ArticlePDF Available

Lifestyle Risk Factors and Cognitive Outcomes from the Multidomain Dementia Risk Reduction Randomized Controlled Trial, Body Brain Life for Cognitive Decline (BBL-CD)

Authors:

Abstract and Figures

BACKGROUND/OBJECTIVES: To evaluate the efficacy of a multidomain intervention to reduce lifestyle risk factors for Alzheimer’s disease (AD) and improve cognition in individuals with subjective cognitive decline (SCD) or mild cognitive impairment (MCI). DESIGN: The study was an 8-week two-arm single-blind proof-of-concept randomized controlled trial. SETTING: Community-dwelling individuals living in Canberra, Australia, and surrounding areas. PARTICIPANTS: Participants were 119 individuals (intervention n = 57; control n = 62) experiencing SCD or MCI. INTERVENTION: The control condition involved four educational modules covering dementia and lifestyle risk factors, Mediterranean diet, physical activity, and cognitive engagement. Participants were instructed to implement this information into their own lifestyle. The intervention condition included the same educational modules and additional active components to assist with the implementation of this information into participants’ lifestyles: dietitian sessions, an exercise physiologist session, and online brain training. MEASUREMENTS: Lifestyle risk factors for AD were assessed using the Australian National University-Alzheimer’s Disease Risk Index (ANU-ADRI), and cognition was assessed using Alzheimer’s Disease Assessment Scale-Cognitive subscale, Pfeffer Functional Activities Questionnaire, Symbol Digit Modalities Test (SDMT), Trail Making Test-B, and Category Fluency. RESULTS: The primary analysis showed that the intervention group had a significantly lower ANU-ADRI score (X2 = 10.84; df = 3; P = .013) and a significantly higher cognition score (X2 = 7.28; df = 2; P = .026) than the control group. A secondary analysis demonstrated that the changes in lifestyle were driven by increases in protective lifestyle factors (X2 = 12.02; df = 3; P = .007), rather than a reduction in risk factors (X2 = 2.93; df = 3; P = .403), and cognitive changes were only apparent for the SDMT (X2 = 6.46; df = 2; P = .040). Results were robust to intention-to-treat analysis controlling for missing data. CONCLUSION: Results support the hypothesis that improvements in lifestyle risk factors for dementia can lead to improvements in cognition over a short time frame with a population experiencing cognitive decline. Outcomes from this trial support the conduct of a larger and longer trial with this participant group.
Content may be subject to copyright.
CLINICAL INVESTIGATION
Lifestyle Risk Factors and Cognitive Outcomes from the
Multidomain Dementia Risk Reduction Randomized Controlled
Trial, Body Brain Life for Cognitive Decline (BBL-CD)
Mitchell McMaster, Psych. Hons.,* Sarang Kim, PhD,
Linda Clare, ScD,
Susan J. Torres, PhD,
§
Nicolas Cherbuin, PhD,* Catherine DʼEste, PhD,
and Kaarin J. Anstey, PhD**
††
BACKGROUND/OBJECTIVES: To evaluate the efcacy
of a multidomain intervention to reduce lifestyle risk factors
for Alzheimers disease (AD) and improve cognition in indi-
viduals with subjective cognitive decline (SCD) or mild cog-
nitive impairment (MCI).
DESIGN: The study was an 8-week two-arm single-blind
proof-of-concept randomized controlled trial.
SETTING: Community-dwelling individuals living in Can-
berra, Australia, and surrounding areas.
PARTICIPANTS: Participants were 119 individuals (inter-
vention n = 57; control n = 62) experiencing SCD or MCI.
INTERVENTION: The control condition involved four
educational modules covering dementia and lifestyle risk
factors, Mediterranean diet, physical activity, and cognitive
engagement. Participants were instructed to implement this
information into their own lifestyle. The intervention condi-
tion included the same educational modules and additional
active components to assist with the implementation of this
information into participantslifestyles: dietitian sessions,
an exercise physiologist session, and online brain training.
MEASUREMENTS: Lifestyle risk factors for AD were
assessed using the Australian National University-Alzheimers
Disease Risk Index (ANU-ADRI), and cognition was assessed
using Alzheimers Disease Assessment Scale-Cognitive sub-
scale, Pfeffer Functional Activities Questionnaire, Symbol Digit
Modalities Test (SDMT), Trail Making Test-B, and Category
Fluency.
RESULTS: The primary analysis showed that the interven-
tion group had a signicantly lower ANU-ADRI score (χ
2
=
10.84; df =3;P= .013) and a signicantly higher cognition
score (χ
2
= 7.28; df =2;P= .026) than the control group. A
secondary analysis demonstrated that the changes in life-
style were driven by increases in protective lifestyle factors
(χ
2
= 12.02; df =3;P= .007), rather than a reduction in
risk factors (χ
2
= 2.93; df =3;P= .403), and cognitive
changes were only apparent for the SDMT (χ
2
= 6.46; df =2;
P= .040). Results were robust to intention-to-treat analysis
controlling for missing data.
CONCLUSION: Results support the hypothesis that
improvements in lifestyle risk factors for dementia can lead
to improvements in cognition over a short time frame with a
population experiencing cognitive decline. Outcomes from
this trial support the conduct of a larger and longer trial with
this participant group. J Am Geriatr Soc 68:2629-
2637, 2020.
Keywords: dementia prevention; lifestyle risk reduction;
subjective cognitive decline; mild cognitive impairment;
nonpharmacological intervention
The number of people with dementia is expected to rise
to 82 million by 2030 and to more than 152 million
by 2050.
1
Research has shown that together lifestyle risk
factors are responsible for between one-third and one-half
of all Alzheimers disease (AD) cases.
2,3
It is imperative that
interventions to reduce risk are designed to limit the num-
ber of people developing dementia. This can be achieved
From the *Centre for Research on Ageing, Health and Wellbeing
(CRAHW), The Australian National University, Canberra, Australia;
Wicking Dementia Research and Education Centre, University of
Tasmania, Hobart, Australia;
Centre for Research in Ageing and Cognitive
Health (REACH), University of Exeter, Exeter, UK;
§
School of Exercise and
Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin
University, Geelong, Victoria, Australia;
National Centre for Epidemiology
and Population Health (NCEPH), The Australian National University,
Canberra, Australia;
School of Medicine and Population Health, The
University of Newcastle, Newcastle, Australia; **Neuroscience Research
Australia (NeuRA), Sydney, Australia; and the
††
School of Psychology,
University of New South Wales, Randwick, NSW, Australia.
Address correspondence to Mitchell McMaster, Centre for Research on
Ageing, Health & Wellbeing, The Australian National University, Florey
Building (54), Mills Road, Acton, ACT 2601, Australia. E-mail: mitchell.
mcmaster@anu.edu.au
DOI: 10.1111/jgs.16762
JAGS 68:2629-2637, 2020
© 2020 The American Geriatrics Society 0002-8614/20/$15.00
through primary prevention, focusing on lowering the
dementia risk for cognitively normal individuals, and sec-
ondary prevention aimed at high-risk individuals beginning
to experience subjective cognitive decline (SCD) or mild
cognitive impairment (MCI).
4
SCD and MCI are considered
to be prodromes of dementia, and emerging evidence clearly
demonstrates mild forms of neuropathology related to
dementia in both conditions.
5
Although some evidence is available from large lifestyle
interventions for the primary prevention of dementia and
AD,
6
systematic reviews have noted the need to further
investigate secondary prevention.
7-9
A meta-analysis by
Bhome et al.
10
specically noted a lack of research evaluat-
ing lifestyle interventions in those with SCD, and previous
randomized controlled trials (RCTs) in this population have
been underpowered to detect the full spectrum of potential
outcomes included.
8
To date, more research has been con-
ducted on individuals with MCI than with SCD. A system-
atic review
11
of lifestyle-focused RCTs in the MCI
population found that all multidomain interventions evalu-
ated were associated with signicant improvements in at
least one cognitive domain, suggesting that these interven-
tions were more promising than interventions focusing on
single domains, such as physical activity or cognitive
engagement alone. The review noted heterogeneity in the
cognitive domains showing improvements and in interven-
tion and assessment methods, and it recommended further
well-designed and adequately powered RCTs.
Objectives
This proof-of-concept study adapts elements of previously
successful trials from other participant groups to the SCD
and MCI groups.
12-16
It is hypothesized that given individu-
alized support, people experiencing cognitive decline can
make meaningful lifestyle changes, and in this prodromal
stage of dementia the brain will still retain sufcient neuro-
plasticity to modify the trajectory of the disease. The aims
of the work were to evaluate the efcacy of the Body Brain
Life for Cognitive Decline (BBL-CD) study to reduce the
overall lifestyle risk of AD and other dementias and to pre-
vent further cognitive decline. Here we report on the pri-
mary outcomes of the BBL-CD study: lifestyle risk of AD
and cognition.
METHODS
Design
The full protocol of the study was published.
15
The study
was an 8-week two-arm single-blind RCT of a lifestyle
modication program to reduce dementia risk for people
experiencing cognitive decline. It was conducted in accor-
dance with the Consolidated Standards of Reporting Trials
(CONSORT) statement
17
and the CONSORT statement for
nonpharmacological interventions.
18
The trial was regis-
tered with the Australian and New Zealand Clinical Trial
Registry (ACTRN12617000792325), and ethical clearance
was provided by the ANU Human Research Ethic Commit-
tee (Protocol No. 2016/360). All participants gave written
informed consent to take part.
