You are on page 1of 45

NeuroToxicology 61 (2017) 143–187

Contents lists available at ScienceDirect

NeuroToxicology

Full Length Article

Risk factors associated with the onset and progression of Alzheimer’s


disease: A systematic review of the evidence
Mona Hersia,b,* , Brittany Irvinea , Pallavi Guptaa , James Gomesa,c,d , Nicholas Birketta,b ,
Daniel Krewskia,b,e
a
McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada
b
School of Epidemiology, Public Health and Preventive Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
c
Environmental Health Research Unit, University of Ottawa, ON, Canada
d
Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, ON, Canada
e
Risk Sciences International, Ottawa, Ontario Canada

A R T I C L E I N F O A B S T R A C T

Article history:
Received 22 March 2017 A systematic review was conducted to identify risk factors associated with the onset and progression of
Accepted 22 March 2017 Alzheimer’s disease (AD). Moderate and high quality systematic reviews were eligible for inclusion.
Available online 29 March 2017 Primary studies reporting on non-genetic risk factors associated with neuropathologically or clinically
confirmed AD were considered. Eighty one systematic reviews reporting on AD onset and 12 reporting on
Keywords: progression satisfied the eligibility criteria. Four hundred and thirty-two relevant primary studies
Alzheimer’s disease reporting on onset were identified; however, only those published between 2010 and 2012 (n = 65) were
Dementia included in the qualitative synthesis. Several factors including statins, light-to-moderate alcohol
Etiology
consumption, compliance with a Mediterranean diet, higher educational attainment, physically and
Systematic review
cognitively stimulating activities, and APOE e2 appeared to be associated with a decreased risk of AD
Onset
Progression onset. The evidence was suggestive of an increased risk of AD associated with head injury in males, age,
Risk factors diabetes mellitus, conjugated equine estrogen use with medroxyprogesterone acetate, current smoking,
and lower social engagement. With respect to genetic factors, APOE e4 remained the strongest predictor
of AD. Physical and cognitive activities were associated with a beneficial effect on cognitive function and
other indicators of dementia progression while higher educational attainment was associated with faster
cognitive decline. Although suggestive of an association, the current evidence for a majority of the
identified putative factors for AD onset and progression was weak, at best due to conflicting findings
across studies or inadequate evidence. Further research is required to confirm the etiological or
protective role of a number of risk factors.
© 2017 Published by Elsevier B.V.

1. Introduction Short- and long-term memory loss represent the most pronounced
cognitive marker of AD, with non-cognitive indicators encompass-
Alzheimer’s disease (AD) is the most common form of dementia ing declining physical capacity and alterations in behaviour
(Lindsay and Anderson, 2003). Dementia is characteristic of a (Gauthier et al., 2005; Aglukkaq, 2011). Characterized by debili-
number of conditions including, but not limited to, AD (Public tating symptoms which can dramatically impact quality of life, AD
Health Agency of Canada, 2010; National Advisory Council on usually results in death within a decade post- diagnosis (Aglukkaq,
Aging, 2004). AD involves irreparable neuronal degeneration 2011). Data suggests that one in ten elderly Canadians had
which gives rise to a myriad of clinically detectable neurological dementia in 2011 with 60–70% of these cases attributed to AD
impairments (Parihar and Hemnani, 2004; Gauthier et al., 2005). (Aglukkaq, 2011). Compared to AD occurrence in 2008, a more than
two-fold increase in AD incidence and prevalence is anticipated by
2038 (Alzheimer Society, 2010).
Despite the abundance of AD research, the etiology of this
* Corresponding author. Present address: Ottawa Hospital Research Institute, The
neurological disease is not yet understood (National Advisory
Ottawa Hospital, Ottawa, ON, Canada.
E-mail address: mhersi@ohri.ca (M. Hersi). Council on Aging, 2004). The objective of the study was to

http://dx.doi.org/10.1016/j.neuro.2017.03.006
0161-813X/© 2017 Published by Elsevier B.V.
144 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

systematically search the current scientific literature to identify factors impacting cognitive and non-cognitive (such as behaviour,
risk factors associated with the onset and progression of AD. physical function, quality of life, mortality) indicators of AD
progression were considered.
2. Methods Primary studies were eligible for inclusion if they reported on
non-genetic risk factors associated with the development of AD.
This review was conducted in accordance to the methodological Only studies which diagnosed AD using neuropathologic exami-
details outlined in the accompanying methods paper (Hersi et al., nation or according to the Diagnostic and Statistical Manual of
2016). The systematic review of risk factors associated with disease Mental Disorders (DSM), the Neurological Disorders and Stroke-
onset involved two phases: (1) an umbrella review of systematic Alzheimer Disease and Related Disorders Association (NINCDS-
reviews (i.e., a systematic search of the literature to identify ADRDA), or the International Classification of Diseases (ICD)
existing systematic reviews); (2) a systematic search of the criteria were considered. Studies reporting solely on non-AD
literature to identify primary studies. For the systematic review of dementias as well as those failing to distinguish AD from all-cause
factors associated with disease progression, only an umbrella dementia or dementia subtypes were excluded. Studies reporting
review of systematic reviews was conducted. on the effect of psychological stress were excluded from the
present report and have been reviewed elsewhere (Hersi, 2015).
2.1. Search methods
2.3. Study screening and data extraction
Separate search strategies were designed to identify: (1)
systematic reviews reporting on risk factors for disease onset; Studies were screened according to the process described by
(2) systematic reviews reporting on risk factors for disease Hersi et al. (2016). For systematic reviews reporting on disease
progression; and (3) primary studies reporting on risk factors onset, data were extracted by one reviewer. A second reviewer
for disease onset. independently extracted data from a randomly selected 10%
Systematic reviews reporting on onset-related factors were sample of reviews. Disagreements were addressed through
identified through a search of electronic databases and grey consensus. Due to limited resources, data from primary studies
literature sources including MEDLINE and MEDLINE In-Process & and systematic reviews reporting on AD progression was extracted
Other Non-Indexed Citations, PubMed, EMBASE, CINAHL, HuGE- by a single reviewer only.
NET, PsychINFO, TOXNET Toxicology Data Network, AARP Ageline,
Cochrane Database of Systematic Reviews, Database of Abstracts of 2.4. Quality appraisal
Reviews of Effects (DARE), AlzGene, ProQuest Digital Dissertations
and Theses database, Google and GoogleScholar. Disease-specific The quality of systematic reviews was assessed according to the
journals including the American Journal of Alzheimer’s Disease AMSTAR criteria (Shea et al., 2009). For systematic reviews
and Other Dementias (1986-December 31, 2011) and the Interna- reporting on AD onset, quality assessment was conducted by
tional Journal of Alzheimer’s Disease (2009-December 31, 2011) one reviewer. A second reviewer independently assessed the
were also searched. Where possible, database specific search quality of a randomly selected 10% sample of reviews. Quality
filters, such as those developed by InterTASC Information Special- assessment of reviews reporting on AD progression was conducted
ists’ Sub-Group and McMaster University’s Health Information entirely by a single reviewer. Low quality reviews with an AMSTAR
Research Unit, were also applied to identify systematic reviews and score of 3 or less were excluded from the qualitative synthesis. The
citations reporting on risk factors associated with onset or quality of primary studies was not evaluated.
progression. The reference lists of a sample of retrieved articles
were reviewed for potentially missed studies. The initial search 2.5. Literature search results
was conducted in September 2011. The OVID AutoAlerts feature
was enabled to ensure that newly indexed relevant articles were The study selection process is outlined in Figs. 1–3 . A total of
identified. The AutoAlerts feature was disabled on December 31, 136 systematic reviews reporting on risk factors associated with
2011. Databases without an alerts feature were re-searched until disease onset (n = 121) and progression (n = 15) were identified. Of
the end of December 2011. The search for systematic reviews on AD these, 93 were deemed to be of high quality and were included in
progression was restricted to MEDLINE and EMBASE and was the qualitative syntheses for disease onset (n = 81) and progression
conducted in March 2013. (n = 12). A list of excluded systematic reviews on onset and
Primary studies reporting on risk factors associated with AD progression, along with reasons for exclusion, can be found in
onset were identified through a search of electronic databases Supplementary material I. One moderate quality review (Daviglus
including MEDLINE and MEDLINE In-Process & Other Non-Indexed et al., 2011) was later excluded from the overview of AD onset as it
Citations, EMBASE, CINAHL and AgeLine. The search was conducted was a manuscript version of an identified Health Technology
in July 2012. The search for risk factors for AD progression was Assessment (HTA) (Williams et al., 2010). Two reviews were also
limited to systematic reviews. All search strategies are provided in later excluded from the overview of AD progression (Isaac et al.,
Supplementary material I. 2008; Aguirre et al., 2013). The review by Isaac et al. (2008) was
excluded because an updated version (Farina et al., 2012) of the
2.2. Eligibility criteria review was identified. The review by Aguirre et al. (2013) was
excluded as it was a duplicate of a Cochrane review (Woods et al.,
For the first phase of the review, systematic reviews reporting 2012). Details regarding the quality appraisal of each systematic
on risk factors associated with AD onset or progression were review on onset and progression are also provided in Supplemen-
eligible for inclusion. French and English language reviews were tary material I.
eligible if a computerized database search was conducted and the A total of 432 relevant primary studies reporting on disease
eligibility criteria of the review were explicitly stated. Reviews onset were identified. The breadth of the primary study literature
which failed to distinguish AD from all-cause dementia or precluded inclusion of all 432 studies. As such, only the most
dementia subtypes were excluded for the systematic review of recent studies (those published from 2010 to 2012) were selected
disease onset. An exception was made for reviews reporting on for inclusion in the qualitative synthesis (n = 65). A list of included
progression. Systematic reviews examining non-pharmacological primary studies is provided in Supplementary material I. Five
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 145

studies, which examined the risk of AD associated with stress, these studies reaching statistical significance. The pooled analysis
were excluded from the present review and were reviewed detected a statistically significant 39% increased risk of AD among
separately (Hersi, 2015). diabetics (RR = 1.39; 95% CI = 1.17, 1.66). Results of the review and
meta-analysis suggest that individuals with diabetes have a
3. Results significantly higher risk of developing Alzheimer's disease.
Profenno et al. (2010) identified eight longitudinal studies
3.1. Risk factors associated with AD onset which reported on the association between DM and risk of AD. Two
additional studies, which reported on the effect of hyperinsuline-
3.1.1. Diabetes mellitus mia without diabetes and borderline diabetes, were also included.
Eight moderate quality systematic reviews reported on the Three of the eight studies reporting on DM detected a statistically
association between diabetes mellitus (DM) and risk of AD significant association. The remaining five studies also found that
(Cukierman et al., 2005; Biessels et al., 2006; Weih et al., 2007; risk of AD was higher among diabetics, however, the results failed
Patterson et al., 2007; Kloppenborg et al., 2008; Lu et al., 2009; to reach statistical significance. The random effects meta-analysis
Profenno et al., 2010; Williams et al., 2010). Table 1 in detected a statistically significant 54% increased risk of AD (effect
Supplementary material II provides an overview of the character- size (ES) = 1.54; 95% CI = 1.33, 1.79; I2 = 3.6%, p = 0.405) associated
istics of these studies and their findings. with DM. When all ten studies were combined, DM, hyper-
Our search for recent primary studies yielded eleven studies insulinemia, and borderline diabetes were associated with a
which reported on diabetes or plasma glucose levels (Ronnemaa statistically significant increased risk of AD (ES = 1.61; 95% CI = 1.40,
et al., 2011; Cheng et al., 2011; Ohara et al., 2011; Kimm et al., 2011; 1.86). Results of the subgroup analysis restricted to studies (n = 5)
Song et al., 2011; Verdelho et al., 2010; N.C. Li et al., 2010a,b; Qiu which adjusted for the potentially confounding effects of body
et al., 2010; Reitz et al., 2010b; Schrijvers et al., 2010; Ahtiluoto mass index (BMI), other vascular risk factors, and stroke yielded
et al., 2010). Table 90 in Supplementary material II provides an consistent findings (ES = 1.64; 95% CI = 1.29, 2.09). Overall, results of
overview of the characteristics of these studies and their findings. the review suggest that diabetes was associated with an increased
risk of AD.
3.1.1.1. Systematic reviews. The earliest systematic review Two systematic reviews (Biessels et al., 2006; Lu et al., 2009)
reporting on DM identified six longitudinal studies (Cukierman and two cohort studies were eligible for inclusion in the HTA
et al., 2005). All six studies reported an increased risk of AD among prepared for the US Agency for Healthcare Research and Quality
diabetics, however, only results from two studies reached (Williams et al., 2010). Results from both systematic reviews
statistical significance. The reviewers concluded that diabetes included in the report suggest that DM increases risk of AD. One of
appears to increase risk of dementia and AD. the two identified cohort studies reported an increased risk of AD
The systematic review reported by Biessels et al. (2006) associated with DM, however, results failed to reach statistical
identified 13 longitudinal cohort studies, 11 of which examined the significance (hazard ratio (HR) = 1.62; 95% CI = 0.98, 2.67). While
association between late-life DM and risk of AD. Nine of the eleven the second study failed to detect an association between diagnosed
studies examining late-life DM reported risk estimates exceeding diabetes and AD, both borderline (HR = 1.87; 95% CI = 1.11, 3.14) and
1.00. Furthermore, results from six of these studies reached undiagnosed diabetes (HR = 3.29; 95% CI = 1.20, 9.01) were
statistical significance with reported risk estimates ranging from associated with a statistically significant increased risk of AD.
1.4 to 2.4. Of the two studies reporting on midlife DM ( < 60 years), Based on the available evidence, the reviewers concluded that DM
one detected a statistically significant increased risk of AD (odds appears to increase the risk of AD.
ratio (OR) = 4.40; p < 0.01) while the other failed to detect an
association (relative risk (RR) = 1.00; 95% CI = 0.50, 2.00). Based on 3.1.1.2. Primary studies. Diabetes was not significantly associated
these findings, the reviewers concluded that diabetes increases with risk of AD according to results of the LADIS prospective cohort
risk of AD. study (HR = 1.34; 95% CI = 0.35, 5.14; p = 0.668) and the Canadian
Seven relevant cohort studies were included in the review Study of Health and Aging (OR = 0.64; 95% CI = 0.39, 1.03) (Verdelho
reported by Weih et al. (2007). Three studies consistently reported et al., 2010; Song et al., 2011). Reitz et al. (2010b) also failed to
a statistically significant increased risk of AD among subjects with detect an association between DM and risk of late-onset AD
diabetes. Four of the remaining studies reported risk estimates (LOAD).
exceeding 1.00, however, the results failed to achieve statistical Six studies detected a significant or marginally significant
significance. The reviewers concluded that diabetes may increase association between DM and AD. Diabetics were at a significantly
risk of AD. increased risk of developing AD according to the Vantaa 85+ study
Patterson et al. (2007) identified two pertinent studies. Both (HR = 2.45; 95% CI = 1.33, 4.53), Kungsholmen project (HR = 1.52;
studies reported risk estimates greater than 1.00, although, both 95% CI = 1.03, 2.25), and the Hisayama Study (HR = 2.05; 95%
failed to reach statistical significance. CI = 1.18, 3.57) (Ahtiluoto et al., 2010; Qiu et al., 2010; Ohara et al.,
Six of the 11 longitudinal studies identified by Kloppenborg 2011). Participants of the Washington Heights-Inwood Columbia
et al. (2008) reported a significant or marginally significant Aging Project (Cheng et al., 2011) reporting a history of type 2
increased risk of AD associated with DM. Two studies reported on diabetes had an 80% increased risk of LOAD according to the
the effect of impaired glucose metabolism. The studies reported minimally adjusted model (HR = 1.80; 95% CI = 1.20, 2.70). Kimm
conflicting results with one reporting an increased risk of AD et al. (2011) detected an increased risk of AD among male
attributed to impaired glucose metabolism while the other failed (HR = 1.60; 95% CI = 1.30, 2.00) and female (HR = 1.40; 95% CI = 1.10,
to detect an association between abnormal glucose levels and AD. 1.70) diabetics. Finally, diabetes was only marginally associated
The reviewers concluded that diabetes was associated with an with an increased risk of incident AD according to results reported
increased risk of AD, vascular dementia, and all-cause dementia. by Li et al. (2010a,b) (HR = 1.04; 95% CI = 1.00, 1.08; p = 0.049).
A total of eight prospective cohort studies investigating the Ohara et al. (2011) utilized data from the Hisayama Study, a
association between diabetes and Alzheimer's disease were longitudinal cohort study of Japanese subjects, to examine the
eligible for inclusion in the systematic review and meta-analysis association between glucose tolerance and risk of AD and other
reported by Lu et al. (2009). Seven of the eight identified studies cognitive outcomes. The study detected an increased risk of AD
reported risk estimates exceeding 1.00 with results from two of associated with higher levels of 2-h post-load glucose but not with
146 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Fig. 1. Screening process for systematic reviews reporting on risk factors for AD onset.

fasting plasma glucose or the remaining individual categories of ascertainment (Biessels et al., 2006; Williams et al., 2010). More
glucose tolerance (Table 90, Supplementary material II). specifically, studies varied with respect to the methods used to
Higher levels of insulin and insulin resistance, but not glucose, ascertain diabetes as well as the timing of the exposure
were associated with an increased risk of AD within three years of measurement (i.e., midlife vs. late-life) (Biessels et al., 2006;
follow-up in the Rotterdam Study (Schrijvers et al., 2010). Williams et al., 2010).
However, insulin and insulin resistance were not associated with We identified nine primary studies which reported on diabetes
risk of AD after three years of follow-up. Fasting glucose remained mellitus and three studies which reported on the effect of high
not associated with AD at subsequent follow-up periods. fasting glucose levels. Six (Ahtiluoto et al., 2010; Qiu et al., 2010;
The Uppsala Longitudinal Study of Adult Men (Ronnemaa et al., Ohara et al., 2011; Cheng et al., 2011; Kimm et al., 2011; Li et al.,
2011) failed to detect an association between higher levels of 2010a,b) of the nine studies reporting on DM detected a significant
fasting plasma glucose, ascertained in mid- and late-life, and or marginally significant association with risk of AD, although
development of AD (Midlife  HR = 1.00; 95% CI = 0.80, 1.20. Late-life results failed to reach statistical significance in the fully adjusted
 HR = 1.00; 95% CI = 0.80, 1.30). model in one study (Cheng et al., 2011). Of the studies reporting on
higher fasting plasma glucose levels, all three failed to detect an
3.1.1.3. Discussion. Evidence summarized in eight moderate- association with AD (Ohara et al., 2011; Schrijvers et al., 2010;
quality systematic reviews is suggestive of an association Ronnemaa et al., 2011). However, one study (Ohara et al., 2011)
between DM and risk of AD. Although a majority of the studies detected an increased risk of AD associated with higher levels of 2-
reported an increased risk of AD associated with AD, results from h post-load glucose and one (Schrijvers et al., 2010) reported that
some studies failed to reach statistical significance. Inconsistent elevated insulin and insulin resistance were associated with an
findings across studies may be attributed to sources of increased short- but not long-term risk of AD.
methodological heterogeneity with respect to exposure
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 147

Fig. 2. Screening process for primary studies reporting on risk factors for AD onset.

3.1.2. Obesity, body mass index, body composition reporting on BMI treated as a continuous variable, only one
Our search for systematic reviews yielded eight moderate reported a statistically significant increased risk of AD with
quality (Weih et al., 2007; Kloppenborg et al., 2008; Profenno et al., increasing BMI.
2010; Williams et al., 2010; Beydoun et al., 2008; Lee et al., 2010; The HTA reported by Williams et al. (2010) identified one
Anstey et al., 2011; Gorospe and Dave, 2007) and three low quality systematic review and three recent cohort studies. Results from
reviews (Pitner, 2005; Chang, 2008; Barnes and Yaffe, 2011) that the systematic review (Beydoun et al., 2008) suggested an
reported on the association between obesity and body mass index increased risk of AD associated with obesity. The three cohort
(BMI) with risk of AD. studies reported a decreased risk of AD associated with late-life
Six primary studies were identified in the second phase of the obesity or overweight BMI. Considered cumulatively, however, the
review (Chen et al., 2010; Reitz et al., 2010b; Ronnemaa et al., 2011; reviewers concluded that obesity did not appear to be associated
Xu et al., 2011; Abellan van Kan et al., 2012; Gelber et al., 2012). The with risk of AD.
study characteristics and findings are summarized in Tables 2 and Three systematic reviews reported a quantitative synthesis of
92 in Supplementary material II. identified evidence. According to the pooled analysis (n = 4)
reported by Beydoun et al. (2008), obesity was associated with
3.1.2.1. Systematic reviews. The evidence summarized in the a marginally significant increased risk of AD (RR = 1.80; 95%
systematic review reported by Weih et al. (2007) is suggestive CI = 1.00, 3.29). The meta-analysis of six longitudinal studies
of an increased risk of AD attributable to obesity, overweight BMI, reported by Profenno et al. (2010) yielded a risk estimate of 1.59
and increasing BMI. Two of three studies, which examined the risk (95% CI = 1.02, 2.48; I2 = 65.5%, p = 0.005). The series of pooled
of AD associated with each unit increase in BMI, reported risk analyses reported by Anstey et al. (2011) detected an increased risk
estimates above 1.00. One study detected an increased risk of AD of AD associated with low (RR = 1.96; 95% CI = 1.32, 2.92; I2 = 69.1%),
associated with obesity (BMI >30 kg/m2) and being overweight overweight (RR = 1.35; 95% CI = 1.19, 1.54; I2 = 92.0%), and obese
(BMI 25–30 kg/m2). (RR = 2.04; 95% CI = 1.59, 2.62; I2 = 82.8%) midlife BMI. Analyses of
Gorospe and Dave (2007) identified a single study which studies reporting on late-life BMI, treated categorically (RR = 1.46;
specifically reported on the risk of developing AD. The study 95% CI = 0.97, 2.21; I2 = 42.3%) and continuously (RR = 0.97; 95%
reported no association between AD and midlife BMI. CI = 0.92, 1.02; I2 = 75.8%) failed to detect an association with AD.
Kloppenborg et al. (2008) identified five relevant longitudinal Anstey et al. (2011) concluded that underweight, overweight, and
studies. There was no association between midlife and late-life obese midlife BMI were all associated with an increased risk of AD.
obesity with risk of developing AD according to two studies. Three In contrast, late-life BMI and obesity were not associated with AD.
studies examined late-life BMI as a continuous variable, with
conflicting findings. One study found that each unit increase in BMI 3.1.2.2. Primary studies. Four studies reported on the effect of BMI
was associated with an increased risk of AD, one reported that on risk of AD. While low (<18.5 kg/m2) BMI was consistently
increasing BMI was protective against the development of AD, and associated with an increased risk of AD among males and females
one study failed to detect an association. across the lifespan, there was no evidence of an association
Lee et al. (2010) identified four longitudinal studies that between overweight/obese (24 kg/m2) BMI and the risk of AD in
reported on the effect of obesity, overweight BMI, and increasing the case-control study reported by Chen et al. (2010). Similarly, the
BMI on AD risk. One of two studies, which reported on the effect of Honolulu-Asia Aging Study and the Uppsala Longitudinal Study of
high BMI, detected a statistically significant increased risk of AD Adult Men failed to detect an association, between AD and higher
among obese and overweight subjects. Of the two studies tertiles of midlife BMI and increasing mid- and late-life BMI
148 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Fig. 3. Screening process for systematic reviews reporting on risk factors for AD progression.

respectively (Gelber et al., 2012; Ronnemaa et al., 2011). The pooled analysis reported in the most recent review (Anstey et al.,
remaining study, based on the Swedish Twin Registry (Xu et al., 2011) failed to detect an association. However, the meta-analysis
2011), reported an increased risk of AD associated with both reported by Anstey et al. (2011) was limited by a small number of
overweight (25–30 kg/m2 OR = 1.91; 95% CI = 1.30, 2.80) and obese identified studies.
( > 30 kg/m2 OR = 3.43; 95% CI = 1.49, 7.90) midlife BMI. When Of the four observational studies reporting on BMI identified in
treated as a continuous variable, BMI was associated with an the second phase of our review, one examined BMI in early life
increased risk of AD (OR = 1.06; 95% CI = 1.01, 1.10). (Chen et al., 2010), four ascertained BMI in midlife (Ronnemaa
The EPIDOS-Toulouse (Abellan van Kan et al., 2012) cohort et al., 2011; Gelber et al., 2011; Xu et al., 2011; Chen et al., 2010) and
study found no association between total lean mass (OR = 0.57; 95% two examined late-life BMI (Ronnemaa et al., 2011; Chen et al.,
CI = 0.24, 1.34) or total fat mass (OR = 2.12; 95% CI = 0.90, 4.99) and 2010). Only one (Xu et al., 2011) of the four studies reporting on
AD. High waist to hip ratio (WHR) was significantly associated with midlife BMI detected an association between higher or increasing
the risk of probable and possible late-onset AD (LOAD) according to BMI and increased risk of AD. Another study (Chen et al., 2010)
results reported by Reitz et al. (2010b) (HR = 2.63; 95% CI = 1.39, detected an increased risk of AD associated with low BMI in early,
4.97). Results were similar when the analysis was restricted to mid-, and late-life. Both studies reporting on the effect of high or
probable LOAD (HR = 3.12; 95% CI = 1.54, 6.33). increasing late-life BMI failed to detect an association. Finally,
while higher waist to hip ratio predicted risk of AD in one study
3.1.2.3. Discussion. Evidence summarized in eight moderate- (Reitz et al., 2010b), late-life total lean mass and total fat mass were
quality systematic reviews suggests that the association not associated with risk of AD in another (Abellan van Kan et al.,
between obesity or higher BMI and risk of AD is inconclusive. 2012).
While some reviews reported evidence of an increased risk of AD
attributed to obesity or higher BMI in midlife, others concluded 3.1.3. Hypertension
that there was no association. The evidence pertaining to late-life Six systematic reviews were identified. Two reviews were
obesity and risk of AD was also conflicting. While recent cohort excluded due to low methodological quality (Purnell et al., 2009;
studies identified by Williams et al. (2010) were suggestive of a Barnes and Yaffe, 2011). Results from one high-quality (Power et al.,
decreased risk of AD associated with higher late-life BMI, the 2011) and three moderate-quality reviews (Patterson et al., 2007;
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 149

Kloppenborg et al., 2008; Williams et al., 2010) were included in increased risk of AD associated with elevated midlife DBP,
the qualitative synthesis. In the second phase of the review, we however, results failed to reach statistical significance. Three
identified nine cohort studies (Ronnemaa et al., 2011; Kimm et al., studies reported on the effect of late-life SBP and DBP. The
2011; Song et al., 2011; Verdelho et al., 2010; Qiu et al., 2010; Reitz reviewers noted the lack of association between categories of SBP
et al., 2010b; Yang et al., 2011; Joas et al., 2012; Chu et al., 2010). The measured in late-life and AD. There was some evidence of an
study characteristics and findings are summarized in Tables 3 and association between higher DBP and a reduced risk of AD. Overall,
91 in the Supplementary material II. results of the review suggested that the association between BP
and risk of AD remained inconclusive (Power et al., 2011).
3.1.3.1. Systematic reviews. Five longitudinal cohort studies
reporting on the effect of blood pressure on AD risk were 3.1.3.2. Primary studies. According to results of the LADIS
eligible for inclusion in the review reported by Patterson et al. prospective cohort study (HR = 0.71; 95% CI = 0.27, 1.90;
(2007). Two studies reported an association between systolic p = 0.499) and the Canadian Study of Health and Aging
hypertension, defined as systolic blood pressure (SBP) greater than (OR = 1.00; 95% CI = 0.75, 1.32), hypertension was not associated
180 mm Hg, and an increased risk of AD. Two studies detected an with risk of AD (Verdelho et al., 2010; Song, Mitnitski, and
increased risk of AD attributed to low (<140 mm Hg) or declining Rockwood, 2011). Similarly, Reitz et al. (2010b) failed to detect an
systolic blood pressure. Low diastolic blood pressure (DBP; association between hypertension and risk of possible and
<65 mm Hg) was also detrimental according to one study. probable LOAD (HR = 1.16; 95% CI = 0.64, 2.11). Results remained
Finally, a single study reported an increased risk of AD consistent when the analysis was restricted to probable LOAD
associated with both high (>84 mm Hg) and low (< 70 mm Hg) (HR = 1.03; 95% CI = 0.54, 1.95). Self-reported hypertension was not
pulse pressure (PP). Overall, results from this review suggest that associated with risk of AD according to results reported by Yang
high and low systolic blood pressure, low diastolic pressure, and et al. (2011) (HR = 0.82; 95% CI = 0.58, 1.15; P = 0.251). The only study
both low and high pulse pressure may increase risk of AD. to detect an association between hypertension and risk of AD
Kloppenborg et al. (2008) identified twelve studies examining reported a detrimental effect associated with stage 2 hypertension
the risk of AD associated with blood pressure. All four studies (HR = 1.40; 95% CI = 1.10, 1.80) among Korean men (Kimm et al.,
reporting on the risk of AD associated with hypertension failed to 2011). Both pre-hypertension (120–139 mmHg systolic and/or 80–
detect an association. Of the five studies reporting on the effect of 89 mmHg diastolic) and stage 1 hypertension (140–159 mmHg
high SBP, one detected an increased risk, one reported a protective systolic/90–99 mmHg diastolic), defined according to the Joint
effect, and three failed to detect an association. Four of five studies National Committee (JNC 7) criteria, were not associated with AD
reporting on high DBP failed to detect an association. The in men. Alternatively, all three categories of hypertension were not
remaining study reported a greater than four-fold increase in associated with risk of AD in the sample of Korean women (see
the risk of AD among those with high DPB. Overall, four of 12 Table 91 in the Supplementary material II).
included longitudinal studies detected a statistically significant Midlife and late-life SBP, treated as a continuous variable, were
association between measures of blood pressure and risk of AD. not associated with risk of AD in the Uppsala Longitudinal Study of
Three of the four studies reported an increased risk of AD Adult Men (Ronnemaa et al., 2011). Yang et al. (2011) also failed to
associated with high or increasing blood pressure while the detect an association between SBP and risk of AD in a longitudinal
remaining study detected a protective effect. analysis (HR = 1.00; 95% CI = 0.99, 1.00; P = 0.294). In contrast, two
Of the ten cohort studies included in the HTA reported by studies detected a detrimental effect attributed to higher SBP. Each
Williams et al. (2010), results from only three studies, which unit increase in SBP was associated with an increased risk of AD
examined two cohorts, detected a statistically significant increased (RR = 1.04; 95% CI = 1.01, 1.08) in an all-male sample of Chinese
risk of AD associated with hypertension. More specifically, two of subjects (Chu et al., 2010). Furthermore, the Kungsholmen project
seven studies detected an increased risk of AD associated with high (Qiu et al., 2010) detected an increased risk of AD attributed to high
SBP. With respect to diastolic blood pressure, only one of seven SBP (140–159 mm Hg  HR = 1.47; 95% CI = 1.02, 2.12. 160 mm Hg
studies reported an increased risk of AD associated with high DBP.  HR = 1.84; 95% CI = 1.06, 3.18).
The remaining studies failed to detect an association. The The effect of DBP was examined in two studies. Using moderate
reviewers concluded that the evidence was suggestive of a lack diastolic pressure (70–94 mm Hg) as a referent, low ( < 70 mm Hg
of association between hypertension and AD.  HR = 1.83; 95% CI = 1.26, 2.65) but not high (95 mm Hg 
Eighteen studies were included in the review reported by Power HR = 0.78; 95% CI = 0.54, 1.12) DBP was significantly associated with
et al. (2011). History of hypertension was not associated with the AD risk in the Kungsholmen project (Qiu et al., 2010). Higher DBP in
risk of AD according to the random effects meta-analysis of ten late-life was marginally associated with risk of AD (HR = 0.98; 95%
studies (RR = 0.98; 95% CI = 0.80, 1.19; I2 = 42%; 95% CI = 0%, 71%). CI = 0.97, 1.00; P = 0.030) according to the longitudinal analysis
According to the pooled analysis of four studies, a 10 mm HG- reported in another study (Yang et al., 2011).
increase in SBP was associated with a marginally significant 5% Results from the Kungsholmen project (Qiu et al., 2010)
reduced risk of AD (RR = 0.95; 95% CI = 0.91, 1.00. I2 = 0%; 95% CI = 0%, suggested that low ( < 70 mm Hg  HR = 1.50; 95% CI = 1.07, 2.12)
68%). A 10 mm HG-increase in DBP was not significantly associated but not high (85 mm Hg  HR = 1.19; 95% CI = 0.83, 1.69) pulse
with risk of AD (RR = 0.94; 95% CI = 0.85, 1.04. I2 = 14%; 95% CI = 0%, pressure was significantly associated with AD when compared
72%). When the analysis was restricted to the three studies that with the middle tertile (70–84 mm Hg). The longitudinal analysis
reported on increasing late-life blood pressure, results remained presented by Yang et al. (2011) failed to detect an association
unchanged. The only study to report on increasing midlife blood between pulse pressure (HR = 1.00; 95% CI = 0.99, 1.01; P = 0.919) or
pressure failed to detect an association between AD and a 10 mm mean arterial pressure (HR = 0.99; 95% CI = 0.98, 1.00; P = 0.060)
HG increase in both SBP (RR = 1.03; 95% CI = 0.80, 1.32) and DBP and risk of AD.
(RR = 1.16; 95% CI = 0.75; 1.81). Among studies that examined the Using data from the Prospective Population Study of Women,
effect of blood pressure treated as a categorical variable, two of Joas et al. (2012) examined the association between blood pressure
three failed to detect an association between midlife SBP and risk “trajectories”, or changes in blood pressure from midlife to late-
of AD. The remaining study detected an increased risk of AD life, and the development of dementia and AD. “All AD” included all
associated with the highest category of SBP (160 mm Hg) subjects diagnosed with AD according to the NINCDS-ADRDA
(RR = 2.60; 95% CI = 1.10, 6.60). All three studies reported an criteria while “pure AD” included subjects with “no
150 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

cerebrovascular or other disease that might contribute to A total of four longitudinal studies were eligible for inclusion in
dementia” (Joas et al., 2012). There was no difference between the systematic review reported by Kloppenborg et al. (2008). The
those who developed AD and those who did not with respect to three studies that reported risk estimates comparing high total
baseline SBP (Pure AD: 0.16 mm Hg; 95% CI = 3.82, 4.14; p = 0.94. serum cholesterol to normal levels reported conflicting findings.
All AD: 2.69 mm Hg; 95% CI = 0.95, 6.33; p = 0.15). Compared to One study failed to detect an association, one study reported a
those who did not develop dementia, there was a greater increase greater than two-fold increased risk of AD associated with high
in SBP between 1968 and 1992 for those who developed pure AD total cholesterol, and the remaining study detected a reduced risk
(Pure AD: 0.32 mm Hg; 95% CI = 0.07, 0.56; p = 0.01. All AD: 0.20 mm of AD among those with high total cholesterol levels. Heterogene-
Hg (95% CI = 0.01, 0.42; p = 0.06) and a greater decrease from 1992 ity across studies, particularly pertaining to the timing of exposure
onwards for both “pure” and “all” AD. ascertainment, was cited as a likely source of the inconsistent
findings (Kloppenborg et al., 2008). The only study which
3.1.3.3. Discussion. According to the results of four moderate/high ascertained exposure at midlife reported an increased risk of AD
quality systematic reviews, the association between hypertension associated with high total cholesterol. Two studies reporting on
and risk of AD is unclear. A majority of studies captured in the late-life total cholesterol failed to detect an association or reported
reviews failed to detect an association between hypertension and a decreased risk of AD associated with higher cholesterol levels.
risk of AD. There is some evidence to suggest that the association The only study to report on total cholesterol considered continu-
between hypertension and AD may be dependent on age (Power ously, detected a 2% increased risk of AD associated with increasing
et al., 2011). While some studies have reported an increased risk of total cholesterol (OR = 1.02; p = 0.03). Lastly, both studies reporting
AD associated with midlife hypertension, results from other on triglycerides failed to detect an association.
studies have detected a protective effect of late-life hypertension Eight prospective cohort studies, four of which examined total
against the development of AD. However, the observed risk cholesterol measured at midlife and five of which examined the
reduction attributed to late-life hypertension is likely due to effect of late-life total cholesterol (TC), were identified in the
sources of bias (Power et al., 2011). More specifically, selection bias review reported by Anstey et al. (2008). Of the four studies
due to losses to follow-up, short lengths of follow-up, confounding, examining midlife total cholesterol, two detected an increased risk
and exposure/outcome misclassification error were all noted as attributed to high (6.5 mmol/l) cholesterol, one reported a
important limitations of primary studies included by Power et al. detrimental effect of decreasing cholesterol levels from mid- to
(2011). late-life, and one failed to detect an association between increasing
Of the five identified primary studies reporting on hyperten- (per 10 mg/dL) total cholesterol and risk of AD. Two of five studies
sion, only one (Kimm et al., 2011) detected an association with risk reporting on late-life TC failed to detect an association. Three of the
of AD, namely, an increased risk among hypertensive males. Two remaining studies, which examined the effect of increasing
studies (Qiu et al., 2010; Chu et al., 2010) reported an increased risk quartiles of late-life TC, were combined in a fixed effects meta-
of AD associated with higher SBP, one (Yang et al., 2011) reported a analysis. The pooled analysis failed to detect an association
protective effect associated with higher DBP in late-life, one (Qiu between AD and increasing quartiles of late-life TC (Second
et al., 2010) reported a detrimental effect associated with low DBP, quartile vs. first quartile  RR = 0.85; 95% CI = 0.67, 1.10. Third
one (Qiu et al., 2010) detected an increased risk associated with quartile vs. first quartile  RR = 1.03; 95% CI = 0.79, 1.35. Fourth
low pulse pressure, and one (Joas et al., 2012) reported differences quartile vs. first quartile  RR = 0.85; 95% CI = 0.65, 1.12). Based on
in blood pressure trajectories from midlife to late-life among the identified evidence, the reviewers concluded that while
subjects with AD as compared to those who did not develop elevated midlife TC was associated with increased risk of AD,
dementia. elevated late-life TC was not.
The HTA reported by Williams et al. (2010) relied solely on the
3.1.4. Cholesterol, hypercholesterolemia findings of the systematic review by Anstey et al. (2008). It was
The effect of cholesterol levels was examined in seven concluded that elevated midlife TC, but not elevated late-life TC,
systematic reviews. Two reviews were of low methodological was associated with an increased risk of AD.
quality and were excluded (Miller and Chacko, 2004; Purnell et al.,
2009). Findings from the remaining five moderate-quality reviews 3.1.4.2. Primary studies. The prospective cohort study reported by
were considered (Weih et al., 2007; Patterson et al., 2007; Reitz et al. (2010a) examined the risk of AD associated with total
Kloppenborg et al., 2008; Anstey et al., 2008; Williams et al., cholesterol, high-density lipoprotein cholesterol (HDL-C), non-
2010). The second phase of the review yielded five relevant cohort high-density lipoprotein cholesterol (Non-HDL-C), and LDL-C. Risk
studies (Reitz et al., 2010a,b; Mielke et al., 2010; Ronnemaa et al., estimates were reported for probable AD and for the combined
2011; Kimm et al., 2011). outcome of possible and probable AD. According to the results of
the fully adjusted model, higher HDL-C was associated with a
3.1.4.1. Systematic reviews. Eight cohort studies reporting on the decreased risk of probable and possible AD; however, only results
association between serum cholesterol and risk of AD were eligible for the highest quartile reached statistical significance (Quartile 2
for inclusion in the review reported by Weih et al. (2007). Two  HR = 0.80; 95% CI = 0.40, 1.50. Quartile 3  HR = 1.10; 95% CI = 0.60,
studies reported that serum cholesterol levels exceeding 6.5 mol/l 1.90. Quartile 4 HR = 0.40; 95% CI = 0.20, 0.90). Analyses of higher
were associated with an approximate three-fold increased risk of total cholesterol, non-HDL-C, and LDL-C levels yielded risk
AD. However, conflicting results were reported in one study that estimates below 1.00; however, results failed to achieve
detected a decreased risk of AD associated with elevated serum statistical significance. Similar results were obtained when
cholesterol. The remaining five studies failed to detect an probable AD was analyzed separately.
association between AD and elevated low-density lipoprotein Low HDL-C was not significantly associated with possible and
cholesterol (LDL-C) (n = 1) or total serum cholesterol (n = 4). probable LOAD (HR = 1.60; 95% CI = 0.77, 3.32) according to Reitz
Both studies included in the review by Patterson et al. (2007) et al. (2010b). Results were similar when the analysis was
detected an increased risk of AD attributed to high midlife serum restricted to probable LOAD (HR = 1.58; 95% CI = 0.71, 3.54).
cholesterol. Based on these findings, the reviewers concluded that Midlife serum cholesterol was not associated with risk of AD
elevated serum cholesterol in midlife may increase risk of AD. after 32 years of follow-up according to the Prospective Population
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 151

Study of Women (>6.5 mmol/l vs. less: HR = 1.48; 95% CI = 0.73, the study suggest that each 1 standard deviation (SD) increase in
2.96) (Mielke et al., 2010). log transformed serum homocysteine at baseline was associated
The study reported by Ronnemaa et al. (2011) examined the risk with risk of AD (RR: 1.80; 95% CI = 1.30, 2.50).
of AD associated with vascular risk factors considered individually The single cohort study included in the review by Patterson
and in combination. Mid- and late-life serum cholesterol, treated et al. (2007) detected a greater than two-fold (RR = 2.11; 95%
as a continuous variable, was not associated with risk of AD CI = 1.19, 3.76) increased risk of AD among those with serum
(Midlife HR = 1.00; 95% CI = 0.90, 1.20. Late-life HR = 1.10; 95% homocysteine levels greater than 15 mmol/L.
CI = 0.90, 1.30). Four community-based cohort studies were identified in the
The association between vascular risk factors and risk of AD HTA by Williams et al. (2010). Three of the four included cohort
among Koreans was prospectively examined by Kimm et al. (2011). studies reported a statistically significant increased risk of AD
Using normal (<5.17 mmol/l) cholesterol as the referent category, associated with elevated homocysteine levels. The remaining
both borderline (HR = 1.20; 95% CI = 1.00, 1.40) and high (HR = 1.20; study reported an increased risk associated with elevated
95% CI = 1.00, 1.50) cholesterol were not significantly associated homocysteine, although results failed to reach statistical signifi-
with risk of AD in men. Similarly, borderline and high cholesterol cance. Results from the three studies that reported on the risk of
did not increase risk of AD in women (Borderline cholesterol AD associated with plasma homocysteine levels exceeding 14 or
HR = 1.00; 95% CI = 0.90, 1.20. High cholesterol HR = 1.10; 95% 15 mmol/L were pooled. According to the findings of the random
CI = 0.90, 1.30). effects meta-analysis, elevated plasma homocysteine levels were
not associated with a statistically significant increased risk of AD
3.1.4.3. Discussion. Five moderate-quality systematic reviews (HR = 1.53; 95% CI = 0.94, 2.49; Q-statistic: 6.378, p = 0.04,
examining the effect of high serum cholesterol on the risk of AD I2 = 68.6%). Results of the pooled analysis were limited by the
were identified. While two earlier systematic reviews concluded small number of eligible studies as well as statistical heterogeneity
that the association between cholesterol and AD was inconclusive, across studies (Williams et al., 2010). Based on these findings, the
conflicting findings across studies was likely due to heterogeneity reviewers concluded that the available evidence did not support an
with respect to the timing of exposure ascertainment (Weih et al., increased risk of AD associated with elevated homocysteine
2007; Kloppenborg et al., 2008). The systematic review by Anstey concentrations.
et al. (2008), which stratified studies based on timing of exposure Four cohort studies were identified and included in the review
ascertainment, concluded that elevated midlife total cholesterol, reported by Dangour et al. (2010). Three of the four included
but not late-life cholesterol, was associated with an increased risk studies reported an increased risk of AD associated with elevated
of AD. Findings of the identified systematic reviews were limited homocysteine levels. The remaining study failed to detect an
by sources of bias in the included observational studies. Firstly, the association between elevated plasma homocysteine concentra-
observational studies failed to control for the potentially tions and risk of AD. Based on the findings of this review, the
confounding effect of lipid lowering medications (Anstey et al., reviewers concluded that there was some evidence to suggest that
2008). Secondly, although a majority of the included studies there is an increased risk of AD associated with elevated serum/
adjusted for other potentially confounding variables, variability in plasma homocysteine concentrations.
the degree of confounding adjustment across the included studies A total of 14 studies identified by Ho et al. (2011) investigated
may have given rise to methodological heterogeneity (Anstey et al., the association between homocysteine levels and AD. Data from 12
2008). Overall, evidence summarized in the identified systematic studies were combined in a random effects meta-analysis. The
reviews suggested that elevated midlife total serum cholesterol quantitative analysis yielded a pooled standardized mean differ-
was associated with an increased risk of AD. However, the analysis ence of 0.59 (95% CI = 0.38, 0.80; I2 = 79.1%) suggesting that subjects
of midlife cholesterol reported by Anstey et al. (2008) should be with AD had significantly higher homocysteine levels compared to
interpreted cautiously due to the small number of included studies. controls. Four studies identified in the review examined the
With respect to the primary studies identified in the second association between elevated homocysteine and subsequent risk
phase of the review, three (Ronnemaa et al., 2011; Reitz et al., of dementia and cognitive decline. However, only one of these
2010a,b) reported on late-life serum cholesterol and two prospective studies focused on the outcome of AD. Results of this
ascertained cholesterol in mid-life (Ronnemaa et al., 2011; Mielke study suggest that elevated homocysteine (>15 mmol/L) was not
et al., 2010). All four cohort studies reporting on mid- and late-life associated with a statistically significant increased risk of AD
total serum cholesterol failed to detect a statistically significant (OR = 1.60; 95% CI = 0.70, 3.50). Overall, results of this systematic
association. Only one (Reitz et al., 2010a) of two (Reitz et al., 2010a, review and meta-analysis suggest that in comparison to controls,
b) studies, which specifically reported on the effect of late-life subjects with AD had significantly higher homocysteine levels. The
HDL–C, detected an association with AD. When evidence from only identified prospective study failed to detect an association.
systematic reviews and primary studies were considered cumula-
tively, the association between elevated serum cholesterol and risk 3.1.5.2. Primary studies. Results from the Cardiovascular Risk
of AD was unclear. Factors, Aging, and Dementia (CAIDE) study suggest that both
increasing homocysteine (OR per 1 mmol/L = 1.19; 95% CI = 1.01,
3.1.5. Homocysteine 1.39) and holotranscobalamin (OR per 1 pmol/l: 0.977 = 95%
Seven systematic reviews were identified (Reutens and CI = 0.958, 0.997) were marginally associated with AD
Sachdev, 2002; Kuo et al., 2005a; Weih et al., 2007; Patterson (Hooshmand et al., 2010). However, homocysteine was no
et al., 2007; Williams et al., 2010; Ho et al., 2011; Dangour et al., longer significantly associated with AD when the analysis was
2010). Two systematic reviews were of low methodological quality adjusted for holotranscobalamin (OR = 1.10; 95% CI = 0.96, 1.25). The
and were excluded (Reutens and Sachdev, 2002; Kuo et al., 2005a). authors concluded that the findings were suggestive of a possible
The search for primary studies yielded one record (Hooshmand association between homocysteine and holotranscobalamin with
et al., 2010). risk of AD.

