Next Article in Journal / Special Issue
Harmonizing Outcomes for Genomic Medicine: Comparison of eMERGE Outcomes to ClinGen Outcome/Intervention Pairs
Previous Article in Journal
Tackling the Consumption of High Sugar Products among Children and Adolescents in the Pacific Islands: Implications for Future Research
Previous Article in Special Issue
Developing a Case-Based Blended Learning Ecosystem to Optimize Precision Medicine: Reducing Overdiagnosis and Overtreatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Precision Medicine for Alzheimer’s Disease Prevention

Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
*
Author to whom correspondence should be addressed.
Healthcare 2018, 6(3), 82; https://doi.org/10.3390/healthcare6030082
Submission received: 30 May 2018 / Revised: 8 July 2018 / Accepted: 10 July 2018 / Published: 13 July 2018
(This article belongs to the Special Issue Precision Public Health and Genomic Medicine)

Abstract

:
Precision medicine is an approach to medical treatment and prevention that takes into account individual variability in genes, environment, and lifestyle and allows for personalization that is based on factors that may affect the response to treatment. Several genetic and epigenetic risk factors have been shown to increase susceptibility to late-onset Alzheimer’s disease (AD). As such, it may be beneficial to integrate genetic risk factors into the AD prevention approach, which in the past has primarily been focused on universal risk-reduction strategies for the general population rather than individualized interventions in a targeted fashion. This review discusses examples of a “one-size-fits-all” versus clinical precision medicine AD prevention strategy, in which the precision medicine approach considers two genes that can be commercially sequenced for polymorphisms associated with AD, apolipoprotein E (APOE), and methylenetetrahydrofolate reductase (MTHFR). Comparing these two distinct approaches provides support for a clinical precision medicine prevention strategy, which may ultimately lead to more favorable patient outcomes as the interventions are targeted to address individualized risks.

1. Introduction to Precision Medicine

The National Institute of Health (NIH), along with several other research centers, has created the Precision Medicine Initiative as a new way of approaching medicine with a targeted and patient-centered focus [1]. Specifically, they have defined precision medicine as an “emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle for each person” [1]. This approach to the practice of medicine has a high potential for treating the nuances of individuals with different genetics, lifestyle factors, and medical comorbidities that may affect their response to treatment. Since its initiation, many fields, including oncology [2] and cardiology [3], have begun refocusing their efforts to more precision-based approaches to practicing medicine. The role that genetics plays in the development of late-onset Alzheimer’s disease (AD) has been widely studied, with one study estimating genetics to account for more than 50% of the phenotypic variance [4]. However, the field of AD prevention has yet to fully advance intervention strategies from universal risk-reduction approaches to targeted interventions based on personalized risk factors, including genetics. In the following discussion, we review examples of a universal “one-size-fits-all” prevention strategy without any distinction that is based on genetics or other personalized risk factors versus a clinical precision medicine approach. From a practical clinical perspective, we have focused on two genes that can be commercially sequenced for polymorphisms that are associated with AD and that physicians may order to help better inform patient care. These include the most well-characterized genetic influencer on late-onset AD risk, apolipoprotein E (APOE), and another potential genetic influencer, methylenetetrahydrofolate reductase (MTHFR). With the increasing ease of both clinical lab-based, as well as direct-to-consumer genetic sequencing, a precision medicine approach that incorporates established genetic factors may be feasible and may also favorably affect patient outcomes by addressing individualized risks as well as pharmacogenomics and nutrigenomic considerations for AD.

2. “One-Size-Fits-All” Approach to AD Prevention

Randomized studies in AD prevention have traditionally used either single or multiple interventions to determine efficacy across a host of clinical outcome measures (e.g., cognitive function, serum biomarkers, brain imaging). The vast majority of these studies have used a “one-size-fits-all” approach to targeting diet, exercise, and other lifestyle factors without accounting for any individual genetic variables. Two large-scale randomized control trials (RCTs), the Multidomain Alzheimer Prevention Trial (MAPT) and Prevention of Dementia by Intensive Vascular Care (PreDIVA) trial did not show improvements in cognitive functioning with lifestyle interventions, including nutrition, physical activity, cognitive engagement, and management of comorbidities [5,6,7]. However, these studies used populations that were already experiencing some degree of cognitive decline or dementia. As AD starts developing in the brain decades before clinical symptoms become apparent [8], these study populations may not have been optimized to benefit from lifestyle modifications since individuals experiencing cognitive decline may already be beyond a critical window for AD prevention [9].
The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) was the first multicenter RCT to investigate the effects of similar lifestyle interventions on cognitive functioning in non-impaired individuals at risk for cognitive decline [10]. The results of the FINGER trial demonstrated that all individuals, regardless of baseline cognition, cardiovascular risk, demographics, or socioeconomic status improved with lifestyle interventions [11,12]. While not included in the initial study, a sub-analysis of the FINGER trial further explored the impact of a particular genetic factor, APOE, on lifestyle interventions in this cohort. This sub-analysis is further discussed in the ‘Precision Medicine Approach to AD Prevention’ section.
Several other prevention studies have shown improvement in cognitive function by implementing universal lifestyle interventions in non-impaired individuals, but with highly variable results. The two categories of interventions with the most robust evidence thus far include nutrition (including dietary patterns and single or multi-nutrients) and physical exercise. The Mediterranean diet is one example of a dietary pattern that has been extensively studied for AD prevention. A recent meta-analysis investigating the impact of the Mediterranean diet on cognitive functioning showed that there was a lower risk of cognitive decline and conversion to mild cognitive impairment (MCI) or AD in subjects with higher adherence to the diet [13]. However, there are also studies that have failed to demonstrate benefits of diet-specific interventions for AD prevention as well as various studies with mixed findings about which particular dietary interventions are the most beneficial [14,15,16].
Another well-studied dietary intervention focusing on single or multi-nutrients in the area of AD prevention has aimed to optimize levels of omega-3 polyunsaturated fatty acids (n-3 PUFA), most specifically, docosahexaenoic acid (DHA) [17,18,19,20,21,22,23]. Epidemiological evidence indicates that regular fish consumption and higher n-3 PUFA levels may reduce the risk for age-associated cognitive decline and AD [24]. Also, higher blood n-3 PUFA levels are protective of cortical structures [25], and chronic fish oil supplementation is associated with increased posterior cingulate activation in non-demented older adults [26]. Additional studies have shown improvement in cognitive function or decreased risk of AD in healthy individuals with DHA supplementation [18,19,20] or with consumption of fish high in omega-3s once per week [20]. However, other studies have suggested that there is no benefit of omega-3 supplementation with regard to cognitive function and AD prevention [22,23].
Another lifestyle intervention that has been studied for AD prevention with variable results is physical activity. A meta-analysis on the role of physical activity in AD prevention concluded that physical activity significantly decreased the risk of developing AD [27]. In addition, an RCT that looked at the impact of physical activity on cognitive functioning demonstrated that individuals who participated in six months of physical activity showed improvement in cognitive functioning up to 18 months later [28]. Researchers have also investigated the timing and intensity of physical activity. One study found that light and vigorous activity in mid-life and light and moderate activity in late-life were associated with lower risks of developing MCI [29]. However, similar to the variations in the nutrition data, there have been varied findings and conclusions about the type and intensity of physical activity that are most effective at reducing the risk of AD, as well as studies demonstrating no risk reduction from physical activity [30,31,32].
The discrepancy in the data on nutritional interventions and physical activity may be related to a lack of a “one-size-fits-all” solution, and modifications in diet, exercise, and other lifestyle factors may need to be personalized to have maximum efficacy. Unlike universal risk reduction approaches, precision medicine strategies allow for incorporation of individual risk factors that may uniquely affect the response to interventions.

