Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Sex differences in Alzheimer disease — the gateway to precision medicine

Abstract

Alzheimer disease (AD) is characterized by wide heterogeneity in cognitive and behavioural syndromes, risk factors and pathophysiological mechanisms. Addressing this phenotypic variation will be crucial for the development of precise and effective therapeutics in AD. Sex-related differences in neural anatomy and function are starting to emerge, and sex might constitute an important factor for AD patient stratification and personalized treatment. Although the effects of sex on AD epidemiology are currently the subject of intense investigation, the notion of sex-specific clinicopathological AD phenotypes is largely unexplored. In this Review, we critically discuss the evidence for sex-related differences in AD symptomatology, progression, biomarkers, risk factor profiles and treatment. The cumulative evidence reviewed indicates sex-specific patterns of disease manifestation as well as sex differences in the rates of cognitive decline and brain atrophy, suggesting that sex is a crucial variable in disease heterogeneity. We discuss critical challenges and knowledge gaps in our current understanding. Elucidating sex differences in disease phenotypes will be instrumental in the development of a ‘precision medicine’ approach in AD, encompassing individual, multimodal, biomarker-driven and sex-sensitive strategies for prevention, detection, drug development and treatment.

Key points

  • Men and women with Alzheimer disease (AD) exhibit different cognitive and psychiatric symptoms, and women show faster cognitive decline after diagnosis of mild cognitive impairment (MCI) or AD dementia.

  • Levels of amyloid-β measured with PET-based brain imaging and with biochemical analysis of cerebrospinal fluid do not differ between the sexes.

  • Brain atrophy rates and patterns differ along the AD continuum between the sexes; in MCI, brain atrophy is faster in women than in men.

  • The prevalence and effects of cerebrovascular, metabolic and socio-economic risk factors for AD are different between men and women.

  • No data are available on sex differences in the efficacy and safety of drugs used in recently completed phase III clinical trials for mild to moderate AD.

  • Systematic studying and reporting of sex differences in disease symptomatology, biomarkers, progression, risk factors and treatment responses will be crucial for the development and implementation of precision medicine in AD.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Brain atrophy in women and men with amnestic mild cognitive impairment.
Fig. 2: Implications of AD sex differences for clinical practice.

Similar content being viewed by others

References

  1. World Health Organization and Alzheimer’s Disease International. Dementia: a public health priority. WHO http://www.who.int/mental_health/publications/dementia_report_2012/en/ (2012).

  2. Prince, M. Wimo, A., Guerchet, M., Ali, G. C., Wu, Y. & Prina, A. M. World Alzheimer Report 2015: the global impact of dementia. An anlaysis of prevalence, incidence, costs and trends. Alzheimer’s Disease International https://www.alz.co.uk/research/WorldAlzheimerReport2015.pdf (2015).

  3. Gauthier, S. et al. Why has therapy development for dementia failed in the last two decades? Alzheimers Dement. 12, 60–64 (2016).

    Article  PubMed  Google Scholar 

  4. Husain, M. Alzheimer’s disease: time to focus on the brain, not just molecules. Brain 140, 251–253 (2017).

    Article  PubMed  Google Scholar 

  5. de Bono, J. S. & Ashworth, A. Translating cancer research into targeted therapeutics. Nature 467, 543–549 (2010).

    Article  PubMed  CAS  Google Scholar 

  6. Vargas, A. J. & Harris, C. C. Biomarker development in the precision medicine era: lung cancer as a case study. Nat. Rev. Cancer 16, 525–537 (2016).

    Article  PubMed  CAS  Google Scholar 

  7. Qian, J., Hyman, B. T. & Betensky, R. A. Neurofibrillary tangle stage and the rate of progression of Alzheimer symptoms: modeling using an autopsy cohort and application to clinical trial design. JAMA Neurol. 74, 540–548 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gamberger, D., Lavrac, N., Srivatsa, S., Tanzi, R. E. & Doraiswamy, P. M. Identification of clusters of rapid and slow decliners among subjects at risk for Alzheimer’s disease. Sci. Rep. 7, 6763 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Escott-Price, V., Myers, A. J., Huentelman, M. & Hardy, J. Polygenic risk score analysis of pathologically confirmed Alzheimer disease. Ann. Neurol. 82, 311–314 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ruigrok, A. N. et al. A meta-analysis of sex differences in human brain structure. Neurosci. Biobehav. Rev. 39, 34–50 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Ingalhalikar, M. et al. Sex differences in the structural connectome of the human brain. Proc. Natl Acad. Sci. USA 111, 823–828 (2014).

    Article  PubMed  CAS  Google Scholar 

  12. Li, R. & Singh, M. Sex differences in cognitive impairment and Alzheimer’s disease. Front. Neuroendocrinol. 35, 385–403 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Cordonnier, C. et al. Stroke in women — from evidence to inequalities. Nat. Rev. Neurol. 13, 521–532 (2017). This paper provides a clear summary of the role of sex differences in clinical practice in the stroke field.

    Article  PubMed  Google Scholar 

  14. Szewczyk-Krolikowski, K. et al. The influence of age and gender on motor and non-motor features of early Parkinson’s disease: initial findings from the Oxford Parkinson Disease Center (OPDC) discovery cohort. Parkinsonism Relat. Disord. 20, 99–105 (2014).

    Article  PubMed  Google Scholar 

  15. Vetvik, K. G. & MacGregor, E. A. Sex differences in the epidemiology, clinical features, and pathophysiology of migraine. Lancet Neurol. 16, 76–87 (2017).

    Article  PubMed  CAS  Google Scholar 

  16. Liu, G. et al. Prediction of cognition in Parkinson’s disease with a clinical-genetic score: a longitudinal analysis of nine cohorts. Lancet Neurol. 16, 620–629 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Hampel, H. et al. Precision pharmacology for Alzheimer’s disease. Pharmacol. Res. 130, 331–365 (2018).