3.1.5.1. Systematic reviews. The systematic review reported by 3.1.5.3. Discussion. Five systematic reviews, collating evidence
Weih et al. (2007) identified a single cohort study which examined from six cohort (Blasko et al., 2008; Luchsinger et al., 2004;
the association between homocysteine and risk of AD. Results of Ravaglia et al., 2005; Seshadri et al., 2002; Annerbo et al., 2006;
152 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Kim et al., 2008), six case-control (Clarke et al., 1998; Koseoglu and longitudinal study design. All four studies reported an increased
Karaman, 2007; Bottiglieri et al., 2001; Religa et al., 2003; risk of AD associated with low ABI (< 0.90); however, results from
Näggaet al., 2003; Hogervorst et al., 2002), and five cross- two studies failed to reach statistical significance and one was only
sectional (Folin et al., 2005; Haan et al., 2007; Quadri et al., marginally significant. Based on the available evidence, the
2004; Storey et al., 2003; Miller et al., 2002) studies were identified reviewers concluded that low ABI, an indicator of cardiovascular
in the present review. Four of the six cohort studies reported an disease, predicted risk of AD.
increased risk of AD associated with elevated homocysteine levels
(Blasko et al., 2008; Ravaglia et al., 2005; Seshadri et al., 2002; 3.1.8. Stroke
Annerbo et al., 2006). The remaining two cohort studies also Two systematic reviews (Patterson et al., 2007; Purnell et al.,
reported risk estimates exceeding 1.00, although the results failed 2009), one of which was excluded for low methodological quality,
to reach statistical significance (Luchsinger et al., 2004; Kim et al., reported on the effect of stroke on the risk of AD. We identified four
2008). The only primary study (Hooshmand et al., 2010) identified relevant primary studies in the second phase of the review (Song
in the second phase of the review detected a marginally significant et al., 2011; Verdelho et al., 2010; Li et al., 2010a,b; Qiu et al., 2010).
association between homocysteine and risk of AD. However,
results failed to reach statistical significance upon adjustment for 3.1.8.1. Systematic reviews. One relevant longitudinal cohort study
holotranscobalamin. was identified by Patterson et al. (2007). According to the results of
Collectively, there is some evidence to suggest that elevated the study, stroke was associated with an 83% increased risk of AD
homocysteine levels may increase the risk of AD. However, (RR = 1.83; 95% CI = 1.14, 2.95).
conflicting findings across primary studies and reviews precludes
a definitive statement. The systematic reviews were limited by the 3.1.8.2. Primary studies. According to results of the LADIS
potential sources of bias affecting the included observational prospective cohort study, previous stroke (HR = 0.22; 95%
studies. Firstly, a number of observational studies failed to adjust CI = 0.05, 0.98; p = 0.051) and stroke during follow-up (HR = 1.35;
for factors which may confound the association between 95% CI = 0.32, 5.70; p = 0.679) were not significantly associated with
homocysteine and AD, namely, vitamin B supplementation, risk of AD (Verdelho et al., 2010). Similarly, the Kungsholmen
smoking status, inactive lifestyle, increasing age, high coffee and project (HR = 1.20; 95% CI = 0.77, 1.89) and Canadian Study of Health
alcohol consumption, and medical conditions including hypothy- and Aging (OR = 0.69; 95% CI = 0.35, 1.36) failed to detect an
roidism and rheumatoid arthritis (Williams et al., 2010; Ho et al., association between stroke and risk of AD (Qiu et al., 2010; Song
2011; Kuo et al., 2005a). Secondly, a majority of the identified et al., 2011). The remaining study reported a two-fold increased
observational studies were case-control or cross-sectional studies, risk of incident AD associated with stroke (HR = 2.00; 95% CI = 1.93,
which are susceptible to reverse causality (Ho et al., 2011). 2.07; p < 0.001) (Li et al., 2010a,b).
Considered cumulatively, the association between elevated
homocysteine and risk of AD was inconclusive. 3.1.8.3. Discussion. There was conflicting evidence pertaining to
the association between stroke and risk of AD. The single cohort
3.1.6. Metabolic syndrome study identified in the systematic review by Patterson et al. (2007)
The effect of metabolic syndrome on risk of AD was examined in suggested an increased risk of AD associated with stroke. Three of
two moderate-quality systematic reviews (Williams et al., 2010; four primary studies identified in the second phase of the review
Hao et al., 2011a,b). The search for primary studies did not yield any failed to detect an association between stroke and AD. Considered
relevant reports. cumulatively, the association between stoke and risk of AD was
inconclusive.
3.1.6.1. Systematic reviews. Two prospective cohort studies were
identified and included in the HTA reported by Williams et al. 3.1.9. Atrial fibrillation
(2010). Both studies failed to detect an association. The reviewers One systematic review (Purnell et al., 2009) was identified that
note that the results of the systematic review were limited by reported on the association between atrial fibrillation and risk of
heterogeneous criteria for exposure ascertainment and the small AD. However, the review was excluded due to low methodological
number of studies available for analysis. quality. Two cohort studies reporting on the risk of AD associated
The systematic review by Hao et al. (2011a,b) examined the with atrial fibrillation were identified (Marengoni et al., 2011;
effect of metabolic syndrome on the risk of dementia. A total of Dublin et al., 2011).
nine relevant prospective cohort studies, three of which reported History of chronic atrial fibrillation did not increase risk of AD
on the risk of developing AD, were eligible for inclusion. All three according to findings from the Kungsholmen Project (Marengoni
studies failed to detect an association between metabolic et al., 2011), a prospective cohort study conducted in Sweden
syndrome and risk of AD. Overall, results of this review suggested (HR = 0.80; 95% CI = 0.40, 1.50).
that metabolic syndrome was not associated with the AD risk. Results from the Adult Changes in Thought study (Dublin et al.,
2011) yielded conflicting results, suggesting an increased risk of AD
3.1.6.2. Discussion. Based on findings from two moderate-quality associated with history of atrial fibrillation (HR = 1.50; 95% CI = 1.16,
systematic reviews, which collated evidence from three cohort 1.94). The risk estimate was attenuated but remained statistically
studies, metabolic syndrome was not associated with risk of AD. significant based on the analysis with the most rigorous
Further research to confirm the association between the metabolic adjustment for potential confounders (HR = 1.41; 95% CI = 1.07,
syndrome and risk of AD is warranted. 1.86).

3.1.7. Ankle Brachial Index (ABI) 3.1.10. Cardiovascular disease


One moderate-quality systematic review examined the associ- Four observational studies were identified that reported on the
ation between low ankle brachial index (ABI), an indicator of broad category of cardiovascular disease (CVD) and certain non-
lower-extremities peripheral artery disease, and risk of dementia stroke subtypes (Li et al., 2010a,b; Qiu et al., 2010; Eriksson et al.,
(Guerchet et al., 2011). A total of 12 studies, four of which examined 2010a; Song et al., 2011) (see Table 95 in Supplementary material
risk of AD, were included in the review. One of the four studies was II.)
cross-sectional while the remaining three studies employed a
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 153

3.1.10.1. Primary studies. According to results reported by Li et al. for trend was statistically significant for all three risk profiles
(2010a,b), cardiovascular disease was not associated with incident indicating that increasing number of vascular factors were
AD (HR = 0.99; 95% CI = 0.96, 1.02; p = 0.505). Similarly, the associated with increasing AD risk.
Canadian Study of Health and Aging failed to detect an Reitz et al. (2010b) developed a summary risk score, based on
association between heart disease and AD (OR = 0.94; 95% vascular factors, to estimate the likelihood of developing LOAD.
CI = 0.69, 1.28) (Song et al., 2011). The Kungsholmen project Sex, age, diabetes, hypertension, current smoking, low HDL-C, high
detected an increased risk of AD associated with heart failure waist to hip ratio, education, ethnicity, and APOE e4 allele were
(HR = 1.54; 95% CI = 1.05, 2.25) (Qiu et al., 2010). considered. Using risk scores between 0 and 14 as a referent, higher
Eriksson et al. (2010a) examined the effect of non-stroke risk scores were associated with greater risk of probable and
cardiovascular disease and myocardial infarction on AD risk. possible LOAD (15–18, HR = 3.74; 95% CI = 1.42, 9.88. 19–22,
According to the longitudinal analysis, there was no association HR = 3.55; 95% CI = 1.31, 9.62. 23–28, HR = 12.57; 95% CI = 5.26,
between non-stroke cardiovascular disease and risk of AD (First 3 30.08. >28, HR = 20.47; 95% CI = 8.38, 49.99). Similar results were
years after CVD, HR = 1.48; 95% CI = 0.83, 2.64. More than 3 years obtained when the analysis was restricted to risk of probable LOAD.
after CVD, HR = 0.67; 95% CI = 0.37, 1.21). However, the analysis The study reported by Ronnemaa et al. (2011) examined the risk
stratified by ApoE4 genotype detected an increased risk of AD of AD associated with vascular risk factors considered individually
associated with CVD among ApoE4 carriers (HR = 2.39; 95% and in combination. Participants enrolled in the Uppsala Longitu-
CI = 1.15, 4.96) but not among non-carriers (HR = 0.76; 95% dinal Study of Adult Men were examined at two baseline
CI = 0.24, 2.42). There was no association between myocardial assessments. Participants were assessed at midlife (average
infarction (MI) alone and risk of AD (First 3 years after MI, age = 49.6 years) and followed for an average of 29.0 years. Subjects
HR = 0.91; 95% CI = 0.35, 2.35. More than 3 years after MI, HR = 0.64; who did not develop dementia by age 70 (average age of late-life
95% CI = 0.27, 1.53). The co-twin control analysis failed to detect an assessment = 71.0 years) were enrolled in the late-life baseline
association between CVD and MI with AD (see Table 95 in assessment and followed for an average of 13.0 years. Systolic
Supplementary material II). blood pressure, fasting plasma glucose, serum cholesterol, body
mass index, and current smoking, measured in midlife and late-
3.1.10.2. Discussion. Of the four identified observational studies life, were not associated with risk of AD. Subjects possessing the
reporting on cardiovascular disease and its non-stroke subtypes, APOE e4 allele were at an increased risk of AD. To examine the
only two detected a positive association. More specifically, results effect of multiple risk factors, subjects with one, two, three or more
of the Kungsholmen project suggest an increased risk of AD existing risk factors, which included current smoking, systolic
associated with heart failure (Qiu et al., 2010). The second study blood pressure >140 mmHg, BMI >28, fasting serum glucose
provided preliminary evidence suggesting an increased risk of AD >7.0 mmol/l, and serum cholesterol >7.0 mmol/l, were compared
associated with non-stroke CVD in ApoE4 carriers (Eriksson et al., to subjects free of these factors. According to the results of the
2010a). The remaining two studies failed to detect an association study, having one or more vascular risk factors at midlife and late-
between CVD and risk of AD. life did not increase risk of AD.

3.1.11. Multiple vascular risk factors 3.1.11.2. Discussion. Two of the three identified studies reported
We did not identify any systematic reviews that examined the that the presence of multiple vascular risk factors (Qiu et al., 2010)
effect of multiple, concurrent vascular risk factors. Three or a higher vascular risk score (Reitz et al., 2010b) could predict the
(Ronnemaa et al., 2011; Qiu et al., 2010; Reitz et al., 2010b) development of AD. The remaining study failed to detect an
relevant cohort studies were identified in the second phase of the association between multiple vascular factors and risk of
review. subsequent AD. Further research is required to clarify the effect
of multiple vascular risk factors on the risk of developing AD.
3.1.11.1. Primary studies. Using data from the Kungsholmen
project, Qiu et al. (2010) examined the risk of AD attributed to 3.1.12. B-type Natriuretic Peptide (BNP)
systolic and diastolic blood pressure, pulse pressure, diabetes and A single primary study was identified which reported on the
pre-diabetes, stroke, and heart failure. The cumulative effect of the association between B-type natriuretic peptide and risk of AD.
aforementioned vascular risk factors was also examined. The According to the results reported by Kerola et al. (2010) higher
overall vascular risk profile was derived based on the number of levels of B-type natriuretic peptide were associated with risk of AD
present vascular risk factors, namely, “high systolic pressure (OR = 1.59; 95% CI = 1.09, 2.30; p = 0.015). Further research is
(160 mm Hg), low diastolic pressure ( < 70 mm Hg), low pulse required to confirm the association.
pressure (<70 mm Hg), diabetes/pre-diabetes, stroke, and heart
failure” (Qiu et al., 2010). Using absence of vascular risk factors as a 3.1.13. Antihypertensive drugs
referent, the presence of two or more vascular risk factors The effect of antihypertensive drugs on risk of AD was examined
significantly increased the risk of AD (One risk factor, HR = 1.09; in four systematic reviews (Standridge, 2004; Shah et al., 2009;
95% CI = 0.75, 1.60. Two risk factors, HR = 1.77; 95% CI = 1.16, 2.71. Williams et al., 2010; Chang-Quan et al., 2011), one of which was
Three or more risk factors, HR = 2.66; 95% CI = 1.39, 5.08). An excluded due to low methodological quality (Standridge, 2004)
atherosclerotic subtype profile was derived based on the presence (see Table 9 in the Supplementary material II). No recent primary
of high systolic pressure, diabetes/pre-diabetes, and stroke. studies were identified.
Presence of one of these factors marginally increased risk of AD
by 33% (HR = 1.33; 95% CI = 1.00, 1.78) while presence of two or 3.1.13.1. Systematic reviews. Six studies investigating the
more factors significantly increased risk of AD by two-fold association between antihypertensive use and incidence or
(HR = 2.09; 95% CI = 1.31, 3.34). Finally, a hypoperfusion risk progression of AD were eligible for inclusion in the systematic
profile was derived based on the presence of low diastolic review reported by Shah et al. (2009). Two studies reported solely
pressure, low pulse pressure, and heart failure. Presence of two on progression of AD and will not be summarized herein. All four
or more of these factors significantly increased risk of AD studies consistently reported risk estimates below 1.00, however;
(HR = 2.06; 95% CI = 1.35, 3.13), whereas presence of a single results from only two studies reached statistical significance.
factor had no significant effect (HR = 1.27; 95% CI = 0.96, 1.69). The p
154 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Eight cohort studies were identified and included in the HTA methodological design, the meta-analysis of five cohort (RR = 0.85;
reported by Williams et al. (2010). Only half of the included cohort 95% CI = 0.71, 1.03; test for heterogeneity: p = 0.76) and three case-
studies detected a statistically significant reduced risk of AD control (RR = 0.84; 95% CI = 0.61, 1.16; test for heterogeneity:
associated with antihypertensive drug use. The reviewers also p = 0.77) studies yielded consistent findings suggesting a lack of
identified a systematic review and meta-analysis restricted to association between aspirin use and risk of AD. The reviewers
randomized controlled trials (RCTs) (McGuinness et al., 2009). concluded that there was some evidence to suggest that NSAIDs,
According to a pooled analysis of three RCTs, antihypertensive drug particularly when used for longer durations, may exert a protective
use was not associated with risk of dementia (OR = 0.89; 95% effect against the development of AD.
CI = 0.69, 1.16). The pooled risk estimate was not specific to AD Szekely et al. (2004) identified seven non-prospective and four
(Williams et al., 2010). prospective studies. Of the seven non-prospective studies, four
Two randomized controlled trials and five cohort studies were studies were cross-sectional studies and three were case-control
eligible for inclusion in the systematic review reported by Chang- studies. All four cross-sectional studies reported a statistically
Quan et al. (2011). Of the seven studies included, six reported a significant reduced risk of AD among NSAID users, with odds ratios
non-significant reduced risk of AD among antihypertensive ranging from 0.29 to 0.43. In contrast, only one case-control study
medication users with risk estimates ranging from 0.55 to 0.89. reported a significant reduced risk of AD associated with NSAID use
The remaining study reported a non-significant increased risk. In (OR = 0.55; 95% CI = 0.37, 0.82). In the pooled analysis of the seven
the pooled analysis combining the results of all seven studies, non-prospective studies representing 12979 subjects, lifetime use
antihypertensive drug use was not associated with AD risk of NSAIDs was associated with a statistically significant 49%
(RR = 0.90; 95% CI = 0.79, 1.03). In the subgroup analysis stratified reduced odds of AD (OR = 0.51; 95% CI = 0.40, 0.66; Q = 5.55;
by methodological study design, antihypertensives were not p = 0.48). NSAID use was associated with a protective effect against
associated with AD in the pooled analysis of cohort studies the development of AD according to the pooled analysis of
(RR = 0.92; 95% CI = 0.79, 1.08) or RCTs (RR = 0.79; 95% CI = 0.53, prospective studies (RR = 0.74; 95% CI = 0.62, 0.89; Q = 4.27,
1.18). p = 0.23). Use of NSAIDs for two or more years was associated
with a greater risk reduction according to the pooled analysis of
3.1.13.2. Discussion. Available data from observational studies, three prospective studies (RR = 0.42; 95% CI = 0.26, 0.66; Q = 1.16,
summarized in three moderate quality systematic reviews, suggest p = 0.56). Overall, results of the review suggested that use of non-
that antihypertensive drugs do not significantly reduce risk of AD. aspirin NSAIDs was associated with a reduced risk of AD (Szekely
Although results from many of the observational studies failed to et al., 2004).
reach statistical significance, a majority of studies reported risk Eleven case-controls studies, which examined the association
estimates below 1.00 (Shah et al., 2009; Chang-Quan et al., 2011). A between NSAID use and prevalent dementia, were identified by de
meta-analysis of three RCTs failed to detect an association between Craen et al. (2005). According to the random effects meta-analysis,
antihypertensive drug use and risk of dementia (McGuinness et al., NSAID use was associated with a statistically significant 49%
2009). Heterogeneity across the included studies with respect to reduced risk of dementia (RR = 0.51; 95% CI = 0.37, 0.70; test for
antihypertensive drug dosage, study methodology, patient heterogeneity: p = 0.001). However, the results of the pooled
characteristics, and length of follow-up were cited as important analysis were not specific to AD as data from three studies that
limitations (Shah et al., 2009). Collectively, there was a lack of focused on all-cause dementia were included in the combined risk
evidence to suggest that antihypertensive drug use was associated estimate. Ten studies examined the association between NSAID use
with a protective effect against the development of AD. and incident dementia. Eight of these studies specifically reported
on the risk of AD. All eight studies reported risk estimates below
3.1.14. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) 1.00, however, only results from two studies reached statistical
Six systematic reviews, which examined the association significance. When data from the ten studies were pooled in a
between NSAID use and risk of AD, were identified (Etminan random effects meta-analysis, NSAID use was associated with a
et al., 2003; Standridge, 2004; Szekely et al., 2004; de Craen et al., 21% reduced risk of incident dementia (RR = 0.79; 95% CI = 0.68,
2005; Patterson et al., 2007; Williams et al., 2010). One low quality 0.92). When stratified by methodological design, NSAID use was
review was excluded (Standridge, 2004). A single primary study associated with a statistically significant reduced risk of incident
was identified in the second phase of the review (Breitner et al., dementia according to the pooled analysis of case-control studies
2011). (RR = 0.77; 95% CI = 0.61, 0.96) and only a marginally significant
reduced risk among cohort studies (RR = 0.79; 95% CI = 0.61, 1.01).
3.1.14.1. Systematic reviews. Six cohort and four case-control Overall, results of this review suggested that NSAID use was
studies investigating the association between NSAIDs and/or associated with a reduced risk of dementia and AD. However, the
aspirin use and AD were included in the systematic review reviewers cautioned that the observed protective effect may have
reported by Etminan et al. (2003). The pooled analysis of nine been attributed to sources of bias.
studies suggested a 28% reduced risk of AD associated with NSAID Patterson et al. (2007) identified a single study reporting on
use (RR = 0.72; 95% CI = 0.56, 0.94; test for heterogeneity: p = 0.06). NSAIDs. According to the results of the longitudinal cohort study,
Analyses stratified by methodological design detected an NSAID use was associated with a 58% (RR = 0.42; 95% CI = 0.26, 0.66)
association among case-control (RR = 0.62; 95% CI = 0.45, 0.82; reduced risk of AD.
test for heterogeneity: p = 0.55) but not cohort (RR = 0.84; 95% One systematic review (Szekely et al., 2004), four cohort
CI = 0.54, 1.05; test for heterogeneity: p = 0.04) studies. Four cohort studies, and two RCTs were identified and included in the HTA
studies provided data pertaining to the length of NSAID use. Short- reported by Williams et al. (2010). Results of the pooled analysis
and intermediate-term NSAID use was not associated with risk of restricted to cohort studies, conducted by Szekely et al. (2004),
AD. Long term use, defined as greater than 12 months in one study suggest that NSAID use was associated with a statistically
and greater or equal to 24 months in the remaining three studies, significant reduced risk of AD. Four additional prospective studies
was associated with a reduced risk of AD (RR = 0.27; 95% CI = 0.13, not included in the review by Szekely et al. (2004), were identified
0.58). No association was detected between aspirin use and risk of by Williams et al. Aspirin was not associated with AD in all three
AD according to the pooled analysis of eight studies (RR = 0.87; 95% cohort studies. Two of the four studies failed to detect an
CI = 0.70, 1.07; test for heterogeneity: p = 0.79). When stratified by association between NSAID use and AD, and one study reported
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 155

a statistically significant reduced risk of AD associated with NSAID the ADAPT trial update identified in the second phase of the review,
use among carriers of one or more APOE e4 alleles. The final study the association between NSAIDs and AD was inconclusive.
failed to detect an association between moderate NSAID use and
risk of AD; however, heavy use was associated with a statistically 3.1.15. Cholinesterase inhibitors
significant increased risk of AD (Williams et al., 2010). Findings Three moderate-quality systematic reviews examined the
from the four cohort studies were combined with the four cohort association between cholinesterase inhibitors (ChEIs) and risk of
studies identified by Szekely et al. (2004). Six of the eight studies conversion from mild cognitive impairment (MCI) to AD (Raschetti
reported risk estimates below 1.00, two of which reached et al., 2007; Diniz et al., 2009; Williams et al., 2010). No relevant
statistical significance. According to the pooled analysis, however, primary studies were identified.
NSAID use was not associated with risk of AD (HR = 0.83; 95%
CI = 0.63, 1.09; Q statistic = 40.84. p < 0.001, I2 = 83%). However, a 3.1.15.1. Systematic reviews. A total of three published and five
sensitivity analysis excluding one study yielded statistically unpublished RCTs examining the effect of ChEIs on conversion
significant results (HR = 0.79; 95% CI = 0.69, 0.91). Findings from from MCI to AD were eligible for inclusion in the systematic review
two RCTs were also considered by Williams et al. According to one reported by Raschetti et al. (2007). All four studies consistently
study, rofecoxib (25 mg/day) increased the risk of AD (HR = 1.49; reported a higher conversion rate for the placebo group; however,
95% CI = 1.08, 2.05). The second study, by the ADAPT Research conversion rates were not significantly different between groups.
Group, reported consistent findings, suggesting an increased risk of Moreover, the relative risks of conversion from MCI to AD, which
dementia associated with NSAID use when compared with placebo could only be calculated for one published (RR = 0.84; 95% CI = 0.57,
(Celecoxib, HR = 4.11; 95% CI = 1.30, 13.0. Naproxen, HR = 3.57; 95% 1.25) and one unpublished (RR = 0.85; 95% CI = 0.64, 1.12) study,
CI = 1.09 to 11.7). Overall, relying on evidence from observational were not statistically significant. Based on the results of this
studies would suggest that the association between NSAIDs and systematic review, use of ChEIs was not associated with a delay in
risk of AD was inconclusive. However, results from two RCTs conversion from MCI to AD. Heterogeneous study populations,
suggest that use of certain NSAIDs, namely, rofecoxib, celecoxib, ambiguity of MCI diagnosis, and unavailable data for two of the
and naproxen, were associated with a deleterious effect (Williams studies included were all cited as important limitations to consider
et al., 2010). Based on the latter finding, the reviewers concluded when interpreting the results of the review (Raschetti et al., 2007).
that NSAID use may increase risk of AD. Four randomized controlled trials were eligible for inclusion in
the review by Diniz et al. (2009). All four studies were also
3.1.14.2. Primary Studies. One primary study was identified which identified and included in the earlier systematic review by
reported on the association between NSAID use and risk of AD Raschetti et al. (2007). However, Diniz et al. (2009) excluded
(Breitner et al., 2011). This study, reported by the Adapt Research two published and two unpublished studies that were included in
Group, was a follow-up to the RCT identified in the systematic the earlier review, citing “conversion from MCI to AD [was] not the
review by Williams et al. (2010). The study was an 18 to 24 month main study outcome” as the basis for exclusion. According to meta-
follow-up of subjects previously randomized to receive naproxen, analysis of the RCTs, 15.4% and 20.4% of subjects in the treated and
celecoxib, or placebo for an average of two years. Results of the placebo groups converted to AD, respectively. Cholinesterase
follow-up were inconsistent with previous results, which inhibitor use was associated with a statistically significant 24%
suggested an increased risk of AD associated with NSAID (RR = 0.76; 95% CI = 0.65, 0.88; Q = 3.40, p = 0.33) decreased risk of
treatment (Celecoxib, HR = 4.11; 95% CI = 1.30, 13.0. Naproxen, progression from MCI to AD. Overall, results of this review suggest
HR = 3.57; 95% CI = 1.09 to 11.7). More specifically, results of the that cholinesterase inhibitor use, particularly for longer durations,
follow-up failed to detect an increased risk of AD among subjects can reduce the risk of progression from MCI to AD (Diniz et al.,
randomized to naproxen (Rate ratio = 0.87; 95% CI = 0.43, 1.70) and 2009).
celecoxib (Rate ratio = 1.20; 95% CI = 0.72, 2.41). Williams et al. (2010), examined the association between
cholinesterase inhibitors and risk of AD. No relevant observational
3.1.14.3. Discussion. Five moderate quality systematic reviews studies were identified. However, the reviewers identified and
examined the effect of NSAID use and risk of AD. Results from included the systematic reviews by Raschetti et al. (2007) and
identified observational studies were meta-analyzed in four of the Diniz et al. (2009). The review by Raschetti et al. (2007) failed to
five systematic reviews. Meta-analyses of cohort studies detect an association between ChEI use and risk of conversion from
conducted by Etminan et al. (2003) and de Craen et al. (2005) MCI to AD. The meta-analysis of three RCTs by Diniz et al. (2009)
failed to reach statistical significance; however, pooled risk detected a 25% reduced risk of conversion from MCI to AD
estimates were below 1.00. The HTA report by Williams et al. associated with ChEIs. Due to the methodological limitations of the
(2010) yielded statistically significant results once the outlying aforementioned meta-analysis, Williams et al. (2010) concluded
cohort study was excluded from the meta-analysis. Finally, the that cholinesterase inhibitor use was not associated with conver-
pooled analysis of cohort studies by Szekely et al. (2004) suggested sion from MCI to AD.
a statistically significant protective effect associated with NSAID
use. As noted by Williams et al. (2010), sources of methodological 3.1.15.2. Discussion. Results from the earliest systematic review
and clinical heterogeneity across observational studies may have suggest that use of ChEIs was not associated with a delay in
contributed to inconsistent findings across primary studies and the conversion from MCI to AD (Raschetti et al., 2007). A more recent
reviews themselves. Overall, pooled analyses of cohort studies review and meta-analysis by Diniz et al. (2009) suggested that long
consistently yielded risk estimates below 1.00. Considered term ChEI use was associated with a statistically significant
cumulatively, evidence from observational studies suggest that reduced risk of progression. However, as noted by Williams et al.
NSAIDs, particularly when used for longer durations (Etminan (2010), the small number of studies and important sources of
et al., 2003), may reduce the risk of AD. methodological heterogeneity warrant cautious interpretation of
Results from RCTs, which are generally susceptible to less bias, the pooled risk estimate derived by Diniz et al. (2009).
present conflicting findings. Two RCTs identified by Williams et al. Consequently, when the available evidence was considered
(2010) detected an increased risk of AD and dementia associated cumulatively, the effect of ChEI use on AD risk among subjects
with NSAID treatment. However, a follow-up of one of the trials with MCI was inconclusive.
failed to detect an increased risk (Breitner et al., 2011). In light of
156 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

3.1.16. Hormone Replacement Therapy (HRT) Williams et al., 2010; Brinker, 2011). Three moderate quality
Three moderate systematic reviews examined the association systematic reviews examined the association between lipid
between hormone replacement therapy (HRT) and risk of AD (Yaffe lowering therapies, particularly statin use, and risk of AD
et al., 1998; LeBlanc et al., 2001; Williams et al., 2010). One low (Patterson et al., 2007; Zhou et al., 2007; Williams et al., 2010).
quality review was excluded (Standridge, 2004). No primary The remaining six low-quality studies were excluded. The search
studies were identified. for recent primary studies yielded one record (Li et al., 2010a,b)
(see Tables 13 and 100 in Supplementary material II).
3.1.16.1. Systematic reviews. Yaffe et al. (1998) identified two
prospective cohort and eight case-control studies that reported on 3.1.17.1. Systematic reviews. Four cohort and two case-control
the association between HRT use and risk of AD. Results of the studies identified by Zhou et al. (2007) examined AD as an
pooled analysis suggest that postmenopausal estrogen therapy use outcome. According to a random effects meta-analysis, statin
was associated with 29% reduced risk of AD (OR = 0.71; 95% therapy was not associated with risk of AD (OR = 0.81; 95%
CI = 0.52, 0.98). Estrogen use reduced risk of AD, albeit only CI = 0.56, 1.16; test for heterogeneity: p = 0.14).
marginally, according to the subgroup analysis restricted to studies Two longitudinal cohort studies identified by Patterson et al.
which used the widely accepted NINCDS-ADRDS criteria for (2007) examined the effect of statin use. Both studies failed to
outcome ascertainment (OR = 0.69; 95% CI = 0.48, 1.01; test for detect an association. One study examined the effect of all lipid
heterogeneity: p = 0.07). Estrogen use reduced risk of AD according lowering drugs. Results from this study suggest that use of lipid
to the pooled analysis of cohort (OR = 0.48; 95% CI = 0.29, 0.81; test lowering drugs was associated with a reduced risk of AD
for heterogeneity p = 0.86), but not case-control (OR = 0.80; 95% (OR = 0.26; 95% CI = 0.08, 0.88).
CI = 0.56, 1.16; test for heterogeneity: p = 0.11) studies. Six studies were eligible for inclusion in the HTA reported by
Two cohort and 10 case-control studies were eligible for Williams et al. (2010). Findings from these six studies were
inclusion in the systematic review reported by LeBlanc et al. included in a random effects meta-analysis, which detected a
(2001). The meta-analysis of all 12 studies suggested that HRT use statistically significant reduced risk of AD associated with statin
was associated with a statistically significant 34% reduced risk of use (HR = 0.73; 95% CI = 0.57, 0.94). There was no evidence of
AD (RR = 0.66; 95% CI = 0.53, 0.82; test for heterogeneity: p > 0.10). statistical heterogeneity (Q statistic = 5.132, p-value = 0.40;
In the sensitivity analysis stratified by methodological design, the I2 = 2.58%) or publication bias. No relevant RCTs were identified.
protective effect of HRT was more pronounced in the pooled Based on the available evidence from observational studies, the
analysis of cohort studies (Cohort, RR = 0.50; 95% CI = 0.30, 0.80. reviewers concluded that statin use appeared to reduce the risk of
Case-control, RR = 0.71; 95% CI = 0.56, 0.91). However, the protec- AD.
tive effect of HRT did not remain statistically significant when the
analysis was restricted to methodologically superior studies 3.1.17.2. Primary studies. Li et al. (2010a,b) prospectively examined
(RR = 0.64; 95% CI = 0.32, 1.06). Lastly, HRT use was associated the effect of statin therapy on risk of AD. According to the results of
with a reduced risk of AD according to the subgroup analysis the study, statin therapy was associated with a reduced risk of AD
excluding studies that ascertained HRT status using proxy (HR = 0.62; 95% CI = 0.40, 0.97). Stratified analysis by age detected a
informant data (RR = 0.72; 95% CI = 0.55, 0.96). protective effect in younger subjects and a detrimental non-
The HTA reported by Williams et al. (2010) identified one significant association in older subjects ( < 80 years of age,
systematic review and two RCTs that examined the effect of HR = 0.44; 95% CI = 0.25, 0.78. 80 years of age, HR = 1.22; 95%
estrogen therapy on the incidence of dementia and AD. Results of CI = 0.61, 2.42). Stratified analysis by APOE e4 status yielded risk
the identified review (LeBlanc et al., 2001) suggest that HRT use estimates below 1.00 for both strata, however, results failed to
may reduce the risk of AD. As noted by Williams et al. (2010), reach statistical significance for non-carriers (Carriers, HR = 0.42;
findings of the included review must be interpreted with caution 95% CI = 0.20, 0.91. Non-carriers, HR = 0.77; 95% CI = 0.42, 1.40.
due to variability across studies with respect to the types and Unknown, HR = 0.84; 95% CI = 0.22, 3.15) (Li et al., 2010a,b).
duration of HRT use. Both identified RCTs provided risk estimates
for all-cause dementia, which encompasses but is not restricted to 3.1.17.3. Discussion. Two of the three identified systematic
AD (Williams et al., 2010). According to the results of these trials, reviews failed to detect a statistically significant protective
conjugated equine estrogen use with (HR = 2.05; 95% CI = 1.21, 3.48) effect of statin use against the development of AD. However, the
but not without (HR = 1.77; 95%: CI = 0.74, 4.23) medroxyproges- most recent systematic review and meta-analysis by Williams et al.
terone acetate was associated with a deleterious effect. Based on (2010) suggests a reduced risk of AD attributed to statin therapy.
these results, the reviewers concluded that estrogen combined These inconsistent findings may be attributed to methodological
with progestin, namely, conjugated equine estrogen use with heterogeneity across the identified reviews with respect to the
medroxyprogesterone acetate, increased risk of dementia. utilized search strategies and inclusion criteria. A recent cohort
study (Li et al., 2010a,b), identified in the second phase of the
3.1.16.2. Discussion. Earlier systematic reviews, which relied review, detected a reduced risk of AD attributed to statin therapy
solely on observational studies, detected a protective effect of use before the age of 80.
HRT (Yaffe et al., 1998; LeBlanc et al., 2001). However, results from
two RCTs identified by Williams et al. (2010) suggest that estrogen 3.1.18. Benzodiazepines
combined with progestin, namely, conjugated equine estrogen use One moderate systematic review examined the association
with medroxyprogesterone acetate, increases risk of AD/dementia. between benzodiazepines and risk of AD (Patterson et al., 2007).
In the absence of progestin, conjugated equine estrogen use did not However, the review did not identify any relevant studies.
increase risk of AD/dementia (Williams et al., 2010). Consequently, there was insufficient evidence to comment on
the association between benzodiazepines and risk of AD.
3.1.17. Lipid lowering therapy/statins
Nine systematic reviews, which examined the effect of lipid 3.1.19. Memantine
lowering therapies, were identified (Miller and Chacko, 2004; One moderate systematic review examined the association
Standridge, 2004; Patatanian and Gales, 2005; Patterson et al., between memantine and risk of AD (Williams et al., 2010). As the
2007; Zhou et al., 2007; Beri et al., 2009; Desilets et al., 2010; review did not identify any relevant primary studies, there is a lack
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 157