3. Precision Medicine Approach to AD Prevention

A precision medicine approach to AD prevention will need to fully utilize the genome in order to make personalized recommendations. In this section, we discuss some of the genetic influencers on late-onset AD that can currently be ordered by a practicing physician and provide examples of a targeted precision medicine approach based on these genetic factors.

4. APOE and AD Prevention

One of the most well-established genetic influencers on late-onset AD risk is APOE [33], which codes for the apolipoprotein E protein [34]. There are three major polymorphisms at the APOE loci: APOE ε2, ε3, and ε4. Studies have shown that APOE genotype significantly impacts the risk of AD. Specifically, the ε4 allele has been associated with an increased risk of AD [35], while the ε2 allele has been associated with a decreased risk [36]. In addition, the risk of developing AD is even greater in individuals with two copies of the ε4 allele when compared to those with only one copy [37].
Several pathophysiologic mechanisms may explain why APOE ε4 is associated with an increased risk of AD and APOE ε2 is associated with a decreased risk. First, the three major alleles code for proteins with different molecular properties that result in different binding properties of apolipoprotein E to β-amyloid. This difference in binding may contribute to the enhanced accumulation of β-amyloid plaques that was observed in ε4 individuals, which is one of the pathologic markers of AD [38]. Furthermore, their distinct molecular properties also result in differences in their ability to bind to and transport lipids. Studies have demonstrated that there are allele-specific interactions of APOE with both LDL and HDL receptors that play an important role in the development of atherosclerosis, which is one of the major risk factors for AD [34]. As the ε4 allele has been estimated to account for 27.3% of late-onset AD risk (with a heritability of 80%) and with emerging evidence that potential risk-reduction interventions may be preferentially effective (or less effective) depending on presence of the ε4 allele, it may be important to incorporate this genotype into the AD prevention approach [39].
There are several AD prevention interventions that can be personalized based on APOE genotype. Although the FINGER trial showed no significant differences in cognitive function between APOE genotypes with their multimodal lifestyle interventions, a within-group analysis of the APOE ε4 allele demonstrated that there was a significant difference in certain treatment versus control scores only for individuals with ε4 alleles [12]. This suggests that some inherent difference exists between individuals with and without APOE ε4 alleles that impacted the effectiveness of the interventions. Therefore, additional trials with larger sample sizes and more statistical power are important in order to discern the impact of APOE on these multimodal interventions.
Other single-factor studies have demonstrated that AD prevention interventions can be targeted based on APOE genotype. A systematic review of studies that altered dietary fat composition showed that changes in total cholesterol, LDL, and HDL were most significant in individuals with APOE ε4 alleles in 15 of the studies [40]. In another study, researchers found that, in response to a Mediterranean diet, both individuals with and without APOE ε4 alleles showed improvements in cognitive functioning, as measured by the Mini Mental State Exam (MMSE), but only individuals without ε4 alleles showed improvement in the clock drawing test, a measure of executive functioning and spatial reasoning [41]. Tailoring strategies to APOE genotype can also be effective for physical activity interventions. For example, one study demonstrated that sedentary individuals with ε4 alleles were at greater risk of developing MCI, whereas physically active individuals without ε4 alleles were at decreased risk [29]. Another study demonstrated that aerobic fitness was correlated with higher cognitive performance in ε4 homozygotes [42]. Similarly, with regard to omega-3 fatty acids, three recent RCTs showed an improvement in cognitive function with DHA supplementation in non-impaired individuals with ε4 alleles [43].
While a comprehensive review of the literature is beyond the scope of this manuscript, these studies demonstrate that, based on APOE genotype, individuals may exhibit more significant responses to different lifestyle interventions. For example, individuals with ε4 alleles may experience greater changes in total cholesterol, LDL, and HDL in response to reductions in dietary fat, whereas individuals without an ε4 allele might show greater improvement in certain cognitive functions from the Mediterranean diet. In addition, physical activity may benefit all individuals but may have increased efficacy for those with ε4 alleles. Similarly, DHA supplementation may also lead to greater improvement in cognitive function in those with at least one ε4 allele. Overall, genotype-specific strategies such as these may benefit patients by using an evidence-based approach and utilizing specific targeted interventions that were shown to be the most effective for individuals with their same genotype. Additional research to further elucidate the role of APOE genotype on different dietary, physical activity, and other lifestyle interventions will be important in the future as the precision medicine approach to AD prevention continues to develop.

