doi:10.1590/s0004-282x2003000300004
37. Bondi MW, Edmonds EC, Jak AJ, Clark LR, Delano-Wood L, McDonald CR, 58. Rey A. L’examen clinique en psychologie [The clinical examination in
et al. Neuropsychological criteria for mild cognitive impairment improves psychology]. Paris (France): Presses Universitaires de France; 1964. French.
diagnostic precision, biomarker associations, and progression rates. 59. Malloy-Diniz LF, Lasmar VA, Gazinelli LS, Fuentes D, Salgado JV.
J Alzheimers Dis. 2014;42(1):275-89. doi:10.3233/JAD-140276 The Rey Auditory-Verbal Learning Test: applicability for the Brazilian
38. Ortega LFV, Aprahamian I, Borges MK, Cação JC, Yassuda MS. Screening elderly population. Braz J Psiquiatry. 2007;29(4):324-29. doi:10.1590/
for Alzheimer’s disease in low-educated or illiterate older adults in Brazil: S1516-44462006005000053
a systematic review. Arq Neuropsiquiatr. 2019;77(4):279-88. doi:10.1590/ 60. Osterrieth PA. Le test de copie d’une figure complexe; contribution à
0004-282X20190024 l’étude de la perception et de la mémoire [Test of copying a complex
39. Folstein MF, Folstein SE, Folstein, McHugh PR. “Mini-mental state”. figure; contribution to the study of perception and memory]. Arch Psychol.
A practical method for grading the cognitive state of patients for the clinician. 1944;30:206-356.
J Psychiatr Res. 1975;12(3):189-98. doi:10.1016/0022-3956(75)90026-6 61. Foss MP, Bastos-Formigheri MS, Speciali JG. Rey’s complex figures for
40. Brucki SM, Nitrini R, Caramelli P, Bertolucci PH, Okamoto IH. Suggestions for the elderly. Aval Psicol. 2010;9(1):53-61.
utilization of the mini-mental state examination in Brazil. Arq Neuropsiquiatr. 62. Hazin I, Leite G, Oliveira RM, Alencar JC, Fichman HC, Marques PN,
2003;61(3B):777-81. doi:10.1590/s0004-282x2003000500014 et al. Brazilian Normative Data on Letter and Category Fluency Tasks:
41. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, effects of gender, age, and geopolitical region. Front Psychol. 2016;7:684.
Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief screening doi:10.3389/fpsyg.2016.00684
tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53(4):695-9. 63. Caramelli P, Carthery-Goulart MT, Porto CS, Fichman HC, Nitrini R.
Errata em: J Am Geriatr Soc. 2019;67(9):1991. doi:10.1111/ Category fluency as a screening test for Alzheimer disease in illiterate
j.1532-5415.2005.53221.x and literate patients. Alzheimer Dis Assoc Disord. 2007;21(1):65-7.
42. Memória CM, Yassuda MS, Nakano EY, Forlenza OV. Brief screening doi:10.1097/WAD.0b013e31802f244f
for mild cognitive impairment: validation of the Brazilian version of the
64. Kaplan EF, Goodglass H, Weintraub S. The Boston Naming Test. 2a ed.
Montreal cognitive assessment. Int J Geriatr Psychiatry. 2013;28(1):34-40.
Philadelphia: Lippincott Williams & Wilkins; 2001.
doi:10.1002/gps.3787
65. Mansur LL, Radanovic M, Araújo GC, Taquemori LY, Greco LL. Boston
43. Teng EL, Larson EB, Lin KN, Graves AB, Liu HC. Screening for dementia:
Naming Test: performance of Brazilian population from São Paulo.
the Cognitive Abilities Screening Instrument – short version (CASI Short).
Pro Fono. 2006;18(1):13-20. doi:10.1590/S0104-56872006000100003
Proceedings of the 106th Annual Convention of the American Psychological
66. Wechsler D. Wechsler Adult Intelligence Scale-III. San Antonio:
Association; 1998 Aug 14-18; San Francisco, US. Washington, DC:
The Psychological Corporation; 1997.
American Psychological Association; 1998.
67. Zimmermann N, Cardoso CO, Trentini CM, Grassi-Oliveira R, Fonseca RP.
44. Damasceno A, Delicio AM, Mazo DFC, Zullo JFD, Scherer P, Ng RTY, et al.
Brazilian preliminary norms and investigation of age and education effects
Validation of the Brazilian version of mini-test CASI-S. Arq Neuropsiquiatr.
on the Modified Wisconsin Card Sorting Test, Stroop Color and Word test
2005;63(2b): 416-21. doi:10.1590/S0004-282X2005000300010
and Digit Span test in adults. Dement Neuropsychol. 2015;9(2):120-7.
45. Nitrini R, Lefèvre BH, Mathias SC, Caramelli P, Carrilho PE,
doi:10.1590/1980-57642015DN92000006
Sauaia N, et al. Neuropsychological tests of simple application for
68. Shulman KI, Shedletsky R, Silver IL. The challenge of time: clock-drawing and
diagnosing dementia. Arq Neuropsiquiatr. 1994;52(4):457-65. doi:10.1590/
s0004-282x1994000400001 cognitive function in the elderly. Int J Geriatr Psychiatry. 1986;1(2):135-40.
46. Nitrini R, Brucki SMD, Yassuda MS, Fichman HC, Caramelli P. doi:10.1002/gps.930010209
The Figure Memory Test: diagnosis of memory impairment in populations 69. Fuzikawa C, Lima-Costa MF, Uchoa E, Barreto SM, Shulman K; Bambuí
with heterogeneous educational background. Dement. Neuropsychol. Health and Ageing Study. A population based study on the intra and inter-rater
2021;15(2):173-85. doi:10.1590/1980-57642021dn15-020004 reliability of the clock drawing test in Brazil: the Bambuí Health and Ageing
47. Mioshi E, Dawson K, Mitchell J, Arnold R, Hodges JR. The Addenbrooke’s Study. Int J Geriatr Psychiatry. 2003;18(5):450-6. doi:10.1002/gps.863
Cognitive Examination Revised (ACE-R): a brief cognitive test battery 70. Pfeffer RI, Kurosaki TT, Harrah CH Jr, Chance JM, Filos S. Measurement
for dementia screening. Int J Geriatr Psychiatry. 2006;21(11):1078-85. of functional activities in older adults in the community. J Gerontol.
doi:10.1002/gps.1610 1982;37(3):323-9. doi:10.1093/geronj/37.3.323
48. Carvalho VA, Caramelli P. Brazilian adaptation of the Addenbrooke’s 71. Herrera E Jr, Caramelli P, Silveira AS, Nitrini R. Epidemiologic survey of
Cognitive Examination-Revised (ACE-R). Dement Neuropsychol. dementia in a community-dwelling Brazilian population. Alzheimer Dis Assoc
2007;1(2):212-216. doi:10.1590/S1980-57642008DN10200015 Disord. 2002;16(2):103-8. doi:10.1097/00002093-200204000-00007
49. Roth M, Tym E, Mountjoy CQ, Huppert FA, Hendrie H, Verma S, et al. 72. Jorm AF, Jacomb PA. The Informant Questionnaire on Cognitive Decline
CAMDEX. A standardised instrument for the diagnosis of mental disorder in the Elderly (IQCODE): socio-demographic correlates, reliability, validity
in the elderly with special reference to the early detection of dementia. and some norms. Psychol Med. 1989;19(4):1015-22. doi:10.1017/
Br J Psychiatry. 1986;149:698-709. doi:10.1192/bjp.149.6.698 s0033291700005742
50. Nunes PV, Diniz BS, Radanovic M, Abreu ID, Borelli DT, Yassuda MS, et al. 73. Sanchez MA, Lawrence RA. Informant Questionnaire on Cognitive
CAMcog as a screening tool for diagnosis of mild cognitive impairment Decline in the Elderly (IQCODE): adaptação transcultural para uso
and dementia in a Brazilian clinical sample of moderate to high education. no Brasil. Cad Saude Publica. 2009; 25(7):1455-65. doi:10.1590/
Int J Geriatr Psychiatry. 2008;23(11):1127-33. doi:10.1002/gps.2038 S0102-311X2009000700003
51. Aprahamian I, Martinelli JE, Cecato J, Izbicki R, Yassuda MS. Can the 74. Loewnstein DA, Bates CB. The Direct Assessment of Functional
CAMCOG be a good cognitive test for patients with Alzheimer’s disease Status-Revised (DAFS-R). Manual for administration and scoring. Miami:
with low levels of education? Int Psychogeriatr. 2011;23(1):96-101. Mount Sinai Medical Center; 2006.
doi:10.1017/S104161021000116X 75. Pereira FS, Oliveira AM, Diniz BS, Forlenza OV, Yassuda MS. Cross-cultural
52. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. adaptation, reliability and validity of the DAFS-R in a sample of Brazilian
Am J Psychiatry. 1984;141(11):1356-64. doi:10.1176/ajp.141.11.1356 older adults. Arch Clin Neuropsychol. 2010;25(4):335-43. doi:10.1093/
53. Schultz RR, Siviero MO, Bertolucci PH. The cognitive subscale of arclin/acq029
the “Alzheimer’s Disease Assessment Scale” in a Brazilian sample. 76. Gauthier L, Gélinas I, McIntyre M, Gauthier S, Laberge H, Dauphinee SW.
Braz J Med Biol Res. 2001;34(10):1295-1302. doi:10.1590/ Disability Assessment for Dementia (DAD) user’s guide; 1994.
S0100-879X2001001000009 77. Bahia VS, Carthery-Goulart MT, Novelli MM, Kato-Narita EM,
54. Morris JC, Heyman A, Mohs RC, Hughes JP, van Belle G, Fillenbaum G, et al. Areza-Fegyveres R, Caramelli P, et al. Functional disability in Alzheimer
The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). disease: a validation study of the Brazilian version of Disability Assessment

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   33


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

for Dementia (DAD-Br). Alzheimer Dis Assoc Disord. 2010;24(3):291-5. 99. Lombardi G, Crescioli G, Cavedo E, Lucenteforte E, Casazza G,
doi:10.1097/WAD.0b013e3181cfc878 Bellatorre AG, et al. Structural magnetic resonance imaging for the early
78. Johnson N, Barion A, Rademaker A, Rehkemper G, Weintraub S. diagnosis of dementia due to Alzheimer’s disease in people with mild
The Activities of Daily Living Questionnaire: a validation study in patients cognitive impairment. Cochrane Database Syst Rev. 2020;3(3):CD009628.
with dementia. Alzheimer Dis Assoc Disord. 2004;18(4):223-30. doi:10.1002/14651858.CD009628.pub2
79. Medeiros ME, RO Guerra. Translation, cultural adaptation and 100. Maul S, Giegling I, Rujescu D. Proton magnetic resonance spectroscopy
psychometric analysis of the Activities of Daily Living Questionnaire (ADLQ) in common dementias – current status and perspectives. Front Psychiatry.
for functional assessment of patients with Alzheimer’s disease. Braz J 2020;11:769. doi:10.3389/fpsyt.2020.00769
Phys Ther. 2009;13(3):257-66. doi:10.1590/S1413-35552009005000027 101. Koedam EL, Lehmann M, van der Flier WM, Scheltens P, Pijnenburg YA,
80. Hindmarch I, Lehfeld H, Jongh P, Erzigkeit H. The Bayer Activities of Daily Fox N, et al. Visual assessment of posterior atrophy development of
Living Scale (B-ADL). Dement Geriatr Cogn Disord. 1998;9(Suppl 2):20-6. an MRI rating scale. Eur Radiol. 2011;21(12):2618-25. doi:10.1007/
doi:10.1159/000051195 s00330-011-2205-4
81. Folquitto JC, Bustamante SEZ, Barros SB, Azevedo D, Lopes MA, 102. Hansson O, Zetterberg H, Buchhave P, Londos E, Blennow K, Minthon L.
Hototian SR, et al. The Bayer: Activities of Daily Living Scale (B-ADL) in the Association between CSF biomarkers and incipient Alzheimer’s disease in
differentiation between mild to moderate dementia and normal aging. Braz J patients with mild cognitive impairment: a follow-up study. Lancet Neurol.
Psychiatry . 2007;29(4):350-3. doi:10.1590/S1516-44462006005000037 2006;5(3):228-34. doi:10.1016/S1474-4422(06)70355-6
103. Diniz BS, Pinto JA Jr, Forlenza OV. Do CSF total tau, phosphorylated
82. Galvin JE, Roe CM, Powlishta KK, Coats MA, Muich SJ, Grant E, et al.
tau, and beta-amyloid 42 help to predict progression of mild cognitive
The AD8: a brief informant interview to detect dementia. Neurology.
impairment to Alzheimer’s disease? A systematic review and meta-
2005;65(4):559-64. doi:10.1212/01.wnl.0000172958.95282.2a
analysis of the literature. World J Biol Psychiatry. 2008;9(3):172-82.
83. Katz S, Ford AB, Moskowitz RW, Jackson BA, Jaffe MW. Studies of
doi:10.1080/15622970701535502
illness in the aged. The index of ADL: a standardized measure of biological
104. Forlenza OV, Radanovic M, Talib LL, Aprahamian I, Diniz BS, Zetterberg H,
and psychosocial function. JAMA. 1963;185:914-9. doi:10.1001/
et al. Cerebrospinal fluid biomarkers in Alzheimer’s disease: diagnostic
jama.1963.03060120024016 accuracy and prediction of dementia. Alzheimers Dement (Amst).
84. Lino VTS, Pereira SRM, Camacho LAB, Ribeiro ST Filho, Buksman S. 2015;1(4):455-63. doi:10.1016/j.dadm.2015.09.003
Cross-cultural adaptation of the Independence in Activities of Daily Living 105. Dorey A, Perret-Liaudet A, Tholance Y, Fourier A, Quadrio I. Cerebrospinal
Index (Katz Index). Cad Saude Publica. 2008;24(1):103-12. doi:10.1590/ fluid Aβ40 improves the interpretation of Aβ42 concentration for diagnosing
s0102-311x2008000100010 Alzheimer’s disease. Front Neurol. 2015;6:247. doi:10.3389/fneur.2015.00247
85. Hughes CP, Berg L, Danziger WL, Coben LA, Martin RL. A new clinical 106. Racine AM, Koscik RL, Nicholas CR, Clark LR, Okonkwo OC, Oh JM,
scale for the staging of dementia. Br J Psychiatry. 1982;140:566-72. et al. Cerebrospinal fluid ratios with Aβ42 predict preclinical brain β-amyloid
doi:10.1192/bjp.140.6.566 accumulation. Alzheimers Dement (Amst). 2016;2:27-38. doi:10.1016/
86. Chaves ML, Camozzato AL, Godinho C, Kochhann R, Schuh A, j.dadm.2015.11.006
Almeida VL, et al. Validity of the clinical dementia rating scale for the 107. Hansson O, Lehmann S, Otto M, Zetterberg H, Lewczuk P. Advantages
detection and staging of dementia in Brazilian patients. Alzheimer Dis and disadvantages of the use of the CSF Amyloid β (Aβ) 42/40 ratio in
Assoc Disord. 2007;21(3):210-7. doi:10.1097/WAD.0b013e31811ff2b4 the diagnosis of Alzheimer’s disease. Alzheimers Res Ther. 2019;11(1):34.
87. O’Bryant SE, Waring SC, Cullum CM, Hall J, Lacritz L, Massman PJ, et al. doi:10.1186/s13195-019-0485-0
Staging dementia using Clinical Dementia Rating Scale Sum of Boxes 108. Silverman DH, Small GW, Phelps ME. Clinical value of neuroimaging in the
scores: a Texas Alzheimer’s research consortium study. Arch Neurol. diagnosis of dementia. Sensitivity and specificity of regional cerebral metabolic
2008;65(8):1091-5. doi:10.1001/archneur.65.8.1091 and other parameters for early identification of Alzheimer’s disease. Clin Positron
88. Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom J, Imaging. 1999;2(3):119-30. doi:10.1016/s1095-0397(99)00020-5
Relkin N, et al. Practice parameter: diagnosis of dementia (an evidence- 109. Silverman DH, Small GW, Chang CY, Lu CS, De Aburto MAK, Chen W,
based review). Report of the Quality Standards Subcommittee of the et al. Positron emission tomography in evaluation of dementia: regional
American Academy of Neurology. Neurology. 2001;56(9):1143-53. brain metabolism and long-term outcome. JAMA. 2001;286(17):2120-7.
doi:10.1212/wnl.56.9.1143 doi:10.1001/jama.286.17.2120
89. Nitrini R, Caramelli P, Bottino CM, Damasceno BP, Brucki SM, Anghinah R, et al. 110. Mosconi L. Brain glucose metabolism in the early and specific diagnosis
Diagnosis of Alzheimer’s disease in Brazil: cognitive and functional evaluation. of Alzheimer’s disease. FDG-PET studies in MCI and AD. Eur J Nucl Med
Recommendations of the Scientific Department of Cognitive Neurology Mol Imaging. 2005;32(4):486-510. doi:10.1007/s00259-005-1762-7
and Aging of the Brazilian Academy of Neurology. Arq Neuropsiquiatr. 111. Schöll M, Damián A, Engler H. Fluorodeoxyglucose PET in neurology and
2005;63(3A):720-7. doi:10.1590/s0004-282x2005000400034 psychiatry. PET Clin. 2014;9(4):371-90. doi:10.1016/j.cpet.2014.07.005
90. van der Flier WM, Scheltens P. Epidemiology and risk factors of dementia. 112. Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP,
J Neurol Neurosurg Psychiatry. 2005;76(Suppl 5):v2-7. doi:10.1136/ et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh
jnnp.2005.082867 Compound-B. Ann Neurol. 2004;55(3):306-19. doi:10.1002/ana.20009
91. Hort J, O’Brien JT, Gainotti G, Pirttila T, Popescu BO, Rektorova I, 113. Schilling LP, Zimmer ER, Shin M, Leuzy A, Pascoal TA, Benedet AL, et al.
et al. EFNS guidelines for the diagnosis and management of Alzheimer’s Imaging Alzheimer’s disease pathophysiology with PET. Dement Neuropsychol.
disease. Eur J Neurol. 2010;17(10):1236-48. doi:10.1111/ 2016;10(2):79-90. doi:10.1590/S1980-5764-2016DN1002003
114. Fleisher AS, Pontecorvo MJ, Devous MD, Lu M, Arora AK, Truocchio SP,
j.1468-1331.2010.03040.x
et al. Positron emission tomography imaging with [18F]flortaucipir and
92. Caramelli P, Teixeira AL, Buchpiguel CA, Lee HW, Livramento JA,
postmortem assessment of Alzheimer disease neuropathologic changes.
Fernandez LL, et al. Diagnosis of Alzheimer’s disease in Brazil:
JAMA Neurol. 2020;77(7):829-39. doi:10.1001/jamaneurol.2020.0528
supplementary exams. Dement Neuropsychol. 2011;5(3):167-77.
115. Cho H, Choi JY, Hwang MS, Lee JH, Kim YJ, Lee HM, et al. Tau PET
doi:10.1590/S1980-57642011DN05030004
in Alzheimer’s disease and mild cognitive impairment. Neurology.
93. Sintini I, Graff-Radford J, Senjem ML, Schwarz CG, Machulda MM, Martin PR,
2016;87(4):375-83. doi:10.1212/WNL.0000000000002892
et al. Longitudinal neuroimaging biomarkers differ across Alzheimer’s disease
116. Counts SE, Ikonomovic MD, Mercado N, Vega IE, Mufson EJ. Biomarkers
phenotypes. Brain. 2020;143(7):2281-94. doi:10.1093/brain/awaa155 for the early detection and progression of Alzheimer’s disease.
94. Barkhof F, Fox NC, Bastos-Leite AJ, Scheltens P. Neuroimaging in Neurotherapeutics. 2017;14(1):35-53. doi:10.1007/s13311-016-0481-z
dementia. Berlin: Springer; 2011. 117. Matsuda H, Shigemoto Y, Sato N. Neuroimaging of Alzheimer’s disease:
95. Amato ACS Filho, Balthazar MLF. Neuroimagem nas demências: como focus on amyloid and tau PET. Jpn J Radiol. 2019;37(11):735-49.
ela pode nos ajudar? In: Neurologia cognitiva e do envelhecimento: doi:10.1007/s11604-019-00867-7
do conhecimento básico à abordagem clínica. São Paulo: Omnifarma; 2016. 118. Ossenkoppele R, Schonhaut DR, Schöll M, Lockhart SN, Ayakta N,
96. Wattjes MP, Henneman WJ, van der Flier WM, de Vries O, Baker SL, et al. Tau PET patterns mirror clinical and neuroanatomical
Träber F, Geurts JJ, et al. Diagnostic imaging of patients in a memory variability in Alzheimer’s disease. Brain. 2016;139(5):1551-67. doi:10.1093/
clinic: comparison of MR imaging and 64-detector row CT. Radiology. brain/aww027
2009;253(1):174-83. doi:10.1148/radiol.2531082262 119. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s
97. Frisoni GB, Fox NC, Jack CR Jr, Scheltens P, Thompson PM. disease drug development pipeline: 2019. Alzheimers Dement (N Y).
The clinical use of structural MRI in Alzheimer’s disease. Nat Rev Neurol. 2019;5(1):272-93. doi:10.1016/j.trci.2019.05.008
2010;6(2):67-77. doi:10.1038/nrneurol.2009.215 120. Jack CR, Jr, Bennett DA, Blennow K, Carrillo MC, Feldman HH, Frisoni GB,
98. Leocadi M, Canu E, Calderaro D, Corbetta D, Filippi M, Agosta F. et al. A/T/N: An unbiased descriptive classification scheme for Alzheimer
An update on magnetic resonance imaging markers in AD. Ther Adv disease biomarkers. Neurology. 2016;87(5):539-47. doi:10.1212/
Neurol Disord. 2020;13:1-17. doi:10.1177/1756286420947986 WNL.0000000000002923

34   Diagnostic Criteria for AD in Brazil.   Schilling LP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):21-35

121. Blennow K. A review of fluid biomarkers for Alzheimer’s disease: moving in preclinical Alzheimer’s disease. Alzheimers Dement. 2014;10(6):844-52.
from CSF to blood. Neurol Ther. 2017;6(Suppl 1):15-24. doi:10.1007/ doi:10.1016/j.jalz.2014.01.001
s40120-017-0073-9 131. Bondi MW, Edmonds EC, Salmon DP. Alzheimer’s disease: past, present,
122. Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, and future. J Int Neuropsychol Soc. 2017;23(9-10):818-31. doi:10.1017/
et al. NIA-AA research framework: toward a biological definition of S135561771700100X
Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535-62. doi:10.1016/ 132. Henriksen K, O’Bryant SE, Hampel H, Trojanowski JQ, Montine TJ,
j.jalz.2018.02.018 Jeromin A, et al. The future of blood-based biomarkers for Alzheimer’s disease.
123. Hampel H, Frank R, Broich K, Teipel SJ, Katz RG, Hardy J, et al. Biomarkers Alzheimers Dement. 2014;10(1):115-31. doi:10.1016/j.jalz.2013.01.013
for Alzheimer’s disease: academic, industry and regulatory perspectives. 133. Bateman RJ, Blennow K, Doody R, Hendrix S, Lovestone S, Salloway S,
Nat Rev Drug Discov. 2010;9(7):560-74. doi:10.1038/nrd3115 et al. Plasma biomarkers of AD emerging as essential tools for drug
124. Wang L, Benzinger TL, Su Y, Christensen J, Friedrichsen K, Aldea P, development: an EU/US CTAD task force report. J Prev Alzheimers Dis.
et al. Evaluation of tau imaging in staging Alzheimer disease and revealing 2019;6(3):169-73. doi:10.14283/jpad.2019.21
interactions between β-Amyloid and tauopathy. JAMA Neurol. 134. Ovod V, Ramsey KN, Mawuenyega KG, Bollinger JG, Hicks T, Schneider T,
2016;73(9):1070-7. doi:10.1001/jamaneurol.2016.2078 et al. Amyloid β concentrations and stable isotope labeling kinetics of
125. Price JL, McKeel DW Jr, Buckles VD, Roe CM, Xiong C, Grundman M, human plasma specific to central nervous system amyloidosis. Alzheimers
et al. Neuropathology of nondemented aging: presumptive evidence for Dement. 2017;13(8):841-9. doi:10.1016/j.jalz.2017.06.2266
preclinical Alzheimer’s disease. Neurobiol Aging. 2009;30(7):1026-36. 135. Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J,
doi:10.1016/j.neurobiolaging.2009.04.002 et al. Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther.
126. Josefsson M, Luna X, Pudas S, Nilsson LG, Nyberg L. Genetic and lifestyle 2013;5(2):9. doi:10.1186/alzrt163
predictors of 15-year longitudinal change in episodic memory. J Am Geriatr 136. Mattsson N, Andreasson U, Zetterberg H, Blennow K, Alzheimer’s Disease
Soc. 2012;60(12):2308-12. doi:10.1111/jgs.12000 Neuroimaging Initiative. Association of plasma neurofilament light with
127. Bateman RJ, Xiong C, Benzinger TLS, Fagan AM, Goate A, Fox NC, et al. neurodegeneration in patients with Alzheimer disease. JAMA Neurol.
Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. 2017;74(5):557-66. doi:10.1001/jamaneurol.2016.6117
N Engl J Med. 2012;367(9):795-804. doi:10.1056/NEJMoa1202753 137. Zhao Y, Xin Y, Meng S, He Z, Hu W. Neurofilament light chain protein
128. Mormino EC, Sperling RA, Holmes AJ, Buckner RL, De Jager PL, in neurodegenerative dementia: a systematic review and network
Smoller JW, et al. Polygenic risk of Alzheimer’s disease is associated with meta-analysis. Neurosci Biobehav Rev. 2019;102:123-38. doi:10.1016/
early- and late-life processes. Neurology. 2016;87(5):481-8. doi:10.1212/ j.neubiorev.2019.04.014
WNL.0000000000002922 138. Siedlecki-Wullich D, Català-Solsona J, Fábregas C, Hernández I,
129. Jack CR Jr, Albert MS, Knopman DS, McKhann GM, Sperling RA, Clarimon J, Lleó A, et al. Altered microRNAs related to synaptic function
Carrillo MC, et al. Introduction to the recommendations from the National as potential plasma biomarkers for Alzheimer’s disease. Alzheimers Res
Institute on Aging-Alzheimer’s Association workgroups on diagnostic Ther. 2019;11(1):46. doi:10.1186/s13195-019-0501-4
guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257-62. 139. Bram JMF, Talib LL, Joaquim HPG, Sarno TA, Gattaz WF, Forlenza OV.
doi:10.1016/j.jalz.2011.03.004 Protein levels of ADAM10, BACE1, and PSEN1 in platelets and leukocytes
130. Jessen F, Amariglio RE, van Boxtel M, Breteler M, Ceccaldi M, Chételat G, of Alzheimer’s disease patients. Eur Arch Psychiatry Clin Neurosci.
et al. A conceptual framework for research on subjective cognitive decline 2019;269(8):963-72. doi:10.1007/s00406-018-0905-3

Schilling LP, et al.   Diagnostic Criteria for AD in Brazil.   35


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S103EN

Diagnosis of frontotemporal dementia:


recommendations of the Scientific Department
of Cognitive Neurology and Aging of the
Brazilian Academy of Neurology
Leonardo Cruz de Souza1,2 , Mirna Lie Hosogi3 , Thais Helena Machado2,4 ,
Maria Teresa Carthery-Goulart3,5 , Mônica Sanches Yassuda3,6 , Jerusa Smid3 ,
Breno José Alencar Pires Barbosa3,7,8 , Lucas Porcello Schilling9,10,11 ,
Marcio Luiz Figueredo Balthazar12 , Norberto Anízio Ferreira Frota13,14 ,
Francisco Assis Carvalho Vale15 , Paulo Caramelli1,2 , Paulo Henrique Ferreira Bertolucci16 ,
Márcia Lorena Fagundes Chaves17,18 , Sonia Maria Dozzi Brucki3 , Ricardo Nitrini3 ,
Valéria Santoro Bahia19 , Leonel Tadao Takada3

Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
1

2
Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Clínica Médica, Grupo de Pesquisa em Neurologia Cognitiva e do Comportamento,
Belo Horizonte MG, Brazil.
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
3

Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Fonoaudiologia, Belo Horizonte MG, Brazil.
4

5
Universidade Federal do ABC, Grupo de Estudos em Neurociência da Linguagem e Cognição, Programa de Pós-Graduação em Neurociência e Cognição,
Centro de Matemática, Computação e Cognição, Santo André SP, Brazil.
Universidade de São Paulo, Gerontologia, Ciências e Humanidades, Escola de Artes, São Paulo SP, Brazil.
6

Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
7

Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.


8

Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
9

10
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
11
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
12
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil.
13
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
14
Universidade de Fortaleza, Fortaleza CE, Brazil.
15
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brazil.
16
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil.
17
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
18
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brazil.
19
Universidade Cidade de São Paulo, São Paulo SP, Brazil.
Correspondence: Leonardo Cruz de Souza; Email: leocruzsouza@ufmg.br.
Conflict of interest: LCS: Participation in advisory board for Biogen. Participation as speaker in symposia sponsored by Biogen; JS: Participation as speaker in symposia
sponsored by Roche laboratory; LPS: Participation in advisory boards for Biogen. Participation as speaker in symposia sponsored by Aché, Apsen and Biogen laboratories;
MLFB: Participation in advisory boards for Biogen. Development of material for continuous medical education and participation as speaker in symposia sponsored by EMS
and Torrent laboratories; PC: Participation as principal investigator in clinical trials for Novo Nordisk and Roche laboratories. Participation in advisory boards for Aché, Biogen,
EMS, Nutricia and Roche laboratories. Development of material for continuous medical education and participation as speaker in symposia sponsored by Aché, Nutricia,
Libbs, Roche, Sandoz, Torrent and Zodiac laboratories; PHFB: Participation in advisory boards for Biogen and Novo Nordisk laboratories. Supervision of training activities for
Biogen, Janssen-Cilag and Novo Nordisk laboratories and for Quintiles. Participation as speaker in symposia sponsored by Apsen, Nutricia, Roche and Sandoz laboratories;
RN: Participation in advisory board for Biogen; MLH, THM, MTCG, MSY, BJAPB, NAFF, FACV, MLFC, SMDB, VSB, LTT: There is no conflict of interest to declare.
Received on August 10, 2021; Received in its final form on December 08, 2021; Accepted on April 27, 2022.

36   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

ABSTRACT. “Frontotemporal dementia” (FTD) is a clinical syndrome characterized by the focal involvement of the frontal and/or temporal lobes. FTD has
three clinical phenotypes: the behavioral variant and two linguistic subtypes, namely, non-fluent/agrammatic primary progressive aphasia (PPA-NF/A) and
semantic PPA (PPA-S). FTD is the second most common cause of dementia in individuals under the age of 65 years. This article presents recommendations
for the diagnosis of FTD in the Brazilian scenario, considering the three levels of complexity of the health system: primary health care, secondary and
tertiary levels. Diagnostic guidelines are proposed, including cognitive testing, behavioral and language assessments, laboratory tests, and neuroimaging.
Keywords: Frontotemporal Dementia; Aphasia, Primary Progressive.

DIAGNÓSTICO DA DEMÊNCIA FRONTOTEMPORAL: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO


DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. A “demência frontotemporal” (DFT) é uma síndrome clínica, cujo denominador comum é o acometimento focal dos lobos frontais e/ou temporais.
A DFT tem três fenótipos clínicos distintos: a variante comportamental e dois subtipos linguísticos, a saber, a afasia progressiva primária não-fluente/
agramática (APP-NF/A) e a afasia progressiva primária semântica (APP-S). A DFT é a segunda causa mais comum de demência em indivíduos com idade
inferior a 65 anos, após a doença de Alzheimer. O presente artigo apresenta recomendações para diagnóstico da DFT no cenário brasileiro, considerando
os três níveis de complexidade do sistema de saúde: atenção primária à saúde e níveis secundários. São propostos protocolos de investigação diagnóstica
abrangendo testagem cognitiva, avaliação comportamental, avaliação fonoaudiológica, exames laboratoriais e de neuroimagem.
Palavras-chave: Demência Frontotemporal; Afasia Progressiva Primária.

INTRODUCTION Such indices make FTD the second most common

“F rontotemporal dementia” (FTD) is defined as


a clinical syndrome whose common denominator
is the focal involvement of the frontal and/or temporal
cause of presenile dementia, after Alzheimer’s
disease (AD). Indeed, the majority of FTD cases are of
young onset, but it is now recognized that up to 30%
lobes. FTD has three distinct clinical phenotypes: of cases start after 65 years old2.
the behavioral variant, and two linguistic subtypes, This article presents recommendations for the
namely, non-fluent/agrammatic primary progressive diagnosis of FTD in the Brazilian scenario, considering
aphasia (PPA-NF/A) and semantic PPA (PPA-S). Recently, the three complexity levels of the health system: primary
the existence of a fourth subtype, the so-called right health care, secondary and tertiary levels. Diagnostic
temporal variant, has been considered. The behavioral guidelines are proposed, covering cognitive testing,
variant (bvFTD) is the most common phenotypic behavioral and language assessments, laboratory tests,
presentation of FTD. and neuroimaging. The right temporal variant, whose
In turn, “frontotemporal lobar degeneration (FTLD)” clinical definition is still in debate5 and is not part of
refers to the histopathological substrate of FTD. the latest diagnostic consensus6,7, is not addressed
The types FTLD-Tau and FTLD-“transactive response in this article.
DNA-binding protein with Mr 43 kDa” (TDP-43) are
the most common ones 1. Other subtypes, such as
DIAGNOSIS
FET family proteins, FUS (fused in sarcoma) and EWS
The diagnostic criteria for FTD, both for the behavioral6
(Ewing’s sarcoma protein), are less frequent.
and the language variants 7 , were established
Thus, “FTD” should be used for clinical descriptions by international expert consensus.
and phenotypes, whereas “FTLD” should be used The diagnosis of bvFTD is based on the identification
to describe the histopathological classification and of progressive cognitive-behavioral decline6. The current
not to refer to the clinical syndrome2. criteria determine three levels of diagnostic reliability
International studies indicate that the prevalence (Table 1): (I) possible diagnosis, for patients presenting
and incidence of FTD compose 15-22 cases/100,000 characteristic cognitive-behavioral alterations, but have
and 1.2-4.1 cases/100,000 population, respectively, neither typical neuroimaging alterations nor manifest
being higher in the 45-64 age group 3,4. There are functional decline; (II) probable diagnosis, for patients
no epidemiological studies that have specifically who, in addition to characteristic cognitive-behavioral
investigated the prevalence of F TD in Brazil, manifestations, have impaired autonomy and evidence
but epidemiological sur veys on dementias of frontotemporal involvement on structural or
indicate a prevalence of 0.18% among individuals functional neuroimaging; and (III) definitive diagnosis,
over 65 years old3. when histopathological changes are observed on brain

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   37


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

biopsy or on post-mortem examination, or patients with of the language network in the language-dominant
evidence of pathogenic mutation. temporal and frontal lobes7,8. In most cases, there is
According to the same consensus6, for possible predominant involvement of the left hemisphere,
or probable diagnosis, the patient must meet at least with rare exceptions (crossed aphasia and/or conditions
three of the following criteria: (I) early disinhibition; that start with neurodegeneration on the right
(II) apathy or early inertia; (III) early loss of empathy/ hemisphere and with initial symptoms of prosopagnosia
sympathy; (IV) perseverative, stereotyped, or early and/or visual agnosia)9. Language disorders involve one
compulsive/ritualistic behavior; (V) hyperorality and or more levels of linguistic processing (phonological,
dietary changes; and (VI) neuropsychological profile semantic, syntactic) and are associated with cognitive
with executive dysfunction and relative preservation (i.e., speech apraxia) and/or motor (dysarthria)
of episodic memor y and visuospatial abilities. speech alterations. In addition, language impairment
The symptom is early if it occurs within the first three impacts communication skills; thus, functional
years after the onset of symptoms. deficits vary depending on the language demands
related to professional occupation and daily activities,
and to the cognitive resources and strategies that
Table 1. Diagnostic Criteria for Frontotemporal Dementia patients use to compensate for the deficits, as well.
(behavioral variant) – adapted from Rascovsky et al. (2011). Environmental factors can also mitigate or enhance
the functional impairment10.
Frontotemporal dementia: behavioral variant Gorno-Tempini et al. (2011) suggested unifying the
Progressive deterioration in behavior and/or cognition, evidenced
nomenclature and defined criteria for the syndromic
by observation or clinical history (requires an informant).
diagnosis of PPA and its three main variants,
Possible diagnosis in terms of clinical manifestations, neuroanatomical
Must meet at least 3 of the 6 criteria listed below: and neuropatholo g ical cor relates. PPA -NF/A
(I) behavioral disinhibition (socially inappropriate behavior; (non-fluent/agrammatic) and PPA-S (semantic) are
impulsiveness; loss of social rules or decorum); part of the clinical syndrome of FTD. On the contrary,
(II) apathy or inertia; the logopenic subtype of PPA (PPA-L) is considered
(III) loss of empathy/sympathy (decreased affective resonance
an atypical presentation of AD. It should be noted,
to the needs and feelings of others; diminished social interest,
reduced interpersonal “heat”); however, that about 1/3 of the patients have mixed
(IV) perseverative, stereotyped, or compulsive/ritualistic behaviors; conditions or do not fit the criteria for these variants11,12.
(V) hyperorality and dietary changes (alteration of food Thus, although the diagnosis of PPA can be accurately
preference, increased consumption of alcohol or cigarettes, distinguished from bvFTD and/or other dementia
oral exploration of objects); conditions, classification into variants requires speech
(VI) neuropsychological profile with executive deficits and relative and language examination by experts. In the present
preservation of episodic memory and visuospatial functions.
consensus, we have chosen the terms PPA-S, PPA-NF/A
Probable diagnosis and PPA-L. Other terminologies are also used, including:
Must meet criteria for possible FTD and have: “semantic variant of PPA,” “non-fluent variant of PPA,”
(I) Significant functional decline, demonstrated and “logopenic variant of PPA.”
in specific inventories; and According to Gorno-Tempini et al. (2011) 7 ,
(II) Evidence of typical FTD alterations on neuroimaging the clinical diagnosis of PPA requires fulfilling three
examination (atrophy or hypometabolism or hypoperfusion criteria: (1) the most prominent clinical feature is
in frontotemporal regions).
language difficulty; (2) language difficulties are the
Definite diagnosis main cause of functional impairment (difficulty
Must meet criteria for possible or probable FTD and have: in communication); and (3) aphasia must be the
(I) Pathological confirmation of frontotemporal lobe degeneration; or most prominent deficit at the beginning of the
(II) Evidence of causative genetic mutation. condition. Furthermore, the pattern of deficits must
not be explained by another neurological or psychiatric
disorder, and patients must not initially present
significant behavioral disturbances or other cognitive
PPA, in turn, is a clinical syndrome characterized impairments. However, deficits in other cognitive
by a language disorder of insidious onset and functions appear in the neuropsychological assessment,
progressive course, which affects the functioning especially in cognitive skills that share neuroanatomical

38   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

correlates with the language network (such as verbal diagnostic levels: one supported by neuroimaging exams
immediate memory; numerical and calculation skills; and another supported by histopathological findings.
ideomotor praxis). However, these deficits should be Next, we propose ways to carry out the diagnostic
milder compared to the language deficit. In addition investigation of FTD (all subtypes) at different levels
to the clinical level, the consensus predicts two other of health care (Figures 1 and 2).

Behavioral or Language Disorder


General practioner
Primary care:

Clinical Interview
Mini-Mental State Exam
Laboratory screening

Brain CT

Detailed Clinical Interview


Brief Cognitive Battery or
Addenbrooke Cognitive Assessment
Specialist (neurologist)

Frontal Assessment Battery or INECO Frontal Screening


Secondary care:

Neuropsychiatric Inventory

Brain MRI If normal


Psychiatric consultation
Lumbar puncture if age < 65 years-old

Behavioral Disorder Language Disorder


Expand cognitive assessment Language Assessment
(RAVLT; Rey Complex Figure; Hayling Test; Mini-SEA; FRS) with Speech therapist
Multidisciplinary team
Tertiary care:

Consider:
FDG-PET
Genetic Investigation
Biomarkers for Alzheimer’s disease

Figure 1. Diagnostic procedures for frontotemporal dementia

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   39


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

PRIMARY HEALTH CARE: SUSPICION OF PPA

Patient with language or speech complaint Patient with memory complaint


(with no functional decline).
Examples: word-finding difficulties, effortful
speech, comprehension disorders
(oral or written language). Complaint of memory Other memory Complaint about
for words complaints memory, language or
other cognitive domain
with functional decline.
Sudden onset Insidious onset Slowly progressive disorder SUSPICION OF PPA

Laboratory screening CT scan

SECUNDARY LEVEL: SYNDROMIC CONFIRMATION

CONFIRMATION OF THE
Neurological evaluation HYPOTHESIS OF PPA
DIAGNOSIS OF PPA

TERTIARY LEVEL: CLASSIFICATION OF PPA

Investigation:
Avaliação Especializada: Neurological follow up
– Biomarkers
– Speech therapy Pharmacological treatment
– Genetics
– Neuropsychology Non-pharmacological treatment
– Functional neuroimaging

Figure 2. Procedures for the diagnosis of Primary Progressive Aphasia

Primary health care a brief neurological examination, looking for focal deficits
that suggest stroke or expansive intracranial lesions.
Clinical assessment (bvFTD and PPA) Regarding progressive language disorders, patients
In primary health care, the patient will be initially and family members seek medical care due to complaints
evaluated by a general practitioner, who must carry out of memory or specific language and/or speech problems
a careful anamnesis in order to identify a condition of (Figure 2). When faced with the memory complaint,
cognitive-behavioral decline or progressive language it is important to differentiate which memory problem
disorder and then refer the patient to the appropriate the patient and family refer to. Often, patients having
workup. Therefore, it is important to describe how difficulty finding words and forgetting the names of
symptoms appear. Due to its degenerative nature, people and objects (essentially linguistic alterations)
the onset of FTD symptoms is usually insidious and report that they have memory failures, but these are
progressive, unlike vascular conditions, in which the not episodic memory alterations (common in AD),
“stepwise deterioration” of symptoms is more common. but rather word memory (lexical access difficulties and
Considering the clinical suspicion of cognitive- anomie), suggesting language impairment. In some
behavioral decline, the physician must actively inquire cases, it is possible that the memory complaint is related
about psychiatric (past admission to psychiatric to the gradual loss of knowledge of the meaning of
institutions) and infectious (syphilis, HIV) antecedents words and concepts (for example: having doubts about
and about the use of psychotropic drugs that may what the words mean, which is a symptom of impaired
interfere with cognitive-behavioral functioning. verbal semantic memory).
Mood symptoms and maniform manifestations should Initial language complaints may be diverse. The most
also be always investigated, as they may indicate frequent are lexical access difficulties and anomia.
a psychiatric cause for the patient’s clinical condition. However, there are also reports of exchange of words,
On physical examination, the physician should perform exchange of phonemes, stuttering, slowing down

40   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

of speech production, language comprehension the presence of dementia, which should be confirmed
difficulties, failures in reading or writing and so forth. in further evaluations in secondary care.
It is important to investigate whether the patient and/or Social cognition involves processing information
informant (e.g., family member) notices an insidious that is relevant to social interaction. In bvFTD, social
worsening of language in relation to premorbid cognition may be altered, as the patient may fail
performance. There is great heterogeneity of language to understand what others are thinking or feeling.
in the population, depending on education, reading and The clinician should ask the caregiver whether the
writing habits, and occupation. It may be tough to detect patient understands the problems and concerns of
difficulties in individuals who, throughout life, had poor those around them (cognitive empathy) and whether
language skills (for example, writing errors, low reading they care, suffer, or rejoice in what happens to
fluency, impoverished vocabulary). Therefore, in some others (emotional empathy). Questions can be asked
cases, it may be necessary to conduct a longitudinal about their ability to socialize and changes in their
follow-up to identify the progression of the condition. moral rules. From this perspective, primary care
Clinical history is fundamental for the diagnosis of PPA. physicians should inquire caregivers and observe
Speech-language disorders, in the initial phase, do not patients regarding typical behavioral changes of FTD,
significantly impact functional activities, and some in addition to other neuropsychiatric manifestations,
patients even keep their work activities. such as delusions and hallucinations, which may
suggest primary psychiatric disorders.
Cognitive and behavioral assessments The performance of patients with PPA on the MMSE
The assessment of cognitive performance represents an will depend on the intensity of the aphasia, as it is
important component of the diagnostic procedures of a test that is highly dependent on language. MMSE
FTD. Cognitive changes in bvFTD can be heterogeneous, results may overestimate cognitive decline, as cognitive
functions such as temporal orientation and memory are
but the diagnostic criteria of Rascovsky et al. (2011)
assessed by language. On the other hand, the language
suggest that the neuropsychological profile should
deficit can be underestimated, since the MMSE linguistic
include executive dysfunction, relative preservation
tasks have low complexity to assess the production and
of episodic memory and of visuospatial functions.
comprehension of words and sentences. Thus, initial
However, in the early stages of the disease, especially
cases of PPA can obtain scores within the expected
among people with a high level of education, executive
range for their schooling.
dysfunction may not be evident 13 . Additionally,
other diseases (including AD) can lead to executive
Laboratory investigation
dysfunction 14. Another challenging factor is that Metabolic and/or infectious disorders (renal or liver
patients with bvFTD can also present episodic memory failure, hypothyroidism, neurosyphilis and HIV
deficit, in a pattern similar to that observed in AD15,16. infection, and so on) that cause neuropsychiatric
Even so, documenting the cognitive profile suggested manifestations can be ruled out through blood tests.
in the international criteria 6 is important for the Thus, all suspected cases of FTD (regardless of the
diagnosis of bvFTD. According to this perspective, phenotypic presentation) should undergo laboratory
identifying a reduction in global cognitive efficiency investigation to screen for reversible causes of cognitive-
and executive dysfunction of magnitude greater than behavioral decline 20 : blood count, vitamin B12,
episodic memory deficits may suggest bvFTD. folic acid, liver, kidney and thyroid functions, protein
In primary care, complaints from patients and their electrophoresis, and anti-HIV serology.
families suggesting the presence of cognitive deficits
should be carefully analyzed, especially those indicating Neuroimaging investigation
difficulties in planning and organizing activities. In primary health care, CT scan can be a useful procedure
The Mini-Mental State Examination (MMSE)17,18 should as an initial propaedeutic. This exam evaluates the
be applied for the global assessment of cognition, presence of frontotemporal lesions (e.g., tumors,
and the score should be interpreted in relation to the hematomas), ventricular dilatation or cerebrovascular
individual’s educational level. Patients with bvFTD lesions that are associated with behavioral symptoms.
are likely to lose points in mental calculation and Computed tomography in bvFTD usually shows
the command sub-items (due to attention deficit); asymmetric increase in sulci and fissures in the frontal
orientation in time and space are usually preserved19, and/or temporal lobes. These findings, however,
as the drawing. Impairment in the MMSE may suggest can only be observed when the disease is at a more

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   41


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

advanced stage. In the linguistic variants, frontoinsular domains can be especially relevant for the diagnosis
atrophy in the dominant hemisphere can be observed, of bvFTD and PPA, respectively.
in the case of the PPA-NF/A variant, and atrophy The Frontal Assessment Battery (FAB)26,27 and the
of temporal poles, in the case of the PPA-S. INECO Frontal Screening (IFS)28 can identify executive
dysfunction and help detect patients with bvFTD.
Secondary level However, these instruments may fail in the differential
diagnosis of dementia subtypes29.
Clinical assessment Regarding social cognition, the physician should ask
At the secondary level, the patient will be evaluated the caregiver if the patient understands the problems
by a neurologist, who must carry out a thorough and concerns of the people around them (cognitive
neurological examination, preceded by a detailed empathy) and if they care, suffer, or are happy with what
anamnesis, which aims to recapitulate the main happens to others (emotional empathy). Questions
elements of clinical and family history. The neurologist about their social skills and changes in their moral
should actively look for signs of parkinsonian rules can also be asked.
syndrome, in addition to changes in oculomotricity The short version of the Neuropsychiatric Inventory
(conjoined down-gaze palsy), which may evoke (NPI)30,31, the NPI-Q32, can be used in the investigation
progressive supranuclear palsy. Similarly, signs of neuropsychiatric symptoms. The NPI-Q can detect
of asymmetric muscle atrophy, fasciculations and the behavioral symptoms of bvFTD. The NPI-Q has been
pyramidal signs (Babinski and Hofmann signs) should validated for the Brazilian population33. The Frontal
be looked for to identify signs of motor neuron disease. Behavioral Inventory can also be used34.
The neurologist should also look for the presence
of primitive signs (grasping, glabellar, snouting) Laboratory investigation
that suggest severe frontal involvement. At the secondary level, the physician must review
all laboratory tests to screen for reversible causes
Cognitive and behavioral assessments of dementia. For pre-senile patients, it may be necessary
At this level of care, the assessment of cognition must to include tests of autoimmune diseases, such as
be comprehensive and evaluate the main cognitive vasculitis and systemic lupus erythematosus, depending
domains. The application of the Brief Cognitive on the patient’s clinical context.
Screening Batter y (BBRC) is recommended 21,22 . For patients suffering from the condition
The BBRC includes the Figure Memory Test, which before turning 65 years old, or for those with a
requires naming and recalling ten figures to assess rapidly progressive decline, lumbar puncture is
episodic memory. In this battery, executive functions mandatory, to rule out inflammatory and/or infectious
are investigated with the Animal Verbal Fluency Test causes of dementia.
and the Clock Drawing Test, which are followed by the
delayed recall of the ten figures previously presented Neuroimaging investigation
(after 5 minutes). Special attention should be paid to In secondary care, magnetic resonance imaging (MRI) of
the delayed recall (cut-off score ≤ 5 pictures), as it is the brain should be performed. On MR images, an increase
a marker of episodic memory impairment. The BBRC in sulci and fissures can be observed, with frontotemporal
can be used in populations with different educational predominance (Figure 3). bvFTD is characterized by early
backgrounds23. Slowness in naming, lack of responses atrophy of frontotemporal regions, affecting the anterior
and/or phonological/semantic errors may indicate cingulate and orbitofrontal cortex35,36. Hippocampus
difficulties in understanding or expressing the language. may be atrophied in bvFTD to a similar degree to that
Episodic memory is preserved in the early stages of PPA. seen in AD37. Some atrophic patterns may suggest
Patients usually perform well in the recognition task, FTD associated with genetic mutations: pronounced
as it does not require oral recall. bitemporal atrophy occurs in patients with a MAPT
The Addenbrooke Cognitive Examination – mutation; markedly asymmetric frontotemporal atrophy is
Revised (ACE-R)24,25 is also an excellent tool for the common in patients with a progranulin (GRN) mutation38.
global assessment of cognition. The ACE-R includes Extensive white matter lesions can also occur in patients
the MMSE. Additionally, it provides individualized with progranulin mutation39. As disease progresses,
scores for attention and orientation, episodic atrophy gets more pronounced in the frontotemporal
memory, verbal fluency, language, and visual-spatial lobes, with relative preservation of the posterior regions.
skills. The scores for the verbal fluency and language In PPA-NF/A, atrophy of the inferior frontal gyrus

42   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

of the dominant hemisphere is detected, whereas in MRI also allows the identification of hypersignal on
PPA-S there is typically focal atrophy of the temporal pole, T2-weighted and FLAIR sequences, which could suggest
uni- or bilaterally. vascular dementia or leukoencephalopathy.