of evidence on which to evaluate the association between case-control studies included in the review failed to detect an
memantine and risk of AD. association between lead exposure and AD. Based on these
findings, the reviewers concluded that lead exposure was not
3.1.20. Vaccinations associated with AD risk.
One moderate systematic review examined the association
between vaccinations and risk of AD (Patterson et al., 2007). One of 3.1.24. Solvents
the two identified longitudinal studies reported that having Three moderate-quality systematic reviews reported on the risk
received any vaccination was associated with a 60% reduced risk of AD associated with occupational exposure to solvents (Graves
of AD (RR = 0.40; 95% CI = 0.17, 0.96). In a second study, vaccination et al., 1991b; Santibanez et al., 2007; Williams et al., 2010). No
against diphtheria or tetanus (RR = 0.41; 95% CI = 0.27, 0.62) and relevant primary studies were identified.
poliomyelitis (RR = 0.60; 95% CI = 0.37, 0.99) was associated with a
decreased risk of AD. In the same study, vaccination against 3.1.24.1. Systematic reviews. The systematic review and re-analysis
influenza was not associated with risk of AD (RR = 0.75; 95% by Graves et al. (1991b) identified three case-control studies that
CI = 0.54, 1.04). reported on the association between occupational exposure to
solvents and risk of AD. Two studies failed to detect an association.
3.1.21. Histamine-2 receptor antagonists A risk estimate could not be calculated for the third study as there
We did not identify any systematic reviews that reported on the were no exposed cases. In the pooled analysis representing 221
association between histamine-2 receptor antagonist (H2RA) use cases and 287 controls, occupational exposure to solvents was not
and risk of AD. Our search for primary studies yielded one relevant associated with AD (RR = 0.76; 95% CI = 0.47, 1.23).
record (Gray et al., 2011). Santibanez et al. (2007) identified 10 case-control and one
The association between H2RA use and risk of AD was cohort study. Only two of the eleven studies detected a statistically
prospectively examined by Gray et al. (2011). The study reported significant increased risk of AD, attributed to occupational
on the effect of cumulative use, intensity of use, and cumulative exposure to solvents. The remaining nine studies failed to detect
use relative to timing of exposure (recent and distant). There was an association. The reviewers suggest that the conflicting findings
no significant association between cumulative H2RA use and risk may be attributed to heterogeneity with respect to the broad
of AD. See Table 9. Categories delineating intensity of use were also category of solvents considered in the included studies.
not associated with risk of AD (see Table 98 in Supplementary Williams et al. (2010) relied on the systematic review by
material II). Only high distant and low recent H2RA use yielded Santibanez et al. (2007). The HTA report concluded that there was
statistically significant findings (HR = 1.66; 95% CI = 1.07, 2.57). The insufficient evidence to support an association between solvents
remaining categories of distant and recent cumulative use were and risk of AD.
not associated with risk of AD. Overall, findings of the study
suggest that H2RA use was not associated with risk of AD. 3.1.24.2. Discussion. Only two of eleven observational studies
included in the identified systematic reviews detected an
3.1.22. Angiotensin receptor blockers association between solvent exposure and risk of AD. Further
We did not identify any systematic reviews that reported on the research, by way of prospective cohort studies which focus on the
association between angiotensin receptor blocker use and risk of effect of specific solvents, is warranted (Santibanez et al., 2007;
AD. The second phase of the review yielded one relevant Williams et al., 2010).
observational study (Li et al., 2010a,b).
Using data from the US Veteran Affairs administrative database, 3.1.25. Electromagnetic Fields (EMF)
Li et al. (2010a,b) prospectively examined the association between Four systematic reviews, which examined the effect of
angiotensin receptor blocker use and risk of AD. Compared to occupational exposure to electromagnetic fields (EMF), were
lisinopril, an angiotensin converting enzyme inhibitor, angiotensin identified (Ahlbom, 2001; Santibanez et al., 2007; García et al.,
receptor blocker use was associated with a statistically significant 2008; Kheifets et al., 2009). One low-quality review was omitted
reduced risk of AD (HR = 0.81; 95% CI = 0.68, 0.96; p = 0.016). (Kheifets et al., 2009) (see Table 19 in Supplementary material II).
Compared to the cardiovascular comparator (drugs “excluding No primary studies were identified.
angiotensin receptor blockers, statins, and converting enzyme
inhibitors”), angiotensin receptor blockers marginally reduced risk 3.1.25.1. Systematic reviews. Four case-control and one cohort
of incident AD (HR = 0.84; 95% CI = 0.71, 1.00; p = 0.045) (Li et al., study were eligible for inclusion in the systematic review reported
2010a,b). by Ahlbom (2001). Two studies were clinical-based and three were
population-based studies. When the results of the two clinical-
3.1.23. Lead based studies were combined in a meta-analysis, occupational
Four systematic reviews reported on the risk of AD associated exposure to high or medium EMF levels was associated with an
with occupational exposure to lead (Graves et al., 1991b; increased risk of AD (RR = 3.20; 95% CI = 1.90, 5.40). According to
Santibanez et al., 2007; Williams et al., 2010; Loef et al., 2011a, the pooled analysis of population-based studies, occupational EMF
b). One review was excluded due to low methodological quality exposure was not associated with risk of AD (RR = 1.20; 95%
(Loef et al., 2011a,b). No relevant primary studies were identified. CI = 0.70, 2.30). When results from all five studies were pooled in a
The systematic review and re-analysis reported by Graves et al. fixed effects meta-analysis, occupational EMF exposure was
(1991b) identified four relevant case-control studies. All four associated with a greater than two-fold increased risk of AD
studies failed to detect a statistically significant association (RR = 2.20; 95% CI = 1.50, 3.20). Although results from two clinical
between lead exposure and AD. However, two studies had only based studies reached statistical significance, three more recent
one exposed case. The pooled analysis including 261 cases and 337 population-based studies, one of which employed a longitudinal
controls suggested that occupational lead exposure was not cohort design, failed to detect an association between EMF
associated with AD (RR = 0.71; 95% CI = 0.36, 1.41). exposure and AD (Ahlbom, 2001).
The HTA (Williams et al., 2010) examined the association Seven studies included in the systematic review by Santibanez
between lead and risk of AD. The reviewers relied on the et al. (2007) provided data pertaining to the association between
systematic review reported by Santibanez et al. (2007). All six occupational EMF exposure and risk of AD. Only two studies by the
158 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

same author detected an increased risk of AD associated with EMF exposure to defoliants and fumigants was associated with a greater
exposure. The remaining five studies failed to detect an associa- than four-fold increased risk of AD (RR = 4.35; 95% CI = 1.05, 17.90).
tion. The HTA reported by Williams et al. (2010) relied on the
Nine case-control studies and five cohort studies reporting on systematic review reported by Santibanez et al. (2007). Results of
the effect of extremely low frequency electric and magnetic fields the identified review suggest that exposure to pesticides may
(ELF-ELM) were eligible for inclusion in the review reported by increase risk of AD.
García et al. (2008). The meta-analysis combining data from all
case-control studies yielded a risk estimate of 2.03 (95% CI = 1.38, 3.1.27.2. Discussion. According to the evidence summarized in
3.00). The meta-analysis combining results from all five cohort three systematic reviews, exposure to pesticides may increase the
studies detected a 62% increased risk of AD associated with ELF- risk of AD. However, results of the identified reviews were limited
EMF exposure (RR = 1.62; 95% CI = 1.16, 2.27; I2 = 54%; test for by the small number of available studies. Consequently, further
heterogeneity: P value = 0.016). research is required to confirm the effect of pesticide exposure on
the development of AD.
3.1.25.2. Discussion. Evidence pertaining to the effect of ELF-EMF
exposure on risk of AD is relatively inconsistent. Results from the 3.1.28. Manual labour
most recent systematic review and meta-analysis (García et al., One moderate quality systematic review reported on the risk of
2008), which incorporated the largest number of studies, provide AD associated with manual labour (Patterson et al., 2007). No
some evidence of an increased risk of AD associated with ELF-EMF primary studies were identified.
exposure. However, these results must be interpreted with caution According to the only relevant study identified by Patterson
due to statistical heterogeneity and publication bias (García et al., et al. (2007), there was no significant association between manual
2008). work and AD (RR = 1.40; 95% CI = 0.90, 2.10).

3.1.26. Aluminum 3.1.29. Mercury, mercury amalgam fillings


Three systematic reviews reported on the risk of AD associated Risk of AD associated with environmental exposure to mercury
with exposure to aluminum (Santibanez et al., 2007; Ferreira et al., or mercury amalgam fillings was examined in two systematic
2008; Williams et al., 2010). One review was excluded due to low reviews. The review on mercury amalgam fillings was excluded
methodological quality (Ferreira et al., 2008). No primary studies due to methodological quality (Bates, 2006).
were identified. The HTA reported by Williams et al. (2010) identified a single
cohort study that reported on the effect of environmental exposure
3.1.26.1. Systematic reviews. All three studies identified by to mercury on risk of AD. The study detected a reduced risk of AD
Santibanez et al. (2007) failed to detect an association between associated with higher blood mercury concentrations (Third
occupational exposure to aluminum and risk of AD. Based on these quartile, OR = 0.41; 95% CI = 0.23, 0.74. Fourth quartile, OR = 0.56;
findings, the reviewers concluded that occupational aluminum 95% CI = 0.32, 0.99).
exposure was not associated with risk of AD.
The HTA reported by Williams et al. (2010) relied on the 3.1.30. Copper, iron
findings reported by Santibanez et al. (2007), which failed to detect One systematic review examined the effect of copper and iron
an association between the variables of interest. The reviewers on the risk of AD (Loef and Walach, 2012a,b). The review was
identified one recent cohort study that reported an increased risk deemed to be of low methodological quality and was excluded.
of AD associated with aluminum from drinking water equal to or
exceeding 0.1 mg/day (RR = 1.34; 95% CI = 1.09, 1.65). The reviewers 3.1.31. Genetic risk factors
concluded that there was insufficient evidence to comment on the Amyloid precursor protein (APP), presenilin 1 (PSEN1) or
effect of aluminum. presenilin 2 (PSEN2) mutations are genetic causes of autosomal-
dominant Alzheimer's disease (Bateman et al., 2011). We identified
3.1.26.2. Discussion. Based on the limited available evidence, the a single review that reported on the 2/2 genotype of presenilin 1
association between aluminum and risk of AD was inconclusive. (Rodriguez-Manotas et al., 2007). Several reviews reporting on
genetic risk factors for AD are also summarized herein.
3.1.27. Pesticides
Three moderate-quality systematic reviews examined the risk 3.1.31.1. Presenilin (PS)-1 2/2 genotype. A total of 38 case-control
of AD associated with exposure to pesticides (Patterson et al., studies examining the association between the presenilin-1 (PS-1)
2007; Santibanez, Bolumar, and Garcia 2007; Williams et al., 2010). 2/2 genotype and late-onset AD were eligible for inclusion in the
No relevant primary studies were identified. review reported by Rodriguez-Manotas et al. (2007). When the
results of all 38 studies were combined in a random effects meta-
3.1.27.1. Systematic reviews. The systematic review by Santibanez analysis, compared to the combined effect of the 1/1 and 1/2
et al. (2007) identified six studies that reported on the association genotypes, the 2/2 genotype was not associated with AD
between occupational pesticide exposure and risk of AD. Of these (OR = 0.99; 95% CI = 0.84, 1.17; test for heterogeneity: x2 = 79.92,
six studies, four case-control studies failed to detect a statistically p < 0.0001, I2 = 53.7%). Stratified analysis by ethnicity detected a
significant association. Results from two high quality cohort marginally significant increased risk among Europeans (OR = 1.19;
studies provided conflicting results, suggesting a positive 95% CI = 1.00, 1.41), a reduced risk among North Americans
association between occupational exposure to pesticides and (OR = 0.79; 95% CI = 0.64, 0.98), and no significant association in
risk of AD. Based on findings of the cohort studies, the review offers Asians (OR = 0.79; 95% CI = 0.54, 1.17; test for heterogeneity: x
2
some evidence of an association between occupational pesticide = 38.83, p = 0.0001, I2 = 69.1%).
exposure and risk of AD (Santibanez et al., 2007).
One longitudinal cohort study identified by Patterson et al. 3.1.31.2. Interleukin-1b. The systematic review reported by Di
(2007) reported on the association between exposure to environ- Bona et al. (2008) examined the association between interleukin-
mental toxins and risk of AD. Results from this study suggest that 1b (IL-1b) SNPs 511 and +3953. For both polymorphisms, the TT
genotype was compared to the TC + CC genotypes. A total of five
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 159

studies reporting on IL-1b +3953 polymorphism were identified. 491A/T polymorphism. The meta-analysis for the allelic
The pooled analysis yielded a risk estimate of 1.60 (95% CI = 1.16, comparison detected an increased risk of AD associated with
2.22; test for heterogeneity: x2 = 2.57, p = 0.63, I2 = 0%) suggesting a the A allele (OR = 1.45; 95% CI = 1.27, 1.65). The pooled analysis
60% increased odds of AD associated with the TT genotype of IL-1b comparing the AA genotype to the AT genotype yielded an OR of
+3953. The meta-analysis restricted to Caucasian-only populations 1.47 (95% CI = 1.27, 1.70). Those carrying the AT genotype had a
yielded a risk estimate of 1.71 (95% CI = 1.21, 2.43). A total of 17 marginally significant 25% (OR = 1.25; 95% CI = 1.00, 1.56) increased
studies reported on IL-1b 511. According to the random effects odds of AD when compared to TT genotype carriers. 491A
meta-analysis, the IL-1beta 511 TT genotype was not associated homozygotes had a 72% (OR = 1.72; 95% CI = 1.39, 2.13) increased
with an increased odds of AD (OR = 1.01; 95% CI = 0.84, 1.21; test for odds of AD when compared to 491T homozygotes. When 491A
heterogeneity: x2 = 25.64, p = 0.06, I2 = 37.6%). When the analysis homozygotes were compared to the AT and TT (AT + TT) genotypes,
was restricted to studies with the highest statistical power (n = 4), 491A homozygotes had a 1.49 times (OR = 1.49; 95% CI = 1.29,
the TT genotype of IL-1b 511 was significantly associated with 1.72; test for heterogeneity: x2 = 77.34, p < 0.001) increased odds
risk of AD (OR = 1.32; 95% CI = 1.03, 1.69; test for heterogeneity: of AD. Finally, in comparison to the TT genotype, the AA and AT
x2 = 0.01, p = 1.00, I2 = 0%). (AA + AT) genotypes were associated with a 57% increased odds of
AD (OR = 1.57; 95% CI = 1.27, 1.94).
3.1.31.3. Interleukin-1a. The systematic review by Rainero et al. Fifteen studies provided data pertaining to the risk of AD
(2004) examined the association between the interleukin-1alpha attributed to the 427T/C polymorphism. The series of allelic and
889C/T gene polymorphism and AD. A total of 11 case-control genotypic comparisons for the 427T/C polymorphism included
studies were eligible for inclusion. According to random effects 2962 cases and 2883 controls. All comparisons were non-
meta-analysis, the TT genotype was associated with an increased significant (see Table 28 in Supplementary material II)A total of
risk of AD when compared to the CT (OR = 1.51; 95% CI = 1.15, 1.99; 16 studies representing 6614 cases and 6363 controls provided
test for heterogeneity: x2 = 15.55, p = 0.11), CC (OR = 1.49; 95% data on the association between the 219T/G polymorphism and
CI = 1.09, 2.03; test for heterogeneity: x2 = 19.32, p = 0.036), and CC/ AD. Each meta-analytic comparison yielded statistically significant
CT (OR = 1.50; 95% CI = 1.12, 2.02) genotypes. Subgroup analysis findings with ORs ranging from 1.21 to 1.51.
according to the fixed effects model detected an increased risk of
early onset AD (see Table 25 in Supplementary material II). Results 3.1.31.7. Apolipoprotein E (APOE) e4. We identified two systematic
failed to reach statistical significance according to the random reviews (Elias-Sonnenschein et al., 2011; Hao et al., 2011a,b) and
effects model. No association was detected between the TT five cohort studies (Reitz et al., 2010b; Chu et al., 2010; Blasko et al.,
genotype and LOAD according to both models. The pooled 2010; Ronnemaa et al., 2011; Yang et al., 2011) that examined the
analysis comparing the CT genotype to the CC genotype was not association between APOE e4 and risk of AD.
statistically significant (OR = 0.98; 95% CI = 0.96, 1.14; test for
heterogeneity: p = 0.20). 3.1.31.7.1. Systematic reviews. Thirty-five studies, reporting on the
association between APOE e4 and risk of progression from MCI to
3.1.31.4. Interleukin-10. The systematic review and meta-analysis AD, were eligible for inclusion in the systematic review conducted
by Zhang et al. (2011) investigated the effect of the 1082G/A by Elias-Sonnenschein et al. (2011). According to the random
polymorphism in the IL-10 gene on AD risk. Twelve studies were effects meta-analysis, possessing at least one APOE e4 allele was
eligible for inclusion. According to the preliminary random effects associated with a 2.29 times (95% CI = 1.58, 4.42) increased risk of
meta-analysis, when compared to the GG genotype, the A allele progression from MCI to AD. When stratified by study type,
(AA + AG) was associated with an increased risk of AD (OR = 1.27; possessing at least one APOE e4 allele was associated with a 2.36,
95% CI = 1.02, 1.58; test for heterogeneity: x2 = 20.74, p = 0.04, 1.83, and 2.64 times increased risk of progression from MCI to AD
I2 = 47.0%). When the analysis was restricted to the 11 European according to 21 clinical studies, six population-based studies, and
studies, the A allele remained significantly associated with an eight ‘other’ studies, respectively.
increased risk of AD (OR = 1.27; 95% CI = 1.01, 1.59; test for The systematic review reported by Hao et al. (2011a,b)
heterogeneity: x2 = 20.72, p = 0.02, I2 = 51.7%). The only Asian examined the association between APOE e4 and risk of AD in
study failed to detect a significant association (OR = 1.37; 95% East Asians. Three studies were eligible for inclusion in a random
CI = 0.32, 5.88). Meta-analyses examining the remaining effects meta-analysis. According to the pooled analysis, APOE e4
comparisons failed to detect statistically significant results carriers had a near 3-fold increased risk of AD (OR = 2.98; 95%
(seeTable 26 in Supplementary material II) CI = 1.84, 4.84; test for heterogeneity: x2 = 6.05, p = 0.05, I2 = 67%).

3.1.31.5. Interleukin-6. Fourteen studies which examined the 3.1.31.7.2. Primary studies. Four studies detected an increased risk
association between IL-6 174G/C polymorphism and risk of AD of AD associated with the APOE e4. The longitudinal cohort study
among Caucasians were included in the systematic review conducted by Yang et al. (2011) detected an increased risk of AD
reported by Han et al. (2011). According to random effects among those possessing one or more APOE e4 allele, with risk
meta-analysis, the GG (OR = 1.35; 95% CI = 1.06, 1.72; test for estimates ranging from 1.65 to 1.69. In an all-male sample of
heterogeneity: I2 = 52.5%, p = 0.01) and GG/GC (OR = 1.27; 95% Chinese subjects (Chu et al., 2010), APOE e4 carriers had a five-fold
CI = 1.05, 1.53) genotypes were associated with risk of AD. However, increased risk of AD compared to non-carriers (RR = 5.04; 95%
results failed to reach statistical significance when one study “that CI = 1.05, 24.10). Possession of an APOE e4 allele, ascertained in
reported a higher detection rate of C allele (57%)" was excluded mid- and late-life, was associated with an increased risk of AD
(Han et al., 2011). Results of the remaining meta-analytic according to findings from the Uppsala Longitudinal Study of Adult
comparisons failed to reach statistical significance. Men (Midlife, HR = 2.60; 95% CI = 1.70, 3.90. Late-life, HR = 2.50; 95%
CI = 1.60, 3.80) (Ronnemaa et al., 2011). Although Reitz et al.
3.1.31.6. Apolipoprotein E (APOE) promoter 491A/T, 427T/C, 219T/ (2010b) detected an increased risk of probable and possible LOAD
G polymorphisms. The association between APOE promoter among carriers of one or more APOE e4 allele (HR = 1.83; 95%
491A/T, 427T/C, 219T/G polymorphisms and risk of AD was CI = 1.02, 3.29), results failed to reach statistical significance when
reviewed by Xin et al. (2010). A total of 32 studies, representing the analysis was restricted to probable LOAD (HR = 1.85; 95%
5789 cases and 5962 controls, provided data pertaining to the
160 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

CI = 0.96, 3.57). The remaining study (Blasko et al., 2010) failed to increased risk of AD. However, an association was detected for
detect an association (OR = 2.30; 95% CI = 0.90, 6.00). Asians (OR = 1.37; 95% CI = 1.12, 1.68) but not Caucasians (OR = 1.05;
95% CI = 0.92, 1.21).
3.1.31.8. Apolipoprotein E (APOE) e2. We did not identify any
systematic reviews which reported on the association between 3.1.31.11. Neuronal sortilin-related receptor gene (SORL1). We
APOE e2 and risk of AD. However, one relevant cohort study was identified two systematic reviews which reported on the
identified in the second phase of the review (Yang et al., 2011). association between neuronal sortilin-related receptor gene
Results from the AlzGene database were also considered. (SORL1) and risk of AD (Ning et al., 2010; Reitz et al., 2011).
The longitudinal cohort study conducted by Yang et al. (2011) Ning et al. (2010) examined the association between neuronal
examined the association between blood pressure, hypertension, SORL1 and LOAD. Four studies were eligible for inclusion. Random
and risk of AD. The association between blood pressure measures, effects meta-analysis of three studies detected an association
namely, self-reported hypertension and objectively measured between SNP rs2070045 and LOAD (OR = 1.35; 95% CI = 1.11, 1.65). In
systolic (SBP) and diastolic blood pressure (DBP), pulse pressure the pooled analysis of all four studies, representing 984 LOAD cases
(PP), and mean arterial pressure (MAP), were analyzed in separate and 1320 control subjects, rs3824968 (OR = 1.29; 95% CI = 1.14, 1.46)
models. The association between APOE e2 status and risk of AD was and rs2282649 (OR = 1.34; 95% CI = 1.19, 1.52) were associated with
examined in each model. According to the risk estimates derived an increased risk of LOAD.
from each model, APOE e2 was not associated with risk of AD. A total of 17 studies representing 20 data sets were eligible for
Results from a pooled analysis (n = 37) summarized in the AlzGene inclusion in the review reported by Reitz et al. (2011). A series of
database (see “AlzGene Top Results” summary) was suggestive of a meta-analyses were conducted to examine the association
protective effect associated with APOE e2 (OR = 0.62; 95% CI = 0.46, between AD and SORL1 SNPs 4, 5, 8, 9, 10, 12, 19, 22, 23, 24, and
0.85; I2 = 64%). 25. SNPs 4, 5, 8, 9, 10, 12, and 19 were significantly associated with
AD in the Caucasian data sets (SNPs 5, 8, 9, 10, 19 increased risk
3.1.31.9. Aldehydedehydrogenase 2 gene (ALDH2) Glu504Lys while SNPs 4 and 12 reduced risk). SNPs 19, 23, 24, and 25 were
polymorphism. Hao et al. (2011a,b) reviewed the association associated with risk of AD in Asians (SNPs 19 and 25 increased risk
between the aldehyde dehydrogenase 2 gene (ALDH2) Glu504Lys while SNPs 23 and 24 were protective).
polymorphism and risk of AD in East Asians. Four studies were
eligible for inclusion. Random effects meta-analysis representing 3.1.31.12. Cathepsin D (CTSD) Ala224Val gene polymorphism. Ntais
821 cases and 1380 controls failed to detect an association et al. (2004) reviewed the association between cathepsin D
(OR = 1.35; 95% CI = 0.75, 2.42; test for heterogeneity: x2 = 23.01, Ala224Val gene polymorphism (CTSD) and risk of AD. Fourteen
p < 0.0001, I2 = 87%). The presence of the ALDH2 genotype studies were eligible for inclusion. The random effects meta-
increased the risk of AD in males (OR = 1.72; 95% CI = 1.10, 2.67) analysis failed to detect a statistically significant difference in AD
but not females (OR = 0.91; 95% CI = 0.33, 2.49) according to risk between the alleles (OR = 1.17; 95% CI = 0.95, 1.44; test for
subgroup analysis of two studies. Results of the review support an heterogeneity: p < 0.01). The remaining comparisons also failed to
association between the GA/AA genotype of ALDH2 and an yield statistically significant findings (see Table 34 in
increased risk of AD in East Asian men (Hao et al., 2011a,b). Supplementary material II).

3.1.31.10. Methylenetetrahydrofolatereductase C677T polymorphism 3.1.31.13. ACE D/I polymorphism. A total of 23 studies, which
We identified two moderate-quality systematic reviews which examined the association between the I allele of the angiotensin-
reported on the association between methylenetetrahydrofolater- converting enzyme (ACE) D/I polymorphism and LOAD, were
eductase C677T polymorphism and risk of AD (Zhang et al., 2010; eligible for inclusion in the systematic review reported by Elkins
Hua et al., 2011). et al. (2004). When the results of all studies were combined in a
Nineteen studies satisfied the eligibility criteria of the review random effects meta-analysis, the I allele of the ACE D/I
reported by Zhang et al. (2010). In the pooled analysis examining polymorphism was associated with an increased risk of AD
the allelic comparison, the T allele of the MTHFR C677T (OR = 1.27; 95% CI = 1.10, 1.47; test for heterogeneity: p < 0.001).
polymorphism was associated with a 15% (OR = 1.15; 95% CI = 1.06, When compared to carriers of the DD genotype, those with the II
1.26) increased odds of AD. However, the subgroup analysis by (OR = 1.27; 95% CI = 1.04, 1.54) and I/D (OR = 1.27; 95% CI = 1.09, 1.47)
ethnicity suggested that the T allele increased risk of AD in East genotypes were also at an increased risk.
Asians (OR = 1.22; 95% CI = 1.08, 1.39) and in “other populations”
(OR = 1.56; 95% CI = 1.14; 2.12) but not in Caucasians (OR = 1.05; 95% 3.1.31.14. Tumor Necrosis Factor (TNF)  a polymorphisms. Di Bona
CI = 0.93, 1.18). T allele carriers were at an increased risk of AD when et al. (2009) systematically reviewed the literature to ascertain the
compared to those carrying the CC genotype (OR = 1.21; 95% association between tumor necrosis factor (TNF)-alpha
CI = 1.07, 1.36). TT genotype carriers had a 32% increased risk of AD polymorphisms and risk of AD. According to random effects
compared to CC genotype carriers (OR = 1.32; 95% CI = 1.11, 1.58). meta-analyses of six studies, TNF-alpha-308 was not associated
There was no significant difference between TT and CT + CC with risk of AD. When compared to the CC (OR = 1.63; 95% CI = 1.07,
genotypes with respect to AD risk (OR = 0.86; 95% CI = 0.69, 1.06). 2.49) and CT + CC genotypes (OR = 1.57; 95% CI = 1.08, 2.29), the TT
Twenty-seven studies, 13 of which were conducted in Asian genotype of TNF-alpha-850 was significantly associated with risk
populations, were included in the systematic review reported by of AD. The remaining comparisons for TNF-alpha-850 failed to
Hua et al. (2011). According to random effects meta-analysis, the T reach statistical significance. When results from the four studies
allele was associated with a 13% (OR = 1.13; 95% CI = 1.04, 1.24) examining TNF-alpha-863 SNP were combined in a random effects
increased odds of AD when compared to the C allele. This meta-analysis, there was no statistically significant difference in
association was true for Asians (OR = 1.28; 95% CI = 1.07, 1.52; test AD risk between the CC genotype and the AC + AA genotypes
for heterogeneity: p = 0.002, I2 = 61.6%) but not Caucasians (OR = (OR = 1.26; 95% CI = 0.86, 1.85). Similarly, results from the pooled
1.02; 95% CI = 0.92, 1.12). Compared to the CT + CC genotypes, the analysis comparing the AG + AA genotypes of the TNF-alpha-238
TT genotype was not associated with risk of AD (OR = 1.09; 95% SNP to the GG genotype were not statistically significant
CI = 0.95, 1.26). In comparison with CC genotype carriers, those (OR = 0.78; 95% CI = 0.57, 1.08). Finally, the pooled analysis
carrying the T allele had an 18% (OR = 1.18; 95% CI = 1.06, 1.32) examining the effect of the 1031 SNP failed to detect a
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 161

statistically significant difference in AD risk between the TT polymorphism of MS4A6A and rs3865444 polymorphism of CD33
genotype and the CT + CC genotypes (OR = 0.62; 95% CI = 0.36, 1.09). were associated with a reduced risk of AD (APOE e2, OR = 0.62; 95%
Overall, results of the review provide no evidence to support an CI = 0.46, 0.85; I2 = 64%. MS4A6A, OR = 0.90; 95% CI = 0.88, 0.93, p-
association between AD and TNF-alpha SNPs 308, 863, 238, value = 1.81E-11; I2 = 23%. CD33, OR = 0.89; 95% CI = 0.86, 0.93, p-
and 1031. However, the TT genotype of the 850 SNP was value = 2.04E-10; I2 = 0%). Finally, the rs11136000, rs3818361, and
associated with an increased risk of AD. rs3851179 polymorphisms of CLU, CR1, and PICALM, respectively,
were associated with AD among Caucasians (CLU, OR = 0.88; 95%
3.1.31.15. PRNP M129V polymorphism. One systematic review CI = 0.86, 0.90, p-value = 3.37E-23; I2 = 0%. CR1, OR = 1.17; 95%
examined the effect of the PRNP M129 V polymorphism on the CI = 1.14, 1.21, p-value = 4.72E-21; I2 = 0%. PICALM, OR = 0.88; 95%
risk of AD (Del Bo et al., 2006). The study was excluded due to low CI = 0.86, 0.90, p-value = 2.85E-20; I2 = 0%).
methodological quality. Consequently, there was inadequate
evidence to comment on the effect of this polymorphism. 3.1.32. Alcohol
We identified ten systematic reviews reporting on the
3.1.31.16. b-amyloid related genes. According to the series of meta- association between alcohol consumption and risk of AD. Three
analyses conducted by Llorca et al. (2008), only the C allele of the reviews were later excluded due to low methodological quality
OLR1 30 -UTR (+1073) polymorphism was associated with AD. The (Standridge, 2004; Purnell et al., 2009; ESHRE Capri Workshop
overall pooled analyses for the remaining polymorphisms (BACE1, Group, 2011). Results of the remaining seven moderate-quality
CYP47, FE65, ACT Ala17Thr, BH 1443 V, OLR1 30 -UTR (+1071), VLDLR systematic reviews were used to evaluate the association between
50 -UTR (CGG-repeat)) failed to yield statistically significant alcohol consumption and risk of AD (Graves et al., 1991a; Weih
findings. However, according to subgroup analyses, the et al., 2007; Patterson et al., 2007; Peters et al., 2008b; Anstey et al.,
association between the CC + CG genotype of the BACE1exon 5 2009; Williams et al., 2010; Lee et al., 2010). Furthermore, two
(G/C) polymorphism was dependent on APOE status. More primary studies were identified (Weyerer et al., 2011; Zhou et al.,
specifically, the CC + CG genotype of the BACE1 exon 5 (G/C) 2011) (see Tables 38 and 107 in Supplementary material II).
polymorphism was associated with a protective effect against AD
among APOE e4 carriers (OR = 0.57; 95% CI = 0.38, 0.88) and a 3.1.32.1. Systematic reviews. The systematic review by Graves et al.
marginally significant increased risk of AD (OR = 1.33; 95% CI = 1.00, (1991a) examined the effect of smoking and alcohol consumption
1.78) among APOE e4 non-carriers. Similarly, the CC genotype of on the risk of AD. Five case-control studies were eligible for
the BACE1 exon 5 (G/C) polymorphism was associated with a inclusion in the pooled analysis. Results of the pooled analysis,
protective effect against AD among APOE e4 carriers (OR = 0.48; adjusted for family history of dementia, suggested that when
95% CI = 0.26, 0.88) and an increased risk of AD among APOE e4 compared to abstinence, mild (RR = 0.97; 95% CI = 0.59, 1.60),
non-carriers (OR = 1.61; 95% CI = 1.11, 2.34). Although the VLDLR 50 - moderate (RR = 0.75; 95% CI = 0.46, 1.20), and high (RR = 0.89; 95%
UTR (CGG-repeat) polymorphism was not associated with AD in CI = 0.52, 1.51) alcohol consumption was not associated with a
the overall pooled analysis, subgroup analysis revealed that reduced risk of AD. A more rigorous attempt to control for potential
genotype 2 was associated with a significant increase risk of AD confounders, namely head injury, education, and family history of
among Asians (OR = 1.70; 95% CI = 1.09, 2.63) and a reduced risk in dementia, yielded consistent findings suggesting a lack of
non-Asians (OR = 0.48; 95% CI = 0.26, 0.86). association between alcohol consumption and risk of AD (Mild,
RR = 0.89; 95% CI = 0.59, 1.47. Moderate, RR = 0.82; 95% CI = 0.49,
3.1.31.17. AlzGene “Top Results”. Our systematic search of the 1.39. Heavy, RR = 0.95; 95% CI = 0.51, 1.81). Although all risk
literature was supplemented with a review of the AlzGene estimates failed to reach statistical significance, the reviewers
database, a frequently updated, publically available, online note that moderate alcohol consumption produced the lowest
database examining genetic variants associated with AD relative risk. Finally, the sensitivity analysis excluding the
(Khoury et al., 2009; Bertram et al., 2007). Studies included in methodologically heterogeneous study also failed to reach
the database are obtained through a comprehensive, systematic statistical significance for all three categories of alcohol
review of the literature. Pooled analyses are regularly updated with consumption (Mild, RR = 0.85; 95% CI = 0.48, 1.50. Moderate,
newly emerging data (Bertram et al., 2007). RR = 1.48; 95% CI = 0.67, 3.23. Heavy, RR = 0.68; 95% CI = 0.31,
Currently, the AlzGene database summarizes evidence from 1.48). Overall, results of the systematic review reported by
1395 primary studies covering 695 genes and 2973 polymor- Graves et al. (1991a), suggested that alcohol consumption was
phisms. Moreover, 320 meta-analyses have been conducted to not associated with risk of AD
date. Only evidence summarized in the “Top Results” section of the Eight studies were eligible for inclusion in the systematic
database were reviewed. Briefly, this section presents polymor- review reported by Weih et al. (2007). Two studies reported a
phisms which are “most strongly associated” with Alzheimer’s reduced risk of AD associated with mild and moderate alcohol
disease according to a quantitative synthesis of the available consumption. All three studies reporting on wine consumption
literature (Bertram et al., 2007). detected a protective effect. One study reported a detrimental
The most strongly associated genes, listed in order of decreasing effect attributed to monthly beer consumption. The remaining
strength of association according to p-value, were APOE e2/3/4, studies failed to detect an association. Overall, findings of the
BIN1, CLU, ABCA7, CR1, PICALM, MS4A6A, CD33, MS4A4E, and review suggest that mild to moderate alcohol consumption may
CD2AP. Compared to the e3 allele, APOE e4 was associated with reduce risk of AD, with a greater benefit associated with moderate
3.69 increased risk of AD (95% CI = 3.30, 4.12, p-value = < 1E-50; consumption.
I2 = 30%). Polymorphism rs744373 of BIN1 was associated with a Two longitudinal cohort studies were eligible for inclusion in
1.17 times increased risk of AD (95% CI = 1.13, 1.20, p-value = 1.59E- the systematic review reported by Patterson et al. (2007). Both
26; I2 = 0%). The rs3764650, rs670139, and rs9349407 polymor- studies detected a reduced risk of AD associated with wine
phisms of the ABCA7, MS4A4E, and CD2AP genes, respectively, consumption. One study detected a 47% (RR = 0.53; 95% CI = 0.30,
were all associated with an increased risk of AD (ABCA7, OR = 1.23; 0.95) reduced risk of AD associated with moderate (250 to 500 ml/
95% CI = 1.18, 1.28, p-value = 8.17E-22; I2 = 0%. MS4A4E, OR = 1.08; day) wine consumption. The other reported consistent findings
95% CI = 1.05, 1.11, p-value = 9.51E-10; I2 = 2%. CD2AP, OR = 1.12; 95% (OR = 0.49; 95% CI = not reported).
CI = 1.08, 1.16, p-value = 2.75E-09; I 2 = 0%). APOE e2, rs610932
162 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Eight longitudinal studies, which reported on risk of incident Compared to occasional drinking, only daily alcohol consump-
AD, were eligible for inclusion in the pooled analysis reported by tion was associated with risk of AD (HR = 1.72; 95% CI = 1.07, 2.75)
Peters et al. (2008b). Results of the random effects meta-analysis according to the cohort study reported by Zhou et al. (2011).
suggested that alcohol consumption was associated with a
protective effect against the development of AD (RR = 0.57; 95% 3.1.32.3. Discussion. Evidence summarized in a majority of the
CI = 0.44, 0.74). However, there was evidence of heterogeneity identified systematic reviews suggest a reduced risk of AD
across studies (I2 = 46%, Cochran Q = 22.23, df = 12, P = 0.035) likely associated with alcohol consumption. Results from two recent
attributed to the heterogeneous definitions of exposure. There was cohort studies confirm the association between alcohol
no evidence of publication bias (Begg-Mazumdar test: consumption and risk of AD. One study detected a reduced risk
Kendall's t = 0.03, P = 0.86; Egger’s test: 0.27; 95% CI = 2.16, of AD associated with light-to-moderate alcohol consumption in
2.71, P = 0.81). late-life (Weyerer et al., 2011). The other reported a detrimental
A total of nine studies, identified by Anstey et al. (2009), effect associated with daily alcohol consumption (Zhou et al.,
provided data on the risk of AD associated with alcohol 2011). Considered cumulatively, alcohol consumption appears to
consumption. Six studies reported on the effect of light to reduce the risk of AD with the most convincing evidence for light-
moderate alcohol consumption. The pooled analysis detected a to-moderate quantities of consumption.
28% reduced risk of AD associated with light to moderate alcohol
consumption (RR = 0.72; 95% CI: 0.61, 0.86; Test for heterogeneity: 3.1.33. Smoking
x2 = 11.43, p = 0.04). Four studies evaluated the effect of heavy/ Twelve systematic reviews examined the association between
excessive alcohol consumption. Heavy alcohol consumption was smoking and risk of Alzheimer’s disease (Graves et al., 1991a;
not associated with risk of AD according to the pooled analysis Almeida et al., 2002; Patterson et al., 2007; Weih et al., 2007;
(RR = 0.92; 95% CI 0.59, 1.45; test for heterogeneity = not statisti- Anstey et al., 2007; Peters et al., 2008c; Purnell et al., 2009;
cally significant). As noted by the reviewers, the failure to detect an Williams et al., 2010; Cataldo et al., 2010; Lee et al., 2010; ESHRE
association may be attributed to sources of selection bias. Two Capri Workshop Group, 2011; Barnes and Yaffe, 2011). Three
studies compared the risk of AD among drinkers compared to non- reviews received a low quality score and were excluded (Purnell
drinkers. Results of the pooled analysis suggested a 34% decreased et al., 2009; ESHRE Capri Workshop Group, 2011; Barnes and Yaffe,
risk (RR = 0.66; 95% CI = 0.47, 0.94) of AD associated with alcohol 2011). The remaining nine reviews were deemed to be of moderate
consumption. The analysis, however, was limited by the small methodological quality with AMSTAR scores ranging from 4 to 7.
number of studies suitable for inclusion (Anstey et al., 2009). Our search for recent observational studies yielded six studies that
The HTA report by Williams et al. (2010) examined the effect of reported on the effect of smoking.
alcohol consumption on risk of AD. The report identified one
systematic review (Anstey et al., 2009). According to findings of the 3.1.33.1. Systematic reviews. The systematic review by Graves et al.
identified review, the report summarizes that light to moderate, (1991a) examined the effect of smoking and alcohol consumption
but not heavy, alcohol consumption protects against the develop- on the risk of AD. According to the pooled analysis, having ever
ment of AD. smoked was associated with a statistically significant 22%
Lee et al. (2010) identified two studies which reported on the (RR = 0.78; 95% CI = 0.62, 0.98) reduced risk of AD. Smoking less
association between alcohol consumption and risk of AD. Alcohol than one pack per day was associated with a 0.84 times (95%
consumption, defined as intake of sake one to two times per week, CI = 0.75, 0.92) reduced risk of AD while excessive smoking ( > 1
was not associated with AD risk according to one of the two studies pack/day) was not protective (RR = 0.81; 95% CI = 0.53, 1.27). When
(RR = not reported). The second study reported that in comparison the outlying study was excluded from the analysis, however,
to abstinence, light (1 serving/month  6 servings/week) to smoking less than one pack per day was no longer associated with
moderate (1–3 servings/day) wine consumption was associated a reduced risk of AD (RR = 0.80; 95% CI = 0.60, 1.07) and excessive
with a 45% (HR = 0.55; 95% CI = 0.34, 0.89) decreased risk of AD. smoking was associated with a marginally significant reduced risk
(RR = 0.66; 95% CI = 0.44, 1.00). Compared to never smokers, those
3.1.32.2. Primary studies. The effect of current alcohol with a low pack-year smoking history (<15.5 pack-years) did not
consumption in late-life was examined in the prospective cohort have a statistically significant reduced risk of AD (RR = 0.73; 95%
study conducted by Weyerer et al. (2011). The effect of varying CI = 0.50, 1.09). In contrast, moderate (15.5–37.0) and high (>37.0)
quantities and types of alcoholic beverages were also examined. pack-year smoking histories were associated with a statistically
Abstainers served as the referent category for all analyses. When significant 0.63 (95% CI = 0.42, 0.95) and 0.50 (95% CI = 0.31, 0.80)
alcohol consumption was treated as a dichotomous variable, reduced risk of AD, respectively. The trend was statistically
current alcohol consumption was associated with a statistically significant (p = 0.0003) suggesting that risk of AD decreased
significant reduced risk of AD (HR = 0.58; 95% CI = 0.38, 0.89). with increasing pack-year history. Based on an analysis of eight
Stratification by quantity of alcohol revealed a reduced risk of AD case-control studies, the reviewers concluded that smoking
among those reporting consumption of 20 to 29 g/day (HR = 0.13; appeared to be associated with a protective effect against the
95% CI = 0.02, 0.95). The remaining categories did not reach development of AD. The reviewers cautioned that prospective
statistical significance (1.9 g/day, HR = 0.61; 95% CI = 0.36, 1.01. 10– studies were required to confirm these findings.
19 g/day, HR = 0.72; 95% CI = 0.37, 1.39. 30–39 g/day, HR = 0.33; 95% The systematic review and meta-analysis conducted by
CI = 0.05, 2.41. 40 g/day, HR = 0.68; 95% CI = 0.16, 2.90). With Almeida et al. (2002) examined the effect of ever smoking on
respect to the type of alcoholic beverage, only those reporting risk of AD. A total of 21 case-control and eight cohort studies were
consumption of a variety of drinks (wine, beer, spirits) were at a eligible for inclusion in the review. Results from the fixed effects
reduced risk of AD (HR = 0.14; 95% CI = 0.03, 0.56). The “wine only” meta-analysis including all 21 case-control studies (n = 5323),
and “beer only” categories failed to reach statistical significance irrespective of study quality, suggested that ever smoking was
(Wine only, HR = 0.76; 95% CI = 0.46, 1.23. Beer only, HR = 0.60; 95% associated with a statistically significant decreased odds (OR =
CI = 0.30, 1.21). Based on these results, the authors concluded that 0.74; 95% CI = 0.66, 0.84; test for heterogeneity: p = 0.246) of AD.
late-life alcohol consumption, in light-to-moderate quantities, Results from the overall pooled analysis of case-control studies
appeared to be associated with a protective effect against the suggest that history of smoking was associated with a protective
development of AD. effect against AD. However, the series of sensitivity analyses
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 163

