Background: Alzheimer’s disease (AD), the most common cause of dementia, affects twice as many women as men. Moreover, sex is increasingly recognised as an important factor for AD, influencing symptom presentation, progression, disease biology, and treatment responses. In parallel, AD biomarkers are becoming more accessible with the discovery of specific and accurate blood-based biomarkers and their incorporation in AD diagnostic frameworks. This narrative review aimed to summarise sex differences in the concentration and interpretation of biofluid biomarkers for AD. Summary: Biological sex may influence both the concentration and interpretation of biofluid biomarkers for AD pathology such as amyloid-β aggregation, tau neurofibrillary tangles, neurodegeneration, or neuroinflammation. While some biofluid biomarkers display consistent sex differences in absolute levels, most biomarker levels have not been found to differ consistently by sex. Nonetheless, even biomarkers that do not differ in absolute levels display sex-specific associations with clinically relevant variables such as brain atrophy, cognitive impairment, and disease progression. Key Message: Sex may influence the interpretation of AD biomarkers depending on their context of use, and more research is required to develop sex-specific guidelines. Future research should aim to study sex differences and sex-specific associations with variables of interest, as well as the underlying factors driving sex differences in AD.

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. Sex is increasingly recognised as an important factor in AD and a key variable for precision medicine approaches [1]. The prevalence of AD is higher in women, and the lifetime risk of AD for women is approximately 20%, twice that of men [2]. This is partially explained by the relatively greater life expectancy in women, as higher age is a major risk factor for AD [2]. Studies on age-adjusted incidence have found conflicting results, and sex differences in AD risk appear to depend on age, birth cohort, and across different geographical regions [3]. Several studies in Europe and Asia suggest higher incidence of AD in women, while many studies in North and South America have not found sex differences in incidence [4, 5]. Nonetheless, biological sex can contribute to differences in the pathophysiological process, presentation of symptoms, and treatment responses [6]. In parallel with the increasing focus on sex differences and sex-specific risk factors for AD, recent years have seen considerable development in biofluid biomarkers for AD pathology, with the emergence of highly precise blood-based biomarkers and their incorporation in the diagnosis of AD [7].

The pathophysiological hallmarks of AD are extracellular amyloid-β (Aβ) plaques, intracellular neurofibrillary tangles composed of phosphorylated tau (p-tau) proteins, and neurodegeneration [8] see (Fig. 1). This is accompanied by changes in several molecular pathways, including inflammation and mitophagy [9, 10]. AD pathology can be identified by analysis of specific proteins in cerebrospinal fluid (CSF) and blood plasma (Fig. 1) years before onset of clinical symptoms [7]. Thus, recently revised criteria for diagnosis and staging of AD by the Alzheimer’s Association Workgroup (AAW-criteria) define AD by the presence of biomarkers for Aβ proteinopathy and p-tau [7] and propose that biomarkers of AD tau proteinopathy, neurodegeneration, and inflammation may inform disease staging, prognosis, or serve as indicators of treatment responses [7].

Fig. 1.

AD pathology and associated fluid (CSF and plasma) biomarkers. Aβ, amyloid-β; AD, Alzheimer’s disease; CSF, cerebrospinal fluid; GFAP, glial fibrillary acidic protein; MTBR-tau, microtubule-binding region tau; NfL, neurofilament light; NTA-tau, N-terminal-associated tau; p-tau, phosphorylated tau.

Fig. 1.

AD pathology and associated fluid (CSF and plasma) biomarkers. Aβ, amyloid-β; AD, Alzheimer’s disease; CSF, cerebrospinal fluid; GFAP, glial fibrillary acidic protein; MTBR-tau, microtubule-binding region tau; NfL, neurofilament light; NTA-tau, N-terminal-associated tau; p-tau, phosphorylated tau.

Close modal

Biological sex may affect both the concentration and interpretation of biofluid biomarkers [6] (see Fig. 2). Certain markers may vary in concentration between males and females, which would necessitate sex-specific cut-offs. Sex might also influence interpretation of biomarkers due to sex-specific associations with clinical presentation, pathology, comorbidities, or progression. While sex differences in biofluid biomarkers have been reviewed previously [6], the last 5 years have been prolific for the AD biomarker field: novel and highly accurate blood-based biomarkers for amyloid and tau pathology have been developed and are already tested in primary care settings [11], and new assays specific to neurofibrillary tangles are emerging in both CSF and plasma [12‒14]. As such, this review expands previous work by considering the impact of biological sex on blood-based biomarkers and recently developed assays. In line with the AAW-criteria, focus will be on sex differences in fluid biomarkers for Aβ proteinopathy; phosphorylated and secreted AD tau; AD tau proteinopathy; neuronal injury; and inflammation (see Fig. 1).

Fig. 2.

Biological sex differences may influence AD and result in sex differences in the concentration and interpretation of AD fluid biomarkers. Sex differences may manifest as differences in absolute biomarker concentration or sex-specific associations with variables of interest such as brain atrophy and disease progression. Aβ, amyloid-β; AD, Alzheimer’s disease; APOE, apolipoprotein E; CSF, cerebrospinal fluid; GFAP, glial fibrillary acidic protein; MTBR-tau, microtubule-binding region tau; NfL, neurofilament light; NTA-tau, N-terminal-associated tau; p-tau, phosphorylated tau.

Fig. 2.

