This subgroup analysis of a randomized clinical trial examines whether the apolipoprotein E ε4 allele modifies the previously reported significant cognitive benefits of a multidomain lifestyle intervention.
Key Points
Question
Are the cognitive benefits of a 2-year multidomain lifestyle intervention affected by the apolipoprotein E ε4 allele?
Findings
In the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability, a randomized clinical trial of 1260 at-risk elderly individuals from the general population, the cognitive benefits of a multidomain intervention (diet, exercise, cognitive training, and vascular risk management) were not significantly different between apolipoprotein E ε4 carriers and noncarriers (test of interaction). Within-group results by apolipoprotein E ε4 carrier status suggested beneficial effects, particularly among carriers.
Meaning
Healthy lifestyle changes may be beneficial for cognition in older at-risk individuals even in the presence of apolipoprotein E–related genetic susceptibility to dementia.
Abstract
Importance
The role of the apolipoprotein E (APOE) ε4 allele as an effect modifier in lifestyle interventions to prevent cognitive impairment is still unclear.
Objective
To examine whether the APOE ε4 allele modifies the previously reported significant cognitive benefits of a multidomain lifestyle intervention (prespecified subgroup analysis).
Design, Setting, and Participants
The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) was a randomized clinical trial in 6 centers across Finland (screening and randomization performed from September 7, 2009, through November 24, 2011; intervention duration, 2 years). Data analysis was performed from August 1, 2015, to March 31, 2016. The study population was at-risk older individuals from the general population. Inclusion criteria were age of 60 to 77 years; Cardiovascular Risk Factors, Aging, and Dementia risk score of at least 6 points; and cognition at a mean level or slightly lower than expected for age. Individuals with dementia or substantial cognitive impairment and conditions that prevented cooperation or safe engagement in the intervention were excluded. APOE genotype data were available for 1175 of the 1260 participants.
Interventions
Participants were randomly assigned in a 1:1 ratio to a multidomain intervention group (diet, exercise, cognitive training, and vascular risk management) or a control group (general health advice). Group allocation was not actively disclosed to participants, and outcome assessors were masked to group allocation.
Main Outcomes and Measures
Primary outcome was change in cognition measured through a comprehensive neuropsychological test battery. Analysis was based on modified intention to treat (participants with at least 1 postbaseline assessment).
Results
A total of 1109 participants (mean [SD] age, 69.3 [4.7] years; 514 [46.3%] female) were included in the analysis: 362 APOE ε4 allele carriers (173 intervention and 189 control) and 747 noncarriers (380 intervention and 367 control). The APOE ε4 carriers and noncarriers were not significantly different at baseline (except for serum cholesterol level). The difference between the intervention and control groups in annual neuropsychological test battery total score change was 0.037 (95% CI, 0.001 to 0.073) among carriers and 0.014 (95% CI, −0.011 to 0.039) among noncarriers. Intervention effect was not significantly different between carriers and noncarriers (0.023; 95% CI, −0.021 to 0.067).
Conclusions and Relevance
Healthy lifestyle changes may be beneficial for cognition in older at-risk individuals even in the presence of APOE-related genetic susceptibility to dementia. Whether such benefits are more pronounced in APOE ε4 carriers compared with noncarriers should be further investigated. The findings also emphasize the importance of early prevention strategies that target multiple modifiable risk factors simultaneously.
Trial Registration
ClinicalTrials.gov Identifier: NCT01041989
Introduction
Dementia and Alzheimer disease (AD) are complex conditions that likely result from interactions between genetic and environmental factors. The apolipoprotein E (APOE) ε4 allele is the strongest known genetic risk factor for sporadic AD. Most available studies have also linked APOE ε4 to an increased rate of late-life cognitive decline in individuals without dementia, although there is variability among the affected cognitive domains reported in different studies. Several modifiable risk factors for dementia have been identified in population-based studies. It is estimated that approximately one-third of all AD dementia cases worldwide could be attributable to low educational level, physical inactivity, obesity, hypertension, diabetes, smoking, and depression. There is evidence that the APOE genotype interacts with modifiable risk factors, but variability in reported findings still precludes firm conclusions. One prevailing hypothesis is that APOE ε4 carriers are more susceptible to the detrimental effects of environmental risk factors. It remains unclear whether ε4 carriers are more likely to benefit from preventive interventions or whether the ε4 allele counteracts potential intervention benefits.
APOE with its 3 isoforms (ε2, ε3, and ε4) has key roles in lipid transport and metabolism, both systemically and in the brain. The ε4 allele has been linked to cardiovascular and neurologic conditions, particularly AD. The connections between the ε4 allele and AD pathophysiologic findings seem to involve a variety of amyloid-dependent (eg, related to amyloid-β production, aggregation, and clearance) and amyloid-independent (eg, effects on tau phosphorylation and neurofibrillary tangle formation, neuroinflammation, oxidative stress, synaptic plasticity and dendritic spine integrity, brain lipid metabolism, and blood-brain barrier permeability) mechanisms. APOE ε4 carriers have brain structural and developmental features (eg, lower cortical gray matter volume in regions particularly affected by AD) that, together with functional features (eg, deficient neuronal maintenance and repair), increase vulnerability to neuropathologic changes and subsequent late-life cognitive decline.