restricted to the most methodologically rigorous case-control case-control and 17 cohort studies) satisfied the inclusion criteria
studies failed to detect an association, suggesting that the observed of the review. The pooled analysis of affiliated cohort studies failed
protective effect may be due to sources of bias. Furthermore, to detect an association (RR = 0.60; 95% CI: 0.27, 1.32), however,
analyses restricted to cohort studies yielded risk estimates industry affiliated case-control studies detected a reduced risk of
exceeding 1.00, suggesting that smokers were at an increased AD associated with cigarette smoke (OR = 0.86; 95% CI: 0.75, 0.98).
risk of AD. Although cohort studies are generally less susceptible to The pooled analysis of unaffiliated case-control (OR = 0.91; 95%
bias, the analysis of cohort studies was based on findings from a CI = 0.75, 1.10) failed to detect an association. Alternatively, when
limited number of heterogeneous studies (Almeida et al., 2002). unaffiliated cohort studies were quantitatively synthesized,
Due to these inconsistent findings, the reviewers concluded that smoking was associated with a detrimental effect (RR = 1.45;
the association between smoking and risk of AD was inconclusive. 95% CI = 1.16, 1.80). Overall, when the analysis was restricted to
The systematic review and meta-analysis by Anstey et al. (2007) unaffiliated cohort studies, smoking appeared to be associated
examined the association between smoking and risk of AD and with a detrimental effect (Cataldo et al., 2010).
other cognitive outcomes. Ten studies, representing 13786 Lee et al. (2010) systematically reviewed the literature for
participants, provided data pertaining to the effect of smoking longitudinal, community-cohort studies which examined the risk
on the development of AD. Current smokers had an increased risk of AD associated with smoking among elderly subjects (65 years).
of AD when compared to never (RR = 1.79; 95% CI = 1.43, 2.23; test Smoking was consistently associated with an increased risk of
for heterogeneity: p = 0.50) and former (RR = 1.70; 95% CI = 1.25, AD in all four included cohort studies.
2.31; test for heterogeneity: p = 0.60) smokers. The pooled risk The HTA report by Williams et al. (2010) examined the effect of
estimate did not reach statistical significance, however, when smoking on risk of AD.
current smoking was compared to the combined category of past The report identified two relevant systematic reviews (Peters
and never smoking (RR = 1.25; 95% CI = 0.49, 3.17; test for et al., 2008c; Anstey et al., 2007), however, only findings reported
heterogeneity: p = 0.03). The aforementioned analysis was limited by Anstey et al. (2011) were considered. According to the results of
by the small number of studies (n = 2) reporting on this association the identified review, current but not former smoking was
as well as obvious heterogeneity (Anstey et al., 2007). Compared to associated with an increased risk of AD. Two cohort studies were
never smoking, ever (RR = 1.21; 95% CI = 0.66, 2.22; test for also identified and included in the report. Both studies reported
heterogeneity: p = 0.01) and former (RR = 1.01; 95% CI = 0.83, risk estimates for current and former smoking and also presented
1.23; test for heterogeneity: p = 0.26) smoking was not associated results stratified by APOE e4 status (Williams et al., 2010). Both
with AD. According to the reviewers, the former and ever smoking studies detected an increased risk of AD associated with current
exposure categories were poorly defined and may include a group but not former smokers. Both studies reported an increased risk of
of people with diverse smoking histories with respect to quantity AD associated with current smoking among APOE e4 non-carriers
and duration of exposure (Anstey et al., 2007). These heteroge- but not among e4 allele carries (Williams et al., 2010). Based on the
neous exposure categories serve as a limitation of the review and available literature, the reviewers concluded that current smoking
may offer an explanation for the null findings for these exposure was associated with an increased risk of AD.
groups (Anstey et al., 2007).
The systematic review conducted by Patterson et al. (2007) 3.1.33.2. Primary studies. Current (HR = 2.03; 95% CI = 1.34, 3.09)
examined the effect of several putative risk factors on the but not past (HR = 1.01; 95% CI = 0.66, 1.55) smoking was associated
development of AD and other cognitive outcomes, including with an increased risk of AD according to the prospective cohort
unspecified dementia and vascular dementia. One cohort study study of Chinese subjects reported by Zhou et al. (2011).
and one systematic review were identified. The cohort study failed Current smoking was significantly associated with risk of
to detect an association (RR = 1.10; 95% CI = 0.94, 1.29). The probable LOAD according to results reported by Reitz et al. (2010b)
identified systematic review and meta-analysis (Almeida et al., (HR = 2.52; 95% CI = 1.03, 6.19). Results failed to reach statistical
2002) restricted to methodologically sound prospective cohort significance, however, when possible LOAD was included in the
studies detected a two-fold increased risk of AD associated with analysis (HR = 1.98; 95% CI = 0.88, 4.49).
smoking (OR = 1.99; 95% CI = 1.33, 2.98). The nested case-control study by Gelber et al. (2012) examined
Five longitudinal studies were identified by Weih et al. (2007). the effect of midlife lifestyle risk factors, namely, BMI, smoking
All three studies that examined the effect of current smoking status, diet, and physical activity on the development of AD among
reported an increased risk of AD. Smoking was not associated with Japanese-American men enrolled in the Honolulu-Asia Aging
risk of AD according to the remaining two studies. Study. Former (OR = 1.15; 95% CI = 0.71, 1.87) and current (OR = 0.97;
The effect of current and former smoking on the risk of AD and 95% CI = 0.59, 1.60) smoking categories were not associated with
other cognitive outcomes were reviewed by Peters et al. (2008c). risk of AD when compared to never smoking.
Eleven studies reporting on the association between smoking and The study reported by Ronnemaa et al. (2011) examined the risk
risk of AD were identified. Seven of the ten studies reported a of AD associated with vascular risk factors considered individually
statistically significant increased risk of AD associated with current and in combination. Current smoking status, reported at midlife
smoking. According to the random effects meta-analysis, current and late-life, was not associated with risk of AD when compared to
smoking was associated with an increased risk of AD (RR = 1.59; non-smoking (Midlife, HR = 1.00; 95% CI = 0.70, 1.40. Late-life,
95% CI = 1.15, 2.20; Cochran Q = 23.25, p = 0.0015). All eight studies HR = 0.80; 95% CI = 0.40, 1.20).
which examined the effect of former smoking on AD risk failed to The association between vascular risk factors and risk of AD
detect an association. In the random effects meta-analysis, former among Koreans was prospectively examined by Kimm et al. (2011).
smokers were not at an increased risk of developing AD when Using never smoking as the referent category, both former
compared to non-smokers (RR = 0.99; 95% CI = 0.81, 1.23; Cochran (HR = 1.00; 95% CI = 0.80, 1.20) and current (HR = 1.10; 95% CI =
Q = 11.27, p = 0.08). 0.90, 3.40) smoking were not associated with risk of AD among
The systematic review and meta-analysis by Cataldo et al. men. Alternatively, current (HR = 1.30; 95% CI = 1.10, 1.50) but not
(2010) examined the risk of Alzheimer's disease associated with former (HR = 1.20; 95% CI = 0.90, 1.50) smoking increased risk of AD
cigarette smoking. The reviewers adjusted for industry affiliation, a in women. Stratified analysis detected a marginally significant
factor which may influence study findings, introduce bias, and lead association between current smoking and risk of AD among
to spurious results (Cataldo et al., 2010). A total of 43 studies (26 women 65 years of age and older (HR = 1.20; 95% CI = 1.00, 1.50) and
164 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

no association among those less than 65 years of age (HR = 1.40; and 76% (OR = 0.24; 95% CI = 0.09, 0.60) reduced risk of AD
95% CI = 0.90, 2.10). Smoking was marginally associated with risk of according to analyses restricted to cohort and case-control
AD among men less than 65 years of age (HR = 1.30; 95% CI = 1.00, studies, respectively.
1.70) but not among those 65 years or older (HR = 0.90; 95% The systematic review by Weih et al. (2007) included seven
CI = 0.70, 1.10). longitudinal studies. Physical activity had a protective effect
The prospective cohort study reported by Rusanen et al. (2011) against the development of AD according to two studies. Four of
examined the effect of midlife smoking on the risk of developing the five remaining studies reported risk estimates below 1.00,
AD in late-life. Risk estimates were derived for former smoking and however, results failed to reach statistical significance. Despite the
for current smoking, which was stratified by quantity/pack(s) number of negative findings, the reviewers concluded that physical
smoked per day. Using non-smokers as the referent category, activity may reduce risk of AD.
former smokers were not at an increased risk of developing AD The systematic review conducted by Patterson et al. (2007)
(HR = 1.00; 95% CI = 0.89, 1.13). With respect to current smoking, examined the effect of several putative risk factors on the
only the heaviest smokers (2 packs/day) were at a significantly development of AD and other cognitive outcomes, including
increased risk of AD (HR = 2.57; 95% CI = 1.63, 4.03). The remaining unspecified and vascular dementia. All three studies included in
current smoking categories were not significantly associated with the review consistently detected a reduced risk of AD attributed to
risk of AD, however, risk estimates exceeded 1.00 for the 0.5-1 physical activity.
pack/day and 1–2 pack/day categories. Overall, the authors Ten studies identified by Rolland et al. (2008) reported on the
concluded that midlife smoking, particularly in heavy quantities, effect of physical activity on the risk of developing AD, although
increased risk of AD in late-life. risk estimates specific to AD were not specified for a majority of
studies. Seven of the ten studies detected an association between
3.1.33.3. Discussion. Considered cumulatively, the identified physical activity, or at least one measure of activity, and risk of AD/
systematic reviews consistently concluded that smoking dementia. The remaining three studies failed to detect an
increased risk of AD. The increased risk of AD, however, was association. Based on the available evidence, the reviewers
only apparent in current smokers. Four of the six observational concluded that physical activity may protect against the develop-
studies identified in the second phase of the review detected a ment of AD and dementia.
detrimental effect attributed to current smoking status. The effect of physical activity on the development of AD, all-
Former smokers, or those reporting to have ever smoked, were cause dementia, and Parkinson’s disease was reviewed by Hamer
not at an increased risk of developing AD. As suggested, failure to and Chida (2009). Six studies were eligible for inclusion in the
detect an association may be attributed to the fact that these pooled analysis for the outcome of AD. Results of the random
exposure categories were poorly defined (Anstey et al., 2007; effects meta-analysis, representing 13771 subjects, suggest that
Peters et al., 2008c). Such heterogeneity may impede the detection physical activity was associated with a 45% reduced risk of AD
of an association between AD and former/ever smoking, if one (RR = 0.55; 95% CI = 0.36, 0.84; test for heterogeneity: x2 = 29.12,
exists (Anstey et al., 2007; Peters et al., 2008c). p< 0.001). Using subgroup analysis, the reviewers detected that
A number of case-control studies included in the identified the protective association was more pronounced in methodologi-
systematic reviews detected a reduced risk of AD attributed to cally rigorous studies. However, risk estimates for the subgroup
smoking. These findings were likely attributed to sources of bias analysis were not reported for the outcome of AD.
inherent in the methodological design of such studies (Cataldo The systematic review by Stern and Konno (2009) examined the
et al., 2010). This assumption was confirmed when findings of case- effect of physical leisure activities on the prevention of all-cause
control studies were compared to those derived from methodo- dementia and AD. Two case-control and 11 cohort studies
logically rigorous prospective studies, which consistently yielded reporting on the risk of AD were eligible for inclusion. The only
conflicting findings (Cataldo et al., 2010). When the analysis was study that reported on activities during early adulthood deter-
restricted to cohort studies, there was consensus regarding the mined that cases were less likely to engage in a range of activities
detrimental effect of smoking. when compared to controls (OR = 2.67; 95% CI = 1.85, 3.85) (Stern
and Konno, 2009; Friedland et al., 2001). With respect to activities
3.1.34. Physical activity engaged in during midlife, three of the four studies reporting on
We identified fifteen systematic reviews reporting on the effect the risk of AD detected a protective effect. Seven of the nine studies
of physical activity (Podewils, 2003; Fratiglioni et al., 2004; reporting on the risk of AD associated with late-life engagement in
Penrose, 2005; Weih et al., 2007, 2010; Patterson et al., 2007; physical activities detected an association. Considered cumula-
Rolland et al., 2008; Hamer and Chida, 2009; Stern and Konno, tively, the reviewers concluded that the available evidence did not
2009; Purnell et al., 2009; Williams et al., 2010; Lee et al., 2010; consistently support a protective effect of physical activities for the
Barnes and Yaffe, 2011; ESHRE Capri Workshop Group, 2011; prevention of AD and all-cause dementia. When the analysis was
Hurley et al., 2011). Six reviews were deemed to be of low restricted to studies reporting on risk of AD specifically, three of
methodological quality according to the AMSTAR criteria (Frati- four and seven of nine studies reporting on midlife and late-life
glioni et al., 2004; Penrose, 2005; Purnell et al., 2009; Hurley et al., physical activities, respectively, detected an association. The
2011; ESHRE Capri Workshop Group, 2011; Barnes and Yaffe, 2011). protective effect was diluted when studies reporting solely on
Data was abstracted from the remaining nine systematic reviews all-cause dementia were also considered (3 of 5 for midlife
and results were qualitatively synthesized. One primary study was activities and 9 of 12 for late-life activities).
identified in the second phase of the review (Gelber et al., 2012) Lee et al. (2010) systematically reviewed the literature for
(see Tables 40 and 104 in Supplementary material II). longitudinal, community-cohort studies which examined the risk
of AD associated with physical activity among elderly subjects
3.1.34.1. Systematic reviews. The effect of physical activity on risk (65 years). Of the six relevant studies, four reported a statistically
of all-cause dementia, AD, and vascular dementia was examined in significant association.
the systematic review and meta-analysis by Podewils et al. (2003). The HTA reviewed the available literature to examine the
Seven cohort and four case-control studies were eligible for association between physical activity and subsequent risk of AD
inclusion. According to the random effects meta-analysis, physical (Williams et al., 2010). Twelve cohort studies were identified, nine
activity was associated with a 41% (RR = 0.59; 95% CI = 0.42, 0.77) of which were eligible for inclusion in a pooled analysis. The
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 165

random effects model yielded a risk estimate of 0.72 (95% CI = 0.53, and the risk of AD. The review identified zero randomized
0.98; Q = 23.25, p = 0.003, I2 = 66%) suggesting a 28% reduced risk of controlled trials.
AD associated with engagement in physical activities. Based on the Six prospective cohort studies reporting on the effect of PUFAs
available evidence, the reviewers concluded that physical activity were identified by Dangour et al. (2010). Only one of the six studies
may reduce risk of AD. The reviewers caution, however, that the reported a statistically significant reduced risk of AD associated
observed protective effect may be attributed to confounding by with fish consumption, omega-3 fatty acids, and a high intake of
other putative protective factors that are associated with a DHA.
physically active lifestyle. Both studies included in the review reported by Lee et al. (2010)
The effect of physical activity on risk of developing AD was consistently demonstrated a reduced risk of AD associated with
reviewed by Weih et al. (2010). PUFAs.
Six studies satisfied the inclusion criteria. All studies reported Williams et al. (2010) collated evidence pertaining to the
risk estimates below 1.00 and all but one reached statistical association between omega-3 fatty acids and risk of AD from
significance. According to the random effects meta-analysis, longitudinal cohort studies. Of the seven included studies
compared to those with a sedentary lifestyle, physically active reporting on the effect of fish consumption, only three reported
subjects had a 41% reduced risk of AD (RR = 0.59; 95% CI = 0.50, a statistically significant reduced risk of AD. Similarly conflicting
0.69). There was no evidence of statistical heterogeneity across findings were observed in the analysis examining the effect of
studies (test for heterogeneity: x2 = 1.84, df = 5, p = 0.87). PUFAs derived from unspecified dietary sources or ascertained
according to plasma/serum levels.
3.1.34.2. Primary studies. We identified a single observational
study reporting on the association between physical activity and 3.1.35.2. Primary studies. The case-cohort study reported by Lopez
risk of AD. The nested case-control study by Gelber et al. (2012) et al. (2011) examined the effect of plasma and dietary
examined the effect of midlife lifestyle risk factors, namely, BMI, docosahexaenoic acid (DHA) and fish consumption on the risk
smoking status, diet, and physical activity, on the development of of AD. Analyses treating plasma and dietary DHA exposure as a
AD among Japanese-American men enrolled in the Honolulu-Asia continuous variable (per log SD increase) yielded risk estimates
Aging Study. Physical activity, treated as a dichotomous variable below 1.00, however, results failed to reach statistical significance
(high vs. low), was not significantly associated with risk of AD (Plasma, OR = 0.76; 95% CI = 0.50, 1.14. Dietary, OR = 0.52; 95%
(OR = 1.44; 95% CI = 0.94, 2.21). CI = 0.32, 0.84). Alternatively, when treated as a categorical
variable, low plasma (OR = 2.33; 95% CI = 1.04, 5.21) and dietary
3.1.34.3. Discussion. Eight of nine identified systematic reviews (OR = 3.43; 95% CI = 1.42, 8.26) DHA significantly increased odds of
consistently concluded that engagement in physical activities may AD. High dietary (OR = 0.28; 95% CI = 0.09, 0.93) and plasma
reduce risk of AD. The only identified observational study failed to (OR = 0.40; 95% CI = 0.15, 1.10) DHA were associated with a reduced
detect an association between physical activity and AD. Despite the odds of AD, although results for plasma DHA failed to reach
various limitations of observational studies, evidence collated in statistical significance. The risk estimate for fish consumption was
several systematic reviews offer convincing evidence regarding the below 1.00, however, results failed to reach statistical significance
protective effect of physical activity against the development of (OR = 0.55; 95% CI = 0.20, 1.48). Considered cumulatively, results of
AD. the study suggest that higher plasma and dietary DHA may reduce
risk of dementia and AD (Lopez et al., 2011).
3.1.35. Polyunsaturated fatty acids (PUFAs)
Nine reviews examining the association between polyunsatu- 3.1.35.3. Discussion. Seven higher quality systematic reviews,
rated fatty acids (PUFAs) and risk of AD were identified (Stand- collating evidence from 12 longitudinal studies reporting on the
ridge, 2004; Issa et al., 2006; Lim et al., 2006; Weih et al., 2007; association between PUFAs and risk of AD, were identified. Larger
Patterson et al., 2007; Fotuhi et al., 2009; Williams et al., 2010; Lee systematic reviews by Williams et al. (2010) and Dangour et al.
et al., 2010; Dangour et al., 2010). Two of the reviews deemed to be (2010) identified a number of negative studies. One recent
of low methodological quality were excluded (Standridge, 2004; observational study, which detected a protective effect
Fotuhi et al., 2009). One primary study was identified in the second associated with higher plasma and dietary DHA, was identified
phase of the review (Lopez et al., 2011). Refer to in the second phase of our review (Lopez et al., 2011). Considered
cumulatively, there was insufficient evidence to support a
3.1.35.1. Systematic reviews. Issa et al. (2006) reviewed the effect beneficial effect of PUFAs against the development of AD.
of omega-3 fatty acid intake on cognitive function and incidence of
dementia. Two of the three included studies reported statistically 3.1.36. Vitamin B12
significant findings suggesting a reduced risk of AD associated with Six systematic reviews examined the association between
fish consumption. One study reported that total n-3 fatty acid vitamin B-12 and risk of AD (Standridge, 2004; Raman et al., 2007;
consumption and a high intake of DHA, but not EPA or ALA, was Purnell et al., 2009; Williams et al., 2010; Dangour et al., 2010; Lee
associated with a reduced risk of AD. et al., 2010). Two reviews were deemed to be of low methodologi-
Weih et al. (2007) identified three longitudinal cohort studies. cal quality and were consequently excluded (Standridge, 2004;
Two of the three included studies reported a statistically Purnell et al., 2009). No primary studies were identified.
significant reduced risk of dietary fish consumption and one
detected a protective effect associated with total n-3 fatty acids. 3.1.36.1. Systematic reviews. The effect of B-vitamins on the risk of
The remaining study reported a marginally significant protective AD was examined by Raman et al. (2007). The reviewers identified
effect. twelve studies which examined the association between blood
Patterson et al. (2007) identified a single study which reported concentrations or dietary intake of B-vitamins and risk of AD. All
on the effect of fish consumption on incident AD. The study seven studies reporting on blood concentrations of vitamin B-12
detected a 70% decreased risk of AD (RR = 0.30; 95% CI = 0.10, 0.90) failed to detect an association. The only study to report on the
associated with fish consumption exceeding 18.5 g/day. effect of dietary intake of B-12 also failed to detect an association
The high-quality Cochrane systematic review conducted by Lim (RR = not reported). Based on these findings, the reviewers
et al. (2006) examined the association between omega-3 fatty acid concluded that vitamin B-12 was not associated with AD.
166 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Five prospective cohort studies identified by Dangour et al. vitamin B-6 and risk of AD. Conflicting findings and the limited
(2010) examined the association between vitamin B-12 and AD number of available studies suggest that the association, if any, was
risk. Serum/plasma vitamin B-12 was not associated with risk of unclear (Raman et al., 2007).
AD according to all three studies reporting on this association.
However, one study, which examined the combined effect of serum 3.1.38. Vitamin B3/Niacin
vitamin B-12 and folate, reported a greater than two-fold increased Two moderate quality systematic reviews, which reported on
risk of AD among those with low serum vitamin B-12 and low the association between vitamin B-3/niacin and risk of AD, were
serum folate (RR = 2.10; 95% CI = 1.20, 3.50). All three studies identified (Williams et al., 2010; Lee et al., 2010). The search for
reporting on dietary intake (food and supplements) of vitamin B- primary studies did not yield any relevant records.
12 failed to detect a statistically significant protective effect
associated with higher consumption. Considered cumulatively, the 3.1.38.1. Systematic reviews. One study was identified by Williams
reviewers concluded that the effect of vitamin B-12 on AD risk was et al. (2010). The highest quintile of total niacin/vitamin B3 intake
inconclusive. was associated with a protective effect against the development of
Four cohort studies examining the effect of vitamin B-12 on AD AD (OR = 0.20; 95% CI = 0.01, 0.70). In the same study, tryptophan
incidence were eligible for inclusion in the HTA reported by (OR = 0.40; 95% CI = 0.10, 0.80), niacin obtained from food
Williams et al. (2010). All four studies failed to detect a statistically (OR = 0.30; 95% CI = 0.10, 0.70), and “niacin equivalents”
significant association between AD and low dietary intake or low (OR = 0.20; 95% CI = 0.10, 0.80) were all associated with a
serum concentrations of vitamin B12. statistically significant reduced risk of AD.
Lee et al. (2010) identified a single cohort study which reported The review by Lee et al. (2010) examined the association
on the effect of vitamin B12. There was no significant association between a number of lifestyle-related risk factors and AD. One of
between serum concentrations of vitamin B-12 and risk of AD the included studies examined the association between niacin
according to the study. intake and risk of AD. Higher intake of niacin was associated with a
statistically significant reduced risk of AD (RR = 0.30; 95% CI = 0.10,
3.1.36.2. Discussion. Results from the identified moderate quality 0.70).
systematic reviews suggest that vitamin B-12 was not associated
with a reduced risk of AD. Although studies that rely on self- 3.1.38.2. Discussion. Only one prospective cohort study examining
reported dietary intake collected by food frequency questionnaires the effect of vitamin B-3/niacin on risk of AD was identified in two
or food diaries are susceptible to exposure measurement error systematic reviews. Results from one recent primary study
(Kristal et al., 2005), primary studies that employed more objective suggests that higher intake of niacin is associated with a
measures for exposure ascertainment, namely serum protective effect against the development of AD.
concentrations, reported consistent findings suggesting a lack of
association between vitamin B-12 and risk of AD. However, 3.1.39. Folic Acid/Folate
conclusions drawn from the reviews were limited by the small Six systematic reviews reporting on the effect of folate were
number of available studies. Further research is required to identified (Standridge, 2004; Weih et al., 2007; Raman et al., 2007;
confirm the effect of vitamin B-12 on the risk of AD. Dangour et al., 2010; Williams et al., 2010; Lee et al., 2010). One
review was of low methodological quality and was excluded
3.1.37. Vitamin B6 (Standridge, 2004). A single primary study was identified
Five systematic reviews examined the association between (Hooshmand et al., 2010).
vitamin B-6 and risk of AD (Standridge, 2004; Raman et al., 2007;
Purnell et al., 2009; Dangour et al., 2010; Williams et al., 2010). Two 3.1.39.1. Systematic reviews. Nine studies identified by Raman
studies were excluded due to low methodological quality (Stand- et al. (2007) examined the association between blood folate levels
ridge, 2004; Purnell et al., 2009). No primary studies were and risk of AD. Four of the nine studies reported a detrimental
identified. effect of low blood folate levels. The two studies that reported on
dietary folate intake reported conflicting findings. One detected an
3.1.37.1. Systematic reviews. Raman et al. (2007) identified two association between low folate intake and AD while the other
studies which reported on the effect of blood concentrations of reported null findings.
vitamin B-6 on risk of AD. Both failed to detect an association. With One study identified by Weih et al. (2007) reported on folate
respect to dietary intake, one study detected an association while intake. According to the study, low folate was associated with a
the other reported null findings. The reviewers concluded that the greater than two-fold increased risk of AD (RR = 2.10; 95% CI = 1.20,
available evidence did not implicate low vitamin B-6 as a risk factor 3.50).
for the development of AD. The systematic review by Dangour et al. (2010) examined the
Both cohort studies included in the HTA reported by Williams association between several nutrients, including folate, and risk of
et al. (2010) failed to detect an association between vitamin B6 and AD. A total of eight cohort studies investigating the association
risk of AD. Based on the identified evidence, the reviewers between serum folate levels or folate intake were identified. Of the
concluded that vitamin B-6 was not associated with risk of AD. five studies investigating the association between serum folate
Of the four prospective cohort studies identified by Dangour levels and AD, only one detected an association (HR = 1.98; 95%
et al. (2010), two studies failed to detect an association between CI = 1.15, 3.40). Two of three studies reporting on dietary and
dietary intake of vitamin B-6 and risk of AD and one study failed to supplemental folate intake detected a protective effect, attributed
detect an association between plasma vitamin B-6 levels and risk to higher intake. Overall, three of the eight included cohort studies
of AD. The remaining cohort study reported conflicting findings, detected a reduced risk of AD associated with higher serum folate
suggesting a protective effect attributed to high vitamin B-6 concentrations and folate intake.
consumption. The reviewers concluded that vitamin B-6 did not Lee et al. (2010) identified two cohort studies. One study
appear to be associated with risk of AD. detected a near two-fold (HR = 1.98; 1.15, 3.40) increased risk of AD
associated with low serum folate concentrations (11.8 nmol/l
3.1.37.2. Discussion. All three moderate-quality systematic vs. >15.2 nmol/l). The remaining study failed to detect an
reviews consistently failed to detect an association between association.
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 167

Williams et al. (2010) examined the effect of folate on the risk of vitamin C and E (HR = 0.36; 95% CI = 0.09, 0.99), however, isolated
AD. Of the two included studies that examined serum folate, low supplementation of vitamin C or vitamin E was not protective
concentrations were associated with a near two-fold increased risk against the development of AD. The final study reported
of AD (HR = 1.98; 95% CI = 1.15, 3.40) in one study and only a conflicting findings, suggesting an increased risk of AD associated
marginally significant 70% (HR = 1.70; 95% CI = 1.00, 3.40) increased with the second and fourth quartile of vitamin E intake. Based on
risk in the second study. High total folate intake was associated the available evidence, the reviewers concluded that there was
with a statistically significant reduced risk of AD in one of two insufficient evidence to suggest that vitamins C and E were
studies. Findings from the review suggested that while low folate related to risk of AD.
serum concentrations were associated with an increased risk of
AD, the association between total folate intake and AD was 3.1.40.2. Primary studies. We identified one relevant primary
inconclusive. study which reported on the effect of vitamins C and E. Using data
from the Rotterdam Study, Devore et al. (2010) prospectively
3.1.39.2. Primary studies. We identified a single primary study examined the effect of dietary intake of vitamins C and E, beta-
which reported on the association between serum folate and risk carotene, and flavonoids on risk of AD. Using the lowest tertile of
of AD (Hooshmand et al., 2010). The Cardiovascular Risk Factors, consumption as a referent, higher tertiles of vitamin C intake were
Aging, and Dementia (CAIDE) study failed to detect an association not associated with risk of AD. The highest tertile of vitamin E
between serum folate and risk of AD (OR = 1.01; 95% CI = 0.87, 1.18). consumption, however, was associated with a statistically
significant reduced risk of AD. These findings suggest that
3.1.39.3. Discussion. A recent primary study failed to detect an higher dietary consumption of vitamin E may confer a
association between serum folate and risk of AD (Hooshmand protective effect against the development of AD.
et al., 2010). Findings from four moderate systematic reviews
suggest that the effect of dietary, supplemental and serum folate 3.1.40.3. Discussion. Based on four moderate quality systematic
concentrations and risk of AD was inconclusive. reviews, the combined and isolated effect of vitamins C and E on
risk of AD was inconclusive. A majority of the primary studies
3.1.40. Vitamins C and E included in the identified systematic reviews reported null
Six systematic reviews examining the effect of vitamins C and/ findings. The only primary study which we identified in the
or E on the risk of AD were identified (Standridge, 2004; Pham and second phase of the review detected a protective effect attributed
Plakogiannis, 2005; Weih et al., 2007; Patterson et al., 2007; to high consumption of vitamin E but not C (Devore et al., 2010).
Williams et al., 2010,,2010). Two reviews were excluded due to low Further research into the effect of dietary and supplemental intake
methodological quality (Standridge, 2004; Pham and Plakogiannis, of vitamin C and E on the risk of AD is required (Williams et al.,
2005). One relevant primary study was identified in the second 2010).
phase of the review (Devore et al., 2010).
3.1.41. Multivitamins
3.1.40.1. Systematic reviews. Three cohort studies identified by
Two systematic reviews reported on the effect of multivitamin
Weih et al. (2007) examined the effect of vitamin C, vitamin E or
use on the risk of AD (Standridge, 2004; Williams et al., 2010). One
the combined effect of these vitamins on the risk of AD. One of two
study was excluded due to low methodological quality (Standridge,
studies reporting on vitamin C detected an association. Two
2004). No primary studies were identified.
studies reported a statistically significant (RR = 0.30; 95% CI = 0.10,
The HTA reviewed the effect of supplemental multivitamin use
0.92) and a marginally significant (RR = 0.82; 95% CI = 0.66, 1.00)
on the risk of incident AD (Williams et al., 2010). Both included
reduced risk of AD associated with increasing dietary vitamin E
cohort studies failed to detect a protective effect attributed to
consumption. One study examined the combined effect of vitamin
multivitamin use.
C and E intake. The combined effect of vitamin C and E intake was
Multivitamin use did not appear to reduce the risk of AD. These
not associated with risk of AD (RR = 1.81; 95% CI = 0.91, 3.63). The
findings were limited by the very small number of available studies
reviewers concluded that the effect of vitamins C and E on AD was
which precludes a definitive conclusion.
unclear.
One study identified by Patterson et al. (2007) examined the
3.1.42. Flavonoids
effect of low serum vitamin E on AD risk. The lowest tertile of
Three systematic reviews examined the association between
serum vitamin E was not significantly associated with an increased
flavonoid intake and risk of AD (Standridge, 2004; Williams et al.,
risk of AD (RR = 2.10; 95% CI: 0.81, 5.54).
2010; Lee et al., 2010). One review was excluded due to low
Lee et al. (2010) identified four prospective cohort studies
methodological quality (Standridge 2004). A single primary study
examining the risk of AD associated with vitamin C and E intake.
was identified in the second phase of the review (Devore et al.,
Three studies failed to detect an association between dietary
2010).
intake of vitamin C and vitamin E and risk of AD. The remaining
study, however, reported a marginally significant 18% reduced risk
3.1.42.1. Systematic reviews. The systematic review by Lee et al.
of AD associated with each 1 SD increase in vitamin C (RR = 0.82;
(2010) examined the association between AD and a number of
95% CI = 0.68, 0.99) and vitamin E (RR = 0.82; 95% CI = 0.66, 1.00)
dietary factors including flavonoids. Two of the three studies
consumption. The reviewers concluded that vitamin E may reduce
identified in the HTA (Williams et al., 2010) were also identified by
risk of AD.
Lee et al. (2010). Both cohort studies failed to detect a statistically
Eleven studies were included in the HTA reported by Williams
significant association between dietary flavonoid consumption
et al. (2010). Six of the included studies failed to detect a
and risk of AD.
statistically significant association between dietary or supple-
Three cohort studies were identified and included in the HTA
mental intake of vitamins C and/or E and risk of AD. Of the
reported by Williams et al. (2010). Two of the three studies failed to
remaining five studies, three reported a marginally or statistically
detect an association. One study reported that higher consumption
significant reduced risk of AD associated with higher intake of
of dietary flavonoids was associated with a statistically significant
vitamin C or vitamin E. One reported a statistically significant
reduced risk of AD (HR = 0.49; 95% CI = 0.26, 0.92).
reduced risk of AD associated with combined supplementation of
168 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

3.1.42.2. Primary studies. A single primary study was identified Rotterdam Study, Devore et al. (2010) prospectively examined the
which reported on the association between flavonoids and risk of effect of dietary intake of vitamins C and E, beta-carotene, and
AD. According to the Rotterdam Study (Devore et al., 2010), there flavonoids on risk of AD. According to the results of the study, there
was no association between flavonoid intake and risk of AD was no association between dietary beta carotene and risk of AD
(Middle tertile, HR = 1.11; 95% CI = 0.85, 1.44. Highest tertile, (Middle tertile, HR = 1.02; 95% CI = 0.79, 1.31. Highest tertile,
HR = 1.09; 95% CI = 0.84, 1.42). HR = 1.07; 95% CI = 0.83, 1.39).