5. MTHFR and AD Prevention

The MTHFR gene, which codes for the methylenetetrahydrofolate reductase protein, is another potential genetic contributor to AD and is also readily available for physicians to order in commercial labs. Several MTHFR polymorphisms have been described in the literature [44], but two polymorphisms, C677T and A1298C, have had the greatest investigation as to their association with AD [45]. These polymorphisms also appear to have a high prevalence in the general population [46], and one study reported that 92.5% of its AD subjects had at least one of these MTHFR polymorphisms [45].
The association between MTHFR polymorphisms and AD may relate to the catalytic role that the MTHFR protein plays as the rate-limiting step in the conversion of homocysteine into methionine, with the B-vitamins folate and cobalamin serving as cofactors [47]. Homocysteine is an amino acid that is involved in inflammation and has been associated with cognitive decline and an increased risk of AD [48,49]. One study in cognitively healthy individuals found that baseline homocysteine levels inversely correlated with cognitive testing scores and rates of cognitive decline over a five-year period [48]. Similarly, another study of 1000 individuals from the Framingham cohort looked at non-impaired individuals at baseline and showed a strong positive correlation between baseline homocysteine and the risk of dementia up to 11 years later [49]. Another longitudinal study showed there was an 88% increased rate of cognitive decline over ten years associated with doubling the homocysteine level from 10 mg/L to 20 mg/L [50].
Changes in the MTHFR protein that alter its catalytic function, such as seen in the C677T and A1298C polymorphisms, result in higher levels of serum homocysteine [51], and therefore, have the potential to increase the risk of AD. Several studies have shown an association between the A1298C polymorphism and an increased risk of AD [52], but not with the C677T polymorphism [52,53]. However, another study showed that the combination of these two polymorphisms with a third A1793G polymorphism, together known as Haplotype C, was associated with a decreased risk of AD [54]. Therefore, further research into the relationship between these polymorphisms and the risk of AD is warranted.
Similar to APOE, MTHFR genotype status may allow for targeted AD prevention interventions. B-vitamin supplementation (cyanocobalamin, folic acid, and B6) has been shown to slow cognitive decline in individuals with elevated homocysteine levels [55,56]. Several trials have studied a combination of B vitamins to determine whether lowering homocysteine can impact cognitive function and/or brain pathology [55]. While there is limited evidence thus far, individuals with one or more MTHFR polymorphisms may potentially benefit from genotype-specific recommendations. For example, as individuals with certain MTHFR polymorphisms have decreased catalytic ability of the MTHFR protein, replacing the traditional B-vitamins with their methylated counterparts (methylcobalamin for cyanocobalamin and methyltetrahydrofolate [5-MTH] for folic acid) that do not require hepatic conversion to active forms may increase the outcomes. One study demonstrated that 5-MTH supplementation in individuals with C677T and A1298C polymorphisms significantly increased the serum folate concentration when compared to folic acid, but it did not result in differences in the serum homocysteine concentration [57]. Additional studies evaluating the impact of methylated B-vitamins for specific MTHFR polymorphisms and AD risk may therefore help to advance the field of precision medicine for AD prevention.

6. Other Genetic Influencers on AD Prevention

In addition to the discussed polymorphisms in the APOE and MTHFR genes, recent genome-wide association studies (GWAS) have identified several other single nucleotide polymorphisms (SNPs) that are associated with an increased risk of AD: CLU, CR1, and PICALM. Although these genes are not yet routinely available for sequencing commercially, the impact of polymorphisms at these loci on dietary interventions for AD prevention has recently been investigated [58,59]. One study demonstrated that improvements in cognitive function in response to the Mediterranean diet differed depending on which polymorphisms an individual had [41]. These findings provide further evidence that genetics may modify the effectiveness of AD prevention interventions. As this trial only investigated the impact of the Mediterranean diet on polymorphisms at these loci, other dietary interventions as well as other lifestyle interventions should be explored in a similar manner. In addition, there are many other known genetic risk factors for AD, such as TOMM40, which have yet to be explored regarding their impact on lifestyle interventions for AD prevention [60]. However, these genes are also not yet routinely commercially available for sequencing. A discussion of all of the genes that are involved in AD risk is beyond the scope of this paper, but it is discussed further in an Alzgene meta-analysis [61].