Previous randomized clinical trials (RCTs) that aimed to prevent cognitive impairment or dementia have yielded mainly negative results, with some positive effects on cognition reported in smaller and/or shorter RCTs of physical activity and/or cognitive training. The effect of the APOE genotype on response to intervention was investigated in some of these RCTs. Beneficial effects among APOE ε4 carriers were reported in patients with mild cognitive impairment treated with donepezil hydrochloride or galantamine hydrobromide, in an RCT of eicosapentaenoic acid and docosahexaenoic acid, in an RCT of docosahexaenoic acid supplementation, and in a small 1-year weight loss RCT in elderly patients with obesity and mild cognitive impairment. Some benefits for APOE ε4 carriers and noncarriers were observed in a trial of Mediterranean diet–based interventions, whereas better effects among APOE ε4 noncarriers were found in a short physical activity RCT. No effects of the APOE genotype on intervention response were found in trials of nonsteroidal anti-inflammatory drugs, statins, gingko biloba, vitamin E, or vitamin B12 supplementation.
The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) was, to our knowledge, the first large, longer-term RCT to report beneficial effects on cognition for a 2-year multidomain lifestyle intervention in 1260 older at-risk individuals from the general population. Herein, we report prespecified analyses of intervention effects on primary and secondary cognitive outcomes by APOE ε4 allele (carriers vs noncarriers).
Methods
Study Participants
The FINGER trial protocol, baseline population characteristics, and primary results have been previously described in detail. The present study is a prespecified subgroup analysis by APOE genotype. Participants were recruited at 6 study sites across Finland from previous population-based observational studies. Eligibility criteria were age of 60 to 77 years; Cardiovascular Risk Factors, Aging, and Dementia risk score of 6 points or higher; the Consortium to Establish a Registry for Alzheimer Disease word list memory task (10 words 3 times) score of 19 words or fewer; Consortium to Establish a Registry for Alzheimer Disease word list recall of 75% or less; or Mini-Mental State Examination score of 26 or fewer of 30 points. Exclusion criteria were previously diagnosed dementia, suspected dementia after clinical assessment by a study physician at the screening visit (individuals recommended for further investigations), Mini-Mental State Examination score of less than 20 points, and conditions that affect safe engagement in the intervention (eg, malignant tumor; major depression; symptomatic cardiovascular disease; revascularization within 1 year); severe vision, hearing, or communicative impairment; conditions that prevent cooperation as judged by the study physician; and coincident participation in another trial. FINGER and this subgroup analysis were approved by the Coordinating Ethics Committee of the Hospital District of Helsinki and Uusimaa. Participants gave written informed consent at screening and baseline visits. All data were deidentified.
Randomization
From September 7, 2009, through November 24, 2011, a total of 2654 individuals were screened for eligibility, and 1260 were randomized 1:1 to the intensive multidomain intervention or regular health advice (ie, control) group. Computer-generated allocation was performed in blocks of 4 (2 persons randomly allocated to each group) at each site. Outcome assessors were masked to allocation and not involved in the intervention. Group allocation was not actively disclosed to participants, and they were advised not to discuss the intervention during testing sessions. Data analysis was performed from August 1, 2015, to March 31, 2016.
Intervention
The control group received regular health advice. Both groups met the study nurse at screening, baseline, and 6, 12, and 24 months (for blood tests and blood pressure, weight, body mass index, and hip and waist circumference measurements) and the study physician at screening and 24 months (for medical history and physical examination). At baseline, the study nurse gave both groups oral and written information and advice on healthy diet and physical, cognitive, and social activities beneficial for vascular risk management and disability prevention. Blood test results were mailed to all participants, together with general written information about the significance of measurements and advice to contact a primary health care practitioner if needed.
The intervention group additionally received 4 intervention components. The nutritional intervention was based on the Finnish Nutrition Recommendations and conducted by study nutritionists (3 individual and 7-9 group sessions). Individual sessions included tailoring of the participant’s diet. Group sessions provided discussions and practical exercises for facilitating lifestyle changes. The physical exercise training program followed international guidelines. Training was guided by study physiotherapists and included aerobic and resistance training and balance exercises. Cognitive training included psychologist-led group sessions and computer-based individual training. The training program was a web-based, in-house–developed program that focused on updating information on memory that was effective in shorter-term RCTs. Social activities were stimulated through the group meetings of all intervention components. Management of metabolic and vascular risk factors was based on national evidence-based guidelines and included additional meetings with the study nurse (at 3, 9, and 18 months) and the study physician (at 3, 6, and 12 months). Study physicians did not prescribe medication but strongly recommended participants to contact their own physician or clinic if needed.