3.1.42.3. Discussion. One recent primary study failed to detect an 3.1.44.3. Discussion. Three moderate quality systematic reviews
association between flavonoids and risk of AD. Results summarized summarizing evidence from five cohort studies suggest that beta-
in two moderate quality systematic reviews indicate that there is carotene and other carotenoids are not associated with a beneficial
insufficient evidence to suggest that flavonoids reduce risk of AD. effect. A recent cohort study also failed to detect an association.
Further research into the effect of flavonoids is required.
3.1.45. Caffeine consumption
3.1.43. Fruit and vegetable intake Three moderate quality systematic reviews examined the effect
Two moderate-quality systematic reviews examined the effect of caffeine consumption on the risk of AD (Barranco Quintana et al.,
of fruit and vegetable intake and risk of incident AD (Williams 2007; Santos et al., 2010; Lee et al., 2010). One relevant primary
et al., 2010; Lee et al., 2010). No primary studies were identified. study was identified in the second phase of the review (Gelber
et al., 2011).
3.1.43.1. Systematic reviews. Two cohort studies were eligible for
inclusion in the HTA reported by Williams et al. (2010). Both 3.1.45.1. Systematic reviews. Barranco Quintana et al. (2007)
studies detected a statistically significant association. One study identified two case-control and two cohort studies. Two of the
reported that consumption of fruit and vegetable juice three or four identified studies detected a protective effect attributed to
more times per week significantly reduced risk of AD (HR = 0.24; coffee consumption. The random effects meta-analysis failed to
95% CI = 0.09, 0.61). Moderate or high intake of fruits and detect an association between coffee consumption and AD (Risk
vegetables in midlife was associated with a reduced risk of AD estimate: 0.79; 95% CI = 0.46, 1.36). However, sensitivity analyses
according to the second study (OR = 0.60; 95% CI = 0.41, 0.86). Based restricted to cohort studies (Risk estimate: 0.73; 95%: CI 0.54, 0.99),
on the available evidence, the reviewers concluded that high studies employing interview questionnaires (Risk estimate: 0.70;
consumption of fruits and vegetables may reduce the risk of AD. 95% CI = 0.55, 0.90), and studies that were less susceptible to bias
However, these findings were limited by the small number of (Risk estimate: 0.58; 95% CI = 0.44, 0.77), all support the protective
available studies. effect of coffee consumption against the development of AD.
Lee et al. (2010) identified one cohort study which examined Because of the limited number of available studies and the
the effect of fruit and vegetable intake on risk of AD. According to presence of heterogeneity across these studies, further research is
the results of this study, consumption of fruit and vegetable juices warranted (Barranco Quintana et al., 2007).
three or more times per week was associated with a 76% Two case-control and three cohort studies were included in the
(HR = 0.24; 95% CI = 0.09, 0.61) decreased risk of AD. systematic review reported by Santos et al. (2010). Among the five
included studies, three failed to detect a reduced risko of AD
3.1.43.2. Discussion. The available evidence, summarized in two associated with coffee consumption. The remaining two studies
moderate-quality systematic reviews, suggest that higher intake of reported a 31% and 60% reduced risk of AD among coffee
fruits and vegetables may decrease risk of AD. However, the small consumers. All three studies reporting tea consumption failed to
number of available studies precludes a conclusive statement detect an association. Caffeine consumption was associated with a
regarding the effect of fruit and vegetable intake on the risk of 38% (RR = 0.62; 95% CI = 0.45, 0.87; test for heterogeneity: p = 0.169;
incident AD (Williams et al., 2010). I2 = 40.5%) reduced risk of AD according to the pooled analysis of
four studies. Findings from this review support the protective
3.1.44. Beta-carotene effect of caffeine consumption. However, as noted by the
Four systematic reviews examined the association between reviewers, methodological heterogeneity across studies serves as
beta-carotene and risk of AD (Standridge, 2004; Weih et al., 2007; an important limitation of the review.
Williams et al., 2010; Lee et al., 2010). One review was excluded The review by Lee et al. (2010) examined the association
(Standridge, 2004). One primary study was identified in the second between a number of lifestyle-related risk factors and AD. Tea
phase of the review (Devore et al., 2010). consumption was not associated with risk of AD according to the
only relevant study.
3.1.44.1. Systematic reviews. One cohort study identified in the
systematic review by Weih et al. (2007) examined the association 3.1.45.2. Primary studies. The nested case-control study conducted
between beta-carotene intake and risk of AD. According to the by Gelber et al. (2011) examined the effect of coffee and caffeine
results of the study, low beta-carotene consumption was not consumption on the risk of developing all-cause dementia, AD,
associated with reduced risk of AD (RR = 0.55; 95% CI = 0.22, 1.35). vascular dementia, and other cognitive outcomes. Higher intake of
The HTA identified four cohort studies that examined the effect coffee was not associated with risk of AD. Higher quintiles of
of carotenoids or beta-carotene on the risk of AD (Williams et al., caffeine consumption, derived from self-reported coffee, tea, and
2010). All four studies failed to detect an association. Findings of cola intake, were also not associated with risk of AD. When caffeine
the report suggest that beta-carotene and other carotenoids are not intake was treated as a continuous variable, each 1 SD (223 mg)
associated with a reduced risk of AD (Williams et al., 2010). increase in consumption was not associated with AD. Results of
The systematic review by Lee et al. (2010) identified four cohort this study suggest that coffee and caffeine consumption were not
studies. All four studies failed to detect an association between associated with risk of AD.
beta-carotene/carotenoids and AD.
3.1.45.3. Discussion. Three moderate quality systematic reviews
3.1.44.2. Primary studies. We identified a single primary study examined the effect of caffeine intake on the risk of AD. Among the
reporting on the effect of beta-carotene. Using data from the six primary studies included in the three reviews, only two studies
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 169

reported a statistically significant association between caffeine 3.1.47. Other dietary patterns
consumption and risk of AD. Moreover, recent cohort studies, We identified a single study that prospectively examined the
including the nested case-control study identified in the second effect of multiple dietary patterns on the risk of developing AD (Gu
phase of the review (Gelber et al., 2011), also failed to detect an et al., 2010b).
association. Although the pooled analysis reported by Santos et al. Of the seven dietary patterns (DP) examined, only the second
(2010) and the subgroup analyses reported by Barranco Quintana DP was associated with risk of AD (HR = 0.54; 95% CI = 0.39, 0.75; p
et al. (2007) were suggestive of a protective effect, these findings < 0.001). This dietary pattern consisted of “higher intakes of salad
were limited by the small number of available studies. Overall, dressing, nuts, fish, tomatoes, poultry, cruciferous vegetables,
based on the conflicting findings of the observational studies, the fruits, and dark and green leafy vegetables and a lower intake of
association between caffeine consumption and risk of AD remains high-fat dairy products, red meat, organ meat, and butter” (Gu
inconclusive. et al., 2010b). Moderate and high compliance to the aforemen-
tioned dietary pattern conferred a protective effect against the
3.1.46. Mediterranean diet development of AD, however, results for moderate compliance
Six systematic reviews examined the association between failed to reach statistical significance (Middle tertile, HR = 0.81;
adherence to the Mediterranean diet and risk of AD (Weih et al., 95% CI = 0.59, 1.12. Highest tertile, HR = 0.62; 95% CI = 0.43, 0.89).
2007; Roman et al., 2008; Sofi et al., 2008, 2010; Williams et al.,
2010; ESHRE Capri Workshop Group, 2011). Two reviews were of 3.1.48. Fatintake, saturated fatty acids
poor methodological quality and were consequently excluded. One Four moderate quality systematic reviews, which examined the
primary study was identified in the second phase of the review (Gu association between total fat, saturated fat, or transunsaturated fat
et al., 2010a). intake and risk of AD, were identified (Weih et al., 2007; Williams
et al., 2010; Lee et al., 2010; Crichton et al., 2010). No primary
3.1.46.1. Systematic reviews. The effect of adherence to the studies were located.
Mediterranean diet was examined in the review by Weih et al.
(2007). Evidence from the only identified study suggested that 3.1.48.1. Systematic reviews. Three cohort study identified in the
high adherence to the Mediterranean was associated with a systematic review by Weih et al. (2007) examined the association
reduced risk of AD (HR = 0.60; 95% CI = 0.40, 0.90). between fat intake and risk of AD. All three studies reported risk
The systematic review by Sofi et al. (2008) examined the effect estimates exceeding 1.00 for total or saturated fat intake, however,
of the Mediterranean dietary pattern on the risk of numerous results from only one study reached statistical significance.
outcomes including Alzheimer's disease, Parkinson's disease, Specifically, the study reported an increased risk of AD
cancer, and mortality. The only study which focused on the associated with saturated and transunsaturated dietary fat but
outcome of AD reported a protective effect attributed to higher not with total fat intake. The reviewers concluded that dietary fat
adherence (RR = 0.83; 95% CI = 0.70, 0.98). may be associated with an increased risk of AD.
The updated review reported by Sofi et al. (2010) identified one Crichton et al. (2010) examined the association between fat
newly published study focused on the risk of AD. The prospective intake from dairy products and risk of AD. The reviewers identified
cohort study failed to detect an association between high a single study which focused on the risk of developing AD.
adherence to a Mediterranean diet and development of AD The study reported that fat intake from milk or sour milk was
(RR = 1.00; 95% CI = 0.71, 1.40). not associated with risk of AD. Moderate intake of saturated fatty
Four cohort studies, three of which examined the same cohort, acids from spreads was associated with a near four-fold increased
were identified by Williams et al. (2010). Three of four identified risk of AD (OR = 3.82; 95% CI = 1.48, 9.87).
studies detected a reduced risk of AD associated with adherence to The review by Lee et al. (2010) examined the association
the Mediterranean diet. Findings from this review suggest that the between a number of lifestyle-related risk factors and AD. Two
Mediterranean diet may exert a protective effect against studies, reporting on the same cohort, examined the association
incident AD, however, further research is required (Williams between saturated fatty acids obtained from spreads and risk of
et al., 2010). AD. Higher saturated fatty acid intake from milk, sour milk, and
spreads was associated with an almost four fold increased odds of
3.1.46.2. Primary studies. According to results of the fully adjusted AD (OR = 3.82; 95% CI = 1.48, 9.87) according to one study. The other
analysis reported by Gu et al. (2010a), higher adherence to the reported a greater than four-fold (OR = 4.34; 95% CI = 1.28, 14.68)
Mediterranean diet, treated as a continuous variable, was increased risk of AD among APOE e4 carriers associated with
associated with a significantly reduced risk of AD (HR = 0.87; higher saturated fatty acid consumption from spreads. Results of
95% CI = 0.78, 0.97). Treated categorically, both moderate and high the review suggest that higher saturated fatty acid consumption,
adherence yielded risk estimates below 1.00, however, only the particularly obtained from spreads, may increase risk of AD.
moderate category reached statistical significance (Middle tertile, Williams et al. (2010) identified two cohort studies that
HR = 0.56; 95% CI = 0.36, 0.86. Highest tertile, HR = 0.68; 95% examined the association between dietary fats (saturated,
CI = 0.42, 1.08). Results of the study suggest that greater monounsaturated, transunsaturated) and risk of AD. One study
adherence to the Mediterranean diet is associated with a detected an increased risk of AD associated with high saturated fat
reduced risk of AD. consumption (RR = 2.20; 95% CI = 1.10, 4.70). High transunsaturated
fat consumption also increased risk of AD (Quintile 2, RR = 2.40;
3.1.46.3. Discussion. Four systematic reviews, collating evidence 95% CI = 1.10, 5.30. Quintile 3, RR = 2.90; 95% CI = 1.20, 7.20). The
from four cohort studies, were identified. A recent cohort study second study detected an increased risk of AD associated with
was also identified in the second phase of the review. Considered saturated fat obtained from spreads, however, only the second
cumulatively, the available evidence suggested that greater quartile reached statistical significance (OR = 3.82; 95% CI = 1.48,
adherence to the Mediterranean diet may be associated with a 9.87). Total and monounsaturated fats were not associated with
reduced risk of AD. However, the limited number of available risk of AD according to both studies. Based on this evidence, the
studies precludes a conclusive statement regarding the effect of reviewers concluded that saturated and transunsaturated fat
this dietary pattern. consumption may be associated with a deleterious effect (Williams
et al., 2010).
170 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

3.1.48.2. Discussion. Considered cumulatively, the available Caamano-Isorna et al. (2006), low levels of education were
evidence suggested that dietary saturated and transunsaturated associated with an increased risk of AD. Findings from two cohort
fats may be associated with an increased risk of AD. However, the studies, published after the review by Caamano-Isorna et al.
small number of available primary studies precludes any definitive (2006), reported consistent findings. One of the studies reported a
statement. statistically significant protective effect attributed to high levels of
education (9 years) (OR = 0.15; 95% CI = 0.05, 0.40). Moderate
3.1.49. Caloric intake levels of education (6–8 years) were associated with a marginally
Two moderate quality systematic reviews examined the significant (OR = 0.49; 95% CI = 0.24, 1.00) reduced risk of AD when
association between caloric intake and risk of AD (Weih et al., compared to low educational attainment. The second study
2007; Williams et al., 2010). detected an increased risk of AD associated with lower educational
attainment ( high school) compared to high levels of education
3.1.51.1. Systematic reviews. Weih et al. (2007) identified a single (graduate school) (OR = 41.48; 95% CI = 4.00, 42.40). Moderate
cohort study that examined the association between caloric intake education (undergraduate degree) was associated with an
and risk of AD. The study detected a marginally significant 50% increased risk of AD, however, results did not reach statistical
increased risk of AD associated with the highest quartile of caloric significance (OR = 2.07; 95% CI = 0.28, 15.10). Based on the evidence
intake (RR = 1.50; 95% CI = 1.00, 2.20). provided by the included systematic review and cohort studies, the
The same primary study identified by Weih et al. (2007), which reviewers concluded that higher educational attainment appeared
suggested an increased risk of AD associated with high total daily to be associated with a protective effect against the development of
caloric intake, was also included in the HRT reported by Williams AD (Williams et al., 2010).
et al. (2010).
3.1.50.2. Primary studies. According to results of the LADIS
3.1.49.2. Discussion. Results from a single cohort study suggest prospective cohort study (Verdelho et al., 2010), educational
that high caloric intake may increase risk of AD. However, as only level, treated as a continuous variable, was not significantly
one primary study was identified in both reviews, further research associated with risk of AD (HR = 0.96; 95% CI = 0.85, 1.09; p = 0.513).
is needed to clarify this association. When compared to more years of education ( > 9 years), fewer
years of education significantly increased risk of possible and
3.1.50. Education probable LOAD (7–9 years, HR = 3.01; 95% CI = 1.32, 6.87. 0–6 years,
Three moderate-quality systematic reviews, which examined HR = 4.51; 95% CI = 2.09, 9.74) according to the prospective study
the association between education and risk of AD, were identified reported by Reitz et al. (2010b). Results remained consistent when
(Caamano-Isorna et al., 2006; Patterson et al., 2007; Williams et al., the analysis was restricted to probable LOAD (7–9 years, HR = 2.93;
2010). Five observational studies were retrieved (Verdelho et al., 95% CI = 1.19, 7.23. 0–6 years, HR = 3.76; 95% CI = 1.61, 8.77).
2010; Reitz et al., 2010b; Song et al., 2011; Yang et al., 2011; Shah The longitudinal cohort study conducted by Yang et al. (2011)
et al., 2011). sought to examine the association between blood pressure,
hypertension, and risk of AD. The association between blood
3.1.50.1. Systematic reviews. The systematic review and meta- pressure measures, namely, self-reported hypertension and
analysis by Caamano-Isorna et al. (2006) examined the association objectively measured systolic (SBP) and diastolic blood pressure
between educational attainment and risk of AD. Nine cohort and (DBP), pulse pressure (PP), and mean arterial pressure (MAP) were
five case-control studies were included. Low educational analyzed in separate models. The association between education,
attainment was associated with an 80% (RR = 1.80; 95% CI = 1.43, reported as a continuous variable (1 year increase), and risk of AD
2.27; test for heterogeneity: Ri statistic: 0.61; Cochrane Q statistic was examined in each model. According to the risk estimates
p-value: <0.0001) increased risk of AD according to the pooled derived from each model, increasing education was not associated
analysis of all 14 studies. In the analysis restricted to cohort studies, with risk of AD (Self-reported HTN model, HR = 1.01; 95% CI = 0.96,
low educational attainment was associated with a 59% increased 1.06. SBP model, HR = 0.99; 95% CI = 0.94, 1.05. DBP model,
risk of AD (RR = 1.59; 95% CI = 1.35, 1.86). The pooled analysis HR = 1.00; 95% CI = 0.95, 1.05. Pulse pressure model, HR = 1.00;
restricted to case-control studies also yielded statistically 95% CI = 0.95, 1.05. Mean arterial pressure model, HR = 1.00; 95%
significant findings, however, the derived risk estimate was CI = 0.95, 1.05).
greater in magnitude (RR = 2.40; 95% CI = 1.32, 4.38). Compared Results from the Rush Memory and Aging Project (Shah et al.,
to the highest level of education, the combined category of low and 2011) failed to detect an association between education and risk of
moderate levels of education was associated with a significant 44% AD (HR = 0.98; 95% CI = 0.91, 1.05).
increased risk of AD (RR = 1.44; 95% CI = 1.24, 1.67; Ri statistic: 0.47; Lower education, defined as less than nine years, was associated
Cochrane Q statistic p-value: 0.073). Stratified analysis by with an increased risk of AD according to results from the Canadian
methodological design also yielded statistically significant Study of Health and Aging (OR = 1.37; 95% CI = 1.02, 1.83) (Song
findings (Cohort studies, RR = 1.32; 95% CI = 1.09, 1.59. Case- et al., 2011).
control studies, RR = 1.66; 95% CI = 1.30, 2.10). Based on the
available evidence, the reviewers concluded that low 3.1.50.3. Discussion. Only two of five observational studies
educational attainment was associated with an increased risk of identified in the second phase of the review detected an
AD (Caamano-Isorna et al., 2006). association between educational attainment and risk of AD.
The systematic review reported by Patterson et al. (2007) However, findings from three moderate-quality systematic
identified a single cohort study which examined the effect of reviews, collating evidence from 16 observational studies,
educational attainment on the risk of developing AD. The study suggested that lower levels of education appeared to increase
detected a decreased risk of AD associated with lower educational risk of AD. Consequently, a majority of the available studies
attainment (<12 years vs. >15 years) (RR = 0.48; 95% CI = 0.27 to supported the hypothesis that higher educational attainment is
0.84). associated with a reduced risk of AD. Whether educational
One systematic review (Caamano-Isorna et al., 2006) and two attainment is a true risk factor for AD or a factor involved in the
cohort studies were included in the HTA reported by Williams et al. alleviation or postponement of clinical symptoms remains an issue
(2010). According to the findings of the systematic review by of continued debate (Caamano-Isorna et al., 2006; Stern, 2006).
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 171

3.1.51. Cognitive leisure activities p = 0.72) subjects were not at a significantly increased risk of AD.
We identified three moderate- (Weih et al., 2007; Williams The size of social networks was not associated with AD risk (4–7
et al., 2010; Stern and Munn, 2010) and two low- (Fratiglioni et al., persons vs. 0–3 persons, RR = 1.24; 95% CI = 0.80, 1.70; p = 0.22.  8
2004; Barnes and Yaffe, 2011) quality systematic reviews that persons vs. 0–3 persons, RR = 1.22; 95% CI = 0.80, 1.70; p = 0.25). The
examined the association between cognitively stimulating leisure ratio of family members to friends was also not significantly
activities and subsequent risk of AD. No primary studies were associated with risk of AD (More family members than friends,
identified. RR = 0.80; 95% CI = 0.60, 1.00; p = 0.12. More friends than family
members, RR = 0.92; 95% CI = 0.60, 1.30; p = 0.68). There was no
3.1.51.1. Systematic reviews. Weih et al. (2007) systematically association between AD and satisfaction with social networks
reviewed the association between cognitive stimulation, leisure (RR = 0.84; 95% CI = 0.30, 1.30; p = 0.21) and feeling misunderstood
activities, and social contacts with risk of developing AD. All six (RR = 0.66; 95% CI = 0.30, 1.40; p = 0.24). Subjects reporting to
identified studies reported risk estimates below 1.00. Based on the receive more than they give were at a marginally significant
available evidence, the reviewers concluded that maintaining reduced risk of AD (RR = 0.47; 95% CI = 0.20, 1.00; p = 0.05). Giving
social contacts and regular engagement in cognitively stimulating more than receiving yielded a risk estimate above 1.00, however,
and other leisurely activities appeared to protect against the results failed to reach statistical significance (RR = 1.16; 95%
development of AD. CI = 0.90, 1.40; p = 0.22).
Stern and Munn (2010) examined the effect of early-, middle-,
and late-life participation in cognitive leisure activities on the risk 3.1.52.3. Discussion. Results summarized in two moderate-quality
of AD. A total of five case-control and one cohort study examined systematic reviews suggest that high social engagement and
the effect of early and middle life (20 to 60 years of age) maintaining social contacts were associated with a protective
participation. All six studies found a statistically significant or effect against the development of AD (Weih et al., 2007; Williams
marginally significant association between AD and at least one et al., 2010). There was evidence to suggest that marriage was
domain of cognitive leisure activity or with cognitive leisure associated with a reduced risk of AD (Williams et al., 2010). Results
activities considered as an all-encompassing category. Five cohort from the only primary study identified in the second phase of the
studies, three of which specifically reported on risk of developing review detected a reduced risk of AD among those who reported
AD, examined the effect of late-life (65 years of age) participation receiving more than they give (Amieva et al., 2010). However, the
in cognitive leisure activities. All three studies detected a study failed to detect an association between size and composition
protective effect attributed to frequent engagement in late-life. (ratio of family to friends, marriage) of social networks with risk of
Based on the available evidence, the reviewers concluded that AD. Considered cumulatively, there was some evidence to suggest
middle and late-life participation in cognitive leisure activities may that lower social engagement was associated with an increased
protect against the development of AD. risk of AD.
Three of four cohort studies identified by Williams et al. (2010)
specifically reported on the risk of developing AD with respect to 3.1.53. Constricted life space
engagement in cognitive leisure activities. All three studies We identified a single prospective study reporting on the
consistently reported a statistically significant reduced risk of association between life space and risk of AD (James et al., 2011).
AD associated with frequent participation. Subjects were recruited from the Rush Memory and Aging Project
and the Minority Aging Research Study. The study detected an
3.1.51.2. Discussion. According to evidence summarized in three increased risk of AD associated with constricted life space
moderate-quality systematic reviews, increased participation in (HR = 1.21; 95% CI = 1.08, 1.36). The risk estimate was slightly
cognitive leisure activities appears to be associated with a reduced attenuated, but remained statistically significant, when the
risk of AD. analysis was adjusted for additional variables, namely, physical
function, disability, depressive symptoms, social networks, vascu-
3.1.52. Social Networks/Engagement lar risk factors and diseases (HR = 1.15, 95% CI = 1.02, 1.31). Overall,
Two moderate- (Weih et al., 2007; Williams et al., 2010) and one results of the study suggest that a higher level of life-space
low- (Fratiglioni, Paillard-Borg, and Winblad 2004) quality constriction was associated with an increased risk of AD.
systematic reviews examined the association between social
engagement and risk of AD. Our search for primary studies 3.1.54. Lifestyle behaviour Patterns/Multiple lifestyle factors
yielded one relevant record (Amieva et al., 2010). We did not identify any systematic reviews reporting on the
effect of multiple, concurrent lifestyle risk factors. The search for
3.1.52.1. Systematic reviews. Weih et al. (2007) identified six primary studies yielded two relevant studies (Norton et al., 2012;
relevant studies. Based on the available evidence, the reviewers Gelber et al., 2012).
concluded that maintaining social contacts and regular
engagement in cognitively stimulating and other leisurely 3.1.54.1. Primary studies. Data from the Cache County Study was
activities were associated with a protective effect against the used to examine the combined effect of diet, exercise, church
development of AD. attendance, social interaction, alcohol consumption, and smoking
Studies identified by Williams et al. (2010) suggest that low status on risk of developing AD (Norton et al., 2012). Using these
social engagement, poor social networks, and loneliness were factors, four behavioural patterns or exposure categories were
associated with an increased risk of AD. Furthermore, never being developed: (a) unhealthy and not religious, (b) healthy and
married, being single and living alone, as well as widowhood and moderately religious, (c) healthy and very religious, and (d)
divorce in late-life were also associated with an increased risk. unhealthy and religious, with the latter category serving as the
reference. Compared to the unhealthy and religious group, the
3.1.52.2. Primary studies. Using data from the PAQUID cohort healthy and moderately religious group (HR = 0.54; 95% CI = 0.34,
study, Amieva et al. (2010) examined the effect of social networks 0.86) as well as the healthy and very religious group (HR = 0.56;
on risk of AD. Compared to married subjects, widowed (RR = 0.92; 95% CI = 0.36, 0.88) were at a reduced risk of AD. The risk estimate
95% CI = 0.70, 1.10; p = 0.53), single (RR = 1.36; 95% CI = 0.70, 2.30; for the unhealthy and not religious group did not reach statistical
p = 0.27) and divorced/separated (RR = 0.88; 95% CI = 0.40, 1.70; significance (HR = 0.59; 95% CI = 0.31, 1.12). The association was not
172 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

modified by APOE e4 status. Results from this longitudinal cohort (OR = 1.35; 95% CI = 0.94, 1.94). Head trauma was not associated
study suggest that certain lifestyle behavioural patterns, which with risk of AD among females (OR = 0.91; 95% CI = 0.56, 1.47). In
take several risk factors into consideration, were associated with a contrast, head trauma increased risk of AD in males (OR = 2.29; 95%
reduced risk of AD. CI = 1.47, 3.58). The increased risk of AD in males (OR = 2.26; 95%
The nested case-control study by Gelber et al. (2012) examined CI = 1.13, 4.53) but not females (OR = 0.92; 95% CI = 0.53, 1.59) was
the effect of lifestyle risk factors, namely, BMI, smoking status, diet, also observed when the analyses were restricted to post Mortimer
and physical activity on the development of AD among Japanese- et al. (1991) studies. Overall, when the results of all 15 studies were
American men enrolled in the Honolulu-Asia Aging Study. The considered cumulatively, the review suggested that prior head
combined effect of the aforementioned factors was also examined. injury with loss of consciousness was associated with an increased
Lifestyle behavioural patterns were derived according to the risk of AD. However, when the analysis was restricted to studies
number of low-risk factors. The following patterns were examined: published after the Mortimer et al. (1991) review, there was no
one low-risk factor (non-smoking), two low-risk factors (non- evidence of an association. The review was able to replicate
smoking and diet score in top 40%), three low-risk factors (non- findings reported by Mortimer et al. (1991), which suggest an
smoking, diet score in top 40%, BMI <25.0 kg/m2), and four low-risk association between head injury and risk of AD in males only.
factors (non-smoking, diet score in top 40%, BMI <25.0 kg/m2, high Three longitudinal cohort studies, identified by Patterson et al.
physical activity). According to the results of the study, the (2007), reported on the association between head injury and AD.
aforementioned patterns were not associated with risk of AD (risk Only one of the three studies detected an association between head
estimates not reported). Overall, the study failed to detect an injury and AD. More specifically, the study reported that moderate
association between putative lifestyle risk factors, considered in and severe head injury, experienced by U.S servicemen during
combination, and risk of AD. World War II, was associated with a greater than two-fold
(HR = 2.32; 95% CI = 1.04, 5.10) and greater than four-fold (HR =
3.1.54.2. Discussion. The identified studies were heterogeneous 4.51; 95% CI = 1.77, 11.47) increased risk of AD, respectively.
with respect to the risk factors considered in the analysis. The HTA reported by Williams et al. (2010) included one
Methodological heterogeneity and the small number of studies previously conducted systematic review by Fleminger et al. (2003)
preclude a definitive statement regarding the effect of multiple, and three additional cohort studies. The included systematic
concurrent lifestyle factors or behaviour patterns. review, which relied on findings from 15 case-control studies,
yielded results suggesting that head injury was associated with an
3.1.55. Head injury increased risk of AD in males but not females. Two recent
Four moderate quality systematic reviews, which examined the prospective cohort studies identified in the HTA failed to detect an
association between head injury and risk of AD, were identified association between self-reported traumatic brain injury and risk
and eligible for inclusion in the present review. of AD. The third cohort study, which was retrospective in nature
and relied on an all-male sample, detected an increased risk of AD
3.1.55.1. Systematic reviews. Seven case-control studies were associated with moderate (Risk estimate: 2.32; 95% CI = 1.04, 5.17)
eligible for inclusion in the systematic review reported by and severe (Risk estimate: 4.51; 95% CI = 1.77, 11.47) traumatic
Mortimer et al. (1991). The meta-analysis combining the results brain injury. Mild TBI, however, was not associated with risk of AD
of the seven included studies detected an increased risk of AD (Risk estimate: 0.76; 95% CI = 0.18, 3.29) according to the same
associated with head trauma involving a loss of consciousness study. The reviewers concluded that traumatic brain injury,
(RR = 1.82; 95% CI = 1.26, 2.67). There was no evidence of statistical particularly in males, appeared to be associated with an increased
heterogeneity. Several subgroup analyses were conducted. Cases risk of AD.
were stratified by family history of dementia, age of onset, and
gender. When cases were stratified by family history, the 3.1.55.2. Discussion. Based on the evidence presented in four
association between head injury and AD was statistically moderate quality systematic reviews, there appears to be an
significant for those with a sporadic (RR = 2.31; 95% CI = 1.17, association between head injury and risk of AD. Subgroup analysis
4.84) but not familial (RR = 1.42; 95% CI = 0.76, 2.71) case of AD. conducted in two reviews suggests that this association was true
However, there was no statistical difference between groups for males but not females. One recent cohort study, which relied on
(x2 = 1.17, p = 0.30). When stratified by age of onset, the association an all-male sample, corroborated this finding (Williams et al.,
between AD and head trauma was similar in both groups 2010).
( < 70 years: RR = 1.95; 95% CI = 1.12, 3.48. 70 years: RR = 1.81;
95% CI = 1.03, 3.25). Lastly, according to the subgroup analysis by 3.1.56. Anaemia, haemoglobin
gender, head trauma was associated with an increased risk of AD in We identified one moderate-quality systematic review (Peters
males (RR = 2.67; 95% CI = 1.64, 4.41) but not in females (RR = 0.85; et al., 2008a) and one cohort study (Shah et al., 2011) that
95% CI = 0.43, 1.70). Overall, this review suggested that head trauma examined the association between anaemia and haemoglobin with
with loss of consciousness was associated with an increased risk of risk of AD.
AD in males.
The systematic review reported by Fleminger et al. (2003) was 3.1.56.1. Systematic reviews. A total of three studies investigating
aimed at updating the earlier review by Mortimer et al. (1991). A the association between haemoglobin or anaemia and risk of
total of 15 case-control studies, seven of which were published dementia, cognitive decline, and AD were eligible for inclusion in
after the Mortimer et al. review, were identified. Results of the the review reported by Peters et al. (2008a). AD was the outcome of
fixed effects meta-analysis (n = 15) suggested an increased odds of interest in only one of the three included publications. The
AD associated with head trauma (OR = 1.58; 95% CI = 1.21, 2.06). publication reported on two studies, one of which was a
Studies published before the Mortimer et al. (1991) systematic retrospective case-control study while the other employed a
review were combined in a subgroup analysis. The pooled analysis longitudinal cohort design. In the retrospective case-control
detected a 90% increased odds of AD associated with head injury analysis, anaemia was associated with an increased odds of AD
(OR = 1.90; 95% CI = 1.29, 2.81). In contrast, head trauma was not (OR = 1.88; 95% CI = 1.17, 3.03). However, when stratified by sex,
associated with AD when the analysis was restricted to the case- anaemia was associated with a statistically significant increased
control studies published after the 1991 systematic review odds of AD in women (OR = 1.96; 95% CI = 1.11, 3.47) but not in men
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 173

(OR = 1.81; 95% CI = 0.75, 4.39). In the longitudinal cohort study by Williams et al. (2010) concluded that depression was associated
the same author, there was no association between anaemia and with an increased risk of AD.
risk of AD (SIR: 0.98; 95% CI = 0.67, 1.37). Sex stratified analysis
failed to detect an association in females (SIR: 0.79; 95% CI = 0.49, 3.1.57.2. Primary studies. Midlife symptoms of depression were
1.23) and males (SIR: 1.49; 95% CI = 0.79, 2.56). Overall, the not associated with risk of AD (RR = 1.06; 95% CI = 0.85, 1.33)
reviewers concluded that anaemia appeared to increase risk of according to the retrospective cohort study reported by Barnes
dementia and cognitive decline. However, when the analysis was et al. (2012). Alternatively, symptoms experienced in late-life alone
restricted to AD, the association was unclear. (RR = 2.06; 95% CI = 1.67, 2.55) and in both mid- and late-life
(RR = 1.99; 95% CI = 1.47, 2.69) were associated with an increased
3.1.56.2. Primary studies. The association between haemoglobin risk of AD. Findings from this study support the association
levels and risk of AD was prospectively assessed by Shah et al. between depression, particularly in late-life, and risk of AD.
(2011). Low haemoglobin or anaemia, defined as <12 g/dL for The prospective cohort study by Kohler et al. (2011) failed to
women and <13 g/dL men, was associated with an increased risk of detect an association between depressive symptoms and risk of AD
AD (HR = 1.60; 95% CI = 1.02, 2.52). Similarly, when compared to among subjects enrolled in the Maastricht Ageing Study. More
subjects with normal haemoglobin levels, those with high specifically, depression, treated as a continuous (OR = 1.04; 95%
haemoglobin (>15.5 g/dL for women, >17.5 g/dL for men) were at CI = 0.98, 1.09) and categorical (OR = 1.81; 95% CI = 0.78, 4.23)
an increased risk of AD (HR = 3.39; 95% CI = 1.25, 9.20). Overall, variable, did not significantly increase risk of AD. As noted by the
results of the study suggest that both low and high haemoglobin authors, failure to detect a significant association may be
levels were associated with a detrimental effect. attributed to the small number of AD cases.
The effect of depressive symptoms at baseline, past and current
3.1.56.3. Discussion. According to the identified systematic review, major depressive episodes, and lifetime treated depression on risk
the association between anaemia and AD was inconclusive. of AD was prospectively examined in the Three-City study (Lenoir
Findings from a newly published cohort study implicated both et al., 2011). There was no association between depressive
high and low haemoglobin levels as a risk factor for the symptoms at baseline and risk of AD (HR = 1.00; 95% CI, 0.70,
development of AD. The small number of available studies 1.60). Similarly, lifetime treated depression and major depressive
precludes a conclusive statement regarding the effect of episodes were not associated with AD. The study was limited by
anaemia and haemoglobin on risk of AD. the small sample size (Lenoir et al., 2011).
Using data obtained from the Adult Changes in Thought study,
3.1.57. Depression Li et al. (2011) examined the association between depressive
We identified three moderate-quality reviews that reported on symptoms, self-reported history of depression, and risk of AD, all-
the association between depression and risk of AD (Patterson et al., cause dementia, and other cognitive outcomes. Depressive
2007; Ownby et al., 2006; Williams et al., 2010). Two low-quality symptoms increased risk of AD (HR = 1.43; 95% CI = 1.05, 1.94),
reviews were excluded (Jorm, 2001; Barnes and Yaffe, 2011). Seven however, there was no association between self-reported history of
relevant primary studies were identified (Blasko et al., 2010; depression and risk of AD.
Dotson et al., 2010; Saczynski et al., 2010; Kohler et al., 2011; Lenoir Using data from the Baltimore Longitudinal Study of Aging,
et al., 2011; Li et al., 2011; Barnes et al., 2012). Dotson et al. (2010) examined the association between recurrent
elevated depressive symptoms (EDS) and risk of AD. Recurrence of
3.1.57.1. Systematic reviews. Twenty studies, representing 102 172 EDS was not associated with risk of AD (HR = 1.06; 95% CI = 0.90,
subjects, were included in the random effects meta-analysis 1.25). Compared to subjects without a history of EDS, those who
reported by Ownby et al. (2006). According to the pooled analysis, had one EDS were at a significantly increased risk of AD (HR = 1.83;
depressed subjects had a two-fold increased risk of AD (OR = 2.02; 95% CI = 1.10, 3.02). Having two or more EDS was associated with an
95% CI = 1.80, 2.26; test for heterogeneity: Egger test, t = 2.75; increased risk of AD, however, results failed to reach statistical
p = 0.01). Subgroup analyses restricted to case-control (n = 9) and significance (HR = 1.47; 95% CI = 0.73, 2.97).
cohort studies (n = 11) yielded similar results (Case-control  Higher scores on the Geriatric Depression Scale were not
OR = 2.03; 95% CI = 1.73, 2.38; test for heterogeneity: p = 0.10. associated with risk of AD according to the VITA study (OR = 1.20;
Cohort  OR = 1.90; 95% CI = 1.55, 2.33; test for heterogeneity: 95% CI = 1.00, 1.50) (Blasko et al., 2010).
p = 0.02). The pooled analysis of retrospective cohort studies The effect of depressive symptoms on risk of AD was examined
(OR = 2.11; 95% CI = 1.82, 2.45) yielded a stronger risk estimate by Saczynski et al. (2010) using data from the Framingham Heart
when compared to that of prospective studies (OR = 1.78; 95% Study. Depression was associated with an increased risk of AD
CI = 1.16, 2.73). Results of the review suggest that depression was (HR = 1.76; 95% CI = 1.03, 3.01; p = 0.039). Treated continuously,
associated with an increased risk of AD. This finding was confirmed each 10 point increase in Center for Epidemiologic Studies
by subgroup analysis restricted to studies which employed widely Depression Scale (CES-D) was associated with an increased risk
accepted criteria for diagnosis of both depression and AD (see of AD (HR = 1.39; 95% CI = 1.11, 1.75; p = 0.005). Depression
Table 62 in Supplementary material II). remained significantly associated with AD following further
Patterson et al. (2007) identified a single study which detected adjustments and in the subgroup analysis excluding subjects with
an association between depression and risk of AD in males possible mild cognitive impairment. Based on these findings, the
(RR = 4.20; 95% CI = 2.10, 8.80) but not females (RR = 1.00; 95% authors concluded that there was evidence to suggest that late-life
CI = 0.50, 1.90). depression was associated with an increased risk of AD.
Williams et al. (2010) relied on findings from the systematic
review reported by Ownby et al. (2006). Five additional studies, 3.1.57.3. Discussion. Evidence summarized in three moderate
published after Ownby et al. (2006), were also considered. Ownby quality systematic reviews was suggestive of an association
et al. (2006) reported an increased risk of AD associated with between history of depression and an increased risk of AD. All
history of depression. All five included primary studies consistent- but one observational study identified in the second phase of the
ly detected a statistically significant increased risk of AD among review reported risk estimates exceeding 1.00. Four of seven cohort
those with a history of depression. Based on these findings, studies detected a statistically significant detrimental effect
associated with depression. Further research is required to
174 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

determine whether depression is a risk factor for AD or an early 3.1.62. Hearing loss, central auditory dysfunction
symptom of the disease (Barnes et al., 2012; Li et al., 2011). No systematic reviews reporting on the association between
hearing loss or impairment were identified. However, our search
for observational studies yielded two relevant cohort studies
3.1.58. Mild Cognitive Impairment (MCI)
(Gates et al., 2011; Lin et al., 2011).
One moderate-quality systematic review examined the risk of
The association between central auditory dysfunction (CAD)
conversion from MCI to AD (Mitchell and Shiri-Feshki, 2009). Two
and risk of AD was examined using data obtained from the Adult
low-quality reviews were excluded from the qualitative synthesis
Changes in Thought (ACT) Study (Gates et al., 2011). CAD was
(Bruscoli and Lovestone, 2004; Mitchell and Shiri-Feshki, 2008).
ascertained according to three auditory tests (SSI-ICM, DDT, DSI)
No recent primary studies were identified.
with moderate and severe CAD defined as achieving scores less
The systematic review by Mitchell and Shiri-Feshki (2009)
than 50% and 80% correct on each test, respectively. Moderate CAD
examined the annual conversion rate from mild cognitive
was associated with an increased risk of AD according to results
impairment (MCI) to AD. A meta-analysis examining the risk of
from the DSI (HR = 6.80; 95% CI = 1.90, 24.10) and DDT (HR = 7.00;
AD among subjects with MCI, in comparison to healthy elderly
95% CI = 1.60, 31.00) tests. While the SSI-CCM produced a risk
controls, was also conducted. Among studies employing the Mayo
estimate above 1.00, the results failed to reach statistical
clinic definition for MCI diagnosis, the annual conversion rate
significance (HR = 2.50; 95% CI = 0.90, 7.50). Severe CAD was
(ACR) was 8.10% (95 CI: 6.30%, 13.40%) in “specialist settings” and
associated with a significant increased risk of AD according to
6.80% (95% CI = 1.90%, 14.50%) in “community settings”. Six studies
the DSI (HR = 9.90; 95% CI = 3.60, 26.70) but not the DDT (HR = 2.20;
examined the risk of AD among individuals diagnosed with mild
95% CI = 0.70, 7.00) or SSI-CCM (HR = 2.10; 95% CI = 0.70, 6.60).
cognitive impairment (MCI) in comparison to healthy elderly
Considered cumulatively, the authors concluded that CAD
controls. According to the results from the random effects meta-
appeared to be a risk factor for AD.
analysis, those with MCI had a near nine-fold increased risk of AD
The association between hearing loss and risk of AD and all-
in comparison to healthy elderly controls (RR = 8.93; 95% CI = 4.18,
cause dementia was prospectively examined by Lin et al. (2011).
19.07).
Treated as a continuous variable, each 10 dB of hearing loss was
associated with an increased risk of dementia, however, results did
3.1.59. Sporadic cerebral amyloid angiopathy not reach statistical significance for AD (HR = 1.20; 95% CI = 0.94,
One identified systematic review (Keage et al., 2009) reported 1.53). As noted by the authors, the analysis was limited by the small
on the effect of sporadic cerebral amyloid angiopathy. However, number of incident AD cases.
the review was of low methodological quality and was subse-
quently excluded. 3.1.63. Declining health and self-Rated health
We identified one cohort study (Song et al., 2011) that reported
on the effect of declining health and another (Montlahuc et al.,
3.1.60. Essential tremor
2011) that examined the association between self-rated health and
One systematic review reported on the association between
risk of AD.
essential tremor and risk of AD (LaRoia and Louis, 2011). Due to low
Using data from the Canadian Study of Health and Aging, Song
methodological quality, the review was excluded.
et al. (2011) examined the risk of AD associated with “health
deficits not known to predict dementia”. A frailty index (FI-NTRF),
3.1.61. Cancer
which considered 19 deficit indicators, was developed to measure
We identified no systematic reviews which reported on the
the exposure of interest. Results from the minimally adjusted
association between cancer and risk of AD. Two cohort studies
model suggest that the FI-NTRF was associated with an increased
were identified in the second phase of the review (Roe et al., 2010;
risk of AD (OR = 1.03; 95% CI = 1.01, 1.05). However, results
Driver et al., 2012).
approached the null when putative risk factors for AD were
According to the Cognition Sub-study of the Cardiovascular
adjusted for in the analysis (OR = 1.01; 95% CI = 1.00, 1.03). Based on
Health Study (CHS), history of cancer among whites was associated
the results of the study, the authors concluded that health deficits
with a statistically significant reduced risk of AD (HR = 0.57; 95%
or declining health may predict risk of AD.
CI = 0.36, 0.90) (Roe et al., 2010). In contrast, history of cancer was
The Three City prospective cohort study detected an increased
associated with an increased risk among minority subjects
risk of AD among subjects who rated their health as poor
(HR = 5.04; 95% CI = 1.99, 12.80). As noted by the author, the latter
(HR = 1.48; 95% CI = 1.00, 2.24) when compared to subjects
finding may be attributed to the small sample size of minority
reporting to be in good health (Montlahuc et al., 2011).
subjects (n = 29) as well as racial differences with respect to cancer
type (Roe et al., 2010).
3.1.64. Mild Parkinsonian Signs
Results reported by Driver et al. (2012) suggest that cancer was
We did not find any systematic reviews which reported on the
associated with a reduced risk of probable AD among subjects
association between mild Parkinsonian signs (MPS) and risk of AD.
enrolled in the Framingham Heart Study (HR = 0.67; 95% CI = 0.47,
One relevant primary study was identified (Louis et al., 2010).
0.97). When results were stratified by type of cancer, only subjects
Louis et al. (2010) prospectively examined the association
with a history of smoking related cancers were at a significantly
between MPS and risk of AD. According to the results of the study,
reduced risk of probable AD (Smoking related cancers, HR = 0.26;
MPSs were significantly associated with risk of AD (HR = 1.78; 95%
95% CI = 0.08, 0.82. Non-smoking related cancers, HR = 0.82; 95%
CI = 1.20, 2.65; p = 0.004).
CI = 0.57, 1.19). However, the association failed to reach statistical
significance when the analysis was adjusted for APOE e4 status,
3.1.65. Spine surgery under general anesthesia
education, and homocysteine level (All cancers, HR = 0.76; 95%
No systematic review was identified that reported on the
CI = 0.52, 1.12. Smoking related cancers, HR = 0.34; 95% CI = 0.11,
association between spine surgery under general anesthesia and
1.08. Non-smoking related cancers, HR = 0.91; 95% CI = 0.61, 1.35).
risk of AD. One relevant primary study was identified (Zuo and Zuo,
No association between cancer and possible AD was detected.
2010).
Based on the results of the study, the authors concluded that
Zuo and Zuo (2010) retrospectively examined the association
cancer appeared to reduce risk of AD.
between spine surgery under general anesthesia and risk of AD.
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 175