7. Conclusions and Future Directions

This review considered examples of two approaches to AD prevention: a universal “one-size-fits-all” approach, which uses generalized prevention strategies for all individuals, and a clinical precision medicine approach, which factors in genotype-specific intervention strategies. While both approaches have merit, utilizing a precision medicine approach offers the opportunity to personalize interventions that are based on factors that may impact the efficacy of the interventions. Genotype-specific intervention strategies, in particular, hold a great deal of promise for advancing the field of AD prevention toward more personalized and effective intervention strategies.
Investigation into the impact of different genetic factors on AD prevention will continue to become more practicable through online genetic repositories that are available to the scientific community. For example, the Alzheimer’s Disease Sequencing Project (ADSP) and Alzheimer’s Disease Neuroimaging Initiative (ADNI) are ongoing large-scale whole-exome and whole-genome sequencing projects in individuals with AD available through the NIA Genetics of Alzheimer’s Disease Data Storage Site (NIAGADS) and the database of Genotypes and Phenotypes (dbGaP) [62]. This increased availability of genetic data will provide additional resources to investigate the impact of various genetic factors on AD prevention interventions in the future.
In addition, there has been an exponential growth in the ability of consumers to order personal genomic testing on their own via a number of commercially available testing kits. In the United States, the Food and Drug Administration approved the first direct-to-consumer tests that provide genetic risk information for a subset of medical conditions, including APOE [63]. Further, despite these commercial tests not being meant for clinical purposes, it has also become more common for patients (and even some physicians) to use a number of online tools to further investigate the raw data provided by these tests. Websites such as Promethiase.com and Snpedia.com may be utilized, although there are currently no professional guidelines and/or standards on how to do this [64].
We should also be mindful of the ethical implications of integrating genetic risk factors into clinical practice. While the Risk Evaluation and Education for Alzheimer’s Disease (REVEAL) study demonstrated that APOE ε4 disclosure to adult children of AD patients did not result in significant short-term psychological effects, the long-term effects have not been evaluated [65,66]. Clinicians should weigh the potential risks and benefits of disclosing genetic risk factors to their patients and should counsel patients accordingly prior to disclosing genotype status [67]. Referral to a certified genetic counselor should also be considered when clinically indicated. Over the last five years at the Alzheimer’s Prevention Clinic (APC) at Weill Cornell Medicine and NewYork-Presbyterian, the majority of patients (over 95%) have consented to receive APOE and MTHFR testing [68]. Counseling is initially provided in person by either of the two treating clinicians (a board-certified Neurologist or Family Nurse Practitioner). Patients are also asked to complete an online course via AlzU.org that explains genetic risk for AD and limitations of these tests [69]. In select cases, when patients have additional questions or concerns about testing, patients may be referred to a genetic counselor. In all patients with a family history that is highly suggestive of early-onset (autosomal dominant) AD, patients are referred to a genetic counselor prior to any genetic testing. Studies are ongoing to determine whether APOE and MTHFR polymorphism disclosure to APC patients impacts outcomes (e.g., compliance with recommendations, psychological measures including anxiety and depression). Furthermore, additional analyses are planned to determine whether clinical outcomes (e.g., cognitive performance, blood biomarkers of AD risk) are differentially impacted by APOE and MTHFR genotype. Generally speaking, the use of genetic testing as a part of clinical evaluation and patient care has been a favorable addition in the opinion of the treating clinicians, although further study is warranted in a broader subset of clinicians and in diverse patient cohorts.
Finally, it is important to consider the limitations of a genetic-based precision medicine approach to AD prevention. The genomic-centered foundation that forms the core of precision medicine reduces diseases to their molecular and cellular processes. However, there are many risk factors for AD in which the exact pathogenesis is not fully understood. A precision medicine approach that relies solely on genetics may miss some of the underlying mechanisms that are important for AD prevention but as of yet are not fully established. In addition to genetics, there are many other important aspects of a precision medicine approach to AD prevention, including medical comorbidities such as hypertension [70,71], diabetes [72], and hyperlipidemia [73,74], which have been associated with an increased risk of developing AD. There are also other lifestyle factors in addition to diet, exercise, and omega-3 fatty acids, such as smoking status, alcohol consumption, and cognitive engagement, which may play a role in AD prevention. Therefore, a precision medicine approach should also encompass recommendations to target these lifestyle factors and medical comorbidities on an individual basis. All of these factors need to be considered together to maximize a precision medicine approach that targets AD prevention strategies to the individual. Ultimately, genetics should be incorporated as one part of an overarching precision medicine approach to individualize AD prevention strategies.

Author Contributions

Conceptualization, C.L.B. and R.S.I.; Writing-Original Draft Preparation, C.L.B.; Writing-Review & Editing, C.L.B., R.S.I., L.M., O.S., A.R., and H.W.; Visualization, C.L.B., R.S.I., and L.M.; Supervision, R.S.I.; Project Administration, O.S. and A.R.; Funding Acquisition, R.S.I. and L.M.

Funding

This research was funded by philanthropic support by the Zuckerman Family Foundation, Women’s Alzheimer’s Movement, David G. Kabiller Charitable Foundation (In Memory of Adele Rubin Tunick and In Memory of Kaisu Ilmanen), Rimora Foundation, the Washkowitz Family in Memory of Alan Washkowitz, proceeds from the Annual Memories for Mary fundraiser organized by David Twardock, and contributions from grateful patients of the Alzheimer’s Prevention Clinic, Weill Cornell Memory Disorders Program; Grant funding by the Weill Cornell Medicine Clinical and Translational Science Center (NIH/NCATS #UL1TR002384), and NIH PO1AG026572. The funders had no role in the study design, data collection or analysis, decision to publish, or preparation of the manuscript.

Conflicts of Interest

R.S.I. has served as a consultant for Lilly, Neurotrack, and 23andMe: DNA Genetic Testing and Analysis; other authors declare no conflict of interest. The funders had no role in the study design, data collection or analysis, decision to publish, or preparation of the manuscript.