Cognitive Outcomes
Standard neuropsychological tests (an extended version of the Neuropsychological Test Battery [NTB]) were administered at baseline and 12 and 24 months by study psychologists. Participants who dropped out during the study were invited to a final 24-month visit for outcome evaluation. Primary outcome was change in cognitive performance measured with NTB total score, including the 14 tests listed below (calculated on a standardized z scale, with higher scores indicating better performance). Secondary outcomes included NTB domain z scores for executive functioning, processing speed, and memory. The executive functioning domain included Category Fluency, Digit Span, Concept Shifting Test (condition C), Trail Making Test (shifting score B-A), and a shortened 40-stimulus version of the original Stroop test (interference score 3-2). The processing speed domain included Letter Digit Substitution, Concept Shifting (condition A), and Stroop (condition 2). The memory domain included Visual Paired Associates, immediate and delayed recall, Logical Memory immediate and delayed recall, and Word List Learning and Delayed Recall. Post hoc analyses were conducted for an abbreviated memory domain that included 4 of 6 tests (2 associative memory and 2 logical memory tests), including longer recall delay (30 minutes instead of 5 minutes) and requiring more complex processing.
APOE Assessment
Genomic DNA was extracted from venous blood samples with Chemagic MSM1 (PerkinElmer) using magnetic beads. APOE genotyping was determined by polymerase chain reaction using TaqMan genotyping assays (Applied Biosystems) for 2 single-nucleotide polymorphisms (rs429358 and rs7412) and an allelic discrimination method on the Applied Biosystems 7500 platform.
Statistical Analysis
Because of the small number of participants with the APOE ε4ε4 genotype (40 individuals, 16 in the control group and 24 in intervention group), participants were categorized as carriers of at least 1 ε4 allele vs noncarriers. For baseline comparisons between the intervention and control groups by APOE ε4 carrier status, the t test and χ2 test were used as appropriate. Zero-skewness log transformation was applied to skewed NTB components. The z scores for tests at each time point were standardized to the baseline mean and SD. The NTB total score and domain scores for executive functioning, processing speed, and memory were obtained by calculating the mean of the individual NTB component z scores. The minimum number of necessary NTB components was set to 8 of 14 for calculating the NTB total score, 3 of 5 for executive functioning, 2 of 3 for processing speed, and 3 of 6 for memory.
Because data included repeated measurements from the same individuals, longitudinal analyses had to take into account within-person and between-person variability over time. Mixed effects regression models (xtmixed command in Stata [StataCorp]) with maximum likelihood estimation were thus used to analyze change in cognitive scores as a function of randomization group, time, APOE genotype (ε4 allele carriers vs noncarriers), and their interactions (group × time, group × APOE, time × APOE, and group × time × APOE). Following guidelines for subgroup analyses in clinical trials, we report the coefficient (95% CI) for the group × time × APOE interaction as the main result (ie, estimated difference in intervention effects between ε4 carriers and noncarriers per year). We also present the effect estimates (95% CI) within each APOE group using the lincom postestimation command after xtmixed in Stata.
Analyses were conducted according to the predefined primary efficacy analysis based on the modified intention-to-treat (mITT) population, including all randomized participants with at least 1 postbaseline observation (APOE genotype data available for 1109 of 1190 participants). Sensitivity analyses were conducted in the ITT population (all randomized participants; APOE genotype data available for 1175 of 1260 participants) and all randomized participants who completed all cognitive evaluations (APOE genotype data available for 1020 of 1094 participants). Level of significance was set to P < .05 in all analyses, and Stata software, version 14 (StataCorp), was used.
Results
Compared with participants without APOE genotype data, participants with available data included fewer physically active individuals (771 [70.2%] vs 66 [81.4%], P = .03) and more individuals with diabetes at baseline (146 [13.2%] vs 4 [5.0%], P = .03). No other significant differences in participants’ baseline characteristics were found by availability of APOE data.
In the mITT population, the number of APOE ε4 carriers was 173 (31.3%) in the intervention group and 189 (33.9%) in the control group (P = .34). Comparisons of population characteristics between the intervention and control groups among ε4 carriers and noncarriers are given in Table 1. Among ε4 carriers, the intervention group had higher baseline diastolic blood pressure (81.08 vs 79.01 mm Hg, P = .048) and lower baseline memory performance (−0.07 vs 0.08, P = .04) compared with the control group (Table 1). Intervention and control groups were not significantly different among ε4 noncarriers.
Table 1. Baseline Characteristics of FINGER Participants.