Participants
Participants were community-dwelling individuals from the
Canberra region, Australia, aged older than 65; owning a
computer with Internet access; having sufcient English
skills; willing to make lifestyle changes to improve health;
and having been diagnosed with MCI by a medical profes-
sional or reporting experiencing SCD. The Jessen criteria
were used for SCD: clinically normal on objective assess-
ment, self/informant-reported cognitive decline, and decline
not better accounted for by a major medical, neurological,
or psychiatric diagnosis.
15
Although potentially limiting in
this age group, a computer with Internet access was essen-
tial for certain intervention components; greater than 50%
of Australians aged 65 and older have access to the Inter-
net, and this gure has been increasing in recent years.
19,20
Exclusion criteria were any major neurological or psy-
chiatric disorder, or other chronic condition that would pre-
vent participation in a lifestyle behavior change program;
and currently participating in any other lifestyle change
interventions. No restrictions were placed on current levels
of or adherence to Mediterranean diet, physical activity, or
cognitive engagement.
Interventions
The active control group completed four online informa-
tional modules to reduce dementia risk. The modules cov-
ered dementia literacy and lifestyle risk, Mediterranean diet,
physical activity, and cognitive engagement. Following each
module, participants were given a week with no education
to allow them to implement the information into their own
lifestyle.
The intervention group completed the same online edu-
cational modules, but in the weeks between undertaking the
modules, the intervention group took part in practical activ-
ities including meeting with a dietitian and exercise physiol-
ogist and completing brain training. These practical
components were designed to assist the participants to
implement the information from the modules more effec-
tively into their lifestyle. Participants had an initial 1-hour
appointment with a dietitian (week 3) and two follow-up
30-minute appointments (weeks 10 and 21) to assist with
adhering to a Mediterranean diet; an initial 1-hour appoint-
ment with an exercise physiologist (week 7) to formulate an
exercise plan, and two follow-up 30-minute appointments
(weeks 10 and 21) to modify as required; and 2 hours
weekly of online brain training (beginning week 5) on the
Brain HQ platform.
21
Further details of the intervention are
provided in the published protocol.
15
However, there was
one deviation from this protocol. The exercise physiologist
was hospitalized on two occasions during the study, and no
suitable replacement could be identied; therefore the two
follow-up appointments were not conducted.
Outcomes
Lifestyle Risk of Alzheimers Disease
Lifestyle risk for AD was assessed using the Australian
National University-Alzheimers Disease Risk Index (ANU-
ADRI). The ANU-ADRI yields scores for 11 lifestyle risk
2630 MCMASTER ET AL. NOVEMBER 2020-VOL. 68, NO. 11 JAGS
factors (age [moderated by sex], low educational attain-
ment, body mass index, diabetes mellitus type II, depres-
sion, cholesterol, traumatic brain injuries, smoking status,
low social engagement, and exposure to pesticides), four
protective factors (alcohol intake, physical activity, cogni-
tive engagement, and sh intake), or an overall score com-
bining both factors.
22
For the ANU-ADRI, lower scores
indicate lower lifestyle risk for all three measures. Partici-
pants completed the ANU-ADRI via computer at the
research teamsofce at baseline (week 0), immediately fol-
lowing the intervention (week 9), at 3-month follow-up
(week 20), and at 6-month follow-up (week 32).
The largest score component of the ANU-ADRI is age
(041 points). To assess the effect of the intervention
adequately, ANU-ADRI scores were calculated at all time
points from the participantsage at baseline. This prevented
any scores from increasing as a result of participants mov-
ing to a higher risk bracket of age, hence obscuring changes
due to lifestyle alterations.
Cognition
The cognitive outcomes for the study were the measures
comprising the Alzheimers Disease Assessment Scale-Cog-
nitive Plus (ADAS-Cog Plus)
23
: the ADAS-Cog 11,
24
Pfeffer
Functional Activities Questionnaire (PFAQ),
25
Trail Mak-
ing Test-B (TMT-B),
26
Symbol Digit Modalities Test
(SDMT),
27
and Category Fluency for vegetables.
28
The
Contacted Research Team
(n = 199)
Baseline Testing
(n = 123)
Enrolled
(n = 135)
Not enrolled (n = 64)
Ineligible (n = 21)
Declined (n = 43)
Incomplete Baseline Testing
(n = 4)
Withdrew (n = 12)
Availability (n = 7)
Health (n = 5)
Withdrawals (n = 8)
Lost to follow-up (n = 5)
Availability (n = 2)
Group allocation (n = 1)
Withdrawals (n = 10)
Availability (n = 6)
Health (n = 3)
Group allocation (n = 1)
Withdrawals (n = 0) Withdrawals (n = 4)
Lost to follow-up (n = 2)
AD diagnosis (n = 1)
Availability (n = 1)
Intervention
(n = 49)
Withdrawals (n = 1)
AD diagnosis (n = 1)
Withdrawals (n = 0)
Immediate Follow-up
(Week 9)
Three-month Follow-up
(week 20)
Six-month Follow-up
(week 32)
Randomization
Control
(n = 52)
Control
(n = 62)
Intervention
(n = 57)
Intervention
(n = 49)
Control
(n = 48)
Control
(n = 48)
Intervention
(n = 48)
Figure 1. Participant owchart for BBL-CD study. Lost to follow-up: These participants could not be contacted/did not respond.
Availability: These participants formally withdrew due to other commitments. Group allocation: These participants formally with-
drew due to the group they were randomized to. AD diagnosis: These participants were diagnosed with Alzheimers disease. [Color
gure can be viewed at wileyonlinelibrary.com]
JAGS NOVEMBER 2020-VOL. 68, NO. 11 BBL-CD: A LIFESTYLE AD RISK REDUCTION RCT 2631
ADAS-Cog Plus measures were selected because they are
sensitive to the decits seen in early stages of cognitive
decline.
23
All cognitive measures were assessed face to face
at baseline (week 0), 3-month follow-up (week 20), and
6-month follow-up (week 32). Further details on the out-
comes can be found in the protocol article.
15
These cognitive measures were combined into a single
composite score by conversion to zscores. These were
calculated based on the baseline mean and standard devia-
tion (SD) of each measure. The zscores were then averaged
across the measures for each participant at each time point.
For the ADAS-Cog, PFAQ, and TMT-B, lower scores indi-
cate better cognitive function; for calculating the composite
score, these were reversed so that increases indicated better
cognitive function for all measures. This composite zscore
as well as the results from the individual cognitive measures
are reported.
Blinding
All testing was carried out by researchers who were blind
to group allocation. Due to the nature of the intervention,
blinding of the participants was not possible. Participants
were asked not to discuss the intervention with any
researchers; if they had any questions, they could discuss
these with the project manager in private.
Statistical Methods
At the nal follow-up, a minimum sample size of 36 partici-
pants per arm was required to detect a difference between
groups of .70 SDs in the primary outcome measures.
Accounting for potential attrition of 10% per follow-up
period gave a baseline target sample size of 120 participants.
All participants were randomized to either the intervention
or control group in a 1:1 ratio, within strata dened by sex,
baseline cognition (above or below median ADAS-Cog
11 score), and baseline lifestyle risk of AD (above or below
median ANU-ADRI score) in permuted blocks of eight. The
randomization sequence was generated by an independent
researcher (R.B.) from www.sealedenvelope.com.
Linear mixed models were used to compare outcomes
between groups at each follow-up time. Each model
included group, time point, and the group ×time point
interaction, as well as stratication variables: sex, ANU-
ADRI strata, and ADAS-Cog 11 strata. The likelihood ratio
test (LRT) was used to assess statistical signicance of the
Figure 2. Scores for Primary Outcomes: Lifestyle risk and Cognition. Adjusted outcomes for Australian National University-
Alzheimers Disease Risk Index (ANU-ADRI) (A) Group ×time interaction: χ
2
=10.84;df =3;P= .012 and a cognitive composite z
score (B) group ×time interaction: χ
2
=7.28;df =2;P= .026. All data points are least square means generated from regression
models after adjusting for strata variables that were not the dependent variable (eg, ANU-ADRI adjusted for sex and cognition strata
and vice versa). Lower ANU-ADRI scores indicate lower lifestyle risk. Higher cognitive composite scores indicate better cognitive
function. Blue dashed lines represent the intervention group, solid red lines represent the control group, and error bars represent 95%
condence intervals. Between-group signicance denoted by *P< .05 and **P<.01.
Table 1. Baseline Characteristics and Primary Outcome
Measures of the Two Groups
Intervention
(n = 57)
Control
(n = 62)
Age, y 72.8 (5.3) 73.3 (5.8)
Female (%) 35 (61.4) 38 (61.3)
Education, y 12.4 (5.3) 14.0 (5.9)
ANU-ADRI total, score
range = 14 to 73
8.3 (10.8) 10.3 (11.6)
Protective factors, score
range = 0 to 14
9.1 (4.2) 8.9 (4.5)
Risk factors, score range = 0 to
73
17.4 (9.0) 19.2 (10.0)
Cognitive composite zscore .091 (.67) .095 (.56)
ADAS-Cog 11, score range = 0
to 70
7.5 (3.6) 7.0 (3.5)
PFAQ, score range = 0 to 15 .8 (1.6) .8 (1.5)
SDMT, score range = 0 to 110 42.1 (9.8) 41.7 (9.2)
TMT-B, s 97.3 (33.2) 99.3 (41.7)
Category uency, score
range = 0 to
14.7 (4.2) 14.5 (4.4)
Note: Cognitive composite zscores were created using baseline means and
SDs for all participants, then averaging across these zscores to form a com-
posite. Participants with missing data on one or more cognitive measures
were not included in the zscore average.