The study failed to detect an association between volatile (Breteler et al., 1991). These findings must be interpreted with
anesthesia and risk of AD (OR = 1.27; 95% CI = 0.52, 3.09; p- caution due to the small number of available studies and reliance
value = 0.603). There was also no association between AD and on case-control studies. Furthermore, the review relied on older
length of hospital stay (OR = 0.99; 95% CI = 0.93, 1.05; p-value = studies published in the late 1980s and early 1990s. Current
0.742) or length of operating room time (OR = 1.00; 95% CI = 0.99, observational studies are required prior to arriving at a definitive
1.01; p-value = 0.960). conclusion regarding the effect of these factors on the risk of AD.
The association between thyroid function and risk of AD was
3.1.66. Various medical factors unclear. One review found a marginally significant increased risk of
Three moderate quality systematic reviews examined the AD associated with hypothyroidism but failed to detect an
association between various medical factors and risk of AD association with thyroid disease (unspecified) and hyperthyroid-
(Breteler et al., 1991; Patterson et al., 2007; Williams et al., ism (Breteler et al., 1991). Results from a single study identified by
2010). Medical factors analyzed in these reviews include thyroid Patterson et al. (2007) detected a greater than three-fold increased
disorders, goitre, atopy, hay fever, rheumatoid arthritis, osteoar- risk of AD associated with low levels of thyroid stimulating
thritis, poliomyelitis, herpes zoster, herpes simplex, encephalitis/ hormone. However, conflicting evidence was reported in the
meningitis, epilepsy, severe headaches/migraines, blood trans- recent prospective study identified in the second phase of the
fusions, general anesthesia, and sleep apnea. review (Forti et al., 2012). More specifically, the study failed to
detect an association between levels of thyroid stimulating
3.1.66.1. Systematic reviews. The systematic review and hormone, hypothyroidism and risk of AD.
“collaborative re-analysis” by Breteler et al. (1991) examined the
association between several exposures pertaining to medical 3.1.67. C-reactive protein
history and risk of AD. According to the re-analysis and pooled risk One moderate-quality systematic review reported on the
estimates of eight case-control studies, there was no association association between c-reactive protein and risk of AD (Kuo
between AD and a majority of the examined medical conditions et al., 2005b).
and procedures. More specifically, thyroid disease (unspecified), The systematic review conducted by Kuo et al. (2005b)
hyperthyroidism, goitre, atopy, hay fever, rheumatoid arthritis, identified two nested case-control studies. One study detected
poliomyelitis, herpes zoster, herpes simplex, encephalitis/ an increased risk of AD among men with higher concentrations of
meningitis, epilepsy, and general anesthesia were not associated CRP. However, the risk estimate derived for the greatest quartile of
with AD. However, the reviewers suggest that failure to detect a concentration was only marginally significant (Second quartile,
significant association may be attributed to inadequate statistical OR = 2.80; 95% CI = 1.30, 6.30. Third quartile, OR = 5.40; 95%
power. While hypothyroidism was associated with a marginally CI = 2.50, 11.70. Fourth quartile, OR = 2.30; 95% CI = 1.00, 5.40).
significant increased risk of AD, osteoarthritis and severe Increasing CRP concentration was not associated with risk of AD
headaches and migraines were associated with a marginally according to the second study (RR = 1.10; 95% CI = 0.96, 1.26).
significant decreased risk of AD. Finally, blood transfusions were Overall, results of the review suggest that CRP concentrations were
associated with a statistically significant 40% decreased risk of AD associated with risk of cognitive decline and dementia. However,
(95% CI = 0.40, 0.90). As noted by the reviewers, however, this when the analysis was restricted to AD, the association was less
finding was likely attributed to selection bias. clear. Although both studies reported risk estimates greater than
Patterson et al. (2007) identified a single cohort study which 1.00, results from only one reached statistical significance.
examined the association between AD and serum thyroid
stimulating hormone. According to the study, a low level of 3.1.68. Plasma clusterin levels
thyroid stimulating hormone was associated with an increased risk No systematic review examined the effect of plasma clusterin
of AD (RR = 3.50; 95% CI = 1.10, 11.50). levels and risk of AD. One relevant primary study was identified.
The HTA (Williams et al., 2010) examined the association The case-cohort study reported by Schrijvers et al. (2011)
between sleep apnea and subsequent risk of AD. However, there examined the association between plasma clusterin levels, treated
were no studies eligible for inclusion. continuously and categorically, and risk of prevalent and incident
AD. Treated as a continuous variable, each 1 SD increase in plasma
3.1.66.2. Primary studies. Our search of the literature identified clusterin levels was associated with prevalent (OR = 1.63; 95%
one prospective cohort study (Forti et al., 2012) that reported on CI = 1.21, 2.20) but not incident (HR = 1.00; 95% CI = 0.85, 1.17) AD.
the association between serum thyroid stimulating hormone When treated as a categorical variable (47.2- 99.5 mg/mL as
(TSH), hypothyroidism and risk of AD. According to the fully referent), only the highest quartile (132.5  198.0 mg/mL) of
adjusted model, hypothyroidism (OR = 1.13; 95% CI = 0.53, 2.38) plasma clusterin was associated with prevalent AD (OR = 2.99; 95%
was not associated with risk of AD. Compared to the lowest tertile CI = 1.25, 7.16). The test for trend was statistically significant,
of serum thyroid stimulating hormone (<2.20 mIU/L), both middle however (p = 0.004). Alternatively, there was no association
(2.20-3.59 mIU/L) and high (>3.59 mIU/L) tertiles of TSH were not between increasing quartiles of plasma clusterin and risk of
associated with risk of AD (Middle, OR = 1.14; 95% CI = 0.50, 2.57. incident AD (see Table 79 in Supplementary material II).
Highest, OR = 1.30; 95% CI = 0.61, 2.77). Treated continuously, each 1
SD increase in log transformed TSH was also not associated with 3.1.69. White matter hyperintensities
risk of AD (OR = 1.13; 95% CI = 0.86, 1.52). One moderate-quality systematic review reported on the
association between white matter hyperintensities (WMH) and
3.1.66.3. Discussion. Evidence summarized in one moderate risk of AD (Debette and Markus, 2010). One primary study, which
quality systematic review suggested that: (a) goitre, atopy, hay reported on the association between white matter changes and risk
fever, rheumatoid arthritis, poliomyelitis, herpes zoster, herpes of AD, was also identified (Verdelho et al., 2010).
simplex, encephalitis/meningitis, epilepsy, and general anesthesia
were not significantly associated with AD, (b) osteoarthritis and 3.1.69.1. Systematic reviews. A total of six studies examining the
severe headaches and migraines were associated with a marginally association between white matter hyperintensities, deep white
significant decreased risk of AD, and (c) blood transfusions were matter hypertensities, and/or periventricular hyperintensities
associated with a statistically significant decreased risk of AD with AD risk were eligible for inclusion in the review reported
176 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

by Debette and Markus (2010). All of the included studies were contrast, both the Rush Memory and Aging Project (Shah et al.,
prospective population-based studies or prospective studies 2011) (HR = 1.49; 95% CI = 0.95, 2.35) and the Canadian Study
conducted in high-risk populations. One population-based study of Health and Aging (Song et al., 2011) (OR = 1.13; 95% CI = 0.85,
detected an association between white matter hyperintensities 1.49) failed to detect an association between male sex and risk of
and risk of AD (WMH >3, HR = 1.50; 95% CI = 1.17, 1.99). The AD.
remaining three studies, all of which examined subjects with MCI,
failed to detect an association. Results from three studies were 3.1.71.3. Discussion. A pooled analysis (n = 7) reported in a
eligible for inclusion in a meta-analysis. The pooled analysis moderate-quality systematic review detected a statistically
detected an increased risk of AD associated with WMH (HR = 1.50; significant increased risk of AD among females. However, all but
95% CI = 1.20, 1.80; p for heterogeneity = 0.74; I2 = 0%). All four one of the six recent primary studies failed to detect an association
studies, which reported on the effect of deep white matter between sex and AD.
hyperintensities (DWMH), failed to detect an association. Two of
three studies, one of which examined subjects with amnestic MCI, 3.1.72. Sex hormones and sex hormone binding globulin
detected an association between periventricular hyperintensities One moderate-quality systematic review reported on the
(PVH) and risk of AD. Overall, results of this review suggest that association between sex hormones and risk of AD (Patterson
WMH were associated with an increased risk of incident AD. et al., 2007). We also identified two cohort studies which
However, this association was not observed in high risk reported on the association between AD and sex hormones or
populations, namely, those with MCI. PVH appeared to increase sex hormone binding globulin (SHBG) (Muller et al., 2010; Chu
risk of AD in high-risk and general populations, while DWMH were et al., 2010).
not associated with risk of AD.
3.1.72.1. Systematic reviews. The systematic review by Patterson
3.1.69.2. Primary studies. According to results of the LADIS
et al. (2007) examined the association between sex hormones and
prospective cohort study (Verdelho et al., 2010), white matter
risk of AD. One of two included studies failed to detect an
change severity was not significantly associated with risk of AD
association between high estrogen levels and risk of AD in females
(Moderate, HR = 0.29; 95% CI = 0.06, 1.41; p = 0.126. Severe,
(RR = 1.30; 95% CI = 0.88, 1.99). The second study reported that
HR = 1.29; 95% CI = 0.46, 3.65; p = 0.632). Medial temporal
higher free testosterone index in males was protective against the
atrophy (MTA), however, was significantly associated with AD
development of AD (RR = 0.74; 95% CI = 0.57 to 0.96).
(p = 0.046). A MTA score of 4, indicating most severe atrophy, was
significantly associated with risk of AD (HR = 15.62; 95% CI = 1.95,
3.1.72.2. Primary studies. A prospective cohort study identified in
124.77; p = 0.010). Lower scores were not significantly associated
the second phase of the review examined the association between
with AD (see Table 87 in Supplementary material II).
testosterone and risk of AD in an all-male sample of Chinese
subjects (Chu et al., 2010). The study detected an association
3.1.70. Age
between late-life baseline serum bioavailable testosterone (BT)
One moderate-quality systematic review reported that age was
level and risk of AD. More specifically, each unit (one nmol/L)
a “significant [predictor] of incidence of AD (F42 = 15.6,
increase of serum BT level was associated with a reduced risk of AD
p = 0.0003 . . . )” (Gao et al., 1998). Seven primary studies reporting
(RR = 0.22; 95% CI = 0.07, 0.69). Serum total testosterone (TT) and
on the effect of age were also identified (Song et al., 2011; Verdelho
SHBG levels were not predictors of AD.
et al., 2010; Li et al., 2010a,b; Reitz et al., 2010b; Yang et al., 2011;
Results reported by Muller et al. (2010) detected an increased
Shah et al., 2011; Zuo and Zuo, 2010). Six studies detected an
risk of AD associated with SHBG (HR = 1.30; 95% CI = 1.10, 1.60).
increased risk of AD associated with age. The remaining study
Higher levels of SHBG increased risk in men (HR = 1.30; 95%
failed to detect an association (Verdelho et al., 2010).
CI = 1.00, 2.00) and women (HR = 1.40; 95% CI = 1.10, 1.90).

3.1.71. Sex
3.1.73. Ethnicity, race
One moderate-quality systematic review examined the associ-
One systematic review, which reported on the effect of
ation between sex and AD (Gao et al., 1998). We also identified six
ethnicity/race, was identified (Shadlen et al., 2000). However,
relevant primary studies.
the study was deemed to be of low methodological quality and was
excluded from the review. Two relevant cohort studies were
3.1.71.1. Systematic reviews. The systematic review and meta-
identified in the second phase of the review (Reitz et al., 2010b;
analysis by Gao et al. (1998) examined the association between
Yang et al., 2011).
age, sex, and risk of AD. According to the pooled analysis of the
Compared to white subjects, black (HR = 1.94; 95% CI = 0.84,
included studies, females had a 56% increased risk of AD (OR = 1.56;
4.52) and Hispanic (HR = 1.69; 95% CI = 0.67, 4.22) subjects were not
95% CI = 1.16, 2.10).
at a significantly increased risk of developing possible and
probable LOAD according to Reitz et al. (2010b). Similar results
3.1.71.2. Primary studies. Zuo and Zuo (2010) retrospectively
were derived when the analysis was restricted to probable LOAD
examined the association between spine surgery under general
(Black, HR = 1.82; 95% CI = 0.69, 4.82. Hispanic, HR = 2.45; 95%
anesthesia and risk of AD. According to the results of the study, sex
CI = 0.90, 6.72).
was not significantly associated with risk of AD post-surgery
The longitudinal cohort study conducted by Yang et al. (2011)
(OR = 1.17; 95% CI = 0.51, 2.69; p-value = 0.702). Similarly, Yang et al.
examined the association between blood pressure, hypertension,
(2011) failed to detect an association between sex and risk of AD.
and risk of AD. The association between blood pressure measures
Female sex was not significantly associated with risk of possible
were analyzed in separate models. The association between race,
and probable LOAD according to results reported by Reitz et al.
treated as a dichotomous variable (Caucasian vs. African-Ameri-
(2010b) (HR = 1.15; 95% CI = 0.61, 2.21). Similar results were derived
can), and risk of AD was examined in each model. According to the
when the analysis was restricted to probable LOAD (HR = 1.14; 95%
risk estimates derived from each model, Caucasians had a reduced
CI = 0.51, 2.54).
risk of AD when compared to African-Americans (see Table 85 in
The VITA study detected an association between male sex and
Supplementary material II).
risk of AD (OR = 3.30; 95% CI = 1.30, 8.40) (Blasko et al., 2010). In
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 177

3.1.74. Maternal age increased risk of AD according to results reported by Leinonen et al.
The systematic review and re-analysis of case-control studies (2010).
reported by Rocca et al. (1991) examined the association between According to the results reported by Sundelöf et al. (2012) there
maternal age and risk of AD. The pooled analysis for early maternal was no association between cystatin C levels in cerebrospinal fluid
age yielded a risk estimate of 1.50, however, it failed to reach (CSF) and risk of AD (OR = 0.83; 95% CI = 0.57, 1.19; p = 0.32). Lower
statistical significance (RR = 1.50; 95% CI = 0.80, 3.00). The pooled Ab42 (OR = 0.09; 95% CI = 0.04, 0.21; p < 0.001) and higher total
analysis detected a marginally significant 70% increased risk of AD (OR = 6.50; 95% CI = 3.31, 12.7; p < 0.001) and phosphorylated
associated with a maternal age of 40 years and above (RR = 1.70; (OR = 4.06; 95% CI = 2.23, 7.39; p < 0.001) t in CSF were significantly
95% CI = 1.00, 2.90). Maternal age between 20 and 24 years associated with AD.
(RR = 0.90; 95% CI = 0.60, 1.40), 30 to 34 years (RR = 0.90; 95% Using data from the VITA study, Blasko et al. (2010) detected
CI = 0.60, 1.30), and 35 to 39 years (RR = 1.00; 95% CI = 0.60, 1.60) that higher baseline levels of plasma Ab42 was associated with
were not associated with AD. Overall, results of the review suggest risk of AD among subjects who were cognitively healthy at baseline
that late maternal age may increase risk of AD. The pooled analysis (OR = 2.00; 95% CI = 1.20, 3.30).
yielded a risk estimate above 1.00 for early maternal age, however, The association between beta-amyloid, midlife blood pressure,
results did not reach statistical significance. and risk of AD was prospectively examined by Shah et al. (2012)
using data from an all-male sample of Japanese-American men.
3.1.75. Parental longevity When treated as a continuous variable, decreasing (per 1 SD)
We did not identify any systematic reviews that reported on the plasma Ab1-40 and Ab1-42 levels were associated with an
association between parental longevity and risk of AD. One cohort increased risk of “all” and “pure” AD (see Table 80 in Supplemen-
study was identified (Lipton et al., 2010). When compared to the tary material II). Treated categorically, the lowest quartiles of Ab1-
offspring of parents with usual survival, having a parent with 40 (All AD, HR = 4.30; 95% CI = 1.90, 9.80. Pure AD, HR = 4.00; 95%
exceptional longevity (85 years) was associated with a protective CI = 1.50, 10.70) and Ab1-42 (All AD, HR = 3.10; 95% CI = 1.20, 8.00.
effect against AD according to results of the fully adjusted model Pure AD, HR = 4.30; 95% CI = 1.20, 15.90) were associated with an
(HR = 0.58; 95% CI = 0.35, 0.96). increased risk of both pure and all AD. Additional analyses detected
that the presence of high diastolic blood pressure strengthened the
3.1.76. Bonemineral density association between lower Ab1-40, Ab1-42 and risk of AD.
No systematic reviews examined the association between bone Considered cumulatively, the study suggested that lower plasma
mineral density (BMD) and risk of AD. One relevant cohort study Ab was associated with an increased risk of AD.
was identified. The prospective cohort study reported by Zhou
et al. (2011) detected an increased risk of AD associated with lower
3.1.78.2. Discussion. Cortical amyloid beta (Leinonen et al., 2010)
quartiles of BMD among Chinese subjects (Quartile 1, HR = 2.68;
and lower Ab42 in CSF (Sundelöf et al., 2012) were associated with
95% CI = 1.53, 4.71. Quartile 2, HR = 2.11; 95% CI = 1.17, 3.80. Quartile
AD according to two studies. Higher plasma levels of Ab42
3, HR = 1.67; 95% CI = 0.90, 3.12). Using the lowest quartile as a
predicted AD risk in the VITA study (Blasko et al., 2010). The second
reference, the study detected that higher rates of bone loss were
study detected a detrimental effect associated with lower plasma
also detrimental. While adiponectin was not associated with AD
Ab42 and Ab40 (Shah et al., 2012). With respect to t, one study
risk (HR = 0.98; 0.96, 1.01), the study detected a reduced risk of AD
(Leinonen et al., 2010) detected an association between presence of
associated with higher levels of plasma leptin (HR = 0.93; 95%
hyperphosphorylated t and amyloid beta in right frontal cortical
CI = 0.91, 0.95).
biopsy samples and risk of AD, while another (Sundelöf et al., 2012)
reported an association between AD and higher total and
3.1.77. Retinal vascular calibers
phosphorylated t in CSF. The only study to report on the effect
No systematic reviews reported on the association between
of cystatin C failed to detect an association with risk of AD
retinal vascular calibers and risk of AD. However, one prospective
(Sundelöf et al., 2012).
cohort study was identified in the second phase of the review (de
Jong et al., 2011). According to the fully adjusted model, each SD
3.1.79. Family History of Dementia, Down’s Syndrome and Parkinson’s
decrease in arteriolar caliber was not associated with risk of AD
disease
without cardiovascular disease (CVD) (HR = 1.02; 95% CI = 0.91,
One moderate-quality systematic review examined the effect of
1.14). Although the risk estimate was greater in magnitude, results
family history of dementia, Down’s syndrome, and Parkinson’s
also failed to reach statistical significance for AD with CVD
disease on the risk of AD (van Duijn et al., 1991).
(HR = 1.27; 95% CI = 0.89, 1.82). The study also failed to detect an
association between venular caliber (per 1 SD increase) and risk of
3.1.79.1. Systematic reviews. A total of 7 case-control studies
AD with (HR = 1.16; 95% CI = 0.82, 1.64) and without (HR = 1.06; 95%
identified by van Duijn et al. (1991) examined the association
CI = 0.95, 1.19) CVD. According to the results of the study, there was
between family history of dementia and AD. The meta-analysis
no significant association between AD and smaller arteriolar
combining the results of all seven studies detected a greater than
calibers or larger venular calibers.
three-fold increased risk of AD among those with a family history
of AD (RR = 3.50; 95% CI = 2.60, 4.60). The association remained
3.1.78. Beta-amyloid, t , cystatin C
statistically significant when data from an outlying study was
We did not identify any systematic reviews that examined the
excluded (RR = 3.60; 95% CI = 2.70, 4.90). Sensitivity analyses by
effect of beta-amyloid, t, or cystatin C and risk of AD. Four
gender detected an increased risk in both males (RR = 3.90; 95%
observational studies were identified in the second phase of the
CI = 2.50, 6.50) and females (RR = 3.30; 95% CI = 2.30, 4.60) with a
review (Blasko et al., 2010; Leinonen et al., 2010; Sundelöf et al.,
first degree relative with dementia. Family history was associated
2012; Shah et al., 2012).
with risk of AD irrespective of age of onset.
Five of the included studies examined the association between
3.1.78.1. Primary studies. Presence of amyloid beta (Ab) alone
family history of Down's syndrome and AD. Risk estimates could
(OR = 10.8; 95% CI = 4.90, 23.8; p < 0.001) and with
only be derived for two studies. The pooled analysis yielded a
hyperphosphorylated t (OR = 68.2; 95% CI = 22.1, 210; p < 0.001)
statistically significant risk estimate (RR = 2.70; 95% CI = 1.20, 5.70).
in right frontal cortical biopsy samples was associated with
178 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

The pooled analysis of two studies detected a marginally 3.2. Risk factors associated with AD progression
significant increased risk of AD among those with a family history
of Parkinson’s disease (RR = 2.40; 95% CI = 1.00, 5.80). The pooled Twelve moderate or high quality systematic reviews satisfied
analysis stratified by sex failed to detect an association in both the inclusion criteria of the present review. Reviews included
males and females (Males  RR = 4.40; 95% CI = 0.90, 20.90. Females examined the effect of education (Paradise et al., 2009; Meng and
 RR = 1.60; 95% CI = 0.50, 4.90). D'Arcy, 2012), APoE4 (Allan and Ebmeier, 2011), folic acid (Dangour
et al., 2010; Malouf et al., 2003; Malouf and Grimley, 2008),vitamin
3.1.79.2. Discussion. The available evidence suggested that a B-12 (Malouf and Areosa, 2003), fatty acids (Issa et al., 2006;
positive history of dementia and Down’s syndrome was Dangour et al., 2010), vitamin E (Farina et al., 2012), lecithin
associated with an increased risk of AD. Parkinson’s disease was (Higgins and Flicker, 2000), physical activity (Heyn et al., 2004;
associated with only a marginally significant increased risk of AD. Littbrand et al., 2011), and cognitive stimulation (Woods et al.,
These findings must be interpreted with caution due to the small 2012) on the progression of AD. Methodological characteristics and
number of available studies and reliance on case-control studies. results of the included systematic reviews are summarized (see
Furthermore, the review relies on older studies published in the Table 122 in Supplementary material II).
late 1980s and early 1990s. Current observational studies are
required prior to arriving at a definitive conclusion regarding the 3.2.1. Education
effect of these factors on the risk of AD. The systematic review by Paradise et al. (2009) examined the
effect of educational attainment on survival in AD. A total of 22
3.1.80. IgM antibodies against PC cohort studies were identified. Using the Oxford Centre for
One primary study was identified that reported on the effect of Evidence Based Medicine quality appraisal criteria, 11 of the
IgM antibodies against phosphorylcholine (anti-PC). The associa- cohort studies were deemed to be of good methodological quality.
tion between low levels of IgM antibodies against PC (anti-PC) Of the good quality reviews, only one study detected an association
and both incident and prevalent AD was examined by Eriksson between educational attainment and survival. More specifically,
et al. (2010b) using a nested case-control and case-control design, the study detected shorter survival in subjects with higher
respectively. The lowest 25th and 10th percentile of serum anti- educational attainment (HR = 1.10; p = 0.01). Of the lower quality
PC were not associated with incident AD. When treated as tertiles, studies, only one study detected an association. This study
lower tertiles of serum anti-PC were not associated with incident detected shorter survival in subjects with higher educational
AD. With respect to prevalent AD, those in the lowest 25th attainment (HR = 1.06; 95% CI = 1.01, 1.11). Collectively, results of
( > 31.6 U/ml vs 31.6 U/ml, OR = 2.70; 95% CI = 1.45, 4.99) and 10th the systematic review do not support an association between
( > 19.6 U/ml vs. 19.6 U/ml, OR = 3.98; 95% CI = 1.81, 8.74) educational attainment and survival in AD (Paradise et al., 2009).
percentile were at a significantly increased risk. Compared to Only two of 22 cohort studies detected an association between
the highest tertile, those in the lowest tertile of serum anti-PC higher educational attainment and shorter survival in subjects
were at a significantly increased risk of prevalent AD (OR = 2.36; with AD (Paradise et al., 2009).
95% CI = 1.14, 4.90). Results of the study suggest low levels of anti- The systematic review by Meng and D'Arcy (2012) examined
PC were associated with prevalent but not incident AD. the effect of education on the onset and progression of AD. The
reviewers identified primary studies which used cognitive decline
3.1.81. Gait speed (n = 20) and mortality (n = 8) as indicators of clinical progression.
A single cohort study reported on the association between With respect to cognitive decline, all but six studies detected an
gait speed and risk of dementia or AD. According to the results of association between higher educational attainment and faster
the EPIDOS-Toulouse cohort study, slow gait speed (OR = 3.38; 95% cognitive decline. The reviewers concluded that higher educational
CI = 1.80, 6.33) was associated with a statistically significant attainment was associated with faster cognitive decline among
increased risk of AD (Abellan van Kan et al., 2012). No subjects with dementia/AD. As noted by the reviewers, this finding
association was detected between AD and total lean mass was in support of the cognitive reserve hypothesis which posits
(OR = 0.57; 95% CI = 0.24, 1.34) or total fat mass (OR = 2.12; 95% that higher educational attainment delays the clinical detection of
CI = 0.90, 4.99). the disease and is correlated with worse brain damage upon
diagnosis and faster cognitive decline. Of the eight studies that
3.1.82. Propositional density reported on the effect of education on mortality, all but three failed
A single primary study was identified that reported on the to detect an association. Two studies detected reduced survival
association between propositional density and risk of AD. among subjects with higher educational attainment and one study
Propositional density, defined as a measure of “complexity of detected reduced survival among AD/dementia subjects with
written and spoken language”, was associated with risk of AD lower educational attainment. Overall, results of the review
according to the nested case-control study reported by suggest that higher educational attainment was associated with
Engelman et al. (2010). Subjects with higher scores of proportional faster cognitive decline but not with mortality (Meng and D'Arcy,
density, measured in early life, were at a reduced risk of AD 2012).
according to the matched analysis for age and sex (OR = 0.16; 95%
CI = 0.03, 0.90; p = 0.02). The association was slightly attenuated 3.2.2. APoE4
after additional adjustments for age (OR = 0.22; 95% CI = 0.03, 1.36; The systematic review and meta-analysis conducted by Allan
p = 0.05). and Ebmeier (2011) examined the effect of APoE4 status on
progression of AD. Progression was defined according to cognitive
3.1.83. Purpose in life decline and mortality. In the random effects meta-analysis
We identified a single primary study which reported on the including 16 studies and a total of 1618 subjects, APOE e4 status
effect of purpose in life. According to the Rush Memory and Aging was not associated with cognitive decline (effect size: 0.025; 95%
Project (Boyle et al., 2010), greater purpose in life was significantly CI = 0.09, 0.14; Q = 16.4; p = 0.36; I2 = 8.41). There was also no
associated with a reduced risk of AD according to the minimally association between APoE4 and mortality according to the random
(HR = 0.48; 95% CI = 0.33, 0.69; p < 0.001) and fully (HR = 0.60; 95% effects meta-analysis including four studies (effect size: 0.74; 95%
CI = 0.39, 0.92; p = 0.02) adjusted models. CI = 0.36, 1.53; Q = 8.83; p = 0.03; I2 = 66.0%). Results of the review
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 179

suggest that APoE4 status does not predict clinical progression 3.2.4. Vitamin B12
of AD. More specifically, there was no difference in cognitive The Cochrane systematic review reported by Malouf and Areosa
decline or mortality when carriers and non-carriers were Sastre (2003) examined the effect of vitamin B-12 supplementa-
compared. tion on cognition in subjects with and without dementia. The
review included three randomized controlled trials, only one of
3.2.3. Folic acid which specifically enrolled subjects with AD. The remaining
The Cochrane Systematic Review reported by Malouf et al. studies examined subjects with cognitive impairment and
(2003) examined the effect of folic acid with or without vitamin B- unspecified dementia. According to the one RCT in AD subjects,
12 on the progression of dementia or onset of cognitive there was no difference between the intervention group (n = 6
impairment. The review included four randomized controlled receiving 10 mg over 5 months) and the placebo group (n = 5) with
trials, two of which reported on progression of dementia/AD. One respect to cognitive function measured by the cognitive domain of
study reported that participants randomized to receive 10 mg of the Alzheimer's Disease Scale (mean difference (MD) = 0.04; 95%
folic acid per day (duration unknown) did not significantly differ CI = 5.95, 6.03). The second RCT also reported no difference in
from the placebo group with respect to cognitive tasks. The second cognition measured by the Mini Mental State Examination (MMSE)
study also failed to detect a significant difference between groups between experimental (10 mg/day or 50 mg/day of cyanocobalamin
(2 mg folic acid plus 1 mg vitamin B12/day vs. placebo for 12 for one month) and placebo among dementia subjects (50mcg vs.
weeks) with respect to cognitive decline, measured according to placebo, MD = 0.60; 95% CI = 3.06 to 1.86, p = 0.63. 10mcg vs.
Mini Mental State Examination (MMSE) (weighted mean differ- placebo, MD = 1.60 95% CI = 3.99 to 0.79, p = 0.19). The final
ence (WMD): 0.39; 95% CI = 0.43, 1.21; p = 0.35) and the cognitive study failed to detect a difference in cognitive function between
domain of the Alzheimer's Disease Scale (WMD: 0.41; 95% intervention (1 mg/week for 4 weeks) and placebo. More
CI = 1.25, 2.07; p = 0.63). There was also no difference between specifically, groups did not differ with respect to MMSE (MD =
groups with respect to activities of daily living, measured by the 0.10; 95% CI: 0.60, 0.80, P = 0.70), the Cambridge Cognitive
Bristol Activity of Daily Living instrument (WMD: 0.57; 95% Examination (MD = 0.60; 95% CI = 2.20 to 0.90; P = 0.43), and the
CI = 1.95, 0.81; p = 0.42). 12-words learning test immediate recall (MD = 0.20; 95% CI:
The Cochrane review (Malouf et al., 2003) was updated in 2008 0.70, 0.30, P = 0.42). Overall, the review suggested that vitamin B-
(Malouf and Grimley, 2008). The reviewers identified two 12 did not improve cognitive function among subjects with AD,
additional RCT studies examining the effect of folic acid on dementia, or cognitive impairment (Malouf and Areosa, 2003). As
progression of dementia/AD. One study failed to detect a difference noted by the reviewers, the identified studies were limited by the
between groups (10 mg folic acid vs. placebo for 10 weeks) with very small sample size of subjects.
respect to measures of memory, language, speed and concentra-
tion. The second study detected no significant difference between 3.2.5. Vitamin E
groups (1 mg folic acid/day vs. placebo for 24 weeks) with respect The Cochrane systematic review by Farina et al. (2012)
to cognitive performance, measured by MMSE (WMD: 0.13; 95% examined the effect of vitamin E supplementation on progression
CI = 1.96, 1.70, P = 0.89) or Digit Symbol Substitution Test (DSST) of MCI and AD. The review included two randomized controlled
(WMD: 0.26; 95% CI = 4.12, 4.64, p = 0.91). Similarly, there was no trials examining treatment effect in subjects with AD and one
difference between groups with respect to behaviour measured by study enrolling subjects with MCI. The RCT reporting on AD
the Social Behaviour Subscale (WMD: 1.38; 95% CI = 1.13, 3.89, examined the outcome of “survival time to any one of four end
p = 0.28). Patients assigned to the intervention group, however, points; death, institutionalisation, change in severity of dementia
performed better on the Instrumental Activities of Daily Living to a CDR of three or loss of two basic activities of daily living”
Scale (WMD: 2.67; 95% CI = 0.25, 5.09, p = 0.031). When data from (Farina et al., 2012). According to the adjusted analysis controlling
the aforementioned study was combined with one of the studies for baseline MMSE, time to the endpoints was delayed in AD
identified in the original review, there was no difference between subjects assigned to the intervention group (2000 IU/day)
groups with respect to MMSE (WMD: 0.30; 95% CI = 0.45, 1.05; compared to the placebo group (RR 0.47; P = 0.001). The second
Test for heterogeneity: x2 = 0.26, df = 1, p = 0.61, I2 = 0.0%). The study examined the effect of 800 IU/day for six months on
second study identified in the updated review failed to detect a cognitive function measured by the Clock drawing test, Blessed
difference between groups (10 mg folic acid vs. placebo for 10 Dementia Scale (BDS), and MMSE. Treatment with vitamin E had
weeks) with respect to measures of memory, language, speed and no effect on cognitive function measured by the Clock drawing test
concentration. Overall, folic acid with or without vitamin B-12 had and the BDS. However, treated subjects who did not respond to
no effect on cognitive decline/performance, or a number of other vitamin E supplementation, assessed by oxidative stress, experi-
outcomes, in subjects with dementia/AD (Malouf et al., 2003; enced a more pronounced decrease in cognitive function measured
Malouf and Grimley, 2008). by MMSE when compared to placebo. There was no difference
The systematic review by Dangour et al. (2010) examined the between placebo and treated subjects who responded. According
association between folic acid supplementation, with or without to the final study, compared to placebo, 2000 IU/day of vitamin E
vitamin B, on cognitive function among subjects with dementia did not prevent conversion of MCI to AD (HR = 1.02; 95%CI: 0.74,
and AD. The review included 10 randomized controlled trials, six 1.41; P = 0.91). Considered cumulatively, the reviewers concluded
of which examined the effect of folic acid combined with B- that vitamin E supplementation did not appear to be efficacious in
vitamins and four of which examined the isolated effect of folic the prevention of AD progression or progression of MCI to AD.
acid. There was methodological heterogeneity across studies with
respect to the tools used to assess cognitive function. Three of the 3.2.6. Fatty acids
four trials reporting on isolated folic acid supplementation The systematic review by Dangour et al. (2010) examined the
reported a beneficial effect. All six studies that examined folic effect of fatty acids on cognitive function among subjects with AD
acid supplementation combined with vitamin B failed to detect a or dementia. Four randomized controlled trials were captured in
beneficial effect. Overall, the reviewers concluded that the effect the review, two of which detected a beneficial effect with respect
of folic acid supplementation was inconclusive (Dangour et al., to cognitive function or quality of life. Based on the conflicting
2010). findings across studies, the reviewers concluded that the effect of
180 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

fatty acid supplementation on cognitive function in AD was functional, behaviour) were combined, a moderate effect size
unclear. was derived (ES = 0.62; 95% CI = 0.55, 0.70, P < 0.001). Results of the
The systematic review by Issa et al. (2006) examined the effect review suggest that physical activity was associated with a
of omega-3 fatty acids on the incidence and treatment of dementia. beneficial effect on several outcomes including cognition, behav-
The review included one randomized controlled trial examining iour, physical function and physical fitness (Heyn et al., 2004).
the effect of omega-3 fatty acid supplementation on cognitive The systematic review by Littbrand et al. (2011) examined the
function. Briefly, subjects assigned to receive 4.3 g/day of effect of physical activity on indicators of progression. Inclusion
docosahexaenoic acid (DHA) experienced improvements in was restricted to randomized controlled trials. A systematic search
cognitive function measured by the Hasegawa’s Dementia Rating of the literature yielded ten relevant studies, four of which were
Scale (HDS-R) andMini Mental State Examination (MMSE). deemed to be of moderate methodological quality according to the
However, all subjects enrolled in the study had vascular dementia, Physiotherapy Evidence Database (PEDro) instrument. The
therefore, the applicability of this trial was limited as our review remaining studies were of low quality. Studies were heterogeneous
was focused on AD. with respect to the interventions under study. One (low-quality) of
four studies detected a difference between intervention and
3.2.7. Lecithin control groups with respect to cognitive function. Of the three
The Cochrane systematic review reported by Higgins and moderate-quality studies reporting on the effect of physical
Flicker (2000) examined the effect of lecithin on the progression of function, two reported beneficial effects associated with exercise.
AD and other dementia subtypes. The review included ten relevant The only study which reported on the effect of exercise on activities
trials examining progression of AD. Studies were heterogeneous of daily living (ADL) reported a beneficial effect, namely, reduced
with respect to the outcome or indicator of progression examined. ADL decline. The reviewers concluded that there was no evidence
Two studies reported on global impression, one reported on of improved cognitive function or mobility associated with
functional performance, two reported on behaviour, one reported physical exercise among subjects with dementia. Alternatively,
on cognitive function, two reported on specific domains of there was preliminary evidence to suggest a beneficial effect of
cognitive function, and three reported on death. The random weight-bearing exercise with respect to walking performance and
effects meta-analysis combining results from two studies failed to ADLs (Littbrand et al., 2011).
detect a difference between groups with respect to global
impression (OR = 3.01; 95% CI = 0.92, 9.81; test for heterogeneity: 3.2.9. Cognitive stimulation
x2 = 0.94, p = 0.33, I2 = 0%). One RCT failed to detect a statistically The Cochrane systematic review by Woods et al. (2012)
significant difference between the intervention and placebo group examined the effect of cognitive stimulation on cognitive and
with respect to functional performance (MD: 0.76; 95% CI = 0.91, non-cognitive outcomes. Fifteen RCTs satisfied the inclusion
2.43). There was no difference in behaviour between groups criteria of the review. Fourteen trials reported on the effect of
according to the pooled analysis of two studies (standard mean cognitive stimulation on cognitive function. The pooled analysis of
difference (SMD) = 0.09; 95% CI = 0.58, 0.77; test for heterogene- the fourteen trials detected a statistically significant beneficial
ity: x2 = 0, p = 0.97, I2 = 0%). There was no difference in cognitive effect of cognitive stimulation (Standard Mean Differrence (SMD):
function between groups according to one trial (OR = 0.91; 95% 0.41; 95% CI = 0.25, 0.57; test for heterogeneity: x2 = 7.98, p = 0.84,
CI = 0.25, 3.34). One RCT failed to detect a difference between I2 = 0%). A pooled analysis (n = 4) detected a beneficial effect of
groups with respect to memory (SMD: 0.40; 95% CI = 0.66, 1.46). cognitive stimulation on communication and social interaction
The pooled analysis of two studies failed to detect a difference (SMD: 0.44; 95% CI = 0.17, 0.71; test for heterogeneity: x2 = 2.24,
between intervention and placebo with respect to cognitive p = 0.52, I2 = 0%). Pooled analyses detected that cognitive stimula-
function measured by the Paired Associated Learning Test (SMD: tion was effective in improving self-reported quality of life (SMD:
0.03; 95% CI = 1.48, 1.42; test for heterogeneity: x2 = 3.83, 0.38; 95% CI = 0.11, 0.65; test for heterogeneity: x2 = 2.15, p = 0.54,
p = 0.05, I2 = 74%). Similarly, lecithin did not exert a beneficial effect I2 = 0%) but not self- (SMD: 0.22; 95% CI = 0.09, 0.53; test for
on orientation according to the pooled analysis of two studies heterogeneity: x2 = 5.30, p = 0.26, I2 = 25%) or staff-reported mood
(SMD: 0.12; 95% CI = 0.84, 0.60; test for heterogeneity: x2 = 1.38, (SMD: 0.05; 95% CI = 0.21, 0.31; test for heterogeneity: x2 = 3.16,
p = 0.24, x2 = 28%). Finally, the only study to provide adequate data p = 0.37, I2 = 5%). Similarly, additional pooled analyses failed to
on death failed to detect a statistically significant difference in detect a difference between intervention and control groups with
mortality between groups (OR = 0.31; 95% CI = 0.03, 3.38). Consid- respect to ADL (SMD = 0.21; 95% CI = 0.05, 0.47; test for
ered cumulatively, the reviewers concluded that lecithin did not heterogeneity: x2 = 0.22, p = 0.97, I2 = 0%), behaviour (SMD = 0.13;
appear to delay the progression of AD. 95% CI = 0.07, 0.32; test for heterogeneity: x2 = 2.61, p = 0.92,
I2 = 0%), or behaviour problems (SMD = 0.14; 95% CI = 0.44, 0.17;
3.2.8. Physical activity test for heterogeneity: x2 = 1.13, p = 0.57, I2 = 0%). The review
The effect of physical activity on cognitive function, physical suggested that cognitive stimulation may be associated with a
function, physical fitness, and behaviour was examined by Heyn beneficial effect with respect to cognitive function, communica-
et al. (2004). Randomized trials examining the effect of physical tion/social interaction, and self-reported quality of life (Woods
activity in older ( > 65 years of age) adults with dementia, AD, or et al., 2012).
other cognitive impairments were sought. Thirty trials satisfied the
eligibility criteria. According to the series of pooled analyses, 4. Conclusion
subjects assigned to the intervention group experienced statisti-
cally significant improvements in functional performance (ES = 4.1. Risk factors associated with AD onset
0.59; 95% CI = 0.43, 0.76, P < 0.001), behaviour (ES = 0.54; 95%
CI = 0.36, 0.72, P < 0.001), and cognitive function (ES = 0.57; 95% According to the available evidence, head injury in males,
CI = 0.38, 0.75, P < 0.001). Exercise training also proved to be depression, mild cognitive impairment, age, diabetes mellitus,
beneficial for physical fitness (ES = 0.69; 95% CI = 0.58, 0.80), a conjugated equine estrogen use with medroxyprogesterone
measure which encompassed several outcomes including cardio- acetate, and exposure to pesticides were all associated with an
vascular fitness, strength, flexibility, and BMI. When all study increased risk of AD. With respect to genetic factors, APOE e4
outcomes (cardiovascular, strength, flexibility, BMI, cognitive, remained the strongest predictor of Alzheimer’s disease. The T/T
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 181

genotype of IL-1b +3953 SNP, the T/T genotype of the IL-1a 889C/ suggest that vitamin B-12 consumed in isolation was not effective
T polymorphism, 1082G/A polymorphism of IL-10 gene, I allele of for the improvement of cognitive function in subjects with
the ACE D/I polymorphism, APOE promoter 491A/T and 219T/G dementia, AD, or cognitive impairment (Malouf and Areosa,
polymorphisms, TT genotype of TNF-a SNP 850, C allele of OLR1 2003). Two reviews reported that there was insufficient evidence
30 -UTR (+1073) polymorphism, rs744373 polymorphism of BIN1, to support a beneficial effect of fatty acid supplementation
rs3764650 polymorphism of ABCA7, rs670139 polymorphism of (Issa et al., 2006; Dangour et al., 2010) and two Cochrane reviews
MS4A4E, and rs9349407 polymorphism of CD2AP were all reported no improvements attributed to vitamin E (Farina et al.,
associated with an increased risk of AD. Several identified genetic 2012) or lecithin (Higgins and Flicker, 2000). Although physical
factors appeared to confer susceptibility to AD in specific activity was not associated with improvements in cognitive
populations. MTHFR C677T polymorphism, SORL1 SNPs function according to one review (Littbrand et al., 2011), an earlier
rs2070045, rs3824968, rs2282649, and VLDLR 50 -UTR genotype systematic review reported a beneficial effect with respect to
2 were associated with an increased risk of AD in Asians, GA/AA cognitive function and other outcomes (Heyn et al., 2004). Finally,
genotype of ALDH2 in East Asian men, PS-1 2/2 genotype in results from a Cochrane review suggested that cognitive stimula-
Europeans, rs3818361 polymorphism of CR1 in Caucasians, SORL1 tion may improve cognitive function, communication/social
SNPs 5, 8, 9, 10, and 19 in whites, SORL1 SNPs 19 and 25 in Asians, interaction, and self-reported quality of life (Woods et al., 2012).
and CC + CG genotype of the BACE1 exon 5 (G/C) polymorphism in Considered cumulatively, there was evidence to suggest that
APOE e4 non-carriers. physical and cognitive activities may exert a beneficial effect
Statin use and several genetic factors, including APOE e2, on cognitive function and other indicators of dementia
rs610932 polymorphism of MS4A6A, and rs3865444 of CD33, were progression, while higher educational attainment may contribute
associated with a reduced risk of AD. Genetic factors that were to faster cognitive decline in subjects with dementia. As our
associated with a protective effect in specific populations include review was restricted to existing systematic reviews, this report
PS-1 2/2 genotype in North Americans, rs11136000 polymorphism does not provide a comprehensive list of putative factors
of CLU and rs3851179 polymorphism of PICALM in Caucasians, associated with AD progression. The identification and synthesis
VLDLR 50 -UTR genotype 2 in non-Asians, SORL1 SNPs 4 and 12 in of primary studies to attain a more comprehensive list of putative
whites, SORL1 SNPs 23 and 24 in Asians, and CC + CG genotype of factors is warranted.
the BACE1 exon 5 (G/C) polymorphism in APOE e4 carriers.
There was no evidence of an association between AD risk and Conflict of interest statement
metabolic syndrome, conjugated equine estrogen use (without
progestin), antihypertensive medication use, lead, and several The authors declare that there are no conflicts of interest.
genetic factors including, but not limited to APOE promoter
427 T/C, CTSD polymorphism, and TNF-a SNPs 308, 863, Update
238, 1031.
With respect to lifestyle factors, current smoking and lower This update identifies systematic reviews reporting on factors
social engagement were identified as factors associated with an associated with the onset or progression of AD published after the
increased risk of AD. The available evidence was suggestive of a original search date of our systematic review. As described in
reduced risk of AD associated with light-to-moderate alcohol methods section, a supplementary search of the Medline database
consumption, compliance with a Mediterranean diet, higher was performed to identify recently published systematic reviews
educational attainment, and regular engagement in physically that may be of interest to reader. Study screening, data extraction,
and cognitively stimulating activities. and quality appraisal of each systematic review was conducted by a
While a number of risk factors appeared to be associated with single reviewer.
risk of AD onset, the associations were weak, at best, for a majority Our supplementary search for systematic reviews on risk
of factors. Conflicting findings across studies or inadequate factors for AD onset included reports published between 2012 and
evidence precluded definitive conclusions with respect to the 2015. We included moderate or high quality (AMSTAR score >3)
effect of a majority of the examined factors on risk of developing systematic reviews reporting on the association between genetic
AD. Certain factors (e.g., mild cognitive impairment and depres- and non-genetic risk factors and onset of AD in adults. We excluded
sion) are suspected prodromes of AD rather than risk factors. narrative reviews, primary research studies, low quality systematic
Finally, as we included multiple reviews reporting on the same risk reviews (AMSTAR score 3), and systematic reviews which failed
factor, there was often considerable overlap of included studies to distinguish AD from all-cause dementia or dementia subtypes.
across reviews. The search yielded 333 citations, 76 of which were deemed
Further research is required to confirm the etiological or potentially relevant. Following full-text review, two primary
protective role of a number of risk factors. research studies were excluded, 18 reviews were excluded due
to low quality, 9 reviews were excluded because they failed to
4.2. Risk factors associated with AD progression distinguish AD from all-cause dementia or dementia subtype, and
one review did not report on risk of AD onset. Two reviews
Twelve systematic reviews were identified that reported on identified in the original search were also excluded from the
factors associated with cognitive and non-cognitive indicators of update. The search for systematic reviews on factors associated
AD progression. Two systematic reviews reported no association with AD progression, published between 2013 and 2015, produced
between educational attainment and mortality (Paradise et al., 241 citations, four of which were deemed potentially relevant. We
2009; Meng and D'Arcy, 2012). However, one review reported an included moderate or high quality systematic reviews reporting on
association between higher educational attainment and faster the association between risk factors and the progression of AD in
cognitive decline (Meng and D'Arcy, 2012). Apolipoprotein E4 adults with a previous AD diagnosis. We excluded narrative
(APoE4) did not predict progression of AD (Allan and Ebmeier, reviews, primary research studies, low quality systematic reviews
2011). Two reviews failed to detect a beneficial effect of folic acid (AMSTAR score 3). Of the four potentially relevant studies, one
supplementation, with or without vitamin B-12, on cognitive was excluded due to low methodological quality. A list of excluded
function (Dangour et al., 2010; Malouf et al., 2003; Malouf and systematic reviews on onset and progression, along with reasons
Grimley, 2008). Similarly, findings of a Cochrane systematic review for exclusion, can be found in Supplementary material III.
182 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

