Effect of APOE and a polygenic risk score on incident dementia and cognitive decline in a healthy older population ================================================================================================================== * Moeen Riaz * Aamira Huq * Joanne Ryan * Suzanne G Orchard * Jane Tiller * Jessica Lockery * Robyn L Woods * Rory Wolfe * Alan E. Renton * Alison M. Goate * Robert Sebra * Eric Schadt * Amy Brodtmann * Raj C Shah * Elsdon Storey * Anne M Murray * John J McNeil * Paul Lacaze ## Abstract **Importance** Few studies have measured the effect of genetic factors on dementia and cognitive decline in a population of healthy older individuals followed prospectively. **Objective** To examine the effect of Apolipoprotein E (*APOE*) genotypes and a polygenic risk score (PRS) on incident dementia and cognitive decline in a longitudinal cohort of healthy older people. **Design, Setting and Participants** Post-hoc genetic analysis of a randomized clinical trial population - the ASPirin in Reducing Events in the Elderly (ASPREE) trial. At enrollment, participants had no history of diagnosed dementia, atherothrombotic cardiovascular disease, or permanent physical disability and were without cognitive impairment. **Main Outcomes and Measures** Dementia (adjudicated trial endpoint) and cognitive decline, defined as a >1.5 standard deviation decline in test score for either global cognition, episodic memory, language/executive function or psychomotor speed, versus baseline scores. Cumulative incidence curves for all-cause dementia and cognitive decline were calculated with mortality as a competing event, stratified by *APOE* genotypes and tertiles of a PRS based on 23 common non-*APOE* variants. **Results** 12,978 participants with European ancestry were included; 54.8% were female, and average age at baseline was 75 years (range 70 to 96). During a median 4.5 years of follow-up, 324 (2.5%) participants developed dementia and 503 (3.8%) died. Cumulative incidence of dementia to age 85 years was estimated to be 7.4% in all participants, 12.6% in *APOE* ε4 heterozygotes, 26.6% in ε4 homozygotes, 9.6% in the high PRS tertile, and 7.3% in the low PRS tertile. *APOE* ε4 heterozygosity/homozygosity was associated with a 2.5/6.3-fold increased risk of dementia and a 1.4/1.8-fold increased risk of cognitive decline, versus ε3/ε3 (P<0.001 for both). A high PRS (top tertile) was associated with a 1.4-fold increase risk of dementia, versus the low tertile (CI 1.04-1.76, P=0.02), but was not associated with cognitive decline risk (CI 0.96-1.22, P = 0.18). **Conclusions and Relevance** Incidence of dementia among healthy older individuals is low across all genotypes; however, *APOE* ε4 and high PRS increase relative risk. *APOE* ε4 is associated with cognitive decline, but PRS is not. **Question** How do genetic factors influence the risk of dementia and cognitive decline among healthy older individuals? **Findings** We studied cumulative incidence of dementia and cognitive decline in 12,978 healthy older individuals without cardiovascular disease or cognitive impairment at enrollment, stratified by *APOE* genotype and a polygenic risk score (PRS). *APOE* ε4 and PRS increased the relative risk of dementia, but cumulative incidence was low across all genotypes. *APOE* genotypes were associated with cognitive decline, but PRS was not. **Meaning** Incidence of dementia is low among healthy older individuals; however, genetic factors still increase relative risk. ## INTRODUCTION Few studies have measured the effect of Apolipoprotein E (*APOE*) genotypes and polygenic risk scores (PRS) on incident dementia and cognitive decline in healthy older people. The ASPREE (ASPirin in Reducing Events in the Elderly) cohort offers the opportunity to measure these effects, as recruited participants had no history of cardiovascular disease, dementia or significant physical disability at enrollment. The ASPREE study was a randomised, placebo-controlled trial to determine whether daily low dose aspirin increased survival, free of dementia or persistent physical disability, in 19,114 healthy community-dwelling older people(1). In 2018, ASPREE reported that over an average 4.5 year follow-up, aspirin did not prolong disability-free survival(2-4) or reduce the risk of dementia or cognitive decline(5). The *APOE* gene is the strongest genetic determinant of all-cause dementia, especially Alzheimer’s disease (AD), with the ε4 allele elevating risk and accelerating age of onset(6-9). The ε4 allele is also associated with cognitive impairment (dysfunction in episodic memory, processing speed, executive function or global cognition) in people without a dementia diagnosis(10-13). Beyond *APOE*, common disease-associated variants identified from genome-wide association studies (GWAS)(14-18) also modify dementia risk, and can be used to calculate a polygenic risk score (PRS)(9, 19-24). Individually, these common genetic variants have low effect sizes, yet when combined into a PRS can enable risk-stratification for dementia indications beyond *APOE* genotype. There is varying evidence for whether a PRS for dementia can also predict cognitive decline(25-28). Incorporating both *APOE* genotypes and PRS, alongside conventional risk factors, may enable more accurate risk prediction(29, 30). This may aid development of therapeutic strategies or prevention, and advance our understanding of the genetic differences between (diagnosed) dementia, and cognitive decline. The predictive performance of PRSs for dementia requires further