References

  1. U.S. National Library of Medicine. What is the Precision Medicine Initiative? 2018. Available online: https://ghr.nlm.nih.gov/primer/precisionmedicine/initiative (accessed on 10 April 2018).
  2. Shin, S.H.; Bode, A.M.; Dong, Z. Precision medicine: The foundation of future cancer therapeutics. npj Precis. Oncol. 2017, 1, 12. [Google Scholar] [CrossRef] [PubMed]
  3. Antman, E.M.; Loscalzo, J. Precision medicine in cardiology. Nat. Rev. Cardiol. 2016, 13, 591–602. [Google Scholar] [CrossRef] [PubMed]
  4. Ridge, P.G.; Hoyt, K.B.; Boehme, K.; Mukherjee, S.; Crane, P.K.; Haines, J.L.; Mayeux, R.; Farrer, L.A.; Pericak-Vance, M.A.; Schellenberg, G.D.; et al. Assessment of the genetic variance of late-onset Alzheimer’s disease. Neurobiol. Aging 2016, 41, 200.e13–200.e20. [Google Scholar] [CrossRef] [PubMed]
  5. Carrié, I.; van Kan, G.A.; Gillette-Guyonnet, S.; Andrieu, S.; Dartigues, J.F.; Touchon, J.; Dantoine, T.; Rouaud, O.; Bonnefoy, M.; Robert, P.; et al. Recruitment strategies for preventive trials. The MAPT study (MultiDomain Alzheimer Preventive Trial). J. Nutr. Health Aging 2012, 16, 355–359. [Google Scholar] [CrossRef] [PubMed]
  6. Gillette-Guyonnet, S.; Andrieu, S.; Dantoine, T.; Dartigues, J.F.; Touchon, J.; Vellas, B. Commentary on “A roadmap for the prevention of dementia II. Leon Thal Symposium 2008.” The Multidomain Alzheimer Preventive Trial (MAPT): A new approach to the prevention of Alzheimer’s disease. Alzheimer’s Dement. 2009, 5, 114–121. [Google Scholar] [CrossRef] [PubMed]
  7. Richard, E.; Van den Heuvel, E.; Moll van Charante, E.P.; Achthoven, L.; Vermeulen, M.; Bindels, P.J.; Van Gool, W.A. Prevention of dementia by intensive vascular care (PreDIVA): A cluster-randomized trial in progress. Alzheimer Dis. Assoc. Disord. 2009, 23, 198–204. [Google Scholar] [CrossRef] [PubMed]
  8. Morris, J.C. Early-stage and preclinical Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2005, 19, 163–165. [Google Scholar] [PubMed]
  9. Schelke, M.W.; Hackett, K.; Chen, J.L.; Shih, C.; Shum, J.; Montgomery, M.E.; Chiang, G.C.; Berkowitz, C.; Seifan, A.; Krikorian, R.; et al. Nutritional interventions for Alzheimer’s prevention: A clinical precision medicine approach. Ann. N. Y. Acad. Sci. 2016, 1367, 50–56. [Google Scholar] [CrossRef] [PubMed]
  10. Kivipelto, M.; Solomon, A.; Ahtiluoto, S.; Ngandu, T.; Lehtisalo, J.; Antikainen, R.; Backman, L.; Hanninen, T.; Jula, A.; Laatikainen, T.; et al. The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER): Study design and progress. Alzheimer’s Dement. 2013, 9, 657–665. [Google Scholar] [CrossRef] [PubMed]
  11. Rosenberg, A.; Ngandu, T.; Rusanen, M.; Antikainen, R.; Backman, L.; Havulinna, S.; Hanninen, T.; Laatikainen, T.; Lehtisalo, J.; Levalahti, E.; et al. Multidomain lifestyle intervention benefits a large elderly population at risk for cognitive decline and dementia regardless of baseline characteristics: The FINGER trial. Alzheimer’s Dement. 2018, 14, 263–270. [Google Scholar] [CrossRef] [PubMed]
  12. Solomon, A.; Turunen, H.; Ngandu, T.; Peltonen, M.; Levalahti, E.; Helisalmi, S.; Antikainen, R.; Backman, L.; Hanninen, T.; Jula, A.; et al. Effect of the Apolipoprotein E Genotype on Cognitive Change During a Multidomain Lifestyle Intervention: A Subgroup Analysis of a Randomized Clinical Trial. JAMA Neurol. 2018, 75, 462–470. [Google Scholar] [CrossRef] [PubMed]
  13. Singh, B.; Parsaik, A.K.; Mielke, M.M.; Erwin, P.J.; Knopman, D.S.; Petersen, R.C.; Roberts, R.O. Association of mediterranean diet with mild cognitive impairment and Alzheimer’s disease: A systematic review and meta-analysis. J. Alzheimer’s Dis. 2014, 39, 271–282. [Google Scholar] [CrossRef] [PubMed]
  14. Kesse-Guyot, E.; Andreeva, V.A.; Lassale, C.; Ferry, M.; Jeandel, C.; Hercberg, S.; Galan, P. Mediterranean diet and cognitive function: A French study. Am. J. Clin. Nutr. 2018, 97, 369–376. [Google Scholar]
  15. Wahl, D.; Cogger, V.C.; Solon-Biet, S.M.; Waern, R.V.; Gokarn, R.; Pulpitel, T.; Cabo, R.; Mattson, M.P.; Raubenheimer, D.; Simpson, S.J.; et al. Nutritional strategies to optimise cognitive function in the aging brain. Ageing Res. Rev. 2016, 31, 80–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Mosconi, L.; McHugh, P.F. Let Food Be Thy Medicine: Diet, Nutrition, and Biomarkers’ Risk of Alzheimer’s Disease. Curr. Nutr. Rep. 2015, 4, 126–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Kulzow, N.; Witte, A.V.; Kerti, L.; Grittner, U.; Schuchardt, J.P.; Hahn, A.; Floel, A. Impact of Omega-3 Fatty Acid Supplementation on Memory Functions in Healthy Older Adults. J. Alzheimer’s Dis. 2016, 51, 713–725. [Google Scholar] [CrossRef] [PubMed]
  18. Abubakari, A.R.; Naderali, M.M.; Naderali, E.K. Omega-3 fatty acid supplementation and cognitive function: Are smaller dosages more beneficial? Int. J. Gen. Med. 2014, 7, 463–473. [Google Scholar] [PubMed]