Biological sex differences may influence AD and result in sex differences in the concentration and interpretation of AD fluid biomarkers. Sex differences may manifest as differences in absolute biomarker concentration or sex-specific associations with variables of interest such as brain atrophy and disease progression. Aβ, amyloid-β; AD, Alzheimer’s disease; APOE, apolipoprotein E; CSF, cerebrospinal fluid; GFAP, glial fibrillary acidic protein; MTBR-tau, microtubule-binding region tau; NfL, neurofilament light; NTA-tau, N-terminal-associated tau; p-tau, phosphorylated tau.

Close modal

This review will focus on the effects of biological sex, defined by the presence and expression of XX (female) and XY (male) sex chromosomes. However, it should be noted that few studies explicitly state how sex is defined in their research, and some still use the terms sex and gender – the social roles associated with being a man or a woman – interchangeably.

Biological Sex Differences Influence Biofluid Biomarker Concentrations and Interpretations

Biological sex can influence the concentration and interpretation of biofluid biomarkers directly, by affecting normal CSF and plasma protein concentrations, or indirectly, through interactions with AD pathology (see Fig. 2). For example, sex differences in brain anatomy may affect biofluid biomarkers. Males have about 10% larger brain volume than females, but whether there are further sex differences across brain regions is debated [15]. However, due to their relatively larger brains, males have greater CSF volumes and larger ventricles. There are also apparent sex differences in the relative percentages of the brain composed of white and grey matter, with the former being higher in males and the latter being higher in females [16]. Sex differences in brain structure and function may contribute to differences in biofluid results for neurodegeneration biomarkers such as CSF neurofilament light chain (NfL), a marker of axonal degeneration. Moreover, these differences may contribute to sex differences in risk for cerebrovascular disease that may interact with or contribute to AD pathology [17].

There are several genetic risk factors for AD, with the apolipoprotein E (APOE) ε4 allele being the most common. Notably, APOE ε4 appears to interact with sex and increase AD risk more in heterozygous female carriers compared to male carriers, with no sex differences in homozygous carriers [18]. Moreover, female APOE ε4 carriers have higher levels of the AD biomarker CSF p-tau compared to male carriers and female noncarriers [19, 20]. The effect of APOE ε4 on sex differences in AD biomarkers appears to be stage dependent, with stronger sex differences in CSF p-tau at early stages for ε4 carriers and at later stages for noncarriers [21]. The APOE ε4 allele influences the relationship between biofluid biomarkers of Aβ and tau pathology, particularly in female carriers with stronger associations between CSF and blood Aβ biomarkers and p-tau biomarkers in carriers [22, 23].

Biological sex appears to interact with and affect AD pathology, in turn affecting the concentration and interpretation of associated biomarkers. Indeed, higher levels of tau deposition are apparent in female brains [24], even when accounting for Aβ plaque loads [25]. Moreover, Aβ and tau pathology are more closely related to brain atrophy and cognitive decline in females [26, 27]. As such, matched levels of Aβ and tau biomarkers might have different implications for males and females in terms of disease progression and clinical presentation, and sex-specific guidelines may be useful for clinical trials and treatment with anti-amyloid therapies [28].

The field of biofluid biomarkers for AD is highly active and in constant development. The discovery of accurate blood-based biomarkers of AD and their incorporation into diagnostic frameworks and clinical practice are poised to revolutionise AD research [7, 11]. Unfortunately, while studies of CSF and blood-based biomarkers typically include both male and female participants, sex differences are rarely examined directly, despite the potential impact of sex on fluid biomarker levels [5]. Rather, it is a common practice to adjust for sex in multivariate analysis, thus eliminating the impact of any sex difference on the results. Moreover, interactions between different AD biomarkers and biological or demographic variables such as sex, race, or socioeconomic status are rarely considered, despite their potential impact on disease stage, prognosis, or treatment responses. The sex differences in biomarkers considered herein are summarised in Table 1, highlighting both sex differences in absolute concentrations (e.g., consistently higher levels in men than in women) or sex-specific interactions with relevant variables such as cognitive decline (e.g., consistently worse memory performance for women with high biomarker levels compared to men).

Table 1.

Summary of the reviewed sex differences in concentration and sex-specific associations for biofluid biomarkers of Aβ pathology, p-tau, neuronal injury, and neuroinflammation