Figure 3. A. Focal atrophy of frontotemporal regions in the behavioral variant of Frontotemporal Dementia; B. Atrophy of the inferior frontal gyrus of the dominant
hemisphere in primary non-fluent/agrammatic progressive aphasia (PPA-NF/A); C. Focal temporal pole atrophy in semantic primary progressive aphasia (PPA-S).

Tertiary level geometric figure, which is later drawn using


visual memory. The copy is used to assess visual-
Clinical Evaluation constructive skills and the subsequent recall of
At the tertiary level of health care, the patient will be the figure is a measure of visual episodic memory;
assisted by a multidisciplinary team. The recommended • Attention and Executive Functions – Trail
clinical assessment consists of anamnesis, clarifying Making Test A and B assess visual attention and
previously obscure points and confirming previous data. shifting, respectively; Wechsler Adult Intelligence
The clinical-neurological examination must follow the Scale (WAIS-III) Forward and Backward Digit
guidelines described for the secondary health level. Span Test assess auditory attention and working
memory, respectively; Wisconsin Card Sorting
Cognitive and behavioral assessments Test assesses working memory and mental
flexibility. These cognitive parameters may be
Behavioral variant (bvFTD) especially important to characterize executive
At this level of care, it may be useful for diagnostic dysfunction, which is part of the formal criteria
purposes to characterize the patient’s neuropsychological of bvFTD6. However, they may not discriminate
profile, with specific and detailed parameters for each between types of dementia43;
domain of cognition. Qualitative observation of the • Inhibitory Control – The Hayling Test assesses
patient’s behavior during the assessment may be especially the individual’s ability to complete sentences
relevant. The presence of unusual behaviors and strategies with words that make sense or with words
during testing, such as impulsiveness, behavioral rigidity, that prevent the logical sense of the sentence.
ritualized or obsessive behaviors, and repetitive speech, The test could help differentiate bvFTD from
can support the diagnosis of bvFTD40. other dementias such as AD, but results are
Next, we suggest instruments composing a minimal inconclusive44. Likewise, the Stroop test may
neuropsychological battery. This proposal can be expanded, not be discriminative43;
according to available time and expert facilities: • Processing Speed – WAIS-III Digit Symbol
• Verbal episodic memory – Rey Auditory-Verbal Substitution test assesses visual attention and
Learning Test41. Scores within the expected range processing speed, which, if altered, can affect
for age and education, or minor impairment, performance in other domains of cognition45;
may be compatible with a diagnosis of bvFTD; • Visuospatial Functions – Rey Complex Figure.
• Visual episodic memory – Rey Complex Figure42, The copy of the figure can be used as a parameter
which requires the individual to copy a complex to assess planning and visuoconstructive skills;

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   43


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

WAIS-III Block Design also assesses visual- Psychiatric Diseases54. There is no validation of this
constr uctive skills and is a timed task ; scale in Brazil, but there is a translation34.
the Visual Object and Space Perception (VOSP) The DAPHNE scale63 is based on the bvFTD diagnostic
test can be used to assess visual-perceptual criteria 6 and on the FBI items. There is only one study
abilities46. According to current criteria, patients to evaluate its application in the differential diagnosis
with bvFTD have unimpaired visuospatial between bvFTD and AD (frontal variant), with good
capabilities6. For example, in the Rey Complex clinical accuracy64. There is no Brazilian validation or
Figure test, patients with bvFTD tend to perform translation of this scale, but it appears to be promising.
well, and errors are usually related to failures in For an accurate differential diagnosis between FTD
organization and planning, while patients with and PPD, a formal evaluation by a psychiatrist with
AD commit visuospatial errors43; experience in degenerative dementias is recommended.
• Language – the Boston Naming Test assesses the
ability to perceive, interpret and name 60 common Linguistic variants (PPA-NF/A and PPA-S)
figures. The importance of using the version The investigation and characterization of language
adapted for Brazil is highlighted47. The WAIS-III and speech impairments should encompass both
Vocabulary test may also be helpful45; spontaneous conversation and the testing of specific
• Social cognition – Although not included language skills, including phonological, lexical/semantic
in current criteria for diagnosing bvFTD, and syntactic levels.
a growing body of evidence suggests that Spontaneous conversation, in addition to the
tests of social cognition may be useful for the elaboration of speech from a picture, provides
differential diagnosis between bvFTD and information on oral fluency and speech motor capacity.
other dementias, such as AD48,49. Studies have Patients with PPA-NF/A often have reduced oral
shown that the Facial Emotion Recognition production; they may manifest articulatory and/or
Test (FERT) and the Faux-Pas Test (which syntactic errors, slow speech rhythm, and difficulty
assesses theory of mind) are useful in the in lexical access. Patients with PPA-S are fluent,
differential diagnosis between bvFTD and AD48,49. with preservation of the syntactic structure, but with
The short version of the Social and Emotional difficulties in lexical access; they also manifest semantic
Assessment (Mini-SEA) 50 – consisting of the and verbal paraphasias, and frequently use generic
FERT and the Faux-Pas test – is an efficient words (“things,” for example), circumlocutions
instrument to differentiate bvFTD from AD, and compensatory gestures7,65.
regardless of executive dysfunction51, apathy52 Variable degree of anomia is a symptom common to
and episodic amnesia53. It also differentiates all PPA subtypes; for this reason, visual confrontation
bvFTD from depressive disorder50, and it is also naming tests should always be used. Not only the final
recommended to distinguish bvFTD from other score, but the types of paraphasias, are important for
primary psychiatric disorders54. Despite the lack the diagnosis of the PPA variant. In PPA-S, the difficulty
of formal validation of the Mini-SEA in Brazil, in naming is accompanied by semantic impairment,
its clinical validity among Brazilian patients has with frequent semantic paraphasias, use of circumlocution
already been demonstrated in research51,52. There and supracategorization (name a dog as an animal,
is a cross-cultural study of FERT with normative for example), while in PPA-NF/A the deficit comes from
data for the Brazilian population55 and a cross- failure to access the lexicon. In PPA-S, it is possible
cultural adaptation of the Faux-Pas test, with the that the patient does not identify the visual stimulus.
assessment of its psychometric properties56; So, in addition to paraphasias, there are answers
• The Frontotemporal Dementia Rating Scale such as “what is this?”, “I don’t understand this one.”
(FRS)57 is a useful scale for staging the disease. In PPA-S, per for mance on specif ic tests of
It has been validated for use in Brazil58. word comprehension is necessar i ly impa ired,
Concerning the investigation of behavioral and performance on object knowledge tests may be
changes, the Frontal Behavioral Inventory (FBI)59,60 altered, especially on less familiar items; patients
is useful in the differential diagnosis between bvFTD with PPA-NF/A have satisfactory results in both
and other dementias and has good psychometric tasks. Conversely, the comprehension of complex
properties60-62. Recently, the FBI has been recommended sentences is frequently altered in PPA-NF/A, but it
for the differential diagnosis between FTD and Primary is generally preserved in PPA-S.

44   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

Repetition of sentences with different lengths and patients over 70 years old, the difficulty of performing
complexities is used to investigate the phonological the exam, and the onerous cost69.
loop of working memory. Patients with PPA-NF/A may Unlike the existing biomarkers for AD, there are
have repetition difficulties due to praxis problems. currently no specific biomarkers of pathophysiological
Individuals with PPA-S, however, show no difficulty changes associated with FTLD. The light chain
in this activity. neurofilament (NFL) has been intensively researched
The analysis of the type of errors in reading i n d e g e n e ra t i v e d e m e n t i a s , i n c l u d i n g F T D.
and writing tasks of regular, irregular words and NFL reflects axonal damage and is increased in FTD,
pseudowords makes it possible to differentiate as in other neurodegenerative diseases, such as AD.
patients with semantic impairment from those with NFL measurement is useful in differentiating FTD from
phonological difficulties. Patients with semantic other non-degenerative conditions such as primary
deficits manifest dyslexia and/or surface dysgraphia, psychiatric disorders 68. NFL dosage also appears
in which irregular words are read and/or written with to correlate with disease severity68. Despite its potential
oral support. The production of sentences and written interest, the measurement of NFL is not currently
texts also contributes to the recognition of syntactic available for use in Brazil.
difficulties. Patients with PPA-NF/A may present Most cases of FTD are sporadic70. However, in about
dyslexia and/or phonological dysgraphia. 40% of cases, a family history of dementia is identified.
Motor and praxis assessment of speech, including In approximately 10 to 15% of patients, an autosomal
diadochokinesia tests, helps the identification of dominant transmission pattern can be found, such as
dysarthria and/or speech apraxia. Speech apraxia mutations in the MAPT gene (“microtubule-associated
is a disturbance in the planning and programming protein tau”), in the progranulin (GRN) gene or the
of sensorimotor commands necessary to perform expansion. In Brazil, we found mutations in the
ar ticulator y movements. It is different from progranulin gene in about a third of familial cases,
dysarthria, which compromises the motor system. while c9orf72 expansions and pathogenic variants in
Speech apraxia is a primarily articulatory disorder, MAPT were found in about 10% of familial cases each70.
and prosody impairment may occur secondarily. Knowledge about monogenic forms of FTD has grown
As it is also responsible for changes in speech sounds, significantly over the last ten years and, currently,
it can be misdiagnosed as phonological impairments more than twenty genes with pathogenic variants
in oral emission. are known as genetic causes of FTD (many of them also
Performance on language tasks is significantly cause amyotrophic lateral sclerosis [ALS]).
impacted by education, occupation, and language The most frequent clinical presentations in
experiences (reading and writing habits, bilingualism, pathogenic variants of the progranulin gene are bvFTD
participation in social activities and hobbies involving (about 40% of cases in a multicenter study), PPA-NF/A
language). Research in Brazil has advanced to validate in 9%, and corticobasal syndrome in 4%. About 8%
and obtain standards for language tests. For a review of patients with this mutation have been diagnosed
of available instruments to assess adult language, with dementia of the Alzheimer type. The syndromes
see Parente et al.66. most frequently associated with c9orf72 expansions
Due to neuropathology, motor deficits are often are bvFTD (31%), FTD with ALS (11%) and ALS (20%).
associated with cognitive and behavioral conditions. In cases of pathogenic variants in the MAPT gene,
Thus, the speech therapy assessment should investigate the most frequently described clinical syndromes are
difficulties related to swallowing. bvFTD (44%), progressive supranuclear palsy (4%),
and Parkinson’s disease (5%)71.
Laboratory investigation Currently, the genetic investigation of familial FTD
For patients who undergo lumbar puncture, the cases is done by requesting the search for pathogenic
investigation of CSF biomarkers (beta-amyloid peptide, variants in gene panels associated with FTD or, preferably,
Tau and P-Tau) is useful to rule out AD, in cases of given the wide range of possible genes related to the disease
difficult differential diagnosis67,68. However, it should in a given family through exome sequencing. Importantly,
be noted that there are methodological issues that the search for c9orf72 expansion cannot be evaluated
must be considered when indicating and interpreting by exome analysis and a separate analysis is needed to
the dosage of biomarkers: the absence of established identify the presence of this mutation. In cases of FTD
universal reference values, analytical variability, with ALS, the search for c9orf72 expansion may even
the significant occurrence of false-positive results in precede exome sequencing. Of note, even in familial cases,

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   45


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

the exome sequencing and expansion search in c9orf72 of the frontal (or dysexecutive) variant of AD 73,
may be negative, and a mutation may not be identified. which is an atypical presentation of the disease
In a recent multicenter study, pathogenic variants in and manifests with apathy, emotional blunting,
the three major genes (GRN, MAPT, c9orf72) were only depressive symptoms, and episodic memory deficit.
identified in 61% of familial cases72. There are also radiotracers that bind to the tau
protein; however, so far, these tracers seem to be
Neuroimaging investigation better at detecting tau deposits related to AD.
At the tertiary level, MRI is also used as the main In conclusion, the diagnosis of different variants
diagnostic imaging method. However, in the early of FTD is mainly based on clinical interviews and the
stages of the bvFTD, structural changes may be subtle assessment of cognitive, linguistic and behavioral
or even absent. In this context, the use of functional aspects. Complementary tests provide valuable
neuroimaging (cerebral perfusion scintigraphy information to support the diagnosis and to rule
and, mainly, positron emission tomography with out causes that may be similar to FTD conditions.
fluorodeoxyglucose) may allow the identification The advent of new biological markers may provide
of alterations that suggest a neurodegenerative greater diagnostic accuracy in the years to come.
process (hyperfusion or hypometabolism), before
the atrophy is undoubted. Functional neuroimaging
exams may show metabolic or perfusion dysfunction in ACKNOWLEDGEMENTS
the prefrontal cortex and temporal poles, depending on PC, LCS and RN are funded by CNPq, Brazil (bolsa de
the clinical type of FTD. Positron emission tomography produtividade em pesquisa).
has better diagnostic accuracy than scintigraphy.
Currently, there are molecular neuroimaging Authors’ contributions. LCS, MLH, THM, MTCG, MSY,
methods of positron emission tomography, which VSB, LTT, JS, BJAPB, LPS: concept; LCS, MLH, THM,
allow the detection of cerebral beta-amyloid MTCG, MSY, VSB, LTT: drafting of the manuscript;
(such as PET with Pittsburgh Compound B, or PiB). BJAPB, FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC,
Although it does not help in the diagnosis of PHFB, RN, SMDB: critical revision of the manuscript
bvFTD, it is a method that allows the diagnosis for important intellectual content.

REFERENCES Brazilian cases. Dement Neuropsychol. 2013;7(1):110-21. doi:10.1590/


S1980-57642013DN70100017.
1. Perry DC, Brown JA, Possin KL, Datta S, Trujillo A, Radke A, et al.
Clinicopathological correlations in behavioural variant frontotemporal 12. Wicklund MR, Duffy JR, Strand EA, Machulda MM, Whitwell JL,
dementia. Brain. 2017;140(12):3329-45. doi:10.1093/brain/awx254. Josephs KA. Quantitative application of the primary progressive aphasia
2. Bang J, Spina S, Miller BL. Frontotemporal dementia. Lancet. consensus criteria. Neurology. 2014;82(13):1119-26. doi:10.1212/
2015;386(10004):1672-82. doi:10.1016/S0140-6736(15)00461-4. WNL.0000000000000261.
3. Custodio N, Herrera-Perez E, Lira D, Montesinos R, Bendezu L. Prevalence 13. Reul S, Lohmann H, Wiendl H, Duning T, Johnen A. Can cognitive
of frontotemporal dementia in community-based studies in Latin America: assessment really discriminate early stages of Alzheimer’s and behavioural
a systematic review. Dement Neuropsychol. 2013;7(1):27-32. doi:10.1590/ variant frontotemporal dementia at initial clinical presentation? Alzheimers
S1980-57642013DN70100005. Res Ther. 2017;9(1):61. doi:10.1186/s13195-017-0287-1.
4. O’Connor CM, Landin-Romero R, Clemson L, Kaizik C, Daveson N, 14. Wong S, Bertoux M, Savage G, Hodges JR, Piguet O, Hornberger M.
Hodges JR,et al. Behavioral-variant frontotemporal dementia: Distinct Comparison of Prefrontal Atrophy and Episodic Memory Performance in
phenotypes with unique functional profiles. Neurology. 2017;89(6):570-77. Dysexecutive Alzheimer’s Disease and Behavioral-Variant Frontotemporal
doi: 10.1212/WNL.0000000000004215. Dementia. J Alzheimers Dis. 2016;51(3):889-903. doi:10.3233/JAD-151016.
5. Ulugut Erkoyun H, Groot C, Heilbron R, Nelissen A, Rossum J, Jutten R, et al. 15. Bertoux M, Cassagnaud P, Lebouvier T, Lebert F, Sarazin M,
A clinical-radiological framework of the right temporal variant of frontotemporal Le Ber I, et al. Does amnesia specifically predict Alzheimer’s pathology?
dementia. Brain. 2020;143(9):2831-43. doi:10.1093/brain/awaa225. A neuropathological study. Neurobiol Aging. 2020;95:123-30.
6. Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, doi:10.1016/j.neurobiolaging.2020.07.011.
Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural 16. Hornberger M, Piguet O. Episodic memory in frontotemporal dementia:
variant of frontotemporal dementia. Brain. 2011;134(Pt 9):2456-77. a critical review. Brain. 2012;135(Pt 3):678-92. doi:10.1093/brain/aws011.
doi:10.1093/brain/awr179. 17. Brucki SMD, Nitrini R, Caramelli P, Bertolucci PHF, Okamoto IH.
7. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, Cappa SF, Suggestions for utilization of the mini-mental state examination
et al. Classification of primary progressive aphasia and its variants. Neurology. in Brazil. Arq Neuropsiquiatr. 2003;61(3B):777-81. doi:10.1590/
2011;76(11):1006-14. doi:10.1212/WNL.0b013e31821103e6. S0004-282X2003000500014.
8. Carthery-Goulart MT. Primary Progressive Aphasia. In: Pachana NA, 18. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical
editora. Encyclopedia of Geropsychology. Singapore: Springer; 2017. method for grading the cognitive state of patients for the clinician.
p. 1-11. doi:10.1007/978-981-287-082-7_315 J Psychiatr Res. 1975;12(3):189-98. doi:10.1016/0022-3956(75)90026-6.
9. Snowden JS, Thompson JC, Neary D. Knowledge of famous faces and 19. Yew B, Alladi S, Shailaja M, Hodges JR, Hornberger M. Lost and forgotten?
names in semantic dementia. Brain. 2004;127(Pt 4):860-72. doi:10.1093/ Orientation versus memory in Alzheimer’s disease and frontotemporal
brain/awh099. dementia. J Alzheimers Dis. 2013;33(2):473-81. doi:10.3233/
10. Springer L. Therapeutic Approaches in Aphasia Rehabilitation. In: Stemmer B, JAD-2012-120769.
Whitaker H, editores. Handbook of the Neuroscience of Language; 20. Caramelli P, Teixeira AL, Buchpiguel CA, Lee HW, Livramento JA,
2008. p. 397-406. Fernandez LL, et al. Diagnosis of Alzheimer’s disease in Brazil:
11. Senaha MLH, Caramelli P, Brucki SMD, Smid J, Takada LT, Porto CS, et al. Supplementary exams. Dement Neuropsychol. 2011;5(3):167-77.
Primary progressive aphasia: classification of variants in 100 consecutive doi:10.1590/S1980-57642011DN05030004.

46   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

21. Nitrini R, Bucki SMD, Yassuda MS, Fichman HC, Caramelli P. 42. Foss MP, Formigheri P, Speciali JG. Heterogeneity of cognitive aging
The Figure Memory Test: diagnosis of memory impairment in populations in Brazilian normal elderls. Dement Neuropsychol. 2009;3(4):344-51.
with heterogeneous educational background. Dement Neuropsychol. doi:10.1590/S1980-57642009DN30400014.
2021;15(2):173-85. doi:10.1590/1980-57642021dn15-020004. 43. Johnen A, Bertoux M. Psychological and Cognitive Markers of Behavioral
22. Nitrini R, Lefèvre BH, Mathias SC, Caramelli P, Carrilho PE, Sauaia N, Variant Frontotemporal Dementia-A Clinical Neuropsychologist’s View on
et al. Neuropsychological tests of simple application for diagnosing Diagnostic Criteria and Beyond. Front Neurol. 2019;10:594. doi:10.3389/
dementia. Arq Neuropsiquiatr. 1994;52(4):457-65. doi:10.1590/ fneur.2019.00594.
s0004-282x1994000400001. 44. Mariano LI, O’Callaghan C, Guimaraes HC, Gambogi LB,
23. Yassuda MS, Silva HS, Lima-Silva TB, Cachioni M, Falcão DVS, Lopes A, Silva TBL, Yassuda MS, et al. Disinhibition in Frontotemporal Dementia
et al. Normative data for the Brief Cognitive Screening Battery stratified and Alzheimer’s Disease: A Neuropsychological and Behavioural
by age and education. Dement Neuropsychol. 2017;11(1):48-53. Investigation. J Int Neuropsychol Soc. 2020;26(2):163-71. doi:10.1017/
doi:10.1590/1980-57642016dn11-010008. S1355617719000973.
24. Carvalho VA, Caramelli P. Brazilian adaptation of the Addenbrooke’s 45. Nascimento E, Figueiredo VLM. WISC-III and WAIS-III: alterations
Cognitive Examination-Revised (ACE-R). Dement Neuropsychol. in the current american original versions of the adaptations for
2007;1(2):212-16. doi:10.1590/s1980-57642008dn10200015. use in Brazil. Psicol Reflex Crit. 2002;15(3):603-12. doi:10.1590/
25. César KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. Addenbrooke’s S0102-79722002000300014.
cognitive examination-revised: normative and accuracy data for seniors 46. Quental NB, Brucki SM, Bueno OF. Visuospatial function in early
with heterogeneous educational level in Brazil. Int Psychogeriatr. Alzheimer’s disease--the use of the Visual Object and Space Perception
2017;29(8):1345-53. doi:10.1017/S1041610217000734. (VOSP) battery. PLoS One. 2013;8(7):e68398. doi:10.1371/journal.
26. Beato R, Amaral-Carvalho V, Guimarães HC, Tumas V, Souza CP, pone.0068398.
Oliveira GN, et al. Frontal assessment battery in a Brazilian sample of 47. Miotto EC, Sato J, Lucia MC, Camargo CH, Scaff M. Development
healthy controls: normative data. Arq Neuropsiquiatr. 2012;70(4):278-80. of an adapted version of the Boston Naming Test for Portuguese
doi:10.1590/s0004-282x2012005000009. speakers. Braz J Psychiatry. 2010;32(3):279-82. doi:10.1590/
27. Dubois B, Slachevsky A, Litvan I, Pillon B. The FAB: a Frontal Assessment s1516-44462010005000006.
Battery at bedside. Neurology. 2000;55(11):1621-6. doi:10.1212/ 48. Bora E, Velakoulis D, Walterfang M. Meta-Analysis of Facial Emotion
wnl.55.11.1621. Recognition in Behavioral Variant Frontotemporal Dementia: Comparison
28. Torralva T, Roca M, Gleichgerrcht E, López P, Manes F. INECO Frontal With Alzheimer Disease and Healthy Controls. J Geriatr Psychiatry Neurol.
Screening (IFS): a brief, sensitive, and specific tool to assess executive 2016;29(4):205-11. doi:10.1177/0891988716640375.
functions in dementia. J Int Neuropsychol Soc. 2009;15(5):777-86. 49. Bora E, Walterfang M, Velakoulis D. Theory of mind in behavioural-variant
doi:10.1017/S1355617709990415. frontotemporal dementia and Alzheimer’s disease: a meta-analysis.
29. Bahia VS, Cecchini MA, Cassimiro L, Viana R, Lima-Silva TB, de Souza LC, J Neurol Neurosurg Psychiatry. 2015;86(7):714-9. doi:10.1136/
et al. The Accuracy of INECO Frontal Screening in the Diagnosis of jnnp-2014-309445.
50. Bertoux M, Delavest M, de Souza LC, Funkiewiez A, Lépine JP,
Executive Dysfunction in Frontotemporal Dementia and Alzheimer
Fossati P, et al. Social Cognition and Emotional Assessment differentiates
Disease. Alzheimer Dis Assoc Disord. 2018;32(4):314-9. doi:10.1097/
frontotemporal dementia from depression. Neurol Neurosurg Psychiatry.
WAD.0000000000000255.
2012;83(4):411-6. doi:10.1136/jnnp-2011-301849.
30. Cummings JL. The Neuropsychiatric Inventory: assessing psychopathology
51. Moura MVB, Mariano LI, Teixeira AL, Caramelli P, de Souza LC. Social
in dementia patients. Neurology 1997;48(5 Suppl 6):S10-6. doi:10.1212/
Cognition Tests Can Discriminate Behavioral Variant Frontotemporal
wnl.48.5_suppl_6.10s.
Dementia From Alzheimer’s Disease Independently of Executive Functioning.
31. Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi DA,
Arch Clin Neuropsychol. 2020;26(5):831-7. doi:10.1093/arclin/acaa084.
Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment
52. Mariano LI, Caramelli P, Guimaraes HC, Gambogi LB, Moura MVB,
of psychopathology in dementia. Neurology. 1994;44(12):2308-14.
Yassuda MS, et al. Can Social Cognition Measurements Differentiate
doi:10.1212/wnl.44.12.2308.
Behavioral Variant Frontotemporal Dementia from Alzheimer’s Disease
32. Kaufer DI, Cummings JL, Ketchel P, Smith V, MacMillan A, Shelley T,
Regardless of Apathy? J Alzheimers Dis. 2020;74(3):817-27. doi:10.3233/
et al. Validation of the NPI-Q, a brief clinical form of the Neuropsychiatric
JAD-190861.
Inventory. J Neuropsychiatry Clin Neurosci. 2000;12(2):233-9.
53. Bertoux M, de Souza LC, O’Callaghan C, Greve A, Sarazin M, Dubois B,
doi:10.1176/jnp.12.2.233. et al. Social Cognition Deficits: The Key to Discriminate Behavioral Variant
33. Camozzato AL, Godinho C, Kochhann R, Massochini G, Frontotemporal Dementia from Alzheimer’s Disease Regardless
Chaves ML. Validity of the Brazilian version of the Neuropsychiatric of Amnesia? Journal of Alzheimers Disease. 2015;49(4):1065-74.
Inventory Questionnaire (NPI-Q). Arq Neuropsiquiatr. 2015;73(1):41-5. doi:10.3233/JAD-150686.
doi:10.1590/0004-282X20140177. 54. Ducharme S, Dols A, Laforce R, Devenney E, Kumfor F, Stock J, et al.
34. Bahia VS, Silva MNM, Viana R, Smid J, Damin AE, Radanovic M, et al. Behavioral Recommendations to distinguish behavioural variant frontotemporal
and activities of daily living inventories in the diagnosis of frontotemporal dementia from psychiatric disorders. Brain. 2020;143(6):1632-50.
lobar degeneration and Alzheimer’s disease. Dement Neuropsychol. doi:10.1093/brain/awaa018.
2008;2(2):108-13. doi:10.1590/S1980-57642009DN20200006. 55. de Souza LC, Bertoux M, Faria ÂRV, Corgosinho LTS, Prado ACA,
35. de Souza LC, Lehéricy S, Dubois B, Stella F, Sarazin M. Neuroimaging in Barbosa IG, et al. The effects of gender, age, schooling, and cultural
dementias. Current Opinion in Psychiatry. 2012;25(6):473-9. doi:10.1097/ background on the identification of facial emotions: a transcultural study. Int
YCO.0b013e328357b9ab. Psychogeriatr. 2018;30(12):1861-70. doi:10.1017/S1041610218000443.
36. Whitwell JL. FTD spectrum: Neuroimaging across the FTD spectrum. 56. Watanabe RGS, Knochenhauer AE, Fabrin MA, Siqueira HH, Martins HF,
Prog Mol Biol Transl Sci. 2019;165:187-223. doi:10.1016/ Oliveira Mello CD, et al. Faux Pas Recognition Test: transcultural adaptation
bs.pmbts.2019.05.009. and evaluation of its psychometric properties in Brazil. Cogn Neuropsychiatry.
37. de Souza LC, Chupin M, Bertoux M, Lehéricy S, Dubois B, Lamari F, 2021;26(5):3321-34. doi:10.1080/13546805.2021.
et al. Is Hippocampal Volume a Good Marker to Differentiate Alzheimer’s 57. Mioshi E, Hsieh S, Savage S, Hornberger M, Hodges JR. Clinical
Disease from Frontotemporal Dementia? Journal of Alzheimers Disease staging and disease progression in frontotemporal dementia. Neurology.
2013;36(1):57-66. doi:10.3233/JAD-122293. 2010;74(20):1591-7. doi:10.1212/WNL.0b013e3181e04070.
38. Cash DM, Bocchetta M, Thomas DL, Dick KM, Swieten JC, Borroni B, 58. Lima-Silva TB, Bahia VS, Cecchini MA, Cassimiro L, Guimarães HC,
et al. Patterns of gray matter atrophy in genetic frontotemporal dementia: Gambogi LB, et al. Validity and Reliability of the Frontotemporal Dementia
results from the GENFI study. Neurobiol Aging. 2018;62:191-6. Rating Scale (FTD-FRS) for the Progression and Staging of Dementia
doi:10.1016/j.neurobiolaging.2017.10.008. in Brazilian Patients. Alzheimer Dis Assoc Disord. 2018;32(3):220-5.
39. Ameur F, Colliot O, Caroppo P, Ströer S, Dormont D, Brice A, et al. doi:10.1097/WAD.0000000000000246.
White matter lesions in FTLD: distinct phenotypes characterize GRN 59. Kertesz A, Nadkarni N, Davidson W, Thomas AW. The Frontal Behavioral
and C9ORF72 mutations. Neurol Genet. 2016;2(1):e47. doi:10.1212/ Inventory in the differential diagnosis of frontotemporal dementia. J Int
NXG.0000000000000047. Neuropsychol Soc. 2000;6(4):460-8. doi:10.1017/s1355617700644041.
40. Harciarek M, Cosentino S. Language, executive function and social 60. Suhonen NM, Hallikainen I, Hanninen T, Jokelainen J, Krüger J, Hall A,
cognition in the diagnosis of frontotemporal dementia syndromes. Int Rev et al. The Modified Frontal Behavioral Inventory (FBI-mod) for Patients with
Psychiatry. 2013;25(2):178-96. doi:10.3109/09540261.2013.763340. Frontotemporal Lobar Degeneration, Alzheimer’s Disease, and Mild Cognitive
41. Malloy-Diniz LF, Lasmar VA, Gazinelli LS, Fuentes D, Salgado JV. Impairment. J Alzheimers Dis. 2017;56(4):1241-51. doi:10.3233/JAD-160983.
The Rey Auditory-Verbal Learning Test: applicability for the Brazilian 61. Konstantinopoulou E, Aretouli E, Ioannidis P, Karacostas D,
elderly population. Braz J Psychiatry. 2007;29(4):324-9. Kosmidis MH. Behavioral disturbances differentiate frontotemporal lobar

de Souza LC, et al.   Diagnosis of frontotemporal dementia.   47


Dement Neuropsychol 2022 September;16(3 Suppl. 1):36-48

degeneration subtypes and Alzheimer’s disease: evidence from the 68. Swift IJ, Sogorb-Esteve A, Heller C, Synofzik M, Otto M, Graff C, et al.
Frontal Behavioral Inventory. Int J Geriatr Psychiatry. 2013;28(9):939-46. Fluid biomarkers in frontotemporal dementia: past, present and future.
doi:10.1002/gps.3907. J Neurol Neurosurg Psychiatry. 2021;92(2):204-15. doi:10.1136/
62. Milan G, Lamenza F, Iavarone A, Galeone F, Lorè E, Falco C, et al. Frontal
jnnp-2020-323520.
Behavioural Inventory in the differential diagnosis of dementia. Acta Neurol
Scand. 2008;117(4):260-5. doi:10.1111/j.1600-0404.2007.00934.x. 69. Frisoni GB, Boccardi M, Barkhof F, Blennow K, Cappa S, Chiotis K,
63. Boutoleau-Bretonniere C, Evrard C, Hardouin JB, Rocher L, Charriau T, et al. Strategic roadmap for an early diagnosis of Alzheimer’s disease
Etcharry-Bouyx F, et al. DAPHNE: A New Tool for the Assessment of the based on biomarkers. Lancet Neurol. 2017;16(8):661-76. doi:10.1016/
Behavioral Variant of Frontotemporal Dementia. Dement Geriatr Cogn Dis S1474-4422(17)30159-X.
Extra. 2015;5(3):503-16. doi:10.1159/000440859.
70. Takada LT. The Genetics of Monogenic Frontotemporal Dementia. Dement
64. Lehingue E, Gueniat J, Jourdaa S, Hardouin JB, Pallardy A,
Courtemanche H, et al. Improving the Diagnosis of the Frontal Variant Neuropsychol 2015;9(3):219-29. doi:10.1590/1980-57642015DN93000003.
of Alzheimer’s Disease with the DAPHNE Scale. J Alzheimers Dis. 71. Moore KM, Nicholas J, Grossman M, McMillan CT, Irwin DJ, Massimo L,
2021;79(1):1735-45. doi:10.3233/JAD-201088. et al. Age at symptom onset and death and disease duration in genetic
65. Montembeault M, Brambati SM, Gorno-Tempini ML, Migliaccio R. Clinical, frontotemporal dementia: an international retrospective cohort study.
Anatomical, and Pathological Features in the Three Variants of Primary
Lancet Neurol. 2020;19(2):145-56. doi:10.1016/S1474-4422(19)30394-1.
Progressive Aphasia: A Review. Front Neurol. 2018;9:692. doi:10.3389/
fneur.2018.00692. 72. Ramos EM, Dokuru DR, Van Berlo V, Wojta K, Wang Q, Huang AY, et al.
66. Parente MAMP, Baradel RR, Fonseca RP, Pereira N, Carthery-Goulart MT. Genetic screening of a large series of North American sporadic and familial
Evolution of language assessment in patients with acquired neurological frontotemporal dementia cases. Alzheimers Dement. 2020;16(1):118-30.
disorders in Brazil. Dement Neuropsychol. 2014;8(3):196-206. doi:10.1002/alz.12011.
doi:10.1590/S1980-57642014DN83000002.
73. Ossenkoppele R, Pijnenburg YA, Perry DC, Cohn-Sheehy BI, Scheltens NM,
67. de Souza LC, Lamari F, Belliard S, Jardel C, Houillier C, De Paz R, et al.
Cerebrospinal fluid biomarkers in the differential diagnosis of Alzheimer’s Vogel JW, et al. The behavioural/dysexecutive variant of Alzheimer’s
disease from other cortical dementias. J Neurol Neurosurg Psychiatry. disease: clinical, neuroimaging and pathological features. Brain.
2011;82(3):240-6. doi:10.1136/jnnp.2010.207183. 2015;138(Pt 9):2732-49. doi:10.1093/brain/awv191.

48   Diagnosis of frontotemporal dementia.   de Souza LC, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S104EN

Diagnosis of vascular
cognitive impairment:
recommendations of the scientific department
of cognitive neurology and aging of the
Brazilian Academy of Neurology
Breno José Alencar Pires Barbosa1,2,3 , José Ibiapina Siqueira Neto4 , Gilberto Sousa Alves5 ,
Felipe Kenji Sudo6 , Claudia Kimie Suemoto7 , Fernanda Tovar-Moll6 , Jerusa Smid3 ,
Lucas Porcello Schilling8,9,10 , Marcio Luiz Figueredo Balthazar11 , Norberto Anízio Ferreira Frota12,13 ,
Leonardo Cruz de Souza14 , Francisco Assis Carvalho Vale15 , Paulo Caramelli14 ,
Paulo Henrique Ferreira Bertolucci16 , Sonia Maria Dozzi Brucki3 , Ricardo Nitrini3 ,
Eliasz Engelhardt17 , Márcia Lorena Fagundes Chaves18,19

1
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
2
Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.
3
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
4
Faculdade de Medicina da Universidade Federal do Ceará. Fortaleza, Brazil.
5
Universidade Federal do Maranhão, Centro de Ciências da Saúde, Departamento de Medicina I, São Luís MA, Brazil.
6
Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro RJ, Brazil.
7
Universidade de São Paulo, Faculdade de Medicina, Divisão de Geriatria, São Paulo SP, Brazil.
8
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
9
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
10
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
11
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil.
12
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
13
Universidade de Fortaleza, Fortaleza CE, Brazil.
14
Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
15
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brazil.
16
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil.
17
Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto e Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil.
18
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
19
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brazil.
Correspondence: Breno José Alencar Pires Barbosa; Email: brenojb@gmail.com.
Conflict of interest: JS: Participation as speaker in symposia sponsored by Roche laboratory; LPS: Participation in advisory boards for Biogen. Participation as speaker in
symposia sponsored by Aché, Apsen and Biogen laboratories; MLFB: Participation in advisory boards for Biogen. Development of material for continuous medical education
and participation as speaker in symposia sponsored by EMS and Torrent laboratories; PC: Participation as principal investigator in clinical trials for Novo Nordisk and Roche
laboratories. Participation in advisory boards for Aché, Biogen, EMS, Nutricia and Roche laboratories. Development of material for continuous medical education and participation
as speaker in symposia sponsored by Aché, Nutricia, Libbs, Roche, Sandoz, Torrent and Zodiac laboratories; PHFB: Participation in advisory boards for Biogen and Novo
Nordisk laboratories. Supervision of training activities for Biogen; Janssen-Cilag and Novo Nordisk laboratories and for Quintiles. Participation as speaker in symposia sponsored
by Apsen, Nutricia, Roche and Sandoz laboratories; LCS: Participation in advisory board for Biogen. Participation as speaker in symposia sponsored by Biogen;
RN: Participation in advisory board for Biogen; BJAPB, JISN, GSA, FKS, CKS, FTM, NAFF, FACV, SMDB, EE, MLFC: There is no conflict of interest to declare.
Received on August 02, 2021; Received in its final form on November 10, 2021; Accepted on April 27, 2022.

Barbosa BJAP, et al.   Vascular cognitive impairment.   49


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

ABSTRACT. Since the publication of the latest recommendations for the diagnosis and treatment of Vascular Dementia by the Brazilian Academy of Neurology
in 2011, significant advances on the terminology and diagnostic criteria have been made. This manuscript is the result of a consensus among experts
appointed by the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (2020-2022). We aimed to update practical
recommendations for the identification, classification, and diagnosis of Vascular Cognitive Impairment (VCI). Searches were performed in the MEDLINE,
Scopus, Scielo, and LILACS databases. This guideline provides a comprehensive review and then synthesizes the main practical guidelines for the diagnosis
of VCI not only for neurologists but also for other professionals involved in the assessment and care of patients with VCI, considering the different levels
of health care (primary, secondary and tertiary) in Brazil.
Keywords: Dementia, Vascular; Cognitive Dysfunction; Cerebral Infarction; Stroke.

DIAGNÓSTICO DO COMPROMETIMENTO COGNITIVO VASCULAR: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO DE NEUROLOGIA COGNITIVA


E DO ENVELHECIMENTO DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. Desde a publicação das últimas recomendações para o diagnóstico e tratamento da Demência Vascular pela Academia Brasileira de Neurologia
em 2011, avanços significativos ocorreram na terminologia e critérios diagnósticos. O presente manuscrito é resultado do consenso entre especialistas
indicados pelo Departamento Científico de Neurologia Cognitiva e do Envelhecimento da Academia Brasileira de Neurologia (2020-2022). O objetivo foi
atualizar as recomendações práticas para a identificação, classificação e diagnóstico do Comprometimento Cognitivo Vascular (CCV). As buscas foram
realizadas nas plataformas MEDLINE, Scopus, Scielo e LILACS. As recomendações buscam fornecer uma ampla revisão sobre o tema, então sintetizar
as evidências para o diagnóstico do CCV não apenas para neurologistas, mas também para outros profissionais de saúde envolvidos na avaliação
e nos cuidados ao paciente com CCV, considerando as diferentes realidades dos níveis de atenção à saúde (primário, secundário e terciário) no Brasil.
Palavras-chave: Demência Vascular; Disfunção Cognitiva; Infarto Cerebral; Acidente Vascular Cerebral.

INTRODUCTION This guideline seeks to provide a comprehensive

V ascular Cognitive Impairment ( VCI) is the


term used to include the entire spectrum of
changes in cognition directly or indirectly related to
review and then synthesize the main practical guidelines
for the diagnosis of VCI not only for neurologists but
also for other professionals involved in the assessment
cerebrovascular disease1. It is a construct proposed and care of patients with VCI, considering the reality
by Vladimir Hachinski (1994) to describe cases different levels of health care (primary, secondary
associated with “cerebrovascular disease” (CVD)2 and and tertiary) in Brazil and Latin America.
partially replace the concept of “Vascular Dementia”
(VD) proposed by Carlo Loeb3. The term VCI identifies
and includes all forms and severity levels of cognitive METHODS
impairment, constituting a continuum of clinical and This manuscript is the result of a consensus among
pathological presentations, from an asymptomatic experts appointed by the Scientific Department
stage (the “brain-at-risk) to dementia VD, including of Cognitive Neurology and Aging of the Brazilian
an intermediate stage of clinical deficits that do Academy of Neurolog y (2020-2022). We aimed
not reach the dementia criteria, initially called the to update practical recommendations for the
“pre-dementia” stage2,4, which was named “Vascular identification, classification, and diagnosis of VCI.
Cognitive Impairment Non-Dementia” (VCIND) 5,6 There are currently several updated guidelines and
or “Vascular Mild Cognitive Impairment” (VMCI)7-9. consensuses on the topic, many of which are cited
Thus, the symptomatic spectrum of the condition and discussed throughout the text, so it was not
comprises VCIND/VMCI and VCI (or VD). our objective to carry out a new systematic review
Since the publication of the latest recommendations or exhaustive classification of the evidence.
for the diagnosis and treatment of Vascular Dementia Searches were performed in the MEDLINE, Scopus,
by the Brazilian Academy of Neurology in 201110-12, Scielo, and LILACS databases until June 2021, using
significant advances on the terminology and diagnostic the descriptors “vascular cognitive impairment”
criteria has been made1,9-15. New markers of structural or “vascular dementia”. We selected mostly articles
and functional neuroimaging allowed the understanding published in the last 10 years, but older relevant
of the heterogeneity of clinical presentations of VCI, publications were also included. Articles in English,
including the compensation mechanisms of neural Portuguese, and Spanish were reviewed. We also
networks 16. We also highlight contributions from revised the reference list of the articles for relevant
Brazilian and Latin American groups to the area, additional references. Review articles were also
which will be highlighted throughout the text. included when applicable.

50   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

EPIDEMIOLOGY AND RISK FACTORS Classic metabolic and cardiovascular RFs are hypertension,
The prevalence of VCI is too complex to estimate, diabetes, dyslipidemia, atrial fibrillation, previous stroke,
because of geographic factors that imply in very metabolic syndrome, obesity, glucose intolerance, elevated
heterogeneous societies, the huge variation in the homocysteine, carotid stenosis, and hyperuricemia.
criteria used for diagnosis, different complementary Toxic RFs include alcoholism, smoking, and other causes
methods used in the investigation, or the scarcity such as low education, sedentary lifestyle, inadequate diet,
of studies, especially in Brazil and in low- and sleep apnea, and depression33.
middle-income countries 17,18. Despite the lack of
standardized diagnostic criteria, making it difficult
to determine its prevalence and risk factors (RF), MECHANISMS AND PATHOPHYSIOLOGY
VD is accepted as the second leading cause of dementia Cerebral vascular injuries comprise the ischemic
in the elderly, ranging from 8-45% of cases19,20. (infarcts, microinfarcts, lacunae, white matter
Studies with post-stroke patients have detected hyperintensities, enlarged perivascular spaces) and
VCIND in 24 to 70% of cases21,22. Considering that hemorrhagic lesions (hemorrhagic infarcts, cerebral
VD usually affects up to a third of individuals who hemorrhages, and microhemorrhages), which present
have suffered a stroke, it is observed that the VCIND in a variable way, with no single neuropathological
segment has a higher prevalence than dementia lesion characterizing VCI. In addition, there are no
conditions23. A Brazilian study evaluated 172 patients widely accepted criteria in relation to the location and
one year after an ischemic stroke and found that number of lesions necessary for the neuropathological
12.2% of cases met the criteria for probable VD24. diagnosis of VCI34-37. Several neuropathologic events
Another study estimated that approximately 5% of seem to contribute to the occurrence of VCI, including
individuals over 65 years of age had VCI, with 2.4% loss of white matter integrity with consequent
in the VCI-ND stage and 1.5% in the DV stage 6. disconnection between strategic areas for cognitive
Similarly, review studies showed that the prevalence networks 16, changes in the coagulation cascade 38
of VMCI ranged between 21 and 30%, affecting and oligodendrocytes39, and changes in endothelial cells
24-75% in cases with diagnosed stroke and 4-19% with alterations in cerebral blood perfusion40.
in those in which stroke had not been reported 25. The presence of lesions on neuroimaging must be
The high prevalence of VCI-ND/VMCI highlights the interpreted considering the clinical context. To cause
importance of this etiology of dementia, especially clinical symptoms, several basic characteristics must be
considering that early diagnosis and treatment met, such as extension, location and number of lesions.
of RF for VCI can prevent, stabilize, or prevent the In addition, other factors can influence the clinical
development of VD19,26-28. A clinicopathological study outcome of injuries, such as diaschisis, compensation
by the Biobank for Aging Studies of the University mechanisms, and cognitive reserve41-43.
of São Paulo described a prevalence of DV of 35%, The parameters for VCI-ND and DV regarding
considering only the presence of chronic infarcts for location, extension, and the number of lesions were
the neuropathological diagnosis, increasing to 49% previously examined by several authors. Lesions
when the presence of moderate to severe small vessel located in limbic-paralimbic regions, heteromodal
disease was included in the neuropathological criteria associative areas, certain subcortical structures,
for VD29. It should be remembered that mixed forms or in their connections tend to produce especially
of vascular pathology with neurodegenerative disease relevant pictures of VCI 44,45 . T hus, lesions in
[e.g., VCI + Alzheimer’s disease (AD)] are also included the following areas are related to clinical symptoms:
in the VCI construct, with important participation in the anterior cerebral artery (affecting the prefrontal
the total prevalence of VCI19,30,31. The possibility that region), the middle cerebral artery (associative areas of
mixed forms have their evolution attenuated and/or the parietal lobe, parietotemporal, temporo-occipital),
delayed through preventive measures is another the posterior cerebral artery (inferotemporal region),
aspect of great importance2,19,3. hippocampus, and thalamus nuclei [anterior, medial
The RFs for VCI are diverse. They are classically dorsal]) 45-47 . As for the white matter, which is
divided into sociodemographic, clinical characteristics, partly made up of long intra-hemispheric bundles,
neuroimaging aspects, and VCI characteristics. the frontosubcortical pathways (fronto-striatum-pale-
Non-modifiable RFs include advanced age, gender, thalamus-frontal circuits) underlying the executive
ethnicity, and genetic aspects (CADASIL, CARASIL, function should be highlighted. Damage to these
VLDL-R, APOE ε-4, HERNS, FABRY, among others). tracts is frequent in cases of VCI, even in the

Barbosa BJAP, et al.   Vascular cognitive impairment.   51


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

early stages 48. It should also be remembered that, In patholog ic studies, the definition of a
regardless of their location, white matter lesions neuropathologic threshold to consider the lesion as
in any location compromise the frontal function49. a cause of cognitive alteration in VCI is a difficult
However, the extent and number of detectable task53,54. The same can be said about VCI as a whole.
lesions have been less studied. The most recent criteria Furthermore, evidence from the recent decades
for VCI pointed to the need for a lower vascular indicates that isolated DV is much less prevalent
load (fewer number of lesions) for the diagnosis of than mixed VCI, a product of degenerative AD type
non-dementia presentations 1,9,14,50. It is important and cerebrovascular lesions55. VCI Changes at any
to remember that white matter hyperintensities stage can be associated with neurodegenerative
(WMH) are not homogeneously constituted and may disorders, such as AD, constituting mixed pictures
present with different degrees of tissue alteration, (such as VD+AD) 23,56, as well as other conditions
with varied rarefaction. This aspect has been described (Frontotemporal lobar degeneration, Dementias
in histopathology and diffusion tensor studies51,52. with Lewy’s Body) (Figure 1 A and B)1.