investigation in well-characterised prospective studies. Predictive performance can be influenced by factors such as ethnicity, age, study recruitment criteria, clinical diagnostic criteria, neuropsychological assessments used, genotyping platform, and genetic variants included(6-9, 19-24, 29-31). More studies of cognitively healthy elderly individuals followed prospectively are required to assess variability and predictive accuracy. Here, we report the effects of *APOE* and PRS on incident dementia and cognitive decline among 12,978 ASPREE participants, where dementia was an exclusionary criterion at entry and adjudicated as a primary trial endpoint. ## METHODS ### Study population Consistent with the ASPREE inclusion criteria(1), participants had no previous history or current diagnosis of atherothrombotic cardiovascular disease, dementia, loss of independence with basic activities of daily living, or life-threatening illness. Participants passed a global cognition screen at enrollment (>77 on the Modified Mini-Mental State (3MS) Examination). Informed consent for genetic analysis was obtained, with ethical approval from the Alfred Hospital Human Research Ethics Committee (390/15) and site-specific Institutional Review Boards (US). ### Incident Dementia Diagnosis After standardized cognition and functional measures, participants reporting memory or cognitive problems were assessed by specialists or prescribed dementia medication (Australia). Following identification of dementia triggers (3MS<78 or a drop of >10.15 points from the participant’s baseline 3MS score, accounting for age and education), additional assessments were conducted, with brain imaging and laboratory analyses collected for adjudication. Each dementia trigger case was reviewed according to the ASPREE protocol for clinical adjudication(4, 5) by an adjudication committee consisting of geriatricians, neurologists and neuropsychologists. Dementia was diagnosed using Diagnostic and Statistical Manual of Mental Disorders, fourth edition criteria. Diagnosis date was recorded as date of trigger. ### Cognitive Decline The ASPREE cognitive battery included the 3MS for general cognition, the Hopkins Verbal Learning Test-Revised (HVLT-R) for episodic memory, the single letter Controlled Oral World Association Test (COWAT) for language and executive function, and the Symbol Digit Modalities Test (SDMT) to measure psychomotor speed. Accredited professionals administered assessments at baseline and year 1, followed biennially during follow-up. As reported previously(5), cognitive decline in participants without a dementia diagnosis was defined as a 1.5 standard deviation decline in 3MS/HVLT-R/SDMT/COWAT compared with baseline scores, sustained over ≥2 timepoints. ### Genotyping and variant analysis 14,052 samples were genotyped using the Axiom 2.0 Precision Medicine Diversity Research Array following standard protocols. 12,978 samples passed quality control (12,343 Australian, 635 US) based on sex, relatedness and ancestry (Non-Finnish Europeans). To estimate population structure, we performed principal component analysis using the 1000 Genomes reference population (Figure S1)(32, 33). Imputation was performed using the haplotype reference consortium European panel(34). Post-imputation quality control removed variants r2<0.3. *APOE* genotype was measured using two directly genotyped variants (rs7412, rs429358) extracted using plink v1.9(35). ### Polygenic risk score PRS was calculated using 23 common variants (15 genotyped, 8 imputed) associated with AD at genome-wide significance, that affect risk independently of *APOE*(17, 24, 36). PRS calculations, using plink v1.9(35), were based on dosage (0,1,2) of SNP effect allele reported from GWAS, multiplied by effect sizes, followed by the sum of products to generate a PRS per participant (Table S1). PRS distribution was divided into low/middle/high tertiles; with mean values of; low −0.56 (range −1.43 to −0.34), middle −0.20 (−0.34 to −0.06), high 0.16 (−0.06 to 1.86)(Figure S2). ### Statistical analysis To determine whether *APOE* genotype frequencies were under selective pressure due to age and/or trial inclusion/exclusion criteria, we performed Hardy-Weinberg equilibrium (HWE) testing. This compared observed genotype frequencies with those expected in a population under no selective pressure, using chi-squared tests. We examined the cumulative incidence of dementia (CID) and cognitive decline, stratified by *APOE* genotype and PRS tertiles. We used ε3 homozygotes as a reference group for *APOE*-stratified analysis, and the low-risk tertile for PRS-stratified analysis. Consistent with other studies(9, 24) we combined *APOE* ε3/ε4:ε2/ε4 into a single group, and ε2/ε2:ε2/ε3 into a single group. We estimated cumulative incidence of all-cause dementia and cognitive decline during an average of 4.5 years of follow-up, using the Cumulative Incidence Function (CIF) of the etm package(37, 38) in R version 3.6.0(39). Data were censored by date of dementia diagnosis, cognitive decline, last contact or death. The age on censored date was used as a time scale in CIF model. Cumulative incidence was calculated up to 95 years, then stratified by *APOE* genotype and PRS tertiles. Dementia and cognitive decline between PRS tertiles was compared for the whole cohort and further stratified by *APOE* genotypes. The dementia and cognitive decline models were estimated independently. We used the Fine and Gray (F&G) method of accounting for competing risk of death, and Cox proportional hazard regression model to