Characteristic | APOE ε4 Carriers | APOE ε4 Noncarriers | P Valuec | P Valued | |||||
---|---|---|---|---|---|---|---|---|---|
Sample Size, Control/Intervention | Controla | Interventiona | P Valueb | Sample Size, Control/Intervention | Controla | Interventiona | |||
Demographic characteristics | |||||||||
Age at baseline, y | 189/173 | 68.74 (4.51) | 69.21 (4.5) | .32 | 367/380 | 69.39 (4.8) | 69.61 (4.7) | .52 | .08 |
Women, No. (%) | 189/173 | 94 (49.7) | 81 (46.8) | .58 | 367/380 | 172 (46.9) | 167 (44.0) | .42 | .35 |
Education length, y | 189/172 | 9.87 (3.2) | 10.29 (3.8) | .25 | 366/380 | 10.12 (3.5) | 9.81 (3.2) | .21 | .62 |
Married or cohabiting, No. (%) | 189/172 | 143 (75.7) | 135 (78.5) | .52 | 367/380 | 281 (76.6) | 273 (71.8) | .14 | .31 |
Vascular factors | |||||||||
Systolic blood pressure, mm Hg | 186/172 | 139.50 (16.22) | 141.06 (18.64) | .40 | 364/378 | 139.27 (15.45) | 139.58 (15.95) | .79 | .43 |
Diastolic blood pressure, mm Hg | 186/172 | 79.01 (9.05) | 81.08 (10.65) | .048 | 364/378 | 80.57 (9.43) | 80.22 (9.23) | .62 | .52 |
Serum total cholesterol level, mg/dL | 189/173 | 204 (42) | 205 (41) | .80 | 365/380 | 197 (38) | 197 (37) | .79 | .001 |
Fasting plasma glucose level, mg/dL | 189/173 | 109 (15) | 108 (13) | .26 | 367/380 | 110 (19) | 111 (15) | .41 | .15 |
2-h Oral glucose tolerance test, mg/dL | 161/150 | 127 (37) | 121 (33) | .10 | 318/327 | 125 (41) | 128 (40) | .38 | .33 |
BMI | 186/172 | 28.07 (4.96) | 28.05 (4.39) | .97 | 364/379 | 28.13 (4.63) | 28.57 (4.63) | .19 | .32 |
Other | |||||||||
HbA1c level, % | 189/172 | 5.55 (0.51) | 5.53 (0.48) | .70 | 365/375 | 5.58 (0.60) | 5.59 (0.58) | .91 | .28 |
CRP level, mg/L | 189/173 | 1.84 (2.89) | 2.29 (4.16) | .22 | 365/380 | 3.07 (10.32) | 2.59 (5.70) | .43 | .09 |
HDL-C level, mg/dL | 189/173 | 55 (15) | 56 (15) | .44 | 365/380 | 57 (14) | 55 (14) | .01 | .95 |
LDL-C level, mg/dL | 189/173 | 125 (38) | 125 (36) | .88 | 365/380 | 117 (32) | 117 (32) | .96 | <.001 |
Triglyceride level, mg/dL | 189/173 | 124 (57) | 120 (53) | .54 | 365/380 | 118 (51) | 122 (55) | .24 | .60 |
Lifestyle factors, No. (%) | |||||||||
Physical activity at least twice per week | 188/170 | 134 (71.3) | 128 (75.3) | .39 | 364/377 | 259 (71.2) | 250 (66.3) | .16 | .13 |
Current smokers | 189/170 | 17 (9.0) | 15 (8.8) | .96 | 366/380 | 29 (7.9) | 41 (10.8) | .18 | .80 |
Alcohol intake at least once per week | 188/171 | 80 (42.6) | 73 (42.0) | .98 | 363/379 | 166 (45.7) | 172 (45.38) | .92 | .36 |
Fish intake at least twice per week | 188/171 | 90 (47.9) | 86 (50.3) | .65 | 365/378 | 191 (52.3) | 208 (55.0) | .46 | .15 |
Daily intake of vegetables | 189/172 | 119 (63.0) | 108 (62.8) | .97 | 366/379 | 226 (61.8) | 231 (61.0) | .82 | .62 |
Self-reported medical conditions, No. (%) | |||||||||
Hypertension | 187/172 | 120 (64.2) | 123 (71.5) | .14 | 363/376 | 242 (66.7) | 248 (66.0) | .84 | .65 |
Hypercholesterolemia | 188/171 | 136 (72.3) | 120 (70.2) | .65 | 364/378 | 250 (68.7) | 246 (65.1) | .30 | .14 |
Diabetes | 188/172 | 27 (14.4) | 24 (14.0) | .91 | 365/378 | 43 (11.8) | 52 (13.8) | .42 | .53 |
History of myocardial infarction | 188/173 | 11 (5.8) | 9 (5.2) | .79 | 365/378 | 19 (5.2) | 18 (4.8) | .78 | .69 |
History of stroke | 188/171 | 8 (4.3) | 10 (5.8) | .49 | 364/378 | 22 (6.0) | 20 (5.3) | .66 | .66 |
Cognition | |||||||||
NTB total score, baseline | 189/173 | 0.04 (0.59) | −0.06 (0.58) | .10 | 367/380 | 0.03 (0.58) | −0.01 (0.55) | .37 | .71 |
NTB total score, month 12 | 187/170 | 0.11 (0.67) | 0.04 (0.66) | .32 | 362/367 | 0.15 (0.66) | 0.14 (0.61) | .85 | .10 |
NTB total score, month 24 | 180/159 | 0.16 (0.70) | 0.15 (0.72) | .97 | 347/357 | 0.24 (0.68) | 0.23 (0.65) | .76 | .07 |