VaD
Vascular
pathology VCI

Neurodegenerative
pathology

Vascular
VCI/ pathology
Mixed
pathologies
AD pathology

VCI: vascular cognitive impairment; VaD: vascular dementia.


Figure 1 A and B. Relationship between vascular cognitive impairment and dementia, adapted99. VCI refers to any degree of cognitive decline related
to cerebrovascular pathology, from the pre-clinical stages (brain at risk), through VMCI and dementia. VCI can be an isolated cause of cognitive decline or,
to some degree, coexist with neurodegenerative pathology such as AD. The term Vascular Dementia refers to the subgroup of patients whose cognitive
decline is mostly cerebrovascular in nature.

THE VASCULAR COGNITIVE IMPAIRMENT SPECTRUM their occurrence is strongly associated with the presence
of vascular-related RF, such as metabolic diseases,
Brain-at-risk smoking, among others 59.
White-matter changes – especially symmetrical bilateral In addition, according to meta-analyses, extensive
punctiform lesions, located in periventricular and deep WMH burden conferred a 73-84% increased risk
subcortical regions – are commonly found in healthy of incident dementia 59,60. Hence, the observation
elderly subjects57. Although often detected in late-life, that these neuroimaging findings precede the onset
WMH on T2 and FLAIR (fluid attenuated inversion of cognitive and behavioral abnormalities suggest
recovery) magnetic resonance imaging (MRI) are not that, similarly to AD61, a preclinical stage may exist
inherent features of normal brain aging58. In fact, in VCI (Figure 2).

52   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

With the high prevalence of cerebrovascular disease recurrence and the “allostatic load” (which refers to the
in older population, determining the odds of cognitive cumulative effects of multiple vascular RF) is needed.
decline attributed to individual or combined biomarkers, The idea that broadly available therapeutic interventions
including the lesion type at neuroimaging (WMH, may effectively participate in the primary prevention of
lacunes, microbleeds, perivascular space dilations, amyloid symptomatic VCI emphasizes the importance of more
angiopathy etc.), the lesion load, the speed of infarct studies aiming at the characterization of this stage62,63.

Brain at risk Vascular and


Vascular dementia
(preclinical phase) mild cognitive impairment

Figure 2. The spectrum of vascular cognitive impairment.

Vascular Cognitive Impairment No-Dementia / Vascular Furthermore, risk of progression towards dementia
Mild Cognitive Impairment (VCIND / VMCI) may vary across individuals classified as VCIND.
Within the VCI spectrum, the earliest symptomatic According to the DSM-5, magnitude of impairment
phase, in which cognitive impairment does not fulfill in neuropsychological testing in this diagnostic
dementia criteria, has been referred to as VCIND5,6 group corresponds to performances between 1 to
or VMCI7-9. Moreover, the 5th edition of the Diagnostic 2 standard deviations (SD) from mean normative
and Statistical Manual of Mental Disorders (DSM-5) values 15. A longitudinal study reported that three
suggested a novel nomenclature, which was endorsed levels of severity could be distinguished within this
by the 11th edition of the International Classification
category: mild VCIND (cognitive deficits up to 1 SD
of Diseases (ICD-11)15,64. In these publications, Vascular
from normative data), moderate VCIND (cognitive
Dementia (VD) was identified as “Major Neurocognitive
impairments of 1.5 SD from normative scores)
Disorder”, whereas VCIND / VMCI was renamed
and severe VCIND (performances up to 2 SD from
as “Mild Neurocognitive Disorder”15,64.
expected scores, considering age and education).
These conditions are associated with substantial risk
of cognitive worsening and progression to dementia65. It has been indicated that higher severity of cognitive
Longitudinal studies reported that 22 to 58% of subjects abnormalities in VCIND correlated with increased odds
diagnosed as VCIND progressed to VD after 2-7 years of transition to dementia70.
of follow-up66-69. In contrast, cognitive recovery was Additionally, the number of vascular-related RF may
detected in 8-45% of the cases, while 38-74% remained impact on cognitive performances, with those with
cognitively stable. Among those in the latest group, more elevated vascular burden presenting poorer scores
30-34% presented subtle cognitive decline, which was in neuropsychological tasks71,72. Table 1 depicts the most
not sufficient for a transition from VCIND to VD66-69. relevant diagnostic criteria for VCIND.

Barbosa BJAP, et al.   Vascular cognitive impairment.   53


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Table 1. Diagnostic Criteria for Vascular Cognitive Impairment, No Dementia (VCIND).


Criteria Description

VCIND
Cognitive impairment in ≥ 1 domain.
Zhao et al., 2010 49
ADLs maintained.
Does not meet accepted criteria for the diagnosis of dementia.

VMCI
Includes the 4 proposed subtypes for MCI: amnesic, amnesic + other domains, non-amnestic single
domain, and non-amnestic multiple domains.
VCCID The VMCI classification must be based on cognitive tests and at least 4 cognitive domains must be
Gorelick et al., 20119 assessed: executive/attention, memory, language, and visuospatial functions.
Classification should be based on presumed decline in cognitive function compared to a previous
baseline and impairment in ≥ 1 cognitive domain.
IADLs can be normal or mildly compromised (regardless of motor/sensory symptoms).

Mild VCD
Evidence of significant cognitive decline in > 1 domain compared to previous level of performance.
Cognitive impairment between 1 and 2 SD below the mean (or between the 3rd and 16th percentile)
VASCOG
(compared to individuals of similar age, sex, education and social-cultural profile).
Sachdev et al., 201414
Fronto-executive deficiencies are more likely to be present.
Preserved IADLs (the individual, although still independent, performs tasks with greater effort and uses
compensation strategies).

Mild VCI
VICCCS
Impairment in ≥ 1 cognitive domain.
Skrobot et al., 20181
BADLs or IADLs maintained or with mild impairment (regardless of motor/sensory symptoms).

CCVND
Cognitive impairment in ≥ 1 cognitive domain.
ABN 2021 Consensus Proposal Cognitive impairment between 1 and 2 SD below the mean (or between the 3rd and 16th percentile).
BADLs maintained (regardless of motor/sensory symptoms).
Preserved IADLs (although with greater effort + compensation strategies).
VCCID: Vascular Contributions to Cognitive Impairment and Dementia; VASCOG: International Society for Vascular Behavioral and Cognitive Disorders; VICCCS: Vascular Impairment of
Cognition Classification Consensus Study; VCIND: vascular cognitive impairment, no dementia; VMCI: vascular mild cognitive impairment; ADL: activities of daily living; BADLs: basic
activities of daily living; IADLs: instrumental activities of daily living; MCI: mild cognitive impairment; mild VCD: mild vascular cognitive disorder; SD: standard deviations.

Vascular Dementia (VD) and its classification the onset of cognitive abnormalities is only mandatory
Current diagnostic criteria for VD require the for the characterization of post-stroke dementia.
occurrence of significant impairment in at least one Other syndromes related to VD have been
cognitive domain (although more domains may be previously described in the literature 73,74. Among
affected), and severe functional disability, including those, small vessel disease associated with Cerebral
difficulties to perform instrumental or basic Amyloid Angiopathy (CAA) deserve to be commented.
activities of daily living1,9,14. Noteworthy, defining In addition to its relationship with cerebral hemorrhage,
thresholds to characterize “severe functional CAA has been linked to AD pathology in post-mortem
disability” might be challenging, since an array analyses75. Cognitive dysfunction may occur in these
of skills are implicated in one’s capacity to exert cases, even without evident brain hemorrhage.
everyday life activities, such as cognitive, behavioral, Some neuroimaging features associated with CAA
sensorial, and motor factors. Table 2 summarizes the include lobar microbleeds, lobar intraparenchymal
main diagnostic criteria for VD. hemorrhage, cortical superficial siderosis, WMH,
Once diagnosed as VD, patients should be convexity subarachnoid hemorrhage, and dilated
investigated for the underlying pathology (Table 3, perivascular spaces74.
Figure 3), and for the level of certainty of the vascular Among genetic syndromes, Cerebral Autosomal
etiology – check the next section for detailed information Dominant Arteriopathy with Subcortical Infarcts
about this theme. Objective 6-month temporal and Leukoencephalopathy (CADASIL), caused by
relationship between a cerebrovascular ictus and mutations in the NOTCH3 gene, ought to be outlined.

54   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

This condition induces deposition of granular may be observed in autosomal recessive mutations
osmiophilic material in the walls of vascular smooth in the HTRA1 gene, which causes Cerebral Autosomal
muscle cells. Clinical manifestations encompass Recessive Arteriopathy with Subcortical Infarcts
early-onset VD, with remarkable decrease in cognitive and Leukoencephalopathy (Cerebral autosomal
speed, executive dysfunction, and attentional deficits, recessive arteriopathy with subcortical infarcts
as well as depression, headache and positive family and leukoencephalopathy – CARASIL). Awareness
history. Brain MRI often evidences substantial white- should be raised to this diagnosis when alopecia and
matter damage, as a result of subcortical infarcts, spondylosis are detected along with the typical signs
and affected temporal poles. A similar phenomenon and symptoms of CADASIL74.

Table 2. Diagnostic Criteria for Vascular Cognitive Impairment – Dementia (VD)


Criteria Description

VD
The diagnosis of VD should be based on a presumption of decline in cognitive function compared to a
VCCID previous baseline and impairment in ≥ 2 cognitive domains sufficient to affect activities of daily living.
Gorelick et al., 20119 The diagnosis of VD must be based on cognitive tests, and at least 4 cognitive domains must be assessed:
executive/attention, memory, language, and visuospatial functions.
Deficits in ADLs must be independent of the motor/sensory sequelae of the vascular event.

Dementia, major VCD


Evidence of significant cognitive decline in > 1 domain compared to previous level of performance.
Cognitive impairment ≥ 2 SD below the average (or below the 3rd percentile) (compared to individuals of
VASCOG
similar age, sex, education and socio-cultural profile)
Sachdev et al., 201414
Fronto-executive deficiencies are more likely to be present.
Enough to interfere with independence (at least requires help with IADLs, e.g., complex tasks such as
managing finances or medications).

Major VCI, VD
VICCCS
Impairment in ≥ 1 cognitive domain.
Skrobot et al., 20181
Significant impairment of IADLs or ABVDs (regardless of motor/sensory symptoms).

Major VCI, VD
Cognitive impairment in ≥ 1 cognitive domain.
ABN 2021 Consensus Proposal
Cognitive impairment ≥ 2 SD below the mean (or below the 3rd percentile).
Significant impairment of IADLs or ADLs (regardless of motor/sensory symptoms).
VCCID: Vascular Contributions to Cognitive Impairment and Dementia; VASCOG: International Society for Vascular Behavioral and Cognitive Disorders; VICCCS: Vascular Impairment of
Cognition Classification Consensus Study; VD: Vascular Dementia; major VCD: major vascular cognitive disorder; VCI: vascular cognitive impairment; ADL: activities of daily living; BADLs:
basic activities of daily living; IADLs: instrumental activities of daily living; SD: standard deviations.

Table 3. Main forms of major VCI / Vascular Dementia, adapted from1.


Classification Description

Presence of new, sudden or subacute cognitive deficit up to 6 months after ischemic or hemorrhagic
stroke. It may be due to different cerebrovascular patterns (e.g., multiple cortico-subcortical infarctions,
Post-stroke dementia
strategic lesions, subcortical vascular dementia, etc.). The temporal relationship between the vascular
event and cognitive decline differentiates this form from VD.

Broad term that encompasses cognitive decline phenotypes combined between VCI and
Mixed dementias neurodegenerative diseases (e.g. VCI-AD, VCI-LBD etc). It is recommended to specify which underlying
pathology is suspected, avoiding the less specific term “mixed dementia.”

Small vessel cerebrovascular disease is the main cause in this group, mainly due to lacunar infarcts and white
Subcortical ischemic vascular dementia
matter lesions. It encompasses the phenotypes described as Binswanger’s Disease and the lacunar state.

Cortical multiple infarct dementia A group characterized by the presence of multiple cortical infarcts and their likely contribution to dementia.

Possible - more appropriate term if neuroimaging is unavailable.


Level of certainty
Probable - in the presence of compatible CT or MR. MR is the method of choice.
VCI: vascular cognitive impairment; VD: vascular dementia; AD: Alzheimer’s disease; LBD: Lewy Body dementia; CT: computer tomography; MR: magnetic resonance.

Barbosa BJAP, et al.   Vascular cognitive impairment.   55


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Post-stroke Subcortical Multi-infarct


Mixed pathologies
dementia ischemic VaD dementia

VaD-AD VaD-DLB VaD-X

VaD: vascular dementia; AD: Alzheimer’s disease; LBD: Lewy Body Dementia.
Figure 3. Classification of forms of vascular dementia according to the VICCCS (Vascular Impairment of Cognition Classification Consensus Study) (1).
Each form will be further stratified into possible and probable (Table 3). Mixed forms can occur in all of the above syndromes, and the suspected
neurodegenerative syndrome should be detailed (VD-AD and VD-LBD were used as examples, with X denoting other possible associations).

CLINICAL MANIFESTATIONS AND DIAGNOSTIC A detailed protocol must be followed to establish


WORKUP OF VCI/DEMENTIA diagnosis, including several steps, i.e., clinical history
T he clinical manifestations of VCI/dementia (clinical, cognitive, neurologic, psychiatric), physical
include cognitive impairment, functional decline, examination (clinical-cardiological, neurologic),
neuropsychiatric symptoms, neurological manifestations neuropsychological assessment (screening, comprehensive
and autonomic dysfunction, in variable proportions and assessment), functional assessment, neuropsychiatric
associations, according to the type, location, number and evaluation, and diagnostic exams (neuroimaging,
extent of the lesions10,11. laboratory testing, among others)9,23,32 (Figure 4).

Anamnesis (global, cognitive, neurological, psychiatric)

Physical Examination (clinical, cardiological, neurovascular)

Secondary level Tertiary level


Primary care
(general neurologist, (research centers,
psychiatrists, geriatricians) specialist in cognitive
and behavioral neurology)

Cognitive and Cognitive screening Comprehensive testing


Expanded evaluation
function evaluation (MoCa or MMSE + BB + SVF + CDT) (MoCa, MMSE + CAMCOG subscale, ACE-R)
(NPS, cognitive protocol)
Depression screening Depression screening
NPI-Q, CDR CES-D ou GDS)

Complementary CSF studies Functional neuroimaging


BAsic laboratory tests
tests Structural neuroimaging Genetic studies
Basic neuroimaging*
Vascular neuroimaging Biomarkers
Biomarkers

MoCa: Montreal cognitive assessment; MMSE: Mini-Mental State Examination; BB: Brief Battery of Cognitive Screening; SVF: semantic verbal fluency; TDR: clock design test; CAMCOG:
Cambridge Cognitive Examination; ACE-R: Addenbrooke’s Cognitive Examination-Revised; NPS: neuropsychological assessment; NPI-Q: Neuropsychiatric Inventory Questionnaire; CDR:
clinical dementia rating; CES-D: Center for Epidemiologic Studies - Depression; GDS: Geriatric Depression Scale.
Figure 4. Flowchart proposed for the assessment and investigation of patients with suspected VCI / VD.
*Basic neuroimaging refers at least to Computed Tomography of the Skull.

56   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Which components of the history are essential in to the corresponding MMSE ones86. A meta-analysis
evaluating patients with suspected VCI and dementia?76 carried out by the Cochrane showed sensitivity
The patient’s history is essential for characterizing of 0.76 and specificity of 0.73 of the Mini-Cog for
cognitive deficits, generating a differential diagnosis, the diagnosis of dementia in general 87 . For the
and determining the cause of dementia. The best way to diagnosis of dementia in the primary care settings,
do this is to identify medical, neurologic and psychiatric the number of studies evaluating the accuracy of the
symptoms as clues to the probable cause of the Mini-Cog was limited.
cognitive changes, establishing the order of appearance, Despite the large amount of short cognitive
the severity and the associated features. Ideally in VD screening tools, few are valid for patients with suspected
the loss of function should be temporally correlated with VCI. A systematic review on screening tests for
cerebrovascular events. A reliable relative/informant the identification of VCI88, the MoCA, the MMSE,
plays an important role in providing information since the Brief Memory and Executive Test – BMET –
cognitive dysfunction may impair the patient’s ability and different versions of the Clock Design Test were
to report accurately. the most widely validated instruments. Based on
VD should be suspected in any patient presenting available evidence, the authors concluded that the
cerebrovascular RF, even if the neurologic examination MoCA was the most accurate and reliable instrument,
does not suggest stroke. A stepwise deterioration may however this finding still needs further validation
be observed. It may be present in patients with silent in our population. The BMET has already been adapted
stroke, in those with several small strokes, or in those for Brazilian individuals89.
with severe diffuse subcortical cerebrovascular disease.
Cognitive assessment tests in the context of VCI
Which methods clinicians should use to detect VCI/VD? Cognitive screening tests used in the assessment
At first, the indiscriminate assessment of elderly of AD, particularly the MMSE, are not ideal for
individuals for dementia is not recommended 77,78. vascular cognitive impairment. Those including the
In the evaluation of older patients for dementia, assessment of frontal, executive, and subcortical
clinicians should use a standardized screening tool, functions are preferred. Modifications of tests originally
along with a brief patient history obtained from developed for AD, such as the Vascular Version of
a reliable informant (a person who is directly in contact the Alzheimer’s Disease Assessment Scale-Cognitive
with the patient). Subscale (VADAS-Cog), may be useful90. Some of them
may be able to differentiate AD from VCI, but even with
Screening tools the use of biomarkers to eliminate the presence of AD
The screening instrument must be easy to use, pathology (e.g., a negative amyloid PET), some overlap
highly sensitive, widely available and supported by may persist between the cognitive changes of vascular
populational data76. The Mini-Mental State Examination cognitive impairment and AD91. Validation data exist
(MMSE) is widely used and contributes to the diagnosis for the MoCA92,93 and for the Addenbrooke Cognitive
of dementia in low prevalence settings. However, Exam Revised Version94, both already validated for the
it should not be used in isolation to confirm or exclude Brazilian population95-97. There are some suggestions
the disease79. To improve diagnostic accuracy in low for protocols of cognitive assessment for the detection
educated populations, we suggest the association of VCI/VD that have been published in the last two
of Brief Cognitive Battery 80-82, which includes an decades and are summarized in Suplementary Material.
interference with a semantic verbal fluency (animals) In Brazil, a previous review on VD was published
and the clock drawing test (CDT). Other options by the Scientific Department of Cognitive Neurology
are the Mini-Cog 83 and the Montreal Cognitive from the Brazilian Academy of Neurology (ABN)10.
Assessment (MoCA)84. In this document, the expert panel emphasized
The Mini-Cog has the benefit of brevity and the that the pattern of cognitive changes was highly
MoCA has the best sensitivity but lower specificity85. variable, requiring sufficient sensitivity from the
The MoCA was originally developed for the detection neuropsychological protocols to detect a wide range of
of MCI and may be difficult for people with moderate domains, mainly executive function. The selected tests
or advanced dementia, as well as for populations must meet criteria of frequency and validity, be freely
with low educational backgrounds. For those whose available, and be well known and sensitive to detect
previous cognitive function was measured with cognitive decline. The protocols must be broad, easy to
the MMSE, there is a tool that links the MoCA scores administer and relatively brief32. The recommendations

Barbosa BJAP, et al.   Vascular cognitive impairment.   57


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

included a brief screening protocol for VCI/VD, executive/activation, language, visuospatial and
consisting of the MMSE, the semantic verbal fluency memory. In addition, tests were selected to examine
test (animals) and the CDT10. The authors also included changes in behavior and mood. The other two protocols
a broader and time-consuming protocol composed of were selected from within the 60-minute protocol to
a wider range of tests (Appendix). This version included be used as a clinical screening tool for suspected VCI
the Cambridge Cognitive Examination (CAMCOG) scale patients. The 5-minute protocol was projected for
with the global and subscale scores, which has been potential use by primary care physicians, nurses,
adapted and validated for the Brazilian population98,99. and other health care professionals. The 5-minute
The National Institute for Neurological Disorders protocol was also designed for large epidemiological
and Stroke (NINDS) and the Canadian Stroke Network studies or clinical trials where sensitivity and
established a working group to define criteria for VCI23. ease of administration are especially important.
In this document, the Neuropsychological Working In addition, once validated the 5-minute protocol
Group proposed three separate protocols which were was also designed to be administered over the phone.
recommended for multicenter investigations with Most of the tests included in these protocols are
VCI patients, one requiring at least 60 minutes, available in Brazil, especially the 5-minute version
a second of 30 minutes and a third of five minutes that is sourced from MoCA.
(Suplementary Material). The longer one, 60 minutes, Based on all the above data, this panel recommends
was developed for use in studies requiring an analysis the use of screening/cognitive assessment tests for the
of cognitive skills by domain, thus the protocol detection of VCI according to the level of health care
contained recommended tests in four domains: in which the patient is inserted (Table 4).

Table 4. Cognitive screening recommendation on vascular cognitive impairment stratified by health care levels.

Health level Cognitive screening recommendation for CCV

General practitioners and professionals from the Family Health Strategy can use screening instruments such
Primary attention as MoCA, or alternatively the MMSE associated with the clock drawing test and semantic verbal fluency (animals)
and screening for depressive symptoms.

Specialist physicians (neurologists, geriatricians, and psychiatrists) who receive patients referred from primary
Secondary Care care can use a broader protocol that includes global function tests (MoCA or MMSE) associated with CAMCOG
subscales, or Addenbrooke battery (ACE-R) and screening for depressive symptoms.

Specialist physicians at referral centers can use expanded assessment including, in addition to a cognitive protocol,
Tertiary care the assessment of neuropsychiatric symptoms (NPI-Q), assessment of severity of dementia (CDR), and screening
for depressive symptoms (CES-D or GDS).

Assessment of functionality home/hobbies and personal care) (CDR FUNC) of


The use of the Clinical Dementia Rating scale (CDR) the CDR presented correlation with the Pfeffer’s
presents some difficulties for cases of vascular nature, Functional Activities Questionnaire (FAQ), CLOX 2,
although the scale has been validated for such cases working memory and abstraction101.
in the Brazilian settings 100. CDR is strongly based A study with the CDR scale defined mild VCI as a MCI
on memory impairment. However, other domains, status of VCI or a CDR 0.5 status with cardiovascular
such as executive function, are especially important disease. Thus, for the assessment of daily life the
in vascular cases. information from caregivers is necessary, especially
A study of subcortical VCI (70% of cases with in the domains (“boxes”) of “community affairs”,
CDR 0.5) showed, in cases of moderate and severe “home and hobbies” and “personal care” 25. Thus,
subcortical lesions, that the sum of the “functional” the use of the CDR scale can be accepted, as long as the
boxes (judgment/problem solving, community affairs, sum of the functional boxes is considered valid.

58   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Laboratory diagnosis A recent systematic review on VMCI (subcortical),


No laboratory test or biomarker is specific for VCI. analyzing studies that assessed lesions qualitatively
Conversely, routine exams can assess comorbidities (WM lesion extension) and semi quantitatively
or RF for cognitive decline. Evaluation of blood (number of lacunes), showed the presence of moderate
count, serology, glucose levels, B12, thyroid, kidney, to severe vascular lesion load106.
and liver function, is often required – as detailed The application of visual scales can help the clinician
in a specific article of the present consensus. in the differential diagnosis, in the anatomical-clinical
A metabolic profile with cholesterol and triglycerides correlation, especially with cognitive alterations,
is also important. AD biomarkers (beta-amyloid, as well as in therapeutic monitoring 107 (Table 5)
tau, and phospho-tau) in the CSF may play a role in (Figure 5). Conversely, the characterization of CVD
through neuroimaging also usually contributes to
selected cases when the presence of mixed pathology
the systemic treatment of the patient. The occurrence
is questionable 102 . O ther CSF measurements
of WM lesions, either periventricular (PVWM) or diffuse
(proteins, electrophoresis) may help to differentiate
(DWM), may exhibit differences in etiopathogenesis;
inf lammator y causes or suggest a blood-brain
for example, a combination of granular ependymitis
barrier dysfunction (e.g., vasculitis or demyelinating (ependymitis granularis) and axonal demyelination
diseases), albeit they are not part of the routine may be related to PVWM 112 , whereas gliosis of
assessment of suspected cases of VCI103. the subependymal areas (subependymal gliosis)
combined with chronic small vessel ischemia seems
Neuroimaging diagnosis to contribute in more significant proportion to the
Neuroimaging is critical to detecting CVD causing occurrence of DWM113.
VCI. MRI is the best and most accurate technique to MRI can offer advantages in observing PVWH and
visualize lesions produced by large vessels (infarctions), DWM through T2 and FLAIR sequences; conversely,
lesions resulting from small vessel diseases (WMH, both CT and MRI seem to identify cortical and lacunar
small subcor tical infarcts, lacunae, enlarged infarcts in a similar way106. Evidence of hyperintensity,
perivascular spaces, cerebral microhemorrhages). once nonspecific, is associated with CVD and AD
Cerebral hemorrhages (lobar, deep) can be well pathology; the volume of WM alterations is regarded
visualized through computed tomography (CT)25. as a possible independent marker of beta-amyloid
Currently, there are no criteria to define the protein accumulation114.
necessar y load of vascular lesions detected by The finding of lesions caused by multiple infarctions
neuroimaging to confirm the presence of VMCI, can direct clinical investigation towards the occurrence
lacking a cutoff point for such definition 104 . of coagulopathies, infectious and parasitic diseases,
Macroscopic findings of conventional neuroimaging alcoholism, and heart failure115,116. Perivascular disease,
(FLAIR sequences, CT) often partially explain the seen as microintensities at the cortico-subcortical
clinical expression (phenotype) of VD and usually junctions, has been independently associated with
represent heterogeneous pathological alterations52. an increased risk for cognitive decline117. In Brazil
and most countries of Latin America, the high
Thus, the presence of hidden aspects (“invisible
occurrence of subcortical ischemic vascular disease
changes”) could contribute to clinical expression,
is usually associated with late diagnosis of systemic
which can be verified by more advanced techniques,
hypertension, diabetes and dyslipidemia, as reported
such as diffusion tensor imaging (DTI) or other
by the ELSA study118.
techniques capable of identifying areas of “WM In conclusion, the evolution of the VCI concept
of normal appearance” 51,105. In addition, advanced from preclinical stages to VMCI and VD turns
MRI techniques with analysis of structural and diagnostic assessment into a challenge for the
functional brain connectivity can contribute to multidisciplinary team. The patient with suspected
possible investigations of how changes in more VMCI should be evaluated from primary care by
complex brain networks can explain the diversity of general practitioners, with complementary work up
clinical presentations in these conditions, even in being carried out at the secondary and tertiary levels
cases with anatomical changes in similar conventional in a horizontal manner, according to the need for
sequences 16, which can optimize the anatomical- more advanced instruments, particularly advanced
clinical correlation in the future. neuroimaging techniques.

Barbosa BJAP, et al.   Vascular cognitive impairment.   59


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Table 5. Main visual scales in CVD.


Scale [reference] Brain region [indication] Method [scores]

Periventricular hyperintensities:
0=absent
1= “hoods” or thin coat
2=smooth halo
White matter hyperintensities 3=periventricular hyperintensities extending into deep white matter
Fazekas et al.114
periventricular and deep Deep white substance:
0=absent
1=point-like foci
2=initial confluence
3=large confluent areas

0=absent
1=[mild] punctiform lesions with maximum unit diameter below 10 mm
and areas of clustered lesions smaller than 20 mm
Deep white matter and subcortical
Modified Fazekas (LADIS)115 2=[moderate] single lesions between 10-20 mm in diameter, areas of
lesions (DSWM)
clustered lesions greater than 20 mm in diameter, no more than connecting
bridges between individual lesions 3=[severe] single lesions or confluent
areas of hyperintensities 20 mm or more in diameter

PVH, DWMH, basal and infratentorial PVH (0-6), WMH (0-24), basal and infratentorial ganglia (both 0-24)
Scheltens et al.116
ganglia hyperintensities Total=0 to 84

White matter lesions:


0 = absence
1= focal lesions
2=initial confluent lesions
3= diffuse involvement of the entire region
Hyperintensities of hemispheres
ARWMC scale117 Basal ganglia:
and basal ganglia
0=absence
1=focal lesions (≥5 mm)
2= >1 focal lesion
3=confluent lesions
Total = 0 to 30

Figure 5. Evaluation of White Substance Hyperintensities according to the Fazekas visual scale107.

60   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

ACKNOWLEDGEMENTS
PC, LCS and RN are funded by CNPq, Brazil (bolsa de CKS, FTM, EE, MLFC: drafting of the manuscript; BJAPB,
produtividade em pesquisa). FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC, PHFB, RN,
Authors’ contributions. BJAPB, JISN, GSA, FKS, CKS, FTM, SMDB: critical revision of the manuscript for important
EE, MLFC, JS, LPS: concept; BJAPB, JISN, GSA, FKS, intellectual content.

REFERENCES
1. Skrobot OA, Black SE, Chen C, DeCarli C, Erkinjuntti T, Ford GA, et al. 19. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S,
Progress toward standardized diagnosis of vascular cognitive impairment: et al. Dementia prevention, intervention, and care: 2020 report of the
Guidelines from the Vascular Impairment of Cognition Classification Lancet Commission. Lancet. 2020;396(10248):413-46. doi:10.1016/
Consensus Study. Alzheimers Dement. 2018;14(3):280-92. doi:10.1016/ S0140-6736(20)30367-6.
j.jalz.2017.09.007. 20. Jørgensen IF, Aguayo-Orozco A, Lademann M, Brunak S. Age-stratified
2. Hachinski V. Vascular dementia: a radical redefinition. Dementia. 1994;5(3-4): longitudinal study of Alzheimer’s and vascular dementia patients.
130-2. doi:10.1159/000106709. Alzheimers Dement. 2020;16(6):908-17. doi:10.1002/alz.12091.
3. Loeb C. Vascular dementia. In: Handbook of clinical neurology. 21. Stephens S, Kenny RA, Rowan E, Allan L, Kalaria RN, Bradbury M, et al.
Amsterdam: Elsevier; 1985. p. 353-69. (Neurobehavioral Disorders; vol. 2). Neuropsychological characteristics of mild vascular cognitive impairment
4. Hachinski P. Preventable senility: a call for action against the vascular and dementia after stroke. Int J Geriat Psychiatry. 2004;19(11):1053-7.
dementias. Lancet. 1992;340(8820):645-8. doi:10.1016/0140- doi:10.1002/gps.1209.
6736(92)92177-h. 22. Rasquin SMC, Oostenbrugge RJ, Verhey FRJ, Lodder J. Vascular mild
5. Ebly EM. Cognitive Impairment in the Nondemented Elderly: Results From cognitive impairment is highly prevalent after lacunar stroke but does not
the Canadian Study of Health and Aging. Arch Neurol. 1995;52(6):612-9. increase over time: a 2-year follow-up Study. Dement Geriatr Cogn Disord.
doi:10.1001/archneur.1995.00540300086018. 2007;24(5):396-401. doi:10.1159/000109747.
6. Rockwood K, Wentzel C, Hachinski V, Hogan DB, MacKnight C, McDowell I. 23. Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL,
Prevalence and outcomes of vascular cognitive impairment. Neurology. Black SE, et al. National Institute of Neurological Disorders and
2000;54(2):447-51. doi:10.1212/wnl.54.2.447. Stroke-Canadian Stroke Network Vascular Cognitive Impairment
7. Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins PV, Harmonization Standards. Stroke. 2006;37(9):2220-41. doi:10.1161/
et al. Current Concepts in Mild Cognitive Impairment. Arch Neurol. 01.STR.0000237236.88823.47.
2001;58(12):1985-92. doi:10.1001/archneur.58. 24. Brucki SMD, Machado MF, Rocha MSG. Vascular Cognitive Impairment
8. Meyer JS, Xu G, Thornby J, Chowdhury MH, Quach M. Is Mild Cognitive (VCI) after non-embolic ischemic stroke during a 12-month follow-up
Impairment Prodromal for Vascular Dementia Like Alzheimer’s Disease? in Brazil. Dement Neuropsychol. 2012;6:164-9. doi:10.1590/
Stroke. 2002;33(8):1981-5. doi:10.1161/01.str.0000024432.34557. S1980-57642012DN06030009.
9. Gorelick PB, Scuteri A, Black SE, DeCarli C, Greenberg SM, Iadecola C, 25. Meguro K, Dodge HH. Vascular Mild Cognitive Impairment: Identifying
et al. Vascular Contributions to Cognitive Impairment and Dementia: Disease in Community-Dwelling Older Adults, Reducing Risk Factors,
A Statement for Healthcare Professionals From the American Heart and Providing Support. The Osaki-Tajiri and Kurihara Projects.
Association/American Stroke Association. Stroke. 2011;42(9):2672-713. Anstey K, Peters R, editors. J Alzheimers Dis. 201913;70(s1):S293-302.
doi:10.1161/STR.0b013e3182299496. doi:10.3233/JAD-180899.
10. Engelhardt E, Tocquer C, André C, Moreira DM, Okamoto IH, Cavalcanti JLS. 26. Hughes TF, Liu A, Jacobsen E, Rosano C, Berman SB, Chang CCH, et al.
Vascular dementia: Cognitive, functional and behavioral assessment. Exercise and the Risk of Mild Cognitive Impairment: Does the Effect
Recommendations of the Scientific Department of Cognitive Neurology and Depend on Vascular Factors? Alzheimer Dis Assoc Disord. 2021;35(1):30-5.
Aging of the Brazilian Academy of Neurology. Part II. Dement Neuropsychol. doi:10.1097/WAD.0000000000000410.
2011;5(4):264-74. doi:10.1590/S1980-57642011DN05040004. 27. Alonso A, Knopman DS, Gottesman RF, Soliman EZ, Shah AJ, O’Neal WT,
11. Engelhardt E, Tocquer C, André C, Moreira DM, Okamoto IH, Cavalcanti JLS. et al. Correlates of Dementia and Mild Cognitive Impairment in Patients
Vascular dementia: Diagnostic criteria and supplementary exams. With Atrial Fibrillation: The Atherosclerosis Risk in Communities
Recommendations of the Scientific Department of Cognitive Neurology and Neurocognitive Study (ARIC–NCS). J Am Heart Assoc. 2017;6(7).
Aging of the Brazilian Academy of Neurology. Part I. Dement Neuropsychol. doi:10.1161/JAHA.117.006014.
2011;5(4):251-63. doi:10.1590/S1980-57642011DN05040003. 28. World Health Organization. Risk reduction of cognitive decline and
12. Brucki SMD, Ferraz AC, Freitas GR, Massaro AR, Radanovic M, dementia: WHO guidelines [Internet]. 2019 [citado em 26 jul 2022].
Schultz RR. Treatment of vascular dementia. Recommendations of the Disponível em: https://www.who.int/publications/i/item/9789241550543
Scientific Department of Cognitive Neurology and Aging of the Brazilian 29. Suemoto CK, Ferretti-Rebustini REL, Rodriguez RD, Leite REP,
Academy of Neurology. Dement Neuropsychol. 2011;5(4):275-87. Soterio L, Brucki SMD, et al. Neuropathological diagnoses and clinical
doi:10.1590/S1980-57642011DN05040005. correlates in older adults in Brazil: A cross-sectional study. PLoS Med.
13. Skrobot OA, O’Brien J, Black S, Chen C, DeCarli C, Erkinjuntti T, et al. 2017;14(3):e1002267. doi:10.1371/journal.pmed.1002267.
The Vascular Impairment of Cognition Classification Consensus Study. 30. Gorelick PB, Counts SE, Nyenhuis D. Vascular cognitive impairment and
Alzheimers Dement. 2017;13(6):624-33. doi:10.1016/j.jalz.2016.10.007. dementia. Biochim Biophys Acta. 2016;1862(5):860-8. doi:10.1016/
14. Sachdev P, Kalaria R, O’Brien J, Skoog I, Alladi S, Black SE, et al. j.bbadis.2015.12.015.
Diagnostic Criteria for Vascular Cognitive Disorders: A VASCOG 31. Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J,
Statement. Alzheimer Dis Assoc Disord. 2014;28(3):206-18. doi:10.1097/ Ames D, et al. Dementia prevention, intervention, and care. Lancet.
WAD.0000000000000034. 2017;390(10113):2673-734. doi:10.1016/S0140-6736(17)31363-6.
15. American Psychiatric Association. Diagnostic and statistical manual of 32. Iadecola C, Duering M, Hachinski V, Joutel A, Pendlebury ST, Schneider JA,
mental disorders. 5a ed. Washington (DC): APA; 2013. doi:10.1176/ et al. Vascular Cognitive Impairment and Dementia. J Am Coll Cardiol.
appi.books.9780890425596.dsm05. 2019;73(25):3326-44. doi:10.1016/j.jacc.2019.04.034.
16. Ter Telgte A, Leijsen EMC, Wiegertjes K, Klijn CJM, Tuladhar AM, Leeuw 33. Grinberg LT. Vascular dementia: current concepts and nomenclature
FE. Cerebral small vessel disease: from a focal to a global perspective. harmonization. Dement Neuropsychol. 2012;6(3):122-6. doi:10.1590/
Nat Rev Neurol. 2018;14(7):387-98. doi:10.1038/s41582-018-0014-y. S1980-57642012DN06030002.
17. Calil V, Elliott E, Borelli WV, Barbosa BJAP, Bram J, Silva FO, et al. Chal- 34. Skrobot OA, Attems J, Esiri M, Hortobágyi T, Ironside JW, Kalaria RN,
lenges in the diagnosis of dementia: insights from the United Kingdom- et al. Vascular cognitive impairment neuropathology guidelines (VCING):
-Brazil Dementia Workshop. Dement Neuropsychol. 2020;14(3):201-8. the contribution of cerebrovascular pathology to cognitive impairment. Brain.
doi:10.1590/1980-57642020dn14-030001. 2016;139(11):2957-69. doi:10.1093/brain/aww214.
18. Rizzi L, Rosset I, Roriz-Cruz M. Global epidemiology of dementia: 35. Jellinger KA. Current Pathogenetic Concepts of Vascular Cognitive
Alzheimer’s and vascular types. Biomed Res Int. 2014;2014:908915. Impairment. J Neurol Neurol Sci Disord. 2016;2(1):10-6. doi:10.17352/
doi:10.1155/2014/908915. jnnsd.000009.

Barbosa BJAP, et al.   Vascular cognitive impairment.   61


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

36. Jokinen H, Koikkalainen J, Laakso HM, Melkas S, Nieminen T, Brander A, 59. Hu HY, Ou YN, Shen XN, Qu Y, Ma YH, Wang ZT, et al. White matter
et al. Global Burden of Small Vessel Disease-Related Brain Changes on hyperintensities and risks of cognitive impairment and dementia:
MRI Predicts Cognitive and Functional Decline. Stroke. 2020;51(1):170-8. A systematic review and meta-analysis of 36 prospective studies. Neurosci
doi:10.1161/STROKEAHA.119.026170. Biobehav Rev. 2021;120:16-27. doi:10.1016/j.neubiorev.2020.11.007.
37. De Luca C, Virtuoso A, Maggio N, Papa M. Neuro-Coagulopathy: Blood 60. Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB,
Coagulation Factors in Central Nervous System Diseases. Int J Mol Sci. et al. NIA-AA Research Framework: Toward a biological definition of
2017;18(10):2128. doi:10.3390/ijms18102128. Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535-62. doi:10.1016/
38. Rajani RM, Williams A. Endothelial cell-oligodendrocyte interactions j.jalz.2018.02.018.
in small vessel disease and aging. Clin Sci. 2017;131(5):369-79. 61. Hachinski V, Einhäupl K, Ganten D, Alladi S, Brayne C, Stephan BCM,
doi:10.1042/CS20160618. et al. Preventing dementia by preventing stroke: The Berlin Manifesto.
39. Ungvari Z, Tarantini S, Kiss T, Wren JD, Giles CB, Griffin CT, et al. Endothelial Alzheimers Dement. 2019;15(7):961-84. doi:10.1016/j.jalz.2019.06.001.
dysfunction and angiogenesis impairment in the ageing vasculature. 62. Azarpazhooh MR, Hachinski V. Vascular cognitive impairment: A preventable
Nat Rev Cardiol. 2018;15(9):555-65. doi:10.1038/s41569-018-0030-z. component of dementia. In: Dekosky ST, Asthana S. Handbook of Clinical
40. Engelhardt E. Brown-Séquard: On neural networks and brain localization Neurology: Geriatric Neurology. Vol. 167. Amsterdam: Elsevier; 2019.
of functions. Dement Neuropsychol. 2014;8(1):79-82. doi:10.1590/ Chapter 20. doi:10.1016/B978-0-12-804766-8.00020-0
S1980-57642014DN81000012. 63. World Health Organization. ICD-11 for Mortality and Morbidity Statistics
41. Jellinger KA. Pathology and pathogenesis of vascular cognitive [Internet]. 2016 [citado em 18 fev 2021]. Disponível em: https://icd.who.int/
impairment – a critical update. Front Aging Neurosci. 2013;5:1-19. browse11/l-m/en
doi:10.3389/fnagi.2013.00017 64. Stephan BC, Matthews FE, Khaw KT, Dufouil C, Brayne C. Beyond mild
42. Shin M, Sohn MK, Lee J, Kim DY, Lee SG, Shin YI, et al. Effect of cognitive impairment: vascular cognitive impairment, no dementia (VCIND).
Cognitive Reserve on Risk of Cognitive Impairment and Recovery After Alzheimers Res Ther. 2009;1(1):4. doi:10.1186/alzrt4.
Stroke: The KOSCO Study. Stroke. 2020;51(1):99-107. doi:10.1161/ 65. Zanetti M, Ballabio C, Abbate C, Cutaia C, Vergani C, Bergamaschini L.
STROKEAHA.119.026829. Mild Cognitive Impairment Subtypes and Vascular Dementia in Community-
43. Mesulam MM, editor. Principles of behavioral and cognitive neurology. Dwelling Elderly People: A 3-Year Follow-Up Study. J Am Geriatr Soc.
2nd ed. Oxford: Oxford University Press; 2000. 2006;54(4):580-6. doi:10.1111/j.1532-5415.2006.00658.x.
44. Zekry D, Duyckaerts C, Belmin J, Geoffre C, Herrmann F, Moulias R, 66. Wentzel C, Rockwood K, MacKnight C, Hachinski V, Hogan DB, Feldman H,
et al. The vascular lesions in vascular and mixed dementia: the weight et al. Progression of impairment in patients with vascular cognitive
of functional neuroanatomy. Neurobiology of Aging. 2003;24(2):213-9. impairment without dementia. Neurology. 2001;57(4):714-6. doi:10.1212/
doi:10.1016/s0197-4580(02)00066-0. wnl.57.4.714.
45. Staekenborg SS, Straaten ECW, Flier WM, Lane R, Barkhof F, 67. Madureira S, Verdelho A, Moleiro C, Santos C, Scheltens P, Gouw A,
Scheltens P. Small vessel versus large vessel vascular dementia: Risk et al. White Matter Changes and Cognitive Decline in a Ten-Year Follow-Up
factors and MRI findings. J Neurol. 2008;255(11):1644-51. doi:10.1007/ Period: A Pilot Study on a Single-Center Cohort from the Leukoaraiosis
and Disability Study. Dement Geriatr Cogn Disord. 2016;41(5-6):303-13.
s00415-008-0944-1.
doi:10.1159/000447121.
46. Straaten ECW, Scheltens Ph, Barkhof F. MRI and CT in the diagnosis of
68. Madureira S. Neuropsychological contribution to the study of White
vascular dementia. Journal of the Neurological Sciences. 2004;226(1-2):
Matter Changes: a 10-year longitudinal study [tese na Internet]. Lisboa:
9-12. doi:10.1016/j.jns.2004.09.003.
Universidade de Lisboa; 2017 [citado em 23 maio 2021]. Disponível
47. Sudo FK, Amado P, Alves GS, Laks J, Engelhardt E. A continuum of
em: https://repositorio.ul.pt/bitstream/10451/28880/1/ulsd730661_td_
executive function deficits in early subcortical vascular cognitive impair-
Sofia_Madureira.pdf
ment: A systematic review and meta-analysis. Dement Neuropsychol.
69. Palmer K, Wang HX, Bäckman L, Winblad B, Fratiglioni L. Differential
2017;11(4):371-80. doi:10.1590/1980-57642016dn11-040006.
Evolution of Cognitive Impairment in Nondemented Older Persons: Results
48. Tullberg M, Fletcher E, DeCarli C, Mungas D, Reed BR, Harvey DJ, et al.
From the Kungsholmen Project. Am J Psychiatry. 2002;159(3):436-42.
White matter lesions impair frontal lobe function regardless of their location.
doi:10.1176/appi.ajp.159.3.436.
Neurology. 2004;63(2):246-53. doi:10.1212/01.wnl.0000130530.55104.b5.
70. Kopchak OO, Bachinskaya NY, Pulyk OR. Vascular risk factors and
49. Zhao QL, Zhou Y, Wang YL, Dong KH, Wang YJ. A new diagnostic algorithm
cognitive functions in the patients with cerebrovascular disease.
for vascular cognitive impairment: the proposed criteria and evaluation of its Wiad Lek. 2020;73(10):2250-4.
reliability and validity. Chin Med J (Engl). 2010;123(3):311-9. 71. W iederkehr S, Laurin D, Simard M, Verreault R, Lindsay J.
50. Gouw AA, Seewann A, Flier WM, Barkhof F, Rozemuller AM, Scheltens P, Vascular Risk Factors and Cognitive Functions in Nondemented
et al. Heterogeneity of small vessel disease: a systematic review of Elderly Individuals. J Geriatr Psychiatry Neurol. 2009;22(3):196-206.
MRI and histopathology correlations. J Neurol Neurosurg Psychiatry. doi:10.1177/0891988709335797.
2011;82(2):126-35. doi:10.1136/jnnp.2009.204685. 72. O’Brien JT, Erkinjuntti T, Reisberg B, Roman G, Sawada T, Pantoni L,
51. Min ZG, Shan HR, Xu L, Yuan DH, Sheng XX, Xie WC, et al. Diffusion et al. Vascular cognitive impairment. Lancet Neurol. 2003;2(2):89-98.
tensor imaging revealed different pathological processes of white matter doi:10.1016/s1474-4422(03)00305-3.
hyperintensities. BMC Neurol. 2021;21(1):128. doi:10.1186/s12883- 73. Graff-Radford J. Vascular Cognitive Impairment. Continuum (Minneap
021-02140-9. Minn). 2019;25(1):147-64. doi:10.1212/CON.0000000000000684.
52. McAleese KE, Alafuzoff I, Charidimou A, De Reuck J, Grinberg LT, 74. Arvanitakis Z, Leurgans SE, Wang Z, Wilson RS, Bennett DA, Schneider JA.
Hainsworth AH, et al. Post-mortem assessment in vascular dementia: Cerebral amyloid angiopathy pathology and cognitive domains in older
advances and aspirations. BMC Med. 2016;14(1):129. doi:10.1186/ persons. Ann Neurol. 2011;69(2):320-7. doi:10.1002/ana.22112.
s12916-016-0676-5. 75. Oh ES, Rabins PV. Dementia. Ann Intern Med. 2019;171(5):ITC33-48.
53. Grinberg LT, Heinsen H. Toward a pathological definition of vascular doi:10.7326/AITC201909030.
dementia. J Neurol Sci. 2010;299(1-2):136-8. doi:10.1016/j.jns.2010.08.055. 76. Ramos AM, Stein AT, Castro Filho ED, Chaves MLF, Okamato I, Nitrini R.
54. Kalaria RN. Neuropathological diagnosis of vascular cognitive impairment Demência do Idoso: diagnóstico na Atenção Primária. São Paulo:
and vascular dementia with implications for Alzheimer’s disease. Associação Médica Brasileira; 2009.
Acta Neuropathol. 2016;131(5):659-85. doi:10.1007/s00401-016-1571-z. 77. Patnode CD, Perdue LA, Rossom RC, Rushkin MC, Redmond N,
55. Rockwood K, Davis H, MacKnight C, Vandorpe R, Gauthier S, Guzman A, Thomas RG, et al. Screening for Cognitive Impairment in Older Adults:
et al. The Consortium to Investigate Vascular Impairment of Cognition: An Evidence Update for the U.S. Preventive Services Task Force [Internet].
Methods and First Findings. Can J Neurol Sci. 2003;30(3):237-43. Rockville (MD): Agency for Healthcare Research and Quality (US); 2020
doi:10.1017/s0317167100002663. [citado em 3 jul 2021]. Evidence Synthesis, No. 189. Disponível em:
56. Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, Frayne R, http://www.ncbi.nlm.nih.gov/books/NBK554654/
et al. Neuroimaging standards for research into small vessel disease 78. Creavin ST, Wisniewski S, Noel-Storr AH, Trevelyan CM, Hampton T,
and its contribution to ageing and neurodegeneration. Lancet Neurol. Rayment D, et al. Mini-Mental State Examination (MMSE) for the
2013;12(8):822-38. doi:10.1016/S1474-4422(13)70124-8. detection of dementia in clinically unevaluated people aged 65 and over in
57. Wardlaw JM, Valdés Hernández MC, Muñoz-Maniega S. What are community and primary care populations. Cochrane Database Syst Rev.
white matter hyperintensities made of? Relevance to vascular cognitive 2016;2016(1):CD011145. doi:10.1002/14651858.CD011145.
impairment. J Am Heart Assoc. 2015;4(6):001140. doi:10.1161/ 79. Nitrini R, Lefèvre BH, Mathias SC, Caramelli P, Carrilho PEM, Sauaia N, et al.
JAHA.114.001140. Testes neuropsicológicos de aplicação simples para o diagnóstico de
58. Debette S, Schilling S, Duperron MG, Larsson SC, Markus HS. Clinical demência. Arq Neuropsiquiatr. 1994;52(4):457-65. doi:10.1590/S0004-
Significance of Magnetic Resonance Imaging Markers of Vascular 282X1994000400001.
Brain Injury: A Systematic Review and Meta-analysis. JAMA Neurol. 80. Nitrini R, Bucki SMD, Yassuda MS, Fichman HC, Caramelli P. The Figure
2019;76(1):81-94. doi:10.1001/jamaneurol.2018.3122. Memory Test: diagnosis of memory impairment in populations with