calculate dementia hazard ratio of both models, for *APOE*, PRS and their interaction, adjusted for age at enrollment (continuous, allowing a quadratic function) and sex(9, 37). We used age on censored date as a time scale in both F&G and Cox models. Hazard ratios for cognitive decline were measured the same way. To test association of *APOE* genotypes and PRS with cohort characteristics, we used a multivariable regression model with variables; age, sex, follow-up time, education, alcohol use, smoking, diabetes mellitus, hypertension, depression (Center for Epidemiological Studies-Depression-10 scale), family history of dementia (father/mother/sibling), body mass index, blood pressure, cholesterol, triglycerides. Bonferroni multiple test correction at P=0.002 significance was applied (0.05/17=0.002). ## RESULTS Characteristics of the 12,978 genotyped participants are shown in Table 1. Overall, 54.8% were female, 47% had educational attainment <12 years, 3% were current smokers, and 25% reported a family history of dementia at enrollment. View this table: [Table 1:](http://medrxiv.org/content/early/2020/10/16/2020.10.11.20210963/T1) Table 1: Characteristics of the ASPREE cohort stratified by *APOE* genotypes and tertiles of a PRS Associations of cohort characteristics with *APOE* genotypes and PRS tertiles are also shown in Table 1. The only associations to survive multiple testing correction was family history of dementia in ε4 heterozygotes/homozygotes, with no cohort characteristics differing between PRS tertiles (Table 1). We found that *APOE* genotype frequencies had deviated from HWE (chi-square=38, P=<0.001)(Table S2), with fewer ε3/ε4 heterozygotes (N=2665 observed, N=2723 expected) and fewer ε4/ε4 homozygotes (N=200 observed, N=239 expected) than expected under HWE. During mean 4.5 years of follow-up (interquartile range 2.1 to 5.7 years, 2779 person-years), we observed 324 (2.5%) incident all-cause dementia cases and 503 (3.8%) deaths (Table 2). For cumulative incidence of dementia (CID), we describe results up to age 85 years, representing an approximate centre point between lower and upper age ranges of the ASPREE population at baseline (70 to 96 years). CID in ASPREE was estimated at 7.4% (CI 6.5 to 8.5). View this table: [Table 2:](http://medrxiv.org/content/early/2020/10/16/2020.10.11.20210963/T2) Table 2: Cumulative incidence of dementia and death in ASPREE In *APOE* genotype-stratified analysis of CID, after adjusting for covariates and death as a competing event, ε4/ε4 genotype was significantly associated with dementia risk (HR 6.38 [CI 3.8-10.7] P=2.0×10−12) compared with ε3/ε3 (Table 3a). Individuals with ε3/ε4 heterozygosity were also at higher risk of dementia (HR 2.51 [CI 1.9-3.1], P=1.5×10−14) compared with ε3/ε3. CID was 26.6% (CI 16.2-42.0) for ε4/ε4 homozygotes, 12.6% (CI 10.2-15.5) for ε3/ε4 heterozygotes, 5.9% (CI 4.8- 7.2) for the common ε3/ε3 genotype group, and 4.0% (CI 2.4-6.5) for the lower-risk ε2/ε2:ε2/ε3 group (Figure 1a). For all *APOE* genotype-stratified results see Table S3. View this table: [Table 3:](http://medrxiv.org/content/early/2020/10/16/2020.10.11.20210963/T3) Table 3: Cox proportional hazard ratio and risk regression models for dementia and cognitive decline risk in the ASPREE cohort ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/10/16/2020.10.11.20210963/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2020/10/16/2020.10.11.20210963/F1) Figure 1. Cumulative incidence of all-cause dementia stratified by *APOE* genotypes and tertiles of a polygenic risk score (PRS). Cumulative incidence curves for all-cause dementia (**a**) and cognitive decline (**b**) were calculated to age 95 years and stratified by *APOE* genotype, with mortality as a competing event. Confidence intervals and participants at risk are shown in Table S3-4. Dementia risk was higher for participants in the high-risk PRS tertile than the low (HR 1.36 [CI 1.0- 1.7], P=0.02)(Table 3a). CID in the high-risk tertile was 9.6% (CI 7.8-11.8) compared with the low tertile 7.3% (CI 5.7-9.3)(Figure 1b; Table S4). At age 95 years, the effect of PRS was more prominent with CID increasing from 17.6% (13.4-23.0) in the low PRS tertile to 30.6% (21.9-41.9) in the high (Table S4). In sub-group analysis, among *APOE* ε3/ε4 heterozygotes, PRS modified dementia risk, with CID increasing from 10.8% (CI 7.2-16.3) in the low PRS tertile to 17.8% (CI 13.2-23.8) in the high (Table S5). Among *APOE* ε4/ε4 homozygotes with high PRS (the highest genetic risk category), CID was 32.2% (CI 11.3-71.6). In ε4/ε4 homozygotes with low PRS, CID was lower at 24.6% (CI 11.2-48.8). For ε3/ε3 homozygotes, CID in the low-risk PRS tertile was 5.7% (CI 3.9-8.3) and in the high-risk 7.6% (CI 5.5-10.5)(Table S5). We compared CID between the highest genetic risk group at age 80 (ε4 carriers with high PRS) and the lowest genetic risk group at age 90 (ε2 carriers with low PRS). CID in the highest genetic risk group at age 80 was 6.1% (CI 4.1-9.0) and in the lowest genetic risk at age 90 was 8.8% (CI 4.5-16.7) (Table S5). This corresponded to an approximately 10-year delay in age-of-onset between these two extreme groups. In sensitivity analysis, we examined interaction between *APOE/*PRS, and found no significant association with incident dementia (P>0.05). A total of 1598 (12.6%) participants had cognitive decline (Table 1). The cumulative incidence of cognitive decline to age 85 years was estimated to be 37.2% (CI 36.4-41.0) in *APOE* ε3/ε3 homozygotes, 35.3% (CI 30.5-39.6) in ε2/ε2 homozygotes, 45.7% (CI 46.5-53.9) in ε3/ε4 heterozygotes, and 52.9% (CI 46.1-76.2) in ε4/ε4 homozygotes (Figure 2a, Table S6). Compared with the ε3/ε3 reference group, cognitive decline risk was significantly higher in ε3/ε4 heterozygotes (HR=1.35 [1.20-1.51], P<0.001) and ε4 homozygotes (HR=1.75 [CI 1.24-2.46]), P<0.001)(Table 3b).PRS was not associated with cognitive decline. Risk of cognitive decline did not significantly increase between low and high PRS tertiles (HR=1.08 [0.96-1.22], P=0.18)(Figure 2b, Table S7-S8). In sensitivity analysis, the interaction effect between *APOE*/PRS for cognitive decline was not significant (P>0.05). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/10/16/2020.10.11.20210963/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/10/16/2020.10.11.20210963/F2) Figure 2: Cumulative incidence of cognitive decline stratified by *APOE* genotypes and tertiles of a polygenic risk score (PRS). Cumulative incidence curves for cognitive decline were calculated with mortality as a competing event, stratified by *APOE* genotypes (**a**) tertiles of a PRS (**b**) based on 23 common non-*APOE* variants. High PRS is shown in red; low PRS in green (mid tertile not shown). Confidence intervals and participants at risk are shown in Table S6-7. ## DISCUSSION In this study, we examined the effect of *APOE* genotypes and PRS on incident dementia and cognitive decline among 12,978 initially healthy older participants. We found that *APOE* ε4 and high PRS were associated with increased relative risk of dementia, but overall, cumulative incidence of dementia was low across all genotype groups. PRS effect on dementia risk was modest and delayed compared with *APOE* ε4, mostly affecting risk after 85 years of age. *APOE* ε4 was associated with cognitive decline, but PRS was not, suggesting that *APOE* genotype has a stronger effect than PRS on both dementia and cognitive decline. We observe that the absence of co-morbidities, atherothrombotic cardiovascular disease and cognitive impairment to age 70 years contributed to the attenuation of incident dementia across all genotypes. The unique ascertainment of the ASPREE population is an important factor in the interpretation of our results. The eligibility criteria excluded individuals with dementia diagnoses and cognitive impairment at enrollment, and individuals with any history or diagnosis of atherothrombotic cardiovascular disease events, major physical disability or life-threatening cancer(1). This produced a highly selected population of healthy older individuals, who at the time of study entry, benefited from the absence of several important dementia risk factors. This selective pressure resulted in deviation from the Hardy-Weinberg equilibrium, with fewer deleterious *APOE* ε4 alleles observed than expected. Selection against ε4 was driven by the age cut-off (>70 years), as well as the strict inclusion/exclusion criteria of the trial. We accordingly observed a low cumulative incidence of all-cause dementia, estimated to be 7.4% to age 85 across all participants. This estimate was approximately half that reported in the community- based Rotterdam study to the same age (15.6%)(9). While acknowledging potential issues with comparing dementia risk between different studies, including differences in population demography, recruitment criteria, diagnostic definitions, and duration of follow-up(7, 9, 24), we consider comparisons between ASPREE and the Rotterdam study warranted. The studies had similar sample sizes, age ranges, sex percentages, diabetes, hypertension, BMI, blood lipids, genetic ancestry and adjudicated dementia cases. Further, both analyses used the same PRS calculations(17, 24, 36). The lower risk of dementia in ASPREE is likely influenced by the selection of healthy participants, depletion of deleterious *APOE* ε4 alleles, and a relatively short follow-up period where healthy selection effects have not yet dissipated. The estimated CID in ASPREE was 26.6% for ε4/ε4 homozygotes, compared with approximately 60% in the Rotterdam study, and 5.9% for ε3/ε4 heterozygotes, compared with approximately 25% in the Rotterdam study(9). These differences in CID are substantial, unlikely to be attributable to confounding factors alone between the studies. Further, in a recent meta-analysis of three population-based cohorts of cognitively normal subjects aged 60-75 years (total N=11,771), the risk of dementia in *APOE* ε4/ε4 homozygotes (N=134) to age 70-75 years was 11.2%(7). In ASPREE, however, the risk of dementia to age 75 in ε4/ε4 (N=200) was only 3.7%. Risk of dementia among ε4/ε4 homozygotes to age 85 years in the Framingham Heart Study (37.6%, N=67) was also considerably higher than ASPREE (26.6% N=200)(7). PRS is more challenging to interpret across studies, given the different PRSs used(7, 9, 21-24). However, we also observed an attenuated effect of PRS on dementia in ASPREE compared with other studies. We observed only a 2.6% difference in CID between low (7.3%) and high (9.6%) PRS tertiles. In the Rotterdam study, the observed difference was 9.0% between low (11.6%) and high (20.4%) tertiles to the same age. In ASPREE, the effect of PRS was more pronounced in *APOE* ε4 carriers, compared with the reference ε3/ε3 group. However, the PRS effect was attenuated and delayed in age-of-onset compared with other studies(9, 40). The PRS effect on dementia risk in ASPREE mostly occurred after the age of 85 years (Figure 2a). We found no significant interaction effect between *APOE* and PRS in ASPREE, unlike the Rotterdam study(9). This again may reflect