Executive functioning, baseline | 188/173 | 0.01 (0.69) | −0.05 (0.67) | .36 | 367/380 | 0.03 (0.68) | −0.03 (0.66) | .23 | .59 |
Executive functioning, month 12 | 186/168 | 0.04 (0.75) | −0.02 (0.68) | .41 | 361/366 | 0.08 (0.74) | 0.08 (0.68) | .89 | .15 |
Executive functioning, month 24 | 179/159 | 0.04 (0.74) | 0.09 (0.75) | .53 | 344/356 | 0.14 (0.72) | 0.11 (0.70) | .56 | .20 |
Memory, baseline | 189/173 | 0.08 (0.65) | −0.07 (0.73) | .04 | 367/380 | 0.01 (0.66) | −0.002 (0.66) | .81 | .96 |
Memory, month 12 | 187/170 | 0.21 (0.77) | 0.09 (0.83) | .16 | 362/368 | 0.24 (0.77) | 0.21 (0.76) | .66 | .12 |
Memory, month 24 | 181/159 | 0.29 (0.85) | 0.25 (0.93) | .72 | 347/357 | 0.40 (0.78) | 0.40 (0.77) | .89 | .02 |
Processing speed, baseline | 189/173 | 0.05 (0.84) | −0.05 (0.80) | .27 | 367/380 | 0.04 (0.83) | −0.01 (0.77) | .42 | .83 |
Processing speed, month 12 | 187/170 | 0.05 (0.89) | 0.03 (0.89) | .86 | 362/367 | 0.08 (0.84) | 0.06 (0.78) | .79 | .58 |
Processing speed, month 24 | 179/159 | 0.07 (0.93) | 0.06 (0.82) | .94 | 347/357 | 0.11 (0.89) | 0.10 (0.86) | .87 | .54 |
Abbreviated memory, baseline | 184/171 | 0.06 (0.76) | −0.07 (0.79) | .10 | 359/375 | 0.02 (0.73) | 0.002 (0.79) | .71 | .79 |
Abbreviated memory, month 12 | 175/167 | 0.08 (0.79) | 0.02 (0.81) | .51 | 352/354 | 0.10 (0.78) | 0.12 (0.81) | .71 | .24 |
Abbreviated memory, month 24 | 174/153 | 0.17 (0.87) | 0.21 (0.92) | .66 | 340/350 | 0.25 (0.79) | 0.28 (0.80) | .66 | .18 |
Abbreviations: APOE, apolipoprotein E; BMI, body mass index (calculated as weight in kilograms divided by height in meters squared); CRP, C-reactive protein; FINGER, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability; HbA1c, hemoglobin A1c; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; NTB, Neuropsychological Test Battery.
SI conversion factors: To convert total cholesterol, HDL-C, and LDL-C to millimoles per liter, multiply by 0.0259; CRP to nanomoles per liter, multiply by 9.524; HbA1c to proportion of hemoglobin, multiply by 0.01; glucose to millimoles per liter, multiply by 0.0555; triglycerides to millimoles per liter, multiply by 0.0113.
Data are presented as mean (SD) unless otherwise specified.
P value for differences between intervention and control groups among APOE ε4 carriers.
P value for differences between intervention and control groups among APOE ε4 noncarriers.
P value for differences between all APOE ε4 carriers and noncarriers.
As expected, APOE ε4 carriers had higher baseline total and low-density lipoprotein cholesterol levels compared with noncarriers (Table 1). No significant differences were found in baseline cognitive performance between ε4 carriers and noncarriers. However, memory performance at month 24 was significantly lower among ε4 carriers (0.27 vs 0.40, P = .02) (Table 1).
Table 2 gives the estimated mean 2-year cognitive change in the intervention and control groups by APOE ε4 carrier status and annual differences between groups (primary analysis, mITT population). Intervention effects (randomization group × time × APOE interaction) did not significantly differ between ε4 carriers and noncarriers for any cognitive domain. Within-group findings by ε4 carrier status indicated that the annual difference between intervention and control groups was significant among ε4 carriers for NTB total score (estimate, 0.037; 95% CI, 0.001 to 0.073; P = .045) and abbreviated memory (estimate, 0.070; 95% CI, 0.006 to 0.135; P = .03) but not among noncarriers (estimates, 0.014 [95% CI, −0.011 to 0.039; P = .28] for NTB total score and 0.022 [95% CI, −0.023 to 0.066; P = .34] for abbreviated memory) (Table 2).
Table 2. Primary and Secondary Cognitive End Points From Baseline to 24 Months.