BiomarkerBiofluidSex differences in concentrationSex-specific associations
42 CSF No sex differences in CU [22, 29Sex-specific associations with hippocampal atrophy, memory performance, and executive function [26
No sex differences in sporadic AD [30‒32Sex-specific associations with CSF p-tau and AD risk in APOE ε4 carriers [19, 22, 29]; no sex-specific interactions with APOE ε4 [33
No sex differences in ADAD [34
Plasma No sex differences in CU [35, 36]; higher levels in CU women [37Sex-specific associations with AD risk and memory decline over time [37
42/40 CSF No sex differences in CU [33Sex and age influenced the relationship between CSF Aβ42/40 and disease progression [33
No sex differences in sporadic AD [30No sex-specific interactions with APOE ε4 [33
No sex differences in ADAD [34
Plasma No sex differences in CU [41]; Lower Aβ42/40 ratios in CU females [42 
No sex differences in sporadic AD [42
p-tau181 CSF No sex difference in CU [23Sex and age influenced the relationship between CSF p-tau181 and risk of MCI [39
No sex differences in sporadic AD [34, 35, 37No sex-specific interactions with APOE ε4 [39
No sex differences in ADAD [38
Plasma No sex differences in CU individuals [40, 41, 49‒51]; lower levels in CU females [53, 54Sex-specific association with Aβ deposition, entorhinal cortex tau deposition, CSF p-tau181, and cognitive decline [51]; no sex-specific association with amyloid load [40
No sex differences in MCI [50, 51Sex-specific association with incident dementia in dementia-free participants with Aβ pathology [51
No sex differences in sporadic AD [51, 52]; lower levels in MCI/dementia females [39, 53, 54
p-tau217 Plasma No sex differences in CU [41, 50, 55]; increased levels in male CU [49Sex-specific association with better global cognitive and memory performance in female CU ADAD mutation carriers [56
No sex differences in MCI or sporadic AD [49Sex-specific association with verbal memory and medial temporal lobe atrophy in CU females [55
No sex differences in ADAD [56
p-tau231 Plasma No sex differences in CU [50 
No sex differences in MCI [50
NfL CSF Higher concentration in males in sporadic AD and other neurodegenerative diseases [63‒66 
 Plasma No baseline sex differences in CU, MCI, or dementia patients [41, 42, 55, 58, 67Sex-specific association with future medial temporal lobe atrophy [55
Lower levels in female ADAD at younger ages [56Sex-specific association with age in female ADAD carriers [56
GFAP Plasma No sex differences in CU [66, 73]; higher levels in CU females [55, 72Sex-specific association with future medial temporal lobe atrophy [55
BiomarkerBiofluidSex differences in concentrationSex-specific associations
42 CSF No sex differences in CU [22, 29Sex-specific associations with hippocampal atrophy, memory performance, and executive function [26
No sex differences in sporadic AD [30‒32Sex-specific associations with CSF p-tau and AD risk in APOE ε4 carriers [19, 22, 29]; no sex-specific interactions with APOE ε4 [33
No sex differences in ADAD [34
Plasma No sex differences in CU [35, 36]; higher levels in CU women [37Sex-specific associations with AD risk and memory decline over time [37
42/40 CSF No sex differences in CU [33Sex and age influenced the relationship between CSF Aβ42/40 and disease progression [33
No sex differences in sporadic AD [30No sex-specific interactions with APOE ε4 [33
No sex differences in ADAD [34
Plasma No sex differences in CU [41]; Lower Aβ42/40 ratios in CU females [42 
No sex differences in sporadic AD [42
p-tau181 CSF No sex difference in CU [23Sex and age influenced the relationship between CSF p-tau181 and risk of MCI [39
No sex differences in sporadic AD [34, 35, 37No sex-specific interactions with APOE ε4 [39
No sex differences in ADAD [38
Plasma No sex differences in CU individuals [40, 41, 49‒51]; lower levels in CU females [53, 54Sex-specific association with Aβ deposition, entorhinal cortex tau deposition, CSF p-tau181, and cognitive decline [51]; no sex-specific association with amyloid load [40
No sex differences in MCI [50, 51Sex-specific association with incident dementia in dementia-free participants with Aβ pathology [51
No sex differences in sporadic AD [51, 52]; lower levels in MCI/dementia females [39, 53, 54
p-tau217 Plasma No sex differences in CU [41, 50, 55]; increased levels in male CU [49Sex-specific association with better global cognitive and memory performance in female CU ADAD mutation carriers [56
No sex differences in MCI or sporadic AD [49Sex-specific association with verbal memory and medial temporal lobe atrophy in CU females [55
No sex differences in ADAD [56
p-tau231 Plasma No sex differences in CU [50 
No sex differences in MCI [50
NfL CSF Higher concentration in males in sporadic AD and other neurodegenerative diseases [63‒66 
 Plasma No baseline sex differences in CU, MCI, or dementia patients [41, 42, 55, 58, 67Sex-specific association with future medial temporal lobe atrophy [55
Lower levels in female ADAD at younger ages [56Sex-specific association with age in female ADAD carriers [56
GFAP Plasma No sex differences in CU [66, 73]; higher levels in CU females [55, 72Sex-specific association with future medial temporal lobe atrophy [55

Biomarkers of neurofibrillary tau tangle pathology are not included as there were no data on sex differences available.

Aβ, amyloid-β; AD, Alzheimer’s disease; ADAD, autosomal dominant Alzheimer’s disease; APOE, apolipoprotein E; CU, cognitively unimpaired; GFAP, glial fibrillary acidic protein; MCI, mild cognitive impairment; NfL, neurofilament light; p-tau, phosphorylated tau.

Aβ Proteinopathy

Low levels of Aβ42 in the CSF are indicative of the presence of amyloid plaques in the brain. CSF Aβ42, or the CSF Aβ42/40 ratio, is used clinically to diagnose the presence of Aβ proteinopathy [30]. In CSF, there is about a 50%-fold change in the Aβ42/40 ratio in the presence of Aβ pathology, whereas in plasma the fold change is limited to 10–15% [7, 31‒33]. This limited fold change makes the Aβ42/40 ratio in plasma a less robust indicator of Aβ proteinopathy; other plasma assays discussed below are preferred for the diagnosis of AD pathology [7].

Several studies have found no sex differences in the concentrations of CSF Aβ42 or the Aβ42/40 ratio in sporadic AD [23, 34‒37] or autosomal dominant AD (ADAD) [38]. However, CSF Aβ42 appears to be differentially associated with hippocampal atrophy, memory performance, and CSF p-tau concentrations in males and females [23, 27]. One study found that while baseline levels of CSF Aβ42/40 did not differ between the sexes, age and sex interacted on the association between CSF Aβ42/40 and disease progression; lower CSF Aβ42/40 was associated with increased risk of disease progression in males, particularly at younger ages, but not in females, irrespective of age [39]. As such, CSF Aβ42 could have different prognostic performances, and in this context of use, sex-specific cut points or even alternative prognostic biomarkers might be required.