62   Vascular cognitive impairment.   Barbosa BJAP, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

heterogeneous educational background. Dement Neuropsychol. diferentes de escolaridade. Aspectos preliminares. Rev Bras Neurol
2021;15(2):173-85. doi:10.1590/1980-57642021dn15-020004. [Internet]. 2013 [citado em 10 nov 2021];48(1):32-6. Disponível em:
81. Nitrini R, Caramelli P, Porto CS, Charchat-Fichman H, Formigoni AP, http://files.bvs.br/upload/S/0101-8469/2013/v49n1/a3588.pdf
Carthery-Goulart MT, et al. Brief cognitive battery in the diagnosis 99. Chaves MLF, Camozzato AL, Godinho C, Kochhann R, Schuh A,
of mild Alzheimer’s disease in subjects with medium and high levels Almeida VL, et al. Validity of the clinical dementia rating scale for the
of education. Dement Neuropsychol. 2007;1(1):32-6. doi:10.1590/ detection and staging of dementia in Brazilian patients. Alzheimer Dis
S1980-57642008DN10100006. Assoc Disord. 2007;21(3):210-7. doi:10.1097/WAD.0b013e31811ff2b4.
82. Borson S, Scanlan J, Brush M, Vitaliano P, Dokmak A. The mini-cog: 100. Sudo FK, Alves GS, Moreira DM, Laks J, Engelhardt E. Subcortical
a cognitive “vital signs” measure for dementia screening in multi-lingual Vascular Cognitive Impairment staged through cdr’s functional subsum
elderly. Int J Geriatr Psychiatry. 2000;15(11):1021-7. doi:10.1002/ (cdr-func): Preliminary results from an outpatient sample. eNeurologicalSci.
1099-1166(200011)15:11<1021::aid-gps234>3.0.co;2-6. 2016;5:7-10. doi:10.1016/j.ensci.2016.06.001.
83. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, 101. Flier WM, Skoog I, Schneider JA, Pantoni L, Mok V, Chen CLH, et al.
Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief screening Vascular cognitive impairment. Nat Rev Dis Primers. 2018;4:18003.
tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53(4):695-9. doi:10.1038/nrdp.2018.3.
doi:10.1111/j.1532-5415.2005.53221.x. 102. Verdelho A, Wardlaw J, Pavlovic A, Pantoni L, Godefroy O, Duering M,
84. Roalf DR, Moberg PJ, Xie SX, Wolk DA, Moelter ST, Arnold SE. et al. Cognitive impairment in patients with cerebrovascular disease:
Comparative accuracies of two common screening instruments for A white paper from the links between stroke ESO Dementia Committee.
classification of Alzheimer’s disease, mild cognitive impairment, and
Eur Stroke J. 2021;6(1):5-17. doi:10.1177/23969873211000258.
healthy aging. Alzheimers Dement. 2013;9(5):529-37. doi:10.1016/
103. Consoli A, Pasi M, Pantoni L. Vascular mild cognitive impairment:
j.jalz.2012.10.001.
concept, definition, and directions for future studies. Aging Clin Exp Res.
85. Saczynski JS, Inouye SK, Guess J, Jones RN, Fong TG, Nemeth E, et al.
2012;24(2):113-6. doi:10.1007/BF03325158.
The Montreal Cognitive Assessment: Creating a Crosswalk with the
104. Poggesi A, Salvadori E, Pantoni L, Pracucci G, Cesari F, Chiti A, et al.
Mini-Mental State Examination. J Am Geriatr Soc. 2015;63(11):2370-4.
Risk and Determinants of Dementia in Patients with Mild Cognitive
doi:10.1111/jgs.13710.
86. Seitz DP, Chan CC, Newton HT, Gill SS, Herrmann N, Smailagic N, et al. Impairment and Brain Subcortical Vascular Changes: A Study of
Mini-Cog for the diagnosis of Alzheimer’s disease dementia and other Clinical, Neuroimaging, and Biological Markers-The VMCI-Tuscany
dementias within a primary care setting. Cochrane Database Syst Rev. Study: Rationale, Design, and Methodology. Int J Alzheimers Dis.
2018;2(2):CD011415. doi:10.1002/14651858.CD011415.pub2. 2012;2012:608013. doi:10.1155/2012/608013.
87. Ghafar MZAA, Miptah HN, O’Caoimh R. Cognitive screening instruments 105. Sudo FK, Alves GS, Tiel C, Ericeira-Valente L, Moreira DM, Laks J, et al.
to identify vascular cognitive impairment: A systematic review. Int J Geriatr Neuroimaging criteria and cognitive performance in vascular mild cognitive
Psychiatry. 2019;34(8):1114-27. doi:10.1002/gps.5136. impairment: A systematic review. Dement Neuropsychol. 2015;9(4):394-404.
88. Gilly Nardy B. Instrumentos de rastreio para o Comprometimento Cognitivo doi:10.1590/1980-57642015DN94000394.
Vascular Subcortical: Revisão da literatura e adaptação do Brief Memory 106. Harper L, Barkhof F, Fox NC, Schott JM. Using visual rating to diagnose
and Executive Test (BMET) ao contexto brasileiro [dissertação na Internet]. dementia: a critical evaluation of MRI atrophy scales. J Neurol Neurosurg
Rio de Janeiro (RJ): Pontifícia Universidade Católica do Rio De Janeiro; Psychiatry. 2015;86(11):1225-33. doi:10.1136/jnnp-2014-310090.
2019 [citado em 12 jul 2021]. Disponível em: http://www.maxwell.vrac. 107. Fazekas F, Chawluk JB, Alavi A, Hurtig HI, Zimmerman RA. MR signal
puc-rio.br/Busca_etds.php?strSecao=resultado&nrSeq=46464@1 abnormalities at 1.5 T in Alzheimer’s dementia and normal aging.
89. Ylikoski R, Jokinen H, Andersen P, Salonen O, Madureira S, Ferro J, et al. AJR Am J Roentgenol. 1987;149(2):351-6. doi:10.2214/ajr.149.2.351.
Comparison of the Alzheimer’s Disease Assessment Scale Cognitive 108. Pantoni L, Basile AM, Pracucci G, Asplund K, Bogousslavsky J, Chabriat H,
Subscale and the Vascular Dementia Assessment Scale in differentiating et al. Impact of age-related cerebral white matter changes on the transition
elderly individuals with different degrees of white matter changes. to disability – the LADIS study: rationale, design and methodology.
The LADIS Study. Dement Geriatr Cogn Disord. 2007;24(2):73-81. Neuroepidemiology. 2005;24(1-2):51-62. doi:10.1159/000081050.
doi:10.1159/000103865. 109. Scheltens P, Barkhof F, Leys D, Pruvo JP, Nauta JJ, Vermersch P, et al.
90. Hong YJ, Yoon B, Shim YS, Han IW, Han SH, Park KH, et al. A semiquantative rating scale for the assessment of signal hyperintensities
Do Alzheimer’s disease (AD) and subcortical ischemic vascular dementia on magnetic resonance imaging. J Neurol Sci. 1993;114(1):7-12.
(SIVD) progress differently? Arch Gerontol Geriatr. 2014;58(3):415-9. doi:10.1016/0022-510x(93)90041-v.
doi:10.1016/j.archger.2013.11.005. 110. Wahlund LO, Barkhof F, Fazekas F, Bronge L, Augustin M, Sjögren M, et al.
91. Pendlebury ST, Cuthbertson FC, Welch SJV, Mehta Z, Rothwell PM. A new rating scale for age-related white matter changes applicable to MRI
Underestimation of cognitive impairment by Mini-Mental State and CT. Stroke. 2001;32(6):1318-22. doi:10.1161/01.str.32.6.1318.
Examination versus the Montreal Cognitive Assessment in patients with 111. Schmidt R, Schmidt H, Haybaeck J, Loitfelder M, Weis S, Cavalieri M, et al.
transient ischemic attack and stroke: a population-based study. Stroke. Heterogeneity in age-related white matter changes. Acta Neuropathol.
2010;41(6):1290-3. doi:10.1161/STROKEAHA.110.579888. 2011;122(2):171-85. doi:10.1007/s00401-011-0851-x.
92. Webb AJS, Pendlebury ST, Li L, Simoni M, Lovett N, Mehta Z, et al. 112. Chowdhury MH, Nagai A, Bokura H, Nakamura E, Kobayashi S,
Validation of the Montreal cognitive assessment versus mini-mental state Yamaguchi S. Age-Related Changes in White Matter Lesions,
examination against hypertension and hypertensive arteriopathy after Hippocampal Atrophy, and Cerebral Microbleeds in Healthy Subjects
transient ischemic attack or minor stroke. Stroke. 2014;45(11):3337-42. Without Major Cerebrovascular Risk Factors. J Stroke Cerebrovasc Dis.
doi:10.1161/STROKEAHA.114.006309. 2011;20(4):302-9. doi:10.1016/j.jstrokecerebrovasdis.2009.12.010.
93. Pendlebury ST, Mariz J, Bull L, Mehta Z, Rothwell PM. MoCA, ACE-R,
113. Kandel BM, Avants BB, Gee JC, McMillan CT, Erus G, Doshi J, et al.
and MMSE versus the National Institute of Neurological Disorders
White matter hyperintensities are more highly associated with pre-
and Stroke-Canadian Stroke Network Vascular Cognitive Impairment
clinical Alzheimer’s disease than imaging and cognitive markers
Harmonization Standards Neuropsychological Battery after TIA and stroke.
of neurodegeneration. Alzheimers Dement (Amst). 2016;4:18-27.
Stroke. 2012;43(2):464-9. doi:10.1161/STROKEAHA.111.633586.
doi:10.1016/j.dadm.2016.03.001.
94. Mioshi E, Dawson K, Mitchell J, Arnold R, Hodges JR. The Addenbrooke’s
114. Havakuk O, King KS, Grazette L, Yoon AJ, Fong M, Bregman N, et al.
Cognitive Examination Revised (ACE-R): a brief cognitive test battery
for dementia screening. Int J Geriatr Psychiatry. 2006;21(11):1078-85. Heart Failure-Induced Brain Injury. J Am Coll Cardiol. 2017;69(12):1609-16.
doi:10.1002/gps.1610. doi:10.1016/j.jacc.2017.01.022.
95. César KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. Addenbrooke’s 115. Doehner W, Ural D, Haeusler KG, Celutkiene J, Bestetti R, Cavusoglu Y,
cognitive examination-revised: normative and accuracy data for et al. Heart and brain interaction in patients with heart failure: overview
seniors with heterogeneous educational level in Brazil. Int Psychogeriatr. and proposal for a taxonomy. A position paper from the Study Group on
2017;29(8):1345-53. doi:10.1017/S1041610217000734. Heart and Brain Interaction of the Heart Failure Association: Heart and
96. César KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. MoCA Test: brain interaction in heart failure. Eur J Heart Fail. 2018;20(2):199-215.
normative and diagnostic accuracy data for seniors with heterogeneous doi:10.1002/ejhf.1100.
educational levels in Brazil. Arq Neuropsiquiatr. 2019;77(11):775-81. 116. Passiak BS, Liu D, Kresge HA, Cambronero FE, Pechman KR, Osborn KE,
doi:10.1590/0004-282X20190130. et al. Perivascular spaces contribute to cognition beyond other small
97. Moreira IFH, Lourenço RA, Soares C, Engelhardt E, Laks J. Cambridge vessel disease markers. Neurology. 2019;92(12):e1309-21. doi:10.1212/
Cognitive Examination: performance of healthy elderly Brazilians with low WNL.0000000000007124.
education levels. Cad Saude Publica. 2009;25(8):1774-80. doi:10.1590/ 117. Teixeira MM, Passos VMA, Barreto SM, Schmidt MI, Duncan BB,
S0102-311X2009000800013. Beleigoli AMR, et al. Association between diabetes and cognitive function
98. Moreira IFH, Bezerra AB, Sudo FK, Alves GS, Ericeira-Valente L, Tiel C, at baseline in the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil).
et al. CAMCOG - valores das subescalas em idosos normais com níveis Sci Rep. 2020;10(1):1596. doi: 10.1038/s41598-020-58332-9.

Barbosa BJAP, et al.   Vascular cognitive impairment.   63


Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

SUPPLEMENTARY MATERIAL
NINDS and the Canadian Stroke Network developed a 5-minute (12 points) subset of the MoCA to identify stroke
patients who developed VCI1. As the MoCA itself, the total scores on this screening test were inversely correlated
with age and positively correlated with education. The Oxford Cognitive Screen is a 15-minute test with fewer
language elements than the MoCA, making it particularly useful in acute stroke patients who may have language
deficits2. The LADIS neuropsychological battery was designed to assess the cognitive performance of a wide range
of functions in a cohort of independently living individuals with age-related white matter changes (ARWMC) during
a 3-year follow-up3. It is a short-comprehensive instrument that can be administered in a single visit. This battery
was chosen based on the test familiarity and the instrument validity to assess cognitive decline in vascular disease
patients. In addition to widely known and validated instruments (MMSE, Stroop Test, and Trail Tests), it included the
VADAS-Cog4; detailed information on global and selective cognitive functioning and time-dependent tasks (Delayed
Recall, Digit Symbol, Digit Extension, Maze, Digit Cancellation and Verbal Fluency) to complement the assessment
of attention, mental processing speed and motor control were provided. These cognitive domains and executive
function are possibly more affected by white matter alterations5. The authors concluded that the neuropsychological
performance of the patients was significantly influenced by age and education, with a higher educational level
being consistently associated with better performance on cognitive tasks. In contrast, older age was associated
with difficulties in memory and executive functions.
Tuscany - Vascular Cognitive Impairment (VCI) is a multicenter, prospective, observational study aiming
to evaluate predictors of transition from VCI (defined by the presence of moderate to severe WML) to dementia6.
The neuropsychological test battery was designed specifically for MCI due to small vessel disease (MCI-SVD),
enabling the automation and standardization of scores and a personalized cognitive profile. For the VCI-Tuscan
neuropsychological battery, tests were selected among those recommended for VCI and the protocols proposed by
the National Institute for Neurological Disorders and Stroke and the Canadian Stroke Network consensus conference
on harmonization standards for VCI7. The definition of cognitive domains followed a confirmatory analysis of
the dimensions assumed theoretically for the battery8, resulting in four cognitive clusters: memory (evaluated by
four cognitive scores), attention and executive functions (five cognitive scores), language (two cognitive scores),
and constructive praxis. Thus, the battery includes two tests of global cognitive functioning and another nine
tests covering these cognitive domains.

REFERENCES
1. Kennedy RE, Wadley VG, McClure LA, Letter AJ, Unverzagt FW, Crowe M, Scale that broaden its scope. The Alzheimer’s Disease Cooperative Study.
et al. Performance of the NINDS-CSN 5-minute protocol in a national Alzheimer Dis Assoc Disord. 1997;11(Suppl 2):S13-21.
population-based sample. J Int Neuropsychol Soc. 2014;20(8):856-67. 6. Poggesi A, Salvadori E, Pantoni L, Pracucci G, Cesari F, Chiti A, et al.
doi:10.1017/S1355617714000733. Risk and Determinants of Dementia in Patients with Mild Cognitive
2. Mancuso M, Demeyere N, Abbruzzese L, Damora A, Varalta V, Pirrotta F, Impairment and Brain Subcortical Vascular Changes: A Study of Clinical,
et al. Using the Oxford Cognitive Screen to Detect Cognitive Impairment Neuroimaging, and Biological Markers-The VMCI-Tuscany Study:
Rationale, Design, and Methodology. Int J Alzheimers Dis. 2012;2012:608013.
in Stroke Patients: A Comparison with the Mini-Mental State Examination.
doi:10.1155/2012/608013.
Front Neurol. 2018;9:101. doi:10.3389/fneur.2018.00101.
7. Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL,
3. Madureira S, Verdelho A, Ferro J, Basile AM, Chabriat H, Erkinjuntti T, et al.
Black SE, et al. National Institute of Neurological Disorders and
Development of a neuropsychological battery for the Leukoaraiosis and
Stroke-Canadian Stroke Network Vascular Cognitive Impairment
Disability in the Elderly Study (LADIS): experience and baseline data. Harmonization Standards. Stroke. 2006;37(9):2220-41. doi:10.1161/
Neuroepidemiology. 2006;27(2):101-16. doi:10.1159/000095381. 01.STR.0000237236.88823.47.
4. Ferris SH. General measures of cognition. Int Psychogeriatr. 8. Salvadori E, Poggesi A, Pracucci G, Inzitari D, Pantoni L; VMCI-Tuscany
2003;15(Suppl 1):215-7. doi:10.1017/S1041610203009220. Study Group. Development and psychometric properties of a
5. Mohs RC, Knopman D, Petersen RC, Ferris SH, Ernesto C, Grundman M, neuropsychological battery for mild cognitive impairment with small vessel
et al. Development of cognitive instruments for use in clinical trials of disease: the VMCI-Tuscany Study. J Alzheimers Dis. 2015;43(4):1313-23.
antidementia drugs: additions to the Alzheimer’s Disease Assessment doi:10.3233/JAD-141449.

64   Vascular cognitive impairment.   Barbosa BJAP, et al.


Table. Characteristics of cognitive assessment protocols in different guidelines - cognitive tests included. References in the text.
LADIS Battery

COMMANDER
Trails
MEEM ADAS-Cog Digit- Digit span Cancellation Stroop
Maze Verbal Fluency AB
symbol WAIS-III Digits

Global Mental Functioning + +

Orientation +

Memory + +

Attention + + + +

Language +

Constructional abilities +

Executive function + + + +

Praxis +

Speed and motor control + + +

MCIV-Tuscany Battery

Rey Test Rey-Osterrieth


Digit- Short Trails
MEEM MoCA (RAVL) Copy Figure - Verbal Fluency Stroop Visual search
symbol story AB
immed - late Retrieval

Global Mental Functioning + +

Orientation

Memory ++ + +

Attention + Executive
+ ++ + +
function

Language +

Constructional abilities +

Continue...
Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Barbosa BJAP, et al.   Vascular cognitive impairment.   65


Table. Continuation.

VCI-VICCCS criteria

Simple
Rey-Osterrieth
Boston Digit- Hopkins Verbal reaction
MEEM Copy Figure- Verbal Fluency NPI-Q CES-D
Naming symbol Learning Test time with
Retrieval
choice

Global Mental Functioning +

Orientation

Memory +

Attention + +

66   Vascular cognitive impairment.   Barbosa BJAP, et al.


Language + +
Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Constructional abilities +

Executive function + + +

Speed and motor control +

Depressive symptoms +

Neuropsychiatric
+
symptoms

Consensus Protocol A/ABN 2011

Clock
MEEM Verbal Fluency
Drawing Test

Global Mental Functioning +

Language +

Constructional abilities +

Executive function +

Continue...
Table. Continuation.

Protocol B of consensus/ABN 2011

CAMCOG Cornell
Digit span Trails Boston Word List -
MEEM (global and Verbal Fluency CLOX NPI Depression CDR
WAIS-III AB Naming CERAD
subscales) Scale

Global Mental Functioning + +

Orientation + +

Memory +

Attention + + +

Language + + +

Constructional abilities +

Executive function + + + +

Praxis +

Gnosia +

Abstract reasoning +

Depressive symptoms +

Neuropsychiatric
+
symptoms

Severity +

Continue...
Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Barbosa BJAP, et al.   Vascular cognitive impairment.   67


Table. Continuation.

NINDS - Canadian Stroke Network Vascular Cognitive Impairment (Hachinski et al., 2006)

30-minute Protocol

Hopkins
Digit- Trails Verbal
MEEM Verbal NPI-Q CES-D
symbol AB Fluency
Learning test

Global Mental Functioning +

Orientation

Memory +

Attention + +

68   Vascular cognitive impairment.   Barbosa BJAP, et al.


Language +
Dement Neuropsychol 2022 September;16(3 Suppl. 1):49-68

Constructional abilities

Executive function + + +

Speed and motor control

Depressive symptoms +

Neuropsychiatric
+
symptoms

5-minute Protocol - MoCA Subtests

Memory Phonemic
task with Orientation Verbal
words Fluency

Orientation +

Memory +

Attention + +

Executive function +
Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S105EN

Diagnosis and management of


Parkinson’s disease dementia and
dementia with Lewy bodies:
recommendations of the Scientific Department
of Cognitive Neurology and Aging of the
Brazilian Academy of Neurology
Jacy Bezerra Parmera1 , Vitor Tumas2 , Henrique Ballalai Ferraz3 , Mariana Spitz4 ,
Maira Tonidandel Barbosa5,6 , Jerusa Smid1 , Breno José Alencar Pires Barbosa1,7,8 ,
Lucas Porcello Schilling9,10,11 , Márcio Luiz Figueiredo Balthazar12 , Leonardo Cruz de Souza13 ,
Francisco Assis Carvalho Vale14 , Paulo Caramelli13 , Paulo Henrique Ferreira Bertolucci3 ,
Márcia Lorena Fagundes Chaves15,16 , Sonia Maria Dozzi Brucki1 , Ricardo Nitrini1 ,
Raphael Machado Castilhos15 , Norberto Anízio Ferreira Frota17,18

1
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
2
Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Neurociências e Ciências do Comportamento, São Paulo, SP, Brazil.
3
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo, SP, Brazil.
4
Universidade do Estado do Rio de Janeiro, Serviço de Neurologia, Rio de Janeiro, RJ, Brazil.
5
Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Medicina Interna, Belo Horizonte, MG, Brazil.
6
Faculdade Ciências Médicas de Minas Gerais, Medicina Geriátrica, Belo Horizonte, MG, Brazil.
7
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife, PE, Brazil.
8
Instituto de Medicina Integral Prof. Fernando Figueira, Recife, PE, Brazil.
9
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre, RS, Brazil.
10
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre, RS, Brazil.
11
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre, RS, Brazil.
12
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas, SP, Brazil.
13
Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte, MG, Brazil.
14
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos, SP, Brazil.
15
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre, RS, Brazil.
16
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Serviço de Neurologia, Porto Alegre, RS, Brazil.
17
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
18
Universidade de Fortaleza, Fortaleza, CE, Brazil.
Correspondence: Jacy Bezerra Parmera; Email: jacy.parmera@hc.fm.usp.br.
Conflict of interest: JS: Participation as speaker in symposia sponsored by Roche laboratory; LPS: Participation in advisory boards for Biogen. Participation as
speaker in symposia sponsored by Aché, Apsen, and Biogen laboratories; MLFB: Participation in advisory boards for Biogen. Development of material for continuous
medical education and participation as speaker in symposia sponsored by EMS and Torrent laboratories; PC: Participation as principal investigator in clinical trials
for Novo Nordisk and Roche laboratories. Participation in advisory boards for Aché, Biogen, EMS, Nutricia, and Roche laboratories. Development of material for
continuous medical education and participation as speaker in symposia sponsored by Aché, Nutricia, Libbs, Roche, Sandoz, Torrent, and Zodiac laboratories; PHFB:
Participation in advisory boards for Biogen and Novo Nordisk laboratories. Supervision of training activities for Biogen, Janssen-Cilag, and Novo Nordisk laboratories
and for Quintiles. Participation as speaker in symposia sponsored by Apsen, Nutricia, Roche, and Sandoz laboratories; LCS: Participation in advisory board for
Biogen. Participation as speaker in symposia sponsored by Biogen; RN: Participation in advisory board for Biogen; JBP, VT, HBF, MS, MTB, BJAPB, FACV, MLFG,
SMDB, RMC, NAFF: There is no conflict of interest to declare.
Received on July 05, 2021; Received in its final form on October 13, 2022; Accepted on April 27, 2022.

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   69


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

ABSTRACT. Parkinson’s disease dementia (PDD) and dementia with Lewy bodies (DLB) represent the second most common type of degenerative dementia
in patients aged 65 years and older, leading to progressive cognitive dysfunction and impaired quality of life. This study aims to provide a consensus based
on a systematic Brazilian literature review and a comprehensive international review concerning PDD and DLB. Moreover, we sought to report on and give
recommendations about the best diagnostic approaches focusing on primary and secondary care. Based on the available data, we recommend clinicians
to apply at least one brief global cognitive instrument to assess PDD, such as the Mini-Mental State Examination and preferably the Montreal Cognitive
Assessment and the Addenbrooke’s Cognitive Examination-Revised. Validated instruments to accurately assess functional abilities in Brazilian PD patients
are still incipient. Further studies should focus on biomarkers with Brazilian cohorts.
Keywords: Consensus; Parkinson Disease; Lewy Bodies; Dementia.

DIAGNÓSTICO E MANEJO DA DEMÊNCIA DA DOENÇA DE PARKINSON E DEMÊNCIA COM CORPOS DE LEWY: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO
DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. A demência da Doença de Parkinson (DDP) e a demência com corpos de Lewy (DCL) representam a segunda causa mais comum de demência
neurodegenerativa em pessoas com mais de 65 anos, ocasionando progressivo declínio cognitivo e comprometimento da qualidade de vida. O presente
estudo tem como objetivo prover um Consenso de especialistas sobre a DDP e DCL, baseado em revisão sistemática da literatura brasileira e revisão
não-sistemática de literatura internacional. Ademais, tal estudo visa a promover informação e conceder recomendações sobre abordagem diagnóstica,
com foco nos níveis de atenção primária e secundária em saúde. Com base nos dados disponíveis, recomendamos que os profissionais realizem pelo
menos um breve instrumento cognitivo global, como o Mini-Exame do Estado Mental, contudo de preferência optem pela Avaliação Cognitiva de Montreal e
o Exame Cognitivo de Addenbrooke-Revisado. Observa-se uma carência de instrumentos validados para a avaliação precisa das habilidades funcionais em
pacientes brasileiros com DDP e DCL. Além disso, mais estudos focando em biomarcadores com coortes brasileiras também demonstram ser necessários.
Palavras-chave: Consenso; Doença de Parkinson; Corpos de Lewy; Demência.

INTRODUCTION predictors of burden in Brazilian PD caregivers are

P arkinson’s disease dementia (PDD) and dementia


with Lewy bodies (DLB) are different clinical
syndromes that share the same pathological hallmark,
cognitive and behavioral symptoms in PD patients,
the time of caregiving, and the occurrence of mood
disorders in caregivers3,4.
namely Lewy body disease, in which post-mortem This work aimed to reach a specialist consensus
examination shows neuronal α-synuclein inclusions based on a systematic Brazilian literature review and
(Lewy bodies) and neuronal loss. The umbrella term a comprehensive international review of other updated
Lewy Body Dementia (LBD) includes both of these and relevant literature on PDD and DLB. Moreover,
syndromes, representing the second most common type we sought to report on and provide a clinical guide with
of degenerative dementia in patients aged 65 years and recommendations focusing on primary and secondary
older and leading to progressive cognitive dysfunction, care in the workup of PDD and DLB.
motor deterioration, and impaired quality of life1,2.
Despite having the same pathological substrate,
Parkinson’s disease (PD) and DLB are classified METHODS
as different diseases based on the temporal This study was conceptualized in meetings held from
relationship of cognitive and motor symptoms. April to June 2021 to design the review process and
Current criteria recommend diagnosing a patient draft the consensus. The consensus group comprised
with PDD when dementia develops in the context seven members experts in the field.
o f well-establishe d PD 1 and w ith DLB when Firstly, we performed a systematic review of the
dementia precedes or coincides within one year of Brazilian literature concerning epidemiological, clinical,
the development of motor symptoms, namely the ancillary tests, and biomarkers and of management
“1-year rule”. This is considered an empirical approach studies regarding PDD and DLB. A systematic
that avoids clinical practice mistakes and clarifies the computer-based literature search using the Start
distinction in research and clinical studies2. program was performed on the PubMed, Scielo,
PDD and DLB significantly affect one’s psychological and PsycINFO electronic databases. For the search,
and social life, decreasing the quality of life for both the medical subject headings [“Parkinson’s disease”]
patients and caregivers. In Brazil, most PD patients OR [“Lewy Body dementia”] OR [“diffuse Lewy Body
are assisted in their homes by family members, disease”] AND [“dementia”] OR [“mild cognitive
who act as informal caregivers. Caregivers of PD impairment”] OR [“cognitive dysfunction”] and (Brazil)
patients face increasing burdens and may develop using English and Portuguese and human studies filters.
burnout, depression, and anxiety. The most significant Inclusion criteria were 1) Brazilian studies referring

70   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

to PD and DLB 2) investigating cognitive or associated RESULTS


features 3) in older adults or those over 18 years old. The search gathered 732 records identified by database
Exclusion criteria were 1) studies focusing on other searching. After an initial exclusion, 106 articles
cognitive primary diagnoses. published in Brazilian cohorts were selected, of which
The consensus group was later divided into three 60 were later excluded due to the reasons explained
subgroups concerning epidemiology and risk factors, in Figure 1.
clinical features and neuropsychological assessment, In total, 46 articles were included in this consensus.
or biomarkers and management, each subgroup in For topics without specific articles in the Brazilian
charge of critically reviewing the literature selected literature, 17 studies, including research articles, review
based on predefined selection criteria. A comprehensive articles, and international consensus, were added
literature search was also performed to add current to the 46 previously selected Brazilian articles, in a
information and knowledge for insufficient data sum of 63 studies.
acquired from the systematic review or if the specialists The PRISMA flow diagram summarizes the search
wanted to enrich the consensus study. (Figure 1).

Identification of studies via databases and registers

Records identified through database searching (n=732)


Identification

Records removed before screening: 626


Duplicated
Case reports
Out of scope

Records screened (n=106)

Records excluded: 46
Screening

Duplicated
Case reports
Letters to the editor
Out of scope

Full-text articles assessed for eligibility (n=60)

Records excluded: 14
Duplicated
Included

Case reports
Letter to the editor

Studies included in review (n=46)

Figure 1. Prisma flow diagram.

Epidemiology Bambuí study5 and 56.7% had it in the Pietá study6.


The prevalence rate of parkinsonism in two community In community-based studies, PDD represented 13.5%
studies with older adults aged 60 years and over of cases with parkinsonism and dementia6. DLB, in turn,
in Brazil ranged from 7.2%5 to 10.6%6, with idiopathic caused less than 5% of cases7,8.
Parkinson’s disease being the most frequent etiology. In tertiary centers-based studies, the frequency
Both studies obser ved a progressive increase of these two etiologies ranged from 3.7% to 15% of
of parkinsonism with age, reaching 30.4% prevalence cases9-13 Similarly, a study based on neuropathological
in the population over 95 years old 6. Around 15% diagnosis cases14 showed that LBD caused 15% of cases
of patients with parkinsonism had dementia in the of dementia, whether isolated or associated with other

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   71


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

pathologies. LBD was also the second most frequent Recommendations for the diagnosis of PDD and DLB
neurodegenerative etiology of dementia and related Dementia is usually defined by clinical diagnostic
to a higher risk of dementia with an odds ratio (OR) criteria. Historically, the most used criteria, even for
of 3.4 (CI: 1.94-5.97). The same study had high specificity patients with PD, were those of the various versions
for clinical diagnosis14. of the Diagnostic and Statistical Manual of Mental
Dementia frequently occurs in PD and its prevalence Disorders (DSM). This concept changed in 2007
increases with disease duration, ranging from 23% when a task force of the Movement Disorder Society
in the first years of the disease to 80% after over (MDS) developed specific diagnostic criteria for PDD1
15 years of symptoms 15-17. Understanding which and recommended procedures to operationalize the
risk factors are associated with the occurrence of diagnosis 18. Since then, clinical studies, including
dementia in PD patients and what differentiates LBD Brazilian studies, have used these criteria and
patients from other etiologies is essential for an earlier procedures the most. We recommend using these
diagnosis and more appropriate therapy for the disease. criteria with a few adaptations marked below with
These aspects will be discussed in the following topics. quotation marks (Table 1).

Table 1. Criteria for the diagnosis of probable and possible Parkinson’s disease dementia1.

The diagnosis is probable when:


A. Core features must be both present.
1. Diagnosis of Parkinson’s disease according to specific “diagnostic criteria” *
2. A syndrome of “cognitive decline” with insidious onset and slow progression, developing within the context of established PD and diagnosed by
history, clinical, and mental examination, defined as:
- Impairment in more than one cognitive domain (including: attention, executive functions, visuo-spatial functions, memory and language)
- Decline from a premorbid level of functioning
- Deficits severe enough to impair daily life (social, occupational, or personal care), regardless of the impairment from motor or autonomic symptoms

B. Associated clinical features.

Typical profile of cognitive deficits including impairment in at least two of the four core cognitive domains (impaired attention which may fluctuate,
impaired executive functions, impairment in visuo-spatial functions, and impaired free recall which usually improves with cueing)
Behavioral features such as apathy, changes in personality and mood, hallucinations, delusions, and excessive daytime sleepiness may be present
(but are not necessary for diagnosis)

C. Features which do not exclude PDD, but make the diagnosis uncertain.

Existence of any other abnormality which may cause cognitive impairment but is not as the cause of dementia, e.g., presence of relevant vascular
disease in imaging.
The time interval between the development of motor and cognitive symptoms is unknown**

D. There are none of the following features suggesting other conditions or diseases as the cause of mental impairment, which would hinder an
accurate diagnosis of PDD: delirium, diagnosis of major depression, evidence for diagnosis of probable vascular dementia.

The diagnosis is possible when:


A. Core features must be both present.

B. The cognitive decline presents with atypical profile of cognitive impairment in one or more domains, such as prominent or receptive (fluent) aphasia
or pure storage-failure type amnesia (memory does not improve with cueing or in recognition tasks) with preserved attention.
or

C. There are features that make the diagnosis uncertain (e.g., presence of relevant vascular disease in imaging).
or

D. Time interval between the development of motor and cognitive symptoms is unknown (“1-year rule”)
or

E. There are features suggesting other conditions or diseases as causes of mental impairment (delirium, diagnosis of major depression, evidence for
diagnosis of probable vascular dementia)
* United Kingdom Parkinson’s Disease Society Brain Bank diagnostic criteria (20) for PD or another validated criterion; ** Refers to the “1-year rule” to differentiate PDD from Lewy body
dementia. PDD develops within the context of established PD or arbitrarily at least a year after the onset of the classic motor symptoms.

72   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

The fourth consensus report of the DLB Consortium, biomarkers. The report classified clinical symptoms and signs
in turn, has recently refined the recommendations for the as core or supportive and weighed biomarkers as indicative
clinical and pathologic diagnosis of DLB19. It incorporated or supportive based on their specificity and the volume
recent developments to increase sensitivity and clearly of high-quality evidence available. We also recommend
distinguished between clinical features and diagnostic the use of these criteria in clinical practice (Table 2).

Table 2. Criteria for the diagnosis of probable and possible dementia with Lewy bodies2.
1. Essential: dementia is required for a diagnosis of DLB and defined as a progressive cognitive decline which affects social and occupational functions
or daily activities. It mainly affects attention, executive function, and visuoperceptual abilities, worsening memory impairment as it progresses.

Core clinical features:


Fluctuating cognition with pronounced variations in alertness.
Recurrent visual hallucinations.
REM sleep behavior disorder.
One or more spontaneous cardinal features of parkinsonism (bradykinesia, rest tremor, or rigidity).*

Supportive clinical features: severe sensitivity to antipsychotic agents, postural instability, repeated falls; syncope or other transient episodes of
unresponsiveness; severe autonomic dysfunction, e.g., constipation, orthostatic hypotension, urinary incontinence; hypersomnia; hyposmia; other
hallucination modalities; systematized delusions; apathy, anxiety, and depression.

Indicative biomarkers:
Reduced dopamine transporter uptake in basal ganglia demonstrated by SPECT or PET.
Abnormal (low uptake) 123iodine-MIBG myocardial scintigraphy.
Polysomnographic confirmation of REM sleep without atonia.

Supportive biomarkers:
Relative preservation of medial temporal lobe structures on CT/MRI scan.
Generalized low uptake on SPECT/PET perfusion/metabolism scan with reduced occipital activity. Cingulate island sign on FDG-PET imaging.
Prominent posterior slow-wave activity on EEG with periodic fluctuations in the pre-alpha/theta range.

Diagnosis is probable when:


Two or more core clinical features are present
OR
Only one core clinical feature is present but with one or more indicative biomarkers.

Diagnosis is possible when:


Only one core clinical feature (with no biomarkers)
One or more indicative biomarkers are present without core clinical features.
*We continue to recommend the existing 1-year rule between the onset of dementia and parkinsonism.

We suggest that the diagnosis of PD should use We recommend adopting the formal criteria for
validated diagnostic criteria, which can be either the PD-mild cognitive impairment (MCI) published
United Kingdom Parkinson’s Disease Society Brain Bank in 201222 to assess the progression of cognitive decline
diagnostic criteria20 or any other diagnostic criteria with in PD patients. Proposed research criteria for diagnosing
established accuracy for diagnosis, such as the recently prodromal DLB have recently introduced three
proposed MDS Parkinson’s Disease criteria21. possible presentations to the prodromal phase: MCI,
delirium-onset, and psychiatric-onset. We recommend
We also suggest maintaining the “1-year rule”
adopting the DLB-MCI criteria19.
for clinical and research purposes, despite the
controversy over whether PDD and DLB are the Clinical features in PDD and DLB
clinical spectra of the same disease. In turn, specialists The evaluations of PDD and DLB present two major
from the MDS have recently proposed not using challenges. Regarding PDD, it is the prediction and early
the “1-year rule” to distinguish PDD from DLB 21. identification of the progression of cognitive decline
Nevertheless, this approach has been criticized and does to dementia. In DLB, it is probably the differentiation
not present general agreement. from Alzheimer’s disease (AD)23.

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   73


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

Parkinson’s Disease is mainly characterized by and vivid dreams. The suspicion is usually confirmed
classic motor symptoms, such as resting tremor, by polysomnography (PSG), but questionnaires can
rigidity, and bradykinesia. However, it is also associated be used clinically if PSG is unavailable. The Brazilian
with many non-motor manifestations that are Portuguese version of the RBD screening questionnaire
well-recognized symptoms and predominant at the late (RBDSQ) for patients with PD may be helpful for
stages of the disease, such as hyposmia, constipation, RBD diagnosis in the country28. This disorder should
depression, and rapid eye movement (REM) sleep be actively questioned since an individual could
behavior disorder (RBD). These may precede the motor develop it up to 15 years before developing dementia
signs whereas dementia and psychosis are common or parkinsonism. Diagnosing this disorder is therefore
in the late stages of the illness1,20. DLB has the same important to improve patient’s quality of life.
non-motor symptoms, but the cognitive decline Hallucinations are one of the few features that
occurs earlier and the motor symptoms are milder, usefully distinguish between DLB/PDD and AD.
sometimes absent. Dementia is essential to diagnose The phenomenology of hallucinations in PDD and
an individual with DLB. DLB is very similar. Visual hallucinations occur twice
PDD and DLB cognitive profiles are characterized as frequently as auditory ones, being mostly complex,
by impairment mainly in attention, executive, formed hallucinations of anonymous people, but they
and visuospatial functions. Behavioral symptoms may also be family members, body parts, animals,
such as affective changes, hallucinations, and apathy or machines1,30. Overall, patients with PDD seem to
are also frequent1. Most studies with Brazilian samples have less frequent or less severe psychiatric symptoms
showed the same pattern of cognitive impairment24,25. than patients with DLB. Such differences, however,
On the other hand, episodic memory is less affected may simply reflect the disparity in the overall dementia
and patients with MCI usually have more difficulty severity between PDD and DLB1,31.
in free recall than cued recall in memory tests (such as Depression, another non-motor symptom frequent
a list of words or pictures). A previous study exploring in PD patients, can worsen executive dysfunction,
visuospatial dysfunctions in PD patients showed that especially in subjects with low schooling level32. Other
62.2% of 35 patients could not copy the pentagon supportive clinical features in DLB are: severe sensitivity
drawing26. Moreover, Machado et al. suggested that to antipsychotic agents, postural instability, repeated falls,
language and visual organization tend to follow motor syncope, or other transient episodes of unresponsiveness;
skills and general cognitive performance in patients severe autonomic dysfunction, hypersomnia, hyposmia,
with DLB27. Poor cognitive performance frequently other hallucination modalities; systematized delusions;
correlates with more advanced stages of the disease, and apathy, anxiety, and depression2.
older age, low schooling level, depression, and poorer
quality of life15,17,28. Cardiovascular risk factors and PDD, Which diagnostic procedures and neuropsychological tests
however, are not correlated with each other29. should be used to diagnose and evaluate PDD and DLB?
The main cognitive feature in DLB is fluctuating Many different methods and instruments can be used
cognition, which represents a fluctuation in attention to assess cognition in patients with PD and DLB.
or level of consciousness. This feature can range We recommend using an adapted version of the
from episodes of inattention and mental confusion, procedures to operationalize the diagnostic criteria
with disorganized thinking and behavior, to lethargy proposed by the MDS for PDD diagnosis 18. These criteria
and excessive daytime sleepiness. DLB and PD have suggested two levels of diagnosis for PDD. In level I,
differences of parkinsonism presentation. DLB presents the diagnosis is based upon using straightforward
more symmetrical parkinsonism, less frequent rest procedures and bedside cognitive tests, which a non-
tremor, and greater postural instability and lower specialist would easily apply in clinical consultations.
responsiveness to levodopa2. In level II, diagnosis would require a more extensive
REM Behavior Disorder (RBD) has a high specificity and specialized neuropsychological evaluation.
for suggesting an alpha-synucleinopathy. RBD is Predictably, a previous Brazilian study showed that
a parasomnia characterized by loss of atony during REM the prevalence of PDD diagnosis was higher when the
sleep. The current DLB2 criteria included this feature more elaborate procedures of level II rather than of
among the core clinical manifestations of the disease. level I were used (23.8% versus 14.9%, respectively).
Clinical suspicion is based on the patient history, This indicates that level I procedures had lower
in which the caregiver usually describes that the patient sensitivity but greater specificity than level II for the
has abnormal vocalizations, aberrant motor behavior, diagnosis of dementia15.

74   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

The MDS proposed practical procedures for level I is available for patients with PD. The MDS primary
diagnosis of PDD based on an algorithm or checklist. recommendation proposed the “pill questionnaire” as
Both of these procedures are advantageous, either for a simple tool to define independence loss. According
clinical routine or clinical research. However, they require to this instrument, patients without functional loss
some adaptations that we suggest in Table 3. are those who can describe in detail their drug schedule
Regarding the assessment of functionality, the best treatment18. However, many studies suggested that the
functional assessment tool for impairment from cognitive questionnaire was insufficiently accurate. Accordingly,
loss in PD patients is still undefined. Most studies two Brazilian studies suggested that the pill questionnaire
used functional assessment instruments applied in was less sensitive than other instruments to detect
other dementias, and no proper validation instrument functional impairment15,33.

Table 3. Modified algorithm for diagnosing Parkinson’s disease dementia at Level I.


Diagnosis of Parkinson’s disease*

Motor symptoms of PD developed at least “1-year” before the onset of dementia

Cognitive deficits severe enough to impact daily living**

Abnormal performance on a brief global cognitive scale***

Cognitive abnormalities affect typical cognitive domains such as: attention, executive functions, visuo-spatial functions, memory and language
(excluding prominent aphasia)

Absence of Major Depression

Absence of delirium

There are no other features that make the diagnosis uncertain or suggest other conditions or diseases as causes of mental impairment (diagnosis of
probable vascular dementia)

If all items are checked, the diagnosis is probable PDD


If item 2 and/or item 5, and/or item 6, and/or item 7, and/or item or 8 are not checked, the diagnosis is possible PDD
*Based on specific diagnostic criteria; **According to any method of functional assessment; ***Based on population normative levels, with scores adjusted for schooling level and other
factors (age, sex, etc.) if necessary.

Studies with Brazilian PD patients have used several as a reference to diagnose functional impairment with
instruments to assess functional abilities, including the the cutoff score of 3.27, the same defined for the cross-
Pfeffer Functional Activities Questionnaire (FAQ), cultural adaptation of the scale for Brazilian patients
the Disability Assessment for Dementia (DAD), with Alzheimer’s disease dementia37.
the Informant Questionnaire on Cognitive Decline Baldivia et al. used DAD to assess functional
in the Elderly (IQCODE), Pill questionnaire, the phone impairment in patients with PD with the cutoff score
call test, and the Direct Assessment of Functional of 94.6, the same previously defined for diagnosis of
Abilities (DAFA)15,33-37. Two studies evaluated the PFAQ Alzheimer’s disease dementia in Brazilian patients15.
accuracy to detect functional impairment in patients In turn, Breder et al. used a modified version of DAD
with PD33,37. Oliveira et al. proposed that a score >2 to evaluate patients with PD. The authors did not
had 23% sensitivity and 74% specificity to predict evaluate a disability but the dependence in activities
abnormal performance on a global cognitive test33. of daily living (ADL). They classified the patient
In turn, Almeida et al. defined a score >3 as the best as dependent if he needed assistance for over half
cutoff for a modified version of the PFAQ (mPFAQ) of the time on at least one activity evaluated due
to diagnose PDD (47% sensitivity and 88% specificity). to cognitive impairment 35. Oliveira et al. showed
In this modified version, the authors suppressed items 5 that close informants usually overestimate patients’
(make coffee) and 6 (prepare a meal) from the original instrumental abilities in ADL. In some patients,
version to minimize the possible effects of motor therefore, the assessment of functional loss might only
symptoms that affect the interpretation of the patient’s be considered reliable with direct assessment tools.
functional assessment37. The authors used the IQCODE The direct assessment of functional abilities might

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   75


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

better predict impairment on a global cognitive scale Diagnostic accuracy also depends on the quality of
than questionnaires33. evidence for normative data of cognitive scales in the
These studies show a lack of validated instruments Brazilian population. Therefore, a less precise approach
to accurately assess functional abilities in Brazilian PD to define cutoff scores for PDD diagnosis relies on scale-
patients. Further studies should focus on developing assessment studies to distinguish between PD without
these instruments. We recommend that despite the dementia and PDD. Souza et al. proposed using the
functional assessment tool used, the examiner should interlocking finger test (ILFT) to screen PD patients for
always be sure that other PD symptoms, such as motor dementia39. ILFT is a simple test in which the patient
symptoms, are not interfering with the patient’s ability is asked to imitate four meaningless bimanual gestures.
to carry out activities. For clinical practice, though the The authors showed that a score <3 (each gesture
pill questionnaire seems to be an interesting procedure earned one point) presented 61% sensitivity and
for screening, it should be complemented by other 85% specificity to diagnose PDD, indicating ILFT as
functional assessments or a clinical interview aimed a practical bedside test to assess cognitive impairment
at functional status. For clinical research studies, in patients with PD39.
authors must precisely define the methods used The Montreal Cognitive Assessment (MoCA) and the
to assess the functional abilities of patients with PD. Addenbrooke’s Cognitive Examination-Revised (ACE-R)
Regarding global cognitive assessment, we suggest are other brief global cognitive instruments that have
using other global cognitive scales besides the Mini- been used in PD and DLB. They have already been
Mental State Examination (MMSE). The MDS has translated and validated for Brazilian populations40-42.
proposed the MMSE as a first-line assessment tool Some studies suggest that these instruments may be
for global cognitive efficiency in PD because of the more effective than the MMSE to detect dementia in
instrument’s simplicity and wide use in dementia. PD patients35,36,38,43,44. However, most studies showed
However, other studies, including some with Brazilian inadequate accuracy to evaluate PD-MCI43. A previous
populations, showed that the MMSE had lower study evaluating the MoCA subtests showed that
sensitivity than other scales to detect cognitive PD patients with low schooling level might have
abnormalities in PD patients34,35,38. Moreover, MMSE difficulty completing the tests, contributing to poor
cutoff scores adjusted for schooling level from normative diagnostic accuracy and affecting the detection
studies performed in the Brazilian population were of MCI45. Table 4 summarizes the findings and cutoff
more accurate for diagnosis of PDD15,34. Other cognitive scores applied with brief global cognitive instruments
scales should also consider schooling level. in Brazilian PD patients.