    Article  PubMed  CAS  Google Scholar 

  18. Snyder, H. M. et al. Sex biology contributions to vulnerability to Alzheimer’s disease: A think tank convened by the Women’s Alzheimer’s Research Initiative. Alzheimers Dement. 12, 1186–1196 (2016).

    Article  PubMed  Google Scholar 

  19. Pike, C. J. Sex and the development of Alzheimer’s disease. J. Neurosci. Res. 95, 671–680 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Mielke, M. M., Vemuri, P. & Rocca, W. A. Clinical epidemiology of Alzheimer’s disease: assessing sex and gender differences. Clin. Epidemiol. 6, 37–48 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Rocca, W. A. Time, sex, gender, history, and dementia. Alzheimer Dis. Assoc. Disord. 31, 76–79 (2017). This paper highlights the current debate in the field of AD epidemiology and the potential role of secular trends in some controversial results.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Gale, S. D., Baxter, L. & Thompson, J. Greater memory impairment in dementing females than males relative to sex-matched healthy controls. J. Clin. Exp. Neuropsychol. 38, 527–533 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Jack, C. R. Jr. et al. Age, sex, and APOE epsilon4 effects on memory, brain structure, and beta-amyloid across the adult life span. JAMA Neurol. 72, 511–519 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. McCarrey, A. C., An, Y., Kitner-Triolo, M. H., Ferrucci, L. & Resnick, S. M. Sex differences in cognitive trajectories in clinically normal older adults. Psychol. Aging 31, 166–175 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Laws, K. R., Irvine, K. & Gale, T. M. Sex differences in cognitive impairment in Alzheimer’s disease. World J. Psychiatry 6, 54–65 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Sundermann, E. E. et al. Better verbal memory in women than men in MCI despite similar levels of hippocampal atrophy. Neurology 86, 1368–1376 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sundermann, E. E. et al. Female advantage in verbal memory: evidence of sex-specific cognitive reserve. Neurology 87, 1916–1924 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Irvine, K., Laws, K. R., Gale, T. M. & Kondel, T. K. Greater cognitive deterioration in women than men with Alzheimer’s disease: a meta analysis. J. Clin. Exp. Neuropsychol 34, 989–998 (2012). This article is a useful meta-analysis that demonstrated the occurrence of sex differences in cognitive decline in AD.

    Article  PubMed  Google Scholar 

  29. Pusswald, G. et al. Gender-specific differences in cognitive profiles of patients with Alzheimer’s disease: results of the Prospective Dementia Registry Austria (PRODEM-Austria). J. Alzheimers Dis. 46, 631–637 (2015).

    Article  PubMed  Google Scholar 

  30. Benke, T. et al. Cognition, gender, and functional abilities in Alzheimer’s disease: how are they related? J. Alzheimers Dis. 35, 247–252 (2013).

    Article  PubMed  Google Scholar 

  31. Holland, D., Desikan, R. S., Dale, A. M. & McEvoy, L. K., Alzheimer’s Disease Neuroimaging Initiative. Higher rates of decline for women and apolipoprotein E epsilon4 carriers. AJNR. Am. J. Neuroradiol. 34, 2287–2293 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Lin, K. A. et al. Marked gender differences in progression of mild cognitive impairment over 8 years. Alzheimers Dement. 1, 103–110 (2015). This article provides strong evidence from the ADNI cohort of faster cognitive decline in women with MCI than in men with MCI.

    Google Scholar 

  33. Tifratene, K., Robert, P., Metelkina, A., Pradier, C. & Dartigues, J. F. Progression of mild cognitive impairment to dementia due to AD in clinical settings. Neurology 85, 331–338 (2015).

    Article  PubMed  Google Scholar 

  34. Pradier, C. et al. The mini mental state examination at the time of Alzheimer’s disease and related disorders diagnosis, according to age, education, gender and place of residence: a cross-sectional study among the French National Alzheimer database. PLoS ONE 9, e103630 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Leening, M. J. et al. Sex differences in lifetime risk and first manifestation of cardiovascular disease: prospective population based cohort study. BMJ 349, g5992 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Karoglu, E. T. et al. Aging alters the molecular dynamics of synapses in a sexually dimorphic pattern in zebrafish (Danio rerio). Neurobiol. Aging 54, 10–21 (2017).

    Article  PubMed  CAS  Google Scholar 

  37. Counts, S. E. et al. Cerebrospinal fluid proNGF: a putative biomarker for early Alzheimer’s disease. Curr. Alzheimer Res. 13, 800–808 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Walker, K. A. et al. Midlife systemic inflammatory markers are associated with late-life brain volume: the ARIC study. Neurology 89, 2262–2270 (2017).

    Article  PubMed  Google Scholar 

  39. Schwarz, J. M., Sholar, P. W. & Bilbo, S. D. Sex differences in microglial colonization of the developing rat brain. J. Neurochem. 120, 948–963 (2012).

    PubMed  PubMed Central  CAS  Google Scholar 

  40. Lenz, K. M., Nugent, B. M., Haliyur, R. & McCarthy, M. M. Microglia are essential to masculinization of brain and behavior. J. Neurosci. 33, 2761–2772 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Villa, A. et al. Sex-specific features of microglia from adult mice. Cell Rep. 23, 3501–3511 (2018).

  42. Ott, B. R., Tate, C. A., Gordon, N. M. & Heindel, W. C. Gender differences in the behavioral manifestations of Alzheimer’s disease. J. Am. Geriatr. Soc. 44, 583–587 (1996).

    Article  PubMed  CAS  Google Scholar 

  43. Mega, M. S., Cummings, J. L., Fiorello, T. & Gornbein, J. The spectrum of behavioral changes in Alzheimer’s disease. Neurology 46, 130–135 (1996).

    Article  PubMed  CAS  Google Scholar 

  44. Ott, B. R., Lapane, K. L. & Gambassi, G. Gender differences in the treatment of behavior problems in Alzheimer’s disease. SAGE Study Group. System. Assess. Geriatr. Drug Epidemiol. Neurol. 54, 427–432 (2000).

    CAS  Google Scholar 

  45. Kitamura, T., Kitamura, M., Hino, S., Tanaka, N. & Kurata, K. Gender differences in clinical manifestations and outcomes among hospitalized patients with behavioral and psychological symptoms of dementia. J. Clin. Psychiatry 73, 1548–1554 (2012).

    Article  PubMed  Google Scholar 

  46. Teri, L., Borson, S., Kiyak, H. A. & Yamagishi, M. Behavioral disturbance, cognitive dysfunction, and functional skill. Prevalence and relationship in Alzheimer’s disease. J. Am. Geriatr. Soc. 37, 109–116 (1989).

    Article  PubMed  CAS  Google Scholar 

  47. Karttunen, K. et al. Neuropsychiatric symptoms and quality of life in patients with very mild and mild Alzheimer’s disease. Int. J. Geriatr. Psychiatry 26, 473–482 (2011).

    Article  PubMed  Google Scholar 

  48. Spalletta, G. et al. Neuropsychiatric symptoms and syndromes in a large cohort of newly diagnosed, untreated patients with Alzheimer disease. Am. J. Geriatr. Psychiatry 18, 1026–1035 (2010).

    Article  PubMed  Google Scholar 

  49. Hollingworth, P. et al. Four components describe behavioral symptoms in 1,120 individuals with late-onset Alzheimer’s disease. J. Am. Geriatr. Soc. 54, 1348–1354 (2006). This paper presents the largest study available documenting sex differences in psychiatric symptoms of AD.

    Article  PubMed  Google Scholar 

  50. Sinforiani, E. et al. Impact of gender differences on the outcome of Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 30, 147–154 (2010).

    Article  PubMed  CAS  Google Scholar 

  51. Jack, C. R. Jr. et al. Age-specific and sex-specific prevalence of cerebral beta-amyloidosis, tauopathy, and neurodegeneration in cognitively unimpaired individuals aged 50–95 years: a cross-sectional study. Lancet Neurol. 16, 435–444 (2017). This landmark study documents sex differences in AD biomarkers across the lifespan.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Scheinin, N. M. et al. Cortical (1)(1)C-PIB uptake is associated with age, APOE genotype, and gender in “healthy aging”. J. Alzheimers Dis. 41, 193–202 (2014).

    Article  PubMed  CAS  Google Scholar 

  53. Cavedo, E. et al. Sex differences in functional and molecular neuroimaging biomarkers of Alzheimer’s disease in a mono-center cohort of cognitively normal older adults with subjective memory complaints. Alzheimers Dement. (in the press).

  54. Gottesman, R. F. et al. The ARIC-PET amyloid imaging study: brain amyloid differences by age, race, sex, and APOE. Neurology 87, 473–480 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Vemuri, P. et al. Evaluation of amyloid protective factors and Alzheimer disease neurodegeneration protective factors in elderly individuals. JAMA Neurol. 74, 718–726 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Barnes, L. L. et al. Sex differences in the clinical manifestations of Alzheimer disease pathology. Arch. Gen. Psychiatry 62, 685–691 (2005). This landmark study examines for the first time sex differences in the clinical manifestation resulting from the accumulation of amyloid plaques and tangles in the brain.

    Article  PubMed  Google Scholar 

  57. Shinohara, M. et al. Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer’s disease. Acta Neuropathol. 132, 225–234 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Mattsson, N. et al. Clinical validity of cerebrospinal fluid Abeta42, tau, and phospho-tau as biomarkers for Alzheimer’s disease in the context of a structured 5-phase development framework. Neurobiol. Aging 52, 196–213 (2017).

    Article  PubMed  CAS  Google Scholar 

  59. Jansen, W. J. et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-nalysis. JAMA 313, 1924–1938 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Salehi, A., Gonzalez Martinez, V. & Swaab, D. F. A sex difference and no effect of ApoE type on the amount of cytoskeletal alterations in the nucleus basalis of Meynert in Alzheimer’s disease. Neurobiol. Aging 19, 505–510 (1998).

    Article  PubMed  CAS  Google Scholar 

  61. Johnson, K. A. et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann. Neurol. 79, 110–119 (2016).

    Article  PubMed  Google Scholar 

  62. Apostolova, L. G. et al. 3D comparison of hippocampal atrophy in amnestic mild cognitive impairment and Alzheimer’s disease. Brain 129, 2867–2873 (2006).

    Article  PubMed  Google Scholar 

  63. Perlaki, G. et al. Are there any gender differences in the hippocampus volume after head-size correction? A volumetric and voxel-based morphometric study. Neurosci. Lett. 570, 119–123 (2014).

    Article  PubMed  CAS  Google Scholar 

  64. Skup, M. et al. Sex differences in grey matter atrophy patterns among AD and aMCI patients: results from ADNI. Neuroimage 56, 890–906 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Hua, X. et al. Sex and age differences in atrophic rates: an ADNI study with n = 1368 MRI scans. Neurobiol. Aging 31, 1463–1480 (2010). This landmark paper shows the faster atrophic rate in women enrolled in the ADNI cohort.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Ardekani, B. A., Convit, A. & Bachman, A. H. Analysis of the MIRIAD data shows sex differences in hippocampal atrophy progression. J. Alzheimers Dis. 50, 847–857 (2016).

    Article  PubMed  Google Scholar 

  67. Koran, M. E., Wagener, M. & Hohman, T. J., Alzheimer’s Neuroimaging Initiative. Sex differences in the association between AD biomarkers and cognitive decline. Brain Imaging Behav. 11, 205–213 (2016).

    Article  Google Scholar 

  68. Karch, A. et al. Stratification by genetic and demographic characteristics improves diagnostic accuracy of cerebrospinal fluid biomarkers in rapidly progressive dementia. J. Alzheimers Dis. 54, 1385–1393 (2016).

    Article  PubMed  CAS  Google Scholar 

  69. Madsen, T. E. et al. Sex-specific stroke incidence over time in the Greater Cincinnati/Northern Kentucky Stroke Study. Neurology 89, 990–996 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Gibson, C. L. Cerebral ischemic stroke: is gender important? J. Cereb. Blood Flow Metab. 33, 1355–1361 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Longstreth, W. T. Jr. et al. Associations between microinfarcts and other macroscopic vascular findings on neuropathologic examination in 2 databases. Alzheimer Dis. Assoc. Disord. 23, 291–294 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  72. National Institute of Mental Health. Major depression. NIMH https://www.nimh.nih.gov/health/statistics/major-depression.shtml (2015).

  73. Mallampalli, M. P. & Carter, C. L. Exploring sex and gender differences in sleep health: a Society for Women’s Health Research Report. J. Womens Health (Larchmt) 23, 553–562 (2014).

    Article  Google Scholar 

  74. Farrer, L. A. et al. 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 278, 1349–1356 (1997).

    Article  PubMed  CAS  Google Scholar 

  75. Altmann, A., Tian, L., Henderson, V. W. & Greicius, M. D., Alzheimer’s Disease Neuroimaging Initiative Investigators. Sex modifies the APOE-related risk of developing Alzheimer disease. Ann. Neurol. 75, 563–573 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Kim, S. et al. Gender differences in risk factors for transition from mild cognitive impairment to Alzheimer’s disease: a CREDOS study. Compr. Psychiatry 62, 114–122 (2015).

    Article  PubMed  Google Scholar 

  77. Neu, S. C. et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 74, 1178–1189 (2017). This highly powered meta-analysis refines our understanding of sex–APOE interactions in AD risk.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Mosconi, L. et al. Perimenopause and emergence of an Alzheimer’s bioenergetic phenotype in brain and periphery. PLoS ONE 12, e0185926 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Ungar, L., Altmann, A. & Greicius, M. D. Apolipoprotein E, gender, and Alzheimer’s disease: an overlooked, but potent and promising interaction. Brain Imaging Behav. 8, 262–273 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Damoiseaux, J. S. et al. Gender modulates the APOE epsilon4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levels. J. Neurosci. 32, 8254–8262 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Heise, V. et al. Apolipoprotein E genotype, gender and age modulate connectivity of the hippocampus in healthy adults. Neuroimage 98, 23–30 (2014).

    Article  PubMed  CAS  Google Scholar 

  82. Sampedro, F. et al. APOE-by-sex interactions on brain structure and metabolism in healthy elderly controls. Oncotarget 6, 26663–26674 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Buckley, R. F. et al. Sex, amyloid, and APOE ε4 and risk of cognitive decline in preclinical Alzheimer’s disease: findings from three well-characterized cohorts. Alzheimers Dement. https://doi.org/10.1016/j.jalz.2018.04.010 (2018).

  84. Hohman, T. J. et al. Sex-specific association of Apolipoprotein E with cerebrospinal fluid levels of tau. JAMA Neurol. https://doi.org/10.1001/jamaneurol.2018.0821 (2018).

  85. Snowdon, D. A. et al. Brain infarction and the clinical expression of Alzheimer disease. The Nun Study. JAMA 277, 813–817 (1997).

    Article  PubMed  CAS  Google Scholar 

  86. Vemuri, P. et al. Vascular and amyloid pathologies are independent predictors of cognitive decline in normal elderly. Brain 138, 761–771 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  87. Roberts, R. O. et al. Association of diabetes with amnestic and nonamnestic mild cognitive impairment. Alzheimers Dement. 10, 18–26 (2014).

    Article  PubMed  Google Scholar 

  88. Gilsanz, P. et al. Female sex, early-onset hypertension, and risk of dementia. Neurology 89, 1886–1893 (2017).

    Article  PubMed  Google Scholar 

  89. Lorius, N. et al. Vascular disease and risk factors are associated with cognitive decline in the alzheimer disease spectrum. Alzheimer Dis. Assoc. Disord. 29, 18–25 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Li, J. et al. Vascular risk factors promote conversion from mild cognitive impairment to Alzheimer disease. Neurology 76, 1485–1491 (2011).

    Article  PubMed  CAS  Google Scholar 

  91. Sachdev, P. S. et al. Risk profiles for mild cognitive impairment vary by age and sex: the Sydney Memory and Ageing study. Am. J. Geriatr. Psychiatry 20, 854–865 (2012).

    Article  PubMed  Google Scholar 

  92. Sundermann, E. E., Katz, M. J. & Lipton, R. B. Sex differences in the relationship between depressive symptoms and risk of amnestic mild cognitive impairment. Am. J. Geriatr. Psychiatry 25, 13–22 (2017).

    Article  PubMed  Google Scholar 

  93. Pankratz, V. S. et al. Predicting the risk of mild cognitive impairment in the Mayo Clinic Study of Aging. Neurology 84, 1433–1442 (2015). This landmark paper specifically examines sex differences in MCI risk.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Artero, S. et al. Risk profiles for mild cognitive impairment and progression to dementia are gender specific. J. Neurol. Neurosurg. Psychiatry 79, 979–984 (2008).

    Article  PubMed  CAS  Google Scholar 

  95. Hayden, K. M. et al. Vascular risk factors for incident Alzheimer disease and vascular dementia: the Cache County study. Alzheimer Dis. Assoc. Disord. 20, 93–100 (2006).

    Article  PubMed  Google Scholar 

  96. Chene, G. et al. Gender and incidence of dementia in the Framingham Heart Study from mid-adult life. Alzheimers Dement. 11, 310–320 (2015).

    Article  PubMed  Google Scholar 

  97. Brown, M. C. et al. Cardiovascular disease risk in women with pre-eclampsia: systematic review and meta-analysis. Eur. J. Epidemiol. 28, 1–19 (2013).

    Article  PubMed  Google Scholar 

  98. Fields, J. A. et al. Preeclampsia and cognitive impairment later in life. Am J Obstet. Gynecol 217, 74.e1–74.e11 (2017).

    Article  Google Scholar 

  99. Buhimschi, I. A. et al. Protein misfolding, congophilia, oligomerization, and defective amyloid processing in preeclampsia. Sci. Transl Med. 6, 245ra292 (2014).

    Article  CAS  Google Scholar 

  100. Rocca, W. A. et al. Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause. Neurology 69, 1074–1083 (2007).

    Article  PubMed  CAS  Google Scholar 

  101. Bove, R. et al. Age at surgical menopause influences cognitive decline and Alzheimer pathology in older women. Neurology 82, 222–229 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  102. Norton, S., Matthews, F. E., Barnes, D. E., Yaffe, K. & Brayne, C. Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol. 13, 788–794 (2014).

    Article  PubMed  Google Scholar 

  103. American Association of University Women. The simple truth about the gender pay gap. AAUW https://www.aauw.org/aauw_check/pdf_download/show_pdf.php?file=The-Simple-Truth (2018).

  104. Brown, J. E., Rhee, A., Saad-Lessler, J. & Oakley, D. Shortchanged in retirement, the continuing challenges to women’s financial future. National Institute on Retirement Security https://www.nirsonline.org/wp-content/uploads/2017/06/final_shortchanged_retirement_report_2016.pdf (2016).

  105. Prince, M. et al. Dementia incidence and mortality in middle-income countries, and associations with indicators of cognitive reserve: a 10/66 Dementia Research Group population-based cohort study. Lancet 380, 50–58 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Swinkels, J., Tilburg, T. V., Verbakel, E. & Broese van Groenou, M. Explaining the gender gap in the caregiving burden of partner caregivers. J. Gerontol. B Psychol. Sci. Soc. Sci. https://doi.org/10.1093/geronb/gbx036 (2017).

  107. Alzheimer’s Association. 2014 Alzheimer’s disease facts and figures. Alzheimers Dement. 10, e47–e92 (2014).

    Article  Google Scholar 

  108. Sharma, N., Chakrabarti, S. & Grover, S. Gender differences in caregiving among family — caregivers of people with mental illnesses. World J. Psychiatry 6, 7–17 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Stahl, S. T., Beach, S. R., Musa, D. & Schulz, R. Living alone and depression: the modifying role of the perceived neighborhood environment. Aging Ment. Health 21, 1065–1071 (2017).

    Article  PubMed  Google Scholar 

  110. Doody, R. S. et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N. Engl. J. Med. 369, 341–350 (2013).

    Article  PubMed  CAS  Google Scholar 

  111. Egan, M. F. et al. Randomized trial of verubecestat for mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 378, 1691–1703 (2018).

    Article  PubMed  CAS  Google Scholar 

  112. Doody, R. S. et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 370, 311–321 (2014).

    Article  PubMed  CAS  Google Scholar 

  113. Salloway, S. et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 370, 322–333 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Religa, D. et al. Dementia diagnosis differs in men and women and depends on age and dementia severity: data from SveDem, the Swedish Dementia Quality Registry. Dement. Geriatr. Cogn. Disord. 33, 90–95 (2012).

    Article  PubMed  Google Scholar 

  115. Ferris, S. et al. Effects of gender on response to treatment with rivastigmine in mild cognitive impairment: a post hoc statistical modeling approach. Gend. Med. 6, 345–355 (2009).

    Article  PubMed  Google Scholar 

  116. MacGowan, S. H., Wilcock, G. K. & Scott, M. Effect of gender and apolipoprotein E genotype on response to anticholinesterase therapy in Alzheimer’s disease. Int. J. Geriatr. Psychiatry 13, 625–630 (1998).

    Article  PubMed  CAS  Google Scholar 

  117. Canevelli, M. et al. Sex and gender differences in the treatment of Alzheimer’s disease: a systematic review of randomized controlled trials. Pharmacol. Res. 115, 218–223 (2017). This systematic review of randomized, double-blind trials with cholinesterase inhibitors and memantine reveals that only 4% of studies examined sex effects in their data sets.

    Article  PubMed  CAS  Google Scholar 

  118. Haywood, W. M. & Mukaetova-Ladinska, E. B. Sex influences on cholinesterase inhibitor treatment in elderly individuals with Alzheimer’s disease. Am. J. Geriatr. Pharmacother. 4, 273–286 (2006).

    Article  PubMed  CAS  Google Scholar 

  119. Davis, M. L. & Barrett, A. M. Selective benefit of donepezil on oral naming in Alzheimer’s disease in men compared to women. CNS Spectr. 14, 175–176 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Buccafusco, J. J., Jackson, W. J., Stone, J. D. & Terry, A. V. Sex dimorphisms in the cognitive-enhancing action of the Alzheimer’s drug donepezil in aged Rhesus monkeys. Neuropharmacology 44, 381–389 (2003).

    Article  PubMed  CAS  Google Scholar 

  121. Scacchi, R., Gambina, G., Broggio, E. & Corbo, R. M. Sex and ESR1 genotype may influence the response to treatment with donepezil and rivastigmine in patients with Alzheimer’s disease. Int. J. Geriatr. Psychiatry 29, 610–615 (2014).

    Article  PubMed  Google Scholar 

  122. Wattmo, C., Londos, E. & Minthon, L. Risk factors that affect life expectancy in Alzheimer’s disease: a 15-year follow-up. Dement. Geriatr. Cogn. Disord. 38, 286–299 (2014).

    Article  PubMed  Google Scholar 

  123. Rhodes, M. E. & Rubin, R. T. Functional sex differences (‘sexual diergism’) of central nervous system cholinergic systems, vasopressin, and hypothalamic-pituitary-adrenal axis activity in mammals: a selective review. Brain Res. Brain Res. Rev. 30, 135–152 (1999).

    Article  PubMed  CAS  Google Scholar 

  124. Counts, S. E., Che, S., Ginsberg, S. D. & Mufson, E. J. Gender differences in neurotrophin and glutamate receptor expression in cholinergic nucleus basalis neurons during the progression of Alzheimer’s disease. J. Chem. Neuroanat. 42, 111–117 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Wang, R. H., Bejar, C. & Weinstock, M. Gender differences in the effect of rivastigmine on brain cholinesterase activity and cognitive function in rats. Neuropharmacology 39, 497–506 (2000).

    Article  PubMed  CAS  Google Scholar 

  126. Smith, C. D., Wright, L. K., Garcia, G. E., Lee, R. B. & Lumley, L. A. Hormone-dependence of sarin lethality in rats: sex differences and stage of the estrous cycle. Toxicol. Appl. Pharmacol. 287, 253–257 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Venerosi, A., Ricceri, L., Tait, S. & Calamandrei, G. Sex dimorphic behaviors as markers of neuroendocrine disruption by environmental chemicals: the case of chlorpyrifos. Neurotoxicology 33, 1420–1426 (2012).

    Article  PubMed  CAS  Google Scholar 

  128. Alves-Amaral, G., Pires-Oliveira, M., Andrade-Lopes, A. L., Chiavegatti, T. & Godinho, R. O. Gender-related differences in circadian rhythm of rat plasma acetyl- and butyrylcholinesterase: effects of sex hormone withdrawal. Chem. Biol. Interact. 186, 9–15 (2010).

    Article  PubMed  CAS  Google Scholar 

  129. Mehta, N. et al. Systematic review of sex-specific reporting of data: cholinesterase inhibitor example. J. Am. Geriatr. Soc. 65, 2213–2219 (2017).

    Article  PubMed  Google Scholar 

  130. Hampel, H. et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 141, 1917–1933 (2018).

    Article  PubMed  Google Scholar 

  131. Ezio, G. & Giancarlo P. Sex and gender differences in the brain cholinergic system and in the response to therapy of Alzheimer disease with cholinesterase inhibitors. Curr. Alzheimer Res. https://doi.org/10.2174/1567205015666180613111504 (2018).

    Article  PubMed  Google Scholar 

  132. Moga, D. C. et al. A comparison of sex differences in psychotropic medication use in older people with Alzheimer’s disease in the US and Finland. Drugs Aging 34, 55–65 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Cooper, C. et al. Inequalities in receipt of mental and physical healthcare in people with dementia in the UK. Age Ageing 46, 393–400 (2017).

    Article  PubMed  Google Scholar 

  134. Cojutti, P., Arnoldo, L., Cattani, G., Brusaferro, S. & Pea, F. Polytherapy and the risk of potentially inappropriate prescriptions (PIPs) among elderly and very elderly patients in three different settings (hospital, community, long-term care facilities) of the Friuli Venezia Giulia region, Italy: are the very elderly at higher risk of PIPs? Pharmacoepidemiol. Drug Saf. 25, (1070–1078 (2016).

    Google Scholar 

  135. Henley, D. B., May, P. C., Dean, R. A. & Siemers, E. R. Development of semagacestat (LY450139), a functional gamma-secretase inhibitor, for the treatment of Alzheimer’s disease. Expert Opin. Pharmacother. 10, 1657–16642 (2009).

    Article  PubMed  CAS  Google Scholar 

  136. Farlow, M. et al. Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease. Alzheimers Dement. 8, 261–271 (2012).

    Article  PubMed  CAS  Google Scholar 

  137. Legato, M. J., Johnson, P. A. & Manson, J. E. Consideration of sex differences in medicine to improve health care and patient outcomes. JAMA 316, 1865–1866 (2016).

    Article  PubMed  Google Scholar 

  138. Berger, J. S. et al. Aspirin for the primary prevention of cardiovascular events in women and men: a sex-specific meta-analysis of randomized controlled trials. JAMA 295, 306–313 (2006).

    Article  PubMed  CAS  Google Scholar 

  139. Claxton, A. et al. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or Alzheimer’s disease. J. Alzheimers Dis. 35, 789–797 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Ngandu, T. et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet 385, 2255–2263 (2015).

    Article  PubMed  Google Scholar 

  141. Andrieu, S. et al. Effect of long-term omega 3 polyunsaturated fatty acid supplementation with or without multidomain intervention on cognitive function in elderly adults with memory complaints (MAPT): a randomised, placebo-controlled trial. Lancet Neurol. 16, 377–389 (2017).

    Article  PubMed  CAS  Google Scholar 

  142. Farlow, M. R. et al. Treatment outcome of tacrine therapy depends on apolipoprotein genotype and gender of the subjects with Alzheimer’s disease. Neurology 50, 669–677 (1998). This landmark paper suggested for the first time a sex–genotype interaction in the effect of tacrine.

    Article  PubMed  CAS  Google Scholar 

  143. Depypere, H., Vierin, A., Weyers, S. & Sieben, A. Alzheimer’s disease, apolipoprotein E and hormone replacement therapy. Maturitas 94, 98–105 (2016). This comprehensive review summarizes the current understanding of HRT and the future challenges.

    Article  PubMed  CAS  Google Scholar 

  144. Burkhardt, M. S. et al. Oestrogen replacement therapy may improve memory functioning in the absence of APOE epsilon4. J. Alzheimers Dis. 6, 221–228 (2004).

    Article  PubMed  CAS  Google Scholar 

  145. Yaffe, K., Haan, M., Byers, A., Tangen, C. & Kuller, L. Estrogen use, APOE, and cognitive decline: evidence of gene-environment interaction. Neurology 54, 1949–1954 (2000).

    Article  PubMed  CAS  Google Scholar 

  146. Ryan, J. et al. Characteristics of hormone therapy, cognitive function, and dementia: the prospective 3C Study. Neurology 73, 1729–1737 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Kang, J. H. & Grodstein, F. Postmenopausal hormone therapy, timing of initiation, APOE and cognitive decline. Neurobiol. Aging 33, 1129–1137 (2012).

    Article  PubMed  CAS  Google Scholar 

  148. Zandi, P. P. et al. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County Study. JAMA 288, 2123–2129 (2002).

    Article  PubMed  CAS  Google Scholar 

  149. Gleason, C. E. et al. Effects of hormone therapy on cognition and mood in recently postmenopausal women: findings from the randomized, controlled KEEPS-Cognitive and Affective Study. PLoS Med. 12, e1001833 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Lista, S. et al. Application of systems theory in longitudinal studies on the origin and progression of Alzheimer’s disease. Methods Mol. Biol. 1303, 49–67 (2016).

    Article  PubMed  Google Scholar 

  151. Kosik, K. S. Personalized medicine for effective Alzheimer disease treatment. JAMA Neurol. 72, 497–498 (2015).

    Article  PubMed  Google Scholar 

  152. Hampel, H. et al. A precision medicine initiative for Alzheimer’s disease: the road ahead to biomarker-guided integrative disease modeling. Climacteric 20, 107–118 (2017). This landmark paper describes the inception of the APMI.

    Article  PubMed  CAS  Google Scholar 

  153. Hampel, H. et al. PRECISION MEDICINE — the golden gate for detection, treatment and prevention of Alzheimer’s disease. J. Prev. Alzheimers Dis. 3, 243–259 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  154. Hebert, L. E., Weuve, J., Scherr, P. A. & Evans, D. A. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology 80, 1778–1783 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  155. Katz, M. J. et al. Age-specific and sex-specific prevalence and incidence of mild cognitive impairment, dementia, and Alzheimer dementia in blacks and whites: a report from the Einstein Aging Study. Alzheimer Dis. Assoc. Disord. 26, 335–343 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Roberts, R. O. et al. The incidence of MCI differs by subtype and is higher in men: the Mayo Clinic Study of Aging. Neurology 78, 342–351 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Lin, K. A. & Doraiswamy, P. M. When Mars versus Venus is not a cliche: gender differences in the neurobiology of Alzheimer’s disease. Front. Neurol. 5, 288 (2014).

    Article  PubMed  Google Scholar 

  158. Dubois, B. et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 13, 614–629 (2014).

    Article  PubMed  Google Scholar 

  159. Lyketsos, C. G. et al. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the cardiovascular health study. JAMA 288, 1475–1483 (2002).

    Article  PubMed  Google Scholar 

  160. Steinberg, M. et al. Vascular risk factors and neuropsychiatric symptoms in Alzheimer’s disease: the Cache County Study. Int. J. Geriatr. Psychiatry 29, 153–159 (2014).

    Article  PubMed  Google Scholar 

  161. Petersen, R. C. Mild cognitive impairment as a diagnostic entity. J. Intern. Med. 256, 183–194 (2004).

    Article  PubMed  CAS  Google Scholar 

  162. Wang, L. et al. Evaluation of tau imaging in staging Alzheimer disease and revealing interactions between beta-amyloid and tauopathy. JAMA Neurol. 73, 1070–1077 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  163. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research & Center for Biologics Evaluation and Research. Early Alzheimer’s disease: developing drugs for treatment. Guidance for industry. Draft guidance. FDA https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM596728.pdf (2018).

  164. European Medicines Agency. Guideline on the clinical investigation of medicines for the treatment of Alzheimer’s disease. EMA http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2018/02/WC500244609.pdf (2018).

  165. Standford University. Gendered innovations terminology. Gendered Innovations http://genderedinnovations.stanford.edu/terms.html (2018).

  166. Jary, D. & Jary, A. Collins Dictionary of Sociology 3rd edn. (Collins, Glasgow, 2005).

  167. World Health Organization. Gender. WHO http://www.who.int/en/news-room/fact-sheets/detail/gender (2015).

  168. Dubal, D. B., Broestl, L. & Worden, K. Sex and gonadal hormones in mouse models of Alzheimer’s disease: what is relevant to the human condition? Biol. Sex. Differ. 3, 24 (2012). This paper is a comprehensive overview of sex differences in the most widely used transgenic models of AD-like amyloidosis.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Middeldorp, J. et al. Preclinical assessment of young blood plasma for Alzheimer disease. JAMA Neurol. 73, 1325–1333 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  170. LaClair, K. D. et al. Treatment with bexarotene, a compound that increases apolipoprotein-E, provides no cognitive benefit in mutant APP/PS1 mice. Mol. Neurodegener. 8, 18 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Melnikova, T. et al. Sex-related dimorphism in dentate gyrus atrophy and behavioral phenotypes in an inducible tTa:APPsi transgenic model of Alzheimer’s disease. Neurobiol. Dis. 96, 171–185 (2016).

    Article  PubMed  CAS  Google Scholar 

  172. Granger, M. W. et al. A TgCRND8 mouse model of Alzheimer’s disease exhibits sexual dimorphisms in behavioral indices of cognitive reserve. J. Alzheimers Dis. 51, 757–773 (2016).

    Article  PubMed  CAS  Google Scholar 

  173. Lewis, J. et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 293, 1487–1491 (2001).

    Article  PubMed  CAS  Google Scholar 

  174. Oikawa, N., Ogino, K., Masumoto, T., Yamaguchi, H. & Yanagisawa, K. Gender effect on the accumulation of hyperphosphorylated tau in the brain of locus-ceruleus-injured APP-transgenic mouse. Neurosci. Lett. 468, 243–247 (2010).

    Article  PubMed  CAS  Google Scholar 

  175. Dumont, M. et al. Behavioral deficit, oxidative stress, and mitochondrial dysfunction precede tau pathology in P301S transgenic mice. FASEB J. 25, 4063–4072 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Yue, M., Hanna, A., Wilson, J., Roder, H. & Janus, C. Sex difference in pathology and memory decline in rTg4510 mouse model of tauopathy. Neurobiol. Aging 32, 590–603 (2011).

    Article  PubMed  CAS  Google Scholar 

  177. Bour, A. et al. Middle-aged human apoE4 targeted-replacement mice show retention deficits on a wide range of spatial memory tasks. Behav. Brain Res. 193, 174–182 (2008).

    Article  PubMed  CAS  Google Scholar 

  178. Reverte, I., Klein, A. B., Ratner, C., Domingo, J. L. & Colomina, M. T. Behavioral phenotype and BDNF differences related to apoE isoforms and sex in young transgenic mice. Exp. Neurol. 237, 116–125 (2012).

    Article  PubMed  CAS  Google Scholar 

  179. Rijpma, A. et al. Sex differences in presynaptic density and neurogenesis in middle-aged ApoE4 and ApoE knockout mice. J. Neurodegener Dis. 2013, 531326 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  180. Cacciottolo, M. et al. The APOE4 allele shows opposite sex bias in microbleeds and Alzheimer’s disease of humans and mice. Neurobiol. Aging 37, 47–57 (2016).

    Article  PubMed  CAS  Google Scholar 

  181. Hampel, H. et al. Revolution of Alzheimer precision neurology. Passageway of systems biology and neurophysiology. J. Alzheimers Dis. 64, S47–S105 (2018).

  182. Hampel, H. et al. Precision medicine and drug development in Alzheimer’s disease: the importance of sexual dimorphism and patient stratification. Front. Neuroendocrinol. https://doi.org/10.1016/j.yfrne.2018.06.001 (2018).

Download references

Acknowledgements

H.H. was supported by the AXA Research Fund, the Fondation Partenariale Sorbonne Université, the Fondation pour la Recherche sur Alzheimer, Paris, France and the programme ‘Investissements d’avenir’ (ANR-10-IAIHU-06; Agence Nationale de la Recherche-10-IA, Agence Institut Hospitalo-Universitaire-6; awarded to H.H.). Further support was provided by the Colam Initiatives and the Fondation pour la Recherche sur Alzheimer, Paris, France (awarded to H.H. and P.A.C.) and the programme ‘PHOENIX’, led by the Sorbonne University Foundation and sponsored by the Fondation pour la Recherche sur Alzheimer (awarded to H.H. and E.C.). H.G. acknowledges support from the Heart and Stroke Foundation of Canada, the Canadian Institutes of Health Research and the Canadian Foundation for Innovation. H.G. is also the holder of an investigator award from the Fonds de Recherche du Québec-Santé. M.T.F. is supported by a research fellowship by the Synapsis Foundation–Alzheimer Research Switzerland (ARS). M.F.I. acknowledges support from the Fonds de Recherche du Québec-Santé and from the Herbert H. Jasper Postdoctoral Research Fellowship from the Groupe de Recherche sur le Système Nerveux Central (GRSNC), Université de Montréal. The authors thank A. Kato (Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland) and L. Kulic (Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland) for encouragement and help with the first draft of the manuscript and A. Herrmann (Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK) for continuous support, insightful discussions and editorial work. The authors thank the contributors to the Alzheimer Precision Medicine Initiative Working Group (Supplementary Box 1). The initial idea and draft of this Review was conceived by the Women’s Brain Project (a non-profit organization advocating for women’s brain and mental health; www.womensbrainproject.com) as part of its advocacy and scientific activity.

Review criteria

We searched PubMed and Google Scholar for articles published in English without time limitations with the search terms “Alzheimer AND gender (or sex or women or female)”, “Amyloid AND gender (or sex or women or female)”, “plaques AND gender (or sex or women or female)”, “tau AND gender (or sex or women or female)”, “atrophy AND Alzheimer AND gender (or sex or women or female)”, “cognitive decline AND gender (or sex or women or female)”, “risk AND Alzheimer AND gender (or sex or women or female)”, “stroke AND Alzheimer AND gender (or sex or women or female)”, “cardiovascular AND Alzheimer AND gender (or sex or women or female)”, “cerebrovascular AND gender (or sex or women or female)”, “diabetes AND Alzheimer AND gender (or sex or women or female)”, “depression AND Alzheimer AND gender (or sex or women or female)” and “APOE AND Alzheimer AND gender (or sex or women or female)”. We also searched in the reference lists of identified articles for additional relevant publications. The final reference list was generated by choosing only papers published since 2012. Papers preceding 2012 were included only if considered by the authors to be landmark studies. Papers were selected on the basis of their perceived relevance to the topics covered in this Review.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

M.T.F., E.G. and H.H. conceived the paper. All authors contributed to the literature search and to the writing. M.T.F., E.C. and P.A.C. designed the figures. E.G., H.H., H.D., H.G. and S.M. provided guidance for specific areas of competence and overall paper design. A.C.S. contributed to the paper with her own expertise and points of view; the views and opinions expressed herein are those of the author and do not reflect the view of the Swiss Agency for Therapeutic Products (Swissmedic).

Corresponding author

Correspondence to Maria Teresa Ferretti.

Ethics declarations

Competing interests

H.H. is a Senior Associate Editor for the journal Alzheimer’s & Dementia. He has received fees for lecturing from Biogen and Roche; research grants from Pfizer, Avid, and MSD Avenir (all three paid to his institution); travel funding from Axovant, Eli Lilly, Functional Neuromodulation, GE Healthcare, Oryzon Genomics and Takeda and Zinfandel; and consultancy fees from Anavex, Axovant, Cytox, Functiona Neuromoduation, GE Healthcare, Jung Diagnostics, Oryzon Genomics and Takeda and Zinfandel. He participated in scientific advisory boards of Axovant, Cytox, Eli Lilly, Functional Neuromodulation, GE Healthcare, Oryzon Genomics, Roche Diagnostics and Takeda and Zinfandel. He is a co-inventor on several patents related to markers and the diagnosis of neurodegenerative disease (numbers 8916388, 8298784, 20120196300, 20100062463, 20100035286, 20090263822, 7547553, 20080206797, 20080199966 and 20080131921) but has received no royalties. All other authors declare no conflicts of interest.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

The Women’s Brain Project: www.womensbrainproject.com

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferretti, M., Iulita, M.F., Cavedo, E. et al. Sex differences in Alzheimer disease — the gateway to precision medicine. Nat Rev Neurol 14, 457–469 (2018). https://doi.org/10.1038/s41582-018-0032-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41582-018-0032-9

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research