the attenuation of genetic effects on dementia risk in ASPREE. A recent analysis of the Framingham cohort also reported no significant interaction between *APOE* and PRS while evaluating dementia risk(40). Further studies with large populations and longer follow-up are required to understand interactions between *APOE* and PRS in modifying dementia risk. Considering the attenuated genetic risk of dementia observed in ASPREE, we query whether other factors further modified risk, beyond the low vascular risk, cognitive screening and absence of cardiovascular disease at baseline. Such factors could include a favourable lifestyle, characterized by healthy diet, regular exercise and high socialisation levels(29, 30). Alternatively, the attenuation could be related to the relatively short follow-up period, during which healthy selection effects had not yet dissipated. Protective genetic loci not included in the PRS may also have contributed to risk modification, including common variants yet to be identified by GWAS, and/or rare high-effect protective variants, including loss-of-function variants in biologically associated genes. There is growing evidence that protection from dementia risk can be conferred by both common and rare genetic variants, especially in the high-risk *APOE* ε4/ε4 group(41, 42). Further studies are required to examine the effect of protective genetic variants for dementia in ASPREE. *APOE* ε4 carrier status was significantly associated with cognitive decline in ASPREE, but PRS was not. This reflects the more modest effect of PRS on cognitive aging, and/or a divergent genetic aetiology versus *APOE* genotype(26). The association between *APOE* ε4 and cognitive decline in non-demented individuals has been reported by several studies using comparable cognitive testing(10-13, 28). However, few studies have reported a significant effect of PRS on cognitive decline alone(25-28). It appears that PRS derived from GWAS of diagnosed dementia/AD cases are not strong predictors of cognitive decline without dementia during aging. However, our approach to quantifying cognitive decline may be insensitive, or might reflect a different biological process. Alternatively, PRS derived from a GWAS of dementia/AD cases may reflect the functional impairment required for dementia diagnosis, rather than the cognitive aspects. Strengths of the study include a well-characterized longitudinal cohort with repeated cognitive assessments and dementia adjudication, genetic data for both *APOE* and PRS variants, longitudinal follow-up to enable survival analysis for dementia and cognition, data available on covariates, adjudicated reports of causes of death to control for competing events, and a large number of initially healthy elderly participants. Limitations of the study include a shorter duration of follow-up compared with other studies(7, 9) (possibly insufficient to overcome a healthy volunteer effect) and limited event numbers in some rarer *APOE* genotype groups. Dementia events were not stratified into AD versus other dementia types. However, very few dementia cases were classified as non-AD in ASPREE (1.2%), with the majority of dementia classified as probable/possible-AD(5). In conclusion, our study found that *APOE* genotypes and PRS effect the relative risk of dementia in a population of healthy older individuals followed prospectively. However, overall CID in the population was low across all genotype groups, reflecting the healthy nature of the population at enrollment. *APOE* ε4 had a stronger effect than PRS on dementia risk. *APOE* genotypes affected cognitive decline, whereas PRS did not. Prospective studies of initially healthy older participants with longer follow-up periods are required to further understand the genetic risk of dementia and cognitive decline during aging, and examine the predictive performance and clinical utility of PRS. ## Supporting information Supplement Details [[supplements/210963_file02.pdf]](pending:yes) ## Data Availability Contact senior author of the paper for data availability ## Funding The ASPREE Healthy Ageing Biobank was supported by a Flagship cluster (including the Commonwealth Scientific and Industrial Research Organisation, Monash University, Menzies Research Institute, Australian National University, University of Melbourne), Monash University and the National Cancer Institute at the National Institutes of Health (supplement to U01AG029824). The ASPREE clinical trial was supported by the National Institute on Aging and the National Cancer Institute at the National Institutes of Health (U01AG029824). the National Health and Medical Research Council (NHMRC) of Australia (334047 and 1127060), Monash University and the Victorian Cancer Agency. We thank the trial staff in Australia and the US, the participants who volunteered for this trial, and the general practitioners and staff of the medical clinics who cared for the participants. We also thank the UK Biobank participants and admin staff. P.L is supported by a National Heart Foundation Future Leader Fellowship (102604). ## Disclosures Dr. Shah serves as a non-compensated member of the Board of Directors of the Alzheimer’s Association – Illinois Chapter. His institution, Rush University Medical Center, receives funds for his role as Site Principal Investigator or Site Sub-Investigator for industry sponsored clinical trials and research studies involving Alzheimer’s disease and dementia sponsored by Amylyx Pharmaceuticals, Inc., Eli Lilly & Co., Inc., Genentech, Inc., Merck & Co., Inc., Navidea