Cognitive End Point by APOE ε4 Carrier Status | Mean (SE) Changea | Difference Between Intervention and Control Groups per Yearb | Difference Between Carriers and Noncarriers per Year (Intervention × Time × APOE) | |||
---|---|---|---|---|---|---|
Control | Intervention | Estimate (95% CI) | P Value | Estimate (95% CI) | P Value | |
NTB total score (primary end point) | ||||||
Carrier | 0.096 (0.025) | 0.170 (0.027) | 0.037 (0.001 to 0.073) | .045 | 0.023 (−0.021 to 0.067) | .30 |
Noncarrier | 0.194 (0.018) | 0.222 (0.018) | 0.014 (−0.011 to 0.039) | .28 | ||
Executive functioning (secondary end point) | ||||||
Carrier | 0.016 (0.032) | 0.105 (0.034) | 0.045 (−0.002 to 0.091) | .059 | 0.022 (−0.034 to 0.078) | .44 |
Noncarrier | 0.079 (0.023) | 0.123 (0.023) | 0.022 (−0.010 to 0.054) | .17 | ||
Processing speed (secondary end point) | ||||||
Carrier | 0.010 (0.035) | 0.077 (0.037) | 0.034 (−0.015 to 0.083) | .18 | 0.013 (−0.047 to 0.073) | .68 |
Noncarrier | 0.051 (0.025) | 0.093 (0.024) | 0.021 (−0.013 to 0.055) | .22 | ||
Memory (secondary end point) | ||||||
Carrier | 0.200 (0.042) | 0.285 (0.044) | 0.042 (−0.017 to 0.102) | .16 | 0.041 (−0.031 to 0.113) | .27 |
Noncarrier | 0.370 (0.030) | 0.373 (0.030) | 0.001 (−0.040 to 0.043) | .95 | ||
Abbreviated memory (post hoc end point) | ||||||
Carrier | 0.099 (0.045) | 0.239 (0.048) | 0.070 (0.006 to 0.135) | .03 | 0.048 (−0.030 to 0.127) | .22 |
Noncarrier | 0.207 (0.033) | 0.250 (0.032) | 0.022 (−0.023 to 0.066) | .34 |
Abbreviations: APOE, apolipoprotein E; NTB, Neuropsychological Test Battery.
A positive mean change indicates improvement.
A positive value of the estimate of differences between intervention and control groups indicates the effect is in favor of the intervention group.
Sensitivity analyses found results similar to the main analyses (eTable 1 in the Supplement). Population characteristics for sensitivity analyses are given in eTable 2 in the Supplement.
Given the complexity of the models, further analyses were conducted to assess the best-fitting model by performing likelihood ratio tests and comparing the Bayesian Information Criterion for the full model with alternative models that excluded nonsignificant interaction terms (eTable 3 in the Supplement). Detailed results of the best-fitting model for each cognitive outcome are given in eTable 4 in the Supplement. The randomization group × time interaction was similar to previously reported intervention effects. The time × APOE interaction was significant for NTB total score and memory, indicating less overall improvement (intervention and control groups together) among ε4 carriers compared with noncarriers (eTable 4 in the Supplement).
Discussion
Results from the 2-year FINGER trial did not show significant differences between APOE ε4 carriers and noncarriers (test of interaction) regarding the previously reported positive intervention effects on cognition. However, within-group findings by APOE ε4 status showed beneficial intervention effects, especially among APOE ε4 carriers for NTB total score and abbreviated memory score, including more complex memory tests. Baseline performance in these cognitive domains was not different between intervention and control or carrier and noncarrier groups.
The APOE ε4 allele is a key genetic risk factor for cognitive decline, AD, and dementia. One of the main concerns regarding dementia prevention strategies is whether genetically susceptible individuals can still benefit from preventive lifestyle interventions. Thus, the current findings have positive practical implications because the APOE ε4 allele did not seem to hinder the intervention benefits.
Subgroup analyses in clinical trials are challenging. Current reporting guidelines have emphasized between-group comparisons (ie, tests of interaction) as a more appropriate approach in assessing potential heterogeneity of intervention effects. Guidelines have also cautioned against claims of heterogeneity based on only within-group results, which should not be overinterpreted. Interpreting subgroup analyses in trials can be difficult for several reasons, including that statistical power for detecting significant interactions may be limited, dividing the trial population into smaller subgroups may also limit power and lead to nonsignificant within-group findings when the overall intervention effect is significant, and multiple subgroup analyses can increase the probability of false-positive findings.
Thus, given the nonsignificant tests of interaction, the promising within-group findings cannot be considered as definitive evidence that the FINGER intervention was significantly more effective among APOE ε4 carriers. However, the lack of significant interactions should be interpreted cautiously because they may result from statistical power limitations, especially if effect sizes are relatively small. Despite the relatively large cohort and higher prevalence of the APOE ε4 allele in Finland compared with other European countries, the sample size may still have limitations concerning interactions. In addition, the FINGER trial included several prespecified subgroup analyses besides APOE. Findings reported here for 5 different cognitive outcomes were not adjusted for multiple testing because interactions were not significant.
Further studies are needed to clarify whether APOE ε4 carriers may benefit more from lifestyle interventions. In the FINGER trial, overall improvement in NTB total score and memory was less pronounced among ε4 carriers compared with noncarriers (intervention and control groups together). The extended 7-year FINGER follow-up will provide additional data for investigating whether the multidomain lifestyle intervention is effective for preventing dementia, whether this effect is modified by APOE genotype, and whether the cognitive change pattern observed among ε4 carriers persists for a longer period.
Strengths and Limitations
The main strengths of this study are the large sample size, longer duration than what is most common in previous dementia prevention trials, thorough randomization and masking, detailed outcome assessments, and choice of target population. The FINGER participants were at-risk older individuals from the general population without dementia or substantial cognitive impairment (cognitive performance <0.5 SD below the mean level for the cognitively normal Finnish population). The multimodal lifestyle intervention thus started early, before the occurrence of significant clinical impairment. This early start date may be particularly important for APOE ε4 carriers, who have increased susceptibility to detrimental effects of unhealthy lifestyle factors through a variety of mechanisms. The multidomain intervention targeted multiple modifiable risk factors simultaneously, thus potentially covering several of these mechanisms.