Abbreviations: ADAS-Cog, Alzheimers Disease Assessment Scale-Cognitive
Subscale; ANU-ADRI, Australian National University-Alzheimers Disease
Risk Index; PFAQ, Pfeffer Functional Activities Scale; SDMT, Symbol Digit
Modalities Test; TMT-B, Trail Making Task-B.
2632 MCMASTER ET AL. NOVEMBER 2020-VOL. 68, NO. 11 JAGS
main effects and interaction term.
29
A statistically signi-
cant interaction term indicated that the between-group dif-
ferences changed over time. Difference in least square mean
outcomes between intervention groups is reported at each
follow-up, with 95% condence intervals (CIs) and
Pvalues from between-group ttests. A secondary analysis
was undertaken to examine the effect of the intervention on
the two subcomponents of the ANU-ADRI, risk and protec-
tive factors, and each of the ve cognitive measures.
The primary analysis was a complete case analysis.
Sensitivity analysis was undertaken as a full intention-to-
treat (ITT) analysis with missing data accounted for using
multivariate imputation by chained equations (MICE).
All preliminary analyses and descriptions of baseline
characteristics were carried out with SPSS v.26.0.
30
Linear
mixed modeling was undertaken in R v.3.6.0
31
using the
lme4,
32
lmerTest,
33
emmeans,
34
and MICE packages,
35
with
graphs creating using ggplot2.
36
RESULTS
Participant Characteristics
Recruitment of participants took place between July 2017
and November 2017; 199 individuals were screened, and
135 were recruited into the study. Of these, 119 participants
completed baseline testing (January 2018) and were ran-
domized into the intervention (n = 57) or control (n = 62)
groups. At the nal 6-month follow-up (November 2018),
48 participants remained in each group of the study. The
full owchart of participants can be found in Figure 1.
At baseline, participants had a mean age of 73.0 years
(SD = 5.5 years) and had 13.3 years (SD = 5.7 years) of
education, and 61% (n = 73) were female. Three (3%) par-
ticipants had a diagnosis of MCI, and all other participants
(n = 116 [97%]) met the criteria for SCD. Baseline charac-
teristics for both groups are shown in Table 1.
Mean differences with 95% CIs and signicance levels
for between-group differences for all variables, at all
follow-up time points, for the primary and secondary ana-
lyses can be found in Supplementary Table S1.
Overall, participants were well able to adhere with
most of the intervention requirements. All participants from
both groups who remained in the intervention until the nal
follow-up completed the four educational modules. For the
intervention group there was mostly strong adherence to
the active interventions. All participants who remained in
the intervention attended the three dietitian appointments
and the exercise physiologist appointment. However, adher-
ence to the brain training component was lower, with 20%
adherence (10.8 hours) of the specied 54 total hours
(27 weeks ×2 hours/week).
Primary Analyses
Lifestyle Risk of Alzheimers Disease (ANU-ADRI)
The LRT analysis showed a signicant group ×time point
interaction (χ
2
=10.84;df =3;P= .013). The between-group
difference was not signicant at immediate follow-up (T2
intervention = 9.80; control = 11.98; difference = 2.18;
t=1.28; P= .204), it was signicant at the 3-month
follow-up (T3 intervention = 7.27; control = 11.35; differ-
ence = 4.08; t=2.36; P= .019), and it was no longer sig-
nicant at the 6-month follow-up (T4 intervention = 7.48;
control = 10.37; difference = 2.88; t=1.66; P= .098)
(Figure 2A). When looking at within-group changes over the
course of the intervention, the control group showed only a
minor reduction in ANU-ADRI scores (0.54) compared
with the larger reduction seen in the intervention group
(2.46); reductions of 2.0 or more ANU-ADRI points are
considered clinically meaningful.
Cognitive Composite zScore
The LRT analyses found a signicant group ×time point
interaction (χ
2
= 7.28; df =2;P= .026). For the between-
group differences, the intervention group had signicantly
higher cognition scores at both follow-up periods (T3 inter-
vention = .159; control = .117; difference = .276; t= 2.62;
P= .010; T4 intervention = .231; control = .014; differ-
ence = .245; t= 2.33; P= .021) (Figure 2B).
Figure 3. Australian National University-Alzheimers Disease Risk Index (ANU-ADRI) risk and protective factors. Adjusted out-
comes for risk factors (A) group ×time point interaction: χ
2
= 2.93; df =3;P= .403 and protective factors (B) group ×time point
interaction: χ
2
= 12.02; df =3;P= .007. All data points are least square means generated from regression models after adjusting
for strata variables (sex and cognition). Lower scores indicate lower lifestyle risk. Blue dashed lines represent the intervention
group, solid red lines represent the control group, and error bars represent 95% condence intervals. Between-group signicance is
denoted by *P< .05 and ***P< .001. [Color gure can be viewed at wileyonlinelibrary.com]
JAGS NOVEMBER 2020-VOL. 68, NO. 11 BBL-CD: A LIFESTYLE AD RISK REDUCTION RCT 2633
Secondary Analyses: Drivers of Signicant Change
Lifestyle Risk and Protective Factor for Alzheimers
Disease (ANU-ADRI)
The outcomes for the ANU-ADRI risk and protective fac-
tors are shown in Figure 3.
Lifestyle Risk Factors for Alzheimers Disease
The LRT analysis found no signicant effects (χ
2
= 2.93;
df =3;P= .403). Although both groups experienced a
within-group reduction of risk factor scores, neither of these
was clinically meaningful or statistically signicant (T2
intervention = 19.3; control = 19.4; difference = .092;
t=.07;P= .945; T3 intervention = 18.5; control = 19.5;
difference = .962; t= .72; P= .472; T4 intervention = 18.6;
control = 18.9; difference = .339; t=.28;P= .783).
Lifestyle Protective Factors for Alzheimers Disease
The LRT analysis showed a statistically signicant group ×
time point interaction (χ
2
= 12.02; df =3;P= .007). The
intervention groups protective scores were signicantly
lower than the control scores at all follow-up periods (T2
intervention = 9.38; control = 7.77; difference = 1.62;
Figure 4. Outcomes for Measures of Cognition. Alzheimers Disease Assessment Scale-Cognitive Subscale (ADAS-Cog 11) (A)
Group ×time point interaction: χ
2
= .73; df =2;P= .696; Pfeffer Functional Activities Questionnaire (PFAQ) (B) χ
2
= .44; df =2;
P= .802; Trail Making Test-B (TMT-B) (C) χ
2
= .94, df =2;P= .625; Symbol Digit Modalities Test (SDMT) (D) χ
2
= 6.459; df =
2; P= .040; Category Fluency (E) χ
2
= 2.04; df =2;P= .360. All data points are least square means generated from regression
models after adjusting for strata variables (sex and Australian National University-Alzheimers Disease Risk Index [ANU-ADRI]).
Lower scores for ADAS-Cog 11, PFAQ, and TMT-B and higher scores for SDMT and Category Fluency indicate better cognitive
function. Blue dashed lines represent the intervention group, solid red lines represent the control group, and error bars represent
95% condence intervals. [Color gure can be viewed at wileyonlinelibrary.com]
2634 MCMASTER ET AL. NOVEMBER 2020-VOL. 68, NO. 11 JAGS
t= 2.10; P= .037; T3 intervention = 11.21; con-
trol = 8.54; difference = 2.67; t= 3.40; P< .001; T4 inter-
vention = 11.05; control = 9.05; difference = 2.00;
t= 2.51; P= .012).
Cognitive Measures
The outcomes for the individual cognitive measures that
made up the cognitive composite zscore are shown in
Figure 4. The measures were ADAS-Cog (Figure 4A), PFAQ
(Figure 4B), TMT-B (Figure 4C), SDMT (Figure 4D), and
Category Fluency (Figure 4E).
When all cognitive measures were analyzed separately
using the LRT analysis, only SDMT showed a signicant
group ×time point interaction (χ
2
= 6.46; df =2;P= .040);
however, neither of the between-group differences was sig-
nicant at follow-up (T3 intervention = 42.4; control = 39.4;
difference = 2.96; t=1.77; P= .078; T4 interven-
tion = 43.6; control = 40.6; difference = 3.29;
t=1.96; P= .052).
Sensitivity Analysis: Intention-to-Treat Analysis
In ITT, which used complete cases following missing data
imputation, outcomes were highly consistent with the pri-
mary and secondary analyses. All between-group differ-
ences at specic time points were retained.
A further analysis was undertaken to examine the impact
that attrition may have had on outcomes. Supplementary
Table S2 shows that only one variable, protective lifestyle fac-
tors, showed a signicant difference for those who withdrew
and those who remained in the study. Although there was a
signicant difference, when this variable was further separated
by intervention group, it showed the signicant difference was
only in the control group (withdrew = 6.19; remained = 9.80;
difference = 3.62; t= 2.91; P= .005), not the intervention
group (withdrew = 8.64; remained = 9.17; difference = .54;
t= .38; P= .704).
DISCUSSION
BBL-CD was a proof-of-concept RCT that adapted a suc-
cessful primary prevention study
12
to the cognitive decline
group and included new components adapted from previ-
ously successful interventions conducted in other partici-
pant groups.
12-16
The main ndings from this study were
that a multidomain lifestyle intervention was able to
decrease exposure to lifestyle risk factors for AD signi-
cantly, and improve cognition in a group experiencing cog-
nitive decline signicantly, relative to a control group. The
results lend support to the hypothesis that secondary pre-
vention interventions may be able to modify the course of
disease progression.