Biopharmaceuticals, and Novartis Pharmaceuticals, Inc. Dr Sebra serves as Vice-President of Technology Development at Sema4. Dr Schadt serves as Chief Executive Officer at Sema4. Dr Goate has consulted for Biogen AbbVie, GSK, Eisai and Denali Therapeutics. No other conflicts were reported. ## Acknowledgements We thank the ASPREE trial staff in Australia and the United States, the ASPREE participants who volunteered for the trial, and the general practitioners and staff of the medical clinics who cared for the participants. ## ABBREVIATIONS AD : Alzheimer’s disease APOE : Apolipoprotein E ASPREE : ASPirin in Reducing Events in the Elderly GWAS : Genome-wide association study PRS : Polygenic risk score HWE : Hardy-Weinberg equilibrium CID : Cumulative incidence of dementia * Received October 11, 2020. * Revision received October 15, 2020. * Accepted October 16, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.McNeil JJ, Woods RL, Nelson MR, Murray AM, Reid CM, Kirpach B, et al. Baseline Characteristics of Participants in the ASPREE (ASPirin in Reducing Events in the Elderly) Study. J Gerontol A Biol Sci Med Sci. 2017 Oct 12;72(11):1586–93. PubMed PMID: 28329340. PMCID: PMC5861878. Epub 2017/03/23. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28329340&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 2. 2.McNeil JJ, Nelson MR, Woods RL, Lockery JE, Wolfe R, Reid CM, et al. Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. N Engl J Med. 2018 Oct 18;379(16):1519–28. PubMed PMID: 30221595. PMCID: PMC6433466. Epub 2018/09/18. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1803955&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 3. 3.McNeil JJ, Wolfe R, Woods RL, Tonkin AM, Donnan GA, Nelson MR, et al. Effect of Aspirin on Cardiovascular Events and Bleeding in the Healthy Elderly. N Engl J Med. 2018 Oct 18;379(16):1509–18. PubMed PMID: 30221597. PMCID: PMC6289056. Epub 2018/09/18. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 4. 4.McNeil JJ, Woods RL, Nelson MR, Reid CM, Kirpach B, Wolfe R, et al. Effect of Aspirin on Disability- free Survival in the Healthy Elderly. N Engl J Med. 2018 Oct 18;379(16):1499–508. PubMed PMID: 30221596. PMCID: PMC6426126. Epub 2018/09/18. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 5. 5.Ryan J, Storey E, Murray AM, Woods RL, Wolfe R, Reid CM, et al. Randomized placebo-controlled trial of the effects of aspirin on dementia and cognitive decline. Neurology. 2020 Mar 25. PubMed PMID: 32213642. Epub 2020/03/28. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibmV1cm9sb2d5IjtzOjU6InJlc2lkIjtzOjk6Ijk1LzMvZTMyMCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzEwLzE2LzIwMjAuMTAuMTEuMjAyMTA5NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 6. 6.Rasmussen KL, Tybjaerg-Hansen A, Nordestgaard BG, Frikke-Schmidt R. Absolute 10-year risk of dementia by age, sex and APOE genotype: a population-based cohort study. CMAJ. 2018 Sep 4;190(35):E1033–E41. PubMed PMID: 30181149. PMCID: PMC6148649. Epub 2018/09/06. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiY21haiI7czo1OiJyZXNpZCI7czoxMjoiMTkwLzM1L0UxMDMzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTAvMTYvMjAyMC4xMC4xMS4yMDIxMDk2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7.Qian J, Wolters FJ, Beiser A, Haan M, Ikram MA, Karlawish J, et al. APOE-related risk of mild cognitive impairment and dementia for prevention trials: An analysis of four cohorts. PLoS Med. 2017 Mar;14(3):e1002254. PubMed PMID: 28323826. PMCID: PMC5360223. Epub 2017/03/23. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1002254&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28323826&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 8. 8.Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH, Pericak-Vance MA, Joo SH, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology. 1993 Aug;43(8):1467–72. PubMed PMID: 8350998. Epub 1993/08/01. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibmV1cm9sb2d5IjtzOjU6InJlc2lkIjtzOjk6IjQzLzgvMTQ2NyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzEwLzE2LzIwMjAuMTAuMTEuMjAyMTA5NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 9. 9.van der Lee SJ, Wolters FJ, Ikram MK, Hofman A, Ikram MA, Amin N, et al. The effect of APOE and other common genetic variants on the onset of Alzheimer’s disease and dementia: a community-based cohort study. Lancet Neurol. 2018 May;17(5):434–44. PubMed PMID: 29555425. Epub 2018/03/21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29555425&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 10. 10.Albrecht MA, Szoeke C, Maruff P, Savage G, Lautenschlager NT, Ellis KA, et al. Longitudinal cognitive decline in the AIBL cohort: The role of APOE epsilon4 status. Neuropsychologia. 2015 Aug;75:411–9. PubMed PMID: 26102189. Epub 2015/06/24. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26102189&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 11. 11.De Jager PL, Shulman JM, Chibnik LB, Keenan BT, Raj T, Wilson RS, et al. A genome-wide scan for common variants affecting the rate of age-related cognitive decline. Neurobiol Aging. 2012 May;33(5):1017 e1–15. PubMed PMID: 22054870. PMCID: PMC3307898. Epub 2011/11/08. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22054870&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 12. 12.Wisdom NM, Callahan JL, Hawkins KA. The effects of apolipoprotein E on non-impaired cognitive functioning: a meta-analysis. Neurobiol Aging. 2011 Jan;32(1):63–74. PubMed PMID: 19285755. Epub 2009/03/17. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neurobiolaging.2009.02.