In plasma, significant sex differences have not been found for SIMOA or LC-MS plasma measurements of Aβ42 [40, 41]. One study found no differences in the Aβ42/40 ratio [41], whereas another found lower Aβ42/40 ratios in females compared to males [42]. Notably, the sex difference in plasma Aβ42/40 levels did not extend to patients with mild cognitive impairment (MCI) or dementia, suggesting that discrepancies between studies might be due to the disease stages studied [42]. Like findings in CSF, sex appears to influence the association between plasma Aβ42 and prognosis. In a longitudinal community-based study, higher plasma Aβ42 levels were associated with reduced future risk of AD in females, but not in males; conversely, lower plasma Aβ42 levels were associated with yearly memory decline exclusively in females [43].

Phosphorylated Tau

High levels of phosphorylated (mid-region) tau 181 in the CSF are used clinically as a biomarker of AD pathology [30]. Recently, different phosphorylated mid-region tau fragments (p-tau181, p-tau217, and p-tau231) have been identified as promising plasma biomarkers of AD pathology; especially, p-tau217 is emerging as an accurate and precise biomarker [44, 45]. Notably, these p-tau fragments become abnormal around the same time as amyloid PET and before the emergence of neurofibrillary tau tangles on tau PET [46, 47]. Secretion of tau fragments phosphorylated at the residues 181, 217, and 231 to plasma could thus represent a reaction to Aβ pathology, linking Aβ proteinopathy to the onset of tau proteinopathy in AD [48]. In the AAW-criteria, these p-tau biomarkers are noted T1 – biofluid analytes of soluble tau fragments that may reflect a reaction to amyloid plaques or soluble Aβ species in plaque penumbra.

Cross-sectional CSF studies of p-tau concentrations generally do not find sex differences [23, 34, 35, 37, 38]. However, females with low CSF Aβ may be more susceptible to increased p-tau concentrations [23, 27]. In line with this, female APOE ε4 carriers with CSF Aβ pathology appear to have higher levels of CSF p-tau181 compared to male carriers [20]. Nonetheless, another study found stronger associations between CSF p-tau181 and progression to MCI in males compared to females, but that this sex difference diminished with age [39].

Analysis of sex difference in plasma p-tau biomarkers has resulted in conflicting findings. Several studies have found no sex differences in plasma p-tau181 in cognitively unimpaired (CU) or MCI participants [40, 49‒51]; in extension, one multiethnic community study and one study of different neurodegenerative diseases also found no sex differences [41, 52]. However, two studies have found lower plasma p-tau181 levels in females compared to males, irrespective of cognitive status or diagnosis [53, 54]. Moreover, while one study found no interaction between sex and amyloid load on plasma p-tau181 levels [40], another found that despite similar plasma p-tau181 levels in males and females, females with higher p-tau181 at baseline had greater Aβ deposition, more entorhinal cortex tau deposition, higher CSF p-tau181, and faster cognitive decline in comparison with males [51]. In dementia-free participants with Aβ pathology, females with higher baseline plasma p-tau181 levels had higher rates of incident dementia relative to males [51].

For plasma p-tau217, several studies have found no sex differences in CU individuals [50, 55], a multiethnic community study [41], patients with different neurodegenerative diseases [52], or PSEN1 carriers [56]. However, one large population-based study found that plasma p-tau217 levels were significantly increased in males compared to females in CU participants, while there were no sex differences in the MCI and dementia groups [49]. In individuals with ADAD, sex and p-tau217 interacted on global cognitive and memory performance among CU mutation carriers, with female carriers performing better than males [56]; this effect was not present among CU noncarriers, suggesting a sex-specific cognitive resilience in female carriers to early accumulation of plasma p-tau217. One small study on 85 CU and 78 MCI participants suggests that a sex-specific association between cognitive performance and p-tau217 may also be present in sporadic AD. This study found that baseline plasma p-tau217 above the threshold indicating early levels of amyloid aggregation predicted verbal memory and medial temporal lobe atrophy in CU females, while CU males exhibited similar rates of decline and atrophy independent of plasma p-tau217 levels [55]. In line with findings from ADAD, these sex-specific interactions were specific to early stages and not present at the MCI stage [55]. Few studies have examined associations between biological sex and p-tau231, but one study examining several p-tau assays found no sex differences in p-tau231 in their cohort of CU and MCI participants over 70 years [50].

Neurofibrillary Tau Tangle Pathology

While the phosphorylated mid-region fragments at residues 181, 217, and 231 become abnormal around the same time as Aβ plaques begin to form, and correlate both with Aβ proteinopathy and tau proteinopathy, novel assays of different tau fragment analytes such as p-tau205, N-terminal-associated tau (NTA-tau) fragments, or microtubule-binding region tau (MTBR-tau243) become abnormal later and correspond better to tau PET [13‒15]. These, together with tau PET, are categorised as T2 markers – markers of AD tau proteinopathy, in the AA framework [7]. These novel biomarkers hold great promise for the study of AD tau proteinopathy across disease stages and in response to treatment interventions, but unfortunately, no study has yet examined sex differences in NTA-tau, p-tau205, or MTBR-tau243, nor if there are sex-specific interactions between these biomarkers and other markers of AD pathology, AD clinical symptoms, prognosis, or other relevant factors. Given accumulating evidence suggesting sex differences in tau proteinopathy in AD evident on tau PET [26, 28], this is a fruitful avenue for further research.