Adherence
The intervention mostly achieved strong adherence. The
lowest levels of adherence were for the brain training com-
ponent. The lower levels of adherence may have been due
to such a large dose of brain training (54 hours) over such
a long period (27 weeks). A meta-analysis of brain training
in the MCI population showed the average dose across the
studies included was 34.2 hours over an average of
14 weeks (according to the study protocols); however, the
meta-analysis provided no information on actual adherence
to the interventions.
37
The Advanced Cognitive Training
for Independent and Vital Elderly (ACTIVE) trial, which
administered 10 to 12.5 hours of speed of processing train-
ing over 5 to 6 weeks, resulted in lower rates of dementia in
a cognitively normal sample of participants aged 65 and
older at 10 years postintervention.
38
This dose was compa-
rable with the actual dose achieved in BBL-CD, albeit over
a longer time frame. Unfortunately, in BBL-CD no qualita-
tive data were collected to determine what the barriers to
higher levels of adherence may have been.
Lifestyle Risk of Alzheimers Disease
By the 3-month follow-up, the intervention group showed a
signicantly lower ANU-ADRI than the control group, but
this difference was not retained at the nal follow-up. The
decline in overall ANU-ADRI score for the intervention
group was 2.7 (T3) and 2.5 (T4) points; a 2-point change
in ANU-ADRI is considered to be clinically meaningful.
39
This demonstrates that clinically relevant lifestyle changes
are feasible over the short term in participants experiencing
cognitive decline. The signicant effects in overall ANU-
ADRI scores were driven by higher levels of protective fac-
tors, rather than lower levels of risk factors. Similar effects
were seen in past multidomain lifestyle interventions.
12,40
The reduction in ANU-ADRI score through increased pro-
tective factors was 2.5 (T3) and 2.3 (T4). In real terms these
changes are similar to the amount of AD risk conferred
between low (0 points) and moderate levels (2 points)
of exercise or the presence of diabetes mellitus type II (+3
points).
39
When missing data were controlled for in the
ITT, all the between-group differences found in previous
analyses were still present, showing that these are robust
ndings. The only scores that may have been affected by
participants withdrawing were protective factors for the
control group. Because the control participants who with-
drew had higher protective scores (ie, less protective), this
may have articially lowered the control group scores.
Given that the only signicant effects were for the interven-
tion group, if anything this articial reduction may have
reduced the magnitude of difference between the control
and intervention groups. This has minimal impact on the
main ndings of this study.
Cognition
At the end of the study, the intervention group had a signi-
cantly higher cognitive composite score, and a signicant
group ×time point interaction effect was observed for both
follow-up periods. When this was investigated further to
determine the specic measures underlying the signicant
effects, only SDMT showed a signicant group ×time point
interaction; however, there were no between-group differ-
ences at specic time points. The most likely explanation
for this is a weak but consistent positive effect across mea-
sures. Greater statistical power through a larger sample size
would be required to assess actual effects on individual cog-
nitive measures. There appeared to be improvements over
time for all cognitive measures in the intervention group,
JAGS NOVEMBER 2020-VOL. 68, NO. 11 BBL-CD: A LIFESTYLE AD RISK REDUCTION RCT 2635
but only the ADAS-Cog and Category Fluency measures
showed improvement for the control group.
In the limited number of multidomain SCD interven-
tions that have taken place, signicant effects have been
found for executive function (color-word Stroop task)
40
and verbal uency (letter uency)
41
but not in cognitive
composite scores. MCI lifestyle interventions show a very
mixed pattern of outcomes with positive and null effects
across executive function, memory, and global cognition.
11
Other multidomain lifestyle interventions in different partic-
ipant groups have found signicant differences in cognitive
composite scores of a similar magnitude (eg, BBL-CD inter-
vention group, z= .25, vs Finnish Geriatric Intervention
Study to Prevent Cognitive Impairment and Disability [FIN-
GER] trial intervention group, z= .20).
6
Importantly, in the
present study, the ITT analysis produced results that were
highly consistent with the primary and secondary analyses,
which is suggestive that signicant effects were not statisti-
cal artifacts due to participant attrition.
Taken together, the intervention group experienced a
statistically signicant and clinically meaningful lower life-
style risk and signicantly higher cognition, relative to the
control group. No signicant differences in lifestyle risk or
cognition were found for the control group. These results
support the hypothesis that individuals in the early stages of
cognitive decline retain sufcient neuroplasticity to achieve
cognitive improvements in the short term. The ultimate goal
of research in this participant group is to demonstrate long-
term improvements in lifestyle risk, cognition, and ultimately
slower rates of cognitive decline and lower rates of conver-
sion to dementia. The outcomes achieved in this study war-
rant the conduct of a larger and longer study to test the
longer term sustainability of improvements in lifestyle and
cognition.
Limitations
The main limitation of this study was the limited follow-up
time of 6 months. Meta-analyses of nonpharmacological
interventions for SCD recommend a minimum follow-up
duration of a year or longer.
8
A second limitation was that
the study overestimated the magnitude of change that
would be observed (.70 SD); hence there may have been
insufcient power to detect small differences.
15
Both limita-
tions can be overcome in future studies.
Implications for Future Research
The pattern of results for lifestyle outcomes illustrate a few
potential areas for renements for this intervention, as well
as other studies in this area. First, lifestyle interventions
commonly report improvement in protective factors, but
signicant reductions in risk factors is an area where clear
improvements are possible. For this reason, it would be
benecial for future research to look at the outcomes of
protective and risk factors separately.
Second, a plateauing of improvement between time
points 3 and 4 for ANU-ADRI (a period where no further
intervention was being implemented) is suggestive of the
need for booster sessionsto maximize and sustain life-
style improvements.
In terms of cognition, future interventions in this area
do need to account for small effect sizes of cognitive out-
comes with adequate sample sizes, so as not to negatively
bias evidence in this developing area. Given the heterogene-
ity of cognitive domains that can be improved through non-
pharmacological interventions, future research should
choose a battery of cognitive tests to cover all potential
domains and also standardize and combine outcomes to
detect any subtle but consistent effects across measures.
More emphasis on measures of everyday function would
also show whether interventions are having an immediate
effect on real-world outcomes.
Given there is some heterogeneity in the SCD and MCI
groups in terms of stability, progression, and remission of
decits, further characterization of participants through
genotyping, neuroimaging, and other biomarkers would be
benecial for any future research.
In conclusion, the present proof-of-concept study
adds evidence to the argument that modifying the life-
style risk of those experiencing cognitive decline can
result in improvements in cognition. The results
obtained are supportive of a larger, longer trial to inves-
tigate the possibilities of sustained improvements in life-
style and cognition, clearly demonstrate cognitive
domains showing improvements, and long-term follow-
up with participants to track cognitive decline and
development of AD and other forms of dementia for a
number of years postintervention.
ACKNOWLEDGMENTS
We thank Richard Burns (R.B.) for his advice and assis-
tance with the randomization of the sample and Ranmalee
Eramudugolla for her advice on clinical diagnoses and
selection criteria. Mitchell McMasters PhD scholarship was
supported by Dementia Australia Research Foundation,
the Australian National University, and Dementia Collabo-
rative Research Centre. Additional research funding was
provided by the NHMRC Centre for Research Excellence
in Cognitive Health, the Australian National University;
Neuroscience Research Australia, University of New South
Wales; the 2017 Royal Commonwealth Society Phyllis
Montgomery Award; NHMRC Fellowship Funds (Kaarin
J. Anstey: APP1102694); and the Dementia Collaborative
Research Centre (funds for precursor study).
Conict of Interest: The authors have declared no conicts
of interest for this article.
Author Contributions: Study concept and design:
McMaster, Kim, Clare, Torres, DEste, and Anstey. Acqui-
sition of data: McMaster. Analysis and interpretation of
data: McMaster, Cherbuin, and DEste. Drafting of the
manuscript: McMaster. Critical revision of the manuscript
for important intellectual content: All authors.
Sponsors Role: None of the funding bodies played any
part in the study design, data collection, analysis, interpreta-
tion, writing, or decision to submit this article for publication.
REFERENCES
1. World Health Organization. Risk Reduction of Cognitive Decline and
Dementia: WHO Guidelines. Geneva, Switzerland: World Health Organiza-
tion; 2019.
2636 MCMASTER ET AL. NOVEMBER 2020-VOL. 68, NO. 11 JAGS
2. Livingstone G, Sommerlad A, Ortega V, et al. Dementia prevention, interven-
tion, and care. Lancet. 2017;390:2673-2734.
3. Barnes DE, Yaffe K. The projected effect of risk factor reduction on
Alzheimers disease prevalence. Lancet Neurol. 2011;10(9):819-828.
4. Fratiglioni L, Winblad B, von Strauss E. Prevention of Alzheimerʼs disease
and dementia. Major ndings from the Kungsholmen Project. Physiol Behav.
2007;92(1):98-104.
5. Shokouhi S, Albert K. Subjective cognitive decline and biomarkers of preclin-
ical Alzheimerʼs disease. Curr Behav Neurosci Rep. 2019;6(4):219-226.
6. Ngandu T, Lehtisalo J, Solomon A, et al. A 2 year multidomain intervention
of diet, exercise, cognitive training, and vascular risk monitoring versus con-
trol to prevent cognitive decline in at-risk elderly people (FINGER): a
randomised controlled trial. Lancet. 2015;385(9984):2255-2263.
7. Horr T, Messinger-Rapport B, Pillai JA. Systematic review of strengths and
limitations of randomized controlled trials for non-pharmacological interven-
tions in mild cognitive impairment: focus on Alzheimerʼs disease. J Nutr
Health Aging. 2015;19(2):141-153.