Table 4. Clinical Brazilian research studies with brief global cognitive instruments in Parkinson’s disease dementia.

Study Sample characteristics Diagnosis of dementia Results

The area under the ROC curve for the MoCA dementia
diagnosis was 0.86 (95%CI= 0.76-0.95). The best cutoff
79 PD patients
score for MoCA to differentiate patients with PDD from
Median age 63 years (28-81)
According to level II of the the others was <21 (sensitivity of 94%, specificity of 68%).
Sobreira et al. 201543 Median schooling
MDS diagnostic criteria The area under the ROC curve for the ACE-R was 0.84
time 6.5 years (1-20)
(95%CI= 0.74-0.94). The best cutoff score for ACE-R
36% male
to differentiate patients with PDD from the others was <76
(sensitivity of 88%, specificity of 68%).

The area under the ROC curve for the MMSE was 0.88
(95% CI: 0.78-0.97). The best cutoff score for MMSE
70 PD patients
to differentiate patients with PDD from the others was ≤24,
Mean age 64.1 years (SD= 9.3)
According to level II of the (sensitivity of 78.5%, specificity of 96.4%).
Rocha et al. 201438 Mean schooling time 5.9 years
MDS diagnostic criteria The area under the ROC curve for the ACE-R was 0.93
(SD= 3.4)
(95% CI: 0.86-0.98). The best cutoff score for ACE-R
57.1% male
to differentiate patients with PDD from the others was ≤72
(sensitivity of 89.3%, specificity of 84.6%)

Continue...

76   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

Table 4. Continuation.

Study Sample characteristics Diagnosis of dementia Results

101 PD patients The area under the ROC curve for the ILFT was 0.761The best
Mean age 62.5 years cutoff score for ILFT to differentiate patients with PDD from
According to MDS
(SD= 12.1) the others was <3 (sensitivity of 61%, specificity of 85%).
Souza et al, 201639 criteria but did say if
Mean schooling time 5.2 years The area under the ROC curve for the MMSE was 0.841. The best
level I or II
(SD= 4.1) cutoff score for MMSE to differentiate patients with PDD from the
42.5% male others was <25 (sensitivity of 80.5%, specificity of 73%).

89 PD patients
The best cutoff score for MoCA to differentiate patients with
Mean age 59 years According to level II of the
Almeida et al, 2019 36
PDD from those with PD-MCI was <18 (sensitivity of 85.5%,
Mean education 8.4 years MDS diagnostic criteria
specificity of 81.6%).
53.9% male

The MoCA test showed an AUC=0.906 area under the


50 PD patients
According to MDS ROC curve with a cutoff score of ≤19 points (sensitivity
Mean age 69.28 (SD= 11.41)
Camargo et al, 201644 criteria but did not of 87.80%, specificity of 88.89%). The MMSE had an 0.936
Mean schooling time 6.9 years
indicate if level I or II area under the curve with a cutoff score of ≤26 points
64% male
(specificity of 66.67%, sensitivity of up to 90.24%).
PD: Parkinson’s disease; PDD: Parkinson’s disease dementia; MDS: Movement Disorders Society; ILFT: interlocking finger test.

Studies conducted in Brazil have assessed the cognitive tests to confirm the PDD and DLB diagnoses. Routine
profile of patients with PD using comprehensive and screening tests are recommended to exclude other
formal neuropsychological tests, such as the Mattis causes of cognitive impairment. We suggest conducting
Dementia Rating Scale (MDRS), Scales for Outcomes in a complete blood count, as suggested in article one,
Parkinson’s Disease-Cognition (SCOPA-COG), Wisconsin and using structural imaging such as magnetic resonance
Card Sorting Test (WCST), Frontal Assessment Battery imaging (MRI) or computed tomography (CT) scans to
(FAB), Verbal Fluency Tests27,46, and Trail Making tests47. exclude other causes for dementia syndromes, such as
A study applying a comprehensive neuropsychological vascular dementia, stroke, and brain tumors.
battery showed cognitive impairment in 56.7% of PD The PDD and DLB biomarkers do not define the
patients in a waiting list for deep brain stimulation (DBS) presence of alpha-synuclein in vivo, but seek to reflect
implantation according to FAB scores and in 76.7% of them the repercussions of the pathophysiological process.
according to MoCA, showing the importance of a formal Specifically for PDD, the current diagnostic criteria do not
cognitive evaluation in this group of candidates to DBS48. recommend structural, functional, or electrophysiological
Moreover, another study showed that the Consortium studies for routine diagnostic purposes in differentiating
to Establish a Registry for Alzheimer’s Disease (CERAD) PD from PDD due to the lack of specificity1. Nevertheless,
neuropsychological battery could effectively assess the current MDS criteria for PD diagnosis21 has included
cognitive deficits in PD patients49. ancillary diagnostic tests.
Based on the available data, we recommend clinicians 123I-Meta-iodo-benzylguanidine scintigraphy
to apply at least one brief cognitive instrument, (123I-MIBG) is a method that provides a functional analysis of
such as MMSE and preferably the MoCA or ACE-R, the sympathetic postganglionic pathway, evaluating in vivo
to investigate PDD and DLB – especially since we the cardiac noradrenergic neurotransmission. 123I-MIBG
found no Brazilian studies or samples concerning scintigraphy documenting cardiac sympathetic denervation
neuropsychological profiles of the latter. However, is a supportive criterion for PD diagnosis, having over
for a tertiary center and other treatment strategies such 80% specificity to distinguish PD from other parkinsonian
as DBS, a comprehensive neuropsychological evaluation conditions21. A study by Leite et al. showed that this
is more appropriate if suitable. method identified cardiac sympathetic neurotransmission
impairment in Brazilian de novo PD patients without
Ancillary investigation and biomarkers clinically defined dysautonomia50. The DLB consortium
Besides analyzing the clinical history and conducting criteria also classifies 123I-MIBG as an indicative biomarker,
neurological examination with brief cognitive showing good sensitivity (69%) and specificity (87%)
instruments, we also recommend applying ancillary values for discriminating probable DLB from probable AD51.

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   77


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

Moreover, normal functional neuroimaging of biomarker for DLB diagnosis. Dopamine Transporter
the presynaptic dopaminergic system is currently (DAT) imaging has well-established utility in
considered an absolute exclusion criterion for the MDS distinguishing DLB from AD, with 78% of sensitivity and
PD criteria. However, the task force mentioned that PD 90% of specificity52. DAT imaging does not distinguish,
diagnosis does not require dopaminergic functional however, between degenerative parkinsonian
imaging21. Reduced dopamine transporter uptake in syndromes, such as progressive supranuclear palsy,
basal ganglia demonstrated by Single Photon Emission corticobasal syndrome, frontotemporal dementia
Computed Tomography (SPECT) or Positron Emission with parkinsonism, among others. Figure 2 shows
Tomography (PET) imaging is another indicative TRODAT-SPECT images in PD patients.

Dopamine transporter uptake in basal ganglia demonstrated by TRODAT-1 SPECT imaging. The superior row shows a normal uptake in the caudate and putamen. The medium row shows an asymmetric
uptake in a PD patient. The lower row shows a bilateral minimal uptake in a DLB patient. (Reproduced with permission from Dr. Artur Coutinho, Nuclear Medicine Center – Inrad – HC-FMUSP).
Figure 2. Dopamine transporter uptake in basal ganglia shown by SPECT or PET.

PSG is the third indicative biomarker for DLB association between global cognitive performance
diagnosis. It aims to confirm the clinical suspicion and wakefulness after sleep onset and the number of
of RBD, showing the loss of atony in the REM sleep stage2. sleep stage changes53.
A study by Sobreira et al. examined the association S u p p o r t i v e b i o m a r k e r s fo r D L B i n c l u d e :
between cognitive status and presence of sleep the relative preservation of medial temporal lobe
disorders in PD patients by PSG, finding a significant structures on CT/MRI scans since patients with

78   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

AD show greater atrophy of medial temporal lobe Furthermore, a prominent posterior slow-wave EEG
structures; the generalized low uptake on SPECT/PET activity with periodic fluctuations in the pre-alpha/theta
perfusion/metabolism scan, which shows reduced range is a supportive biomarker for DLB diagnosis56.
occipital activity; and the posterior cingulate island Some Brazilian studies have investigated the quantitative
sign on FDG-PET imaging. Figure 3 shows FDG-PET EEG changes concerning cognitive decline in PD patients,
showing increased posterior theta and delta amplitude in
images in a DLB patient54. MCI in Parkinson’s Disease
patients with MCI-PD or PDD57,58 and differences in EEG
presents a similar typical pattern of hypometabolism, power and coherence in AD and PDD59,60.
mainly in the posterior regions of the brain.
Therefore, the absence of this typical pattern hints Recommendation for primary and secondary care
at an alternative diagnosis, including depression or Figure 4 presents a flowchart of the approach to patients
an atypical parkinsonian syndrome55. with parkinsonism and dementia.

Upper row – Images on the left with standard axial view and images on the right with [18F]FDG-PET 3D-stereotactic surface projection (3D-SSP, Cortex ID Suite software,
GE Healthcare):occipital lobe metabolism is typically preserved in AD. Hypometabolism in AD usually occurs in bilateral temporoparietal regions. Lower row – Images on the left with
standard axial view and images on the right with [18F]FDG-PET 3D-stereotactic surface projection (3D-SSP, Cortex ID Suite software, GE Healthcare): DLB shows occipital hypometabolism.
Relative preservation of the posterior cingulate region, representing the “cingulate island sign”.
(Reproduced with permission from Dr. Artur Coutinho, Nuclear Medicine Center – Inrad – HC-FMUSP).
Figure 3. [18F]FDG-PET images in Alzheimer’s disease (AD) and Dementia with Lewy Bodies (DLB).

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   79


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

PD patients Probable/possible DLB

Risk factors:
Cognitive impairment and one of the following features:
Old age
Visual hallucinations
Low schooling
Fluctuating cognition (Spontaneous alertness and
Worse motor performance
concentration impairment; excessive daytime sleepiness)
Visual hallucinations
Primary care

Parkinsonism (one sign only)


Memory complaints
REM sleep behavior Disorders
Visuospatial dysfunction

Cognitive and functional


assessment (MMSE; FAQ)

Request neuroimage (brain CT or MRI) and screening blood test


Refer to secondary care

Clinical features and ancillary diagnostic test confirm diagnosis No


Secondary care

Start specific therapy Expand cognitive assessment (ACE-R; MoCA)


Assessment of RDB (specific questionnaires)

Persistence of diagnostic or clinical follow up difficulties. Refer to tertiary care


Tertiary care

Consider performing: Neuropsychological evaluation: More specific


FDG-PET Mattis scale therapeutic adjustment
TRODAT-SPECT SCOOPA-Cog
EEG RAVLT
Polysomnography NPI

Figure 4. Flowchart of the approach to patients with parkinsonism and dementia.

ACKNOWLEDGEMENTS
PC, LCS and RN are funded by CNPq, Brazil (bolsa de MTB, RMC, NAFF: drafting the manuscript; BJAPB,
produtividade em pesquisa). FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC, PHFB, RN,
Authors’ contributions. JBP, VT, HBF, MS, MTB, RMC, SMDB: critical revision of the manuscript for important
NAFF, JS, BJAPB, LPS: concept; JBP, VT, HBF, MS, intellectual content.

REFERENCES
1. Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C, Mizuno Y, et al. 4. Nascimento ER, Barbosa MA, Brasil VV, Sousa ALL, Amaral GF,
Clinical diagnostic criteria for dementia associated with Parkinson’s Jácomo PJ. Qualidade de vida de quem cuida de portadores de demência
disease. Mov Disord. 2007;22(12):1689-707. doi:10.1002/mds.21507. com corpos de Lewy. J Bras Psiquiatr. 2013;62(2):144-52. doi:10.1590/
2. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J, Weintraub D, S0047-20852013000200008
et al. Diagnosis and management of dementia with Lewy bodies: Fourth
5. Barbosa MT, Caramelli P, Maia DP, Cunningham MCQ, Guerra HL,
consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100.
Lima-Costa MF, et al. Parkinsonism and Parkinson’s disease in the elderly:
doi:10.1212/WNL.0000000000004058.
3. Carod-Artal FJ, Mesquita HM, Ziomkowski S, Martinez-Martin P. Burden A community-based survey in Brazil (the Bambuí Study). Mov Disord.
and health-related quality of life among caregivers of Brazilian Parkinson’s 2006;21(6):800-8. doi:10.1002/mds.20806.
disease patients. Parkinsonism Relat Disord. 2013;19(11):943-8. 6. Vale TC, Barbosa MT, Resende EPF, Maia DP, Cunningham MCQ,
doi:10.1016/j.parkreldis.2013.06.005. Guimarães HC, et al. Parkinsonism in a population-based study of

80   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

individuals aged 75+ years: The Pietà study. Parkinsonism Relat Disord. 27. Olchik MR, Ayres A, Ghisi M, Schuh AFS, Rieder CRM. The impact of
2018;56:76-81. doi:10.1016/j.parkreldis.2018.06.030. cognitive performance on quality of life in individuals with Parkinson’s
7. Herrera E Jr, Caramelli P, Silveira ASB, Nitrini R. Epidemiologic disease. Dement Neuropsychol. 2016;10(4):303-9. doi:10.1590/
survey of dementia in a community-dwelling Brazilian population. S1980-5764-2016DN1004008.
Alzheimer Dis Assoc Disord. 2002;16(2):103-8. doi:10.1097/00002093- 28. Pena-Pereira MA, Sobreira-Neto MA, Sobreira E, Chagas MHN,
200204000-00007. Oliveira DS, Rodrigues GR, et al. Validation of the brazilian Portuguese
8. Bottino CMC, Azevedo D Jr, Tatsch M, Hototian SR, Moscoso MA, version of the rapid eye movement sleep behavior disorder screening
Folquitto J, et al. Estimate of dementia prevalence in a community sample questionnaire (RBDSQ-BR). Arq Neuropsiquiatr. 2020;78(10):629-37.
from São Paulo, Brazil. Dement Geriatr Cogn Disord. 2008;26(4):291-9. doi:10.1590/0004-282X20200125.
doi:10.1159/000161053. 29. Schelp AO, Mendes-Chiloff CL, Bazan R, Paduan VC, Pioltini AB.
9. Takada LT, Caramelli P, Radanovic M, Anghinah R, Hartmann APBJ, Metabolic syndrome and dementia associated with Parkinson’s disease:
Guariglia CC, et al. Prevalence of potentially reversible dementias in impact of age and hypertension. Arq Neuropsiquiatr. 2012;70(2):114-8.
a dementia outpatient clinic of a tertiary university-affiliated hospital doi:10.1590/S0004-282X2012000200008.
in Brazil. Arq Neuropsiquiatr. 2003;61(4):925-9. doi:10.1590/ 30. Aarsland D, Cummings JL, Larsen JP. Neuropsychiatric differences
s0004-282x2003000600007. between Parkinson’s disease with dementia and Alzheimer’s
10. Souza RKM, Barboza AF, Gasperin G, Garcia HDBP, Barcellos PM, disease. Int J Geriatr Psychiatry. 2001;16(2):184-91. doi:10.1002/
Nisihara R. Prevalence of dementia in patients seen at a private hospital 1099-1166(200102)16:2<184::aid-gps304>3.0.co;2-k.
in the Southern Region of Brazil. Einstein (Sao Paulo). 2019;18:eAO4752. 31. Oliveira FF, Machado FC, Sampaio G, Marin SMC, Naffah-Mazzacoratti MG,
doi:10.31744/einstein_journal/2020AO4752. Bertolucci PHF. Neuropsychiatric feature profiles of patients with Lewy
11. Correa Ribeiro PC, Souza Lopes C, Alves Lourenço R. Prevalence of body dementia. Clin Neurol Neurosurg. 2020;194:105832. doi:10.1016/
dementia in elderly clients of a private health care plan: A study of the j.clineuro.2020.105832.
FIBRA-RJ, Brazil. Dement Geriatr Cogn Disord. 2013;35(1-2):77-86. 32. Kummer A, Harsányi E, Dias FMV, Cardoso F, Caramelli P, Teixeira AL.
doi:10.1159/000345984. Depression impairs executive functioning in parkinson disease patients
12. Camargo CHF, Retzlaff G, Justus FF, Resende M. Patients with dementia with low educational level. Cogn Behav Neurol. 2009;22(3):167-72.
syndrome in public and private services in southern Brazil. Dement e doi:10.1097/WNN.0b013e3181b278f9.
Neuropsychol. 2015;9(1):64-70. doi:0.1590/S1980-57642015DN91000010 33. Oliveira GSR, Bressan L, Balarini F, Jesuino e Silva RSJ, Brito MMCM,
13. Vieira RT, Barros NM, Caixeta L, Machado S, Silva AC, Nardi AE. Clinical Foss MP, et al. Direct and indirect assessment of functional abilities in
diagnosis of 80 cases of dementia in a university hospital TT. J Bras patients with parkinson’s disease transitioning to dementia. Dement
Psiquiatr. 2013;62(2):139-43. doi:10.1590/S0047-20852013000200007. Neuropsychol. 2020;14(2):171-7. doi:10.1590/1980-57642020dn14-020011.
14. Suemoto CK, Ferretti-Rebustini REL, Rodriguez RD, Leite REP, 34. Oliveira GN, Souza CP, Foss MP, Tumas V. An analysis of the cognitive
Soterio L, Brucki SMD, et al. Neuropathological diagnoses and clinical items of the movement disorders society checklist for the diagnosis of
correlates in older adults in Brazil: A cross-sectional study. PLoS Med. dementia in patients with Parkinson’s disease. Parkinsionism Relat Disord.
2017;14(3):e1002267. doi:10.1371/journal.pmed.1002267. 2015;21(10):1260-3. doi:10.1016/j.parkreldis.2015.08.005.
15. Baldivia B, Brucki SMD, Batistela S, Esper JC, Augusto CD, Rocha MSG.
35. Breder R, Leite MAA, Pinto JA, Cavalcante IP, Pessoa BL, Neves MAO.
Dementia in parkinson’s disease: a Brazilian sample. Arq Neuropsiquiatr.
Low Sensitivity of the Mini-Mental State Examination for Cognitive
2011;69(5):733-8. doi:10.1590/s0004-282x2011000600002.
Assessment of Brazilian Patients with Parkinson Disease. J Geriatr
16. Tedrus GMAS, Fonseca LC, Letro GH, Bossoni AS, Samara AB. Dementia
Psychiatry Neurol. 2017;30(6):311-5. doi:10.1177/0891988717731826.
and mild cognitive impairment in Parkinson’s disease. Arq Neuropsiquiatr.
36. Almeida KJ, Sá Carvalho LCL, Holanda Monteiro THOD, Gonçalves Júnior PCJ,
2009;67(3-B):957-9. doi:10.1590/S0004-282X2009000600038.
Campos-Sousa RN. Cut-off points of the Portuguese version of the
17. Camargo CHF, Jobbins VA, Serpa RA, Berbetz FA, Sabatini JS,
Montreal Cognitive Assessment for cognitive evaluation in Parkinson’s
Teive HAG. Association between olfactory loss and cognitive deficits
disease. Dement Neuropsychol. 2019;13(2):210-5. doi:10.1590/
in Parkinson’s disease. Clin Neurol Neurosurg. 2018;173:120-3.
1980-57642018dn13-020010.
doi:10.1016/j.clineuro.2018.08.018.
37. Almeida KJ, Macêdo LP, Lopes JLMLJ, Bor-Seng-Shu E, Campos-Sousa RN,
18. Dubois B, Burn D, Goetz C, Aarsland D, Brown RG, Broe GA,
Barbosa ER. Modified Pfeffer Questionnaire for Functional Assessment
et al. Diagnostic procedures for Parkinson’s disease dementia:
in Parkinson Disease. J Geriatr Psychiatry Neurol. 2017;30(5):261-6.
recommendations from the movement disorder society task force.
Mov Disord. 2007;22(16):2314-24. doi:10.1002/mds.21844. doi:10.1177/0891988717720298.
19. McKeith IG, Ferman TJ, Thomas AJ, Blanc F, Boeve BF, Fujishiro H, et al. 38. Rocha MSG, Bassetti EM, Oliveira MO, Kuark RGB, Estevam NM,
Research criteria for the diagnosis of prodromal dementia with Lewy bodies. Brucki SMD. Addenbrooke’s Cognitive Examination-Revised is accurate
Neurology. 2020;94(17):743-55. doi:10.1212/WNL.0000000000009323. for detecting dementia in Parkinson’s disease patients with low
20. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical educational level. Dement Neuropsychol. 2014;8(1):20-5. doi:10.1590/
diagnosis of idiopathic Parkinson’s disease: A clinico-pathological S1980-57642014DN81000004.
study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181-4. 39. Souza CP, Oliveira GN, Foss MP, Tumas V. The interlocking finger test in
doi:10.1136/jnnp.55.3.181. patients with Parkinson’s disease and healthy subjects. J Clin Neurosci.
21. Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. 2016;29:145-8. doi:10.1016/j.jocn.2015.09.026.
MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 40. Amaral-Carvalho V, Caramelli P. Normative data for healthy middle-
2015;30(12):1591-601. doi:10.1002/mds.26424. aged and elderly performance on the Addenbrooke Cognitive
22. Litvan I, Goldman JG, Tröster AI, Schmand BA, Weintraub D, Petersen RC, Examination-Revised. Cogn Behav Neurol. 2012;25(2):72-6. doi:10.1097/
et al. Diagnostic criteria for mild cognitive impairment in Parkinson’s WNN.0b013e318259594b.
disease: Movement Disorder Society Task Force guidelines. Mov Disord. 41. César KG, Yassuda MS, Porto FHG, Brucki SMD, Nitrini R. Addenbrooke’s
2012;27(3):349-56. doi:10.1002/mds.24893. cognitive examination-revised: normative and accuracy data for seniors
23. Walker Z, Possin KL, Boeve BF, Aarsland D. Lewy body dementias. Lancet. with heterogeneous educational level in Brazil. Int Psychogeriatr.
2015;386(10004):1683-97. doi:10.1016/S0140-6736(15)00462-6. 2017;29(8):1345-53. doi:10.1017/S1041610217000734.
24. Barbosa ER, LeFèvre BH, Comerlatti LR, Scaff M, Canelas HM. 42. Apolinario D, Santos MF, Sassaki E, Pegoraro F, Pedrini AVA, Cestari B,
Neuropsychologic dysfunctions in Parkinson’s disease: a study of 64 et al. Normative data for the Montreal Cognitive Assessment (MoCA) and
cases. Arq Neuropsiquiatr. 1987;45(2):109-18. doi:10.1590/S0004- the Memory Index Score (MoCA-MIS) in Brazil: Adjusting the nonlinear
282X1987000200003. effects of education with fractional polynomials. Int J Geriatr Psychiatry.
25. Oliveira FF, Machado FC, Sampaio G, Marin SM, Chen ES, Smith MC, 2018;33(7):893-9. doi:10.1002/gps.4866.
et al. Contrasts Between Patients With Lewy Body Dementia Syndromes 43. Sobreira E, Pena-Pereira MA, Eckeli AL, Sobreira-Neto MA, Chagas MHN,
and APOE-ε3/ε3 Patients With Late-onset Alzheimer Disease Dementia. Foss MP, et al. Screening of cognitive impairment in patients with
Neurologist. 2015;20(2):35-41. doi:10.1097/nrl.000000000000004. parkinson’s disease: Diagnostic validity of the Brazilian versions of
26. Pena MCS, Sobreira EST, Souza CP, Oliveira GN, Tumas V, Vale FAC. the montreal cognitive assessment and the addenbrooke’s cognitive
Visuospatial cognitive tests for the evaluation of patients with Parkinson’s examination-revised. Arq Neuropsiquiatr. 2015;73(11):929-33.
disease. Dement Neuropsychol. 2008;2(3):201-5. doi:10.1590/ doi:10.1590/0004-282X20150156.
S1980-57642009DN20300007. 44. Camargo CHF, Tolentino ES, Bronzini A, Ladeira MA, Lima R,
27. Machado FC, Oliveira FF, Marin SMC, Sampaio G, Bertolucci PHF. Schultz-Pereira GL, et al. Comparison of the use of screening tools for
Correlates of neuropsychiatric and motor tests with language assessment evaluating cognitive impairment in patients with Parkinson’s disease.
in patients with lewy body dementia. Rev Psiquiatr Clin. 2020;47(3):75-81. Dement Neuropsychol. 2016;10(4):344-50. doi:10.1590/s1980-5764-
doi:10.1590/0101-60830000000236. 2016dn1004015.

Parmera JB, et al.   Brazilian consensus on Parkinson’s disease dementia.   81


Dement Neuropsychol 2022 September;16(3 Suppl. 1):69-82

45. Tumas V, Borges V, Ballalai-Ferraz H, Zabetian CP, Mata IF, Brito MMC, 53. Sobreira EST, Sobreira-Neto MA, Pena-Pereira MA, Chagas MHN,
et al. Some aspects of the validity of the Montreal Cognitive Assessment Fernandes RMF, Eckeli AL, et al. Global cognitive performance is associated
(MoCA) for evaluating cognitive impairment in Brazilian patients with with sleep efficiency measured by polysomnography in patients with
Parkinson’s disease. Dement Neuropsychol. 2016;10(4):333-8. Parkinson’s disease. Psychiatry Clin Neurosci. 2019;73(5):248-53.
doi:10.1590/S1980-5764-2016DN1004013. doi:10.1111/pcn.12819.
46. Sobreira EST, Pena MCS, Silva Filho JH, Souza CP, Oliveira GN, 54. Higuchi M, Tashiro M, Arai H, Okamura N, Hara S, Higuchi S, et al. Glucose
Tumas V, et al. Executive cognitive tests for the evaluation of patients hypometabolism and neuropathological correlates in brains of dementia
with Parkinson’s disease. Dement Neuropsychol. 2008;2(3):206-10. with Lewy bodies. Exp Neurol. 2000;162(2):247-56. doi:10.1006/
doi:10.1590/S1980-57642009DN20300008. exnr.2000.7342.
47. Olchik MR, Ghisi M, Freiry AM, Ayres A, Schuh AFS, Rieder CRM, et al. 55. Nobili F, Arbizu J, Bouwman F, Drzezga A, Agosta F, Nestor P, et al.
Comparison trail making test between individuals with parkinson’s disease European Association of Nuclear Medicine and European Academy of
and health controls: Suggestions of cutoff point. Psychol Neurosci. Neurology recommendations for the use of brain 18 F-fluorodeoxyglucose
2017;10(1):77-82. doi:10.1037/pne0000076. positron emission tomography in neurodegenerative cognitive impairment
48. Machado FA, Rieder CR, Hilbig A, Reppold CT. Neuropsychological and dementia: Delphi consensus. Eur J Neurol. 2018;25(10):1201-1217.
profile of Parkinson’s disease patients selected for deep brain stimulation doi:10.1111/ene.13728.
surgery. Dement Neuropsychol. 2016;10(4):296-302. doi:10.1590/S1980- 56. Schumacher J, Taylor JP, Hamilton CA, Firbank M, Cromarty RA,
5764-2016DN1004007. Donaghy PC, et al. Quantitative EEG as a biomarker in mild cognitive
49. Camargo CHF, Bronzini A, Tolentino ES, Medyk C, Schultz-Pereira GL. impairment with Lewy bodies. Alzheimers Res Ther. 2020;12(1):82.
Can the CERAD neuropsychological battery be used to assess cognitive doi:10.1186/s13195-020-00650-1.
impairment in parkinson’s disease? Arq Neuropsiquiatr. 2018;76(3):145-9. 57. Fonseca LC, Tedrus GMAS, Letro GH, Bossoni AS. Dementia, mild cognitive
doi:10.1590/0004-282X20180003. impairment and quantitative EEG in patients with Parkinson’s disease. Clin
50. Leite MAA, Nascimento OJM, Pereira JS, Amaral C, Mesquita CT, EEG Neurosci. 2009;40(3):168-72. doi:10.1177/155005940904000309.
Azevedo JC, et al. Cardiac 123I-MIBG uptake in De novo Brazilian 58. Pereira JS, Pimentel MLV, Gomes PLH. Distúrbios cognitivos na doença
patients with Parkinson’s disease without clinically defined dysautonomia. de Parkinson: correlações eletrencefalográficas. Arq Neuropsiquiatr.
Arq Neuropsiquiatr. 2014;72(6):430-4. doi:10.1590/0004-282x20140042. 1995;53(1):11-5. doi:10.1590/S0004-282X1995000100002.
51. Yoshita M, Arai H, Arai H, Arai T, Asada T, Fujishiro H, et al. Diagnostic 59. Sandmann MC, Piana ER, Sousa DS, Bittencourt PRM. Digital EEG
accuracy of123I-meta-iodobenzylguanidine myocardial scintigraphy with brain mapping in Alzheimer’s dementia and Parkinson’s disease:
in dementia with lewy bodies: A multicenter study. PLoS One. a prospective controlled study. Arq Neuropsiquiatr. 1996;54(1):50-6.
2015;10(3):e0120540. doi:10.1371/journal.pone.0120540. doi:10.1590/S0004-282X1996000100009.
52. McKeith I, O’Brien J, Walker Z, Tatsch K, Booij J, Darcourt J, et al. 60. Fonseca LC, Tedrus GMAS, Carvas PN, Machado ECFA. Comparison of
Sensitivity and specificity of dopamine transporter imaging with 123I-FP- quantitative EEG between patients with Alzheimer’s disease and those with
CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Parkinson’s disease dementia. Clin Neurophysiol. 2013;124(10):1970-4.
Lancet Neurol. 2007;6(4):305-13. doi:10.1016/S1474-4422(07)70057-1. doi:10.1016/j.clinph.2013.05.001.

82   Brazilian consensus on Parkinson’s disease dementia.   Parmera JB, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S106EN

Treatment of dementia:
recommendations of the Scientific Department
of Cognitive Neurology and Aging of the
Brazilian Academy of Neurology
Paulo Caramelli1 , Valeska Marinho2 , Jerson Laks3,4 , Marcus Vinicius Della Coletta5 ,
Florindo Stella6,7 , Einstein Francisco Camargos8,9 , Jerusa Smid10 ,
Breno José Alencar Pires Barbosa10,11,12 , Lucas Porcello Schilling13,14,15 ,
Marcio Luiz Figueredo Balthazar16 , Norberto Anízio Ferreira Frota17,18 , Leonardo Cruz de Souza1 ,
Francisco Assis Carvalho Vale19 , Márcia Lorena Fagundes Chaves20,21 , Sonia Maria Dozzi Brucki10 ,
Ricardo Nitrini10 ,Helen Bedinoto Durgante22 , Paulo Henrique Ferreira Bertolucci23

1
Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
2
Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Centro para Doença de Alzheimer, Rio de Janeiro RJ, Brazil.
3
Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Programa de Pós-Graduação em Psiquiatria e Saúde Mental, Rio de Janeiro RJ, Brazil.
4
Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Rio de Janeiro RJ, Brazil.
5
Universidade do Estado do Amazonas, Manaus AM, Brazil.
6
Universidade Estadual Paulista, Instituto de Biociências, Campus de Rio Claro SP, Brazil.
7
Universidade de São Paulo, Faculdade de Medicina, Departamento e Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil.
8
Hospital Universitário de Brasília, Centro de Medicina do Idoso, Brasília DF, Brazil.
9
Universidade de Brasília, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Brasília DF, Brazil.
10
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
11
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
12
Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.
13
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
14
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
15
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
16
Universidade Estadual de Campinas, Departamento de Neurologia, Faculdade de Ciências Médicas, Campinas SP, Brazil.
17
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
18
Universidade de Fortaleza, Fortaleza CE, Brazil.
19
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brazil.
20
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
21
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brazil.
22
Universidade Federal de Pelotas, Pelotas RS, Brazil.
23
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
Correspondence: Paulo Caramelli; Email: caramelli@ufmg.br.
Conflict of interest: PC: Participation as principal investigator in clinical trials for Novo Nordisk and Roche laboratories. Participation in advisory boards for Aché,
Biogen, EMS, Nutricia and Roche laboratories. Development of continuing medical education materials and participation as speaker in symposia sponsored by
Aché, Nutricia, Libbs, Roche, Sandoz, Torrent and Zodiac laboratories; VM: Development of continuing medical education materials and participation as speaker in
symposia sponsored by Biogen, Sandoz and Torrent laboratories; JS: Participation as speaker in symposia sponsored by Roche laboratory; LPS: Participation in
advisory boards for Biogen. Participation as speaker in symposia sponsored by Aché, Apsen and Biogen laboratories; MLFB: Participation in advisory boards for
Biogen. Development of continuing medical education materials and participation as speaker in symposia sponsored by EMS and Torrent laboratories; PHFB:
Participation in advisory boards for Biogen and Novo Nordisk laboratories. Supervision of training activities for Biogen, Janssen-Cilag and Novo Nordisk laboratories
and for Quintiles. Participation as speaker in symposia sponsored by Apsen, Nutricia, Roche and Sandoz laboratories; LCS: Participation in advisory board for Biogen.
Participation as speaker in symposia sponsored by Biogen; RN: Participation in advisory board for Biogen; JL, MVDC, FS, EFC, BJAPB, NAFF, FACV, MLFC, SMDB,
HBD: No conflict of interest to declare.
Received on February 20, 2022; Received in its final form on April 27, 2022; Accepted on April 27, 2022.

Caramelli P, et al.   Dementia care.   83


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

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.

TRATAMENTO DA DEMÊNCIA: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO DA ACADEMIA


BRASILEIRA DE NEUROLOGIA
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 an early deficit in cholinergic activity in mild to

R esearch estimates that by 2050 about 152 million


people will have dementia, or one new case every
three seconds, mostly from low- and middle-income
moderate stages of dementia due to AD6. Cholines-
terase inhibitors (ChEI) donepezil, galantamine, and
rivastigmine inhibit the ChE enzyme and increase
countries (LMIC). Currently, about two thirds of people acetylcholine availability in the brain. These drugs are
diagnosed with dementia (PwD) live in LMIC1, added to approved for treating mild and moderate AD dementia,
a high estimate of underdiagnosed cases—approximate- but have also shown positive effects in severe stages7.
ly 800.000 people in Brazil—according to recent data2. For moderate-to-severe AD dementia, studies recom-
Dementia is one of the leading causes of disability in mend combining ChEI with the glutamatergic NMDA
older adults, with significant impacts on the autonomy and receptor antagonist memantine.
quality of life (QoL) of PwD and their families. Neurode- Despite modest effects, use of ChEI and meman-
generative diseases and cerebrovascular disorders are the tine have shown improvement in disease symptoms
main causes of dementia, with Alzheimer’s disease (AD) or slower progression when compared with placebo.
accounting for more than half of the cases3. In Brazil, Clinical benefits can be seen in cognitive, behavioral,
AD ranked among the top ten causes of deaths in 20194. and functional domains5,7. Despite conflicting evidence,
Other degenerative causes include frontotemporal demen- research shows that all ChEI may reduce AD mortality
tia (FTD), Parkinson’s disease (PD) dementia (PDD) and
and galantamine may reduce the risk of conversion to
dementia with Lewy bodies (DLB).
severe dementia8.
Although we currently lack a cure for neurodegen-
When prescribing these drugs, physicians must
erative or vascular dementias, some pharmacological
clarify to PwD and caregivers that the expected clinical
and non-pharmacological interventions can contribute
to alleviate symptoms, slow disease progression and effects are usually modest, sometimes characterized
improve QoL5. Therapeutic approaches are based on only by stabilization or slower worsening. Therapy
etiology, symptom profile and stage of dementia. Thus, should begin with the lowest dosage to avoid adverse
this manuscript presents updated information and rec- events (AEs) and be increased at least every four weeks
ommendations on current available treatments in Brazil for ChEI and one week for memantine. Regarding ChEI,
for dementia due to AD, vascular cognitive impairment usage should reach the therapeutic doses, i.e., 5-10mg
(VCI), FTD, PDD and DLB. 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 pre-
TREATMENT OF DEMENTIA DUE TO AD scribed for selected cases). Ideally, the highest dose
within the therapeutic range of each drug should be
Pharmacological treatment targeted, given the greater potential for clinical ben-
Degeneration of basal forebrain cholinergic neurons efits. Figure 1 illustrates the dose regimen for drug
precedes clinical manifestations in AD, thus implying therapy of AD dementia.

84   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

4 weeks 4 weeks 4 weeks

Donepezil Donepezil
tablet 5mg 1x/d tablet 10mg 1x/d

Galantamine Galantamine Galantamine


capsule 8mg 1x/d capsule 16mg 1x/d capsule 24mg 1x/d

Rivastigmine
Rivastigmine Rivastigmine capsule 6mg 2x/d
Rivastigmine
capsule 1.5mg 2x/d or capsule 3mg 2x/d or
capsule 4.5mg 2x/d
patch 4.6mg 1x/d patch 9.5mg 1x/d Rivastigmine
patch 9.5mg 1x/d1

Memantine2 Memantine Memantine Memantine


1 week 1 week 1 week
5mg 1x/d 5mg 2x/d 2x/d – 10mg and 5mg 10mg 2x/d or 20mg 1x/d

Treatment of comorbidities
Non-phamacological interventions
1
In case of improvement or stabilization, the highest dose of rivastigmine patch can be postponed; 2memantine is indicated for treating moderate or severe AD dementia.
Figure 1. Overview of pharmacological treatment for AD dementia.

Common AEs consist of nausea, vomiting, diarrhea, • Caution is required when prescribing ChEI to
headache and insomnia, and are less likely to occur individuals with chronic obstructive pulmonary
if medication is taken after a meal. Donepezil should disease or asthma;
be administered at bedtime; in case of insomnia or • Discontinuation or switching (in the case of ChEI)
nightmares, it can be taken in the morning. Less com- shall be considered when there is no clinical ben-
mon, but more serious AEs are bradyarrhythmia and efit or in case of relevant AEs;
syncope. Treatment effects and duration vary from • Avoid abrupt discontinuation of ChEI or memantine;
person to person, and any positive effect may be lost • In the case of AEs with higher doses, dose reduc-
over time. No research has confirmed the superiority of tion may be considered, noting whether there is
one ChEI over the others, despite some specificities on a decrease in AEs as well as changes in cognitive,
the mechanisms of action and frequency/types of AEs. functional, or neuropsychiatric symptoms;
Switching to another ChEI is encouraged, particularly
• Discontinue medication in the absence of clinical
when no clinical benefit is observed after a few months
benefits or if AEs persist.
of follow-up or in case of AEs that do not resolve. With
progression to moderate dementia, the association
Pharmacological treatment of neuropsychiatric symptoms
with memantine is indicated5,9,10. Memantine is usually
Neuropsychiatric symptoms may precede the cogni-
well tolerated; the most common AEs are drowsiness,
dizziness and headache5,7. tive symptoms in AD, becoming more common at the
second half of the mild dementia stage, increasing
Practical issues for prescribing and discontinuing ChEI along the moderate stage and usually decreasing at
and memantine severe stage.
• ChEI increase vagal tone and are contraindicated Clinical trials specifically addressing neuropsychi-
in patients with baseline bradycardia or known atric symptoms in AD have been disappointing. The
cardiac conduction system disorder (i.e., atrio- benefits of ChEI tend to favor specific neuropsychiatric
ventricular block > 1st degree). Caution is required symptoms, such as depression, anxiety, dysphoria,
when any of the three ChEI are used in combina- and apathy11. For depression, which may be present in
tion with drugs that induce bradycardia or alter more than half of AD patients, there is some evidence
atrioventricular conduction (e.g., beta blockers or for the indication of sertraline or mirtazapine as the
calcium channel blockers); best antidepressant options12. A recent clinical trial,

Caramelli P, et al.   Dementia care.   85


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

however, observed a trend to increased mortality with scanter. Studies with brexpiprazole are ongoing, but
mirtazapine used to treat agitation in AD dementia13. evidence of efficacy and safety has not been definitively
Aggressiveness is a common neuropsychiatric established to date17. Although some studies suggest
symptom which may contribute to agitation and in- that cannabinoids may have potential positive effects in
creased caregiver burden. Treatment encompasses both neurodegenerative diseases, there is no current evidence
pharmacological and non-pharmacological interven- to support their clinical use in dementia18.
tions (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 Pharmacological treatment for AD-related conditions
initial effect, it may be lost after some time. Given the AD may have associated complications. Parkinson-
absence of approved pharmacological treatment for ism may be present in 9% to 70%, depending on the
neuropsychiatric symptoms in AD dementia, a sequence disease stage. There are differences with typical PD,
of drugs may be considered for treatment of agitation with tremor being mild or absent, and usually pre-
and aggressiveness14. According to this algorithm14, senting symmetrical bradykinesia and rigidity from
first-line treatment would be atypical antipsychotics, the start. Levodopa may be tried, but response is less
the first choice being risperidone, followed by aripip- likely than in PD.
razole or quetiapine. If these drugs fail or are offset by Between 10% and 20% of AD patients will have at
AEs, carbamazepine may be an option. In case of a new least one seizure. Newer anticonvulsants (e.g., lamo-
failure the next step would be citalopram, followed by trigine or levetiracetam) are first-line treatments for
gabapentin and prazosin. Citalopram is a good thera- epilepsy in PwD19.
peutic option for agitation. Benzodiazepines must be
avoided in dementia due to AD. Non-pharmacological treatments for AD:
Another common neuropsychiatric symptom is cognitive symptoms
sleep disorder. The first step to a successful treat-
ment of sleep disorder in AD is to ask why and how Cognitive stimulation therapy (CST)
it occurs. A sleep disorder may be the consequence Recommended treatment according to the National
of a sleep wake cycle inversion, and in this case sleep Institute for Health and Clinical Excellence of the
United Kingdom20 to stimulate language, memory,
hygiene and non-pharmacological interventions may
executive functions; approved by the Alzheimer’s
be helpful. For pain or another discomfort, the cause
Disease International and used in many countries21.
should be addressed; issues related to sundowning,
CST was shown to improve cognition, QoL, neuropsy-
fragmented sleep or difficulty in starting sleep should
chiatric symptoms in PwD from LMIC22,23. CST and its
be addressed. The sleep may be interrupted by de-
implementation manual have been adapted for use in
lusions and hallucinations, when an antipsychotic,
Brazil24 and the intervention was found to be feasible
particularly with sedative effects (e.g., quetiapine)
and beneficial to improve mood in PwD in a recent
may be useful. There is no evidence that hypnotics
clinical trial25.
or mirtazapine have positive effects. Trazodone
might improve sleep, although the effects are usually Multicomponent/multidisciplinary cognitive rehabilitation
modest15. A recent Brazilian study has shown that program (MCRP)
short-term use (i.e., 14 days) of zopiclone or zolp- MCRP includes cognitive rehabilitation, stimulation and
idem may be clinically helpful in treating insomnia computer-assisted therapy, physical training, physio-
in AD dementia16. therapy, reading and games. This program may decrease
Delusions and hallucinations may be present in up to depressive symptoms and improve QoL indicators of
one third of AD patients and indicate poorer prognosis, PwD/caregivers. However, MCRP effects on cognition
with increased functional impairment and higher mor- are questionable23,26.
tality. Treatment of these neuropsychiatric symptoms
includes avoiding typical antipsychotics, due to AEs and Reality orientation (RO)
cognitive worsening. There is evidence of a modest effect Neurosensory stimulation to reorient daily activities may
of ChEI, but often there is a need of using antipsychotics, improve cognition and adherence to pharmacological
and in this case the evidence is stronger for risperidone treatment in mild-moderate AD dementia27,28. Results for
and olanzapine, while for quetiapine the evidence is neuropsychiatric symptoms are inconclusive29.

86   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

Non-pharmacological treatments for AD: blood flow, hippocampal volume, neurogenesis, also
neuropsychiatric symptoms neuropsychiatric symptoms41. Occupational therapy
To increase the chance of success with a non-pharma- (error-reduction techniques)42 and physiotherapy43,44
cological treatment it is essential to adopt an individu- tend to enhance functioning and delay physical
alized approach that considers: decline; results are inconclusive for improvement
• When and how the neuropsychiatric symptom in cognition. Early interventions targeting lifestyle
started? (Was there a trigger?) modifiable risk factors (arterial hypertension, di-
• How did it evolve? (Are there associated symptoms?) abetes, obesity, physical inactivity, high alcohol
• How did it end? consumption, smoking, social isolation), clinical
• What are the associated worsening and improving conditions (hearing loss and depression), and poor
factors? nutrient intake, may work as protective measures for
reducing AD risk1,10,45,46.
Psychosocial/psychoeducational interventions
These are group or person-centered approaches to in-
form the management and care of dementia, including TREATMENT OF VASCULAR COGNITIVE IMPAIRMENT
emotional and social stimulation of PwD/caregivers. The general principles of approach and treatment for VCI
Face-to-face or technology-based multicomponent are the same as for dementia due to AD. They encompass
psychoeducational and Cognitive-Behavioral Therapy the treatment of comorbidities, including neuropsychiat-
approaches (cognitive reframing, self-monitoring, ric symptoms, providing information and support to PwD
mood management techniques, mindfulness-based and caregivers, and maintaining independence.
training) may improve neuropsychiatric symptoms
(agitation, restlessness, anxiety, sleep disturbances), Primary and secondary prevention
delay the progression of cognitive impairment, and The multidisciplinary team should focus primarily on
benefit caregiver confidence, self-efficacy, burden, controlling the main risk factors for new cerebrovascular
depression, stress levels, and QoL of the dyad30-33. Posi- events and cognitive impairment in VCI. Although we
tive Psychology practices are recommended for mental recognize risk factors for post-stroke dementia, such as
health promotion of PwD/caregivers according to the low education and diabetes mellitus47, factors for stroke
Alzheimer’s Association Psychosocial Measurement itself have long been known and controllable, including
Workgroup 33 and the European INTERDEM Social smoking, arterial hypertension, diabetes mellitus and
Health Taskforce34,35. dyslipidemia (notably high LDL-cholesterol levels)48.
However, in recent decades several studies have
Reminiscence therapy (RT) proposed measuring the weight of these risk factors on
RT is a group-based intervention to recall autobiograph- the appearance of new strokes, on post-stroke cognition
ical memories. It is used for mild-moderate dementia and functionality, often with conflicting results. Based
with modest evidence to improve depressive symptoms, on these observations we recommend:
mood, cognition22, and well-being36. Lacking theoretical • Blood pressure control: There is strong evidence
basis and methodological systematization make RT that increased blood pressure levels are associated
effects questionable37. with stroke48. There are still no studies that have
confirmed the impact of blood pressure control on
Music therapy (MT)/dancing worsening cognitive impairment. Intensive blood
Significant positive effects were found of MT on mem- pressure control (systolic pressure of 120 versus
ory38, emotional/mood22 and neuropsychiatric symp- 140 mmHg) does not show a reduction in the
toms27. Dancing may also lead to beneficial effects on incidence of dementia (all etiologies). In frail
cognition and psychical health20. older adults, with symptoms of orthostatic hy-
potension and increased risk of falls, individu-
Physical-related/lifestyle-related interventions alize blood pressure targets to avoid symptoms
Repetitive transcranial magnetic stimulation (rTMS) of hypotension;
and acupuncture seem to improve cognitive functions, • Glycemic control: There is strong evidence that
whereas acupuncture, exercise and light therapy pre- increased blood glucose levels are associated
sented potential to enhance functional performance22 with stroke 48. However, there is no evidence
and cognition 39,40 . Long-term exercise improves that strict glycemic control in older adults

Caramelli P, et al.   Dementia care.   87


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

prevents cardiovascular events. Studies have • Diet and supplements: A balanced diet, rich in
shown that tight blood glucose control in older vegetables and fruits, impacts many organ systems.
adults was associated with greater frailty and One recent systematic review with metanalysis has
mortality, with no benefit in the evolution of shown that high adherence to the Mediterranean
dementia49,50. Acceptable fasting glycemic values​​ diet reduced the risk of global cognitive decline in
in individuals with VCI should be around up non-demented older adults59. The use of vitamin
to 150 mg/dL and postprandial < 180 mg/dL, and mineral supplements should only be indicated
as well as glycated hemoglobin targets should in cases with insufficiency. It is not recommended
be less stringent (e.g., < 8%)51; to use vitamin supplements for the prevention or
• Antiplatelet agents: There is strong evidence treatment of VCI.
to support using these agents for secondary
prevention of non-embolic stroke, either ace- Pharmacological treatment
tylsalicylic acid (81 to 100mg/day) or clopi-
dogrel (75mg/day)52. In cases of VCI without ChEI and memantine
evidence of previous stroke, therapy should The pharmacological treatment commonly prescribed
be individualized 53, especially in individuals in VCI, particularly vascular dementia, is mainly based
with significant risk of falls. In recent years, on ChEI and memantine. These medications are rou-
no studies have been published providing sup- tinely used in clinical practice to treat cognitive decline,
port for the benefits of antiaggregants in the functionality changes and neuropsychiatric symptoms.
evolution of VCI54; However, randomized controlled clinical trials have
• Statins: Statins are of interest in the secondary shown limited efficacy of these drugs.
prevention of cerebrovascular disease due to both Randomized, placebo-controlled studies demon-
their lipid-lowering and pleiotropic effects on strate little efficacy of ChEI in pure vascular demen-
vascular function, combining to inhibit athero- tia60,61. There is a statistically significant but clini-
sclerosis. There is good evidence that statins given cally slight cognitive benefit for cognitive functions,
late in life to people at risk for vascular disease mainly processing speed and executive functions62.
do not prevent cognitive decline or dementia55. In general, there is a slight attenuation of cognitive
For individuals with VCI and a history of ischemic decline compared to placebo61. Some studies report
(non-embolic) stroke, statin use should follow favorable results regarding functional performance,
secondary prevention recommendations with but the gain is not consistent. Improvement of neuro-
individual risk analysis. The use of statins in older psychiatric symptoms, when present, is also limited.
adults and subjects with vascular risk factors is On the other hand, agitation has been observed as an
not recommended for the sole purpose of primary AE of ChEI and memantine63,64.
prevention or treatment of dementia; Favorable results of ChEI seem more evident in
• Multidisciplinary interventions: In a system- mixed dementia (vascular + AD) compared to ‘pure’
atic review of 15 prospective studies with over vascular dementia65. Galantamine is approved in Brazil
30,000 individuals without dementia, involve- for treatment of mild to moderate AD dementia with
ment in at least one physical activity of minimal concomitant significant cerebrovascular disease and has
to moderate intensity was associated with a 35% shown positive effects on cognitive speed and QoL of
reduction in the relative risk of cognitive decline individuals with mixed dementia in a controlled clini-
in one to 12 years of follow-up56. In another study cal trial in the country66. Possible benefits of ChEI in
with 639 older adults without disability and with mixed dementia, although limited, are attributed to the
cerebral white matter changes, physical activity reduction of cholinergic depletion caused by concomi-
reduced the risk of cognitive and functional defi- tant AD60,65,66. There is insufficient clarity regarding the
cits, regardless of the severity of white matter differences in efficacy of ChEI, as well as memantine,
changes or other risk factors57. Hence, practice between multiple infarcts and subcortical vascular
of physical activity at mild to moderate intensity dementia64,67-69. Initiation of ChEI treatment in patients
should be strongly recommended to all individuals with severe dementia is not recommended.
at the primary care level as a means of primary and Among the AEs of ChEI already described above,
secondary prevention of VCI. On the other hand, bradycardia and arterial hypotension are the main
evidence that physical rehabilitation reduces post- conditions associated that require special attention in
stroke cognitive loss is still insufficient58; patients with vascular dementia62,65,67-71.