In the 2018 NIA-AA research framework for AD, CSF levels of total tau were considered biomarkers of neurodegeneration [11]. However, in the most recent AA framework, total tau is not included as a fluid biomarker of neurodegeneration [57]. Increased levels of plasma or CSF total tau are complex to interpret: on the one hand, CSF and plasma total tau increase early in AD and correlate closely with fluid p-tau, suggesting that total tau could be a biomarker of tau proteinopathy. On the other hand, total tau is not specific to AD and increases in response to, for example, head trauma, cerebral infarction, and proteinopathies such as Creutzfeldt-Jacob disease, suggesting that it is rather a marker of neurodegeneration. As such, sex differences in CSF or total tau could be indicative of sex differences in tau proteinopathy, neurodegeneration, or potentially other comorbidities that influence total tau levels. Highlighting this challenge is a host of contrasting findings on sex differences in total tau biomarkers, with some studies finding lower CSF total tau in males [27, 37] and others in females [38]. Similarly, while some studies have reported higher plasma total tau levels in females [42, 58, 59], other studies have found no sex differences [60, 61].

Neuronal Injury

Neurodegeneration is a key step in the progression of AD pathology but is also central in other forms of dementia such as Lewy body disorder or frontotemporal dementia. As such, abnormalities in biomarkers of neuronal injury – the N category – are not specific to AD and occur in several other conditions such as non-AD neurodegenerative diseases, after traumatic injury and after ischemic injury among others [7]. NfL is a marker of axonal injury and a reliable marker of neurodegeneration that may be measured in plasma and CSF. Plasma and CSF levels of NfL are strongly correlated with each other and similarly associate with markers of brain atrophy such as cortical thinning or white matter atrophy [62].

Several studies have reported higher levels of CSF NfL in males with AD and in other neurodegenerative diseases [63‒66]. This difference could be due to greater vascular pathology in males or a larger proportion of brain white matter, but the exact cause is not known.

Notably, despite the strong correlation between plasma and CSF NfL levels, the sex differences in CSF NfL levels do not appear to translate to findings in plasma, with several studies reporting no baseline sex differences in CU, MCI, or dementia patients in plasma NfL levels [41, 42, 55, 58, 67]. The cause of discordance between CSF and plasma NfL levels is not known. This underscores that beyond examining correspondence between plasma, CSF, and neuroimaging biomarkers, examining sex differences in these modalities is important to understand their utility in picking up sex-specific disease trajectories and the need for sex-specific guidelines.

Few studies have examined sex-specific associations between plasma NfL and AD-related outcomes such as brain atrophy or clinical symptoms. One study found that higher NfL was associated with more medial temporal lobe atrophy over time exclusively in females but did not find evidence of sex-specific effects on cognitive trajectories [55]. This highlights potentially different prognostic utilities of the plasma NfL as a marker of neurodegeneration in males and females. In ADAD, female PSEN1 carriers had lower NfL levels at younger ages but show greater increases with age compared to male carriers [56].

Neuroinflammation

Like markers of neuronal injury, biomarkers of neuroinflammation represent markers of nonspecific processes involved in AD pathology. A growing body of evidence shows that immune and inflammatory mechanisms are important in AD, with both reactive astrocytes and active microglia being linked to amyloid and tau pathology [9]. Biomarkers of inflammation might reflect either astrocyte or microglia activity. Biomarkers of a vast range of inflammatory pathways have been intensively studied in CSF, serum, and plasma, with apparent sex differences in, for example, systemic inflammation [68], the kynurenine pathway [69], matrix metalloproteinases [70], and microglial activity [71]. However, there is still no consensus on reliable biomarkers of AD-associated inflammation. One marker that has been incorporated in the AAW-criteria under the I – inflammation category – is glial fibrillary acid protein (GFAP), a marker of astrocytic reactivity.

GFAP can be measured in plasma or CSF, but plasma levels appear to be a more robust indicator of Aβ pathology [72]. While altered GFAP levels are not specific to AD, they are associated with early amyloid PET, higher risk of incident dementia, and faster rates of cognitive decline [47, 73, 74]. Some studies report no significant associations between sex and plasma GFAP levels [66, 73], while others have found elevated plasma GFAP in CU females compared to males [55, 72] and female-specific associations with future medial temporal lobe atrophy [55].

While some AD biomarkers display consistent differences in concentration between males and females, others do not appear to differ by sex (Table 1). Remarkably, even biomarkers that show no baseline difference display sex-specific associations with brain atrophy, cognitive performance, and/or disease progression (Table 1). For biomarkers with consistent sex differences in concentration, such as CSF NfL levels, sex-specific cut-offs may be appropriate. For the biomarkers showing sex-specific associations with relevant clinical variables such as disease progression, more research is required to develop sex-specific guidelines for different contexts of use, such as clinical trial enrolment or disease staging. This has key implications for future research.