  19. Yurko-Mauro, K.; Alexander, D.D.; van Elswyk, M.E. Docosahexaenoic acid and adult memory: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0120391. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, Y.; Chen, J.; Qiu, J.; Li, Y.; Wang, J.; Jiao, J. Intakes of fish and polyunsaturated fatty acids and mild-to-severe cognitive impairment risks: A dose-response meta-analysis of 21 cohort studies. Am. J. Clin. Nutr. 2016, 103, 330–340. [Google Scholar] [CrossRef] [PubMed]
  21. Morris, M.C.; Brockman, J.; Schneider, J.A.; Wang, Y.; Bennett, D.A.; Tangney, C.C.; van de Rest, O. Association of Seafood Consumption, Brain Mercury Level, and APOE epsilon4 Status with Brain Neuropathology in Older Adults. JAMA 2016, 315, 489–497. [Google Scholar] [CrossRef] [PubMed]
  22. Sydenham, E.; Dangour, A.D.; Lim, W.S. Omega 3 fatty acid for the prevention of cognitive decline and dementia. Cochrane Database Syst. Rev. 2012, 13, CD005379. [Google Scholar]
  23. Jiao, J.; Li, Q.; Chu, J.; Zeng, W.; Yang, M.; Zhu, S. Effect of n-3 PUFA supplementation on cognitive function throughout the life span from infancy to old age: A systematic review and meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2014, 100, 1422–1436. [Google Scholar] [CrossRef] [PubMed]
  24. Cunnane, S.C.; Plourde, M.; Pifferi, F.; Begin, M.; Feart, C.; Barberger-Gateau, P. Fish, docosahexaenoic acid and Alzheimer’s disease. Prog. Lipid Res. 2009, 48, 239–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. McNamara, R.K.; Asch, R.H.; Lindquist, D.M.; Krikorian, R. Role of polyunsaturated fatty acids in human brain structure and function across the lifespan: An update on neuroimaging findings. Prostaglandins Leukot Essent. Fatty Acids 2017. [Google Scholar] [CrossRef] [PubMed]
  26. Boespflug, E.L.; McNamara, R.K.; Eliassen, J.C.; Schidler, M.D.; Krikorian, R. Fish Oil Supplementation Increases Event-Related Posterior Cingulate Activation in Older Adults with Subjective Memory Impairment. J. Nutr. Health Aging 2016, 20, 161–169. [Google Scholar] [CrossRef] [PubMed]
  27. Hamer, M.; Chida, Y. Physical activity and risk of neurodegenerative disease: A systematic review of prospective evidence. Psychol. Med. 2009, 39, 3–11. [Google Scholar] [CrossRef] [PubMed]
  28. Lautenschlager, N.T.; Cox, K.L.; Flicker, L.; Foster, J.K.; van Bockxmeer, F.M.; Xiao, J.; Greenop, K.R.; Almeida, O.P. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: A randomized trial. JAMA 2008, 300, 1027–1037. [Google Scholar] [CrossRef] [PubMed]
  29. Krell-Roesch, J.; Pink, A.; Roberts, R.O.; Stokin, G.B.; Mielke, M.M.; Spangehl, K.A.; Bartley, M.M.; Knopman, D.S.; Christianson, T.J.; Petersen, R.C.; et al. Timing of Physical Activity, Apolipoprotein E epsilon4 Genotype, and Risk of Incident Mild Cognitive Impairment. J. Am. Geriatr. Soc. 2016, 64, 2479–2486. [Google Scholar] [CrossRef] [PubMed]
  30. Sink, K.M.; Espeland, M.A.; Castro, C.M.; Church, T.; Cohen, R.; Dodson, J.A.; Guralnik, J.; Hendrie, H.C.; Jennings, J.; Katula, J.; et al. Effect of a 24-Month Physical Activity Intervention vs Health Education on Cognitive Outcomes in Sedentary Older Adults: The LIFE Randomized Trial. JAMA 2015, 314, 781–790. [Google Scholar] [CrossRef] [PubMed]
  31. Sachs, B.C.; Skinner, J.S.; Sink, K.M.; Craft, S.; Baker, L.D. High intensity aerobic exercise improves performance on computer tests of executive function in adults with mild cognitive impairment: implications for cognitive assessment in clinical trials. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2016, 12, 428. [Google Scholar] [CrossRef]
  32. Chang, Y.K.; Pan, C.Y.; Chen, F.T.; Tsai, C.L.; Huang, C.C. Effect of resistance-exercise training on cognitive function in healthy older adults: A review. J. Aging Phys. Act. 2012, 20, 497–517. [Google Scholar] [CrossRef] [PubMed]
  33. Ballard, C.; Gauthier, S.; Corbett, A.; Brayne, C.; Aarsland, D.; Jones, E. Alzheimer’s disease. Lancet 2011, 377, 1019–1031. [Google Scholar] [CrossRef]
  34. Mahley, R.W. Apolipoprotein E: Cholesterol transport protein with expanding role in cell biology. Science 1988, 240, 622–630. [Google Scholar] [CrossRef] [PubMed]
  35. Corder, E.H.; Saunders, A.M.; Strittmatter, W.J.; Schmechel, D.E.; Gaskell, P.C.; Small, G.W.; Roses, A.D.; Haines, J.L.; Pericak-Vance, M.A. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993, 261, 921–923. [Google Scholar] [CrossRef] [PubMed]
  36. Talbot, C.; Lendon, C.; Craddock, N.; Shears, S.; Morris, J.C.; Goate, A. Protection against Alzheimer’s disease with apoE epsilon 2. Lancet 1994, 343, 1432–1433. [Google Scholar] [CrossRef]
  37. Farrer, L.A.; Cupples, L.A.; Haines, J.L.; Hyman, B.; Kukull, W.A.; Mayeux, R.; Myers, R.H.; Pericak-Vance, M.A.; Risch, N.; van Duijn, C.M. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 1997, 278, 1349–1356. [Google Scholar] [CrossRef] [PubMed]
  38. Strittmatter, W.J.; Weisgraher, K.H.; Huang, D.Y.; Dong, L.M.; Salvesen, G.S.; Pericak-Vance, M.; Schmechel, D.; Saunders, A.M.; Goldgaber, D.; Roses, A.D. Binding of human apolipoprotein E to synthetic amyloid beta peptide: Isoform-specific effects and implications for late-onset Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 8098–8102. [Google Scholar] [CrossRef] [PubMed]