88   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

In current clinical practice, memantine is com- Considering that there are no specific treatments,
monly added to ChEI in subjects with moderate or care should focus on managing behavioral symptoms
severe vascular dementia. However, the benefits of and disability, and reducing caregiver burden that is
memantine are very limited or controversial for con- high in bvFTD. In a recent Brazilian study including
trolling cognitive and neuropsychiatric symptoms, 20 bvFTD and 30 AD individuals, Lima-Silva et al.
as well as for slowing cognitive or functional decline61. found out that bvFTD subjects presented higher levels
For this reason, the drug has not been approved for of neuropsychiatric symptoms and that their caregivers
treatment of vascular dementia. The few randomized, experienced higher levels of distress than AD caregivers.
placebo-controlled studies report that memantine is Moreover, bvFTD caregiver’s distress and burden were
well-tolerated and that drowsiness has been the AE related to cognitive and functional impairment79.
of major concern72. Multidisciplinary management may be needed
throughout the disease process, but this has not been
Treatment of neuropsychiatric symptoms in VCI specifically established for bvFTD treatment. A mul-
Overall, the neuropsychiatric symptoms in VCI are tidisciplinary team may offer a more comprehensive
like those observed in AD, and the treatment is sim- care management and seems to be an essential
ilar 14,73-75. The non-pharmacological interventions tool for dealing with other degenerative diseases80.
include occupational therapy, cognitive stimulation, However, the needs in bvFTD may be more specific
art therapy, involvement in social interaction activ- and studies regarding their management should be
ities, physical activity, and psychological support for conducted. Social, financial, psychological counseling
PwD and caregivers. may be required to address specific issues and more
Regarding pharmacological interventions, general knowledge on those topics should be gathered to help
services’ organization. For instance, the emergence
recommendations follow the commentaries made
of swallowing problems during dementia progression
above (AD section), namely, to avoid benzodiazepines,
may require specialized evaluation. Problems with
to prescribe antipsychotics in low doses and for strict-
inappropriate eating speed, passivity, coughing, and
ly necessary periods, antidepressants for depression,
choking beginning in the mild stages and following
anxiety, panic and mild agitation, and anticonvulsants
the worsening cognitive and behavioral declines were
when indicated.
recently reported and add information about specific
The pharmacological treatment of patients with
treatment needs81.
vascular dementia should be regularly monitored for Regarding pharmacological approaches, a recent
cardiovascular signs and symptoms. systematic review gathered information aiming to
identify pharmacological interventions that could
be used to treat the more troublesome behaviors in
TREATMENT OF FRONTOTEMPORAL DEMENTIA bvFTD. Trieu et al.78 found 23 studies (11 randomized
(BEHAVIORAL AND LANGUAGE VARIANTS) controlled trials, eight open-label studies, one proof-
Frontotemporal dementia (FTD) encompasses different of-concept study, and three case series) involving a
clinical syndromes associated with progressive behav- sample of 573 individuals. Sixteen out of the 23 stud-
ioral/personality deterioration or language disorders, ies used pharmacological interventions, resulting
and is considered the second most frequent cause of in benefits, as measured by the Neuropsychiatric
young-onset dementia76. Inventory, for trazodone, citalopram, rivastigmine,
paroxetine, ameliorating symptoms such as disinhi-
bition, hyperorality, and depression.
Treatment of behavioral variant frontotemporal dementia Two reviews82,83 published by Brazilian authors re-
The clinical syndrome associated with behavioral and port that most studies have small sample sizes, short
personality changes called behavioral variant (bvFTD) duration of treatment, and non-uniform measures while
is the most common clinical presentation and specific evaluating efficacy and tolerability.
diagnostic criteria are available for clinical use77. Portugal et al.82 conducted a systematic review and
There are no approved treatments for bvFTD, either reported better results for drugs with serotonergic
disease-modifying drugs or symptomatic treatments action, such as selective serotonin reuptake inhibi-
to ameliorate disturbing symptoms. Only a few small tors or SSRIs (paroxetine, citalopram, fluvoxamine)
studies have shown benefits of symptomatic pharma- and trazodone to treat behavioral symptoms, but not
cological approaches78. cognition. Authors suggest that using SSRIs as the

Caramelli P, et al.   Dementia care.   89


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

first-line treatment seems to be the best practice with left posterior temporal/parietal atrophy, and slow
until better data on evidence-based strategies to rely speech with long-word finding pauses, anomia,
upon are available. and repetition deficits84.
In a narrative review aiming to guide clinical prac- Up to this moment, there are no disease-modify-
tice, Gambogi et al.83 conducted a literature search ing agents approved for these language syndromes
on bvFTD treatment. According to drug class, ChEI associated with neurodegenerative diseases. Symp-
and memantine do not have significant therapeutic tomatic treatments aiming to improve language dis-
actions. Antidepressants (SSRIs and trazodone) im- orders have been shown to enhance oral and written
proved behavioral symptoms with better tolerability; naming abilities85. Language training therapies im-
antipsychotics reduced agitation (quetiapine, risperi- prove naming accuracy for trained items at short- and
done, aripiprazole), improper behaviors (risperidone long-term follow-up and are considered the standard
and aripiprazole), and scores on the Neuropsychiatric treatment for PPA85,86.
Inventory (olanzapine) in uncontrolled cases series There are very few studies on PPA treatment in
and clinical reports. However, mortality issues related Brazil and Latin-America, with small sample sizes,
to antipsychotic use in dementia, cardiovascular risks, demonstrating the lack of local information about
and motor AEs limit their use in bvFTD. Psychostim- language therapies in PPA87. Notwithstanding, an im-
ulants for apathy and anticonvulsants for abnormal portant piece of information was gathered by a Brazil-
behaviors were investigated, but must be used with ian systematic review organized by Carthery-Goulart
caution due to AEs. In a symptom-focused approach, et al.88. The literature on evidence-based strategies of
authors suggest SSRIs or trazodone as the first option language rehabilitation in PPA was summarized and
for disinhibition, compulsive/perseverative behaviors, the language behavioral treatments classified into
stereotyped behaviors, and hyperorality. Psychostimu-
impairment-directed and functional interventions.
lants may be an option for apathy and antipsychotics
The results suggest, as a practical option, the rec-
should be used with care and reserved to manage
ommendations for the use of impairment-directed
challenging and hazardous behaviors.
therapies aimed at naming and lexical retrieval in
In summary, several low-quality reports are fo-
svPPA. In relation to nfvPPA and lvPPA, the small
cusing on symptomatic treatment for bvFTD and
number of studies limits conclusions about the best
no specific treatment proved to be effective to date.
therapeutic option for such variants88.
ChEI and memantine should not be used as they
While impairment-directed intervention targets
have not shown any significant therapeutic action.
the remediation or slowing the progression of specific
SSRIs or trazodone seem the best practice to manage
behavioral problems as a first step, and the use of language and speech impairments, the functional inter-
second-generation antipsychotics should be reserved ventions focus on communication, including environ-
for the management of psychotic manifestations, mental modifications. The functional interventions key
troublesome or hazardous behaviors. components rely on building communication strategies
and practicing the strategies with a communication
Treatment of primary progressive aphasias partner. However, the frequency and dosage of such
Primary progressive aphasia (PPA) refers to clinical syn- interventions using rigorous outcomes is an unmet need
dromes characterized by an insidious and progressive up to this moment89.
loss of language abilities associated with neurodegen- The maintenance of therapy gains, in the absence of
erative diseases. The three variants differ in anatomic, continuous training, is more evident in the short-term
phenotypic presentation, and underlying pathology84. than long-term in all PPA variants and is influenced by
The non-fluent/agrammatic variant PPA (nfvPPA) treatment length and session frequency. Generalization
is associated with atrophy of left frontal lobe, insula, to untreated items varies following each PPA subtype and
and supplementary motor areas, and progressive, occurs more often in nfvPPA and lvPPA than in svPPA90.
effortful, non-fluent speech, with syntactic deficits, Language therapies usually focus on enhancing
grammatical errors, and omissions. The semantic abilities in trained activities or tasks and such training
variant PPA (svPPA) is associated with left anterior may limit benefits to the practiced domains86,88. Recent
temporal lobe atrophy and fluent speech with deficits reviews have shown a potential benefit for non-invasive
in lexical retrieval, naming, and word comprehension. brain stimulation combined with language therapies in
The logopenic variant PPA (lvPPA) presents AD as the improving oral or written naming accuracy for trained
pathological substrate in most cases and is associated and untrained items85,86.

90   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

As a practical orientation cognitive-linguistic As for treatment of neuropsychiatric symptoms and


interventions targeting impairment and aiming at syndromes, stimuli regulation should be tried before
remediation and symptoms’ improvement seem the any medication is applied. Excessive stimulation, or the
best practice. In this sense, a recent Brazilian study lack of proper stimulation, can worsen orientation and
investigated the short- and mid-term effects of four behavior. Room light, physical exercise, cognitive stim-
training programs directed to language and speech defi- ulation, calm environment and maintenance of clinical
cits in 18 PPA individuals (different subtypes) carried status can help control behavioral problems before
out during four months (two weekly sessions). All cases medication comes into place100. Even if medication is
improved performance on trained items during the ac- needed, all these measures should be enforced.
tive phase of treatment. Statistically significant clinical The clinical diagnosis, differentiating PDD from
benefits were observed in 13 individuals, while for five, DLB, can be a challenge, even for the specialist, since
the results were maintained. However, generalization to the two conditions seem to be different spectra of the
untrained items or to other tasks were observed only same pathological family, being mainly differentiated
in two individuals91. by the clinical course.
The cortical and subcortical distribution of Lewy
bodies will lead to different clinical presentations.
TREATMENT OF PARKINSON’S DISEASE Although it seems somewhat theoretical, this distinc-
DEMENTIA AND DEMENTIA WITH LEWY BODIES tion is essential in some details of the medication man-
PD and DLB are neurodegenerative disorders that share agement inherent to each situation. Diagnostic criteria
some common features such as dopaminergic deple- for PDD and DLB have been published before96,97.
tion and movement disorders92. Lewy body inclusions
throughout the cortex ensue the typical triad of the Lewy body diseases spectrum
dementia syndrome in DLB: dementia, complex visual Due to the inherent characteristics of PDD and DLB,
hallucinations and parkinsonism. PDD, in turn, starts several clinical-pathological correlation studies include
with a dysexecutive syndrome93,94. the two conditions as different spectra of the same patho-
DLB may show parkinsonism later in the course logical process. In the present consensus, we consider the
of the disease, whereas complex visual hallucinations two diseases from this viewpoint, i.e., different involve-
appear early, together with delirium without other ments of a common pathological substrate. In PDD, the
clinical causes. PD first symptoms, on the other hand, accumulation of Lewy bodies is mainly concentrated in
encompass asymmetric bradykinesia, tremor and subcortical areas, while in DLB the pathological process
muscle rigidity95. Memory complaints are not the first affects cortical areas (Figure 2) 101-103.
symptoms in DLB or in PDD, as usually observed in
dementia due to AD and VCI. PDD typically presents Lewy body diseases
with a frontal dysexecutive syndrome95,96.
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 DLB PD PDD
malignant syndrome and because they may aggravate
parkinsonism, regardless of the dose prescribed. When
psychosis exists in PDD, levodopa and other antipar- Cortical Nigral
kinsonian drugs should be re-evaluated, before pre-
scribing any antipsychotics to deal with the behavioral Figure 2. Topographic distribution pattern of Lewy bodies in dementia
and mood symptoms97,98. with Lewy bodies (DLB), Parkinson’s disease (PD) and dementia
Cognitive and neuropsychiatric symptoms in associated with Parkinson’s disease (PDD).
PDD and DLB are treated with rivastigmine, the only
approved ChEI for these indications98,99. Memantine Management of DLB
appears to be associated with some improvement in There is no specific disease-modifying treatment for DLB.
behavioral aspects, but studies have not reported ben- Non-pharmacological and pharmacological interventions
efits for cognition in these individuals98,99. Memantine can be used in symptomatic management. There are no
has not been approved for treatment of DLB or PDD. specific non-pharmacological interventions in DLB100,104.

Caramelli P, et al.   Dementia care.   91


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

Motor symptoms symptoms profile and severity of the dementia syn-


Levodopa is the drug of choice to treat motor symp- drome, as well as the clinical and personal history of
toms. Slow titration is important to achieve some mo- each individual.
tor response with minimal psychiatric AEs. Response It is important to inform the PwD and their families
to levodopa in DLB can be limited, especially in the about the potential expected benefits of the overall
later stages, due to the predominance of levodopa treatment, as well as the possible AEs of the prescribed
non-responsive symptoms. Dopamine agonists are not drugs. Regular follow-up is critical (ideally every three
recommended due to the risk of worsening hallucina- to four months in the beginning of treatment and
tions. Anticholinergics are contraindicated97-99,105,106. around every six months later on) to evaluate the clini-
cal effects and whether adjustments (e.g., drug dosing,
Neuropsychiatric symptoms additional pharmacological and non-pharmacological
The use of antipsychotics must be done very judiciously, interventions) are needed.
due to the high sensitivity of these patients, with im- Given that the modest effects of the available treat-
portant paradoxical reactions107. Typical antipsychotics ments and the lack of approved disease-specific drug
are contraindicated. Atypical antipsychotics, specifically therapies, prevention of dementia is a key principle.
quetiapine and clozapine, can be used, always under Several modifiable risk factors have been identified
close supervision, to detect AEs97,99,108. Of special note, and lifestyle and clinical interventions (e.g., adequate
clozapine may cause neutropenia in about 3% of cases, control of hypertension and diabetes, regular phys-
which demands weekly monitoring of blood cell count ical activity, diet programs, cognitive stimulation
within the first 18 weeks of treatment. activities) shall be recommended for middle-age and
As for ChEI (mainly rivastigmine), besides the cog- older adults to reduce the risk of cognitive impair-
nitive benefits several studies have shown positive ef- ment and dementia, thus contributing to mitigate the
fects on neuropsychiatric symptoms, particularly visual personal, socioeconomic and public health impacts of
hallucinations, delusions, and apathy. It is essential to these devastating disorders.45
be aware that the withdrawal of ChEI, for any reason,
may be associated with cognitive deterioration99,109-111.
ACKNOWLEDGEMENTS
PC, JL, LCS and RN are funded by CNPq, Brazil (bolsa
CONCLUSIONS de produtividade em pesquisa).
Although current treatments for neurodegenerative
and vascular dementias are only symptomatic, several Authors’ contributions. PC, VM, JL, MVDC, FS, EFC, JS,
evidenced-based therapies are available and may provide BJAPB, LPS, HBD, PHFB: concept; PC, VM, JL, MVDC,
benefits to Pwd and to their caregivers. The clinician FS, EFC, HBD, PHFB: drafting of the manuscript;
must combine the best pharmacological options with BJAPB, FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC,
the best available non-pharmacological interventions, PHFB, RN, SMDB: critical revision of the manuscript
taking into consideration the underlying etiology, for important intellectual content.

REFERENCES
1. Mukadam N, Sommerlad A, Huntley J, Livingston G. Population attributab- 6. Bartus RT. On neurodegenerative diseases, models, and treatment stra-
le fractions for risk factors for dementia in low-income and middle-income tegies: lessons learned and lessons forgotten a generation following the
countries: an analysis using cross-sectional survey data. Lancet Glob cholinergic hypothesis. Exp Neurol. 2000;163(2):495-529. doi:10.1006/
Health. 2019;7(5):e596-603. doi:10.1016/S2214-109X(19)30074-9. exnr.2000.7397.
2. Nakamura AE, Opaleye D, Tani G, Ferri CP. Dementia underdiag- 7. Small G, Bullock R. Defining optimal treatment with cholinesterase
nosis in Brazil. Lancet. 2015;385(9966):418-9. doi:10.1016/S0140- inhibitors in Alzheimer’s disease. Alzheimers Dement. 2011;7(2):177-84.
6736(15)60153-2. doi:10.1016/j.jalz.2010.03.016.
8. Xu H, Garcia-Ptacek S, Jönsson L, Wilmo A, Nordström P, Eriks-
3. Nitrini R, Bottino CM, Albala C, Custodio Capuñay NS, Ketzoian C, Llibre
dotter M. Long-term effects of cholinesterase inhibitors on cognitive
Rodriguez JJ, et al. Prevalence of dementia in Latin America: a collaborative
decline and mortality. Neurology. 2021;96(e):2220-30. doi:10.1212/
study of population-based cohorts. Int Psychogeriatr. 2009;21(4):622-30.
WNL.0000000000011832.
doi:10.1017/S1041610209009430. 9. Ministério da Saúde (BR). Secretaria de Atenção à Saúde. Portaria Con-
4. Institute of Health Metrics [homepage on the Internet]. Brazil. 2018 [cited junta nº 13, de 28 de novembro de 2017. Aprova o Protocolo Clínico e
2022 Aug 22]. Available from: http://www.healthdata.org/brazil Diretrizes Terapêuticas da Doença de Alzheimer. Diário Oficial da União.
5. Engelhardt E, Brucki SMD, Cavalcanti JLS, Forlenza OV, Laks J, Vale FAC. 2017 Dec 8;1:201.
Treatment of Alzheimer’s Disease: recommendations and suggestions 10. Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, et al.
of the Scientific Department of Cognitive Neurology and Aging of the Early diagnosis and treatment of Alzheimer’s disease: New definitions and
Brazilian Academy of Neurology. Arq Neuropsiquiatr. 2005;63(4):1104-12. challenges. Braz J Psychiatry. 2020;42(4):431-41. doi:10.1590/1516-
doi:10.1590/S0004-282X2005000600035. 4446-2019-0735.

92   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

11. Vale FAC, Corrêa Neto Y, Bertolucci PHF, Machado JCB, Silva DJ, Allam N, COVID-19 Pandemic and post-pandemic assistance. Front Psychiatry.
et al. Treatment of Alzheimer’s disease in Brazil: II. Behavioral and psycho- 2020;11:577871. doi:10.3389/fpsyt.2020.577871.
logical symptoms of dementia. Dement Neuropsychol. 2011;5(3):189-97. 31. Duan Y, Lu L, Chen J, Wu C, Liang J, Zheng Y, et al. Psychosocial
doi:10.1590/S1980-57642011DN05030006. interventions for Alzheimer’s disease cognitive symptoms: a Bayesian
12. He Y, Li H, Huang J, Huang S, Bai Y, Li Y, et al. Efficacy of antidepres- network meta-analysis. BMC Geriatr. 2018;18(1):175. doi:10.1186/
sant drugs in the treatment of depression in Alzheimer disease patients: s12877-018-0864-6.
A systematic review and network meta-analysis. J Psychopharmacol. 32. Piersol CV, Canton K, Connor SE, Giller I, Lipman S, Sager S. Effectiveness
2021;35(8):901-9. doi:10.1177/02698811211030181. of interventions for caregivers of people with Alzheimer’s Disease and
13. Banerjee S, High J, Stirling S, Shepstone L, Swart AM, Telling T, related major neurocognitive disorders: a systematic review. Am J Occup
et al. Study of mirtazapine for agitated behaviours in dementia Ther. 2017;71(5):7105180020p1-p10. doi:10.5014/ajot.2017.027581.
(SYMBAD): a randomised, double-blind, placebo-controlled trial. Lancet. 33. Shim M, Tilley JL, Im S, Price K, Gonzalez A. A systematic review of mind-
2021;398(10310):1487-97. doi:10.1016/S0140-6736(21)01210-1. fulness-based interventions for patients with mild cognitive impairment or
14. Davies SJ, Burhan AM, Kim D, Gerretsen P, Graff-Guerrero A, Woo VL, dementia and caregivers. J Geriatr Psychiatry Neurol. 2021;34(6):528-54.
et al. Sequential drug treatment algorithm for agitation and aggression in doi:10.1177/0891988720957104.
Alzheimer’s and mixed dementia. J Psychopharmacol. 2018;32(5):509-23. 34. Gaugler JE, Bain LJ, Mitchell L, Finlay J, Fazio S, Jutkowitz E & Alzhei-
doi:10.1177/0269881117744996. mer’s Association Psychosocial Measurement Workgroup. Reconsidering
15. Camargos EF, Louzada LL, Quintas JL, Naves JO, Louzada FM, Nóbrega frameworks of Alzheimer’s dementia when assessing psychosocial out-
OT. Trazodone improves sleep parameters in Alzheimer disease patients: comes. Alzheimers Dement. 2019;5:388-97. https://doi.org/10.1016/
a randomized, double-blind, and placebo-controlled study. Am J Geriatr j.trci.2019.02.008.
Psychiatry. 2014;22(12):1565-74. doi:10.1016/j.jagp.2013.12.174. 35. Dröes RM, Chattat R, Diaz A, Gove D, Graff M, Murphy K. Social health
16. Louzada LL, Machado FV, Quintas JL, Ribeiro GA, Silva MV, Mendonça- and dementia: a European consensus on the operationalization of the
-Silva DL, et al. The efficacy and safety of zolpidem and zopiclone to concept and directions for research and practice. Aging Ment Health.
treat insomnia in Alzheimer’s disease: a randomized, triple-blind, pla- 2017;21(1):4-17. doi:10.1080/13607863.2016.1254596.
cebo-controlled trial. Neuropsychopharmacology. 2022;47(2):570-9. 36. Oksnebjerg L, Diaz-Ponce A, Gove D, Moniz-Cook E, Mountain G, Chattat
doi:10.1038/s41386-021-01191-3. R, et al. Towards capturing meaningful outcomes for people with dementia
17. Caraci F, Santagati M, Caruso G, Cannavò D, Leggio GM, Salomone S, in psychosocial intervention research: a pan-European consultation. Health
et al. New antipsychotic drugs for the treatment of agitation and psychosis Expect. 2018;21(6):1056-65. doi:10.1111/hex.12799.
in Alzheimer’s disease: focus on brexpiprazole and pimavanserin. F1000Res. 37. Arroyo-Anlló EM, Díaz-Marta JP, Chamorro Sánchez J. Técnicas de
2020;9:F1000 Faculty Rev-686. doi:10.12688/f1000research.22662.1. rehabilitación neuropsicológica en demencias: hacia la ciber-rehabilitación
18. Brucki SMD, Adoni T, Almeida CMO, Andrade DC, Anghinah R, Barbosa neuropsicológica. Pensam Psicol. 2012;10(1):107-27.
LM, et al. Cannabinoids in Neurology – Position paper from Scientific 38. Moreira SV, Reis Justi FR, Moreira M. Can musical intervention im-
Departments from Brazilian Academy of Neurology. Arq Neuropsiquiatr. prove memory in Alzheimer’s patients? Evidence from a systematic
2021;79(4):354-69. doi:10.1590/0004-282X-ANP-2020-0432. review. Dement Neuropsychol. 2018;12(2):133-42. doi:10.1590/
19. Frederiksen KS, Cooper C, Frisoni GB, Frölich L, Georges J, Kramber- 1980-57642018dn12-020005.
ger MG, et al. A European Academy of Neurology guideline on medical 39. Jia RX, Liang JH, Xu Y, Wang YQ. Effects of physical activity and exercise
management issues in dementia. Eur J Neurol. 2020;27(10):1805-20. on the cognitive function of patients with Alzheimer disease: a meta-a-
doi:10.1111/ene.14412. nalysis. BMC Geriatr. 2019;19(1):181. doi:10.1186/s12877-019-1175-2.
20. York Health Economics Consortium. NICE: Overview of Systematic Re- 40. Wang LY, Pei J, Zhan YJ, Cai YW. Overview of meta-analyses of five
views of Non-pharmacological Interventions for Dementia [Internet]. Final non-pharmacological interventions for Alzheimer’s Disease. Front Aging
Report. Heslington: University of York; 2017 [cited 2022 Aug 22]. Available Neurosci. 2020;12:594432. doi:10.3389/fnagi.2020.594432.
from: https://www.nice.org.uk/guidance/ng97/evidence/appendix-o-yhe- 41. Meng Q, Lin MS, Tzeng IS. Relationship between exercise and Alzhei-
c-report-pdf-174697045530 mer’s disease: a narrative literature review. Front Neurosci. 2020;14:131.
21. Aguirre E, Spector A, Orrell M. Guidelines for adapting cognitive stimulation doi:10.3389/fnins.2020.00131.
therapy to other cultures. Clin Interv Aging. 2014;9:1003-7. doi:10.2147/ 42. Smallfield S, Heckenlaible C. Effectiveness of occupational therapy
CIA.S61849. interventions to enhance occupational performance for adults with Alzhei-
22. Meyer C, O’Keefe F. Non-pharmacological interventions for people with mer’s Disease and related major neurocognitive disorders: a systematic
dementia: A review of reviews. Dementia (London). 2020;19(6):1927-54. review. Am J Occup Ther. 2017;71(5):7105180010p1-p9. doi:10.5014/
doi:10.1177/1471301218813234. ajot.2017.024752.
23. Stoner CR, Lakshminarayanan M, Durgante H, Spector A. Psychosocial 43. Marques CLS, Borgato MH, Moura Neto E, Bazan R, Luvizutto GJ. Phy-
interventions for dementia in low- and middle-income countries (LMICs): sical therapy in patients with Alzheimer’s disease: a systematic review of
a systematic review of effectiveness and implementation readiness. Aging randomized controlled clinical trials. Fisioter Pesqui. 2019;26(3):311-21.
Ment Health. 2021;25(3):408-19. doi:10.1080/13607863.2019.1695742. doi:10.1590/1809-2950/18037226032019.
24. Bertrand E, Naylor R, Laks J, Marinho V, Spector A, Mograbi DC. Cog- 44. Zhu XC, Yu Y, Wang HF, Jiang T, Cao L, Wang C, et al. Physiotherapy
nitive stimulation therapy for Brazilian people with dementia: examination intervention in Alzheimer’s disease: systematic review and meta-analysis.
of implementation’ issues and cultural adaptation. Aging Ment Health. J Alzheimers Dis. 2015;44(1):163-74. doi:10.3233/JAD-141377.
2019;23(10):1400-4. doi:10.1080/13607863.2018.1488944. 45. Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H,
25. Marinho V, Bertrand E, Naylor R, Bomilcar I, Laks J, Spector A, et al. et al. World-Wide FINGERS Network: A global approach to risk reduction
Cognitive stimulation therapy for people with dementia in Brazil (CST-Bra- and prevention of dementia. Alzheimers Dement. 2020;16(7):1078-94.
sil): Results from a single blind randomized controlled trial. Int J Geriatr doi:10.1002/alz.12123.
Psychiatry. 2021;36(2):286-93. doi:10.1002/gps.5421. 46. Moreira SC, Jansen AK, Silva FM. Dietary interventions and cognition
26. Madureira BG, Pereira MG, Avelino PR, Costa HS, Menezes KKP. Efeitos of Alzheimer’s disease patients: a systematic review of randomized
de programas de reabilitação multidisciplinar no tratamento de pacientes controlled trial. Dement Neuropsychol. 2020;14(3):258-82. doi:10.1590/
com doença de Alzheimer: uma revisão sistemática. Cad Saude Coletiva. 1980-57642020dn14-030008.
2018;26(2):222-32. doi:10.1590/1414-462x201800020446. 47. Pendlebury ST, Rothwell PM. Prevalence, incidence, and factors asso-
27. Cammisuli DM, Danti S, Bosinelli F, Cipriani G. Non-pharmacological inter- ciated with pre-stroke and post-stroke dementia: a systematic review
ventions for people with Alzheimer’s disease: A critical review of the scien- and meta-analysis. Lancet Neurol. 2009;8(11):1006-18. doi:10.1016/
tific literature from the last ten years. Eur Geriatr Med. 2016,7(1):57-64. S1474-4422(09)70236-4.
doi:10.1016/j.eurger.2016.01.002. 48. Boehme AK, Esenwa C, Elkind MSV. Stroke risk factors, geneti-
28. Carrion C, Folkvord F, Anastasiadou D, Aymerich M. Cognitive therapy cs, and prevention. Circ Res. 2017;120(3):472-95. doi:10.1161/
for dementia patients: a systematic review. Dement Geriatr Cogn Disord. CIRCRESAHA.116.308398.
2018;46(1-2):1-26. doi:10.1159/000490851. 49. ADVANCE Collaborative Group; Patel A, MacMahon S, Chalmers J, Neal
29. Chiu H-Y, Chen P-Y, Chen Y-T, Huang H-C. Reality orientation therapy B, Billot L, et al. Intensive blood glucose control and vascular outcomes
benefits cognition in older people with dementia: A meta-analysis. Int J in patients with type 2 diabetes. N Engl J Med. 2008;358(24):2560-72.
Nurs Stud. 2018;86:20-8. doi:10.1016/j.ijnurstu.2018.06.008. doi:10.1056/NEJMoa0802987.
30. Alves GS, Casali ME, Veras AB, Carrilho CG, Bruno Costa E, Rodri- 50. Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, et al.
gues VM, et al. A systematic review of home-setting psychoeducation Effects of intensive glucose lowering on brain structure and function in
interventions for behavioral changes in dementia: some lessons for the people with type 2 diabetes (ACCORD MIND): a randomised open-label

Caramelli P, et al.   Dementia care.   93


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

substudy. Lancet Neurol. 2011;10(11):969-77. doi:10.1016/S1474- dementia. Am J Alzheimers Dis Other Demen. 2008;23(2):167-76.
4422(11)70188-0. doi:10.1177/1533317507312558.
51. Costa e Forti A, Pires AC, Pittito BA, Gerchman F, Oliveira JEP, Zajden- 70. Auchus A, Brashear HR, Salloway S, Korczyn A, De Deyn PP, Gass-
verg L, et al. Diretrizes da Sociedade Brasileira de Diabetes: 2019-2020. mann-Mayer C; GAL-INT-26 Study Group. Galantamine treatment of
São Paulo: Clannad; 2019. vascular dementia: a randomized trial. Neurology. 2007;69(5):448-58.
52. Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz doi:10.1212/01.wnl.0000266625.31615.f6.
MD, et al. Guidelines for the prevention of stroke in patients with stroke 71. Narasimhalu K, Effendy S, Sim C, Lee JM, Chen I, Hia SB, et al. A rando-
and transient ischemic attack. Stroke. 2014;45(7):2160-236. doi:10.1161/ mized controlled trial of rivastigmine in patients with cognitive impairment
STR.0000000000000024. no dementia because of cerebrovascular disease. Acta Neurol Scand.
53. Smith EE, Saposnik G, Biessels GJ, Doubal FN, Fornage M, Gorelick PB, 2010;121(4):217-24. doi:10.1111/j.1600-0404.2009.01263.x.
et al. Prevention of stroke in patients with silent cerebrovascular disease: a 72. Wilcock G, Möbius HJ, Stöffler A; MMM 500 group. A double-blind, place-
scientific statement for healthcare professionals from the American Heart bo-controlled multicentre study of memantine in mild to moderate vascular
Association/American Stroke Association. Stroke. 2017;48(2):e44-71. dementia (MMM500). Int Clin Psychopharmacol. 2002;17(6):297-305.
doi:10.1161/STR.0000000000000116. doi:10.1097/00004850-200211000-00005.
54. Davis KAS, Bishara D, Molokhia M, Mueller C, Perera G, Stewart RJ. 73. Reus VI, Fochtmann LJ, Eyler AE, Hilty DM, Horvitz-Lennon M, Jibson MD,
Aspirin in people with dementia, long-term benefits, and harms: a sys- et al. The American Psychiatric Association practice guideline on the use
tematic review. Eur J Clin Pharmacol. 2021;77(7):943-54. doi:10.1007/ of antipsychotics to treat agitation or psychosis in patients with dementia.
s00228-021-03089-x. Am J Psychiatry. 2016;173(5):543-6. doi:10.1176/appi.ajp.2015.173501.
55. McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Sta- 74. Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral
tins for the prevention of dementia. Cochrane Database Syst Rev. and psychological symptoms in people with Alzheimer’s disease: an
2014;(7):CD007514. doi:10.1002/14651858.CD007514.pub3. international Delphi consensus. Int Psychogeriatr. 2019;31(1):83-90.
56. Sofi F, Valecchi D, Bacci D, Abbate R, Gensini GF, Casini A, et al. Physical doi:10.1017/S1041610218000534.
activity and risk of cognitive decline: a meta-analysis of prospective studies. 75. Oliveira LF, Camargos EF, Martini LLL, Machado FV, Novaes MRCG. Use of
J Int Med. 2011;269(1):107-17. doi:10.1111/j.1365-2796.2010.02281.x. psychotropic agents to treat agitation and aggression in Brazilian patients
57. Verdelho A, Madureira S, Ferro JM, Baezner H, Blahak C, Poggesi A, et al. with Alzheimer’s disease: A naturalistic and multicenter study. Psychiatry
Physical activity prevents progression for cognitive impairment and vascu- Res. 2021;295:113591. doi:10.1016/j.psychres.2020.113591.
lar dementia: results from the LADIS (Leukoaraiosis and Disability) study. 76. Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal de-
Stroke. 2012;43(12):3331-5. doi:10.1161/STROKEAHA.112.661793. mentia. Int Rev of Psychiatry. 2013;25(2):130-7. doi:10.3109/0954026
58. Gillespie DC, Bowen A, Chung CS, Cockburn J, Knapp P, Pollock A. 1.2013.776523.
Rehabilitation for post-stroke cognitive impairment: an overview of re- 77. Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus
commendations arising from systematic reviews of current evidence. Clin J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant
Rehabil. 2015;29(2):120-8. doi:10.1177/0269215514538982. of frontotemporal dementia. Brain. 2011;134(Pt 9):2456-77. doi:10.1093/
brain/awr179.
59. Coelho-Júnior HJ, Trichopoulou A, Panza F. Cross-sectional and longitu-
78. Trieu C, Gossink F, Stek ML, Scheltens P, Pijnenburg YAL, Dols A.
dinal associations between adherence to Mediterranean diet with physical
Effectiveness of pharmacological interventions for symptoms of behavioral
performance and cognitive function in older adults: A systematic review
variant frontotemporal dementia: a systematic review. Cogn Behav Neurol.
and meta-analysis. Ageing Res Rev. 2021;70:101395. doi:10.1016/
2020;33(1):1-15. doi:10.1097/WNN.0000000000000217.
j.arr.2021.101395.
79. Lima-Silva TB, Bahia VS, Carvalho VA, Guimarães HC, Caramelli P, Balthazar
60. Brucki SMD, Ferraz AC, Freitas GR, Massaro AR, Radanovic M, Schultz
ML, et al. Neuropsychiatric symptoms, caregiver burden and distress in
RR. Treatment of vascular dementia. Recommendations of the Scientific
behavioral-variant frontotemporal dementia and Alzheimer’s disease. Dement
Department of Cognitive Neurology and Aging of the Brazilian Academy
Geriatr Cogn Disord. 2015;40(5-6):268-75. doi:10.1159/000437351.
of Neurology. Dement Neuropsychol. 2011;5(4):275-87. doi:10.1590/
80. Wylie MA, Shnall A, Onyike CU, Huey ED. Management of frontotemporal
S1980-57642011DN05040005.
dementia in mental health and multidisciplinary settings. Int Rev Psychiatry.
61. Smith EE, Barber P, Field TS, Ganesh A, Hachinski V, Hogan DB, et al.
2013;25(2):230-6. doi:10.3109/09540261.2013.776949.
Canadian Consensus Conference on Diagnosis and Treatment of Dementia
81. Marin SMC, Mansur LL, Oliveira FF, Marin LF, Wajman JR, Bahia VS, et al.
(CCCDTD) 5: Guidelines for management of vascular cognitive impairment.
Swallowing in behavioral variant frontotemporal dementia. Arq Neuropsi-
Alzheimers Dement (N Y). 2020;6(1):e12056. doi:10.1002/trc2.12056. quiatr. 2021;79(1):8-14. doi:10.1590/0004-282X20200060.
62. Dichgans M, Markus HS, Salloway S, Verkkoniemi A, Moline M, Wang 82. Portugal MG, Marinho V, Laks J. Pharmacological treatment of fron-
Q, et al. Donepezil in patients with subcortical vascular cognitive im- totemporal lobar degeneration: systematic review. Braz J Psychiatry.
pairment: a randomised double-blind trial in CADASIL. Lancet Neurol. 2011:33(1):81-90. doi:10.1590/s1516-44462011000100016.
2008;7(4):310-8. doi:10.1016/S1474-4422(08)70046-2. 83. Gambogi LB, Guimarães HC, Souza LC, Caramelli P. Treatment
63. Black S, Román GC, Geldmacher DS, Salloway S, Hecker J, Burns A, of the behavioral variant of frontotemporal dementia: a narrative
et al. Efficacy and tolerability of donepezil in vascular dementia: positive review. Dement Neuropsychol. 2021;15(3):331-8. doi:10.1590/
results of a 24-week, multicenter, international, randomized, placebo- 1980-57642021dn15-030004.
-controlled clinical trial. Stroke. 2003;34(10):2323-30. doi:10.1161/ 84. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, Cappa
01.STR.0000091396.95360.E1. SF, et al. Classification of primary progressive aphasia and its variants.
64. Orgogozo JM, Rigaud AS, Stöffler A, Möbius HJ, Forette F. Efficacy Neurology. 2011;76(11):1006-14. doi:10.1212/WNL.0b013e31821103e6.
and safety of memantine in patients with mild to moderate vascular 85. Cotelli M, Manenti R, Ferrari C, Gobbi E, Macis A, Cappa SF. Effectiveness
dementia: a randomized, placebo-controlled trial (MMM 300). Stroke. of language training and non-invasive brain stimulation on oral and written
2002;33(7):1834-9. doi:10.1161/01.str.0000020094.08790.49. naming performance in Primary Progressive Aphasia: A meta-analysis
65. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. and systematic review. Neurosci Biobehav Rev. 2020;108:498-525.
Efficacy of galantamine in probable vascular dementia and Alzheimer’s doi:10.1016/j.neubiorev.2019.12.003.
disease combined with cerebrovascular disease: a randomised trial. 86. Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive brain sti-
Lancet. 2002;359(9314):1283-90. doi:10.1016/S0140-6736(02)08267-3. mulation (tDCS or TMS) paired with language therapy in the treatment
66. Caramelli P, Laks J, Palmini AL, Nitrini R, Chaves ML, Forlenza OV, et al. of primary progressive aphasia: an exploratory meta-analysis. Brain Sci.
Effects of galantamine and galantamine combined with nimodipine on cog- 2020;10(9):597. doi:10.3390/brainsci10090597.
nitive speed and quality of life in mixed dementia: a 24-week, randomized, 87. Beber BC, Brandão L, Chaves MLF. A warning to the Brazilian Speech-
placebo-controlled exploratory trial (the REMIX study). Arq Neuropsiquiatr. -Language Pathology and Audiology community about the importance
2014;72(6):411-7. doi:10.1590/0004-282x20140055. of scientific and clinical activities in primary progressive aphasia. CoDAS.
67. Ballard C, Sauter M, Scheltens P, He Y, Barkhof F, Straaten EC, et al. 2015;27(5):505-8. doi:10.1590/2317-1782/20152015081.
Efficacy, safety and tolerability of rivastigmine capsules in patients with 88. Carthery-Goulart MT, Silveira AC, Machado TH, Mansur LL, Parente
probable vascular dementia: the VantagE study. Curr Med Res Opin. MAMP, Senaha MLH, et al. Nonpharmacological interventions for cognitive
2008;24(9):2561-74. doi:10.1185/03007990802328142. impairments following primary progressive aphasia: A systematic review
68. Román GC, Salloway S, Black SE, Royall DR, Decarli C, Weiner MW, et al. of the literature. Dement Neuropsychol. 2013;7(1):122-31. doi:10.1590/
Randomized, placebo-controlled, clinical trial of donepezil in vascular de- S1980-57642013DN70100018.
mentia: differential effects by hippocampal size. Stroke. 2010;41(6):1213- 89. Volkmer A, Spector A, Meitanis V, Warren JD, Beeke S. Effects of func-
21. doi:10.1161/STROKEAHA.109.570077. tional communication interventions for people with primary progressive
69. Moretti R, Torre P, Antonello RM, Cazzato G, Pizzolato G. Different res- aphasia and their caregivers: A systematic review. Aging Ment Health.
ponses to rivastigmine in subcortical vascular dementia and multi-infarct 2020;24(9):1381-93. doi:10.1080/13607863.2019.1617246.

94   Dementia care.   Caramelli P, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):83-95

90. Cadório I, Lousada M, Martins P, Figueiredo D. Generalization and a systematic review. Psychol Med. 2018;48(11):1749-58. doi:10.1017/
maintenance of treatment gains in primary progressive aphasia (PPA): S0033291717003257.
A systematic review. Int J Lang Commun Disord. 2017;52(5):543-60. 101. Dugger BN, Boeve BF, Murray ME, Parisi JE, Fujishiro H, Dickson DW,
doi:10.1111/1460-6984.12310. et al. Rapid eye movement sleep behavior disorder and subtypes in autop-
91. Machado TH, Carthery-Goulart MT, Campanha AC, Caramelli P. Cognitive sy-confirmed dementia with Lewy bodies. Mov Disord. 2012;27(1):72-8.
intervention strategies directed to speech and language deficits in primary doi:10.1002/mds.24003.
progressive aphasia: practice-based evidence from 18 cases. Brain Sci. 102. Ohma E, Ikuta F. Parkinson’s disease: distribution of Lewy bodies and
2021;11(10):1268. doi:10.3390/brainsci11101268. monoamine neuron system. Acta Neuropathol. 1976;34(4):311-9.
92. Aarsland D. Cognitive impairment in Parkinson’s disease and dementia doi:10.1007/BF00696560.
with Lewy bodies. Parkinsonism Relat Disord. 2016;22(Suppl 1):S144-8. 103. Hughes AJ, Daniel SE, Blankson S, Lees AJ. A clinicopathological study
doi:10.1016/j.parkreldis.2015.09.034. of 100 cases of Parkinson’s disease. J Neurol Neurosurg Psychiatry.
93. Burn DJ. Cortical Lewy body disease and Parkinson’s disease 1992;55(3):181-4. doi:10.1136/jnnp.55.3.181.
dementia. Curr Opin Neurol. 2006;19(6):572-9. doi:10.1097/01. 104. Rongve A, Aarsland D. Management of Parkinson’s disease de-
wco.0000247607.34697.a2. mentia: practical considerations. Drugs Aging. 2006;23(10):807-22.
94. Outeiro TF, Koss DJ, Erskine D, Walker L, Kurzawa-Akanbi M, Burn D, doi:10.2165/00002512-200623100-00004.
et al. Dementia with Lewy bodies: an update and outlook. Mol Neurode- 105. Caviness JN, Lue L, Adler CH, Walker DG. Parkinson’s disease dementia
gener. 2019;14(1):5. doi:10.1186/s13024-019-0306-8. and potential therapeutic strategies. CNS Neurosci Ther. 2011;17(1):32-44.
95. Jellinger KA. Dementia with Lewy bodies and Parkinson’s disease-de- doi:10.1111/j.1755-5949.2010.00216.x.
mentia: current concepts and controversies. J Neural Transm (Vienna). 106. Gauthier S. Pharmacotherapy of Parkinson disease dementia and
2018;125(4):615-50. doi:10.1007/s00702-017-1821-9. Lewy body dementia. Front Neurol Neurosci. 2009;24:135-9.
96. Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C, Mizuno Y, et al. doi:10.1159/000197892.
Clinical diagnostic criteria for dementia associated with Parkinson’s 107. Garcia-Ptacek S, Kramberger MG. Parkinson disease
disease. Mov Disord. 2007;22(12):1689-707. doi:10.1002/mds.21507. and dementia. J Geriatr Psychiatry Neurol. 2016;29(5):261-70.
97. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, doi:10.1177/0891988716654985.
et al. Diagnosis and management of dementia with Lewy bodies: Fourth 108. Sezgin M, Bilgic B, Tinaz S, Emre M. Parkinson’s disease demen-
consensus report of the DLB Consortium. Neurology. 2017;89(1):88-100. tia and Lewy body disease. Semin Neurol. 2019;39(2):274-82.
doi:10.1212/WNL.0000000000004058. doi:10.1055/s-0039-1678579.
98. Broadstock M, Ballard C, Corbett A. Latest treatment options for Alzhei- 109. Taylor JP, McKeith IG, Burn DJ, Boeve BF, Weintraub D, Bamford C, et al.
mer’s disease, Parkinson’s disease dementia and dementia with Lewy New evidence on the management of Lewy body dementia. Lancet Neurol.
bodies. Expert Opin Pharmacother. 2014;15(13):1797-810. doi:10.1517 2020;19(2):157-69. doi:10.1016/S1474-4422(19)30153-X.
/14656566.2014.936848. 110. Velayudhan L, Ffytche D, Ballard C, Aarsland D. New therapeutic strate-
99. Stinton C, McKeith I, Taylor JP, Lafortune L, Mioshi E, Mak E, et al. Phar- gies for Lewy body dementias. Curr Neurol Neurosci Rep. 2017;17(9):68.
macological management of Lewy body dementia: a systematic review doi:10.1007/s11910-017-0778-2.
and meta-analysis. Am J Psychiatry. 2015;172(8):731-42. doi:10.1176/ 111. Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase inhibitors
appi.ajp.2015.14121582. for dementia with Lewy bodies, Parkinson’s disease dementia and cog-
100. Connors MH, Quinto L, McKeith I, Brodaty H, Allan L, Bamford C, nitive impairment in Parkinson’s disease. Cochrane Database Syst Rev.
et al. Non-pharmacological interventions for Lewy body dementia: 2012;(3):CD006504. doi:10.1002/14651858.CD006504.pub2.