Firstly, biomarker data should be reported separately for males and females, and group differences should be assessed if the sample size is adequate. This is especially important for emerging biomarkers such as NTA-tau or MTBR-tau243, where little is known about sex differences in concentration. Centrally, beyond examining group differences in biomarkers, sex-specific associations with variables of interest should be explored in well-powered studies. Furthermore, research should consider the upstream factors influencing these sex differences. From a precision medicine perspective, research should reach beyond enrolling equal numbers of males and females towards considering sex-specific biology that may interact with disease pathology [1]. Factors such as menopausal status; hormonal contraception or hormone treatment; reproductive events; genetics and sex-genotype interactions; and sex differences in the pattern of comorbidities [6, 20, 75] may be relevant for disease aetiology, prognosis, and treatment responses. For example, in cognitively normal populations, perimenopausal and postmenopausal women exhibit increased amyloid deposition and reduced brain volume in AD-related brain regions compared to premenopausal females and age-matched males; this difference is more pronounced in APOE ε4 carriers [76]. This indicates a progressive increase in AD risk as females undergo menopausal changes, implying that endocrine ageing may exacerbate the effects of chronologic ageing in the female brain [76]. Females who experience early menopause (naturally or iatrogenic) represent the highest risk cluster for late-life cognitive decline and dementia within the menopause population [1, 77]. As a result, studies examining sex differences at the preclinical stage or in ADAD may find divergent results if their proportion of pre-, peri-, and postmenopausal females differ notably.

Beyond examining the effects of biological sex, an intersectional approach may be required to fully determine the impact of sex and gender on AD. The concepts of sex and gender interact to influence health and disease, and their effects may also differ depending on social and demographic variables such as geography, culture, race and ethnicity, age, or socioeconomic status [78‒80]. Disentangling these concepts in research is a significant challenge but may enrich our understanding of AD and other diseases. Indeed, attempts to create gender metrics based on data such as education levels and self-reported depression have been linked to cardiovascular health, with higher “femininity scores” being associated with an angina diagnosis predating a heart attack in men in the UK biobank [81]. It should be noted that the current review is not a systematic review and, therefore, may exclude relevant articles. As such, future systematic reviews and meta-analysis on sex differences in biofluid biomarkers should be conducted. While sample size and power are central challenges when studying the effects of biological sex differences and interacting identities such as gender and race, more nuanced reporting of these categories could lead to future well-powered meta-analyses.

Biological sex influences the interpretation of biofluid biomarkers, even for biomarkers where baseline sex differences in absolute levels are not consistently reported. Potential sex-specific guidelines may be required depending on context of use; this is becoming increasingly relevant with the growing emphasis on biomarkers in the diagnosis and staging of AD. Intensive work is ongoing to validate and harmonise blood-based biomarkers across different assays as diagnostic tools in specialist memory clinics and primary care. The effects of sex should be considered in this work. The biomarker field should move beyond controlling for the effects of sex to viewing it as a variable of interest, and sex differences and sex-specific interactions should be explored. Ultimately, future research should aim to identify the upstream mechanisms driving interactions between sex biology and disease pathology.

The author has no conflicts of interest to declare.

This work was funded by the Norwegian Health Association (Grant No. 25633). The funder had no role in the manuscript conception, planning, writing, or decision to publish.

M.A. conceived and wrote the manuscript.