A total of 45 systematic reviews satisfied the eligibility criteria Other factors associated with AD onset: A total of eight
for the update on AD onset. Of these, 23 reviews reported on systematic reviews assessed the association between various
genetic factors associated with AD onset and 22 reported on dietary factors and incident AD. Four reviews (Cooper et al., 2015;
non-genetic risk factors. Three systematic reviews were included Singh et al., 2014; Psaltopoulou et al., 2013; Lourida et al., 2013)
in the update on AD progression. A list of systematic reviews reported statistically significant protective effects of Mediterra-
identified in the updated search is provided in Supplementary nean diet on the development of AD, or conversion from mild
material III. Findings of the systematic reviews are briefly cognitive impairment to AD. Other studies reported significant
summarized below. For more details on non genetic and genetic protective effect of high fish intake, n-3 fatty acids, higher fruit and
risk factors see Table 123 and 124 respectively in Supplementary vegetable consumption, vitamins C and E (Loef et al., 2012; Li et al.,
material II. 2012; Beydoun et al., 2014; Wu et al., 2015). Evidence with respect
Genetic factors associated with AD onset: A total of 23 to the protective effects of antioxidants, beta-carotene and caffeine
systematic reviews/meta-analysis were identified that quantita- consumption on the risk of AD onset were conflicting (Beydoun
tively assessed the association of several genes (CETP, CYP2D6*4, et al., 2014).
CYP2D6, APOE, ESR1, HFE, BDNF, CR1, cathepsin D,Interleukin-1A, A total of six systematic reviews reported associations between
APOC1, ACT, PLAU, UBQLN1, ACE, ABCA1, NEDD9, TF and VEGF) with various drugs and the risk of AD. Three reviews (Macedo et al.,
the risk of AD onset. Three systematic reviews investigated the 2014; Song et al., 2013; Wong et al., 2013) assessed the potential
association between different APOE alleles and risk of AD onset. Fei protective effect of statin use on the development of AD. Of these,
and Jianhua (2013) observed a positive and significant association Macedo et al. (2014) and Wong et al. (2013) reported pooled effect
between APOE e4 allele and the risk of progression from MCI to AD. sizes, both of which were statistically significant. Wang (2015)
Liu et al. (2014) reported that APOE e4 and APOE e3 alleles were reported that use of NSAIDs and ASA were associated with a
positively and negatively associated with AD in Chinese and Indian significantly lower risk of developing AD in observational studies,
population, respectively. However, no significant association of however the only RCT identified found no significant benefit of
APOE e2 allele with AD was observed in the Indian population celecoxib. Yang et al. (2013) reviewed the effect of raloxifene on the
(Agarwal and Tripathi, 2014). Three systematic reviews assessed development of AD. They identified only one study, which reported
the association between the ESR1 polymorphisms [PvuII polymor- a statistically significant benefit, but concluded that there was
phism (T/C, rs223493); Xbal (A/G, rs9340799)] and the risk of AD in insufficient evidence to confirm benefit. One systematic review
different populations. In the meta-analysis by Cheng et al. (2014), a (O’Brien et al., 2014) which included one randomized trial and
positive association was noted between the ESR1 Pvu II polymor- eight observational studies reported that postmenopausal hor-
phism in the Caucasian population, whereas, no association was monal therapy was not associated with AD.
found between the ESR1 XbaI polymorphisms and risk of AD in any Two reviews reported associations between co-morbid diseases
population. Pan et al. (2014) reported a weak protective role of the and risk of AD. Diniz et al. (2013) and Cheng et al. (2012) reported
Xbal ESR1 polymorphism in the Asian [but not Caucasian] that late-life depression and diabetes were significantly associated
population. Similarly, Wang (2014) reported a weak protective with the development of AD, respectively.
role of the PvuII ESR1 polymorphism on AD onset in the Caucasian Six reviews reported on associations between various serum
population, but failed to find a significant association between XbaI biomarkers or nutrient levels and risk of AD. Low Apo E levels
ESR1 polymorphism and risk of AD. Lin et al. (2014) investigated (Wang et al., 2014), high CRP levels (Koyama et al., 2013), high
the association between two BDNF gene polymorphisms and risk homocysteine levels (Beydoun et al., 2014) but not elevated IL-6
of AD in different subgroups and found that while the A allele of levels (Koyama et al., 2013) were significantly associated with the
rs6265 BDNF polymorphism was associated with increased the risk development of AD. Annweiler et al. (2013) reported that vitamin D
of AD in the Caucasian females and female LOAD patients, the levels were lower in patients with AD, potentially implicating this
rs2030324 BDNF polymorphism was not associated with the risk of vitamin in the development of AD. No conclusions could be drawn
AD in any subgroup. Liu et al. (2013) investigated the association on associations between vitamin B12 and development of AD
between the two VEGF polymorphisms [-2578C/A polymorphism; (O'Leary et al., 2012).
1154G/A polymorphism] and risk of AD based on their APO e4 Three additional reviews of various modifiable risk factors were
status. The authors reported that VEGF 2578C/A and VEGF- identified. Zhong et al. (2015) reported that current and ever
1154G/A polymorphisms were associated with increased and smokers demonstrated a significantly elevated risk of developing
decreased risk of AD when the APOE e4 status was negative, AD, while former smokers did not. The risk was most pronounced
respectively (Liu et al., 2013). in individuals that were APOE e4 non-carriers. Beydoun et al.
The following other positive associations with AD were (2014) also reviewed the association between smoking and AD,
identified in the overall population or different subgroups: CETP and found that current or ever smokers were significantly more
rs5882A>G; CYP2D6*4A/G; H63D HFE; IL-1A 22889C/T; likely to develop AD than never smokers. Beydoun et al. (2014) also
rs11568822 APOC1; rs2227564 PLAU; UBQ-8i UBQLN1[LOAD risk]; reported on the effects of physical activity, alcohol consumption,
TNFa G308A [Chinese population]; rs4291 ACE [Risk of SAD in and educational attainment. While the evidence for alcohol was
Caucaisan and in older cases]; TF rs1049296; ACT 15A/T [AD as found to be conflicting, physical activity and higher education
well as LOAD in Caucasians]; rs6656401 and rs3818361 CR1 [LOAD [over versus under 8 years] demonstrated statistically significant
risk] (Chen et al., 2014; Lu et al., 2014; Lin et al., 2014; Luo et al. protective effect. Vergara et al. (2013) studied the association
2014; Qin et al., 2012; Zhou et al,, 2014; Guan et al., 2012; Wu et al., between occupational exposure to low frequency magnetic field
2013; Zhang and Jia, 2014; Wang et al., 2015a,b, 2013a). No exposure and AD. The authors found that such exposure was
association with AD onset could be demonstrated for the following associated with a significantly higher risk of developing AD,
polymorphisms in the overall population or in any subgroup: CETP although limitations such as heterogeneity across studies and
rs708272 T > C, C282Y HFE, C224T cathepsin D; rs4343 and evidence of publication bias prevented firm conclusions from
rs1800764 ACE; rs2230806, rs4149313 and rs2230808 ABCA 1 being drawn.
(Chen et al., 2014; Lin et al., 2012; Mo et al., 2014; Wang et al., Factors associated with AD progression: Three out of four
2013a,b, 2015a). NEDD9 rs760678 polymorphism was found to be systematic reviews reporting on AD progression had an AMSTAR
associated with decreased risk of AD in the Caucasian subgroup but score of >3. These systematic reviews reported that natural
not the Asian subgroup (Wang et al., 2012). medicine (Gingko), exercise and occupational therapy may have
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 183

beneficial effects in AD patients (Yang, 2014; Farin, 2014; Mclaren, Bateman, R.J., Aisen, P.S., De Strooper, B., Fox, N.C., Lemere, C.A., Ringman, J.M.,
2014). Salloway, S., Sperling, R.A., Windisch, M., Xiong, C., 2011. Autosomal-dominant
Alzheimer's disease: a review and proposal for the prevention of Alzheimer's
disease. Alzheimer's Res.Ther. 3 (1), 1.
Acknowledgments Bates, M.N., 2006. Mercury amalgam dental fillings: an epidemiological assessment.
Int. J. Hyg. Environ. Health 209 (4), 309–316.
Beri, A., Sural, N., Mahajan, S.B., 2009. Non-atheroprotective effects of statins: a
This project was funded by the Public Health Agency of Canada systematic review. Am. J. Cardiovasc. Drugs 9 (6), 361–370.
in association with Neurological Health Charities Canada through a Bertram, L., McQueen, M.B., Mullin, K., Blacker, D., Tanzi, R.E., 2007. Systematic
contribution agreement administered through the University of meta-analyses of Alzheimer disease genetic association studies: the AlzGene
database. Nat. Genet. 39 (1), 17–23.
Ottawa. We thank Dr. Lynn Beattie for her consultation in preparing Beydoun, M.A., Beydoun, H.A., Wang, Y., 2008. Obesity and central obesity as risk
this manuscript. We also thank Lindsey Sikora for her assistance in factors for incident dementia and its subtypes: a systematic review and meta-
developing the search strategy. Daniel Krewski is the Natural analysis. Obes. Rev. 9 (3), 204–218.
Beydoun, M.A., Beydoun, H.A., Gamaldo, A.A., Teel, A., Zonderman, A.B., Wang, Y.,
Sciences and Engineering Research Council of Canada Chair in Risk
2014. Epidemiologic studies of modifiable factors associated with cognition and
Science at the University of Ottawa. dementia: systematic review and meta-analysis. BMC Public Health 14, 643.
Biessels, G.J., Staekenborg, S., Brunner, E., Brayne, C., Scheltens, P., 2006. Risk of
Appendix A. Supplementary data dementia in diabetes mellitus: a systematic review. Lancet Neurol. 5 (1), 64–74.
Blasko, I., Jellinger, K., Kemmler, G., Krampla, W., Jungwirth, S., Wichart, I., Tragl, K.
H., Fischer, P., 2008. Conversion from cognitive health to mild cognitive
Supplementary data associated with this article can be impairment and Alzheimer's disease: prediction by plasma amyloid beta 42,
found, in the online version, at http://dx.doi.org/10.1016/j. medial temporal lobe atrophy and homocysteine. Neurobiol. Aging 29 (1), 1–11.
Blasko, I., Kemmler, G., Jungwirth, S., Wichart, I., Krampla, W., Weissgram, S.,
neuro.2017.03.006. Jellinger, K., Tragl, K.H., Fischer, P., 2010. Plasma amyloid beta-42 independently
predicts both late-onset depression and Alzheimer’s disease. Am. J. Geriatr.
References Psychiatry 18 (11), 973–982.
Bottiglieri, T., Parnetti, L., Arning, E., Ortiz, T., Amici, S., Lanari, A., Gallai, V., 2001.
Plasma total homocysteine levels and the C677T mutation in the
Abellan van Kan, G., Rolland, Y., Gillette-Guyonnet, S., Gardette, V., Annweiler, C.,
methylenetetrahydrofolate reductase (MTHFR) gene: a study in an Italian
Beauchet, O., Andrieu, S., Vellas, B., 2012. Gait speed, body composition, and
population with dementia. Mech. Ageing Dev. 122 (16), 2013–2023.
dementia. The EPIDOS-Toulouse cohort. J. Gerontol. 67 (4), 425–432.
Boyle, P.A., Buchman, A.S., Barnes, L.L., Bennett, D.A., 2010. Effect of purpose in life
Agarwal, R., Tripathi, C.B., 2014. Association of apolipoprotein E genetic variation in
on risk of incident Alzheimer disease and mild cognitive impairment in
Alzheimer's disease in Indian population: a meta-analysis. Am. J. Alzheimer's
community-dwelling older persons. Arch. Gen. Psychiatry 67 (3), 304–310.
Dis. Other Dementias 29 (7), 575–582.
Breitner, J.C., Baker, L.D., Montine, T.J., Meinert, C.L., Lyketsos, C.G., Ashe, K.H.,
Aglukkaq, L., 2011. Alzheimer's Awareness Month: January 2011. Health Canada.
Brandt, J., et al., 2011. Extended results of the Alzheimer's disease anti-
http://www.hc-sc.gc.ca/ahc-asc/minist/messages/_2011/2011_01_04-eng.php.
inflammatory prevention trial. Alzheimer's Dementia 7 (4), 402–411.
Aguirre, E., Woods, R.T., Spector, A., Orrell, M., 2013. Cognitive stimulation for
Breteler, M.M., van Duijn, C.M., Chandra, V., Fratiglioni, L., Graves, A.B., Heyman, A.,
dementia: a systematic review of the evidence of effectiveness from
Jorm, A.F., et al., 1991. Medical history and the risk of Alzheimer's disease: a
randomised controlled trials. Ageing Res. Rev. 12 (1), 253–262.
collaborative re-analysis of case-control studies. EURODEM Risk Factors
Ahlbom, A., 2001. Neurodegenerative diseases and depressive symptoms in relation
Research Group. Int. J. Epidemiol. 20 (Suppl. 2), S36–S42.
to EMF. Bioelectromagnetics S132–43.
Brinker, K., 2011. Serum Lipids and Statin Therapy in Relation to Alzheimer’s
Ahtiluoto, S., Polvikoski, T., Peltonen, M., Solomon, A., Tuomilehto, J., Winblad, B.,
Disease: Exploring the Relationship Between Elevated Serum Cholesterol Levels
Sulkava, R., Kivipelto, M., 2010. Diabetes, Alzheimer disease, and vascular
and Alzheimer’s Disease Dementia. Diss. Cornell University.
dementia: a population-based neuropathologic study. Neurology 75 (13), 1195–
Bruscoli, M., Lovestone, S., 2004. Is MCI really just early dementia? A systematic
1202.
review of conversion studies. Int. Psychogeriatr. 16 (2), 129–140.
Allan, C.L., Ebmeier, K.P., 2011. The influence of ApoE4 on clinical progression of
Caamano-Isorna, F., Corral, M., Montes-Martinez, A., Takkouche, B., 2006. Education
dementia: a meta-analysis. Int. J. Geriatr. Psychiatry 26 (5), 520–526.
and dementia: a meta-analytic study. Neuroepidemiology 26 (4), 226–232.
Almeida, O.P., Hulse, G.K., Lawrence, D., Flicker, L., 2002. Smoking as a risk factor for
Cataldo, J.K., Prochaska, J.J., Glantz, S.A., 2010. Cigarette smoking is a risk factor for
Alzheimer's disease: contrasting evidence from a systematic review of case-
Alzheimer's Disease: an analysis controlling for tobacco industry affiliation. J.
control and cohort studies. Addiction 97 (1), 15–28.
Alzheimer's Disease 19 (2), 465–480.
Alzheimer Society. Rising Tide: The Impact of Dementia on Canadian Society. 2010.
Chang, C.L., 2008. Obesity and alzheimer's disease. Hong Kong J. Psychiatry 18 (1),
http://www.alzheimer.ca//media/Files/national/Advocacy/ASC_Rising%
28–35.
20Tide Executive%20Summary_Eng.ashx.
Chang-Quan, H., Hui, W., Chao-Min, W., Zheng-Rong, W., Jun-Wen, G., Yong-Hong, L.,
Amieva, H., Stoykova, R., Matharan, F., Helmer, C., Antonucci, T.C., Dartigues, J.F.,
Yan-You, L., Qing-Xiu, L., 2011. The association of antihypertensive medication
2010. What aspects of social network are protective for dementia? Not the
use with risk of cognitive decline and dementia: a meta-analysis of longitudinal
quantity but the quality of social interactions is protective up to 15 years later.
studies. Int. J. Clin. Pract. 65 (12), 1295–1305.
Psychosom. Med. 72 (9), 905–911.
Chen, Y.C., Chen, T.F., Yip, P.K., Hu, C.Y., Chu, Y.M., Chen, J.H., 2010. Body mass index
Annerbo, S., Wahlund, L.O., Lökk, J., 2006. The significance of thyroid-stimulating
(BMI) at an early age and the risk of dementia. Archiv. Gerontol. Geriatrics 50
hormone and homocysteine in the development of Alzheimer’s disease in mild
(Suppl. 1), S48–S52.
cognitive impairment: a 6-year follow-up study. Am. J. Alzheimer’s Dis. Other
Chen, J.J., Li, Y.M., Zou, W.Y., Fu, J.L., 2014. Relationships between CETP genetic
Dementias 21 (3), 182–188.
polymorphisms and Alzheimer's disease risk: a meta-analysis. DNA Cell Biol. 33,
Annweiler, C., Llewellyn, D.J., Beauchet, O., 2013. Low serum vitamin D
807–815.
concentrations in Alzheimer's disease: a systematic review and meta-analysis. J.
Cheng, D., Noble, J., Tang, M.X., Schupf, N., Mayeux, R., Luchsinger, J.A., 2011. Type 2
Alzheimer's Dis. 33, 659–674.
diabetes and late-onset Alzheimer's disease. Dementia Geriatric Cognit. Disord.
Anstey, K.J., von Sanden, C., Salim, A., O’Kearney, R., 2007. Smoking as a risk factor for
31 (6), 424–430.
dementia and cognitive decline: a meta-analysis of prospective studies. Am. J.
Cheng, G., Huang, C., Deng, H., Wang, H., 2012. Diabetes as a risk factor for dementia
Epidemiol. 166 (4), 367–378.
and mild cognitive impairment: a meta-analysis of longitudinal studies. Intern.
Anstey, K.J., Lipnicki, D.M., Low, L.F., 2008. Cholesterol as a risk factor for dementia
Med. J. 42, 484–491.
and cognitive decline: a systematic review of prospective studies with meta-
Cheng, D., Liang, B., Hao, Y., Zhou, W., 2014. Estrogen receptor alpha gene
analysis. Am. J. Geriatr. Psychiatry 16 (5), 343–354.
polymorphisms and risk of Alzheimer's disease: evidence from a meta-analysis.
Anstey, K.J., Mack, H.A., Cherbuin, N., 2009. Alcohol consumption as a risk factor for
Clin. Interv. Aging 9, 1031–1038.
dementia and cognitive decline: meta-analysis of prospective studies. Am. J.
Chu, L.W., Tam, S., Wong, R.L., Yik, P.Y., Song, Y., Cheung, B.M., Morley, J.E., Lam, K.S.,
Geriatr. Psychiatry 17 (7), 542–555.
2010. Bioavailable testosterone predicts a lower risk of Alzheimer's disease in
Anstey, K.J., Cherbuin, N., Budge, M., Young, J., 2011. Body mass index in midlife and
older men. J. Alzheimer's Dis. 21 (4), 1335–1345.
late-life as a risk factor for dementia: a meta-analysis of prospective studies.
Clarke, R., Smith, A.D., Jobst, K.A., Refsum, H., Sutton, L., Ueland, P.M., 1998. Folate,
Obes. Rev. 12 (5), e426–37.
vitamin B12, and serum total homocysteine levels in confirmed Alzheimer
Barnes, D.E., Yaffe, K., 2011. The projected effect of risk factor reduction on
disease. Arch. Neurol. 55 (11), 1449–1455.
Alzheimer's disease prevalence. Lancet Neurol. 10 (9), 819–828.
Cooper, C., Sommerlad, A., Lyketsos, C.G., Livingston, G., 2015. Modifiable predictors
Barnes, D.E., Yaffe, K., Byers, A.L., McCormick, M., Schaefer, C., Whitmer, R.A., et al.,
of dementia in mild cognitive impairment: a systematic review and meta-
2012. Midlife vs. late-life depressive symptoms and risk of dementia:
analysis. Am. J. Psychiatry 172, 323–334.
Differential effects for Alzheimer’s disease and vascular dementia. Arch. Gen.
Crichton, G.E., Bryan, J., Murphy, K.J., Buckley, J., 2010. Review of dairy consumption
Psychiatry 69 (5), 493–498.
and cognitive performance in adults: findings and methodological issues.
Barranco Quintana, J.L., Allam, M.F., Serrano Del Castillo, A., Fernandez-Crehuet
Dementia Geriatric Cognit. Disord. 30 (4), 352–361.
Navajas, R., 2007. Alzheimer's disease and coffee: a quantitative review. Neurol.
Res. 29 (1), 91–95.
184 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

Cukierman, T., Gerstein, H.C., Williamson, J.D., 2005. Cognitive decline and dementia Fotuhi, M., Mohassel, P., Yaffe, K., 2009. Fish consumption, long-chain omega-3 fatty
in diabetes—systematic overview of prospective observational studies. acids and risk of cognitive decline or Alzheimer disease: a complex association.
Diabetologia 48 (12), 2460–2469. Nat. Clin. Pract. Neurol. 5 (3), 140–152.
Dangour, A.D., Whitehouse, P.J., Rafferty, K., Mitchell, S.A., Smith, L., Hawkesworth, Fratiglioni, L., Paillard-Borg, S., Winblad, B., 2004. An active and socially integrated
S., Vellas, B., 2010. B-vitamins and fatty acids in the prevention and treatment of lifestyle in late life might protect against dementia. Lancet Neurol. 3 (6), 343–
Alzheimer's disease and dementia: a systematic review. J. Alzheimer's Dis. 22 353.
(1), 205–224. Friedland, R.P., Fritsch, T., Smyth, K.A., Koss, E., Lerner, A.J., Chen, C.H., Petot, G.J.,
Daviglus, M.L., Plassman, B.L., Pirzada, A., Bell, C.C., Bowen, P.E., Burke, J.R., Connolly Debanne, S.M., 2001. Patients with Alzheimer's disease have reduced activities
Jr., E.S., et al., 2011. Risk factors and preventive interventions for Alzheimer in midlife compared with healthy control-group members. Proc. Natl. Acad. Sci.
disease: state of the science. Arch. Neurol. 68 (9), 1185–1190. U. S. A. 98 (6), 3440–3445.
Debette, S., Markus, H.S., 2010. The clinical importance of white matter Gao, S., Hendrie, H.C., Hall, K.S., Hui, S., 1998. The relationships between age, sex, and
hyperintensities on brain magnetic resonance imaging: systematic review and the incidence of dementia and Alzheimer disease: a meta-analysis. Arch. Gen.
meta-analysis. BMJ 341, c3666. Psychiatry 55 (9), 809–815.
Del Bo, R., Scarlato, M., Ghezzi, S., Martinelli-Boneschi, F., Fenoglio, C., Galimberti, G., García, A.M., Sisternas, A., Hoyos, S.P., 2008. Occupational exposure to extremely
Galbiati, S., et al., 2006. Is M129 V of PRNP gene associated with Alzheimer's low frequency elective and magnetic fields and Alzheimer disease: a meta-
disease? A case-control study and a meta-analysis. Neurobiol. Aging 27 (5) analysis. Int. J. Epidemiol. 37 (2), 329–340.
(770. e1-770.e5). Gates, G.A., Anderson, M.L., McCurry, S.M., Feeney, M.P., Larson, E.B., 2011. Central
Desilets, A.R., Koslowski, D.A., Dunican, K.C., 2010. The role of statins in the auditory dysfunction as a harbinger of Alzheimer dementia. Arch. Otolaryngol.
prevention and treatment of Alzheimer’s disease. J. Pharm. Technol. 26, 276– Head Neck Surg. 137 (4), 390–395.
284. Gauthier, S., Wirth, Y., Mobius, H.J., 2005. Effects of memantine on behavioural
Devore, E.E., Grodstein, F., van Rooij, F.J., Hofman, A., Stampfer, M.J., Witteman, J.C., symptoms in Alzheimer's disease patients: an analysis of the Neuropsychiatric
Breteler, M.M., 2010. Dietary antioxidants and long-term risk of dementia. Arch. Inventory (NPI) data of two randomised, controlled studies. Int. J. Geriatr.
Neurol. 67 (7), 819–825. Psychiatry 20 (5), 459–464.
Di Bona, D., Plaia, A., Vasto, S., Cavallone, L., Lescai, F., Franceschi, C., Licastro, F., Gelber, R.P., Petrovitch, H., Masaki, K.H., Ross, G.W., White, L.R., 2011. Coffee intake in
Colonna-Romano, G., Lio, D., Candore, G., Caruso, C., 2008. Association between midlife and risk of dementia and its neuropathologic correlates. J. Alzheimer's
the interleukin-1b polymorphisms and Alzheimer's disease: a systematic Dis. 23 (4), 607–615.
review and meta-analysis. Brain Res. Rev. 59 (1), 155–163. Gelber, R.P., Petrovitch, H., Masaki, K.H., Abbott, R.D., Ross, G.W., Launer, L.J., White,
Di Bona, D., Candore, G., Franceschi, C., Licastro, F., Colonna-Romano, G., Cammà, C., L.R., 2012. Lifestyle and the risk of dementia in Japanese-american men. J. Am.
Lio, D., Caruso, C., 2009. Systematic review by meta-analyses on the possible Geriatr. Soc. 60 (1), 118–123.
role of TNF-b polymorphisms in association with Alzheimer’s disease. Brain Gorospe, E.C., Dave, J.K., 2007. The risk of dementia with increased body mass index.
Res. Rev. 61 (2), 60–68. Age Ageing 36 (1), 23–29.
Diniz, B.S., Pinto, J.A. Jr., Gonzaga, M.L., Guimaraes, F.M., Gattaz, W.F., Forlenza, O.V., Graves, A.B., van Duijn, C.M., Chandra, V., Fratiglioni, L., Heyman, A., Jorm, A.F.,
2009. To treat or not to treat? A meta-analysis of the use of cholinesterase Kokmen, E., et al., 1991a. Alcohol and tobacco consumption as risk factors for
inhibitors in mild cognitive impairment for delaying progression to Alzheimer's Alzheimer's disease: a collaborative re-analysis of case-control studies.
disease. Eur. Archi. Psychiatry Clin. Neurosci. 259 (4), 248–256. EURODEM Risk Factors Research Group. Int. J. Epidemiol. 20 (Suppl. 2), S48–S57.
Diniz, B.S., Butters, M.A., Albert, S.M., Dew, M.A., Reynolds 3rd, C.F., 2013. Late-life Graves, A.B., van Duijn, C.M., Chandra, V., Fratiglioni, L., Heyman, A., Jorm, A.F.,
depression and risk of vascular dementia and Alzheimer's disease: systematic Kokmen, E., et al., 1991b. Occupational exposures to solvents and lead as risk
review and meta-analysis of community-based cohort studies. Br. J. Psychiatry factors for Alzheimer's disease: a collaborative re-analysis of case-control
202, 329–335. studies. EURODEM Risk Factors Research Group. Int. J. Epidemiol. 20 (Suppl. 2),
Dotson, V.M., Beydoun, M.A., Zonderman, A.B., 2010. Recurrent depressive S58–S61.
symptoms and the incidence of dementia and mild cognitive impairment. Gray, S.L., Walker, R., Dublin, S., Haneuse, S., Crane, P.K., Breitner, J.C., Bowen, J.,
Neurology 75 (1), 27–34. McCormick, W., Larson, E.B., 2011. Histamine-2 receptor agonist use and
Driver, J.A., Beiser, A., Au, R., Kreger, B.E., Splansky, G.L., Kurth, T., Kiel, D.P., Lu, K.P., incident dementia in an older. J. Am. Geriatric Soci. 59 (2), 251–257.
Seshadri, S., Wolf, P.A., 2012. Inverse association between cancer and Gu, Y., Luchsinger, J.A., Stern, Y., Scarmeas, N., 2010a. Mediterranean diet,
Alzheimer's disease: results from the Framingham Heart Study. BMJ 344, e1442. inflammatory and metabolic biomarkers, and risk of Alzheimer's disease. J.
Dublin, S., Anderson, M.L., Haneuse, S.J., Heckbert, S.R., Crane, P.K., Breitner, J.C., Alzheimer's Dis. 22 (2), 483–492.
McCormick, W., et al., 2011. Atrial fibrillation and risk of dementia: a prospective Gu, Y., Nieves, J.W., Stern, Y., Luchsinger, J.A., Scarmeas, N., 2010b. Food combination
cohort study. J. Am. Geriatr. Soc. 59 (8), 1369–1375. and Alzheimer’s disease risk: a protective diet. Arch. Neurol. 67 (6), 699–706.
ESHRE Capri Workshop Groupi, 2011. Perimenopausal risk factors and future health. Guan, F., Gu, J., Hu, F., Zhu, Y., Wang, W., 2012. Association between alpha1-
Hum. Reprod. Update 17 (5), 706–717. antichymotrypsin signal peptide 15A/T polymorphism and the risk of
Elias-Sonnenschein, L.S., Viechtbauer, W., Ramakers, I.H., Verhey, F.R., Visser, P.J., Alzheimer's disease: a meta-analysis. Mol. Biol. Rep. 39, 6661–6669.
2011. Predictive value of APOE-4 allele for progression from MCI to AD-type Guerchet, M., Aboyans, V., Nubukpo, P., Lacroix, P., Clement, J.P., Preux, P.M., 2011.
dementia: a meta-analysis Journal of Neurology. Neurosurg. Psychiatry 82 (10), Ankle-brachial index as a marker of cognitive impairment and dementia in
1149–1156. general population. A systematic review. Atherosclerosis 216 (2), 251–257.
Elkins, J.S., Douglas, V.C., Johnston, S.C., 2004. Alzheimer disease risk and genetic Haan, M.N., Miller, J.W., Aiello, A.E., Whitmer, R.A., Jagust, W.J., Mungas, D.M., Allen,
variation in ACE: a meta-analysis. Neurology 62 (3), 363–368. L.H., Green, R., 2007. Homocysteine, B vitamins, and the incidence of dementia
Engelman, M., Agree, E.M., Meoni, L.A., Klag, M.J., 2010. Propositional density and and cognitive impairment: results from the Sacramento Area Latino Study on
cognitive function in later life: findings from the Precursors Study. J. Gerontol. Aging. Am. J. Clin. Nutr. 85 (2), 511–517.
Ser. B-Psychol.Sci. Soc. Sci. 65 (6), 706–711. Hamer, M., Chida, Y., 2009. Physical activity and risk of neurodegenerative disease: a
Eriksson, U.K., Bennet, A.M., Gatz, M., Dickman, P.W., Pedersen, N.L., 2010a. systematic review of prospective evidence. Psychol. Med. 39 (1), 3–11.
Nonstroke cardiovascular disease and risk of Alzheimer disease and dementia. Han, X.M., Wang, C.H., Sima, X., Liu, S.Y., 2011. Interleukin-6 174G/C polymorphism
Alzheimer Dis. Assoc. Disord. 24 (3), 213–219. and the risk of Alzheimer's disease in Caucasians: a meta-analysis. Neurosci.
Eriksson, U.K., Sjoberg, B.G., Bennet, A.M., de Faire, U., Pedersen, N.L., Frostegard, J., Lett. 504 (1), 4–8.
2010b. Low levels of antibodies against phosphorylcholine in Alzheimer's Hao, P.P., Chen, Y.G., Wang, J.L., Wang, X.L., Zhang, Y., 2011a. Meta-analysis of
disease. J. Alzheimer's Dis. 21 (2), 577–584. aldehyde dehydrogenase 2 gene polymorphism and Alzheimer's disease in East
Etminan, M., Gill, S., Samii, A., 2003. Effect of non-steroidal anti-inflammatory drugs Asians. Can. J. Neurol. Sci. 38 (3), 500–506.
on risk of Alzheimer's disease: systematic review and meta-analysis of Hao, Z., Wu, B., Wang, D., Liu, M., 2011b. Association between metabolic syndrome
observational studies. BMJ 327 (7407), 128. and cognitive decline: a systematic review of prospective population-based
Farina, N., Isaac, M.G., Clark, A.R., Rusted, J., Tabet, N., 2012. Vitamin E for Alzheimer's studies. Acta Neuropsychiatrica 23 (2), 69–74.
dementia and mild cognitive impairment. Cochrane Database Syst. Rev. 11, Hersi, M., Quach, P., Wang, M.D., Gomes, J., Gaskin, J., Krewski, D., 2016. Systematic
CD002854. reviews of factors associated with the onset and progression of neurological
Fei, M., Jianhua, W., 2013. Apolipoprotein epsilon4-allele as a significant risk factor conditions in humans: a methodological overview. Neurotoxicology (in this
for conversion from mild cognitive impairment to Alzheimer's disease: a meta- issue).
analysis of prospective studies. J. Mol. Neurosci. 50, 257–263. Hersi, M., 2015. Lifestyle-related Risk Factors for the Onset of Alzheimer’s Disease: a
Ferreira, P.C., Piai Kde, A., Takayanagui, A.M., Segura-Munoz, S.I., 2008. Aluminum as Systematic Review of the Literature. Thesis. University of Ottawa.
a risk factor for Alzheimer's disease. Rev. Lat. Am. Enfermagem 16 (1), 151–157. Heyn, P., Abreu, B.C., Ottenbacher, K.J., 2004. The effects of exercise training on
Fleminger, S., Oliver, D.L., Lovestone, S., Rabe-Hesketh, S., Giora, A., 2003. Head elderly persons with cognitive impairment and dementia: a meta-analysis.
injury as a risk factor for Alzheimer's disease: the evidence 10 years on; a partial Arch. Phys. Med. Rehab. 85 (10), 1694–1704.
replication Journal of Neurology. Neurosurg. Psychiatry 74 (7), 857–862. Higgins, J.P., Flicker, L., 2000. Lecithin for dementia and cognitive impairment.
Folin, M., Baiguera, S., Gallucci, M., Conconi, M.T., Di Liddo, R., Zanardo, A., Cochrane Database Syst. Rev. 4, CD001015.
Parnigotto, P.P., 2005. A cross-sectional study of homocysteine-, NO-levels, and Ho, R.C., Cheung, M.W., Fu, E., Win, H.H., Zaw, M.H., Ng, A., Mak, A., 2011. Is high
CT-findings in Alzheimer dementia, vascular dementia and controls. homocysteine level a risk factor for cognitive decline in elderly? A systematic
Biogerontology 6 (4), 255–260. review, meta-analysis, and meta-regression. Am. J. Geriatric Psychiatry 19 (7),
Forti, P., Olivelli, V., Rietti, E., Maltoni, B., Pirazzoli, G., Gatti, R., Gioia, M.G., Ravaglia, 607–617.
G., 2012. Serum thyroid-stimulating hormone as a predictor of cognitive Hogervorst, E., Ribeiro, H.M., Molyneux, A., Budge, M., Smith, A.D., 2002. Plasma
impairment in an elderly cohort. Gerontology 58 (1), 41–49. homocysteine levels, cerebrovascular risk factors, and cerebral white matter
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 185