8. Smart CM, Karr JE, Areshenkoff CN, et al. Non-pharmacologic interven-
tions for older adults with subjective cognitive decline: systematic review,
meta-analysis, and preliminary recommendations. Neuropsychol Rev. 2017;
27(3):245-257.
9. Williams JW, Plassman BL, Burke J, Holsinger T, Benjamin S. Prevention
Alzheimerʼs Disease and Cognitive Decline. Rockville, MD: Agency for
Healthcare Research and Quality; 2010.
10. Bhome R, Berry AJ, Huntley JD, Howard RJ. Interventions for subjective
cognitive decline: systematic review and meta-analysis. BMJ Open. 2018;8
(7):e021610.
11. Huckans M, Hutson L, Twamley E, Jak A, Kaye J, Storzbach D. Efcacy of
cognitive rehabilitation therapies for mild cognitive impairment (MCI) in
older adults: working toward a theoretical model and evidence-based inter-
ventions. Neuropsychol Rev. 2013;23(1):63-80.
12. Anstey KJ, Bahar-Fuchs A, Herath P, et al. Body brain life: a randomized con-
trolled trial of an online dementia risk reduction intervention in middle-aged
adults at risk of Alzheimers disease. Alzheimers Dement (N Y). 2015;1(1):72-80.
13. Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular
disease with a Mediterranean diet. N Engl J Med. 2013;368(14):1279-1290.
14. Kim S, McMaster M, Torres S, et al. Protocol for a pragmatic randomised
controlled trial of body brain lifegeneral practice and a lifestyle modica-
tion programme to decrease dementia risk exposure in a primary care set-
ting. BMJ Open. 2018;8(3):e019329.
15. McMaster M, Kim S, Clare L, Torres SJ, DʼEste C, Anstey KJ. Body, brain,
life for cognitive decline (BBL-CD): protocol for a multidomain dementia
risk reduction randomized controlled trial for subjective cognitive decline
and mild cognitive impairment. Clin Interv Aging. 2018;13:2397-2406.
16. Rebok GW, Ball K, Guey LT, et al. Ten-year effects of the ACTIVE cognitive
training trial on cognition and everyday functioning in older adults. J Am
Geriatr Soc. 2014;62(1):16-24.
17. Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated
guidelines for reporting parallel group randomised trials. BMC Med. 2010;8
(18):1-9.
18. Boutron I, Moher D, Altman DG, Schulz KF, Ravaud P. Methods and pro-
cesses of the CONSORT group: example of an extension for trials assessing
nonpharmacologic treatments. Ann Intern Med. 2008;148(4):W-60-W-66.
19. Australian Bureau of Statistics. Household Use of Information Technology,
Australia, 201617 (#8146.0). https://www.abs.gov.au/ausstats/abs@.nsf/mf/
8146.0. Published 2017. Accessed June 11, 2020.
20. McCallum, J., Rees, K. & Maccora, J. (2017) Bridging the Senior Digital
Divide. Brisbane, Australia: National Seniors Australia. https://
nationalseniors.com.au/uploads/12172826PAR_BridgingTheDigitalDivide_
FNWeb.pdf. Accessed June 11, 2020.
21. Posit Sceince. BrainHQ. http://www.brainhq.com. Accessed May 4, 2020.
22. Anstey KJ, Cherbuin N, Herath PM. Development of a new method for
assessing global risk of Alzheimerʼs disease for use in population health
approaches to prevention. Prev Sci. 2013;14(4):411-421.
23. Skinner J, Carvalho JO, Potter GG, et al. The Alzheimerʼs Disease Assessment
Scale-Cognitive-Plus (ADAS-Cog-Plus): an expansion of the ADAS-Cog to
improve responsiveness in MCI. Brain Imaging Behav. 2012;6(4):489-501.
24. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimerʼs disease.
Am J Psychiatry. 1984;141(11):1356-1364.
25. Pfeffer R, Kurosaki T, Harrah C Jr, Chance J, Filos S. Measurement of func-
tional activities in older adults in the community. J Gerontol. 1982;37(3):
323-329.
26. Reitan RM. Trail Making Test: Manual for Administration and Scoring.
Reitan Neuropsychology Laboratory; 1992.
27. Smith A. Symbol digit modalities test. Los Angeles, CA: Western Psychologi-
cal Services; 1982.
28. Lucas JA, Ivnik RJ, Smith GE, et al. Mayoʼs older Americans normative stud-
ies: category uency norms. J Clin Exp Neuropsychol. 1998;20(2):194-200.
29. Winter B. Linear models and linear mixed effects models in R with lingui-
stic applications [arXiv:13085499]. https://arxivorg/ftp/arxiv/papers/1308/
13085499pdf.
30. IBM Corp. IBM SPSS Statistics for Windows, Version 26.0. Armonk, NY:
IBM Corp.; 2019.
31. R Core Team. R: A Language and Environment for Statistical Computing.
Vienna, Austria: R Foundation for Statistical Computing; 2017.
32. Bates D, Maechler M, Bolker B, Walker S. Fitting linear mixed-effects
models using lme4. J Stat Softw. 2015;67(1):1-48.
33. Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest package: tests in
linear mixed effects models. J Stat Softw. 2017;82(13):1-26.
34. Estimated Marginal Means, emmeansR package [computer program].
https://mran.microsoft.com/snapshot/2017-12-11/web/packages/emmeans/
emmeans.pdf. Accessed April 6, 2020.
35. van Buuren S, Groothuis-Oudshoorn K. Mice: multivariate imputation by
chained equations in R. J Stat Softw. 2011;45(3):1-67.
36. Wickham H. ggplot2: Elegant Graphics for Data Analysis. New York, NY:
Springer; 2016.
37. Hill NT, Mowszowski L, Naismith SL, Chadwick VL, Valenzuela M,
Lampit A. Computerized cognitive training in older adults with mild cogni-
tive impairment or dementia: a systematic review and meta-analysis.
Am J Psychiatry. 2017;174(4):329-340.
38. Edwards JD, Xu H, Clark DO, Guey LT, Ross LA, Unverzagt FW. Speed of
processing training results in lower risk of dementia. Alzheimers Dement
(N Y). 2017;3(4):603-611.
39. Anstey KJ, Cherbuin N, Herath PM, et al. A self-report risk index to predict
occurrence of dementia in three independent cohorts of older adults: the
ANU-ADRI. PLoS One. 2014;9(1):e86141.
40. Park J, Jeon S, Kim S, et al. A multidomain intervention for modifying lifestyle
habits reduces the dementia risk in community-dwelling older adults: a single-
blinded randomized controlled pilot study. J Alzheimer Dis. 2019;70(1):51-60.
41. Small GW, Silverman DH, Siddarth P, et al. Effects of a 14-day healthy lon-
gevity lifestyle program on cognition and brain function. Am J Geriatr Psy-
chiatry. 2006;14(6):538-545.
SUPPORTING INFORMATION
Additional Supporting Information may be found in the
online version of this article.
Supplementary Table S1: Least square differences in
means adjusted for strata variables that were not the depen-
dent variable (eg, ANU-ADRI adjusted for sex and cognition
strata and vice versa). The Pvalues are for ttests for between-
group differences. Lower scores for ANU-ADRI, protective
factors, and risk factors all indicate lower levels of lifestyle
risk. Lower scores for ADAS-Cog 11, PFAQ, and TMT-B,
and higher scores for SDMT and Category Fluency indicate
better cognitive function. Values shown in bold are signicant
at P< .05. ANU-ADRI, Australian National University-
Alzheimers Disease Risk Index; ADAS-Cog, AlzheimersDis-
ease Assessment Scale- Cognitive Subscale; PFAQ, Pfeffer
Functional Assessment Questionnaire; TMT-B, Trail Making
Test-B; SDMT, Symbol Digit Modality Test.
Supplementary Table S2: The Pvalues are for ttests for
between-group differences. Lower scores for ANU-ADRI, pro-
tective factors, and risk factors all indicate lower levels of life-
style risk. Lower scores for ADAS-Cog 11, PFAQ, and
TMT-B, and higher scores for SDMT and Category Fluency
indicate better cognitive function. Values shown in bold are
signicant at P< .05. ANU-ADRI, Australian National Uni-
versity-Alzheimers Disease Risk Index; ADAS-Cog,
Alzheimers Disease Assessment Scale-Cognitive Subscale;
PFAQ, Pfeffer Functional Assessment Questionnaire; TMT-B,
Trail Making Test-B; SDMT, Symbol Digit Modalities Test.
JAGS NOVEMBER 2020-VOL. 68, NO. 11 BBL-CD: A LIFESTYLE AD RISK REDUCTION RCT 2637
... Since cognitive decline has multiple causes, multidomain interventions which target several modifiable factors simultaneously are considered appropriate for addressing concurrent age-related deterioration in mental and physical functions. Up to the present, there were several randomized controlled trials (RCT) devoted to determining the effectiveness of multidomain intervention on improving cognition or reducing dementia incidence in older adults with SCD [6][7][8] or those at risk of cognitive decline [9,10]. Although some of the trials failed to find significant effects on cognitive decline [7,8], there were still many studies showing that the combination of different components could significantly enhance cognitive performance in older adults with SCD. ...
... A recent RCT study also showed that immediately after a 9-month multidomain intervention of meditation, physical exercise, cognitive training, and nutrition counseling, cognitive performance was significantly enhanced in community-dwelling older adults who were at risk of cognitive decline [11]. Several studies employing comparable multidomain intervention approaches but over a shorter training duration (e.g., 2 to 6 months) have also yielded promising outcomes in older adults with SCD [9,12]. Upon reviewing these studies, neuropsychological assessments served as the primary outcomes. ...