  39. Van Cauwenberghe, C.; Vab Broeckhoven, C.; Sleegers, K. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet. Med. 2016, 18, 421–430. [Google Scholar] [CrossRef] [PubMed]
  40. Masson, L.F.; McNeill, G.; Avenell, A. Genetic variation and the lipid response to dietary intervention: A systematic review. Am. J. Clin. Nutr. 2003, 77, 1098–1111. [Google Scholar] [CrossRef] [PubMed]
  41. Martinez-Lapiscina, E.H.; Galbete, C.; Corella, D.; Toledo, E.; Buil-Cosiales, P.; Salas-Salvado, J.; Ros, E.; Martinez-Gonzalez, M.A. Genotype patterns at CLU, CR1, PICALM and APOE, cognition and Mediterranean diet: The PREDIMED-NAVARRA trial. Genes Nutr. 2014, 9, 393. [Google Scholar] [CrossRef] [PubMed]
  42. Etnier, J.L.; Caselli, R.J.; Reiman, E.M.; Alexander, G.E.; Sibley, B.A.; Tessier, D.; McLemore, E.C. Cognitive performance in older women relative to ApoE-epsilon4 genotype and aerobic fitness. Med. Sci. Sports Exerc. 2007, 39, 199–207. [Google Scholar] [CrossRef] [PubMed]
  43. Yassine, H.N.; Braskie, M.N.; Mack, W.J.; Castor, K.J.; Fonteh, A.N.; Schneider, L.S.; Harrington, M.G.; Chui, H.C. Association of Docosahexaenoic Acid Supplementation with Alzheimer Disease Stage in Apolipoprotein E ε4 Carriers: A Review. JAMA Neurol. 2017, 74, 339–347. [Google Scholar] [CrossRef] [PubMed]
  44. Sibani, S.; Christensen, B.; O’Ferrall, E.; Saadi, I.; Hiou-Tim, F.; Rosenblatt, D.S.; Rozen, R. Characterization of six novel mutations in the methylenetetrahydrofolate reductase (MTHFR) gene in patients with homocystinuria. Hum. Mutat. 2000, 15, 280–287. [Google Scholar] [CrossRef]
  45. Roman, G.C. MTHFR Gene Mutations: A Potential Marker of Late-Onset Alzheimer’s Disease? J. Alzheimer’s Dis. 2015, 47, 323–327. [Google Scholar] [CrossRef] [PubMed]
  46. Romero-Sánchez, C.; Gomez-Gutierrez, A.; Gomez, P.E.; Casas-Gomez, M.C.; Briceno, I. C677T (RS1801133) MTHFR gene polymorphism frequency in a colombian population. Colomb. Med. 2015, 46, 75–79. [Google Scholar] [PubMed]
  47. Online Mendelian Inheritance in Man (OMIM). An Online Catalog of Human Genes and Genetic Disorders. 2018. Available online: https://www.omim.org/ (accessed on 5 May 2018).
  48. McCaddon, A.; Hudson, P.; Davies, G.; Hughes, A.; Williams, J.H.; Wilkinson, C. Homocysteine and cognitive decline in healthy elderly. Dement. Geriatr. Cogn. Disord. 2001, 12, 309–313. [Google Scholar] [CrossRef] [PubMed]
  49. Seshadri, S.; Beiser, A.; Selhub, J.; Jacques, P.F.; Rosenberg, I.H.; D’Agostino, R.B.; Wilson, P.W.; Wolf, P.A. Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N. Engl. J. Med. 2002, 346, 476–483. [Google Scholar] [CrossRef] [PubMed]
  50. Clarke, R.; Birks, J.; Nexo, E.; Ueland, P.M.; Schneede, J.; Scott, J.; Molloy, A.; Evans, J.G. Low vitamin B-12 status and risk of cognitive decline in older adults. Am. J. Clin. Nutr. 2007, 86, 1384–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Weisberg, I.; Tran, P.; Christensen, B.; Sibani, S.; Rozen, R. A Second Genetic Polymorphism in Methylenetetrahydrofolate Reductase (MTHFR) Associated with Decreased Enzyme Activity. Mol. Genet. Metab. 1998, 64, 169–172. [Google Scholar] [CrossRef] [PubMed]
  52. Mansouri, L.; Fekih-Mrissa, N.; Klai, S.; Mansour, M.; Gritli, N.; Mrissa, R. Association of methylenetetrahydrofolate reductase polymorphisms with susceptibility to Alzheimer’s disease. Clin. Neurol. Neurosurg. 2013, 115, 1693–1696. [Google Scholar] [CrossRef] [PubMed]
  53. Seripa, D.; Forno, G.D.; Matera, M.G.; Gravina, C.; Margaglione, M.; Palermo, M.T.; Wekstein, D.R.; Antuono, P.; Avis, D.G.; Daniele, A.; et al. Methylenetetrahydrofolate reductase and angiotensin converting enzyme gene polymorphisms in two genetically and diagnostically distinct cohort of Alzheimer patients. Neurobiol. Aging 2003, 24, 933–939. [Google Scholar] [CrossRef]
  54. Wakutani, Y.; Kowa, H.; Kusumi, M.; Nakaso, K.; Yasui, K.; Isoe-Wada, K.; Yano, H.; Urakami, K.; Takeshima, T.; Nakashima, K. A haplotype of the methylenetetrahydrofolate reductase gene is protective against late-onset Alzheimer’s disease. Neurobiol. Aging 2004, 25, 291–294. [Google Scholar] [CrossRef]
  55. Smith, A.D.; Smith, S.M.; de Jager, C.A.; Whitbread, P.; Johnston, C.; Agacinski, G.; Oulhaj, A.; Bradley, K.M.; Jacoby, R.; Refsum, H. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: A randomized controlled trial. PLoS ONE 2010, 5, e12244. [Google Scholar] [CrossRef] [PubMed]
  56. Douaud, G.; Refsum, H.; de Jager, C.A.; Jacoby, R.; Nichols, T.E.; Smith, S.M.; Smith, A.D. Preventing Alzheimer’s disease-related gray matter atrophy by B-vitamin treatment. Proc. Natl. Acad. Sci. USA 2013, 110, 9523–9528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hekmatdoost, A.; Vahid, F.; Yari, Z.; Sadeghi, M.; Eini-Zinab, H.; Lakpour, N.; Arefi, S. Methyltetrahydrofolate vs Folic Acid Supplementation in Idiopathic Recurrent Miscarriage with Respect to Methylenetetrahydrofolate Reductase C677T and A1298C Polymorphisms: A Randomized Controlled Trial. PLoS ONE 2015, 10, e0143569. [Google Scholar] [CrossRef] [PubMed]