This study has some limitations. Despite the relatively large cohort, there may be statistical power limitations for tests of interaction. The exact mechanisms of the within-group effects for APOE ε4 carriers could not be determined. Findings may not necessarily apply to individuals who already have substantial cognitive impairment because they were excluded from the trial.
Conclusions
Results from the FINGER trial suggest that healthy lifestyle changes could be beneficial for cognition in older at-risk individuals even in the presence of APOE-related genetic susceptibility to dementia. Whether such benefits are more pronounced in APOE ε4 carriers compared with noncarriers should be further investigated. The findings also emphasize the importance of early prevention strategies that target multiple modifiable risk factors simultaneously.
References
- 1.Winblad B, Amouyel P, Andrieu S, et al. . Defeating Alzheimer’s disease and other dementias: a priority for European science and society. Lancet Neurol. 2016;15(5):455-532. [DOI] [PubMed] [Google Scholar]
- 2.AlzForum http://www.alzgene.org/. Accessed January 7, 2017.
- 3.Agency for Healthcare Research and Quality Alzheimer’s Disease and Cognitive Decline. http://www.ahrq.gov/research/findings/evidence-based-reports/alzcogtp.html. Accessed January 7, 2017.
- 4.Norton S, Matthews FE, Barnes DE, Yaffe K, Brayne C. Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol. 2014;13(8):788-794. [DOI] [PubMed] [Google Scholar]
- 5.Kivipelto M, Rovio S, Ngandu T, et al. . Apolipoprotein E epsilon4 magnifies lifestyle risks for dementia: a population-based study. J Cell Mol Med. 2008;12(6B):2762-2771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Huang Y, Mahley RW Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's disease. Neurobiol Dis 2014;72(pt A):3-12. [DOI] [PMC free article] [PubMed]
- 7.Yu JT, Tan L, Hardy J. Apolipoprotein E in Alzheimer’s disease: an update. Annu Rev Neurosci. 2014;37:79-100. [DOI] [PubMed] [Google Scholar]
- 8.Willis SL, Tennstedt SL, Marsiske M, et al. ; ACTIVE Study Group . Long-term effects of cognitive training on everyday functional outcomes in older adults. JAMA. 2006;296(23):2805-2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Roig M, Nordbrandt S, Geertsen SS, Nielsen JB. The effects of cardiovascular exercise on human memory: a review with meta-analysis. Neurosci Biobehav Rev. 2013;37(8):1645-1666. [DOI] [PubMed] [Google Scholar]
- 10.Lampit A, Hallock H, Valenzuela M. Computerized cognitive training in cognitively healthy older adults: a systematic review and meta-analysis of effect modifiers. PLoS Med. 2014;11(11):e1001756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Colcombe S, Kramer AF. Fitness effects on the cognitive function of older adults: a meta-analytic study. Psychol Sci. 2003;14(2):125-130. [DOI] [PubMed] [Google Scholar]
- 12.Smith PJ, Blumenthal JA, Hoffman BM, et al. . Aerobic exercise and neurocognitive performance: a meta-analytic review of randomized controlled trials. Psychosom Med. 2010;72(3):239-252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fiatarone Singh MA, Gates N, Saigal N, et al. . The Study of Mental and Resistance Training (SMART) study—resistance training and/or cognitive training in mild cognitive impairment: a randomized, double-blind, double-sham controlled trial. J Am Med Dir Assoc. 2014;15(12):873-880. [DOI] [PubMed] [Google Scholar]
- 14.Petersen RC, Thomas RG, Grundman M, et al. ; Alzheimer’s Disease Cooperative Study Group . Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352(23):2379-2388. [DOI] [PubMed] [Google Scholar]
- 15.Prins ND, van der Flier WA, Knol DL, et al. . The effect of galantamine on brain atrophy rate in subjects with mild cognitive impairment is modified by apolipoprotein E genotype: post-hoc analysis of data from a randomized controlled trial. Alzheimers Res Ther. 2014;6(4):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.van de Rest O, Geleijnse JM, Kok FJ, et al. . Effect of fish oil on cognitive performance in older subjects: a randomized, controlled trial. Neurology. 2008;71(6):430-438. [DOI] [PubMed] [Google Scholar]
- 17.Stonehouse W, Conlon CA, Podd J, et al. . DHA supplementation improved both memory and reaction time in healthy young adults: a randomized controlled trial. Am J Clin Nutr. 2013;97(5):1134-1143. [DOI] [PubMed] [Google Scholar]
- 18.Horie NC, Serrao VT, Simon SS, et al. . Cognitive effects of intentional weight loss in elderly obese individuals with mild cognitive impairment. J Clin Endocrinol Metab. 2016;101(3):1104-1112. [DOI] [PubMed] [Google Scholar]