... (4) Go-Nogo task: This task was designed to assess the ability of response inhibition. During the ERP recordings, the subjects were instructed to press a response button when they saw Go trials (i.e., 1,2,4,5,6,8,9), and withhold their responses to Nogo trials (i.e., 3, 7). The task consisted of about 85% Go trials and about 15% Nogo trials with ISIs between 1400-2200 ms. ...
Article
Full-text available
Background: Multidomain intervention may delay or ameliorate cognitive decline in older adults at risk of Alzheimer’s disease, particularly in the memory and inhibitory functions. However, no study systematically investigates the changes of brain function in cognitively-normal elderly with subjective cognitive decline (SCD) when they receive multidomain intervention. Objective: We aimed to examine whether a multidomain intervention could improve neuropsychological function and neurophysiological activities related to memory and inhibitory function in SCD subjects. Methods: Eight clusters with a total of 50 community-dwelling SCD older adults were single-blind, randomized into intervention group, which received physical and cognitive training, or control group, which received treatment as usual. For the neuropsychological function, a composite Z score from six cognitive tests was calculated and compared between two groups. For the neurophysiological activities, event-related potentials (ERPs) of memory function, including mismatch negativity (MMN) and memory-P3, as well as ERPs of inhibitory function, including sensory gating (SG) and inhibition-P3, were measured. Assessments were performed at baseline (T1), end of the intervention (T2), and 6 months after T2 (T3). Results: For the neuropsychological function, the effect was not observed after the intervention. For the neurophysiological activities, improved MMN responses of ΔT2–T1 were observed in the intervention group versus the control group. The multidomain intervention produced a sustained effect on memory-P3 latencies of ΔT3–T1. However, there were no significant differences in changes of SG and inhibition-P3 between intervention and control groups. Conclusions: While not impactful on neuropsychological function, multidomain intervention enhances specific neurophysiological activities associated with memory function.
... These results helped to pave the way for remote delivery to be considered an equally viable option within this area of health promotion. Currently, we see digital components employed in a variety of ways, including distributing educational materials through online modules (e.g., [12][13][14]), offering computerbased cognitive training (e.g., [12,13,15,16]), incorporating coach-led advice via videoconferencing [17], as well as tablet-based physical activities [18]. However, optimal intervention features such as intensity, duration, and personalization remain unclear across all types of interventions [19], and currently, no research has explored user preferences in the context of digital dementia risk reduction interventions for people with cognitive concerns. ...
... An American study conducted during COVID also reported high rates of technology use for people with subjective cognitive concerns (94.1% using smartphones, 96.8 using desktops or laptops) [35]. With these high access rates and technology increasingly featured in dementia risk reduction interventions (e.g., [8,14,36,37]), ensuring interventions are tailored and suitable for people with cognitive impairments is vital. ...
Article
Full-text available
Background: Digital dementia risk reduction interventions are cost-effective and scalable. However, it is unknown how they are perceived by people already experiencing cognitive concerns or decline. Objective: To understand the current use, interest, and preferences for online learning courses and interest in learning about factors influencing brain health and dementia risk among adults ≥45. To explore potential differences between individuals experiencing cognitive concerns and those without. Methods: Adults aged 45 and older completed a survey on technology use and healthy ageing (n = 249, Mean age = 65.6, 76.3% female). The Memory Assessment Clinic-Questionnaire was used to assess subjective memory decline, and 153 participants met the study criteria for cognitive concerns (≥25). Results: Almost all participants (98.4%) reported using two or more digital devices, and 51.8% reported increasing device usage following COVID-19. Most (92.1%) were interested in learning about healthy living and memory within an online course, and over 80% indicated a high interest in learning about dementia risk factors. People with cognitive concerns were more likely to report using a ‘routine or system’ to aid memory than people without (82.4% versus 62.9%, p = 0.001). However, no significant difference was found in technology use, course preferences, or interest in learning about different risk factors. Conclusions: We conclude that adults 45 years and over are interested in online methods for learning about brain health and offer unique insights into adapting dementia prevention programs for cognitive concerns.
... If CQ is related to brain activities as advocated by CQ developers [14,15], it is worthwhile to test the association between lifestyle variables and CQ. This is because research in brain science has clarified the relationship between lifestyles and brain conditions [16][17][18], which indicates the consequent association between lifestyles and CQ. Based on this understanding, in this research, we test the association of CQ with such lifestyle variables, adding to the variables related to international experience and personality traits by using businesspersons who were recruited in Japan. ...
... However, it has also been indicated that these two factors may not be sufficient to explain the variance in CQ [3,5]. Regarding CQ closely related to the activities of the brain [14,15], elements of lifestyle could be considered as another possible antecedent of CQ because research in brain science has clarified the relationship between lifestyles and brain conditions [16][17][18], which indicates the consequent association between lifestyles and CQ. This research tests the association of CQ with lifestyle-related variables using businesspersons in Japan, a country that has so far been culturally rather homogenous but is gradually diversifying with the introduction of foreign labor forces in recent years. ...
Article
Full-text available
A growing amount of research is being conducted on cultural intelligence (CQ), which is the ability to adjust and adapt successfully to a variety of surroundings. CQ is a vital quality for people in diversified societies, as are seen today. However, it is still unclear how an individual can develop or strengthen CQ because previous studies have shown that variables such as foreign experience and personality are not exclusively sufficient as CQ antecedents. If CQ can be treated health-scientifically, as the CQ developers argue, diet and exercise that are effective in keeping the body and brain healthy may also correlate with CQ. It is of great significance to clarify the relationship between lifestyle and CQ by applying recent previous research showing the relationship between CQ and both the brain and intelligence, as well as between lifestyle and both the brain and intelligence. Using data derived from 142 Japanese businesspersons, the results of structural equation modeling indicate that lifestyles (dietary balance and walking frequency) are significantly associated with CQ after controlling for personalities (curiosity and grit) and international experiences (length of studying and working abroad), which have been used as predictors of CQ in previous studies. Furthermore, the moderation test showed that the effect of dietary balance on increasing CQ is greater for people with less overseas experience, indicating that dietary balance compensates for lack of overseas experience in the effect of maintaining the level of CQ. These suggest the effectiveness of a health-scientific approach to the influencing factors of CQ. This study is the first to show that CQ is influenced by lifestyle such as walking and dieting, in addition to personality and overseas experience, and will contribute to the future development of health science and cross-cultural research.
... The mnemonic strategy training in the multicomponent intervention of this study may have played a role in the improvement of memory function because it mainly trains participants in memory skills and helps them to store memories more effectively. These results are consistent with studies on the effects of mnemonic strategy training with older adults with SCD or MCI, 40,41 and imaging results of mnemonic strategy training have shown improvements in brain activity. 15,42 In a study of older adults with SCD, participants tended to spontaneously use more mnemonic strategies to compensate for the memory decline they experienced, and using the strategies in everyday life situations made them more beneficial. ...
... Cognitive Decline. 9 The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) trial combined regular aerobic exercise, aspects of the Mediterranean diet (MeDi) and DASH diets, a computerized brain training program, and regular health monitoring. [10][11][12][13] In their cohort of adults aged 60-77, with cardiovascular risk factors and some evidence of neurocognitive weakness based on the Consortium to Establish a Registry for Alzheimer's Disease neuropsychological battery (CERAD-NB), this study provided evidence of neurocognition improvements resulting from the 2-year intervention. ...
Article
Full-text available
INTRODUCTION The Finnish Geriatric Intervention Study (FINGER) led to the global dementia risk reduction initiative: World‐Wide FINGERS (WW‐FINGERS). As part of WW‐FINGERS, the Australian AU‐ARROW study mirrors aspects of FINGER, as well as US‐POINTER. METHOD AU‐ARROW is a randomized, single‐blind, multisite, 2‐year clinical trial ( n = 600; aged 55–79). The multimodal lifestyle intervention group will engage in aerobic exercise, resistance training and stretching, dietary advice to encourage MIND diet adherence, BrainHQ cognitive training, and medical monitoring and health education. The Health Education and Coaching group will receive occasional health education sessions. The primary outcome measure is the change in a global composite cognitive score. Extra value will emanate from blood biomarker analysis, positron emission tomography (PET) imaging, brain magnetic resonance imaging (MRI), and retinal biomarker tests. DISCUSSION The finalized AU‐ARROW protocol is expected to allow development of an evidence‐based innovative treatment plan to reduce cognitive decline and dementia risk, and effective transfer of research outcomes into Australian health policy. Highlights Study protocol for a single‐blind, randomized controlled trial, the AU‐ARROW Study. The AU‐ARROW Study is a member of the World‐Wide FINGERS (WW‐FINGERS) initiative. AU‐ARROW's primary outcome measure is change in a global composite cognitive score. Extra significance from amyloid PET imaging, brain MRI, and retinal biomarker tests. Leading to development of an innovative treatment plan to reduce cognitive decline.
... Tobacco use also places individuals at an increased risk of vascular disease and increases the risk for AD development [42]. Occupations primarily outdoors, focused on using substances for cleaning and manufacturing, or involving routine use of pesticides place participants at an increased exposure level to trace elements and contribute to the development of cognitive disorders [8,14,[43][44][45]. These factors may place participants at a higher risk than others in the population and should be considered to accurately evaluate the risk of cognitive disorders from trace elements in the water. ...