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19285755&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000285406000007&link_type=ISI) 13. 13.Reas ET, Laughlin GA, Bergstrom J, Kritz-Silverstein D, Barrett-Connor E, McEvoy LK. Effects of APOE on cognitive aging in community-dwelling older adults. Neuropsychology. 2019 Mar;33(3):406–16. PubMed PMID: 30730162. PMCID: PMC6513314. Epub 2019/02/08. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1037/neu0000501&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30730162&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 14. 14.Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nature genetics. 2009;41(10):1088. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.440&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19734902&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000270330400011&link_type=ISI) 15. 15.Hollingworth P, Harold D, Sims R, Gerrish A, Lambert J-C, Carrasquillo MM, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nature genetics. 2011;43(5):429. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.803&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21460840&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000289972600011&link_type=ISI) 16. 16.Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Abeta, tau, immunity and lipid processing. Nat Genet. 2019 Mar;51(3):414–30. PubMed PMID: 30820047. PMCID: PMC6463297. Epub 2019/03/02. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-019-0358-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30820047&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 17. 17.Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013 Dec;45(12):1452–8. PubMed PMID: 24162737. PMCID: PMC3896259. Epub 2013/10/29. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.2802&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24162737&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 18. 18.Naj AC, Jun G, Beecham GW, Wang L-S, Vardarajan BN, Buros J, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nature genetics. 2011;43(5):436–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.801&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21460841&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 19. 19.Escott-Price V, Sims R, Bannister C, Harold D, Vronskaya M, Majounie E, et al. Common polygenic variation enhances risk prediction for Alzheimer’s disease. Brain. 2015 Dec;138(Pt 12):3673–84. PubMed PMID: 26490334. PMCID: PMC5006219. Epub 2015/10/23. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awv268&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26490334&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 20. 20.Sleegers K, Bettens K, De Roeck A, Van Cauwenberghe C, Cuyvers E, Verheijen J, et al. A 22-single nucleotide polymorphism Alzheimer’s disease risk score correlates with family history, onset age, and cerebrospinal fluid Abeta42. Alzheimers Dement. 2015 Dec;11(12):1452–60. PubMed PMID: 26086184. Epub 2015/06/19. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26086184&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 21. 21.Chouraki V, Reitz C, Maury F, Bis JC, Bellenguez C, Yu L, et al. Evaluation of a Genetic Risk Score to Improve Risk Prediction for Alzheimer’s Disease. J Alzheimers Dis. 2016 Jun 18;53(3):921–32. PubMed PMID: 27340842. PMCID: PMC5036102. Epub 2016/06/25. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27340842&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 22. 22.Tan CH, Hyman BT, Tan JJX, Hess CP, Dillon WP, Schellenberg GD, et al. Polygenic hazard scores in preclinical Alzheimer disease. Ann Neurol. 2017 Sep;82(3):484–8. PubMed PMID: 28940650. PMCID: PMC5758043. Epub 2017/09/25. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.25029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28940650&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 23. 23.Cruchaga C, Del-Aguila JL, Saef B, Black K, Fernandez MV, Budde J, et al. Polygenic risk score of sporadic late-onset Alzheimer’s disease reveals a shared architecture with the familial and early-onset forms. Alzheimers Dement. 2018 Feb;14(2):205–14. PubMed PMID: 28943286. PMCID: PMC5803427. Epub 2017/09/26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2017.08.013&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28943286&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 24. 24.Desikan RS, Fan CC, Wang Y, Schork AJ, Cabral HJ, Cupples LA, et al. Genetic assessment of age- associated Alzheimer disease risk: Development and validation of a polygenic hazard score. PLoS Med. 2017 Mar;14(3):e1002258. PubMed PMID: 28323831. PMCID: PMC5360219. Epub 2017/03/23. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1002258&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28323831&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 25. 25.Marden JR, Mayeda ER, Walter S, Vivot A, Tchetgen EJT, Kawachi I, et al. Using an Alzheimer’s Disease polygenic risk score to predict memory decline in black and white Americans over 14 years of follow-up Running head: AD polygenic risk score predicting memory decline. Alzheimer disease and associated disorders. 2016;30(3):195. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/WAD.0000000000000137&link_type=DOI) 26. 26.Harris SE, Davies G, Luciano M, Payton A, Fox HC, Haggarty P, et al. Polygenic risk for Alzheimer’s disease is not associated with cognitive ability or cognitive aging in non-demented older people. Journal of Alzheimer’s Disease. 2014;39(3):565–74. 27. 