  58. Harold, D.; Abraham, R.; Hollingworth, P.; Sims, R.; Gerrish, A.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; Dowzell, K.; Williams, A.; et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1088–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Lambert, J.C.; Heath, S.; Even, G.; Campion, D.; Sleegers, K.; Hiltunen, M.; Combarros, O.; Zelenika, D.; Bullido, M.J.; Tavernier, B.; et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
  60. Lutz, M.W.; Crenshaw, D.G.; Saunders, A.M.; Roses, A.D. Genetic variation at a single locus and age of onset for Alzheimer’s disease. Alzheimer’s Dement. 2010, 6, 125–131. [Google Scholar] [CrossRef] [PubMed]
  61. Bertram, L.; McQueen, M.B.; Mullin, K.; Blacker, D.; Tanzi, R.E. Systematic meta-analyses of Alzheimer disease genetic association studies: The AlzGene database. Nat. Genet. 2007, 39, 17–23. [Google Scholar] [CrossRef] [PubMed]
  62. Reitz, C. Genetic diagnosis and prognosis of Alzheimer’s disease: Challenges and opportunities. Expert Rev. Mol. Diagn. 2015, 15, 339–348. [Google Scholar] [CrossRef] [PubMed]
  63. U.S. Food Drug Administration. Press Announcements—FDA Allows Marketing of First Direct-to-Consumer Tests that Provide Genetic Risk Information for Certain Conditions; U.S. Food Drug Administration: Silver Spring, MD, USA, 2018.
  64. Watershed DNA. Filtering a Promethease Report: One Genetic Counselor’s Strategy. 2018. Available online: https://www.watersheddna.com/blog-and-news/filtering-a-promethease-report-one-genetic-counselors-strategy (accessed on 25 June 2018).
  65. Cupples, L.A.; Farrer, L.A.; Sadovnick, A.D.; Relkin, N.; Whitehouse, P.; Green, R.C. Estimating risk curves for first-degree relatives of patients with Alzheimer’s disease: The REVEAL study. Genet. Med. 2004, 6, 192–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Green, R.C.; Roberts, J.S.; Cupples, L.A.; Relkin, N.R.; Whitehouse, P.J.; Brown, T.; Eckert, S.L.; Butson, M.; Sadovnick, A.D.; Quaid, K.A.; et al. Disclosure of APOE genotype for risk of Alzheimer’s disease. N. Engl. J. Med. 2009, 361, 245–254. [Google Scholar] [CrossRef] [PubMed]
  67. Stites, S. Cognitively Healthy Individuals Want to Know Their Risk for Alzheimer’s Disease: What Should We Do? J. Alzheimer’s Dis. 2018, 62, 499–502. [Google Scholar] [CrossRef] [PubMed]
  68. Seifan, A.; Isaacson, R. The Alzheimer’s Prevention Clinic at Weill Cornell Medical College/New York—Presbyterian Hospital: Risk Stratification and Personalized Early Intervention. J. Prev. Alzheimer’s Dis. 2015, 2, 254–266. [Google Scholar]
  69. Isaacson, R.S.; Haynes, N.; Seifan, A.; Larsen, D.; Christiansen, S.; Berger, J.C.; Safdieh, J.E.; Lunde, A.M.; Luo, A.; Kramps, M.; et al. Alzheimer’s Prevention Education: If We Build It, Will They Come? www.AlzU.org. J. Prev. Alzheimer’s Dis. 2014, 1, 91–98. [Google Scholar]
  70. Gabin, J.M.; Tambs, K.; Saltvedt, I.; Sund, E.; Holmen, J. Association between blood pressure and Alzheimer disease measured up to 27 years prior to diagnosis: The HUNT Study. Alzheimer’s Res. Ther. 2017, 9, 37. [Google Scholar] [CrossRef] [PubMed]
  71. Morris, M.C.; Scherr, P.A.; Hebert, L.E.; Glynn, R.J.; Bennett, D.A.; Evans, D.A. Association of incident Alzheimer disease and blood pressure measured from 13 years before to 2 years after diagnosis in a large community study. Arch. Neurol. 2001, 58, 1640–1646. [Google Scholar] [CrossRef] [PubMed]
  72. De Nazareth, A.M. Type 2 diabetes mellitus in the pathophysiology of Alzheimer’s disease. Dement. Neuropsychol. 2017, 11, 105–113. [Google Scholar] [CrossRef] [PubMed]
  73. Kivipelto, M.; Helkala, E.L.; Laakso, M.P.; Hanninen, T.; Hallikainen, M.; Alhainen, K.; Soininen, H.; Tuomilehto, J.; Nissinen, A. Midlife vascular risk factors and Alzheimer’s disease in later life: Longitudinal, population based study. BMJ 2001, 322, 1447–1451. [Google Scholar] [CrossRef] [PubMed]
  74. Solomon, A.; Kivipelto, M.; Wolozin, B.; Zhou, J.; Whitmer, R.A. Midlife serum cholesterol and increased risk of Alzheimer’s and vascular dementia three decades later. Dement. Geriatr. Cogn. Disord. 2009, 28, 75–80. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Berkowitz, C.L.; Mosconi, L.; Scheyer, O.; Rahman, A.; Hristov, H.; Isaacson, R.S. Precision Medicine for Alzheimer’s Disease Prevention. Healthcare 2018, 6, 82. https://doi.org/10.3390/healthcare6030082

AMA Style

Berkowitz CL, Mosconi L, Scheyer O, Rahman A, Hristov H, Isaacson RS. Precision Medicine for Alzheimer’s Disease Prevention. Healthcare. 2018; 6(3):82. https://doi.org/10.3390/healthcare6030082

Chicago/Turabian Style

Berkowitz, Cara L., Lisa Mosconi, Olivia Scheyer, Aneela Rahman, Hollie Hristov, and Richard S. Isaacson. 2018. "Precision Medicine for Alzheimer’s Disease Prevention" Healthcare 6, no. 3: 82. https://doi.org/10.3390/healthcare6030082

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop