• This record comes from PubMed

Alzheimer disease seen through the lens of sex and gender

. 2025 May ; 21 (5) : 235-249. [epub] 20250414

Language English Country Great Britain, England Media print-electronic

Document type Journal Article, Review

Links

PubMed 40229578
DOI 10.1038/s41582-025-01071-0
PII: 10.1038/s41582-025-01071-0
Knihovny.cz E-resources

Alzheimer disease (AD) is a life-limiting neurodegenerative disorder that disproportionately affects women. Indeed, sex and gender are emerging as crucial modifiers of diagnostic and therapeutic pathways in AD. This Review provides an overview of the interactions of sex and gender with important developments in AD and offers insights into priorities for future research to facilitate the development and implementation of personalized approaches in the shifting paradigm of AD care. In particular, this Review focuses on the influence of sex and gender on important advances in the treatment and diagnosis of AD, including disease-modifying therapies, fluid-based biomarkers, cognitive assessment tools and multidomain lifestyle interventional studies.

See more in PubMed

Ferretti, M. T. et al. Sex differences in Alzheimer disease — the gateway to precision medicine. Nat. Rev. Neurol 14, 457–469 (2018). PubMed DOI

Nebel, R. A. et al. Understanding the impact of sex and gender in Alzheimer’s disease: a call to action. Alzheimers Dement. 14, 1171–1183 (2018). PubMed DOI

Buckley, R. F. et al. Sex, amyloid, and APOEε4 and risk of cognitive decline in preclinical Alzheimer’s disease: findings from three well‐characterized cohorts. Alzheimers Dement. 14, 1193–1203 (2018). PubMed DOI

Buoncervello, M. et al. Preclinical models in the study of sex differences. Clin. Sci. 131, 449–469 (2017). DOI

Zhu, D., Montagne, A. & Zhao, Z. Alzheimer’s pathogenic mechanisms and underlying sex difference. Cell. Mol. Life Sci. 78, 4907–4920 (2021). PubMed DOI PMC

Ullah, M. F. et al. Impact of sex differences and gender specificity on behavioral characteristics and pathophysiology of neurodegenerative disorders. Neurosci. Biobehav. Rev. 102, 95–105 (2019). PubMed DOI

Petersen, E. et al. Feature robustness and sex differences in medical imaging: a case study in MRI-based Alzheimer’s disease detection. In Medical Image Computing and Computer Assisted Intervention — MICCAI 2022. Lecture Notes in Computer Science (eds Wang, L. et al.) 13431, 88–98 (Springer, 2022).

Arbel, I., Bingham, K. S. & Dawson, D. R. A scoping review of literature on sex and gender differences among dementia spousal caregivers. Gerontologist 59, e802–e815 (2019). PubMed DOI

Xiong, C. et al. Sex and gender differences in caregiving burden experienced by family caregivers of persons with dementia: a systematic review. PLoS ONE 15, e0231848 (2020). PubMed DOI PMC

Gove, D. et al. Sex, Gender and Sexuality in the Context of Dementia: A Discussion Paper https://www.alzheimer-europe.org/sites/default/files/2022-02/2022-02-25%20Alzheimer%20Europe%20ethics%20report%202021.pdf (Alzheimer Europe, 2022).

Livingston, G. et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 396, 413–446 (2020). PubMed DOI PMC

Podcasy, J. L. & Epperson, C. N. Considering sex and gender in Alzheimer disease and other dementias. Dialogues Clin. Neurosci. 18, 437–446 (2016). PubMed DOI PMC

May, J. T. & Harris, M. L. Health experiences of sexual and gender minority people living with dementia and their care partners: protocol for a scoping review. JMIR Res. Protoc. 12, e44918 (2023). PubMed DOI PMC

Madsen, T. E. et al. Article commentary: sex- and gender-based medicine: the need for precise terminology. Gender Genome 1, 122–128 (2017). DOI

Burton, A., Mohan, S., Puplampu-Dove, Y. & Tse, T. Characterizing gender eligibility descriptions for clinical trials registered on ClinicalTrials.gov. JAMA 330, 2019 (2023). PubMed DOI PMC

Heidari, S., Babor, T. F., De Castro, P., Tort, S. & Curno, M. Sex and gender equity in research: rationale for the SAGER guidelines and recommended use. Res. Integr. Peer Rev. 1, 2 (2016). PubMed DOI PMC

Gabelli, C. & Codemo, A. Gender differences in cognitive decline and Alzheimer’s disease. Ital. J. Gend.-Specif. Med. 1, 21–28 (2015).

Rebbeck, T. R., Mahal, B., Maxwell, K. N., Garraway, I. P. & Yamoah, K. The distinct impacts of race and genetic ancestry on health. Nat. Med. 28, 890–893 (2022). PubMed DOI

Huque, H. et al. Could country-level factors explain sex differences in dementia incidence and prevalence? A systematic review and meta-analysis. J. Alzheimers Dis. 91, 1231–1241 (2023). PubMed DOI PMC

Au, B., Dale-McGrath, S. & Tierney, M. C. Sex differences in the prevalence and incidence of mild cognitive impairment: a meta-analysis. Ageing Res. Rev. 35, 176–199 (2017). PubMed DOI

Peeters, G., Katelekha, K., Lawlor, B. & Demnitz, N. Sex differences in the incidence and prevalence of young‐onset Alzheimer’s disease: a meta‐analysis. Int. J. Geriatr. Psychiatry https://doi.org/10.1002/gps.5612 (2022).

Gustavsson, A. et al. Global estimates on the number of persons across the Alzheimer’s disease continuum. Alzheimers Dement. 19, 658–670 (2023). PubMed DOI

Patwardhan, V. et al. Differences across the lifespan between females and males in the top 20 causes of disease burden globally: a systematic analysis of the Global Burden of Disease Study 2021. Lancet Public Health 9, e282–e294 (2024). PubMed DOI PMC

Verghese, P. B., Castellano, J. M. & Holtzman, D. M. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol. 10, 241–252 (2011). PubMed DOI PMC

Altmann, A., Tian, L., Henderson, V. W. & Greicius, M. D. Sex modifies the APOE‐related risk of developing Alzheimer disease. Ann. Neurol. 75, 563–573 (2014). PubMed DOI PMC

Hohman, T. J. et al. Sex-specific association of apolipoprotein e with cerebrospinal fluid levels of tau. JAMA Neurol. 75, 989–998 (2018). PubMed DOI PMC

Neu, S. C. et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease. JAMA Neurol. 74, 1178–1189 (2017). PubMed DOI PMC

Corder, E. H. et al. The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism. Ann. N. Y. Acad. Sci. 1019, 24–28 (2004). PubMed DOI

Fratiglioni, L., Grut, M., Forsell, Y., Viitanen, M. & Winblad, B. Clinical diagnosis of Alzheimer’s disease and other dementias in a population survey. Agreement and causes of disagreement in applying Diagnostic and Statistical Manual of Mental Disorders, Revised Third Edition, criteria. Arch. Neurol. 49, 927–932 (1992). PubMed DOI

Sindi, S. et al. Midlife work-related stress increases dementia risk in later life: the CAIDE 30-year study. J. Gerontol. B Psychol. Sci. Soc. Sci. 72, 1044–1053 (2017). PubMed

Beckman, N., Waern, M., Gustafson, D. & Skoog, I. Secular trends in self reported sexual activity and satisfaction in Swedish 70 year olds: cross sectional survey of four populations, 1971–2001. Br. Med. J. 337, a279 (2008). DOI

Sindi, S. et al. Sex differences in dementia and response to a lifestyle intervention: evidence from Nordic population‐based studies and a prevention trial. Alzheimers Dement. 17, 1166–1178 (2021). PubMed DOI

Van Cauwenberghe, C., Van Broeckhoven, C. & Sleegers, K. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet. Med. 18, 421–430 (2016). PubMed DOI

Edwards, G. A. I. I. I., Gamez, N., Escobedo, G. Jr., Calderon, O. & Moreno-Gonzalez, I. Modifiable risk factors for Alzheimer’s disease. Front. Aging Neurosci. 11, 146 (2019). DOI

Zhang, X.-X. et al. The epidemiology of Alzheimer’s disease modifiable risk factors and prevention. J. Prev. Alzheimers Dis. 8, 313–321 (2021). PubMed DOI

Livingston, G. et al. Dementia prevention, intervention, and care: 2024 report of the Lancet standing commission. Lancet 404, 572–628 (2024). PubMed DOI

Mejia-Arango, S., Garcia-Cifuentes, E., Samper-Ternent, R., Borda, M. G. & Cano-Gutierrez, C. A. Socioeconomic disparities and gender inequalities in dementia: a community-dwelling population study from a middle-income country. J. Cross Cult. Gerontol. 36, 105–118 (2021). PubMed DOI

Fitzhugh, M. C. & Pa, J. Women with hearing loss show increased dementia risk and brain atrophy. Alzheimers Dement. 15, e12499 (2023).

Ilinca, S. & Suzuki, E. in Sex and Gender Differences in Alzheimer’s Disease (eds. Ferretti, M. T. et al.) 333–360 (Elsevier, 2021).

Arenaza-Urquijo, E. M. et al. Sex and gender differences in cognitive resilience to aging and Alzheimer’s disease. Alzheimers Dement. 20, 5695–5719 (2024). PubMed DOI PMC

Wadhwa, D. & Bonfert, A. Tracing global trends in education: a tale of old and new gender gaps. The World Bank https://genderdata.worldbank.org/en/data-stories/a-tale-of-old-and-new-gender-gaps (2024).

Mielke, M. M. et al. Consideration of sex and gender in Alzheimer’s disease and related disorders from a global perspective. Alzheimers Dement. 18, 2707–2724 (2022). PubMed DOI

Bloomberg, M. et al. Comparison of sex differences in cognitive function in older adults between high- and middle-income countries and the role of education: a population-based multicohort study. Age Ageing 52, afad019 (2023). PubMed DOI PMC

Geraets, A. F. J. & Leist, A. K. Sex/gender and socioeconomic differences in modifiable risk factors for dementia. Sci. Rep. 13, 80 (2023). PubMed DOI PMC

Klee, M., Leist, A. K., Veldsman, M., Ranson, J. M. & Llewellyn, D. J. Socioeconomic deprivation, genetic risk, and incident dementia. Am. J. Prev. Med. 64, 621–630 (2023). PubMed DOI PMC

Munoz Boudet, A. M. et al. Gender Differences in Poverty and Household Composition through the Life-Cycle: a Global Perspective. Policy Research Working Paper No. 8360 (World Bank, 2018); http://hdl.handle.net/10986/29426 .

Quintana, G. R. & Pfaus, J. G. Do sex and gender have separate identities? Arch. Sex. Behav. 53, 2957–2975 (2024). PubMed DOI PMC

Baez, S., Castro-Aldrete, L., Britton, G., Ibañez, A. & Santuccione-Chadha, A. Enhancing brain health in the Global South through a sex and gender lens. Nat. Ment. Health 2, 1308–1317 (2024). PubMed DOI PMC

Kametani, F. & Hasegawa, M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front. Neurosci. 12, 25 (2018). PubMed DOI PMC

Makin, S. The amyloid hypothesis on trial. Nature 559, S4–S7 (2018). PubMed DOI

Selkoe, D. J. & Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608 (2016). PubMed DOI PMC

Hardy, J. A. & Higgins, G. A. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992). PubMed DOI

Leuzy, A. et al. 2020 update on the clinical validity of cerebrospinal fluid amyloid, tau, and phospho-tau as biomarkers for Alzheimer’s disease in the context of a structured 5-phase development framework. Eur. J. Nucl. Med. Mol. Imaging 48, 2121–2139 (2021). PubMed DOI PMC

Fagan, A. M. et al. Longitudinal change in CSF biomarkers in autosomal-dominant Alzheimer’s disease. Sci. Transl. Med. 6, 226ra30 (2014). PubMed DOI PMC

Johnson, E. C. B. et al. Cerebrospinal fluid proteomics define the natural history of autosomal dominant Alzheimer’s disease. Nat. Med. 29, 1979–1988 (2023). PubMed DOI PMC

Malpetti, M. et al. Gender differences in healthy aging and Alzheimer’s dementia: a PubMed DOI PMC

Filon, J. R. et al. Gender differences in Alzheimer disease: brain atrophy, histopathology burden, and cognition. J. Neuropathol. Exp. Neurol. 75, 748–754 (2016). PubMed DOI PMC

Liesinger, A. M. et al. Sex and age interact to determine clinicopathologic differences in Alzheimer’s disease. Acta Neuropathol. 136, 873–885 (2018). PubMed DOI PMC

Hu, Y. et al. Sex differences in the neuropathological hallmarks of Alzheimer’s disease: focus on cognitively intact elderly individuals. Neuropathol. Appl. Neurobiol. 47, 958–966 (2021). PubMed DOI PMC

Serrano, G. E. et al. Correlation of presynaptic and postsynaptic proteins with pathology in Alzheimer’s disease. Int. J. Mol. Sci. 25, 3130 (2024). PubMed DOI PMC

Zea‐Sevilla, M. A., Uceda‐Heras, A. & Rabano, A. The imprint of sex on the heterogeneity of Alzheimer’s disease sex differences in advanced Alzheimer’s disease: a clinical‐pathological study. Alzheimers Dement. 16, e040781 (2020). DOI

Kouri, N. et al. Clinicopathologic heterogeneity and glial activation patterns in Alzheimer disease. JAMA Neurol. 81, 619–629 (2024). PubMed DOI PMC

Gao, C., Jiang, J., Tan, Y. & Chen, S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 8, 359 (2023). PubMed DOI PMC

Reed, E. G. & Keller-Norrell, P. R. Minding the gap: exploring neuroinflammatory and microglial sex differences in Alzheimer’s disease. Int. J. Mol. Sci. 24, 17377 (2023). PubMed DOI PMC

Smith, R. et al. The accumulation rate of tau aggregates is higher in females and younger amyloid-positive subjects. Brain 143, 3805–3815 (2020). PubMed DOI PMC

Wang, Y.-T. et al. Sex-specific modulation of amyloid-β on tau phosphorylation underlies faster tangle accumulation in females. Brain 147, 1497–1510 (2024). PubMed DOI

Buckley, R. F. et al. Sex differences in the association of global amyloid and regional tau deposition measured by positron emission tomography in clinically normal older adults. JAMA Neurol. 76, 542–551 (2019). PubMed DOI PMC

Lee, J. et al. Sex-related reserve hypothesis in Alzheimer’s disease: changes in cortical thickness with a five-year longitudinal follow-up. J. Alzheimers Dis. 65, 641–649 (2018). PubMed DOI PMC

Williamson, J. et al. Sex difference in brain functional connectivity of hippocampus in Alzheimer’s disease. Geroscience 46, 563–572 (2024). PubMed DOI

Holland, D., Desikan, R. S., Dale, A. M. & McEvoy, L. K. Higher rates of decline for women and apolipoprotein E ε4 carriers. Am. J. Neuroradiol. 34, 2287–2293 (2013). PubMed DOI PMC

Lin, K. A. & Doraiswamy, P. M. When Mars versus Venus is not a cliché: gender differences in the neurobiology of Alzheimer’s disease. Front. Neurol. 5, 288 (2015). PubMed DOI PMC

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). PubMed DOI

Levine, D. A. et al. Sex differences in cognitive decline among US adults. JAMA Netw. Open 4, e210169 (2021). PubMed DOI PMC

Sundermann, E. E. et al. Sex-specific norms for verbal memory tests may improve diagnostic accuracy of amnestic MCI. Neurology 93, e1881–e1889 (2019). PubMed DOI PMC

Oliver, M. D., Morrison, C., Kamal, F., Graham, J. & Dadar, M. Subjective cognitive decline is a better marker for future cognitive decline in females than in males. Alzheimers Res. Ther. 14, 197 (2022). PubMed DOI PMC

Sims, J. R. et al. Donanemab in early symptomatic Alzheimer disease: the TRAILBLAZER-ALZ 2 randomized clinical trial. JAMA 330, 512–527 (2023). PubMed DOI PMC

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). PubMed DOI

Cohen, D. et al. Sex differences in the psychiatric manifestations of Alzheimer’s disease. J. Am. Geriatr. Soc. 41, 229–232 (1993). PubMed DOI

Tao, Y. et al. Sex differences in the neuropsychiatric symptoms of patients with Alzheimer’s disease. Am. J. Alzheimers Dis. Other Dement. 33, 450–457 (2018). DOI

Nagata, T. et al. Psychosocial or clinico‐demographic factors related to neuropsychiatric symptoms in patients with Alzheimer’s disease needing interventional treatment: analysis of the CATIE‐AD study. Int. J. Geriatr. Psychiatry 32, 1264–1271 (2017). PubMed DOI

Eikelboom, W. S. et al. Sex differences in neuropsychiatric symptoms in Alzheimer’s disease dementia: a meta-analysis. Alzheimers Res. Ther. 14, 48 (2022). PubMed DOI PMC

Ferretti, M. T., Santuccione-Chadha, A. & Hampel, H. Account for sex in brain research for precision medicine. Nature 569, 40 (2019). PubMed DOI

Rocca, W. A., Grossardt, B. R. & Shuster, L. T. Oophorectomy, estrogen, and dementia: a 2014 update. Mol. Cell. Endocrinol. 389, 7–12 (2014). PubMed DOI PMC

Nerattini, M. et al. Systematic review and meta-analysis of the effects of menopause hormone therapy on risk of Alzheimer’s disease and dementia. Front. Aging Neurosci. 15, 1260427 (2023). PubMed DOI PMC

Liao, H. et al. Association of earlier age at menopause with risk of incident dementia, brain structural indices and the potential mediators: a prospective community-based cohort study. eClinicalMedicine 60, 102033 (2023). PubMed DOI PMC

Basit, S., Wohlfahrt, J. & Boyd, H. A. Pre-eclampsia and risk of dementia later in life: nationwide cohort study. Br. Med. J. 363, k4109 (2018). DOI

Testo, A. A., McBride, C., Bernstein, I. M. & Dumas, J. A. Preeclampsia and its relationship to pathological brain aging. Front. Physiol. 13, 979547 (2022). PubMed DOI PMC

Depypere, H. et al. Coping with menopausal symptoms: an internet survey of Belgian postmenopausal women. Maturitas 90, 24–30 (2016). PubMed DOI

Dattani, S., Rodés-Guirao, L., Ritchie, H., Ortiz-Ospina, E. & Roser, M. Life expectancy. Our World in Data https://ourworldindata.org/life-expectancy (2023).

Appiah, D., Nwabuo, C. C., Ebong, I. A., Wellons, M. F. & Winters, S. J. Trends in age at natural menopause and reproductive life span among US women, 1959–2018. JAMA 325, 1328–1330 (2021). PubMed DOI PMC

Amundsen, D. W. & Diers, C. J. The age of menopause in medieval Europe. Hum. Biol. 45, 605–612 (1973). PubMed

Maki, P. M. The timing of estrogen therapy after ovariectomy — implications for neurocognitive function. Nat. Clin. Pract. Endocrinol. Metab. 4, 494–495 (2008). PubMed DOI

Depypere, H., Vierin, A., Weyers, S. & Sieben, A. Alzheimer’s disease, apolipoprotein E and hormone replacement therapy. Maturitas 94, 98–105 (2016). PubMed DOI

Jauregi‐Zinkunegi, A. et al. Menopausal hormone therapy is associated with worse levels of Alzheimer’s disease biomarkers in APOE ε4‐carrying women: an observational study. Alzheimers Dement. 21, e14456 (2025). PubMed DOI PMC

Maki, P. M., Panay, N. & Simon, J. A. Sleep disturbance associated with the menopause. Menopause 31, 724–733 (2024). PubMed DOI

Cheng, Y.-S. et al. Pharmacologic and hormonal treatments for menopausal sleep disturbances: a network meta-analysis of 43 randomized controlled trials and 32,271 menopausal women. Sleep Med. Rev. 57, 101469 (2021). PubMed DOI

Saeed, Y. & Abbott, S. M. Circadian disruption associated with Alzheimer’s disease. Curr. Neurol. Neurosci. Rep. 17, 29 (2017). PubMed DOI

Depypere, H. et al. Menopause hormone therapy significantly alters pathophysiological biomarkers of Alzheimer’s disease. Alzheimers Dement. 19, 1320–1330 (2023). PubMed DOI

Rahman, A. et al. Sex-driven modifiers of Alzheimer risk: a multimodality brain imaging study. Neurology 95, e166–e178 (2020). PubMed DOI PMC

Brinton, R. D., Yao, J., Yin, F., Mack, W. J. & Cadenas, E. Perimenopause as a neurological transition state. Nat. Rev. Endocrinol. 11, 393–405 (2015). PubMed DOI PMC

Chen, L. et al. Menopausal hormone therapy does not improve some domains of memory: a systematic review and meta-analysis. Front. Endocrinol. 13, 894883 (2022). DOI

Stute, P. et al. Cognitive health after menopause: does menopausal hormone therapy affect it? Best Pract. Res. Clin. Endocrinol. Metab. 35, 101565 (2021). PubMed DOI

Craig, M. C., Maki, P. M. & Murphy, D. G. The Women’s Health Initiative Memory Study: findings and implications for treatment. Lancet Neurol. 4, 190–194 (2005). PubMed DOI

Shumaker, S. A. Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: women's health initiative memory study. JAMA 291, 2947–2958 (2004). PubMed DOI

Manson, J. E. et al. Menopausal hormone therapy and long-term all-cause and cause-specific mortality. JAMA 318, 927–938 (2017). PubMed DOI PMC

Valen-Sendstad, A. et al. Effects of hormone therapy on depressive symptoms and cognitive functions in women with Alzheimer disease: a 12 month randomized, double-blind, placebo-controlled study of low-dose estradiol and norethisterone. Am. J. Geriatr. Psychiatry 18, 11–20 (2010). PubMed DOI

Saleh, R. N. M., Hornberger, M., Ritchie, C. W. & Minihane, A. M. Hormone replacement therapy is associated with improved cognition and larger brain volumes in at-risk APOE4 women: results from the European Prevention of Alzheimer’s Disease (EPAD) cohort. Alzheimers Res. Ther. 15, 10 (2023). PubMed DOI PMC

Kantarci, K. et al. Early postmenopausal transdermal 17β-estradiol therapy and amyloid-β deposition. J. Alzheimers Dis. 53, 547–556 (2016). PubMed DOI PMC

Pa, J. et al. Effects of sex, APOE4, and lifestyle activities on cognitive reserve in older adults. Neurology 99, e789–e798 (2022). PubMed DOI PMC

AARP & National Alliance for Caregiving. Caregiving in the United States 2020. AARP https://doi.org/10.26419/ppi.00103.001 (2020).

Abken, E., Ferretti, M. T., Castro-Aldrete, L., Santuccione Chadha, A. & Tartaglia, M. C. The impact of informant-related characteristics including sex/gender on assessment of Alzheimer’s disease symptoms and severity. Front. Glob. Womens Health 5, 1326881 (2024). PubMed DOI PMC

Roberto, K. A. & Jarrott, S. E. Family caregivers of older adults: a life span perspective. Fam. Relat. 57, 100–111 (2008). DOI

Kroenke, K. & Mangelsdorff, A. D. Common symptoms in ambulatory care: incidence, evaluation, therapy, and outcome. Am. J. Med. 86, 262–266 (1989). PubMed DOI

Kroenke, K. & Spitzer, R. L. Gender differences in the reporting of physical and somatoform symptoms. Psychosom. Med. 60, 150–155 (1998). PubMed DOI

Steenland, K. et al. Late-life depression as a risk factor for mild cognitive impairment or Alzheimer’s disease in 30 US Alzheimer’s disease centers. J. Alzheimers Dis. 31, 265–275 (2012). PubMed DOI PMC

Ritchie, K. et al. The clinical picture of Alzheimer’s Disease in the decade before diagnosis. J. Clin. Psychiatry 77, e305–e311 (2016). PubMed DOI

Ownby, R. L., Crocco, E., Acevedo, A., John, V. & Loewenstein, D. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch. Gen. Psychiatry 63, 530–538 (2006). PubMed DOI PMC

Yee, J. L. & Schulz, R. Gender differences in psychiatric morbidity among family caregivers. Gerontologist 40, 147–164 (2000). PubMed DOI

Westergaard, D., Moseley, P., Sørup, F. K. H., Baldi, P. & Brunak, S. Population-wide analysis of differences in disease progression patterns in men and women. Nat. Commun. 10, 666 (2019). PubMed DOI PMC

Vargas-Gonzalez, J.-C., Chadha, A. S., Castro-Aldrete, L., Ferretti, M. T. & Tartaglia, M. C. Informant characteristics influence Clinical Dementia Rating Sum of Boxes scores-based staging of Alzheimer’s disease. Nat. Aging 4, 1538–1543 (2024). PubMed DOI

Gómez-Isla, T. & Frosch, M. P. Lesions without symptoms: understanding resilience to Alzheimer disease neuropathological changes. Nat. Rev. Neurol. 18, 323–332 (2022). PubMed DOI PMC

Sperling, R. A. et al. Trial of solanezumab in preclinical Alzheimer’s disease. N. Engl. J. Med. 389, 1096–1107 (2023). PubMed DOI PMC

Miller, D. I. & Halpern, D. F. The new science of cognitive sex differences. Trends Cogn. Sci. 18, 37–45 (2014). PubMed DOI

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). PubMed DOI PMC

Sundermann, E. E. et al. Does the female advantage in verbal memory contribute to underestimating Alzheimer’s disease pathology in women versus men? J. Alzheimers Dis. 56, 947–957 (2017). PubMed DOI PMC

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

Berent-Spillson, A. et al. Hormonal environment affects cognition independent of age during the menopause transition. J. Clin. Endocrinol. Metab. 97, E1686–E1694 (2012). PubMed DOI PMC

Avila-Varela, D. S. et al. Whole-brain dynamics across the menstrual cycle: the role of hormonal fluctuations and age in healthy women. npj Women’s Health 2, 8 (2024). DOI

Global Education Monitoring Report Team. Global Education Monitoring Report 2020: Gender Report, A New Generation: 25 Years of Efforts for Gender Equality in Education (UNESCO, 2020).

Ardila, A. Cultural values underlying psychometric cognitive testing. Neuropsychol. Rev. 15, 185–195 (2005). PubMed DOI

Boone, K. B., Victor, T. L., Wen, J., Razani, J. & Pontón, M. The association between neuropsychological scores and ethnicity, language, and acculturation variables in a large patient population. Arch. Clin. Neuropsychol. 22, 355–365 (2007). PubMed DOI

Gallegos, M. et al. 45 years of the mini-mental state examination (MMSE): a perspective from Ibero-America. Dement. Neuropsychol. 16, 384–387 (2022). PubMed DOI PMC

Kueper, J. K., Speechley, M. & Montero-Odasso, M. The Alzheimer’s disease assessment scale–cognitive subscale (ADAS-Cog): modifications and responsiveness in pre-dementia populations. A narrative review. J. Alzheimers Dis. 63, 423–444 (2018). PubMed DOI PMC

Evans, J., Dutt, A. & Fernández, A. L. in Understanding Cross-Cultural Neuropsychology: Science, Testing, and Challenges (eds. Fernández, A. L. & Evans, J.) 174–185 (Routledge, 2022).

Ponsford, J. International growth of neuropsychology. Neuropsychology 31, 921–933 (2017). PubMed DOI

Manly, J. J. Advantages and disadvantages of separate norms for African Americans. Clin. Neuropsychol. 19, 270–275 (2005). PubMed DOI

Storey, J. E., Rowland, J. T. J., Conforti, D. A. & Dickson, H. G. The Rowland Universal Dementia Assessment Scale (RUDAS): a multicultural cognitive assessment scale. Int. Psychogeriatr. 16, 13–31 (2004). PubMed DOI

Kempler, D., Teng, E. L., Taussig, M. & Dick, M. B. The common objects memory test (COMT): a simple test with cross-cultural applicability. J. Int. Neuropsychol. Soc. 16, 537–545 (2010). PubMed DOI

Yaffe, K. et al. Predictors of maintaining cognitive function in older adults: the Health ABC Study. Neurology 72, 2029–2035 (2009). PubMed DOI PMC

Zahodne, L. B. et al. Late-life memory trajectories in relation to incident dementia and regional brain atrophy. J. Neurol. 262, 2484–2490 (2015). PubMed DOI PMC

Gamaldo, A. A. & Allaire, J. C. Daily fluctuations in everyday cognition. J. Aging Health 28, 834–849 (2016). PubMed DOI

Hultsch, D. F., MacDonald, S. W. S., Hunter, M. A., Levy-Bencheton, J. & Strauss, E. Intraindividual variability in cognitive performance in older adults: comparison of adults with mild dementia, adults with arthritis, and healthy adults. Neuropsychology 14, 588–598 (2000). PubMed DOI

Muurling, M. et al. Augmented reality versus standard tests to assess cognition and function in early Alzheimer’s disease. npj Digit. Med. 6, 234 (2023). PubMed DOI PMC

Laricchia, F. Global smartphone penetration rate as share of population from 2016 to 2023. Statista https://www.statista.com/statistics/203734/global-smartphone-penetration-per-capita-since-2005/ (2024).

Hassenstab, J. et al. Remote cognitive assessment approaches in the Dominantly Inherited Alzheimer Network (DIAN). Alzheimers Dement. 16, e038144 (2020). DOI

Sliwinski, M. J. et al. Reliability and validity of ambulatory cognitive assessments. Assessment 25, 14–30 (2018). PubMed DOI

Kaye, J. et al. Unobtrusive measurement of daily computer use to detect mild cognitive impairment. Alzheimer Dement. 10, 10–17 (2014). DOI

Oatley, G., Choudhury, T. & Buckman, P. Smart textiles for improved quality of life and cognitive assessment. Sensors 21, 8008 (2021). PubMed DOI PMC

Wakim, N. I., Braun, T. M., Kaye, J. A. & Dodge, H. H. Choosing the right time granularity for analysis of digital biomarker trajectories. Alzheimers Dement. 6, e12094 (2020).

Aschenbrenner, A. J. et al. Neuropsychological correlates of changes in driving behavior among clinically healthy older adults. J. Gerontol. B Psychol. Sci. Soc. Sci. 77, 1769–1778 (2022). PubMed DOI PMC

Cho, S. et al. Lexical and acoustic speech features relating to Alzheimer disease pathol. Neurology 99, e313–e322 (2022). PubMed DOI PMC

Tavabi, N. et al. Cognitive digital biomarkers from automated transcription of spoken language. J. Prev. Alzheimers Dis. 9, 791–800 (2022). PubMed DOI

Mueller, K. D. et al. Amyloid β associations with connected speech in cognitively unimpaired adults. Alzheimers Dement. 13, e12203 (2021).

Hajjar, I. et al. Development of digital voice biomarkers and associations with cognition, cerebrospinal biomarkers, and neural representation in early Alzheimer’s disease. Alzheimers Dement. 15, e12393 (2023).

Mahon, E. & Lachman, M. E. Voice biomarkers as indicators of cognitive changes in middle and later adulthood. Neurobiol. Aging 119, 22–35 (2022). PubMed DOI PMC

Amini, S. et al. Automated detection of mild cognitive impairment and dementia from voice recordings: a natural language processing approach. Alzheimers Dement. 19, 946–955 (2023). PubMed DOI

Moret-Tatay, C. et al. A pilot screening for cognitive impairment through voice technology (WAY2AGE). BMC Psychol. 11, 170 (2023). PubMed DOI PMC

Zhao, X. et al. A voice recognition-based digital cognitive screener for dementia detection in the community: development and validation study. Front. Psychiatry 13, 899729 (2022). PubMed DOI PMC

Young, C. B. et al. Speech patterns during memory recall relates to early tau burden across adulthood. Alzheimers Dement. 20, 2552–2563 (2024). PubMed DOI PMC

Harms, R. L. et al. Digital biomarkers and sex impacts in Alzheimer’s disease management — potential utility for innovative 3P medicine approach. EPMA J. 13, 299–313 (2022). PubMed DOI PMC

Cirillo, D. et al. Sex and gender differences and biases in artificial intelligence for biomedicine and healthcare. npj Digit. Med. 3, 81 (2020). PubMed DOI PMC

Piau, A., Wild, K., Mattek, N. & Kaye, J. Current state of digital biomarker technologies for real-life, home-based monitoring of cognitive function for mild cognitive impairment to mild Alzheimer disease and implications for clinical care: systematic review. J. Med. Internet Res. 21, e12785 (2019). PubMed DOI PMC

Nicosia, J. et al. Bridging the technological divide: stigmas and challenges with technology in digital brain health studies of older adults. Front. Digit. Health 4, 880055 (2022). PubMed DOI PMC

Ford, E., Milne, R. & Curlewis, K. Ethical issues when using digital biomarkers and artificial intelligence for the early detection of dementia. Wiley Interdiscip. Rev. Data Min. Knowl. Discov. 13, e1492 (2023). PubMed DOI PMC

Hatahet, O. & Seghier, M. L. The validity of studying healthy aging with cognitive tests measuring different constructs. Sci. Rep. 14, 23880 (2024). PubMed DOI PMC

Fernández, A. L. & Abe, J. Bias in cross-cultural neuropsychological testing: problems and possible solutions. Cult. Brain 6, 1–35 (2018). DOI

Romero, H. R. et al. Challenges in the neuropsychological assessment of ethnic minorities: summit proceedings. Clin. Neuropsychol. 23, 761–779 (2009). PubMed DOI

Hampel, H. et al. The amyloid-β pathway in Alzheimer’s disease. Mol. Psychiatry 26, 5481–5503 (2021). PubMed DOI PMC

Hampel, H. et al. The β-secretase BACE1 in Alzheimer’s disease. Biol. Psychiatry 89, 745–756 (2021). PubMed DOI

Janelidze, S. et al. Head-to-head comparison of 8 plasma amyloid-β 42/40 assays in Alzheimer disease. JAMA Neurol. 78, 1375–1382 (2021). PubMed DOI

Vergallo, A. et al. Plasma β-secretase1 concentrations correlate with basal forebrain atrophy and neurodegeneration in cognitively healthy individuals at risk for AD. Alzheimers Dement. 17, 629–640 (2021). PubMed DOI

Vergallo, A. et al. Plasma amyloid β 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer’s disease. Alzheimers Dement. 15, 764–775 (2019). PubMed DOI

Jack, C. R. et al. Age-specific and sex-specific prevalence of cerebral β-amyloidosis, tauopathy, and neurodegeneration in cognitively unimpaired individuals aged 50–95 years: a cross-sectional study. Lancet Neurol. 16, 435–444 (2017). PubMed DOI PMC

Mielke, M. M. Consideration of sex differences in the measurement and interpretation of Alzheimer disease-related biofluid-based biomarkers. J. Appl. Lab. Med. 5, 158–169 (2020). PubMed DOI PMC

Zetterberg, H. et al. Association of cerebrospinal fluid neurofilament light concentration with Alzheimer disease progression. JAMA Neurol. 73, 60–67 (2016). PubMed DOI PMC

Bridel, C. et al. Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: a systematic review and meta-analysis. JAMA Neurol. 76, 1035–1048 (2019). PubMed DOI PMC

Skillbäck, T. et al. Sex differences in CSF biomarkers for neurodegeneration and blood–brain barrier integrity. Alzheimers Dement. 13, e12141 (2021).

Gur, R. C. et al. Sex differences in brain gray and white matter in healthy young adults: correlations with cognitive performance. J. Neurosci. 19, 4065–4072 (1999). PubMed DOI PMC

Lotze, M. et al. Novel findings from 2,838 adult brains on sex differences in gray matter brain volume. Sci. Rep. 9, 1671 (2019). PubMed DOI PMC

Keshavan, A. et al. Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70. Brain 144, 434–449 (2020).

Brickman, A. M. et al. Plasma p‐tau PubMed DOI

Mielke, M. M. et al. Performance of plasma phosphorylated tau 181 and 217 in the community. Nat. Med. 28, 1398–1405 (2022). PubMed DOI PMC

Tsiknia, A. A. et al. Sex differences in plasma p-tau PubMed DOI PMC

Liu, C. et al. Sex‐specific biomarkers in Alzheimer’s disease progression: Framingham Heart Study. Alzheimers Dement. 14, e12369 (2022).

Hampel, H. et al. Precision medicine and drug development in Alzheimer’s disease: the importance of sexual dimorphism and patient stratification. Front. Neuroendocrinol. 50, 31–51 (2018). PubMed DOI

Mosconi, L. et al. Increased Alzheimer’s risk during the menopause transition: a 3-year longitudinal brain imaging study. PLoS ONE 13, e0207885 (2018). PubMed DOI PMC

Wang, Y.-T. T. et al. Interactive rather than independent effect of APOE and sex potentiates tau deposition in women. Brain Commun. 3, fcab126 (2021). PubMed DOI PMC

Li, X. et al. Sex difference in network topology and education correlated with sex difference in cognition during the disease process of Alzheimer. Front. Aging Neurosci. 13, 639529 (2021). PubMed DOI PMC

Cavedo, E. et al. Sex differences in functional and molecular neuroimaging biomarkers of Alzheimer’s disease in cognitively normal older adults with subjective memory complaints. Alzheimers Dement. 14, 1204–1215 (2018). PubMed DOI

Damoiseaux, J. S. et al. Gender modulates the APOE ε4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levels. J. Neurosci. 32, 8254–8262 (2012). PubMed DOI PMC

Vergallo, A. et al. Brain Aβ load association and sexual dimorphism of plasma BACE1 concentrations in cognitively normal individuals at risk for AD. Alzheimers Dement. 15, 1274–1285 (2019). PubMed DOI

Yiannopoulou, K. G. & Papageorgiou, S. G. Current and future treatments in Alzheimer disease: an update. J. Cent. Nerv. Syst. Dis. 12, 1179573520907397 (2020). PubMed DOI PMC

Marucci, G. et al. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 190, 108352 (2021). PubMed DOI

Boxer, A. L. & Sperling, R. Accelerating Alzheimer’s therapeutic development: the past and future of clinical trials. Cell 186, 4757–4772 (2023). PubMed DOI PMC

Bayer, A. J. et al. Evaluation of the safety and immunogenicity of synthetic Aβ42 (AN1792) in patients with AD. Neurology 64, 94–101 (2005). PubMed DOI

van Dyck, C. H. et al. Lecanemab in early Alzheimer’s disease. N. Engl. J. Med. 388, 9–21 (2023). PubMed DOI

Bateman, R. J. et al. Two phase 3 trials of gantenerumab in early Alzheimer’s disease. N. Engl. J. Med. 389, 1862–1876 (2023). PubMed DOI PMC

Budd Haeberlein, S. et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. J. Prev. Alzheimers Dis. 9, 197–221 (2022). PubMed DOI

Wang, Y. An insider’s perspective on FDA approval of aducanumab. Alzheimers Dement. 9, e12382 (2023).

Mahase, E. Alzheimer’s disease: lecanemab gets full FDA approval and black box safety warning. Br. Med. J. 382, 1580 (2023). DOI

US Food and Drug Administration. FDA approves treatment for adults with Alzheimer’s disease. FDA https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-adults-alzheimers-disease (2024).

US Food and Drug Administration. Women in clinical trials: research and policy from the FDA Office of Women’s Health. FDA https://www.fda.gov/consumers/diverse-women-clinical-trials/women-clinical-trials-research-and-policy (2025).

Abdelnour, C. et al. Gender and sex bias in clinical trial recruitment in Alzheimer’s disease: findings from Fundació ACE. Alzheimers Dement. 16, e041234 (2020). DOI

Buckley, R. F., Gong, J. & Woodward, M. A call to action to address sex differences in Alzheimer disease clinical trials. JAMA Neurol. 80, 769–770 (2023). PubMed DOI PMC

Satizabal, C. L. et al. Incidence of dementia over three decades in the Framingham Heart Study. N. Engl. J. Med. 374, 523–532 (2016). PubMed DOI PMC

Pinho-Gomes, A.-C., Gong, J., Harris, K., Woodward, M. & Carcel, C. Dementia clinical trials over the past decade: are women fairly represented? BMJ Neurol. Open 4, e000261 (2022). PubMed DOI PMC

Martinkova, J. et al. Proportion of women and reporting of outcomes by sex in clinical trials for Alzheimer disease. JAMA Netw. Open 4, e2124124 (2021). PubMed DOI

Thanasopoulou, A. et al. GRADUATE I and II: subgroup analyses from two phase III studies evaluating the efficacy and safety of subcutaneous gantenerumab in early Alzheimer’s disease (AD). N. Engl. J. Med. 389, 1862–1876 (2023). PubMed PMC

Buckley, R. F. et al. Menopause status moderates sex differences in tau burden: a Framingham PET study. Ann. Neurol. 92, 11–22 (2022). PubMed DOI PMC

Shokouhi, S., Taylor, W. D., Albert, K., Kang, H. & Newhouse, P. A. In vivo network models identify sex differences in the spread of tau pathology across the brain. Alzheimers Dement. 12, e12016 (2020).

Rogers, M. B. What happens after amyloid plaque removal? Who benefits most? In International Conference on Alzheimer’s and Parkinson’s Diseases 2023 (ed. Rogers, M. B.) Part 6 of 19 https://www.alzforum.org/news/conference-coverage/what-happens-after-amyloid-plaque-removal-who-benefits-most (Alzforum, 2023).

16th Clinical Trials on Alzheimer’s Disease (CTAD) Boston, MA (USA) October 24–27, 2023: Symposia. J. Prev. Alzheimers Dis. 10 (Suppl. 1), 4–55 (2023).

Sperling, R. A. et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement. 7, 367–385 (2011). PubMed DOI

Barakos, J. et al. Detection and management of amyloid-related imaging abnormalities in patients with Alzheimer’s disease treated with anti-amyloid β therapy. J. Prev. Alzheimers Dis. 9, 211–220 (2022). PubMed DOI

Salloway, S. et al. Amyloid-related imaging abnormalities in 2 phase 3 studies evaluating aducanumab in patients with early Alzheimer disease. JAMA Neurol. 79, 13–21 (2022). PubMed DOI

Mintun, M. A. et al. Donanemab in early Alzheimer’s disease. N. Engl. J. Med. 384, 1691–1704 (2021). PubMed DOI

Swanson, C. J. et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer’s disease with lecanemab, an anti-Aβ protofibril antibody. Alzheimers Res. Ther. 13, 80 (2021). PubMed DOI PMC

Ostrowitzki, S. et al. A phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease. Alzheimers Res. Ther. 9, 95 (2017). PubMed DOI PMC

Cummings, J. et al. Aducanumab: appropriate use recommendations update. J. Prev. Alzheimers Dis. 9, 221–230 (2022). PubMed DOI PMC

Solopova, E. et al. Fatal iatrogenic cerebral β-amyloid-related arteritis in a woman treated with lecanemab for Alzheimer’s disease. Nat. Commun. 14, 8220 (2023). PubMed DOI PMC

Loomis, S. J. et al. Genome-wide association studies of ARIA from the aducanumab phase 3 ENGAGE and EMERGE studies. Neurology 102, e207919 (2024). PubMed DOI

Isaacson, R. S. et al. Individualized clinical management of patients at risk for Alzheimer’s dementia. Alzheimers Dement. 15, 1588–1602 (2019). PubMed DOI

Saif, N. et al. Sex-driven differences in the effectiveness of individualized clinical management of Alzheimer’s disease risk. J. Prev. Alzheimers Dis. 9, 731–774 (2022). PubMed DOI

Nucci, D. et al. Association between Mediterranean diet and dementia and Alzheimer disease: a systematic review with meta-analysis. Aging Clin. Exp. Res. 36, 77 (2024). PubMed DOI PMC

Chen, L.-J., Sha, S., Stocker, H., Brenner, H. & Schöttker, B. The associations of serum vitamin D status and vitamin D supplements use with all-cause dementia, Alzheimer’s disease, and vascular dementia: a UK Biobank based prospective cohort study. Am. J. Clin. Nutr. 119, 1052–1064 (2024). PubMed DOI PMC

Shi, L. et al. Sleep disturbances increase the risk of dementia: a systematic review and meta-analysis. Sleep Med. Rev. 40, 4–16 (2018). PubMed DOI

Coley, N., Giulioli, C., Aisen, P. S., Vellas, B. & Andrieu, S. Randomised controlled trials for the prevention of cognitive decline or dementia: a systematic review. Ageing Res. Rev. 82, 101777 (2022). PubMed DOI

Noach, S., Witteman, B., Boss, H. M. & Janse, A. Effects of multidomain lifestyle interventions on cognitive decline and Alzheimer’s disease prevention: a literature review and future recommendations. Cereb. Circ. Cogn. Behav. 4, 100166 (2023). PubMed PMC

Xu, Z. et al. A pilot feasibility randomized controlled trial on combining mind-body physical exercise, cognitive training, and nurse-led risk factor modification to reduce cognitive decline among older adults with mild cognitive impairment in primary care. PeerJ 8, e9845 (2020). PubMed DOI PMC

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). PubMed DOI

Chen, L. et al. Efficacy of multidomain interventions to improve physical frailty, depression and cognition: data from cluster‐randomized controlled trials. J. Cachexia Sarcopenia Muscle 11, 650–662 (2020). PubMed DOI PMC

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). PubMed DOI

Meng, X. et al. Multidomain lifestyle interventions for cognition and the risk of dementia: a systematic review and meta-analysis. Int. J. Nurs. Stud. 130, 104236 (2022). PubMed DOI

Zülke, A. E. et al. Gender-specific design and effectiveness of non-pharmacological interventions against cognitive decline – systematic review and meta-analysis of randomized controlled trials. J. Prev. Alzheimers Dis. 10, 69–82 (2022). DOI

Weiss, E. M., Kemmler, G., Deisenhammer, E. A., Fleischhacker, W. W. & Delazer, M. Sex differences in cognitive functions. Pers. Individ. Differ. 35, 863–875 (2003). DOI

Hirnstein, M., Stuebs, J., Moè, A. & Hausmann, M. Sex/gender differences in verbal fluency and verbal-episodic memory: a meta-analysis. Persp. Psychol. Sci. 18, 67–90 (2023). DOI

Ferretti, M. T. et al. Maximizing utility of neuropsychological measures in sex-specific predictive models of incident Alzheimer’s disease in the Framingham Heart Study. Alzheimers Dement. 20, 1112–1122 (2024). PubMed DOI

van Charante, E. P. M. et al. Effectiveness of a 6-year multidomain vascular care intervention to prevent dementia (preDIVA): a cluster-randomised controlled trial. Lancet 388, 797–805 (2016). DOI

den Brok, M. G. H. E. et al. The effect of multidomain interventions on global cognition, symptoms of depression and apathy — a pooled analysis of two randomized controlled trials. J. Prev. Alzheimers Dis. 9, 96–103 (2022). DOI

Belleville, S. et al. Is more always better? Dose effect in a multidomain intervention in older adults at risk of dementia. Alzheimers Dement. 18, 2140–2150 (2022). PubMed DOI

Park, H. K. et al. South Korean study to prevent cognitive impairment and protect brain health through lifestyle intervention in at-risk elderly people: protocol of a multicenter, randomized controlled feasibility trial. J. Clin. Neurol. 16, 292–303 (2020). PubMed DOI PMC

Xu, X. et al. The Singapore geriatric intervention study to reduce cognitive decline and physical frailty (SINGER): study design and protocol. J. Prev. Alzheimers Dis. 9, 40–48 (2022). PubMed DOI PMC

Kivipelto, M. et al. World‐Wide FINGERS network: a global approach to risk reduction and prevention of dementia. Alzheimers Dement. 16, 1078–1094 (2020). PubMed DOI

Röhr, S., Kivipelto, M., Mangialasche, F., Ngandu, T. & Riedel-Heller, S. G. Multidomain interventions for risk reduction and prevention of cognitive decline and dementia: current developments. Curr. Opin. Psychiatry 35, 285–292 (2022). PubMed DOI

Rosenberg, A., Mangialasche, F., Ngandu, T., Solomon, A. & Kivipelto, M. Multidomain interventions to prevent cognitive impairment, Alzheimer’s disease, and dementia: from FINGER to World-Wide FINGERS. J. Prev. Alzheimers Dis. 7, 29–36 (2020). PubMed DOI

Kivipelto, M., Mangialasche, F. & Ngandu, T. Lifestyle interventions to prevent cognitive impairment, dementia and Alzheimer disease. Nat. Rev. Neurol. 14, 653–666 (2018). PubMed DOI

Zülke, A. E. et al. A multidomain intervention against cognitive decline in an at‐risk‐population in Germany: results from the cluster‐randomized AgeWell.de trial. Alzheimers Dement. 20, 615–628 (2024). PubMed DOI

Crivelli, L. et al. Latin American initiative for lifestyle intervention to prevent cognitive decline (LatAm‐FINGERS): study design and harmonization. Alzheimers Dement. 19, 4046–4060 (2023). PubMed DOI

Nichols, E. et al. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden of Disease Study 2019. Lancet Public Health 7, e105–e125 (2022). DOI

Custodio, N., Allegri, R., Lopera, F. & Caramelli, P. Need to adapt Alzheimer’s disease criteria in Latin America. Alzheimers Dement. 20, 8206–8208 (2024). PubMed DOI PMC

van Wanrooij, L. L. et al. Pooling individual participant data from randomized controlled trials: exploring potential loss of information. PLoS ONE 15, e0232970 (2020). PubMed DOI PMC

Research news: FINGERS Plus for women: a new collaboration just announced at Lausanne XI. Dementia Researcher https://www.dementiaresearcher.nihr.ac.uk/fingers-plus-for-women-a-new-collaboration/ (2024).

Feldman, H. H. et al. Protocol for the brain health support program study of the Canadian therapeutic platform trial for multidomain interventions to prevent dementia (CAN-THUMBS UP): a prospective 12-month intervention study. J. Prev. Alzheimers Dis. 10, 875–885 (2023). PubMed DOI

Hadoux, X. et al. Non-invasive in vivo hyperspectral imaging of the retina for potential biomarker use in Alzheimer’s disease. Nat. Commun. 10, 4227 (2019). PubMed DOI PMC

Cheung, C. Y. et al. A deep learning model for detection of Alzheimer’s disease based on retinal photographs: a retrospective, multicentre case-control study. Lancet Digit. Health 4, e806–e815 (2022). PubMed DOI

Hinman, J. D. et al. Placental growth factor as a sensitive biomarker for vascular cognitive impairment. Alzheimers Dement. 19, 3519–3527 (2023). PubMed DOI

Milà-Alomà, M. et al. Plasma p-tau PubMed PMC

Buerger, K. et al. CSF phosphorylated tau protein correlates with neocortical neurofibrillary pathology in Alzheimer’s disease. Brain 129, 3035–3041 (2006). PubMed DOI

Hampel, H. et al. Correlation of cerebrospinal fluid levels of tau protein phosphorylated at threonine 231 with rates of hippocampal atrophy in Alzheimer disease. Arch. Neurol. 62, 770–773 (2005). PubMed DOI

Ashton, N. J. et al. Diagnostic accuracy of a plasma phosphorylated tau 217 immunoassay for Alzheimer disease pathology. JAMA Neurol. 81, 255–263 (2024). PubMed DOI PMC

Canals-Gispert, L. et al. Impact of gender on the willingness to participate in clinical trials and undergo related procedures in individuals from an Alzheimer’s prevention research cohort. Alzheimers Res. Ther. 16, 263 (2024). PubMed DOI PMC

Peters, S. A. E. & Woodward, M. A roadmap for sex- and gender-disaggregated health research. BMC Med. 21, 354 (2023). PubMed DOI PMC

Vasquez-Avila, K., Pacheco-Barrios, K., Sampaio de Melo, P. & Fregni, F. Addressing the critical role of gender identity and sex in the planning, analysis, and conduct of clinical trials. Princ. Pract. Clin. Res. 7, 59–62 (2021). PubMed PMC

Aranda, M. P. et al. A call to address structural barriers to Hispanic/Latino representation in clinical trials on Alzheimer’s disease and related dementias: a micro‐meso‐macro perspective. Alzheimers Dement. 9, e12389 (2023).

Ferretti, M. T. et al. Sex and gender differences in Alzheimer’s disease: current challenges and implications for clinical practice. Eur. J. Neurol. 27, 928–943 (2020). PubMed DOI

Buffenstein, I. et al. Demographic recruitment bias of adults in United States randomized clinical trials by disease categories between 2008 to 2019: a systematic review and meta-analysis. Sci. Rep. 13, 42 (2023). PubMed DOI PMC

Hentzen, N. B. et al. Mapping of European activities on the integration of sex and gender factors in neurology and neuroscience. Eur. J. Neurol. 29, 2572–2579 (2022). DOI

Mazure, C. M. The white house initiative on women’s health research. JAMA Intern. Med. 184, 1277–1278 (2024). PubMed DOI

Yu, J.-T. et al. Evidence-based prevention of Alzheimer’s disease: systematic review and meta-analysis of 243 observational prospective studies and 153 randomised controlled trials. J. Neurol. Neurosurg. Psychiatry 91, 1201–1209 (2020). PubMed DOI

Booi, L. et al. Protocol for the next generation brain health survey: an international survey of attitudes, understanding and exposure to brain health risk factors in young adults. Discov. Publ. Health 21, 244 (2024). DOI

Bassetti, C. L. A. et al. The swiss brain health plan 2023–2033. Clin. Transl. Neurosci. 7, 38 (2023). DOI

Guo, L., Zhong, M. B., Zhang, L., Zhang, B. & Cai, D. Sex differences in Alzheimer’s disease: insights from the multiomics landscape. Biol. Psychiatry 91, 61–71 (2022). PubMed DOI

Toro, C. A., Zhang, L., Cao, J. & Cai, D. Sex differences in Alzheimer’s disease: understanding the molecular impact. Brain Res. 1719, 194–207 (2019). PubMed DOI PMC

Rahman, A. et al. Sex and gender driven modifiers of Alzheimer’s: the role for estrogenic control across age, race, medical, and lifestyle risks. Front. Aging Neurosci. 11, 315 (2019). PubMed DOI PMC

Gore, K. L., Cherney, S., Shih, R. A. & Girven, R. S. Could dementia in the national security workforce create a security threat? RAND Corporation http://www.jstor.org/stable/resrep48835 (2023).

Ortiz-Ospina, E., Joe Hasell, J. & Roser, M. Economic inequality by gender. Our World in Data https://ourworldindata.org/economic-inequality-by-gender (2024).

Fornwagner, H., Grosskopf, B., Lauf, A., Schöller, V. & Städter, S. On the robustness of gender differences in economic behavior. Sci. Rep. 12, 21549 (2022). PubMed DOI PMC

Fuchs, V. R. Sex differences in economic well-being. Science 232, 459–464 (1986). PubMed DOI

Arias, D., Saxena, S. & Verguet, S. Quantifying the global burden of mental disorders and their economic value. eClinicalMedicine 54, 101675 (2022). PubMed DOI PMC

Ocañas, S. R. et al. Chromosomal and gonadal factors regulate microglial sex effects in the aging brain. Brain Res. Bull. 195, 157–171 (2023). PubMed DOI PMC

Ackley, S. F. et al. Discordance in chromosomal and self-reported sex in the UK Biobank: implications for transgender- and intersex-inclusive data collection. Proc. Natl Acad. Sci. USA 120, e2218700120 (2023). PubMed DOI PMC

Hennekam, R. C. M. et al. Elements of morphology: standard terminology for the external genitalia. Am. J. Med. Genet. A 161, 1238–1263 (2013). DOI

Lee, B. H., Eid, R. S., Hodges, T. E., Barth, C. & Galea, L. A. M. Leveraging research into sex differences and steroid hormones to improve brain health. Nat. Rev. Endocrinol. 21, 214–229 (2024). PubMed DOI

Lam, C. S. P. How to incorporate sex and gender into the design of cardiovascular clinical trials. Circulation 145, 499–501 (2022). PubMed DOI

Dumitrescu, L., Mayeda, E. R., Sharman, K., Moore, A. M. & Hohman, T. J. Sex differences in the genetic architecture of Alzheimer’s disease. Curr. Genet. Med. Rep. 7, 13–21 (2019). PubMed DOI PMC

Find record

Citation metrics

Loading data ...

Archiving options

Loading data ...