27.Chaudhury S, Brookes KJ, Patel T, Fallows A, Guetta-Baranes T, Turton JC, et al. Alzheimer’s disease polygenic risk score as a predictor of conversion from mild-cognitive impairment. Translational psychiatry. 2019;9(1):1–7. 28. 28.Verhaaren BF, Vernooij MW, Koudstaal PJ, Uitterlinden AG, van Duijn CM, Hofman A, et al. Alzheimer’s disease genes and cognition in the nondemented general population. Biol Psychiatry. 2013 Mar 1;73(5):429–34. PubMed PMID: 22592056. Epub 2012/05/18. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22592056&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 29. 29.Lourida I, Hannon E, Littlejohns TJ, Langa KM, Hypponen E, Kuzma E, et al. Association of Lifestyle and Genetic Risk With Incidence of Dementia. JAMA. 2019 Jul 14. PubMed PMID: 31302669. PMCID: PMC6628594. Epub 2019/07/16. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2019.9879&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31302669&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 30. 30.Licher S, Ahmad S, Karamujic-Comic H, Voortman T, Leening MJG, Ikram MA, et al. Genetic predisposition, modifiable-risk-factor profile and long-term dementia risk in the general population. Nat Med. 2019 Sep;25(9):1364–9. PubMed PMID: 31451782. PMCID: PMC6739225. Epub 2019/08/28. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31451782&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 31. 31.Leonenko G, Sims R, Shoai M, Frizzati A, Bossu P, Spalletta G, et al. Polygenic risk and hazard scores for Alzheimer’s disease prediction. Ann Clin Transl Neurol. 2019 Mar;6(3):456–65. PubMed PMID: 30911569. PMCID: PMC6414493 some SNPs identified as associated with AD; VE-P - personal fees from Consultancy, outside the submitted work. GL, RS, MS, AF, PB, GS, and NF have nothing to report. Epub 2019/03/27. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30911569&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 32. 32.Zheng X, Levine D, Shen J, Gogarten SM, Laurie C, Weir BS. A high-performance computing toolset for relatedness and principal component analysis of SNP data. Bioinformatics. 2012 Dec 15;28(24):3326–8. PubMed PMID: 23060615. PMCID: PMC3519454. Epub 2012/10/13. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bts606&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23060615&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000312105300026&link_type=ISI) 33. 33.Genomes Project C, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, et al. A global reference for human genetic variation. Nature. 2015 Oct 1;526(7571):68–74. PubMed PMID: 26432245. PMCID: PMC4750478. Epub 2015/10/04. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature15393&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26432245&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 34. 34.Das S, Forer L, Schonherr S, Sidore C, Locke AE, Kwong A, et al. Next-generation genotype imputation service and methods. Nat Genet. 2016 Oct;48(10):1284–7. PubMed PMID: 27571263. PMCID: PMC5157836. Epub 2016/08/30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3656&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27571263&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 35. 35.Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7. PubMed PMID: 25722852. PMCID: PMC4342193. Epub 2015/02/28. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13742-015-0047-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25722852&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 36. 36.Ruiz A, Dols-Icardo O, Bullido MJ, Pastor P, Rodríguez-Rodríguez E, de Munain AL, et al. Assessing the role of the TREM2 p. R47H variant as a risk factor for Alzheimer’s disease and frontotemporal dementia. Neurobiology of aging. 2014;35(2):444. e1-. e4. 37. 37.Meister R, Schaefer C. Statistical methods for estimating the probability of spontaneous abortion in observational studies--analyzing pregnancies exposed to coumarin derivatives. Reprod Toxicol. 2008 Sep;26(1):31–5. PubMed PMID: 18620043. Epub 2008/07/16. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.reprotox.2008.06.006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18620043&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom) 38. 38.Allignol A, Schumacher M, Beyersmann J. Empirical transition matrix of multi-state models: the etm package. Journal of Statistical Software. 2011;38(4):1–15. 39. 39.Team RC. R: A language and environment for statistical computing. 2013. 40. 40.Peloso GM, Beiser AS, Satizabal CL, Xanthakis V, Vasan RS, Pase MP, et al. Cardiovascular health, genetic risk, and risk of dementia in the Framingham Heart Study. Neurology. 2020 Jul 20. PubMed PMID: 32690788. Epub 2020/07/22. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibmV1cm9sb2d5IjtzOjU6InJlc2lkIjtzOjExOiI5NS8xMC9lMTM0MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzEwLzE2LzIwMjAuMTAuMTEuMjAyMTA5NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 41. 41.Huq AJ, Fransquet P, Laws SM, Ryan J, Sebra R, Masters CL, et al. Genetic resilience to Alzheimer’s disease in APOE ε4 homozygotes: A systematic review. Alzheimer’s & Dementia. 2019;15(12):1612–23. 42. 42.Belloy ME, Napolioni V, Han SS, Le Guen Y, Greicius MD, Alzheimer’s Disease Neuroimaging I. Association of Klotho-VS Heterozygosity With Risk of Alzheimer Disease in Individuals Who Carry APOE4. JAMA Neurol. 2020 Apr 13. PubMed PMID: 32282020. PMCID: PMC7154955. Epub 2020/04/14. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32282020&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F10%2F16%2F2020.10.11.20210963.atom)