Caramelli P, et al.   Dementia care.   95


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115 Consensus
https://doi.org/10.1590/1980-5764-DN-2022-S107EN

Management in severe dementia:


recommendations of the Scientific Department
of Cognitive Neurology and Aging of the
Brazilian Academy of Neurology
Sonia Maria Dozzi Brucki1 , Ivan Aprahamian2,3,4 , Wyllians Vendramini Borelli5 ,
Victor Calil da Silveira6,7 , Ceres Eloah de Lucena Ferretti1 , Jerusa Smid1 ,
Breno José Alencar Pires Barbosa1,8,9 , Lucas Porcello Schilling10,11,12 ,
Márcio Luiz Figueiredo Balthazar13 , Norberto Anízio Ferreira Frota14,15 ,
Leonardo Cruz de Souza16 , Francisco Assis Carvalho Vale17 , Paulo Caramelli16 ,
Paulo Henrique Ferreira Bertolucci18 , Márcia Lorena Fagundes Chaves19,20 ,
Ricardo Nitrini1 , Rodrigo Rizek Schultz21,22 , Lilian Schafirovits Morillo23

1
Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil.
2
Faculdade de Medicina de Jundiaí, Departamento de Medicina Interna, Divisão de Geriatria, Grupo de Investigação sobre Multimorbidade e Saúde Mental no
Envelhecimento, Jundiaí SP, Brazil.
3
University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands.
4
Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Serviço de Geriatria, Laboratorio de Investigação Médica em Envelhecimento, São Paulo SP, Brazil.
5
Hospital de Clínicas de Porto Alegre, Departamento de Neurologia, Porto Alegre RS, Brazil.
6
Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro RJ, Brazil.
7
Hospital Glória D’Or, Rio de Janeiro RJ, Brazil.
8
Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brazil.
9
Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brazil.
10
Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil.
11
Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
12
Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brazil.
13
Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil.
14
Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil.
15
Universidade de Fortaleza, Fortaleza CE, Brazil.
16
Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brazil.
17
Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brazil.
18
Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil.
19
Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.
20
Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brazil.
21
Associação Brasileira de Alzheimer, São Paulo SP, Brazil.
22
Universidade Santo Amaro, Departamento de Neurologia, São Paulo SP, Brazil.
23
Universidade de São Paulo, Hospital das Clínicas, Serviço de Geriatria, São Paulo SP, Brazil.
Correspondence: Sonia Brucki; Email: sonia.brucki@gmail.com.
Conflict of interest: JS: Participation as speaker in symposia sponsored by the Roche laboratory; LPS: Participation in advisory boards for Biogen. Participation
as speaker in symposia sponsored by the Aché, Apsen, and Biogen laboratories; MLFB: Participation in advisory boards for Biogen. Development of material for
continuous medical education and participation as speaker in symposia sponsored by the EMS and Torrent laboratories; PC: Participation as principal investigator
in clinical trials for the Novo Nordisk and Roche laboratories. Participation in advisory boards for the Aché, Biogen, EMS, Nutricia, and Roche laboratories. Development
of material for continuous medical education and participation as speaker in symposia sponsored by the Aché, Nutricia, Libbs, Roche, Sandoz, Torrent, and Zodiac
laboratories; PHFB: Participation in advisory boards for the Biogen and Novo Nordisk laboratories. Supervision of training activities for the Biogen, Janssen-Cilag,
and Novo Nordisk laboratories and for Quintiles. Participation as speaker in symposia sponsored by the Apsen, Nutricia, Roche, and Sandoz laboratories; LCS:
Participation in advisory board for Biogen. Participation as speaker in symposia sponsored by Biogen; RN: Participation in advisory board for Biogen; SMDB, IP,
WVB, VC, CELF, BJAPB, NAFF, FACV, MLFC, RRS, LSM: There is no conflict of interest to declare.
Received on August 08, 2021; Received in its final form on October 04, 2021; Accepted on April 27, 2022.

96   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

ABSTRACT. Alzheimer’s disease (AD) and other neurodegenerative dementias have a progressive course, impairing cognition, functional capacity, and behavior.
Most studies have focused on AD. Severe dementia is associated with increased age, higher morbidity-mortality, and rising costs of care. It is fundamental
to recognize that severe dementia is the longest period of progression, with patients living for many years in this stage. It is the most heterogeneous phase
in the process, with different abilities and life expectancies. This practice guideline focuses on severe dementia to improve management and care in this stage
of dementia. As it is a long period in the continuum of dementia, clinical practice should consider non-pharmacological and pharmacological approaches.
Multidisciplinary interventions (physical therapy, speech therapy, nutrition, nursing, and others) are essential, besides educational and support to caregivers.
Keywords: Dementia; Palliative Care; Behavior; Cognition.

MANEJO DAS DEMÊNCIAS EM FASE AVANÇADA: RECOMENDAÇÕES DO DEPARTAMENTO CIENTÍFICO DE NEUROLOGIA COGNITIVA E DO ENVELHECIMENTO
DA ACADEMIA BRASILEIRA DE NEUROLOGIA
RESUMO. A doença de Alzheimer (DA) e outras demências neurodegenerativas têm um curso progressivo com comprometimento da cognição, capacidade
funcional e comportamento. A maioria dos estudos enfocou a DA. A demência grave está associada ao aumento da idade, maior morbimortalidade e aumento
dos custos de cuidados. É fundamental reconhecer que a demência grave é o período mais longo de progressão, com o paciente vivendo muitos anos
nesta fase. É a fase mais heterogênea do processo, com diferentes habilidades e expectativa de vida. Esta diretriz de prática concentra-se na demência
grave para melhorar o manejo e o cuidado nessa fase da demência. Como um longo período no continuum da demência, as abordagens não farmacológicas
e farmacológicas devem ser consideradas. Intervenções multidisciplinares (fisioterapia, fonoaudiologia, nutrição, enfermagem, entre outras) são essenciais,
além de educacionais e de apoio aos cuidadores.
Palavras-chave: Demência; Cuidados Paliativos; Comportamento; Cognição.

INTRODUCTION This practice guideline focuses on severe dementia

A lzheimer’s disease (AD) and other neurodegenerative


dementias have a progressive course, impairing
cognition, functional capacity, and behavior. Most studies
to improve management and care in this stage
of the disease. As a long period in the continuum
of dementia, clinical practice should consider non-
have focused on AD. Severe dementia is associated pharmacological and pharmacological approaches.
with increased age, higher morbidity-mortality, Multidisciplinary interventions (physical therapy,
and rising costs of care. Severe stages could account for speech therapy, nutrition, nursing, and others)
70 to 80% of total treatment expenses1,2. Degenerative are essential, besides educational and support
dementias slowly and progressively worsen. In its severe to caregivers. Primary, secondary, and tertiary care
stage, patients show a higher dependency for all basic center professionals could use this practice guideline .
activities of daily living and incapacity for instrumental
activities of daily living.
Around 30 to 60% of patients with dementia (PWD) STAGING SEVERE DEMENTIA
and 90% of individuals residing in long-term care We find many different situations and stages of severe
facilities are in its moderate-late stage3-5. dementia. A bedridden patient with dysphagia is different
The progression of dementia is associated with from a walking and dependent but communicative and
a progressive dependency on caregivers, with loss without dysphagia patient.
of capacity to provide self-care in basic activities of daily The Clinical Dementia Rating (CDR) permits
living. Usually, severe AD patients will score below ten rating the severity of AD and other dementias on
on mini mental examination (MMSE) and moderately a five-point scale from 0 (normal) to 3 (severe stage)10.
severely with 10 to 155. Severe dementia could cause The final score is obtained after interviews with PWD
complications such as immobility, swallowing disorders, and informants. It evaluates memory, orientation,
malnutrition, and fragility. This situation can increase judgment, problem solving, community affairs, home,
the risk of pneumonia, which has been found as hobbies, self-care. Chaves et al.11 validated this study
a common cause of death in PWD5-9. for Brazilian Portuguese.
It is fundamental to recognize that severe dementia The Alzheimer Disease Cooperative Study of activities
is the longest period of progression, with patients living of daily living (ADCS-ADL sev) is a good measure of
for many years in this stage. It is the most heterogeneous activities of daily living (ADL) which strongly correlates
phase in the process, with different abilities and life with cognition and severity of dementia in moderate
expectancies7. Around 17% of PWD older than 75 years to severe AD. Its scores vary from 0 to 54 points,
are living with a very severe stage of dementia8. divided into 19 items12.

Brucki SMD, et al.   Management in severe dementia.   97


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

A simple scale that assesses gradual severity and (stage 7); the latter is subdivided into seven situations
progression of the disability during follow-up is (A to G) (Box 1)14.
the Global Deterioration Scale (GDS) 13. This scale In advanced dementia, we observe impairment
has seven global stages, from normality to severe to basic daily activities. The Katz Scale is often
impairment; stage 6 represents severe dementia, used to measure performance in bathing, dressing,
and stage 7, very severe13. toileting, transferring, continence, and feeding;
The Functional Assessment Staging (FAST) characterizing severe dementia. Table 1 summarizes
assesses decline from normal (stage 1) to severe stages all these instruments.

Box 1. Functional Assessment Staging (FAST).


1 No difficulty either subjectively or objectively.

2 Complains of forgetting location of objects. Subjective work difficulties.

3 Decreased job functioning evident to co-workers. Difficulty in traveling to new locations. Decreased organizational capacity.

Decreased ability to perform complex tasks, e.g., planning dinner for guests, handling personal finances (such as forgetting to pay bills),
4
difficulty shopping, etc.

5 Requires assistance in choosing proper clothing.

6a Improperly puts on clothes without assistance.

6b Unable to properly bathe (shower) (e.g., difficulty adjusting bathwater (shower) temperature.

6c Inability to handle toileting mechanics.

6d Urinary incontinence.

6e Fecal incontinence.

Ability to speak limited to approximately a half a dozen intelligible different words or fewer in the course of an average day or in the course
7a
of an intensive interview.

7b Speech ability limited to the use of a single intelligible word in an average day or in the course of an interview.

7c Loss of ambulatory ability (unable to walk without personal assistance).

7d Unable to sit up without assistance.

7e Loss of ability to smile.

7f Loss of ability to independently hold up their head.

Table 1. Instruments to evaluate severe dementia.


Test Measure Score Answered by Comments

SMMSE Global cognition 0-30 points Patient Good for measuring cognition

0 none
Global State
0.5 questionable
Domains: memory, orientation, judgment,
CDR 1 mild Caregiver and patient Good for follow-up
problem solving, community affairs,
2 moderate
home and hobbies, and personal care
3 severe

0-100 points
SIB Global cognition Patient Allows to evaluate very severe patients
(<63 severe impairment)

ADL Allows to evaluate severe to very


ADCS-ADL 0-54 points Caregiver
19 ADL of moderate to severe dementia severe patients

Stages 6-7: divided into incapacity


FAST Functionality 1-7 Health professional
progression

98   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

COGNITIVE EVALUATION and acute medical conditions), caregivers (e.g., stress,


Cognitive evaluation in advanced dementia is relevant lack of politeness, and communication skills), and the
to measure pharmacologic and non-pharmacologic environment (e.g., under/overstimulation and lack of
management and interventions. MMSE is the most routine and activities). Consequently, it is rational and
frequently used instrument in brief cognitive evaluation evidence-based that the first treatment step against
in dementia. However, for moderate-late stages, BPSD includes non-pharmacological measures 26 .
we could observe a floor-effect, with few modifications A systematic review and meta-analysis points to an
over time below 10 points. At this point, we must effect size at least equivalent to those of psychotropic
consider other tests. The GDS described above could drugs, and it is safer26. However, in clinical practice,
be an instrument for cognitive evaluation but severe- non-pharmacological measures are yet to be fully
MMSE (SMMSE) or Severe Impairment Battery (SIB)15,16 implemented due to several economic and cultural
could offer more detailed and objective assessments. reasons. The next session will better explore this
The SIB, consisting of 40 questions, evaluates specific topic. Clinical practice recommends the
nine areas of cognition: social interaction, memory, observation of BPSD symptoms at different hours,
orientation, language, attention, praxis, visuospatial with varying caregivers, and multiple environments.
ability, construction, and orientation to name. Scores Moreover, research has also suggested using BPSD
vary from 0 to 100.15 measurement instruments, such as the Neuropsychiatric
SMMSE is a brief test requiring minimal training Inventory (including the clinician version or NPI-C)
and no special materials, but educational attainment and the BEHAVE-AD, due to multiple concurrent and
influences it. Its scores vary from 0 to 30 and its sometimes complex symptoms.
items are divided into autobiographical knowledge, After non-pharmacological measures or concomitantly
executive function, language, verbal f luenc y, to them, pharmacological treatment is possible and
and spelling16, 17 (Table 1). common due to safety reasons or BPSD severity.
If only one physician monitors cognitive status, they In general, response rates to different classes of
may ask the same questions to observe the progression medications are heterogenous and of small therapeutic
of deterioration, such as age, date of birth, names of effect. The overall evidence points to a small number
children or spouse, food or soccer team preferences. of psychotropic drugs which reasonably improve
BPSD. We recommend that these drugs be prescribed
at low doses and that clinicians avoid, if possible,
TREATMENT OF BEHAVIOR AND THE polypharmacotherapy. Table 2 summarizes our expert
PSYCHOLOGICAL SYMPTOMS OF DEMENTIA consensus-based recommendation for the use of
Behavior and psychological symptoms of dementia psychotropic medication in BPSD associated with AD.
(BPSD) comprehends the neuropsychiatric Treating cognitive impairment with cholinesterase
manifestations besides cognition, affecting between inhibitors and memantine shows slight improvement in
60 and 90% of people with dementia18. BPSD includes early and moderate stages of AD, according to a recent
a wide spectrum of conditions: apathy, depression, systematic review and meta-analysis27. Effects are small
anxiety, sleep disorders, psychosis, agitation, aggression, and benefits to which family members and caregivers
wandering and motor manifestations, disinhibition, referred may amount to a placebo effect. The benefits
and many others. Generally, BPSD prevalence increases of these drugs for severe dementia are questionable but
with disease severity although epidemiologic studies in may exist and thus, clinicians must weigh the decision
this topic mostly include small sample sizes, especially of maintaining or ceasing such treatment27. In moderate
among those with AD18,19. Moreover, severe forms to severe AD, low to insufficient evidence suggested
of BPSD like delusions, hallucinations, agitation, that cholinesterase inhibitors and add-on memantine
aggression, and aberrant motor conditions are more inconsistently improved cognition and global clinical
common in moderate and severe dementia20. Multiple impression, compared to placebo. However, the evidence
adverse outcomes have been associated with BPSD, is questionable and deserves further exploration in
such as cognitive and functional impairment, caregiver clinical trials. Cholinesterase inhibitors and memantine
burnout, nursing home placement, and mortality21-25. also slightly improve BPSD, especially in the early and
However, research has scarcely explored outcomes moderate stages of AD and include apathy, anxiety,
in severe forms of dementia. and depressive symptoms. In total, three systematic
The occurrence and maintenance of BPSD relies reviews with meta-analysis28-30 have summarized this
on three factors, namely patients (e.g., hunger, pain, evidence. In general, attending physicians may consider

Brucki SMD, et al.   Management in severe dementia.   99


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

prescribing and maximizing both cholinesterase (small sample size from three studies) favoring the
inhibitors and memantine to improve cognitive function use of methylphenidate to reduce this symptom in the
and BPSD before considering other psychotropic Apathy Evaluation Scale36. However, apathy is part of
medication. Nonetheless, we emphasize that evidence the phenotype of more severe dementia, and the overall
is insufficient to decide in favor or against this position. cognitive decline summed with other bothersome BPSD
Citalopram and risperidone have produced favorable in this stage results in a questionable pharmacological
evidence for agitation in AD. Citalopram improved treatment of apathy.
moderate agitation, if administered between 10-30mg A systematic review and meta-analysis 37 was
a day31. Other antidepressant agents, such as sertraline unable to recommend the use of antidepressants to
and trazodone, improved agitation32. However, we must treat depression in BPSD based on its lack of efficacy
stress their potential limitations as one review included in respond to or reduce depressive symptomatology.
fewer patients with severe AD, still showing cognitive Benzodiazepines, anticonvulsants, and cannabidiols
performance and a prolonged QT interval in patients are also ineffective in pharmacologically controlling
receiving citalopram 30mg31. Ongoing S-CitAD, which BPSD due to their lack of efficiency or adverse reactions,
evaluates escitalopram for agitation, may show greater including cognitive decline. Finally, dextromethorphan/
safety and a better cognitive profile. Risperidone, quinidine and prazosin showed evidence of improving
mostly at low doses (0.5-1mg), and selective serotonin agitation in AD 38,39 and pimavanserin reduced
reuptake inhibitors (SSRI), as a class, alleviated psychosis in AD 40 . However, these three agents
agitation in patients with dementia, according to a are unavailable in Brazil.
systematic review and meta-analysis32-34. B ox 2 s ummar iz es our overall treatment
The evidence for the use of antipsychotics for recommendation for cognitive dysfunction and BPSD.
BPSD is limited even for agitation and aggression. In conclusion, taken together, current evidence shows
Data from a systematic review showed that aripiprazole low certainty for prescribing cholinesterase inhibitors
was the safest and most effective antipsychotic and memantine in severe dementia. The use of agents
versus placebo, and it was associated with improved such as SSRIs, risperidone, and aripiprazole for
outcomes on the NPI, the Brief Psychiatric Rating agitation, aggression, and psychosis also showed a small
Scale (BPRS), and the Cohen-Mansfield Agitation or uncertain effect when we consider severe dementia.
Inventory (CMAI)35. Quetiapine improved outcomes on Non-pharmacological measures, including activities
the BPRS, and risperidone was associated with improved of daily living and care routines, proper feeding, pain
outcomes on the CMAI. Differences between atypical control, music therapy, physical therapies, and caregiver
antipsychotics were insignificant for effectiveness, education and support, seem to be safer and more
death, or cardiovascular adverse events35. effective41. The use of pharmacological agents should
Apathy is a common BPSD, especially in early- consist of single agents, in small doses, and for a short
stage dementia. The literature reports pooled evidence period to control the targeted BPSD (Table 2).

Box 2. Overall recommendation for the non-pharmacological and pharmacological treatment of cognitive dysfunction and behavior and psychological
symptoms of dementia (BPSD) in severe Alzheimer’s dementia.
1. Consider withdrawing cholinesterase inhibitors and memantine in severe dementia in the absence of clear benefits to cognition or BPSD

2. Education and support for caregivers

3. Well established routine of daily care and activities

4. If possible, consider music therapy and any form of physical activity

5. Investigate causes for cognitive fluctuations or BPSD

6. Consider pain control before prescribing psychotropic agents

7. If agitated or aggressive, consider citalopram, sertraline or trazodone

8. If agitation or aggression persists with antidepressants, consider antipsychotics such as risperidone, aripiprazole or quetiapine

9. Always reevaluate withdrawing psychotropics for BPSD after symptom control

100   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

Table 2. Consensus-based recommendation for the use of psychotropic medication in specific behavior and psychological symptoms of dementia (BPSD)
associated with severe Alzheimer’s dementia.
BPSD Suggested dose range* Side effects

Citalopram** 10-20mg/day, single dose

Sertraline 50-100mg/day, single dose nausea, diarrhea, headache, increased risk of falls
Agitation or
aggression Trazodone*** 25-100mg/day, single/partial doses

extrapyramidal side effects, weight gain, metabolic abnormalities,


Risperidone 0.25-1mg/day, single dose
hyperprolactinemia

extrapyramidal side effects, weight gain, metabolic abnormalities,


Risperidone 1-3mg/day, single dose
hyperprolactinemia
Psychosis
Quetiapine 100-200mg/day, single/ partial doses drowsiness, weight gain, metabolic abnormalities

Aripiprazole 10-30mg/day, single dose nausea, weight gain, headache, somnolence, akathisia
*Doses are recommended based on clinical trials and personal experience, considering the pharmacological properties of these agents; **May consider escitalopram due to its greater
safety and better cognitive profile. It is also more commonly prescribed in Brazil than citalopram; ***Although it has less evidence than the other agents, this drug shows a good balance
between safety and effectiveness in clinical practice. Extended-release formulation is better during the day to avoid somnolence.

NON-PHARMACOLOGICAL TREATMENT interdisciplinary assessment and conduct to make the


best decision, case by case47.
Nonverbal communication Family doctors and multidisciplinar y teams
“Perhaps it’s the most important part of caring. Because it (previously trained by specialists) in primary health care
leads to reflection, because it provides the manifestation units can theoretically recognize the needs and monitor
of human nature, present in each of the professionals patients with severe dementia who were discharged
involved with care. It is the deepest empathy and the need from the secondary care service. However, professionals
to show affection, affection and, above all, respect for working in this sector still face difficulties, related to
the other. Holding the hands of someone who may have their knowledge to perform diagnoses and administer
their mind lost, in another dimension perhaps, gently pharmacological and non-pharmacological treatment,
touching the thin skin of an already weakened body, looking especially in mild and severe AD phases48.
deeply into their eyes and letting the feelings present in Due to the problems in the health, economic,
this exchange of glances lead to the understanding of what and social sectors in Brazil and the need for
is needed to do. It is the meeting of the human and bioethics, biopsychosocial support (especially during this serious
health crisis49), reference and assistance centers have
a feeling of lightness and of certainty that the task is being
created projects which aim to minimize the direct
carried out with respect and with the dignity that every
and indirect social costs of PWD caregivers 50,51 .
human being deserves.” (Ceres Ferretti)
These projects are in line with most guidelines from
The literature is very rich in papers discussing
international consensus meetings of associations52-57,
non-pharmacological approaches to psychological and
universities58,59, and scientific committees57,60.
behavioral symptoms in dementia – BPSD, which arise A l l e x p e r t co n s e n s u s e s d i s c u s s t h e n e e d
with the evolution of different dementia syndromes42,43. for psychoeducational programs and sug gest
However, unfortunately, the literature still lacks studies recommendations which contribute to the education
discussing the possibilities and effectiveness of these of caregivers about systematized and appropriate
approaches in severe dementia, or even the difficulty models for different behavioral disorders. The great gap,
of conceptualizing and adequately treating people however, is still the approach to the severe phase
with it44-46. Some behaviors are more prevalent in the of AD. Knowing the causes of SCPD in this phase
initial phase of the disease, others in its intermediate is the objective, and solving them, or better yet,
phase and some in its severe phase. Its last phase preventing them is the goal 61. The starting point
signals a possible combination of factors intrinsic is understanding possible social and more individual
to patients (comorbidities) and caregivers (stress) and family barriers to the construction and practice of these
extrinsic (environmental) ones, which require multi and models, those aimed at reducing these behaviors and,

Brucki SMD, et al.   Management in severe dementia.   101


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

consequently, the comfort and quality of life of patients Disease Research Center and the Center for Innovative
with severe dementia and their caregivers62. Care in Aging with guidelines to identify needs
for the practice of care58.
Assessment Models As mentioned above, clinical practice still lacks
In Brazil, the nursing model in dementia care includes a consensus on the definition of severe dementia,
all stages of disease evolution63. Research has shown sometimes confused with moderate dementia.
that the protocol is useful in its main objective, However, studies agree on possible causes that give
which is to assess patients and their caregivers. rise to common behaviors, such as agitation, apathy,
Composed of two stages, patients and caregivers focus hallucinations, delusions, “unmotivated” crying,
on patient-centered care models, in combination with fear, anxiety, contracted body, and resistance to care,
the caregiver-centered care model64. among others58,62.
Kales (2015)58 offers a review with an interesting Several studies report that most factors responsible
conceptual model of factors which require evaluation, for SPCD in severe dementia originate from patients’
leading to the reflection on the union of disciplines comorbidities, caregivers’ lack of knowledge and
combining different areas of knowledge such stress, and the environment. The inability to verbally
as neurology, geriatrics, psychiatry, and gerontology communicate can lead to the behaviors in the same way
in a multidisciplinary view (Figure 1). The same that the rapid recognition of any change in the pattern
review shows “DICE - Describe, Investigate, Create, of patients’ usual behavior can prevent progressing
and Evaluate,” proposed by the University of Michigan of the SPCD and interfering with the quality of life of
in partnership with the Johns Hopkins Alzheimer’s both patients and caregivers (Table 3)58,65.

Neurodegeneration associated with dementia Greater vulnerability to stressors Psychological and Behavioral
Symptoms of Dementia (BPSD)

– Change in the ability to interact with others Caregiver-related factors Factors related to the environment
and environment;
– Disruption of brain neurocircuits – Stress, overload, depression – Overstimulation or understimulation
– Lack of education about dementia; – Lack of activity and structure
– Communication; – Lack os establishing routines
– Incompatibility of expectations
Factors related to patient
and severity of dementia
– Premorbid personality/psychiatric illnesses
– Acute medical problems
(ICU, pneumonia, dehydration, constipation)
– Unmet needs
(pain, sleep problems, fear...)

Figure 1. Depiction of a conceptual model for different interactions resulting in BPSD. Adapted from Kales (2015)58.

Table 3. Recognition and Management Recommendations for Psychological and Behavioral Symptoms in Severe Dementia. Adapted of Kales (2015)58.
In the patient In the caregiver In the environment Recommendations

Caregivers’ training;
Health education;
Lack of knowledge
Poor positioning Identification and elimination of causal factors in patients and the environment;
Health education
Pain Excessive noise Promotion of physical comfort;
Lack of education
Abrupt movements Information to the responsible physician and members of the health team;
in dementia
Elimination of excessive noise;
Gentle patient mobilization.

Continue...

102   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

Table 3. Continuation.

In the patient In the caregiver In the environment Recommendations

Caregivers’ training;
Health education;
Urinary Lack of knowledge
Inadequate hygiene Optimization of fractional hydration (1.5-2L/day) or at medical discretion;
infection Stress
Double incontinence;
Hygiene, body, and environmental measures.

Dark, closed or dirty Caregivers’ training;


Respiratory
Lack of knowledge environment Preventive measures;
Infection
Cold temperature Passive exercises.

Adequate bathroom Training in hygiene measures and health education to caregivers;


Constipation Lack of knowledge hygiene, security, Medical and nutritional approach;
and privacy Fractioned hydration.

Inadequate
Refusal Environment Training of caregivers and health education;
communication
of care adaptation and care Caregivers must be careful with all stimulus and approaches.
and conducts

Lack of recognition
concerning pain,
Observation Training and health education;
Sleep hunger, coldness, heat,
Change in patients’ Adequacy of auditory environmental stimuli;
disturbances mobilization in bed,
behavioral patterns Medical evaluation.
and previous sleep
problems

* All caregivers must be trained in all activities proposed by the health team to offer their consent and understand the importance of each proposed intervention. Elaborated by Ferretti (2021).

Comorbidities in advanced dementia adults without this disease. These comorbidities impact
In general, older adults, and especially those with survival throughout patients’ later years74,75 (Table 4).
dementia, face exclusion from clinical trials on
pharmacological interventions and the medical
literature is scarce in good-quality data regarding Table 4. Conditions associated with higher mortality among subjects.
the advanced stages of dementia since the focus
Number of diagnoses
of guidelines and randomized, controlled studies
and intervention are aimed at premorbid, very mild, Male
and mild stages of dementias66,67. Age
Although research has suggested the possibility of
AD patients being healthier68, current evidence points Lung infections
to the opposite: people with dementia (PWD) show Parkinsonism
a significantly higher prevalence and medication use69-73.
Previous Stroke
Clinical diseases impact quality of life (by, for example,
worsening mobility) and patient survival regardless of Atrial fibrillation
dementia, both in institutionalized individuals and those Malignancy
who remain in the community, increasing the number
of searches for emergency rooms, hospitalizations,
hospital costs, and health expenditures69,70. As for the severity of dementia, results pointed
Older adults with dementia are three times more to a significantly lower two-year survival rate among
likely to have four or more concurrent chronic diseases most dependent patients, regardless of their number of
and annual expenses, up to 3.3 times higher than in older comorbidities. We find a positive correlation between

Brucki SMD, et al.   Management in severe dementia.   103


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

number of comorbidities and dementia severity the progressive impairment of language, memory,
independently but not the impact of the interaction and criticism the efficiency in reporting symptoms and
between them; the more advanced phases compete complaints is lower, less valued or commented upon
with higher health expenditures73. and dependent on a third party not always attentive
Schubert et al. highlight the possibility that the or accurate (the caregiver). Adherence to medication
lesser search for diagnoses and treating comorbidities and non-pharmacological guidelines is also erratic
in PWD on an outpatient basis may be one of the and variable, by patients and caregivers69,81. Moreover,
conditions responsible for its aggravation, showing once undiagnosed and addressed with an adequate
the need to search for advanced care units, which could care plan at an outpatient level, such illnesses become
be avoided under a less nihilistic outlook72,76,77(Box 3). more complicated and patients end up being taken
to emergency rooms, wards, and Intensive Care Units,
often with treatments that are disproportionate to their
Box 3. Chronic conditions most frequently associated in PWD. condition. advanced disease69,81.
Thus, often, despite having a high number of
• Heart failure
comorbidities, patients with dementia are less
• Hypertension likely to have their health problems diagnosed and
• Diabetes mellitus treated, including those that may directly impact
their functionality, such as auditory and visual
• Neoplasms sensory alterations71. Often, by the simple mention
• Kidney failure of the diagnosis of dementia, especially if advanced
(albeit moderately), such individuals are deprived of
• Cerebral and coronary artery diseases
beneficial proportional treatments due to discrimination
• Atrial fibrillation since the diagnosis suggests that these patients would
• Lung diseases fail to obtain the benefit of a certain procedure,
even though they might live long enough to experience
the complications of the disease, thus sometimes
Dementia in comorbidities imposing suffering which could have been avoided72,82.
As previously mentioned, dementia directly impacts The reduction in survival expectancy should limit
the survival of individuals regardless of the presence the administration of treatments unable to improve
of comorbidities 75 . Dementia decreases survival patients’ symptoms or quality of life. Thus, clinical
after acute myocardial infarction, with greater renal practice should rethink primary preventive treatments
decompensation, higher rate of acute lung edema, in light of the prognosis to avoid the unnecessary
showing the delay in hospitalization and the lesser use of medications and polypharmacy and control
prescription of antiplatelet agents and beta-blockers diseases such as hypertension and diabetes mellitus,
at hospital discharge77. Dementia also predicted higher and dyslipidemia. Moreover, other secondary prevention
mortality in patients with heart failure78. measures must comply with current guidelines
The frequent coexistence between Advanced indicating less strict goals for older adults with advanced
Dementia and Frailty syndrome could be one of the ages and those with lower survival expectations83.
ways to explain, at least partially, the worse prognosis
of comorbidities among PWD79. As it is known, although Comorbidities of dementia
the definition of frailty is still a matter of debate The more advanced stages of dementias, especially
in the current medical literature, frail individuals, in its terminal stages, induce the increase of morbid
especially the elderly, have less functional reserve conditions such as lung infections due to bronchial
of physiological organs and systems and less capacity aspiration or other causes, immobility, dysphagia,
to face challenges and organic overload, having urinary tract and skin infections, pressure injuries, falls,
greater difficulty in maintaining homeostasis when malnutrition, and various dental problems84.
faced with acute illnesses, interventions of any kind, Maintaining patients’ dignity, regardless of their
diagnostic or therapeutic80. cognitive status, treating and preventing reversible
The management of comorbidities in dementia, complications (considering individual and family values),
on the other hand, faces several difficulties regardless and establishing, as early as possible, a plan to consistently
of what has been exposed so far: starting with care for these patients with the best technical-scientific
making a diagnosis in these individuals, who, due to knowledge, are the pillars to treat individuals with

104   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

any diagnosis, including dementia, whatever their etiology, disturbances, agitation, depression, weight loss,
patients’ socioeconomic condition or stage81,82,84,85. and decreased mobility90. Studies have shown that
analgesics better control agitation in PWD and pain
Cancer and dementia than neuroleptics, especially in moderate-advanced
Both cancer and dementia have increased prevalence dementia. They also reduce aggressiveness and pain
with age and lead to very complex health care needs without worsening patients’ cognition 91,92.
and worse outcomes in people suffering from these To optimize pain assessment in PWD and, therefore,
diseases than those without these comorbidities86. its identification, a study developed a specific pain
Estimates of the prevalence of the association between scale for severely demented patients, later validated
the two diseases vary in the literature. An estimate for Brazilian Portuguese90. The instrument (PAINAD)
suggests that 7.5% of individuals over 75 years old live evaluates five items: breathing; negative vocalizations;
with both diagnoses86,87. People living with this dual facial expressions; body language; and comfortability,
diagnosis are less likely to receive screening, staging, observing the patient for five minutes in different
curative treatment, and adequate pain management daily situations: during rest, in pleasurable activities
than cancer patients without dementia86,87. Moreover, and moments of care; and 30 minutes after analgesic
they have late diagnoses, a lower survival rate after it, medication. Multidisciplinary teams can use it after
and a greater number of comorbidities than those living training without impeding its complexity.
only with cancer or dementia87. Thus, evaluating and treating pain in patients
Oncology services poorly identify dementia and with advanced dementia is a challenge that clinical
its patients often receive limited therapeutic options. practice must face and, whenever suspected due
Oncology teams feel insecure about managing these to its manifestations, approach and treat it.
patients since dementia carries more complex decision-
making. Finally, studies highlight the important role Oral health
families play in promoting greater success in treating People with dementia have the same oral problems
and managing cancer in patients with dementia86. as the general population. Good oral health positively
Cancer diagnosis and treatment for patients in very inf luences individuals’ overall health, dignity,
advanced stages of dementia refrain from administering self-esteem, social integration, and nutrition. Studies
screening and very invasive measures. Even so, clinical show the effects of oral problems in patients with
practice must establish a care plan to ensure quality dementia with difficulty chewing due to missing dental
at the end of life for people who have aged with these elements and consequent refusal to eat, behavioral
two diseases: pain management, prevention of avoidable changes (such as withdrawal and aggressiveness
complications, and family support. due to pain – as previously mentioned), and other
changes. The nature of dementia and its severity, social
Approach to pain in advanced dementia functioning, behavioral aspects, adherence to oral cavity
Older adults with dementia shown a 32-53% estimated care, and caregivers’ ability of caregivers to replace
prevalence of pain, higher among those with diseases them in this care may compromise the conditions
known to cause pain, such as osteoarthritis, fractures, for maintaining oral health93 (Table 5).
peripheral arterial disease, and cancer. This prevalence Overall health and comfort are closely linked to oral
can reach 83%¨in those living in ILPIs. No study has health in the terminal stages of neurodegenerative
shown that dementia affects pain sensitivity; rather, diseases 94 (Table 6). Oral diseases worsen general
what it alters is individuals’ ability to report what comfort, cause pain, affect cognition and behavior,
they are feeling88. and alter quality and life expectancy of people with
Research considers self-reports as the gold standard dementia. The risk of aspiration pneumonia increases
in pain diagnosis. Patients with dementia show an in the presence of oral factors such as poor hygiene,
unacceptable underdiagnosis and undertreated pain88-90. meager coronal and cer vical teeth, periodontal
Among institutionalized older adults, 25% of those who disease, and the presence of microbes in saliva,
daily complained of pain had received no analgesics. whereas a clean and healthy oral cavity significantly
Among those with hip fractures and dementia, a student reduces its occurrence95,96.
found that the prescribed opioid was a third of the normal Caregivers play a central role in the oral health
dose and, therefore, insufficient 89,91. of patients with advanced dementia. The quality
Untreated pain manifests itself via secondary of hygiene care and the perception of problems
symptoms in patients with dementia, such as sleep which may arise depend almost exclusively on them.

Brucki SMD, et al.   Management in severe dementia.   105


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

Studies have shown a poor understanding of the which are associated with more rapidly progressing
importance of oral health and its subsequent problems, cognitive impairment and increased mortality 98.
such as patients’ (often aggressive) resistance further Therefore, all PWD should receive routine assessments
complicate the adherence of caregivers to these demands. of their nutritional problems. Several instruments
Education, motivation, and the offer of strategies can identify whether patients are undernourished, such
was associated with improved oral hygiene in patients as the Mini Nutritional Assessment (MNA) and the
with severe dementia97. Regular odonatological visits, Malnutrition Universal Screening Tool (MUST)99,100.
caregivers’ education and access to care facilities An unstructured evaluation is, however, also a useful
in day centers, outpatient clinics, and home services are option, particularly under limited time, common in
strategies which can ensure better oral health for these Brazilian primary care consultations. These evaluations
patients, benefitting their global health and quality should include anthropometric measurements (weight,
of life increasing their expectations of survival. height, and body mass index) and questions about food
and fluid intake, dietary habits, and adversative feeding
behaviors. Laboratory tests are also helpful to better
Table 5. The main changes seen in the oral cavity of patients with evaluate patients’ nutritional status, including full
dementia are: blood count, electrolytes, B12, urea, creatinine, glucose,
albumin, and ferritin.
• Poor hygiene
Individuals with advanced dementia may show
• Gingivitis with accumulation of bacterial plaque, calculi, and bleeding adversative feeding behaviors that can importantly
• Caries contribute to undernutrition, such as refusal
to eat, wandering, and agitation 101. Research has
• Fractures with remaining roots and eventual infection little evidence that interventions to modify mealtime
• Ulcers, gingival hyperplasia, and lack of taste due to psychotropic environments can improve food and fluid intake for
drugs often used to control symptoms in these patients. these patients102,103. However, given their favorable
risk-benefit and the possibility of improving patients’
quality of life, employing such measures is reasonable104.
Table 6. Factors which negatively influence oral health93.
Box 4 describes our suggested interventions.
• The severity of dementia

• Previous dental history - care and diseases


Box 4. Measures to improve mealtime environment in advanced dementia.
• Ability to receive / consent to oral hygiene care by caregivers
or dental teams Refrain from rushing; good interaction between patients
and caregivers increases food intake
• Knowledge of patients or their caregivers about the importance
of oral health Offer food patients like

• Lack of patient/caregiver motivation If possible, the same caregiver should always be responsible
for assisting patient’s meals
• Impacts of medications on the oral cavity (xerostomia)
Try to set a pleasant, homelike environment with improved lighting
• Lack of information on how to access teams and familiar music
• Degree of knowledge/training of oral health teams regarding Although it is useful to maintain a routine, it is advisable to wait until
dementia and aging patients are calm before offering them food and fluids.
• Teams’ failure to develop strategies and long-term care plans. Use high-contrast colored tableware
• Scarcity of appropriate care facilities for small/medium Consider offering regular snacks and small meals
dental surgeries, day centers, and at home.

Oral Nutritional Supplements (ONS) can increase


macronutrient intake and avoid undernutrition in older
NUTRITIONAL ISSUES IN ADVANCED DEMENTIA adults, including persons with advanced dementia.
As a meta-analysis shows, ONS can increase weight in
Undernutrition in advanced dementia persons with dementia, showing few gastrointestinal side
As dementia advances into a severe stage, feeding effects. Note, however, that the literature lacks enough
difficulties become more common and bring about evidence to aclaimffirm that ONS decrease mortality
important problems such as undernutrition and weight loss, or cognitive deterioration among persons with dementia105.

106   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

Dysphagia in advanced dementia to fail to improve mortality in individuals with advanced


Oropharyngeal dysphagia is a serious and common dementia116-118, may lead to higher levels of discomfort119,
problem in advanced dementia, as it may be an and its complications are responsible for almost half of
important cause of undernutrition and increase the all emergency department visits among patients with
risk of respiratory infections and death106. Early stages advanced dementia 120. Tube feeding also seems to
of dementia may show subtle aspirations and even go increase the risk of pressure ulcers and are unhelpful
unnoticed by patients or caregivers107. Therefore, speech to heal existing pressure ulcers121,122. Finally, enteral
and language therapists should take part in managing feeding with a PEG tube fails to decrease caregivers’
PWD as early as possible, even in the absence of burden121 and raise a perception of better end-of-life
swallowing complaints. The most important instrument care among relatives of individuals with advanced
for diagnosing dysphagia on a day-to-day basis is the dementia123. Current evidence still remains unclear,
Clinical Swallow Evaluation (CSE). it includes both however, on whether such adverse outcomes are related
questionnaires on swallowing problems and a motor to the late use of PEG tubes and if patients in less severe
and sensory examination of all oral structures involved stages of advanced dementia could benefit from it 117,124.
in bolus formation 108,109. More specific situations The decision to withhold tube feeding in advanced
(e.g., to diagnose more accurately aspiration) may dementia is ethical and based on current scientific
require employing an instrumental assessment, such as evidence but should not be viewed as a one-size-fits-all
videofluoroscopic swallow studies (VFSS) and fiberoptic way to approach the matter. The discussion should
endoscopic evaluation of swallowing (FEES)108-110. include families and, whenever possible, patients,
The use of thickeners to change the consistency and consider their social, cultural, and religious values.
of fluids offered to patients with advanced dementia In other words, the decision to use or withhold use
can help to mitigate the consequences of dysphagia. PEG tube feeding must be individualized. Whenever
In 2018, a meta-analysis concluded that thickening tube feeding is contraindicated, assisted oral feeding is
the fluids offered to patients with advanced dementia encouraged. The concept of “comfort feeding only”125
may have an immediate positive effect on swallowing is useful in such situations: it is based on feeding with
and may decrease the three-month incidence of comfort as its main goal, i.e., focusing on satisfaction
pneumonia 111. It noted, however, that long-term and stopping whenever feeding is distressing.
benefits are uncertain due to scarce evidence 111.
Research also lacks enough evidence to recommend
a specific thickness (i.e., nectar- or honey-thick) over the END-OF-LIFE CARE IN DEMENTIA
other112. Adopting a chin-down posture while drinking People with advanced dementia may possibly include a
liquids is another useful intervention in dysphagia great part of the patients living with dementia in Brazil.
and may be as effective as fluid thickening to decrease However, trustful estimates are still impossible due to
the incidence of pneumonia, especially in individuals the lack of epidemiological data and the profile of this
with milder dysphagia112,113. population. Advanced dementia patients show a low
income, associated with a high disease burden profile,
Tube feeding in advanced dementia often lacking the assistance of formal caregivers126.
Usin g or w i t h hold ing t ub e fe e d ing re m a i n s Delivery of care to PWD is essentially provided by family
one of the most controversial topics in the management members in Brazil127, unlike European countries with
of advanced dementia. Caregivers often misinterpret a public health system, such as the United Kingdom128.
the recommendation to not insert a percutaneous Thus, unique characteristics of the Brazilian people and
endoscopic gastrostomy (PEG) as a recommendation its continental-size health system demand adapted
to withdraw all types of care, which might compromise recommendations for appropriate end-of-life care
ongoing treatment and even harm the confidence of people with dementia.
in healthcare providers. Of note, such recommendations End-of-life care, or palliative care, is a therapeutic
are based mostly on observational studies and not on strategy to maintain a person’s quality of life by
randomized controlled trials, as these are unavailable relieving discomfort or stress in a life-limiting condition.
due to ethical reasons114,115. This subtopic details a few steps to aid general
Most of the available evidence suggest that tube practitioners and primary care physicians: (1) accurately
feeding fails to benefit patients with advanced dementia identify the moment of defining end-of-life care,
and may even harm them. The use of PEG tubes seems (2) plan next steps with patients and their family

Brucki SMD, et al.   Management in severe dementia.   107


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

members, and (3) provide mental and physical assistance members must immediately increase awareness
to patients with terminal illness. of end-of-life care. They should raise a community
spirit, reach local policies, and increase their voice
Identifying the moment of end-of-life caring on a nationwide level. Philanthropic institutions
Recognizing individuals with dementia in advanced such as “Associação Brasileira de Alzheimer” (ABraz)
disease stage is a cornerstone to provide adequate may provide information about local hospices or
strategies. In Brazil, almost 80% of individuals low-cost services, though they often have limited
with dementia are still undiagnosed129. A sensitive, regional actions. All levels of care should also address
accurate feeling that forgetfulness may not be solely spiritual needs, as sect leaders should aid planning
associated with the aging process is the first step at the time of diagnosis134.
to provide proper care of patients with severe dementia.
Ideally, the moment of discussing palliative care is Providing mental and physical assistance for patients
as early as dementia is diagnosed, focusing on patients’ (and caregivers)
wishes. Family members are essentially involved in Assisting people with terminal dementia should consider
deliberating decisions when individuals no longer may several levels of care135. The relatively weak healthcare
take complex decisions130. structure for patients with terminal diseases compels
Primary care physicians should have a high suspicion families to establish a plan of care for persons with
of dementia even in patients without cognitive dementia. The whole primary care team should perform
complaints131. Once they identify the cognitive decline, timely planning discussions, including stratifying care
the next step is defining its stage. Clinical features vary demanded by both patients (when cognitively able
in severe stage dementia, including mutism, impaired to take decisions) and caregivers136.
per os, and severe gait disturbances. Recurrent hospital Ideally, they shall create a plan of care whenever
admissions or demanding frequent medical assistance patients receive a diagnosis of end-of-life dementia.
may be early signs of end-of-life disease, especially They must assess patients’ understanding and
involving a recent diagnosis of cancer, heart failure judgement to evaluate their wishes; if impossible,
or chronic obstructive pulmonary disease. Primary this decision should involve their families or caregivers.
healthcare professionals should also pay attention Patients with end-of-life dementia benefit from
to mental status of patients in home care appointments, care levels divided into five stages, ranging from critical
as a great number of individuals were home assisted, care including invasive procedures to supportive care
lacking appropriate follow-up132. and comfort measures only137,138 . Care level must
consider patients’ wishes regarding invasive procedures
Planning the approach to patients and their families in general, including orotracheal tube, central venous
The Brazilian public healthcare system has a major infra- catheter, cardiopulmonary resuscitation, and ICU
structure to provide an interdisciplinary approach to admission (Table 7). Caring should be individualized
families with patients in the end-of-life dementia stage. and performed together with families.
Primary care facilities are strategically located close to Education for caregivers and family members is
the local community, which increases confidence and a fundamental point that must be thoroughly discussed.
access to healthcare. However, most public facilities lack Instructions such as available sources of information
psychologists, physical therapists, speech therapists about end-of-life dementia, where to ask for help,
or social workers. when to ask for assistance (or be taken to the ER),
Once the diagnosis of end-of-life dementia and what is expected and what is not. Assistant physicians
is established, the next priority is to discuss an may provide adequate end-of-life care, including
interdisciplinary approach to provide optimal assistance adequate management of pain, agitation/aggression,
to loved ones. Only a minority of older adults have risk of aspiration, and need for a feeding tube,
access to long-term care institutions 133. Although dyspnea, and pneumonia139.
interdisciplinary care is essential to mitigate end-of-life Caregivers’ mental health must also be addressed.
discomforts involving different healthcare professionals, Since they usually are a family member, they often show
Clinical practice should implement a broad care for a psychological distress associated with the caregivers’
those with access to private physical, occupational, burden 140,141. Caregiver assistance should focus on
psychological, and speech therapy. providing a better quality of life for the entire family.
For most patients with limited access to these Avoiding psychological distress should be a priority,
services, we must discuss a few points. Firstly, family and we must suggest them to perform rotation shifts

108   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

between caregivers (other family members or half-time Thus, end-of-care dementia demands unique,
support caregivers). Mental health hotlines have also individualized strategies which assistant physicians
been progressively implemented to assist those with should recognize. Early identification, conjoint
psychological needs in Brazil142, especially after the planning with families, and with level of care staging
COVID-19 pandemic143. are cornerstones of adequate dementia care.

Table 7. Medical Orders for Scope of Treatment (MOST), in Portuguese, adapted to patients with end-of-life dementia stage. Staging must consider wishes
to admission in a critical care unit, orotracheal intubation and cardiac resuscitation.
Plan demanded by the patient Medical record Plan of care Level of care

I want my life to be preserved using


ICU admission, including orotracheal
all measures of care available, Full intensive care
intubation and CPR
when indicated

I want my life to be preserved,


Critical care:
except when my heart stops. ICU admission.
- Attempt to support life with
I accept ICU admission and all Intensive care without CPR Excluding:
artificial measures in ICUs.
measures, including intubation, Defibrillation and CPR
- These orders in general are
except CPR if my heart stops.
not used in case of natural
I want my life to be preserved, end-of-life outcomes.
ICU admission
except when my heart stops. Intensive care without
Non-invasive ventilation and high-flow O2
I accept ICU admission and all orotracheal intubation or
Excluding:
measures, except CPR if my heart CPR
Intubation, defibrillation, and CPR
stops or in case of intubation.

I want to be treated for reversible


causes but without measures of
artificial life support.
If I show an irreversible condition, Ward care, including treatment for
I want to receive the treatment Advanced ward care reversible causes.
available, except for invasive Excluding: ICU admission, intubation or CPR Ward care:
procedures (intubation or CPR). - Support artificial measures
I want to receive this care will not be initiated if patients’
in this ward. heart or breathing stops
(i.e., intubation or CPR)
I understand death as a natural,
expected event. I accept receiving
Exclusive symptoms management
care to decrease pain or other Basic ward care
and comfort measures
symptoms. I would like to receive
this set of care whenever available.