- 19.Martínez-Lapiscina EH, Galbete C, Corella D, et al. . Genotype patterns at CLU, CR1, PICALM and APOE, cognition and Mediterranean diet: the PREDIMED-NAVARRA trial. Genes Nutr. 2014;9(3):393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lautenschlager NT, Cox KL, Flicker L, et al. . Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial. JAMA. 2008;300(9):1027-1037. [DOI] [PubMed] [Google Scholar]
- 21.Drye LT, Zandi PP. Role of APOE and age at enrollment in the Alzheimer’s Disease Anti-Inflammatory Prevention Trial (ADAPT). Dement Geriatr Cogn Dis Extra. 2012;2(1):304-311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.McGuinness B, Craig D, Bullock R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev. 2016;(1):CD003160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Snitz BE, O’Meara ES, Carlson MC, et al. ; Ginkgo Evaluation of Memory (GEM) Study Investigators . Ginkgo biloba for preventing cognitive decline in older adults: a randomized trial. JAMA. 2009;302(24):2663-2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Smith AD, Smith SM, de Jager CA, et al. . Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PLoS One. 2010;5(9):e12244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ngandu T, Lehtisalo J, Solomon A, et al. . A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet. 2015;385(9984):2255-2263. [DOI] [PubMed] [Google Scholar]
- 26.Kivipelto M, Solomon A, Ahtiluoto S, et al. . The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER): study design and progress. Alzheimers Dement. 2013;9(6):657-665. [DOI] [PubMed] [Google Scholar]
- 27.Ngandu T, Lehtisalo J, Levälahti E, et al. . Recruitment and baseline characteristics of participants in the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER): a randomized controlled lifestyle trial. Int J Environ Res Public Health. 2014;11(9):9345-9360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Vartiainen E, Laatikainen T, Peltonen M, et al. . Thirty-five-year trends in cardiovascular risk factors in Finland. Int J Epidemiol. 2010;39(2):504-518. [DOI] [PubMed] [Google Scholar]
- 29.Saaristo T, Peltonen M, Keinänen-Kiukaanniemi S, et al. ; FIN-D2D Study Group . National type 2 diabetes prevention programme in Finland: FIN-D2D. Int J Circumpolar Health. 2007;66(2):101-112. [DOI] [PubMed] [Google Scholar]
- 30.Kivipelto M, Ngandu T, Laatikainen T, Winblad B, Soininen H, Tuomilehto J. Risk score for the prediction of dementia risk in 20 years among middle aged people: a longitudinal, population-based study. Lancet Neurol. 2006;5(9):735-741. [DOI] [PubMed] [Google Scholar]
- 31.Morris JC, Heyman A, Mohs RC, et al. ; Consortium to Establish a Registry for Alzheimer’s Disease (CERAD) . Part I: clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989;39(9):1159-1165. [DOI] [PubMed] [Google Scholar]
- 32.National Nutrition Council Finnish Nutrition Recommendations: Diet and Physical Activity in Balance. Helsinki, Finland: Edita Publishing; 2005. [Google Scholar]
- 33.Nelson ME, Rejeski WJ, Blair SN, et al. ; American College of Sports Medicine; American Heart Association . Physical activity and public health in older adults: recommendation from the American College of Sports Medicine and the American Heart Association. Circulation. 2007;116(9):1094-1105. [DOI] [PubMed] [Google Scholar]
- 34.Komulainen P, Kivipelto M, Lakka TA, et al. . Exercise, fitness and cognition—a randomised controlled trial in older individuals: the DR’s EXTRA study. Eur Geriatr Med. 2010;1:266-272. [Google Scholar]
- 35.Dahlin E, Neely AS, Larsson A, Bäckman L, Nyberg L. Transfer of learning after updating training mediated by the striatum. Science. 2008;320(5882):1510-1512. [DOI] [PubMed] [Google Scholar]
- 36.Bäckman L, Nyberg L, Soveri A, et al. . Effects of working-memory training on striatal dopamine release. Science. 2011;333(6043):718. [DOI] [PubMed] [Google Scholar]
- 37.Gavelin HM, Boraxbekk C-J, Stenlund T, Järvholm LS, Neely AS. Effects of a process-based cognitive training intervention for patients with stress-related exhaustion. Stress. 2015;18(5):578-588. [DOI] [PubMed] [Google Scholar]
- 38.Harrison J, Minassian SL, Jenkins L, Black RS, Koller M, Grundman M. A neuropsychological test battery for use in Alzheimer disease clinical trials. Arch Neurol. 2007;64(9):1323-1329. [DOI] [PubMed] [Google Scholar]
- 39.De la Vega FM, Lazaruk KD, Rhodes MD, Wenz MH. Assessment of two flexible and compatible SNP genotyping platforms: TaqMan SNP Genotyping Assays and the SNPlex Genotyping System. Mutat Res. 2005;573(1-2):111-135. [DOI] [PubMed] [Google Scholar]
- 40.Wang R, Lagakos SW, Ware JH, Hunter DJ, Drazen JM. Statistics in medicine–reporting of subgroup analyses in clinical trials. N Engl J Med. 2007;357(21):2189-2194. [DOI] [PubMed] [Google Scholar]
- 41.Corbo RM, Scacchi R. Apolipoprotein E (APOE) allele distribution in the world: is APOE*4 a ‘thrifty’ allele? Ann Hum Genet. 1999;63(pt 4):301-310. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.