Article
Full-text available
The prevalence of dementia increases with nearly 10 million new cases each year, with Alzheimer’s disease contributing to 60–70% of cases. Environmental factors such as drinking water have been evaluated to determine if a relationship exists between trace elements in drinking water and the risk of developing cognitive disorders in the elderly. The purpose of the current systematic review was to evaluate an association between the composition of drinking water and cognitive function in the elderly. In accordance with the Preferred Reporting Items for Systematic Review and Meta-Analyses guidelines, a literature search was conducted using PubMed and CINAHL databases. A total of 10 studies were included in the current systematic review. Aluminum is the most commonly evaluated trace element in studies (n = 8), followed by silica (n = 5), calcium (n = 4), and fluoride (n = 4). Aluminum exposure showed an increased risk of cognitive decline in four studies, with no association reported in the other studies. Higher silica and pH levels were shown to be protective against a decline in cognitive function. A similar protective effect of calcium was found in two studies. Future research should measure multiple trace mineral levels in all water sources to evaluate the impact on cognitive function.
Article
Full-text available
A tendência de inversão da pirâmide etária traz importante preocupação na incidência de doenças em idosos, destacando-se o comprometimento cognitivo relacionado às síndromes demenciais. Diversos fatores de risco estão associados ao declínio cognitivo nesta população. Dentre os modificáveis, a prática de atividade física e hábitos alimentares saudáveis configuram-se importantes medidas potencialmente protetoras à saúde mental. Avaliar a influência da atividade física e hábitos alimentares sobre a função cognitiva em idosos. Revisão integrativa feita a partir de buscas na literatura nas bases de dados “PubMed” e “Scielo”. 20 trabalhos foram incluídos a partir de uma avaliação criteriosa relacionada ao tema proposto. Os trabalhos mostraram uma tendência de melhora da cognição em idosos a partir da prática de atividades físicas, utilizando como comparação diferentes escalas, como o MoCa, MEEM e o ADAS-Cog. No que tange a alimentação, também foi observada uma tendência de melhora de testes cognitivos a partir de hábitos alimentares específicos. Em ambos os fatores analisados, alguns trabalhos não obtiveram diferenças significativas na análise comparativa (p>0,05). A influência da atividade física no funcionamento cognitivo dos idosos foi testada por meio de exercícios de força, aeróbicos e de flexibilidade, com diferentes frequências e intensidades, o que dificultou o efeito comparativo. Porém, foram descobertos achados interessantes sobre o efeito de redução do declínio cognitivo e melhora de diversos índices - como memória, atenção, habilidades visuais e espaciais - desse grupo ao realizar as atividades. Ademais, os artigos analisados apresentam que o padrão dietético com maior qualidade nutricional, aliada a hábitos de vida saudáveis, favorece a prevenção de doenças e comprometimento da funcionalidade e dos domínios cognitivos. Entre os limitantes dessas análises está o curto período de acompanhamento, desconsiderar a heterogeneidade dos padrões alimentares e a falta de quantificação da influência isolada da alimentação na promoção desses benefícios Os estudos analisados mostraram uma tendência de efeito protetor da atividade física e adoção de alimentação saudável sobre a função cognitiva em idosos, sendo Importante destacar os diferentes fatores limitantes dos trabalhos observados.
Article
Background The ‘Australian National University Alzheimer's Disease Risk Index’ (ANU‐ADRI) assesses the risk of developing Alzheimer's disease (AD) and is a potential tool for its prevention. Objectives The aim of this study is to adapt the ANU‐ADRI‐SF (the short version of ANU‐ADRI) into the Turkish language and Turkish cultural context. Methods The study was methodological and involved the translation and intercultural adaptation of the ANU‐ADRI‐SF into the Turkish language. The study included 384 community‐based participants from a province in the Western Black Sea Region of Türkiye. Data was collected via an online form prepared using Google Forms. Results The index was translated from its original language, English, into Turkish and then retranslated to English by bilingual translators. It was then reviewed and evaluated for possible issues related to translation and degrees of equivalence. When TR‐ANU‐ADRI‐SF levels were compared according to sex, the mean risk scores were found to be 11.25 ± 7.02 for males and 11.69 ± 7.99 for females. After cross‐cultural adaptation, the TR‐ANU‐ADRI‐SF was conceptually intelligible to Turkish adults. Conclusions The TR‐ANU‐ADRI‐SF is a valid and reliable AD risk assessment tool. Implications for practice Given the increase in AD and its impact on people's health, there is a great need for strategies to be implemented by health professionals to improve the lifestyle of the adult population. For use in conjunction with these strategies, a localised AD risk assessment tool that can be applied by clinicians or by individual patients has been adapted and introduced to the Turkish literature.
Article
Background Studies have shown apolipoprotein E (APOE) genotype disclosure to be safe and well-tolerated in cognitively unimpaired (CU) older adults. This study aimed to examine the effect of the disclosure process on decisions about future directives and health behaviors in community-dwelling CU older adults from the Butler Alzheimer’s Prevention Registry (BAPR). Methods CU APOE E4 non-carriers (n = 106) and carriers (n = 80) aged 58-78 completed in-person psychological readiness screening to undergo APOE disclosure. Follow-up assessments were completed online 3 days, 6 weeks, and 6 months post-disclosure. The primary outcomes were future directives, dietary habits, and physical activity scores. Results Disclosure was associated with decision making on future directives in E4 carriers ( t = 3.59, P = .01) at 6 months compared to baseline, but not non-carriers. Family history of memory impairment, SCD endorsement, and education consistently predicted scores on future directives. A significant interaction between E4+ and SCD endorsement on future directive scores was noted (OR = 163.06, 9.5-2,799.8). E4 + carrier status was associated with physical activity ( W = 60,148, P = .005) but not dietary habits scores. Conclusions Our findings indicate that disclosure led to a change in future directives but not protective health behaviors, specifically in E4 carriers. Future work will explore whether pairing disclosure with education about the role of lifestyle factors in AD risk and providing guidelines on making risk-lowering lifestyle modifications as an intervention approach leads to positive change.
Article
Background and purpose The complex aetiology of Alzheimer's disease suggests prevention potential. Risk scores have potential as risk stratification tools and surrogate outcomes in multimodal interventions targeting specific at‐risk populations. The Australian National University Alzheimer's Disease Risk Index (ANU‐ADRI) was tested in relation to cognition and its suitability as a surrogate outcome in a multidomain lifestyle randomized controlled trial, in older adults at risk of dementia. Methods In this post hoc analysis of the Finnish Intervention Study to Prevent Cognitive Impairment and Disability (FINGER), ANU‐ADRI was calculated at baseline, 12, and 24 months ( n = 1174). The association between ANU‐ADRI and cognition (at baseline and over time), the intervention effect on changes in ANU‐ADRI, and the potential impact of baseline ANU‐ADRI on the intervention effect on changes in cognition were assessed using linear mixed models with maximum likelihood estimation. Results A higher ANU‐ADRI was significantly related to worse cognition, at baseline (e.g., estimate for global cognition [95% confidence interval] was −0.028 [−0.032 to −0.025]) and over the 2‐year study (e.g., estimate for 2‐year changes in ANU‐ADRI and per‐year changes in global cognition [95% confidence interval] was −0.068 [−0.026 to −0.108]). No significant beneficial intervention effect was reported for ANU‐ADRI, and baseline ANU‐ADRI did not significantly affect the response to the intervention on changes in cognition. Conclusions The ANU‐ADRI was effective for the risk prediction of cognitive decline. Risk scores may be crucial for the success of novel dementia prevention strategies, but their algorithm, the target population, and the intervention design should be carefully considered when choosing the appropriate tool for each context.
Article
Full-text available
Purpose of Review Here, we have provided an overview of previous biomarker studies to support the importance of subjective cognitive decline (SCD) in early identification of at-risk subjects and differentiation between normal and pathological aging, including Alzheimer’s disease (AD). We have identified several major areas that would require future research to address current gaps in knowledge and increase the value of SCD in preclinical AD. Recent Findings SCD in clinically normal individuals has received increasing attention by clinicians and AD researchers. SCD is etiologically heterogeneous. Biomarker studies have and will continue to increase our knowledge of the biological factors contributing to the manifestation and progression of SCD in AD. Summary Future research with current and new generations of biomarkers is needed to disentangle the biological basis of SCD, improve the utility of SCD for early AD diagnosis, and understand factors that influence SCD characteristics, including sex differences and cognitive reserve.
Article
Full-text available
Background With no cure for dementia and the number of people living with the condition predicted to rapidly rise, there is an urgent need for dementia risk reduction and prevention interventions. Modifiable lifestyle risk factors have been identified as playing a major role in the development of dementia; hence, interventions addressing these risk factors represent a significant opportunity to reduce the number of people developing dementia. Relatively few interventions have been trialed in older participants with cognitive decline (secondary prevention). Objectives This study evaluates the efficacy and feasibility of a multidomain lifestyle risk reduction intervention for people with subjective cognitive decline (SCD) and mild cognitive impairment (MCI). Methods This study is an 8-week, two-arm, single-blind, randomized controlled trial (RCT) of a lifestyle modification program to reduce dementia risk. The active control group receives the following four online educational modules: dementia literacy and lifestyle risk, Mediterranean diet (MeDi), cognitive engagement and physical activity. The intervention group also completes the same educational modules but receives additional practical components including sessions with a dietitian, online brain training and sessions with an exercise physiologist to assist with lifestyle modification. Results Primary outcome measures are cognition (The Alzheimer’s Disease Assessment Scale-Cognitive-Plus [ADAS-Cog-Plus]) and a composite lifestyle risk factor score for Alzheimer’s disease (Australian National University – Alzheimer’s Disease Risk Index [ANU-ADRI]). Secondary outcome measures are motivation to change lifestyle (Motivation to Change Lifestyle and Health Behaviour for Dementia Risk Reduction [MCLHB-DRR]) and health-related quality of life (36-item Short Form Health Survey [SF-36]). Feasibility will be determined through adherence to diet (Mediterranean Diet Adherence Screener [MEDAS] and Australian Recommended Food Score [ARFS]), cognitive engagement (BrainHQ-derived statistics) and physical activity interventions (physical activity calendars). Outcomes are measured at baseline, immediately post-intervention and at 3- and 6-month follow-up by researchers blind to group allocation. Discussion If successful and feasible, secondary prevention lifestyle interventions could provide a targeted, cost-effective way to reduce the number of people with cognitive decline going on to develop Alzheimer’s disease (AD) and other dementias.