1.
Ferretti
MT
,
Iulita
MF
,
Cavedo
E
,
Chiesa
PA
,
Schumacher Dimech
A
,
Santuccione Chadha
A
, et al
.
Sex differences in Alzheimer disease: the gateway to precision medicine
.
Nat Rev Neurol
.
2018
;
14
(
8
):
457
69
.
2.
Chêne
G
,
Beiser
A
,
Au
R
,
Preis
SR
,
Wolf
PA
,
Dufouil
C
, et al
.
Gender and incidence of dementia in the Framingham Heart Study from mid-adult life
.
Alzheimers Dement
.
2015
;
11
(
3
):
310
20
.
3.
Alzheimer’s disease facts and figures
.
Alzheimer Dementia
.
2023
;
19
(
4
):
1598
695
.
4.
Saleh
RNM
,
Hornberger
M
,
Ritchie
CW
,
Minihane
AM
.
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
.
2023
;
15
(
1
):
10
.
5.
Mielke
MM
,
Aggarwal
NT
,
Vila-Castelar
C
,
Agarwal
P
,
Arenaza-Urquijo
EM
,
Brett
B
, et al
.
Consideration of sex and gender in Alzheimer’s disease and related disorders from a global perspective
.
Alzheimers Dement
.
2022
;
18
(
12
):
2707
24
.
6.
Mielke
MM
.
Consideration of sex differences in the measurement and interpretation of alzheimer disease-related biofluid-based biomarkers
.
J Appl Lab Med
.
2020
;
5
(
1
):
158
69
.
7.
Jack
CR
Jr
,
Andrews
JS
,
Beach
TG
,
Buracchio
T
,
Dunn
B
,
Graf
A
, et al
.
Revised criteria for diagnosis and staging of Alzheimer’s disease: alzheimer’s Association Workgroup
.
Alzheimers Dement
.
2024
;
20
(
8
):
5143
69
.
8.
Serrano-Pozo
A
,
Frosch
MP
,
Masliah
E
,
Hyman
BT
.
Neuropathological alterations in Alzheimer disease
.
Cold Spring Harb Perspect Med
.
2011
;
1
(
1
):
a006189
.
9.
Heneka
MT
,
Carson
MJ
,
El Khoury
J
,
Landreth
GE
,
Brosseron
F
,
Feinstein
DL
, et al
.
Neuroinflammation in Alzheimer’s disease
.
Lancet Neurol
.
2015
;
14
(
4
):
388
405
.
10.
Fang
EF
,
Hou
Y
,
Palikaras
K
,
Adriaanse
BA
,
Kerr
JS
,
Yang
B
, et al
.
Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease
.
Nat Neurosci
.
2019
;
22
(
3
):
401
12
.
11.
Jack
CR
Jr
,
Bennett
DA
,
Blennow
K
,
Carrillo
MC
,
Dunn
B
,
Haeberlein
SB
, et al
.
NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease
.
Alzheimers Dement
.
2018
;
14
(
4
):
535
62
.
12.
Palmqvist
S
,
Tideman
P
,
Mattsson-Carlgren
N
,
Schindler
SE
,
Smith
R
,
Ossenkoppele
R
, et al
.
Blood biomarkers to detect alzheimer disease in primary care and secondary care
.
Jama
.
2024
;
332
(
15
):
1245
57
.
13.
Horie
K
,
Barthélemy
NR
,
Spina
S
,
VandeVrede
L
,
He
Y
,
Paterson
RW
, et al
.
CSF tau microtubule-binding region identifies pathological changes in primary tauopathies
.
Nat Med
.
2022
;
28
(
12
):
2547
54
.
14.
Horie
K
,
Salvadó
G
,
Barthélemy
NR
,
Janelidze
S
,
Li
Y
,
He
Y
, et al
.
CSF MTBR-tau243 is a specific biomarker of tau tangle pathology in Alzheimer’s disease
.
Nat Med
.
2023
;
29
(
8
):
1954
63
.
15.
Lantero-Rodriguez
J
,
Salvadó
G
,
Snellman
A
,
Montoliu-Gaya
L
,
Brum
WS
,
Benedet
AL
, et al
.
Plasma N-terminal containing tau fragments (NTA-tau): a biomarker of tau deposition in Alzheimer’s Disease
.
Mol Neurodegener
.
2024
;
19
(
1
):
19
.
16.
DeCasien
AR
,
Guma
E
,
Liu
S
,
Raznahan
A
.
Sex differences in the human brain: a roadmap for more careful analysis and interpretation of a biological reality
.
Biol Sex Differ
.
2022
;
13
(
1
):
43
.
17.
Gur
RC
,
Turetsky
BI
,
Matsui
M
,
Yan
M
,
Bilker
W
,
Hughett
P
, et al
.
Sex differences in brain gray and white matter in healthy young adults: correlations with cognitive performance
.
J Neurosci
.
1999
;
19
(
10
):
4065
72
.
18.
Morrison
C
,
Dadar
M
,
Collins
DL
,
Alzheimer’s Disease Neuroimaging Initiative
.
Sex differences in risk factors, burden, and outcomes of cerebrovascular disease in Alzheimer’s disease populations
.
Alzheimers Dement
.
2024
;
20
(
1
):
34
46
.
19.
Neu
SC
,
Pa
J
,
Kukull
W
,
Beekly
D
,
Kuzma
A
,
Gangadharan
P
, et al
.
Apolipoprotein E genotype and sex risk factors for alzheimer disease: a meta-analysis
.
JAMA Neurol
.
2017
;
74
(
10
):
1178
89
.
20.
Hohman
TJ
,
Dumitrescu
L
,
Barnes
LL
,
Thambisetty
M
,
Beecham
G
,
Kunkle
B
, et al
.
Sex-specific association of apolipoprotein E with cerebrospinal fluid levels of tau
.
JAMA Neurol
.
2018
;
75
(
8
):
989
98
.
21.
Duarte-Guterman
P
,
Albert
AY
,
Barha
CK
,
Galea
LAM
;
On Behalf Of The Alzheimer’s Disease Neuroimaging Initiative
.
Sex influences the effects of APOE genotype and Alzheimer’s diagnosis on neuropathology and memory
.
Psychoneuroendocrinology
.
2021
;
129
:
105248
.
22.
Babapour Mofrad
R
,
Tijms
BM
,
Scheltens
P
,
Barkhof
F
,
van der Flier
WM
,
Sikkes
SAM
, et al
.
Sex differences in CSF biomarkers vary by Alzheimer disease stage and APOE ε4 genotype
.
Neurology
.
2020
;
95
(
17
):
e2378
88
.
23.