changes (Leukoaraiosis) in patients with Alzheimer disease. Arch. Neurol. 59 Li, N.C., Lee, A., Whitmer, R.A., Kivipelto, M., Lawler, E., Kazis, L.E., Wolozin, B., 2010b.
(5), 787–793. Use of angiotensin receptor blockers and risk of dementia in a predominantly
Hooshmand, B., Solomon, A., Kåreholt, I., Leiviskä, J., Rusanen, M., Ahtiluoto, S., male population: prospective cohort analysis. BMJ 340, b5465.
Winblad, B., Laatikainen, T., Soininen, H., Kivipelto, M., 2010. Homocysteine and Li, G., Wang, L.Y., Shofer, J.B., Thompson, M.L., Peskind, E.R., McCormick, W., Bowen, J.
holotranscobalamin and the risk of Alzheimer’s disease: a longitudinal study. D., Crane, P.K., Larson, E.B., 2011. Temporal relationship between depression and
Neurology 75 (16), 1408–1414. dementia: findings from a large community-based 15-year follow-up study.
Hua, Y., Zhao, H., Kong, Y., Ye, M., 2011. Association between the MTHFR gene and Arch. Gen. Psychiatry 68 (9), 970–977.
Alzheimer's disease: a meta-analysis. Int. J. Neurosci. 121 (8), 462–471. Li, F.J., Shen, L., Ji, H.F., 2012. Dietary intakes of vitamin E, vitamin C, and beta-
Hurley, B.F., Hanson, E.D., Sheaff, A.K., 2011. Strength training as a countermeasure carotene and risk of Alzheimer's disease: a meta-analysis. J. Alzheimer's Dis. 31,
to aging muscle and chronic disease. Sports Med. 41 (4), 289–306. 253–258.
Isaac, M.G., Quinn, R., Tabet, N., 2008. Vitamin E for Alzheimer's disease and mild Lim, W.S., Gammack, J.K., Van Niekerk, J.K., Dangour, A., 2006. Omega 3 fatty acid for
cognitive impairment. Cochrane Database Syst. Rev. 3, CD002854. the prevention of dementia. Cochrane Database Syst. Rev. 1, CD005379.
Issa, A.M., Mojica, W.A., Morton, S.C., Traina, S., Newberry, S.J., Hilton, L.G., Garland, Lin, F.R., Metter, E.J., O'Brien, R.J., Resnick, S.M., Zonderman, A.B., Ferrucci, L., 2011.
R.H., Maclean, C.H., 2006. The efficacy of omega-3 fatty acids on cognitive Hearing loss and incident dementia. Arch. Neurol. 68 (2), 214–220.
function in aging and dementia: a systematic review. Dementia Geriatric Lin, M., Zhao, L., Fan, J., Lian, X.G., Ye, J.X., Wu, L., et al., 2012. Association between
Cognit. Disord. 21 (2), 88–96. HFE polymorphisms and susceptibility to Alzheimer's disease: a meta-analysis
James, B.D., Boyle, P.A., Buchman, A.S., Barnes, L.L., Bennett, D.A., 2011. Life space and of 22 studies including 4,365 cases and 8,652 controls. Mol. Biol. Rep. 39, 3089–
risk of Alzheimer’s disease, mild cognitive impairment, and cognitive decline in 3095.
old age. Am. J. Geriatr. Psychiatry 19 (11), 961–969. Lin, Y., Cheng, S., Xie, Z., Zhang, D., 2014. Association of rs6265 and rs2030324
Joas, E., Backman, K., Gustafson, D., Ostling, S., Waern, M., Guo, X., Skoog, I., 2012. polymorphisms in brain-derived neurotrophic factor gene with Alzheimer's
Blood pressure trajectories from midlife to late life in relation to dementia in disease: a meta-analysis. PLoS One [Electron. Resour.] 9, e94961.
women followed for 37 years. Hypertension 59 (4), 796–801. Lindsay, J., Anderson, L., 2003. Dementia and Alzheimer's Disease. Women's Health
Jorm, A.F., 2001. History of depression as a risk factor for dementia: an updated Surveillance Report. Public Health Agency of Canada. http://www.phac-aspc.gc.
review. Aust. New Zealand J. Psychiatry 35 (6), 776–781. ca/publicat/whsr-rssf/chap_19-eng.php.
Keage, H.A., Carare, R.O., Friedland, R.P., Ince, P.G., Love, S., Nicoll, J.A., Wharton, S.B., Lipton, R.B., Hirsch, J., Katz, M.J., Wang, C., Sanders, A.E., Verghese, J., Barzilai, N.,
Weller, R.O., Brayne, C., 2009. Population studies of sporadic cerebral amyloid Derby, C.A., 2010. Exceptional parental longevity associated with lower risk of
angiopathy and dementia: a systematic review. BMC Neurol. 9, 3. Alzheimer's disease and memory decline. J. Am. Geriatr. Soc. 58 (6), 1043–1049.
Kerola, T., Nieminen, T., Hartikainen, S., Sulkava, R., Vuolteenaho, O., Kettunen, R., Littbrand, H., Stenvall, M., Rosendahl, E., 2011. Applicability and effects of physical
2010. B-type natriuretic peptide as a predictor of declining cognitive function exercise on physical and cognitive functions and activities of daily living among
and dementia a cohort study of an elderly general population with a 5-year people with dementia: a systematic review. Am. J. Phys. Med. Rehab. 90 (6),
follow-up. Ann. Med. 42 (3), 207–215. 495–518.
Kheifets, L., Bowman, J.D., Checkoway, H., Feychting, M., Harrington, J.M., Kavet, R., Liu, Y., Zhang, J., Zeng, Z., 2013. Vascular endothelial growth factor polymorphisms
Marsh, G., Mezei, G., Renew, D.C., van Wijngaarden, E., 2009. Future needs of and risk of Alzheimer's disease: a meta-analysis. Gene 518, 296–302.
occupational epidemiology of extremely low frequency electric and magnetic Liu, M., Bian, C., Zhang, J., Wen, F., 2014. Apolipoprotein E gene polymorphism and
fields: review and recommendations. Occup. Environ. Med. 66 (2), 72–80. Alzheimer's disease in Chinese population: a meta-analysis. Sci. Rep. 4, 4383.
Khoury, M.J., Bertram, L., Boffetta, P., Butterworth, A.S., Chanock, S.J., Dolan, S.M., Llorca, J., Rodriguez-Rodriguez, E., Dierssen-Sotos, T., Delgado-Rodriguez, M.,
Fortier, I., Carcia-Closas, M., Gwinn, M., et al., 2009. Genome-wide association Berciano, J., Combarros, O., 2008. Meta-analysis of genetic variability in the
studies, field synopses, and the development of the knowledge base on genetic beta-amyloid production, aggregation and degradation metabolic pathways
variation and human disease. Am. J. Epidemiol. 170 (3), 269–279. and the risk of Alzheimer's disease. Acta Neurol. Scand. 117 (1), 1–14.
Kim, J.M., Stewart, R., Kim, S.W., Shin, I.S., Yang, S.J., Shin, H.Y., Yoon, J.S., 2008. Loef, M., Walach, H., 2012a. Copper and iron in Alzheimer's disease: a systematic
Changes in folate, vitamin B12 and homocysteine associated with incident review and its dietary implications. Br. J. Nutr. 107 (1), 7–19.
dementia. J. Neurol. Neurosurg. Psychiatry 79 (8), 864–868. Loef, M., Walach, H., 2012b. Fruit, vegetables and prevention of cognitive decline or
Kimm, H., Lee, P.H., Shin, Y.J., Park, K.S., Jo, J., Lee, Y., Kang, H.C., Jee, S.H., 2011. Midlife dementia: a systematic review of cohort studies. J. Nutr. Health Aging 16 (7),
and late-life vascular risk factors and dementia in Korean men and women. 626–630.
Arch. Gerontol. Geriatr. 52 (3), e117–e122. Loef, M., Mendoza, L.F., Walach, H., 2011a. Lead (Pb) and the risk of Alzheimer’s
Kloppenborg, R.P., van den Berg, E., Kappelle, L.J., Biessels, G.J., 2008. Diabetes and disease or cognitive decline: a systematic review. Tox. Rev. 30 (4), 103–114.
other vascular risk factors for dementia: which factor matters most? A Loef, M., Schrauzer, G.N., Walach, H., 2011b. Selenium and Alzheimer's disease: a
systematic review. Eur. J. Pharmacol. 585 (1), 97–108. systematic review. J. Alzheimer's Dis. 26 (1), 81–104.
Kohler, S., van Boxtel, M., Jolles, J., Verhey, F., 2011. Depressive symptoms and risk for Loef, M., von Stillfried, N., Walach, H., 2012. Zinc diet and Alzheimer's disease: a
dementia: a 9-year follow-up of the Maastricht Aging Study. Am. J. Geriatr. systematic review. Nutr. Neurosci. 15 (5), 2–12.
Psychiatry 19 (10), 902–905. Lopez, L.B., Kritz-Silverstein, D., Barrett Connor, E., 2011. High dietary and plasma
Koseoglu, E., Karaman, Y., 2007. Relations between homocysteine, folate and levels of the omega-3 fatty acid docosahexaenoic acid are associated with
vitamin B12 in vascular dementia and in Alzheimer disease. Clin. Biochem. 40 decreased dementia risk: the Rancho Bernardo study. J. Nutr. Health Aging 15
(12), 859–863. (1), 25–31.
Koyama, A., O'Brien, J., Weuve, J., Blacker, D., Metti, A.L., Yaffe, K., 2013. The role of Louis, E.D., Tang, M.X., Schupf, N., 2010. Mild parkinsonian signs are associated with
peripheral inflammatory markers in dementia and Alzheimer's disease: a meta- increased risk of dementia in a prospective, population-based study of elders.
analysis. J. Gerontol. Ser. A-Biol. Sci. Med. Sci. 68, 433–440. Mov. Disord. 25 (2), 172–178.
Kristal, A.R., Peters, U., Potter, J.D., 2005. Is it time to abandon the food frequency Lourida, I., Soni, M., Thompson-Coon, J., Purandare, N., Lang, I.A., Ukoumunne, O.C.,
questionnaire? Cancer Epidemiol. Biomarkers Prevention 14 (12), 2826–2828. et al., 2013. Mediterranean diet, cognitive function, and dementia: a systematic
Kuo, H.K., Sorond, F.A., Chen, J.H., Hashmi, A., Milberg, W.P., Lipsitz, L.A., 2005a. The review. Epidemiology 24, 479–489.
role of homocysteine in multisystem age-related problems: a systematic Lu, F.P., Lin, K.P., Kuo, H.K., 2009. Diabetes and the risk of multi-system aging
review. J. Gerontol. Ser. A-Biol. Sci. Med. Sci. 60 (9), 1190–1201. phenotypes: a systematic review and meta-analysis. PLoS ONE [Electron.
Kuo, H.K., Yen, C.J., Chang, C.H., Kuo, C.K., Chen, J.H., Sorond, F., 2005b. Relation of C- Resour.] 4 (1), e4144.
reactive protein to stroke, cognitive disorders, and depression in the general Lu, Y., et al., 2014. Quantitative assessment of CYP2D6 polymorphisms and risk of
population: systematic review and meta-analysis. Lancet Neurol. 4 (6), 371– Alzheimer's disease: a meta-analysis. J. Neurol. Sci. 343, 15–22.
380. Luchsinger, J.A., Tang, M.X., Shea, S., Miller, J., Green, R., Mayeux, R., 2004. Plasma
LaRoia, H., Louis, E.D., 2011. Association between essential tremor and other homocysteine levels and risk of Alzheimer disease. Neurology 62 (11), 1972–
neurodegenerative diseases: what is the epidemiological evidence? 1976.
Neuroepidemiology 37 (1), 1–10. Luo, J., Li, S., Qin, X., Song, L., Peng, Q., Chen, S., et al., 2014. Meta-analysis of the
LeBlanc, E.S., Janowsky, J., Chan, B.K., Nelson, H.D., 2001. Hormone replacement association between CR1 polymorphisms and risk of late-onset Alzheimer's
therapy and cognition: systematic review and meta-analysis. JAMA 285 (11), disease. Neurosci. Lett. 578, 165–170.
1489–1499. Macedo, A.F., Taylor, F.C., Casas, J.P., Adler, A., Prieto-Merino, D., Ebrahim, S., 2014.
Lee, Y., Back, J.H., Kim, J., Kim, S.H., Na, D.L., Cheong, H.K., Hong, C.H., Kim, Y.G., 2010. Unintended effects of statins from observational studies in the general
Systematic review of health behavioral risks and cognitive health in older population: systematic review and meta-analysis. BMC Med. 12, 51.
adults. Int. Psychogeriatr. 22 (2), 174–187. Malouf, R., Areosa Sastre, A., 2003. Vitamin B12 for cognition. Cochrane Database
Leinonen, V., Koivisto, A.M., Savolainen, S., Rummukainen, J., Tamminen, J.N., Syst. Rev. 3, CD004326.
Tillgren, T., Vainikka, S., et al., 2010. Amyloid and (proteins in cortical brain Malouf, R., Grimley, E.J., 2008. Folic acid with or without vitamin B12 for the
biopsy and Alzheimer's disease. Ann. Neurol. 68 (4), 446–453. prevention and treatment of healthy elderly and demented people. Cochrane
Lenoir, H., Dufouil, C., Auriacombe, S., Lacombe, J.M., Dartigues, J.F., Ritchie, K., Database Syst. Rev. 4, CD004514.
Tzourio, C., 2011. Depression history, depressive symptoms, and incident Malouf, M., Grimley, E.J., Areosa, S.A., 2003. Folic acid with or without vitamin B12
dementia: the 3C Study. J. Alzheimer's Dis. 26 (1), 27–38. for cognition and dementia. Cochrane Database Syst. Rev. 4, CD004514.
Li, G., Shofer, J.B., Rhew, I.C., Kukull, W.A., Peskind, E.R., McCormick, W., Bowen, J.D., Marengoni, A., Qiu, C., Winblad, B., Fratiglioni, L., 2011. Atrial fibrillation, stroke and
et al., 2010a. Age-varying association between statin use and incident dementia in the very old: a population-based study. Neurobiol. Aging 32 (7),
Alzheimer's disease. J. Am. Geriatr. Soc. 58 (7), 1311–1317. 1336–1337.
186 M. Hersi et al. / NeuroToxicology 61 (2017) 143–187

McGuinness, B., Todd, S., Passmore, P., Bullock, R., 2009. Blood pressure lowering in Pham, D.Q., Plakogiannis, R., 2005. Vitamin E supplementation in Alzheimer's
patients without prior cerebrovascular disease for prevention of cognitive disease, Parkinson's disease, tardive dyskinesia, and cataract: part 2. Ann.
impairment and dementia. Cochrane Database Syst. Rev. 4, CD004034. Pharmacother. 39 (12), 2065–2072.
Meng, X., D'Arcy, C., 2012. Education and dementia in the context of the cognitive Pitner, J.K., 2005. Obesity in the elderly. Consult. Pharm. 20 (6), 498–513.
reserve hypothesis: a systematic review with meta-analyses and qualitative Podewils, L.J., 2003. Physical Activity and Dementia Risk: a Systematic Review.
analyses. PLoS ONE [Electron. Resour.] 7 (6), e38268. (Diss., Johns Hopkins University).
Mielke, M.M., Zandi, P.P., Shao, H., Waern, M., Ostling, S., Guo, X., Bjorkelund, C., Power, M.C., Weuve, J., Gagne, J.J., McQueen, M.B., Viswanathan, A., Blacker, D., 2011.
Lissner, L., Skoog, I., Gustafson, D.R., 2010. The 32-year relationship between The association between blood pressure and incident alzheimer disease: a
cholesterol and dementia from midlife to late life. Neurology 75 (21), 1888– systematic review and meta-analysis. Epidemiology 22 (5), 646–659.
1895. Profenno, L.A., Porsteinsson, A.P., Faraone, S.V., 2010. Meta-analysis of Alzheimer's
Miller, L.J., Chacko, R., 2004. The role of cholesterol and statins in Alzheimer's disease risk with obesity, diabetes, and related disorders. Biol. Psychiatry 67 (6),
disease. Ann. Pharmacother. 38 (1), 91–98. 505–512.
Miller, J.W., Green, R., Mungas, D.M., Reed, B.R., Jagust, W.J., 2002. Homocysteine, Psaltopoulou, T., Sergentanis, T.N., Panagiotakos, D.B., Sergentanis, I.N., Kosti, R.,
vitamin B6, and vascular disease in AD patients. Neurology 58 (10), 1471–1475. Scarmeas, N., 2013. Mediterranean diet, stroke, cognitive impairment, and
Mitchell, A.J., Shiri-Feshki, M., 2008. Temporal trends in the long term risk of depression: a meta-analysis. Ann. Neurol. 74, 580–591.
progression of mild cognitive impairment: a pooled analysis Journal of Public Health Agency of Canada, 2010. Chapter 3: The Health and Well-being of
Neurology. Neurosurg. Psychiatry 79 (12), 1386–1391. Canadian Seniors. The Chief Public Health Officer's Report on The State of Public
Mitchell, A.J., Shiri-Feshki, M., 2009. Rate of progression of mild cognitive Health in Canada 2010. . Accessed 28 Oct. 2013 http://www.phac-aspc.gc.ca/
impairment to dementia–meta-analysis of 41 robust inception cohort studies. cphorsphc-respcacsp/2010/fr-rc/cphorsphc-respcacsp-06-eng.php.
Acta Psychiatr. Scand. 119 (4), 252–265. Purnell, C., Callahan, C.M., Hendrie, H.C., 2009. Cardiovascular risk factors and
Mo, C., Peng, Q., Sui, J., Wang, J., Deng, Y., Xie, L., et al., 2014. Lack of association incident alzheimer’s disease: a systematic review of the literature. Alzheimer
between cathepsin D C224T polymorphism and Alzheimer's disease risk: an Dis. Assoc. Disord. 23 (1), 1–10.
update meta-analysis. BMC Neurol. 14, 13. Qin, X., Peng, Q., Zeng, Z., Chen, Z., Lin, L., Deng, Y., et al., 2012. Interleukin-1A 889C/
Montlahuc, C., Soumare, A., Dufouil, C., Berr, C., Dartigues, J.F., Poncet, M., Tzourio, C., T polymorphism and risk of Alzheimer's disease: a meta-analysis based on 32
Alperovitch, A., 2011. Self-rated health and risk of incident dementia: a case-control studies. J. Neurol. 259, 1519–1529.
community-based elderly cohort, the 3C study. Neurology 77 (15), 1457–1464. Qiu, C., Xu, W., Winblad, B., Fratiglioni, L., 2010. Vascular risk profiles for dementia
Mortimer, J.A., van Duijn, C.M., Chandra, V., Fratiglioni, L., Graves, A.B., Heyman, A., and Alzheimer's disease in very old people: a population-based longitudinal
Jorm, A.F., et al., 1991. Head trauma as a risk factor for Alzheimer's disease: a study. J. Alzheimer's Dis. 20 (1), 293–300.
collaborative re-analysis of case-control studies. EURODEM Risk Factors Quadri, P., Fragiacomo, C., Pezzati, R., Zanda, E., Forloni, G., Tettamanti, M., Lucca, U.,
Research Group. Int. J. Epidemiol. 20 (Suppl.2), S28–S35. 2004. Homocysteine, folate, and vitamin B-12 in mild cognitive impairment,
Muller, M., Schupf, N., Manly, J.J., Mayeux, R., Luchsinger, J.A., 2010. Sex hormone Alzheimer disease, and vascular dementia. Am. J. Clin. Nutr. 80 (1), 114–122.
binding globulin and incident Alzheimer's disease in elderly men and women. Rainero, I., Bo, M., Ferrero, M., Valfrè, W., Vaula, G., Pinessi, L., 2004. Association
Neurobiol. Aging 31 (10), 1758–1765. between the interleukin-1a gene and Alzheimer’s disease: a meta-analysis.
Nägga, K., Rajani, R., Mårdh, E., Borch, K., Mårdh, S., Marcusson, J., 2003. Cobalamin, Neurobiol. Aging 25 (10), 1293–1298.
folate, methylmalonic acid, homocysteine, and gastritis markers in dementia. Raman, G., Tatsioni, A., Chung, M., Rosenberg, I.H., Lau, J., Lichtenstein, A.H., Balk, E.
Dement. Geriatr. Cogn. Disord. 16 (4), 269–275. M., 2007. Heterogeneity and lack of good quality studies limit association
National Advisory Council on Aging (Canada), 2004. The NACA Position on between folate, vitamins B-6 ad B-12, and cognitive function. J. Nutr. 137 (7),
Alzheimer Disease and Related Dementias. . http://books2. scholarsportal.info/ 1789–1794.
viewdoc.html?id=/ebooks/ebooks1/gibson_chrc/2010-08-06/1/10089389. Raschetti, R., Albanese, E., Vanacore, N., Maggini, M., 2007. Cholinesterase inhibitors
Ning, M., Yang, Y., Zhang, Z., Chen, Z., Zhao, T., Zhang, D., Zhou, D., Xu, J., Liu, Z., Wang, in mild cognitive impairment: a systematic review of randomised trials. PLoS
Y., Liu, Y., Zhao, X., Li, W., Li, S., He, L., 2010. Amyloid-b-Related genes SORL1 and Med. Public Lib. Sci. 4 (11), e338.
ACE are genetically associated with risk for late-onset alzheimer disease in the Ravaglia, G., Forti, P., Maioli, F., Martelli, M., Servadei, L., Brunetti, N., Porcellini, E.,
chinese population. Alzheimer Dis. Relat. Disord. 24 (4), 390–396. Licastro, F., 2005. Homocysteine and folate as risk factors for dementia and
Norton, M.C., Dew, J., Smith, H., Fauth, E., Piercy, K.W., Breitner, J.C., Tschanz, J., Alzheimer disease. Am. J. Clin. Nutr. 82 (3), 636–643.
Wengreen, H., Welsh-Bohmer, K., Cache County Investigators, 2012. Lifestyle Reitz, C., Tang, M.X., Schupf, N., Manly, J.J., Mayeux, R., Luchsinger, J.A., 2010a.
behavior pattern is associated with different levels of risk for incident dementia Association of higher levels of high-density lipoprotein cholesterol in elderly
and Alzheimer's disease: the Cache County study. J. Am. Geriatr. Soc. 60 (3), individuals and lower risk of late-onset Alzheimer disease. Arch. Neurol. 67 (12),
405–412. 1491–1497.
Ntais, C., Polycarpou, A., Ioannidis, J.P., 2004. Meta-analysis of the association of the Reitz, C., Tang, M.X., Schupf, N., Manly, J.J., Mayeux, R., Luchsinger, J.A., 2010b. b: a
cathepsin D Ala224Val gene polymorphism with the risk of Alzheimer's disease: summary risk score for the prediction of Alzheimer disease in elderly persons.
a HuGE gene-disease association review. Am. J. Epidemiol. 159 (6), 527–536. Arch. Neurol. 67 (7), 835–841.
O'Brien, J., Jackson, J.W., Grodstein, F., Blacker, D., Weuve, J., 2014. Postmenopausal Reitz, C., Cheng, R., Rogaeva, E., Lee, J.H., Tokuhiro, S., Zou, F., Bettens, K., et al., 2011.
hormone therapy is not associated with risk of all-cause dementia and Meta-analysis of the association between variants in SORL1 and Alzheimer
Alzheimer's disease. Epidemiol. Rev. 36, 83–103. disease. Arch. Neurol. 68 (1), 99–106.
O'Leary, F., Allman-Farinelli, M., Samman, S., 2012. Vitamin B12 status: cognitive Religa, D., Styczynska, M., Peplonska, B., Gabryelewicz, T., Pfeffer, A., Chodakowska,
decline and dementia: a systematic review of prospective cohort studies. Br. J. M., Luczywek, E., Wasiak, B., Stepien, K., Golebiowski, M., Winblad, B.,
Nutr. 108, 1948–1961. Barcikowska, M., et al., 2003. Homocysteine, apolopoproteine E and
Ohara, T., Doi, Y., Ninomiya, T., Hirakawa, Y., Hata, J., Iwaki, T., Kanba, S., Kiyohara, Y., methylenetetrahydrofolate reductase in Alzheimer’s disease and mild cognitive
2011. Glucose tolerance status and risk of dementia in the community: the impairment. Dement. Geriatr. Cogn. Disord. 16 (2), 4–70.
Hisayama study. Neurology 77 (12), 1126–1134. Reutens, S., Sachdev, P., 2002. Homocysteine in neuropsychiatric disorders of the
Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006. Depression and elderly. Int. J. Geriatr. Psychiatry 17 (9), 859–864.
risk for Alzheimer disease: systematic review, meta-analysis, and Rocca, W.A., van Duijn, C.M., Clayton, D., Chandra, V., Fratiglioni, L., Graves, A.B.,
metaregression analysis. Arch. Gen. Psychiatry 63 (5), 530–538. Heyman, A., et al., 1991. Maternal age and Alzheimer's disease: a collaborative
Pan, Y., Li, Y., Shen, H., 2014. Meta-analysis of the association between re-analysis of case-control studies: EURODEM Risk Factors Research Group. Int.
polymorphisms of estrogen receptor alpha genes rs9340799 and rs2234693 and J. Epidemiol. 20 (Suppl. 2), S21–7.
Alzheimer's disease: evidence from 23 articles. Am. J. Alzheimer's Dis. Other Rodriguez-Manotas, M., Amorin-Diaz, M., Canizares-Hernandez, F., Ruiz-Espejo, F.,
Dementias 29, 704–711. Martinez-Vidal, S., Gonzalez-Sarmiento, R., Martinez-Hernandez, P., Cabezas-
Paradise, M., Cooper, C., Livingston, G., 2009. Systematic review of the effect of Herrera, J., 2007. Association study and meta-analysis of Alzheimer's disease
education on survival in Alzheimer's disease. Int. Psychogeriatr. 21 (1), 25–32. risk and presenilin-1 intronic polymorphism. Brain Res. 1170, 119–128.
Parihar, M.S., Hemnani, T., 2004. Alzheimer's disease pathogenesis and therapeutic Roe, C.M., Fitzpatrick, A.L., Xiong, C., Sieh, W., Kuller, L., Miller, J.P., Williams, M.M.,
interventions. J. Clin. Neurosci. 11 (5), 456–467. Kopan, R., Behrens, M.I., Morris, J.C., 2010. Cancer linked to Alzheimer disease
Patatanian, E., Gales, M.A., 2005. The future of statins: alzheimer's disease? Consult. but not vascular dementia. Neurology 74 (2), 106–112.
Pharm. 20 (8), 663–673. Rolland, Y., Abellan van Kan, G., Vellas, B., 2008. Physical activity and Alzheimer's
Patterson, C., Feightner, J., Garcia, A., MacKnight, C., 2007. General risk factors for disease: from prevention to therapeutic perspectives. J. Am. Med. Dir. Assoc. 9
dementia: a systematic evidence review. Alzheimer's Dementia 3 (4), 341–347. (6), 390–405.
Penrose, F.K., 2005. Can exercise affect cognitive functioning in Alzheimer ‘s Roman, B., Carta, L., Martinez-Gonzalez, M.A., Serra-Majem, L., 2008. Effectiveness
disease? A review of the literature. Activities Adaptation Aging 29 (4), 15–40. of the Mediterranean diet in the elderly. Clin. Interv. Aging 3 (1), 97–109.
Peters, R., Burch, L., Warner, J., Beckett, N., Poulter, R., Bulpitt, C., 2008a. Ronnemaa, E., Zethelius, B., Lannfelt, L., Kilander, L., 2011. Vascular risk factors and
Haemoglobin, anaemia, dementia and cognitive decline in the elderly, a dementia: 40-year follow-up of a population-based cohort. Dementia Geriatric
systematic review. BMC Geriatr. 8, 18. Cognit. Disord. 31 (6), 460–466.
Peters, R., Peters, J., Warner, J., Beckett, N., Bulpitt, C., 2008b. Alcohol, dementia and Rusanen, M., Kivipelto, M., Quesenberry, C.P.Jr., Zhou, J., Whitmer, R.A., 2011. Heavy
cognitive decline in the elderly: a systematic review. Age Ageing 37 (5), 505– smoking in midlife and long-term risk of Alzheimer disease and vascular
512. dementia. Arch. Intern. Med. 171 (4), 333–339.
Peters, R., Poulter, R., Warner, J., Beckett, N., Burch, L., Bulpitt, C., 2008c. Smoking, Saczynski, J.S., Beiser, A., Seshadri, S., Auerbach, S., Wolf, P.A., Au, R., 2010. Depressive
dementia, and cognitive decline in the elderly, a systematic review. BMC Geriatr. symptoms and risk of dementia: the Framingham Heart Study. Neurology 75
8, 36. (1), 35–41.
M. Hersi et al. / NeuroToxicology 61 (2017) 143–187 187

Santibanez, M., Bolumar, F., Garcia, A.M., 2007. Occupational risk factors in Wang, J., Tan, L., Wang, H.F., Tan, C.C., Meng, X.F., Wang, C., et al., 2015a. Anti-
Alzheimer's disease: a review assessing the quality of published inflammatory drugs and risk of Alzheimer's disease: an updated systematic
epidemiological studies. Occup. Environ. Med. 64 (11), 723–732. review and meta-analysis. J. Alzheimer's Dis. 44, 385–396.
Santos, C., Costa, J., Santos, J., Vaz-Carneiro, A., Lunet, N., 2010. Caffeine intake and Wang, X.B., Cui, N.H., Gao, J.J., Qiu, X.P., Yang, N., Zheng, F., 2015b. Angiotensin-
dementia: systematic review and meta-analysis. J. Alzheimer's Dis. 20 (Suppl 1), converting enzyme gene polymorphisms and risk for sporadic Alzheimer's
S187–204. disease: a meta-analysis. J. Neural Transm. 122, 211–224.
Schrijvers, E.M., Witteman, J.C., Sijbrands, E.J., Hofman, A., Koudstaal, P.J., Breteler, Wang, T., 2014. Meta-analysis of PvuII: XbaI variants in ESR1 gene and the risk of
M.M., 2010. Insulin metabolism and the risk of Alzheimer disease: the Alzheimer's disease: the regional European difference. Neurosci. Lett. 574, 41–
Rotterdam Study. Neurology 75 (22), 1982–1987. 46.
Schrijvers, E.M., Koudstaal, P.J., Hofman, A., Breteler, M.M., et al., 2011. Plasma Wang, T., 2015. TNF-alpha G308A polymorphism and the susceptibility to
clusterin and the risk of Alzheimer disease. J. Am. Med. Assoc. 305 (13), 1322– Alzheimer's disease: an updated meta-analysis. Arch. Med. Res. 46, 24–30 (e1).
1326. Weih, M., Wiltfang, J., Kornhuber, J., 2007. Non-pharmacologic prevention of
Seshadri, S., Beiser, A., Selhub, J., Jacques, P.F., Rosenberg, I.H., D'Agostino, R.B., Alzheimer's disease: nutritional and life-style risk factors. J. Neural Transm. 114
Wilson, P.W., Wolf, P.A., 2002. Plasma homocysteine as a risk factor for dementia (9), 1187–1197.
and Alzheimer's disease. New Engl. J. Med. 346 (7), 476–483. Weih, M., Degirmenci, Ü., Kreil, S., Kornhuber, J., 2010. Physical activity and
Shadlen, M.F., Larson, E.B., Yukawa, M., 2000. The epidemiology of Alzheimer's Alzheimer’s disease: a meta-analysis of cohort studies. GeroPsych 23 (1), 17–20.
disease and vascular dementia in Japanese and African-American populations: Weyerer, S., Schaufele, M., Wiese, B., Maier, W., Tebarth, F., van den Bussche, H.,
the search for etiological clues. Neurobiol. Aging 21 (2), 171–181. Pentzek, M., et al., 2011. Current alcohol consumption and its relationship to
Shah, K., Qureshi, S.U., Johnson, M., Parikh, N., Schulz, P.E., Kunik, M.E., 2009. Does incident dementia: results from a 3-year follow-up study among primary care
use of antihypertensive drugs affect the incidence or progression of dementia? attenders aged 75 years and older. Age Ageing 40 (4), 456–463.
A systematic review. Am. J. Geriatr. Pharmacother. 7 (5), 250–261. Williams, J.W., Plassman, B.L., Burke, J., Benjamin, S., 2010. Preventing Alzheimer’s
Shah, R.C., Buchman, A.S., Wilson, R.S., Leurgans, S.E., Bennett, D.A., 2011. Disease and Cognitive Decline. Evidence Report/Technology Assessment N0.
Hemoglobin level in older persons and incident Alzheimer disease: prospective 193. (Prepared by the Duke Evidence-based Practice Center Under Contract No.
cohort analysis. Neurology 77 (3), 219–226. HHSA 290–2007-10066-I.) AHRQ Publication No. 10-E005. Agency for
Shah, N.S., Vidal, J.S., Masaki, K., Petrovitch, H., Ross, G.W., Tilley, C., DeMattos, R.B., Healthcare Research and Quality, Rockville, MD.
Tracy, R.P., White, L.R., Launer, L.J., 2012. Midlife blood pressure, plasma Wong, W.B., Lin, V.W., Boudreau, D., Devine, E.B., 2013. Statins in the prevention of
b-Amyloid and the risk for alzheimer’s disease: the Honolulu asia aging study. dementia and Alzheimer's disease: a meta-analysis of observational studies and
Hypertension 59 (4), 780–786. an assessment of confounding. Pharmacoepidemiol. Drug Saf. 22, 345–358.
Shea, B.J., Hamel, C., Wells, G.A., Bouter, L.M., Kristjansson, E., Grimshaw, J., Henry, D. Woods, B., Aguirre, E., Spector, A.E., Orrell, M., 2012. Cognitive stimulation to
A., Boers, M., 2009. AMSTAR is a reliable and valid measurement tool to assess improve cognitive functioning in people with dementia. Cochrane Database
the methodological quality of systematic reviews. J. Clin. Epidemiol. 62, 1013– Syst. Rev. 2, CD005562.
1020. Wu, W., Jiang, H., Wang, M., Zhang, D., 2013. Meta-analysis of the association
Singh, B., Parsaik, A.K., Mielke, M.M., Erwin, P.J., Knopman, D.S., Petersen, R.C., et al., between urokinase-plasminogen activator gene rs2227564 polymorphism and
2014. Association of mediterranean diet with mild cognitive impairment and Alzheimer's disease. Am. J. Alzheimer's Dis. Other Dementias 28, 517–523.
Alzheimer's disease: a systematic review ad meta-analysis. J. Alzheimer's Dis. Wu, S., Ding, Y., Wu, F., Li, R., Hou, J., Mao, P., 2015. Omega-3 fatty acids intake and
39, 271–282. risks of dementia and Alzheimer's disease: a meta-analysis. Neurosci. Biobehav.
Sofi, F., Cesari, F., Abbate, R., Gensini, G.F., Casini, A., 2008. Adherence to Rev. 48, 1–9.
Mediterranean diet and health status: meta-analysis. BMJ 337, a1344. Xin, X.Y., Ding, J.Q., Chen, S.D., 2010. Apolipoprotein E promoter polymorphisms and
Sofi, F., Abbate, R., Gensini, G.F., Casini, A., 2010. Accruing evidence on benefits of risk of Alzheimer's disease: evidence from meta-analysis. J. Alzheimer's Dis. 19
adherence to the Mediterranean diet on health: an updated systematic review (4), 1283–1294.
and meta-analysis. Am. J. Clin. Nutr. 92 (5), 1189–1196. Xu, W.L., Atti, A.R., Gatz, M., Pedersen, N.L., Johansson, B., Fratiglioni, L., 2011. Midlife
Song, X., Mitnitski, A., Rockwood, K., 2011. Nontraditional risk factors predict overweight and obesity increase late-life dementia risk: a population-based
Alzheimer disease. Neurology 77 (3), 227–234. twin study. Neurology 76 (18), 1568–1574.
Song, Y., Nie, H., Xu, Y., Zhang, L., Wu, Y., 2013. Association of statin use with risk of Yaffe, K., Sawaya, G., Lieberburg, I., Grady, D., 1998. Estrogen therapy in
dementia: a meta-analysis of prospective cohort studies. Geriatrics Gerontol. postmenopausal women: effects on cognitive function and dementia. JAMA 279
Int. 13, 817–824. (9), 688–695.
Standridge, J.B., 2004. Pharmacotherapeutic approaches to the prevention of Yang, Y.H., Roe, C.M., Morris, J.C., 2011. Relationship between late-life hypertension,
Alzheimer's disease. Am. J. Geriatr. Pharmacother. 2 (2), 119–132. blood pressure, and Alzheimer's disease. Am. J. Alzheimer's Dis. Other
Stern, C., Konno, R., 2009. Physical leisure activities and their role in preventing Dementias 26 (6), 457–462.
dementia: a systematic review. Int. J. Evide. Based Healthcare 7 (4), 270–282. Yang, Z.D., Yu, J., Zhang, Q., 2013. Effects of raloxifene on cognition, mental health,
Stern, C., Munn, Z., 2010. Cognitive leisure activities and their role in preventing sleep and sexual function in menopausal women: a systematic review of
dementia: a systematic review. Inte. J. Evid. Based Healthcare 8 (1), 2–17. randomized controlled trials. Maturitas 75, 341–348.
Stern, Y., 2006. Cognitive reserve and Alzheimer disease. Alzheimer Dis. Assoc. Zhang, T., Jia, Y., 2014. Meta-analysis of Ubiquilin1 gene polymorphism and
Disord. 20 (3 (Sulppl. 2)), S69–S74. Alzheimer's disease risk. Med. Sci. Monit. 20, 2250–2255.
Storey, S.G., Suryadevara, V., Aronow, W.S., Ahn, C., 2003. Association of plasma Zhang, M.Y., Miao, L., Li, Y.S., Hu, G.Y., 2010. Meta-analysis of the
homocysteine in elderly persons with atherosclerotic vascular disease and methylenetetrahydrofolate reductase C677T polymorphism and susceptibility
dementia, atherosclerotic vascular disease without dementia, dementia to Alzheimer's disease. Neurosci. Res. 68 (2), 142–150.
without atherosclerotic vascular disease, and no dementia or atherosclerotic Zhang, Y., Zhang, J., Tian, C., Xiao, Y., Li, X., He, C., Huang, J., Fan, H., 2011. The 1082G/
vascular disease. J. Gerontol. 58 (12), 1135–1136. A polymorphism in IL-10 gene is associated with risk of Alzheimer's disease: a
Sundelöf, J., Sundström, J., Hansson, O., Eriksdotter-Jönhagen, M., Giedraitis, V., meta-analysis. J. Neurol. Sci. 303 (1-2), 133–138.
Larsson, A., Degerman-Gunnarsson, M., Ingelsson, M., Minthon, L., Blennow, K., Zhong, G., Wang, Y., Zhang, Y., Guo, J.J., Zhao, Y., 2015. Smoking is associated with an
Kilander, L., Basun, H., Lannfelt, L., 2012. Cystatin C levels are positively increased risk of dementia: a meta-analysis of prospective cohort studies with
correlated with Ab42 and t levels in cerebrospinal fluid in persons with investigation of potential effect modifiers. PLoS ONE [Electron. Resour.] 10,
Alzheimer’s disease, mild cognitive impairment, and healthy controls. J. e0118333.
Alzheimer’s Dis. 21 (2), 471–478. Zhou, B., Teramukai, S., Fukushima, M., 2007. Prevention and treatment of dementia
Szekely, C.A., Thorne, J.E., Zandi, P.P., Ek, M., Messias, E., Breitner, J.C., Goodman, S.N., or Alzheimer's disease by statins: a meta-analysis. Dementia Geriatric Cognit.
2004. Nonsteroidal anti-inflammatory drugs for the prevention of Alzheimer's Disord. 23 (3), 194–201.
disease: a systematic review. Neuroepidemiology 23 (4), 159–169. Zhou, R., Deng, J., Zhang, M., Zhou, H.D., Wang, Y.J., 2011. Association between bone
Verdelho, A., Madureira, S., Moleiro, C., Ferro, J.M., Santos, C.O., Erkinjuntti, T., mineral density and the risk of Alzheimer's disease. J. Alzheimer's Dis. 24 (1),
Pantoni, L., et al., 2010. White matter changes and diabetes predict cognitive 101–108.
decline in the elderly: the LADIS study. Neurology 75 (2), 160–167. Zhou, Q., Zhao, F., Lv, Z.P., Zheng, C.G., Zheng, W.D., Sun, L., et al., 2014. Association
Vergara, X., Kheifets, L., Greenland, S., Oksuzyan, S., Cho, Y.S., Mezei, G., 2013. between APOC1 polymorphism and Alzheimer's disease: a case-control study
Occupational exposure to extremely low-frequency magnetic fields and and meta-analysis. PLoS ONE [Electron. Resour.] 9, e87017.
neurodegenerative disease: a meta-analysis. J. Occup. Environ. Med. 55, 135–146. Zuo, C., Zuo, Z., 2010. Spine Surgery under general anesthesia may not increase the
Wang, Y., Bi, L., Wang, H., Li, Y., Di, Q., Xu, W., et al., 2012. NEDD9 rs760678 risk of Alzheimer's disease. Dementia Geriatric Cognit. Disord. 29 (3), 233–239.
polymorphism and the risk of Alzheimer's disease: a meta-analysis. Neurosci. de Craen, A.J., Gussekloo, J., Vrijsen, B., Westendorp, R.G., 2005. Meta-analysis of
Lett. 527, 121–125. nonsteroidal antiinflammatory drug use and risk of dementia. Am. J. Epidemiol.
Wang, X.F., Cao, Y.W., Feng, Z.Z., Fu, D., Ma, Y.S., Zhang, F., et al., 2013a. Quantitative 161 (2), 114–120.
assessment of the effect of ABCA1 gene polymorphism on the risk of de Jong, F.J., Schrijvers, E.M., Ikram, M.K., Koudstaal, P.J., de Jong, P.T., Hofman, A.,
Alzheimer's disease. Mol. Biol. Rep. 40, 779–785. Vingerling, J.R., Breteler, M.M., 2011. Retinal vascular caliber and risk of
Wang, Y., Xu, S., Liu, Z., Lai, C., Xie, Z., Zhao, C., et al., 2013b. Meta-analysis on the dementia: the Rotterdam study. Neurology 76 (9), 816–821.
association between the TF gene rs1049296 and AD. Can. J. Neurol. Sci. 40 (5), van Duijn, C.M., Clayton, D., Chandra, V., Fratiglioni, L., Graves, A.B., Heyman, A.,
691–697. Jorm, A.F., et al., 1991. Familial aggregation of Alzheimer's disease and related
Wang, C., Yu, J.T., Wang, H.F., Jiang, T., Tan, C.C., Meng, X.F., et al., 2014. Meta-analysis disorders: a collaborative re-analysis of case-control studies. Int. J. Epidemiol.
of peripheral blood apolipoprotein E levels in Alzheimer's disease. PLoS ONE 20 (Suppl. 2), S13–S20.
[Electron. Resour.] 9, e89041.

You might also like