ETHICAL AND LEGAL ASPECTS individuals, close ones, and healthcare providers
Countless ethical and legal issues arise and evolve and concern future preferences about their medical
regarding the stage and severity of neurodegenerative treatment. Although referring to advanced dementia
diseases. These disorders compromise the psychological and contemplating favorable intentions, they are
well-being and behavior of patients, impacting their unable to predict all possible scenarios for the most
quality of life, and physically and emotionally harming appropriate decision-making. Patients have their own
their families. Dementias generate loss of autonomy characteristics; diseases have different courses, and the
and inability to make decisions, pillars of medical future is unpredictable146.
ethics which support the management of diagnostic Over the past few years, several countries have
or therapeutic medical approaches144. implemented laws regulating care for the dying. Without
In its advanced stage, PWD are unable to care for a doubt, the issue is controversial, especially regarding
themselves145. Advance care planning (ACP) involves shortening patients’ life; colorful debates address
a dynamic process based on a dialogue between euthanasia, though very timidly in Brazil. In patients

Brucki SMD, et al.   Management in severe dementia.   109


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

with severe dementia, the situation becomes more and its quantitative analyses. In dementia, substitute
complex due to patients’ inability to make decisions and decision makers (SDM) are authorized to have frank
report their feelings and suffering147. conversations to predict and document patients’ desire
The literature on severe dementia is significantly prior to their disability. Even when a SDM is appointed
reduced, containing few studies with reliable indicators to provide legally effective consent or refusal, this fails
and guidelines directing healthcare providers in this to render patients’ preferences ethically irrelevant150.
moment of innumerable doubts and uncertainties. In accordance with the DMC issue, doctors must
The lack of scientific evidence predisposes the prescription understand legal consequences, as they are responsible
of aggressive therapies without concern for their for patients and will always be asked, when necessary,
insignificance, especially in intercurrent diseases, such as to report information officially attesting the disease,
the lack of results showing the effectiveness of artificial as well as briefly describing patients’ conditions
nutrition and hydration. Thus, to avoid procedures within expectations. One of these situations is the
incompatible with the dignity of the human life, we need possibility of a guardianship procedure for individuals
to advance concepts such as which prognostic criteria best with dementia, depriving them of the legal capacity
fit survival, how to assess suffering and quality of life in to make decisions and manage their assets. The Brazilian
this special population, and for how long the medications judicial determination of disability (a legal institution
available could prolong life148. provided for in the legislation) finds only a small number
In this sense, evoking the Hippocratic maxim of interdicted patients at an advanced stage of dementia.
of “primum non nocere,” i.e., above all do no harm, The main intention of the interdiction is to protect
is always an appropriate judgment145. individuals with a legally significant disability. The Civil
It is essential to reflect on the development of Procedure Code regulates these guardianship processes,
technologies that prolong life. Clinical practice may “interdiction” in the Brazilian legal language. This means
falsely perceive that this has been accompanied by that Brazil faces a significant lack of legal responsibility
an improvement in the quality of life. Living longer in severe dementia, probably resulting in numerous
fails to necessarily mean patients have had an adequate inappropriate measures which fial to reflect patients’
quality of life, as the opposite is more often the case149. real needs and interests151.
Therefore, when questioned, people claim that living
with quality of life is more relevant than living longer Topics to deal with ethical dilemmas in the advanced
but the measures adopted in many assistance services stages of chronic diseases such as dementias
may be incompatible with this desire. Living without The concept called “Jonsen’s 4-topic” is a practical
a minimum of quality is unacceptable for a considerable approach and structure used by many ethics committees
portion of the cognitively competent population. to resolve clinical ethical dilemmas. It includes
In severe dementia, cardiopulmonary resuscitation a categorization into four similarly weighted quadrants
and other life support measures may seem futile and composed of information, facts, and descriptions
should be carefully evaluated according to the patients’ (Box 5)144,145,151,152.
previous wishes. Refusal to eat and dysphagia are late In conclusion, the general approach to patients
manifestations and uncomfortable ethical dilemmas for with advanced dementia is, as can be seen from the
doctors who care for older adults. The decision to insert above, quite complex. The range of information,
food via an alternative route is complex and dependent difficulties in diagnosis, peculiarities in treatments,
on a multidimensional understanding144. and the mandatory inclusion of family members
In the end, our approach must be proportional in decision-making impose extensive and frequent
to patients’ needs and based on bioethical principles. care to guarantee these individuals the best care based
A bioethical perspective that studies life not only from on the best evidence and, in the absence of such,
a biological point of view but also from a biographical the best experience and common sense available.
one, with the maintenance of life as a right rather To this end, we suggest that patients and their caregivers
than obligation149. are evaluated very closely, at least every three or four
months, to maintain their best possible functionality
Decision-making capacity and maximum comfort, and that the following aspects
Ethically, screening and evaluating individuals for skills receive attention. Respectful care, based on the best
such as decision-making capacity (DMC) should be evidence, individual and family values, ​​and the search
patient-centered and based on a functional assessment, to clarify any remaining doubts should guide the medical
rather than on expectations from an instrument or scale practice before patients and their loved ones.

110   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

Box 5. Bioethical topics.


Medical indication Patient preference
benefits X harms/favoring patients principle of autonomy/honor and respect the patients’ wishes
It refers to the practice of doing good and benefiting others against acts It applies the ethical principle of autonomy and examines patients’ previously
which may be harmful, helping health professionals choose available expressed or assumed beliefs and preferences. It aims to honor and respect
treatments and examining how each alternative raises the possibility of patients’ wishes as much as possible within acceptable limits. In severe
success and favors patients. dementia, it would be supported by an advance directive, with the need
for a SDM as a family member or friend to help with decision-making.
In the absence of advanced directives, it is essential to request the presence
of an SDM. This enables caregivers to determine how patients would like
to be cared for if they were able to decide, whatever it may be. It would
involve their morals, hopes, aspirations, values, principles, and spirituality.
Therefore, sensitivity of those responsible is paramount, which is practically
the current rule in Brazil.

Quality of life Contextual features


Dignity/safety and comfort socio-economic factors/political and religious preferences
The various treatments available should provide a better quality of life (QOL). This last quadrant refers to the exacerbation of ethical dilemmas due
Dimensioning QOL is complex and unique for everyone, even with the help to socio-economic, political, cultural, and religious factors that frequently
of family members’ judgment, care must be taken not to prolong life with appear in advanced dementia and are reflected in care and decision-making.
suffering. Dignity, safety, and comfort must prevail.

ACKNOWLEDGEMENTS
PC, LCS and RN are funded by CNPq, Brazil (bolsa de CELF, RRS, LSM: drafting the manuscript; BJAPB,
produtividade em pesquisa). FACV, JS, LCS, LPS, MLFB, MLFC, NAFF, PC, PHFB, RN,
Authors’ contributions. SMDB, IA, WVB, VC, CELF, RRS, SMDB: critical revision of the manuscript for important
LSM, JS, BJAPB, LPS: concept; SMDB, IA, WVB, VC, intellectual content.

REFERENCES 12. Galasko D, Schmitt F, Thomas R, Jin S, Bennett D, Ferris S. Detailed


assessment of activities of daily living in moderate to severe Alzheimer’s
1. Wimo A, Ljunggren G, Winblad B. Costs of dementia and dementia
disease. J Int Neuropsychol Soc. 2005;11(4):446-53. doi:10.1017/
care: a review. Int J Geriatr Psychiatry. 1997;12(8):841-56. doi:10.1002/
S1355617705050502.
(SICI)1099-1166(199708)12:8<841::AID-GPS652>3.0.CO;2-R.
13. Reisberg B, Ferris SH, Leon MJ, Crook T. The Global deterioration scale
2. Ferretti C, Sarti FM, Nitrini R, Ferreira FF, Brucki SMD. An assessment of
(GDS) for assessment of primary degenerative dementia. Am J Psychiatry.
direct and indirect costs of dementia in Brazil. PLoS ONE. 2018;13(3):1-15.
1982;139(9): 1136-39. doi:10.1176/ajp.139.9.1136.
doi:10.1371/journal.pone.0193209.
14. Reiberg B. Functional Assessment Staging (FAST). Psychopharmacol Bull.
3. Canadian Study of Health and Aging. Study methods and prevalence
1988;24(4):653-59.
of dementia. Can Med Assoc J. 1994;150:899-913.
15. Saxton J, McGonigle-Gibson KL, Swihart AA, Miller VJ, Boller F.
4. Schmitt FA, Wichems CH. A systematic review of assessment and
Assessment of the severely impaired patient: description and validation of
treatment of moderate to severe Alzheimer’s disease. J Clin Psychiatry.
a new neuropsychological test battery. Psychol Assess. 1990;2(3):298-303.
2006;8(3):158-59. doi:10.4088/pcc.v08n0306.
doi:10.1037/1040-3590.2.3.298.
5. Herrmann N, Gauthier S. Diagnosis and treatment of dementia: 16. Harrel LE, Marson D, Chatterjee A, Parrish JA. The Severe Mini-Mental
6. Management of severe Alzheimer’s disease. Can Med Assoc J. State Examination: a new neuropsychologic instrument for the bedside
2008;179(12):1279-87. doi:10.1503/cmaj.070804. assessment of severely impaired patients with Alzheimer disease.
6. Feldman HH, Woodward M. The staging and assessment of moderate Alzheimer Dis Assoc Disord. 2000;14(3):168-75. doi:10.1097/
to severe Alzheimer disease. Neurology. 2005;65(6suppl3):S10-7. 00002093-200007000-00008.
doi:10.1212/WNL.65.6_suppl_3.S10. 17. Wajman JR, Bertolucci PHF: Comparison between neuropsychological
7. Volicer L. Management of Alzheimer’s Disease and End-of-life issues. Clin evaluation instruments for severe dementia. Arq Neuropsiquiatr.
Geriatr Med. 2001;17(2):377-91. doi:10.1016/S0749-0690(05)70074-4. 2006;64(3b): 736-40. doi:10.1590/S0004-282X2006000500007.
8. Voisin T, Vellas B. Diagnosis and treatment of patients with Severe 18. Lyketsos CG, Steinberg M, Tschanz JT, Norton MC, Steffens DC,
Alzheimer’s disease. Drugs Aging. 2009;26:135-44. doi:10.2165/ Breitner JC. Mental and behavioral disturbances in dementia: findings
0002512-200926020-00005. from the Cache County Study on Memory in Aging. Am J Psychiatry.
9. Arrighi HM, Neumann PJ, Lieberburg IM, Townsend RJ. Lethality of 2000;157(5):708-14. doi:10.1176/appi.ajp.157.5.708.
Alzheimer’s disease and its impact on nursing home placement. Alzheimer 19. Fossey J, Ballard C, Juszczak E, James I, Alder N, Jacoby R, Howard R.
Dis Assoc Disord. 2010;24(1):90–5. doi:10.1097/WAD.0b013e31819fe7d1. Effect of enhanced psychosocial care on antipsychotic use in nursing
10. Morris, JC. The Clinical Dementia Rating (CDR): current version and scoring home residents with severe dementia: cluster randomised trial. Br Med J.
rules. Neurology. 1993;43(11):2412-14. doi:10.1212/wnl.43.11.2412-a. 2006;332(7544):756-61. doi:10.1136/bmj.38782.575868.7C.
11. Chaves MLF, Camozzato AL, Godinho C, Kochhann R, Schuh A, 20. Selbaek G, Kirkevold Ø, Engedal K. The prevalence of psychiatric
Almeida VL, et al. Validity of clinical dementia rating scale for the detection symptoms and behavioural disturbances and the use of psychotropic drugs
and stasging dementia in Brazilian patients. Alzheimer Dis Assoc Disord. in Norwegian nursing homes. Int J Geriatr Psychiatry. 2007;22(9),843-49.
2007;21(3):210-17. doi:10.1097/WAD.0b013e31811ff2b4. doi:10.1002/gps.1749.

Brucki SMD, et al.   Management in severe dementia.   111


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

21. Siafarikas N, Selbaek G, Fladby T, Šaltyt BJ, Auning E, Aarsland D. Fre- 40. Wang LY, Shofer JB, Rohde K, Hart KL, Hoff DJ, McFall YH, et al.
quency and subgroups of neuropsychiatric symptoms in mild cognitive Prazosin for the treatment of behavioral symptoms in patients with
impairment and different stages of dementia in Alzheimer’s disease. Alzheimer disease with agitation and aggression. Am J Geriatr Psychiatry.
Int Psychogeriatr. 2018;30(1),103-13. doi:10.1017/S1041610217001879. 2009;17(9):744-51. doi:10.1097/JGP.0b013e3181ab8c61.
22. Cerejeira J, Lagarto L, Mukaetova-Ladinska EB. Behavioral and 41. Ballard C, Youakim JM, Coate B, Stankovic S. Pimavanserin in Alzheimer’s
psychological symptoms of dementia. Front Neurol. 2012;3(73):1-21. Disease Psychosis: Efficacy in Patients with More Pronounced Psychotic
doi:10.3389/fneur.2012.00073. Symptoms. J Prev Alzheimers Dis. 2019;6(1):27-33. doi:10.14283/
23. Toot S, Swinson T, Devine M, Challis D, Orrell M. Causes of nursing jpad.2018.30.
home placement for older people with dementia: A systematic review 42. Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral
and meta-analysis. Int Psychogeriatr. 2017;29(2),195-208. doi:10.1017/ and psychological symptoms in people with Alzheimer’s disease:
S1041610216001654. an international Delphi consensus. Int Psychogeriatr. 2019;31(1):83-90.
24. Kamiya M, Sakurai T, Ogama N, Maki Y, Toba K. Factors associated with doi:10.1017/S1041610218000534.
increased caregivers’ burden in several cognitive stages of Alzheimer’s 43. Bessey LJ, Walaszek A. Management of Behavioral and Psychological
disease. Geriatr Gerontol Int. 2014;14(52):45–55. doi:10.1111/ggi.12260. Symptoms of Dementia. Curr Psychiatry Rep. 2019;21(8):66. doi:10.1007/
25. Bränsvik V, Granvik E, Minthon L, Nordström P, Nägga K. Mortality in s11920-019-1049-5.
patients with behavioural and psychological symptoms of dementia: 44. Reese TR, Thiel DJ, Cocker KE. Behavioral Disorders in Dementia:
a registry-based study. Aging Ment Health. 2021;25(6):1101-9. Appropriate Nondrug Interventions and Antipsychotic Use. Am Fam
doi:10.1080/13607863.2020.1727848. Physician. 2016;94(4):276-82.
26. Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological 45. Vellas B, Gauthier S, Allain H, Andrieu S, Aquino J-P, Berrut G, et al.
interventions for neuropsychiatric symptoms of dementia. Am J Psychiatry. Consensus sur la démence de type Alzheimer au stade severe. Presse
2012;169(9):946-53. doi:10.1176/appi.ajp.2012.11101529. Med 2005;34(20):1545-55. doi:10.1016/S0755-4982(05)84221-6.
27. Fink HA, Linskens EJ, MacDonald R, Silverman PC, McCarten JR, 46. Rongen S, Krammers C, O´mahony D, Feuth TB, Rikkert MGMO, Ahmed
Talley KMC, et al. Benefits and Harms of Prescription Drugs and AIA. Potentially inappropriate prescribing in older patients admitted
Supplements for Treatment of Clinical Alzheimer-Type Dementia. to psychiatric hospital. Int J Geriatr Psychiatry. 2016;31(2):137-45.
Ann Intern Med. 2020;172(10):656-68. doi:10.7326/M19-3887. doi:10.1002/gps.4302.
28. Kennedy RE, Cutter GR, Fowler ME, Schneider LS. Association of 47. McGilton KS, Rochon E, Sidani S, Shaw A, Ben-David BM, Saragosa M,
Concomitant Use of Cholinesterase Inhibitors or Memantine With Cognitive et al. Can we help care providers communicate more effectively with persons
Decline in Alzheimer Clinical Trials: A Meta-analysis. JAMA Netw Open. having dementia living in long-term care homes? Am J Alzheimers Dis
Other Demen. 2017 Feb;32(1):41-50. doi:10.1177/1533317516680899.
2018;1(7):1-10. doi:10.1001/jamanetworkopen.2018.4080.
48. Hickey JV. Neurological and Neurosurgical Nursing. 5th ed. Philadelphia:
29. Chen R, Chan PT, Chu H, Lin YC, Chang PC, Chen CY, Chou KR.
Lippincott Williams & Wilkins, 2003: 1-816.
Treatment effects between monotherapy of donepezil versus combination
49. New hope for advancing neuropalliative care. Lancet Neurol.
with memantine for Alzheimer disease: A meta-analysis. PLoS One.
2021;20(6):409. doi:10.1016/S1474-4422(21)00142-3.
2017;12(8):1-14. doi:10.1371/journal.pone.0183586.
50. Tsolaki M, Papaliagkas V, Anogeianakis G, Bernabei R, Emre M, Frolich L,
30. Wang J, Yu JT, Wang HF, Meng XF, Wang C, Tan CC, Tan L.
et al. Consensus statement on dementia education and training in Europe.
Pharmacological treatment of neuropsychiatric symptoms in Alzheimer’s
J Nutr Health Aging. 2010;14(2):131-35. doi:10.1007/s12603-009-0238-z.
disease: a systematic review and meta-analysis. J Neurol Neurosurg
51. Costa GD, Santos OG, Oliveira MAC. Knowledge, attitudes, and qualification
Psychiatry. 2015;86(1):101-9. doi:10.1136/jnnp-2014-308112.
needs of primary health care professionals in the care of dementia. Rev Bras
31. Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase inhibitors
Enferm. 2020;73(Suppl 3):1-9. doi:10.1590/0034-7167-2020-0330.
in the treatment of neuropsychiatric symptoms and functional impairment
52. Keng A, Brown EE, Rostas A, Rajji TK, Pollock BG, Mulsant BH,
in Alzheimer disease: a meta-analysis. JAMA. 2003;289(2):210-6.
et al. Effectively Caring for Individuals with Behavioral and Psychological
doi:10.1001/jama.289.2.210.
Symptoms of Dementia During the Covid-19 Pandemic. Front Psychiatry.
32. Porsteinsson AP, Drye LT, Pollock BG, Devanand DP, Frangakis C, Ismail Z,
2020;11:1-9. doi:10.3389/fpsyt.2020.573367.
et al. Effect of citalopram on agitation in Alzheimer disease: the CitAD
53. Ferretti C, Nitrini R, Brucki SMD. Virtual Support in Dementia: A possible
randomized clinical trial. JAMA. 2014;311(7):682-91. doi:10.1001/
viable strategy for caregivers. Front Neurol. 2021;12:1-7. doi:10.3389/
jama.2014.93.
fneur.2021.662253.
33. Henry G, Williamson D, Tampi RR. Efficacy and tolerability of 54. Egan KJ, Pinto-Bruno AC, Bighelli I, Berg-Weger M, van Straten A,
antidepressants in the treatment of behavioral and psychological Albanese E, et al. Online Training Support Programs Designed to Improve
symptoms of dementia: a literature review of evidence. Am J Alzheimers Mental Health and Reduce Burden Among Caregivers of People With
Dis Other Demen. 2011;26(3):169-83. doi:10.1177/1533317511402051. Dementia: A Systematic Review. J Am Med Dir Assoc. 2018;19(3):200-6.
34. Ehrhardt S, Porsteinsson AP, Munro CA, Rosenberg PB, Pollock BG, doi:10.1016/j.jamda.2017.10.023.
Devanand DP, et al. Escitalopram for agitation in Alzheimer’s disease 55. Reus VI, Forchtmann LJ, Eyler E, Hilty DM, Horvitz-Lennon M, Jibson MD,
(S-CitAD): Methods and design of an investigator-initiated, randomized, et al. The American Psychiatric Association Practice Guideline on the Use
controlled, multicenter clinical trial. Alzheimers Dement. 2019;15(11):1427-36. of Antipsychotics to Treat Agitation or Psychosis in Patients With Dementia.
doi:10.1016/j.jalz.2019.06.4946. Am J Psychiatry. 2016;173(5):543-6. doi: 10.1176/appi.ajp.2015.173501.
35. Kongpakwattana K, Sawangjit R, Tawankanjanachot I, Bell JS, Hilmer SN, 56. National Institute for Health and Care Excellence. Dementia: A NICE.
Chaiyakunapruk N. Pharmacological treatments for alleviating agitation Suportting people with dementia and their carers.in health and social
in dementia: a systematic review and network meta-analysis. Br J Clin care. Leicester (UK): British Psychological Society; 2012: 1-392.
Pharmacol. 2018;84(7):1445-56. doi:10.1111/bcp.13604. 57. International Psychogeriatric Association [Internet]. Milwalkee (WI): IPA;
36. Yunusa I, Alsumali A, Garba AE, Regestein QR, Eguale T. Assessment of 2015 [citado 23 ago. 2022]. Disponível em: https://www.ipa-online.org/
Reported Comparative Effectiveness and Safety of Atypical Antipsychotics 58. American Geriatrics Society and American Association for Geriatric
in the Treatment of Behavioral and Psychological Symptoms of Psychiatry. Consensus Statement on Improving the Quality of Mental
Dementia: A Network Meta-analysis. JAMA Netw Open. 2019;2(3):1-14. Health Care in U.S. Nursing homes: management of depression and
doi:10.1001/jamanetworkopen.2019.0828. behavioral symptoms associated with dementia. J Am Geriatr Soc.
37. Ruthirakuhan MT, Herrmann N, Abraham EH, Chan S, Lanctôt KL. 2003;51(9):1287-98. doi:10.1046/j.1532-5415.2003.51415.x.
Pharmacological interventions for apathy in Alzheimer’s disease. Cochrane 59. American Medical Association, American Academy of Neurology
Database Syst Rev. 2018;5(5):CD012197. doi:10.1002/14651858. Institute and American Psychiatric Association. Dementia management
CD012197.pub2. quality measurement set update [Internet]. Chicago (IL): American
38. Orgeta V, Tabet N, Nilforooshan R, Howard R. Efficacy of Antidepressants for Medical Association; 2018 [citado 23 ago. 2022]. Disponível em:
Depression in Alzheimer’s Disease: Systematic Review and Meta-Analysis. https://www.aan.com/siteassets/home-page/policy-and-guidelines/
J Alzheimers Dis. 2017;58(3):725-33. doi:10.3233/JAD-161247. quality/quality-measures/2018-dementia-management-measures.pdf
39. Cummings JL, Lyketsos CG, Peskind ER, Porsteinsson AP, Mintzer JE, 60. Ismail Z, Black SE, Camicioli R, Chertkow H, Herrmann N, Laforce R,
Scharre DW, et al. Effect of Dextromethorphan-Quinidine on Agitation in et al. Recommendations of the 5th Canadian Consensus Conference
Patients With Alzheimer Disease Dementia: A Randomized Clinical Trial. on the diagnosis and treatment of dementia. Alzheimers Dement.
JAMA. 2015;314(12):1242-54. doi:10.1001/jama.2015.10214. 2020;16(8):1182-95. doi:10.1002/alz.12105.

112   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

61. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management 84. Husebo B, Ballard C, Sandvik R, Nilsen OB, Aarsland D. Efficacy of treating
of behavioral and psychological symptoms of dementia. Br Med J. pain to reduce behavioural disturbances in residents of nursing homes
2015;350:1-16. doi:10.1136/bmj.h369. with dementia: cluster randomised clinical trial. Br Med J, 2011;343:1-11.
62. Brodaty H, Arasaratnam C. Meta-Analysis of Non-Pharmacological doi:10.1136/bmj.d4065.
Interventions for Neuropsychiatric Symptoms of Dementia. Am J Psychiatry. 85. Hyland K, Fiske J, Matthews N. Nutritional and dental health management
2012;169(9):946-53. doi:10.1176/appi.ajp.2012.11101529. in Parkinson’s disease. J Community Nurs. 2000;14(1):28-32.
63. Livingston G, HuntleyJ, Sommerlad A, Ames D, Ballard C, Banerjee S, 86. James PA, Oparil S, Carter BL, William CC, Handler J, Lackland DT, et al.
et al. Dementia prevention, intervention, and care: 2020 report of Evidence-Based Guideline for the Management of High Blood Pressure in
the Lancet Comission. Lancet. 2020;396:413-46. doi:10.1016/ Adults – Report From the Panel Members Appointed to the Eighth Joint
S0140-6736(20)30367-6. National Committee (JNC 8). JAMA. 2014;311(5):507-20. doi:10.1001/
64. Caramelli P, Bottino CMC. Tratando os sintomas comportamentais e jama.2013.284427.
psicológicos da demência (SCPD). Conferência Clínica. J Bras Psiquiatr. 87. Lee DS, Austin PC, Rouleau JL, Liu PP, Naimark D, Tu JV. Predicting
2007;56(2):83-7. doi:10.1590/S0047-20852007000200002. Mortality Among Hospitalized Patients for Heart Failure – derivation and
65. Eisenmann Y, Golla H, Schmidt H, Voltz R, Perrar KM. Palliative Care variation of a clinical model. JAMA. 2003;290(19):2581-87. doi:10.1001/
in Advanced Dementia. Front Psychiatry. 2020;11:1-13. doi:10.3389/ jama.290.19.2581.
fpsyt.2020.00699. 88. Lyketsos CG, Colenda CC, Beck C, Blank K, Doraiswamy MP,
66. Ferretti CEL. Intervenções de Enfermagem nas Doenças Neurodegenerativas. Kalunian DA, et al. Position Statement of the American Association
In: Koizume MS, Diccini S (orgs). Enfermagem em Neurociência: for Geriatric Psychiatry Regarding Principles of Care for Patients With
Fundamentos para a prática clínica. São Paulo (SP): Atheneu; 2006: 1-672. Dementia Resulting From Alzheimer Disease. Am J Geriatr Psychiatry.
67. Bessey LJ, Walaszek A. Management of behavioral and psychological 2006;14(7):561-73. doi:10.1097/01.JGP.0000221334.65330.55.
symptoms of dementia. Curr Psychiatry Rep. 2019;21(8):66. doi:10.1007/ 89. McWillimas L, Farrell C, Grande G, Keady J, Swarbrick C, Yorke J.
s11920-019-1049-5. A systematic review of the prevalence of comorbid cancer and dementia
68. de Vugt ME, Stevens F, Aalten P, Lousberg R, Jaspers N, Winkens I, and its implication for cancer-related care. Aging Ment Health.
et al. Do caregiver management strategies influence patient behavior? 2017;22(10):1254-71. doi:10.1080/13607863.2017.1348476.
Int J Geriatr Psychiatry. 2004;19(1):85-92. doi:10.1002/gps.1044. 90. Manchery N, Subbiah GK, Nagappan N, Premnath P. Are oral health
69. Abreu W, Tolson D, Jackson GA, Staines H, Costa N. The relationship education for carers effective in the oral hygiene management of elderly
between frailty, functional dependence, and healthcare needs among with dementia? A systematic review. Dent Res J. 2020;17(1):1-9.
community-dwelling people with moderate to severe dementia. Health 91. Mitchell SL. Advanced Dementia. N Engl J Med. 2015;372:2533-40.
Soc Care Community. 2018;27(3):642-53. doi:10.1111/hsc.12678. doi:10.1056/NEJMcp1412652.
70. Aguero-Torres H, Fratiglioni L, Guo Z, Viitanen M, Winblad B. Prognostic 92. Morrison RS, Siu AL. A comparison of pain and its treatment in advanced
Factors in Very Old Demented Adults: A Seven-Year Follow-Up dementia and cognitively intact patients with hip fracture. J Pain Symptom
Manag. 2000;19(4):240-8. doi:10.1016/S0885-3924(00)00113-5.
From a Population-Based Survey in Stockholm. J Am Geriatr Soc.
93. Reisberg B, Wegiel J, Franssen E, Kadiyala S, Auer S, Souren L,et al.
1988;46(4):444-52. doi:10.1111/j.1532-5415.1998.tb02464.x.
Clinical Features of Severe Dementia: Staging. In: Burns A, Winblad B
71. Allen RS, Thorn BE, Fisher EF, Gerstle J, Quarles K, Bourgeois MS, et al.
(orgs). Severe Dementia. Hoboken: John Wiley & Sons Ltd, 2006: 1-260.
Prescription and Dosage of Analgesic Medication in Relation to Resident
94. Schubert CC, Bustani M, Callahan CM, Perkins AJ, Carney CP,
Behaviors in the Nursing Home. J Am Geriatr Soc. 2003;51(4):534-8.
Fox C, et al. Comorbidity Profile of Dementia Patients in Primary Care:
doi:10.1046/j.1532-5415.2003.51164.x.
Are They Sicker? J Am Geriatr Soc. 2006;54(1):104-9. doi:10.1111/
72. Bauer K, Schwarzkopf L, Graessel E, Holle R. A claims data-based
j.1532-5415.2005.00543.x.
comparison of comorbidity in individual with and without dementia. BMC
95. Sloan, FA, Trogdon JG, Curtis LH, Schulman KA. The Effect of Dementia
Geriatrics. 2014;14(10):1-13. doi:10.1186/1471-2318-14-10.
on Outcomes and Process of Care for Medicare Beneficiaries Admitted
73. Bynum JP, Rabins PV, Weller W, Niefeld M, Anderson GF, Wu AW.
with Acute Myocardial Infarction. J Am Geriatr Soc. 2004;52(2):173-81.
The Relationship Between a Dementia Diagnosis, Chronic Ilness, Medical
doi:10.1111/j.1532-5415.2004.52052.x.
Expenditures and Hospital use. J Am Geriatr Soc. 2004;52(2):187-94.
96. Surr C, Griffiths AW, Kelley R, Ashley L, Cowdell F, Henry A, et al.
doi:10.1111/j.1532-5415.2004.52054.x.
Navigating cancer treatment and care when living with comorbid dementia:
74. Cintra, MTG, Rezende, NA; Torres, HOG. Advanced Dementia in a sample
an ethnographic study. Support Care Cancer. 2021;29:2571-79.
of Brazilian elderly: Sociodemographic and morbidity analysis. Rev Assoc
doi:10.1007/s00520-020-05735-z.
Med Bras. 2016;62(8):735-41. doi:10.1590/1806-9282.62.08.735. 97. Terpenning MS, Taylor GW, Lopatin DE, Kerr CK, Dominguez BL,
75. Doraiswamy PM, Leon J, Cummings J, Marin D, Neumann P. Prevalence Loesche WJ. Aspiration pneumonia: dental and oral risk factors in an older
and Impact of Medical Comorbity in Alzheimer’s Disease. J Gerontol A Biol veteran population. J Am Geriatr Soc. 2001;49(5):557-89. doi:10.1046/
Sci Med Sci. 2002;57(3):173-7. doi:10.1093/gerona/57.3.m173. j.1532-5415.2001.49113.x.
76. Downs M, Small N, Froggatt K. Person-centred Care for People with 98. Fiske J, Frenkel H, Griffiths J, Jones V; British Society of Gerodontology;
Severe Dementia. In: Burns A, Winblad B (orgs). Severe Dementia. British Society for Disability and Oral Health. Guidelines for the
New York: Wiley; 2007: 1-260. development of local standards of oral health care for people with
77. Fiske J, Griffiths J, Jamieson R, Manger D. Guidelines for oral health care dementia. Gerodontology. 2006;23 Suppl 1:3-32. doi:10.1111/
for long-stay patients and residents. Gerodontology. 2008;17(1):55-64. j.1741-2358.2006.00140.x.
doi:10.1111/j.1741-2358.2000.00055.x. 99. van Dijk PTM, Dippell DWJ, Meulen JHP, Habbema JDF. Comorbidity and
78. Fulton AT, Rhodes-Kropf J, Corcoran AM, Chau D, Castillo EH. Palliative Its Effects on Mortality in Nursing Home Patients with Dementia. J Nerv
Care for Patitents With Dementia in Long-Term Care. Clin Geriatr Med. Ment Dis. 1996;184(3):180-7. doi:10.1097/00005053-199603000-00007.
2011;27(2):153-70. doi:10.1016/j.cger.2011.01.002. 100. Volicer L, McKee A, Hewitt S. Dementia. Neurol Clin. 2001;19(4):867-85.
79. Gauthier S, Patterson C, Chertkow H, Gordon M, Herrmann N, Rockood doi:10.1016/S0733-8619(05)70051-7.
K, Rosa-Neto K, Soucy JP. 4th Canadian Consensus Conference 101. Warden V, Hurley AC, Volicer L. Development and Psychometric Evaluation
on the Diagnosis and Treatment of Dementia. Can J Neurol Sci. of the Pain Assessment in Advanced Dementia (PAINAD). Scale J Am Med
2012;39(6Suppl5):1-8. doi:10.1017/s0317167100015183. Dir Assoc. 2003;4:9-15. doi:10.1097/01.JAM.0000043422.31640.F7.
80. Herrera E, Caramelli P, Silveira ASB, Nitrini R. Epidemiologic survey of 102. Wolf-Klein GP, Silverstone F, Brod MS, Levy A, Foley CJ, Termotto V, Breuer
Dementia in a community-dwelling Brazilian population. Alzheimer Dis Assoc J. Are Alzheimer Patients Healthier? J Am Geriatr Soc. 1988;36(3):219-24.
Disord. 2002; 16(2):103-8. doi:10.1097/00002093-200204000-00007. doi:10.1111/j.1532-5415.1988.tb01804.x.
81. Herrman N, Gauthier S Diagnosis and treatment of dementia: 6. 103. Xue QI. The Frailty Syndrome: Definition and Natural History. Clin Geriatr
Management of severe Alzheimer disease. Can Med Am J. Med. 2011;27(1):1-15. doi:10.1016/j.cger.2010.08.009.
2008;179(12):1279-87. doi:10.1503/cmaj.070804. 104. Zhu CW, Consentino S, Ornstein KA, Gu Y, Andrews H, Stern Y. Interactive
82. Hill JW, Futterman R, Duttagupta S, Mastey V, Lloyd JR, Fillit H. Alzheimer’s Effect of Dementia Severity and Comorbidities on Medical Expenditures.
disease and related dementias increase costs of comorbidities in managed J Alzheimers Dis. 2017;57(1):301-15. doi:10.3233/JAD-161077.
Medicare. Neurology. 2002;58(1):62-70. doi:10.1212/WNL.58.1.62. 105. White H, Pieper C, Schmader K. The association of weight change
83. Horgas AL, Elliott A, Marsiske M. Pain Assessment in Persons with in Alzheimer’s disease with severity of disease and mortality:
Dementia: Relationship between Self-report and Behavioral Observation. A longitudinal analysis. J Americ Geriatr Soc. 1998;46(10):1223-7.
J Am Geriatr Soc. 2009;57(1):126-32. doi:10.1111/j.1532-5415.2008.02071.x. doi:10.1111/j.1532-5415.1998.tb04537.x.

Brucki SMD, et al.   Management in severe dementia.   113


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

106. Hanson LC, Ersek M, Lin FC, Carey TS. Outcomes of feeding problems 127. Pasman HRW, Onwuteaka-Philipsen BD, Kriegsman DMW, Ooms ME,
in advanced dementia in a nursing home population. J Am Geriatr Soc. Ribbe MW, van der Wal G. Discomfort in nursing home patients with severe
2013;61(10):1692–7. doi:10.1111/jgs.12448. dementia in whom artificial nutrition and hydration is forgone. Arch Intern
107. Rubenstein LZ, Harker JO, Salvà A, Guigoz Y, Vellas B. Screening Med. 2005;165(15):1729-35. doi:10.1001/archinte.165.15.1729.
for undernutrition in geriatric practice: Developing the short-form 128. Givens JL, Selby K, Goldfeld KS, Mitchell SL. Hospital transfers of
mini-nutritional assessment (MNA-SF). J Geront Series A Biolog Sci nursing home residents with advanced dementia. J Am Geriatr Soc.
Med Sci. 2001;56(6):M366-72. doi:10.1093/gerona/56.6.m366. 2012;60(5):905-9. doi:10.1111/j.1532-5415.2012.03919.x.
108. Stratton RJ, Hackston A, Longmore D, Dixon R, Price S, Stroud M, et al. 129. Cintra MTG, Rezende NA, Torres HO, Cintra MTG, Rezende NA, Torres OG.
Malnutrition in hospital outpatients and inpatients: Prevalence, concurrent Advanced dementia in a sample of Brazilian elderly: Sociodemographic
validity and ease of use of the “malnutrition universal screening tool” and morbidity analysis. Rev Assoc Méd Bras. 2016;62(8):735-41.
(‘MUST’) for adults. British J Nut. 2004;92(5):799–808. doi:10.1079/ doi:10.1590/1806-9282.62.08.735.
bjn20041258. 130. Teno JM, Gozalo P, Mitchell SL, Kuo S, Fulton AT, Mor V. Feeding Tubes
109. Volkert D, Chourdakis M, Faxen-Irving G, Frühwald T, Landi F, and the Prevention or Healing of Pressure Ulcers. Arch Intern Med.
Suominen, et al. ESPEN guidelines on nutrition in dementia. Clin Nutr. 2012;172(9):697–701. doi:10.1001/archinternmed.2012.1200.
2015;34(6):1052–73. doi:10.1016/j.clnu.2015.09.004. 131. Teno JM, Mitchell SL, Kuo SK, Gozalo PL, Rhodes RL, Lima JC,
110. Herke M, Fink A, Langer G, Wustmann T, Watzke S, Hanff AM, et al. et al. Decision-making and outcomes of feeding tube insertion:
Environmental and behavioural modifications for improving food and A five-state study. J Am Geriatr Soc. 2011;59(5):881–6. doi:10.1111/
fluid intake in people with dementia. Cochrane Database Syst Rev. j.1532-5415.2011.03385.x.
2018;7(7):CD011542. doi:10.1002/14651858.CD011542.pub2. 132. Goldberg LS, Altman KW. The role of gastrostomy tube placement in
111. Liu W, Cheon J, Thomas SA. Interventions on mealtime difficulties in advanced dementia with dysphagia: A critical review. Clin Interv Aging.
older adults with dementia: A systematic review. Int J Nurs Studies. 2014;9:1733-9. doi:10.2147/CIA.S53153.
2014;51(1):14–27. doi:10.1016/j.ijnurstu.2012.12.021. 133. Palecek EJ, Teno JM, Casarett DJ, Hanson LC, Rhodes RL, Mitchell SL.
112. Abdelhamid A, Bunn D, Copley M, Cowap V, Dickinson A, Gray L, et al. Comfort Feeding Only: A Proposal to Bring Clarity to Decision-Making
Effectiveness of interventions to directly support food and drink intake Regarding Difficulty with Eating for Persons with Advanced Dementia. J Am
in people with dementia: Systematic review and meta-analysis. BMC Geriatr Soc. 2010;58(3):580–4. doi:10.1111/j.1532-5415.2010.02740.x.
Geriatrics. 2016;16(1):26. doi:10.1186/s12877-016-0196-3. 134. Azevedo LVDS, Calandri IL, Slachevsky A, Graviotto HG, Vieira MCS,
113. Prince M, Albanese E, Guerchet M, Prina M. Nutrition and dementia: Andrade CB, et al. Impact of Social Isolation on People with Dementia
A review of available research. Chertsey: Compass group, 2014. and Their Family Caregivers. J Alzheimers Dis. 2021;81(2):607-17.
114. Espinosa-Val MC, Martín-Martínez A, Graupera M, Arias O, doi:10.3233/JAD-201580.
Elvira A, Cabré M, et al. Prevalence, Risk Factors, and Complications 135. Bamford C, Lee R, McLellan E, Poole M, Harrison-Dening K, Hughes J,
of Oropharyngeal Dysphagia in Older Patients with Dementia. Nutrients. et al. What enables good end of life care for people with dementia?
2020;12(3):1-15. doi:10.3390/nu12030863. A multi-method qualitative study with key stakeholders. BMC Geriatr.
115. Özsürekci C, Arslan SS, Demir N, Çalı kan H, engül Ayçiçek G, Kılınç HE, 2018;18(1):302. doi:10.1186/s12877-018-0983-0.
et al. Timing of Dysphagia Screening in Alzheimer’s Dementia. J Parenter 136. Borelli WV, Augustin MC, Oliveira PBF, Reggiani LC, Bandeira-de-Mello RG,
Enteral Nutr. 2020;44(3):516–24. doi:10.1002/jpen.1664. Schumacher-Schuh AF, et al. Neuropsychiatric Symptoms in Patients with
116. Alagiakrishnan K, Bhanji RA, Kurian M. Evaluation and management of Dementia Associated with Increased Psychological Distress in Caregivers
oropharyngeal dysphagia in different types of dementia: A systematic During the COVID-19 Pandemic. J Alzheimers Dis. 2021;80(4):1705-12.
review. Archives Gerontology Geriatrics. 2013;56(1):1–9. doi:10.1016/ doi:10.3233/JAD-201513.
j.archger.2012.04.011. 137. Cintra MT, Rezende NA, Torres HO. Advanced dementia in a sample of
117. Boccardi, V., Ruggiero, C., Patriti, A., Marano, L. Diagnostic Assessment Brazilian elderly: Sociodemographic and morbidity analysis. Rev Assoc
and Management of Dysphagia in Patients with Alzheimer’s Disease. Méd Bras. 2016;62(8):735-41. doi:10.1590/1806-9282.62.08.735.
J Alzheimers Dis. 2016;50(4):947–55. doi:10.3233/JAD-150931. 138. Citko J, Moss AH, Carley M, Tolle S. The National POLST Paradigm
118. Egan A, Andrews C, Lowit A. Dysphagia and mealtime difficulties Initiative. J Palliat Med. 2011;14(2):241-2. doi:10.1089/jpm.2010.9730.
in dementia: Speech and language therapists’ practices and perspectives. 139. Dezorzi LW, Raymundo MM, Goldim JR, Oliveira CAV. Spirituality in
Int J Lang Commun Disord. 2020;55(5):777–92. doi:10.1111/ the continuing education of healthcare professionals: An approach to
1460-6984.12563. palliative care. Palliat Support Care. 2019;17(6):662-7. doi: 10.1017/
119. Flynn E, Smith CH, Walsh CD, Walshe M. Modifying the consistency of food S1478951519000117.
and fluids for swallowing difficulties in dementia. Cochrane Database System 140. Durgante H, Contreras ML, Backhouse T, Mavrodaris A, Ferreira MG,
Rev. 2018;9(9):CD011077. doi:10.1002/14651858.CD011077.pub2. Paulo DLV, et al. Challenges in dementia care: comparing key issues from
120. Robbins J, Gensler G, Hind J, Logemann JA, Lindblad AS, Brandt D, Brazil and the United Kingdom. Dement Neuropsychol, 2020;14(3):216-22.
et al. Comparison of 2 interventions for liquid aspiration on pneumonia doi:10.1590/1980-57642020dn14-030003.
incidence: A randomized trial. Ann Intern Med. 2008;148(7):509-18. 141. Harzheim E, Gonçalves MR, Umpierre RN, Silva Siqueira AC, Katz N,
doi:10.7326/0003-4819-148-7-200804010-00007. Agostinho MR, et al. Telehealth in Rio Grande do Sul, Brazil: Bridging the
121. Saconato, M., Chiari, B. M., Lederman, H. M., Gonçalves, M. I. R. Gaps. Telemed E-Health. 2016;22(11):938-44. doi:10.1089/tmj.2015.0210.
Effectiveness of Chin-tuck Maneuver to Facilitate Swallowing in 142. Hickman SE, Keevern E, Hammes BJ. Use of the physician orders for life-
Neurologic Dysphagia. Int Arch Otorhinolaryngol. 2016;20(1):13–7. sustaining treatment program in the clinical setting: a systematic review of
doi:10.1055/s-0035-1564721. the literature. J Am Geriatr Soc. 2015;63(2):341-50. doi:10.1111/jgs.13248.
122. Ijaopo EO, Ijaopo RO. Tube feeding in individuals with advanced 143. Marcucci FCI, Cabrera MAS, Perilla AB, Brun MM, Barros, EML,
dementia: a review of its burdens and perceived benefits. J Aging Res. Martins VM, Rosenberg JP, Yates P. Identification and characteristics of
2019:7272067. doi:10.1155/2019/7272067. patients with palliative care needs in Brazilian primary care. BMC Palliative
123. Orlandoni P, Jukic Peladic N, Cherubini A. Enteral nutrition in advanced Care. 2016;15(1), 1-10. doi:10.1186/s12904-016-0125-4.
dementia: An unresolved dilemma in clinical practice. Eur Geriatr Med. 144. Nakamura AE, Opaleye D, Tani G, Ferri CP. Dementia underdiagnosis in
2020;11(2):191–4. doi:10.1007/s41999-020-00292-4. Brazil. Lancet, 2015;385(9966):418-9. doi:10.1016/S0140-6736(15)60153-2.
124. Murphy LM, Lipman TO. Percutaneous endoscopic gastrostomy 145. Nitrini R, Barbosa MT, Brucki SMD, Yassuda MS, Caramelli P. Current
does not prolong survival in patients with dementia. Arch Intern Med. trends and challenges on dementia management and research in Latin
2003;163(11):1351–3. doi:10.1001/archinte.163.11.1351. America. J Global Health, 2020;10(1):1-10. doi:10.7189/jogh.10.010362.
125. Ribeiro Salomon, AL, Novaes MR. Outcomes of enteral nutrition for 146. Sampson EL, Candy B, Davis S, Gola AB, Harrington J, King M,et al.
patients with advanced dementia: A systematic review. J Nutr Health Living and dying with advanced dementia: A prospective cohort study
Aging. 2015;19(2):169–77. doi:10.1007/s12603-014-0517-1. of symptoms, service use and care at the end of life. Palliat Med,
126. Sanders, DS, Carter MJ, D’Silva J, James G, Bolton RP, Bardhan KD. 2017;32(3):668-81. doi:10.1177/0269216317726443.
Survival analysis in percutaneous endoscopic gastrostomy feeding: 147. Schafirovits-Morillo L, Suemoto CK. Severe dementia: A review on diagnoses,
A worse outcome in patients with dementia. Am J Gastroenterol. therapeutic management and ethical issues. Dement Neuropsychol,
2000;95(6):1472-5. doi:10.1111/j.1572-0241.2000.02079.x. 2010;4(3):158-64. doi:10.1590/S1980-57642010DN40300003.

114   Management in severe dementia.   Brucki SMD, et al.


Dement Neuropsychol 2022 September;16(3 Suppl. 1):96-115

148. Viana BM, Bicalho MAC, Moraes EN, Romano-Silva MA. Twenty-four-year 153. Cipriani G, Di Fiorino M. Euthanasia and other end of life in patients
demographic trends of a Brazilian long-term care institution for the suffering from dementia. Leg Med. 2019;40:54-9. doi:10.1016/
aged. J Am Med Dir Assoc. 2015;16(2):174.e1-6. doi:10.1016/ j.legalmed.2019.07.007.
j.jamda.2014.11.012. 154. Congedo M, Causarano R, Alberti F, Bonito V, Borghi L, Colombi L,
149. Wittmann-Vieira R, Goldim JR. Bioética e cuidados paliativos: tomada et al. Ethical issues in end of life treatments for patients with dementia.
de decisões e qualidade de vida. Acta Paul Enferm. 2012;25(3):334–9. Eur J Neurol. 2010;17:774-9. doi:10.1111/j.1468-1331.2010.02991.x.
doi:10.1590/S0103-21002012000300003. 155. Fontaneda AJD, Sainz MIL. Problemas éticos y legales en la demência
150. Low JA, Ho E. Managing ethical dilemmas in end-stage neurodegenerative
severa. El derecho a morir en paz. Rev Esp Ger Gerontol. 2009;44(2):43-
diseases. Geriatrics. 2017;2(1):1-7. doi:10.3390/geriatrics2010008.
7. doi:10.1016/j.regg.2009.06.011.
151. Passmore MJ, Ho A, Gallagher R. Behavioral and psychological symptoms
in moderate to severe Alzheimer’s disease: a palliative care approach 156. Allen W. Medical ethics issues in dementia and end of life. Curr Psychiatry
emphasizing recognition of personhood and preservation of dignity. Rep. 2020;22(6):31. doi:10.1007/s11920-020-01150-7.
J Alzheimers Dis. 2012;29(1):1-13. doi:10.3233/JAD-2012-111424. 157. Bello DVME, Schultz RR. Dementia and legal determination of capacity. Arq
152. Piers R, Albers G, Gilissen J, De Lepeleire J, Steyaert J, van Mechelen W, Neuro-Psiquiatr. 2017;75(6):349-53. doi:10.1590/0004-282X20170061.
et al. Advance care planning in dementia: recommendations for heal- 158. Toh HJ, Low JA, Lim ZY, Lim Y, Siddiqui S, Tan L. Jonsen’s four topics
thcare professionals. BMC Palliat Care. 2018;17(1):88. doi:10.1186/ approach as a framework for clinical ethics consultation. Asian Bioeth.
s12904-018-0332-2. 2018;10(1):37-51. doi:10.1007/s41649-018-0047-y.

Brucki SMD, et al.   Management in severe dementia.   115

You might also like