Article
Full-text available
Objectives This review provides a broad overview of the effectiveness of interventions for subjective cognitive decline (SCD) in improving psychological well-being, metacognition and objective cognitive performance. Methods Databases including PubMed, Web of Science and Cochrane Systematic Reviews were searched up to August 2017 to identify randomised controlled trials evaluating interventions for SCD. Interventions were categorised as psychological, cognitive, lifestyle or pharmacological. Outcomes of interest included psychological well-being, metacognitive ability and objective cognitive performance. To assess the risk of bias, three authors independently rated study validity using criteria based on the Critical Appraisal Skills Programme. Random-effects meta-analyses were undertaken where three or more studies investigated similar interventions and reported comparable outcomes. Results Twenty studies met inclusion criteria and 16 had sufficient data for inclusion in the meta-analyses. Of these, only seven were rated as being high quality. Group psychological interventions significantly improved psychological well-being (g=0.40, 95% CI 0.03 to 0.76; p=0.03) but the improvement they conferred on metacognitive ability was not statistically significant (g=0.26, 95% CI −0.22 to 0.73; p=0.28). Overall, cognitive training interventions led to a small, statistically significant improvement in objective cognitive performance (g=0.13, 95% CI 0.01 to 0.25; p=0.03). However, the pooled effect sizes of studies using active control groups (g=0.02, 95% CI −0.19 to 0.22; p=0.85) or reporting global cognitive measures (g=0.06, 95% CI –0.19 to 0.31; p=0.66) were non-significant. Conclusions There is a lack of high-quality research in this field. Group psychological interventions improve psychological well-being and may also improve metacognition. A large, high-quality study is indicated to investigate this further. There is no evidence to suggest that cognitive interventions improve global cognitive performance and the clinical utility of small improvements in specific cognitive domains is questionable. There is a lack of research considering lifestyle interventions and poor quality evidence for pharmacological interventions. PROSPERO registration number CRD42017079391.
Article
Full-text available
Introduction It has been estimated that a 10%–25% reduction in seven key risk factors could potentially prevent 1.1–3.0 million Alzheimer’s disease cases globally. In addition, as dementia is preceded by more subtle cognitive deficits which have substantial social and economic impact, effective preventative interventions would likely have more extensive benefits. The current study evaluates in primary care a multidomain risk-reduction intervention targeting adults with high risk of developing dementia. Methods and analysis A randomised controlled trial (RCT) is being conducted to evaluate three intervention programmes using a pragmatic approach suitable to the clinic: (1) a 12-week online and face-to-face dementia risk-reduction intervention (Body Brain Life—General Practice (BBL-GP)); (2) a 6-week face-to-face group lifestyle modification programme (LMP); and (3) a 12-week email-only programme providing general health information. We aim to recruit 240 participants, aged 18 and over, to undergo a comprehensive cognitive and physical assessment at baseline and follow-ups (postintervention, 18, 36 and 62 weeks). The primary outcome is dementia risk measured with the modified version of the Australian National University—Alzheimer’s Disease Risk Index Short Form. Secondary outcomes are cognitive function measured with Trails A and B, and the Digit Symbol Modalities Test; physical activity with moderate-vigorous physical activity and the International Physical Activity Questionnaire; depression with the Centre for Epidemiological Studies Depression; cost evaluation with the 12-item Short Form Health Survey, Framingham Coronary Heart Disease Risk Score and Australian Type 2 Diabetes Risk Assessment Tool; diet quality with the Australian Recommended Food Score; and sleep quality with the Pittsburgh Sleep Quality Index. Ethics and dissemination This RCT is a novel pragmatic intervention applied in a primary care setting to reduce the dementia risk exposure in adults at high risk. If successful, BBL-GP and LMP will provide a versatile, evidence-based package that can be easily and quickly rolled out to other primary care settings and which can be scaled up at relatively low cost compared with other strategies involving intensive interventions. Trial registration number ACTRN12616000868482
Article
Full-text available
Introduction Cognitive training improves cognitive performance and delays functional impairment, but its effects on dementia are not known. We examined whether three different types of cognitive training lowered the risk of dementia across 10 years of follow-up relative to control and if greater number of training sessions attended was associated with lower dementia risk. Methods The Advanced Cognitive Training in Vital Elderly (NCT00298558) study was a randomized controlled trial (N = 2802) among initially healthy older adults, which examined the efficacy of three cognitive training programs (memory, reasoning, or speed of processing) relative to a no-contact control condition. Up to 10 training sessions were delivered over 6 weeks with up to four sessions of booster training delivered at 11 months and a second set of up to four booster sessions at 35 months. Outcome assessments were taken immediately after intervention and at intervals over 10 years. Dementia was defined using a combination of interview- and performance-based methods. Results A total of 260 cases of dementia were identified during the follow-up. Speed training resulted in reduced risk of dementia (hazard ratio [HR] 0.71, 95% confidence interval [CI] 0.50–0.998, P = .049) compared to control, but memory and reasoning training did not (HR 0.79, 95% CI 0.57–1.11, P = .177 and HR 0.79, 95% CI 0.56–1.10, P = .163, respectively). Each additional speed training session was associated with a 10% lower hazard for dementia (unadjusted HR, 0.90; 95% CI, 0.85–0.95, P < .001). Discussion Initially, healthy older adults randomized to speed of processing cognitive training had a 29% reduction in their risk of dementia after 10 years of follow-up compared to the untreated control group.
Article
The Covid-19 crisis has added importance to contact-free transactions and maximum digitalization in every aspect of life. However, vast sections of societies in almost every country are not having access to modern information & communication technology. This has resulted in a phenomenon known as the Digital Divide. Various socio-economic factors contribute to widening of this divide. If digitalization has to succeed faster, we need to bridge this gap as fast as possible. All stakeholders of ICT need to pool their resources and eff orts to help in bridging the digital divide, by enhancing digital literacy and making it more aff ordable. This Paper is based on the speech delivered in the International online conference «New Digital Reality: Science and Education, Law, Security, Economics and Finance» held on July 6–10, 2020.
Article
We aimed to examine the feasibility and effectiveness of a multidomain intervention including intensive and maintenance programs for reducing the risk of dementia in at-risk older adults. Community-dwelling older adults (aged ≥60 years) without dementia but having several risk factors for dementia (N = 32; 89% female; mean age±standard deviation, 76.8±4.7 years) were assigned to three parallel programs: intensive plus maintenance (INT+MNT), intensive only (INT-only), and active control. Subjects in INT+MNT and INT-only groups participated in a 4-week intensive group-based lifestyle modification program that focused on physical activity, vascular risk factors, dietary habits, cognitive activities, and social engagement. INT+MNT participants underwent an additional 20-week maintenance program to consolidate modified habits. The modified Australian National University-Alzheimer's Disease Risk Index (ANU-ADRI) score was used as the primary outcome measure for dementia risk. The changes in ANU-ADRI scores exhibited a significant group-by-time interaction: the INT+MNT group showed significant improvement at 24 weeks (β= -6.05; SE = 1.86; p = 0.002), while the INT-only group did not. Additional exploratory analyses showed that the reduction in ANU-ADRI scores was caused by changes in protective factors rather than in risk factors. The INT + MNT group also showed greater improvement in executive function at 4 and 24 weeks (both p = 0.044), whereas changes in global cognitive function did not reach significance (p = 0.055). A 24-week multidomain dementia prevention involving a maintenance strategy for sustaining modified lifestyle habits reduced the risk of dementia and improved executive function in at-risk older adults.
Article
Acting now on dementia prevention, intervention, and care will vastly improve living and dying for individuals with dementia and their families, and in doing so, will transform the future for society. Dementia is the greatest global challenge for health and social care in the 21st century. It occurs mainly in people older than 65 years, so increases in numbers and costs are driven, worldwide, by increased longevity resulting from the welcome reduction in people dying prematurely. The Lancet Commission on Dementia Prevention, Intervention, and Care met to consolidate the huge strides that have been made and the emerging knowledge as to what we should do to prevent and manage dementia. Globally, about 47 million people were living with dementia in 2015, and this number is projected to triple by 2050. Dementia affects the individuals with the condition, who gradually lose their abilities, as well as their relatives and other supporters, who have to cope with seeing a family member or friend become ill and decline, while responding to their needs, such as increasing dependency and changes in behaviour. Additionally, it affects the wider society because people with dementia also require health and social care. The 2015 global cost of dementia was estimated to be US$818 billion, and this figure will continue to increase as the number of people with dementia rises. Nearly 85% of costs are related to family and social, rather than medical, care. It might be that new medical care in the future, including public health measures, could replace and possibly reduce some of this cost.