Buckley
RF
,
Mormino
EC
,
Chhatwal
J
,
Schultz
AP
,
Rabin
JS
,
Rentz
DM
, et al
.
Associations between baseline amyloid, sex, and APOE on subsequent tau accumulation in cerebrospinal fluid
.
Neurobiol Aging
.
2019
;
78
:
178
85
.
24.
Cai
Y
,
Fu
P
,
Zhang
X
,
Alzheimer’s Disease Neuroimaging Initiative
.
Association of plasma phosphor-tau181 with Aβ levels may vary by APOE ε4 status and sex among non-demented old adults
.
Neurosci Lett
.
2023
;
802
:
137161
.
25.
Liesinger
AM
,
Graff-Radford
NR
,
Duara
R
,
Carter
RE
,
Hanna Al-Shaikh
FS
,
Koga
S
, et al
.
Sex and age interact to determine clinicopathologic differences in Alzheimer’s disease
.
Acta Neuropathol
.
2018
;
136
(
6
):
873
85
.
26.
Buckley
RF
,
Mormino
EC
,
Rabin
JS
,
Hohman
TJ
,
Landau
S
,
Hanseeuw
BJ
, 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
.
2019
;
76
(
5
):
542
51
.
27.
Koran
MEI
,
Wagener
M
,
Hohman
TJ
,
Alzheimer’s Neuroimaging Initiative
.
Sex differences in the association between AD biomarkers and cognitive decline
.
Brain Imaging Behav
.
2017
;
11
(
1
):
205
13
.
28.
Buckley
RF
,
Scott
MR
,
Jacobs
HIL
,
Schultz
AP
,
Properzi
MJ
,
Amariglio
RE
, et al
.
Sex mediates relationships between regional tau pathology and cognitive decline
.
Ann Neurol
.
2020
;
88
(
5
):
921
32
.
29.
Wang
Y-T
,
Therriault
J
,
Servaes
S
,
Tissot
C
,
Rahmouni
N
,
Macedo
AC
, et al
.
Sex-specific modulation of amyloid-β on tau phosphorylation underlies faster tangle accumulation in females
.
Brain
.
2024
;
147
(
4
):
1497
510
.
30.
McKhann
GM
,
Knopman
DS
,
Chertkow
H
,
Hyman
BT
,
Jack
CR
Jr
,
Kawas
CH
, et al
.
The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease
.
Alzheimers Dement
.
2011
;
7
(
3
):
263
9
.
31.
Schindler
SE
,
Bollinger
JG
,
Ovod
V
,
Mawuenyega
KG
,
Li
Y
,
Gordon
BA
, et al
.
High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis
.
Neurology
.
2019
;
93
(
17
):
e1647
59
.
32.
Hansson
O
,
Seibyl
J
,
Stomrud
E
,
Zetterberg
H
,
Trojanowski
JQ
,
Bittner
T
, et al
.
CSF biomarkers of Alzheimer’s disease concord with amyloid-β PET and predict clinical progression: a study of fully automated immunoassays in BioFINDER and ADNI cohorts
.
Alzheimers Dement
.
2018
;
14
(
11
):
1470
81
.
33.
Janelidze
S
,
Teunissen
CE
,
Zetterberg
H
,
Allué
JA
,
Sarasa
L
,
Eichenlaub
U
, et al
.
Head-to-Head comparison of 8 plasma amyloid-β 42/40 assays in alzheimer disease
.
JAMA Neurol
.
2021
;
78
(
11
):
1375
82
.
34.
Bouter
C
,
Vogelgsang
J
,
Wiltfang
J
.
Comparison between amyloid-PET and CSF amyloid-β biomarkers in a clinical cohort with memory deficits
.
Clin Chim Acta
.
2019
;
492
:
62
8
.
35.
Schoonenboom
NS
,
Pijnenburg
YAL
,
Mulder
C
,
Rosso
SM
,
Van Elk
EJ
,
Van Kamp
GJ
, et al
.
Amyloid beta(1-42) and phosphorylated tau in CSF as markers for early-onset Alzheimer disease
.
Neurology
.
2004
;
62
(
9
):
1580
4
.
36.
Li
G
,
Shofer
JB
,
Petrie
EC
,
Yu
CE
,
Wilkinson
CW
,
Figlewicz
DP
, et al
.
Cerebrospinal fluid biomarkers for Alzheimer’s and vascular disease vary by age, gender, and APOE genotype in cognitively normal adults
.
Alzheimers Res Ther
.
2017
;
9
(
1
):
48
.
37.
Cousins
KAQ
,
Arezoumandan
S
,
Shellikeri
S
,
Ohm
D
,
Shaw
LM
,
Grossman
M
, et al
.
CSF biomarkers of alzheimer disease in patients with concomitant α-synuclein pathology
.
Neurology
.
2022
;
99
(
20
):
e2303
12
.
38.
Wagemann
O
,
Li
Y
,
Hassenstab
J
,
Aschenbrenner
AJ
,
McKay
NS
,
Gordon
BA
, et al
.
Investigation of sex differences in mutation carriers of the dominantly inherited alzheimer network
.
Alzheimers Dement
.
2024
;
20
(
1
):
47
62
.
39.
Greenberg
BD
,
Pettigrew
C
,
Soldan
A
,
Wang
J
,
Wang
MC
,
Darrow
JA
, et al
.
CSF alzheimer disease biomarkers: time-varying relationships with MCI symptom onset and associations with age, sex, and ApoE4
.
Neurology
.
2022
;
99
(
15
):
e1640
50
.
40.
Keshavan
A
,
Pannee
J
,
Karikari
TK
,
Rodriguez
JL
,
Ashton
NJ
,
Nicholas
JM
, et al
.
Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70
.
Brain
.
2021
;
144
(
2
):
434
49
.
41.
Brickman
AM
,
Manly
JJ
,
Honig
LS
,
Sanchez
D
,
Reyes-Dumeyer
D
,
Lantigua
RA
, et al
.
Plasma p-tau181, p-tau217, and other blood-based Alzheimer’s disease biomarkers in a multi-ethnic, community study
.
Alzheimers Dement
.
2021
;
17
(
8
):
1353
64
.
42.
Syrjanen
JA
,
Campbell
MR
,
Algeciras-Schimnich
A
,
Vemuri
P
,
Graff-Radford
J
,
Machulda
MM
, et al
.
Associations of amyloid and neurodegeneration plasma biomarkers with comorbidities
.
Alzheimers Dement
.
2022
;
18
(
6
):
1128
40
.
43.
Liu
C
,
Li
Y
,
Nwosu
A
,
Ang
TFA
,
Liu
Y
,
Devine
S
, et al
.
Sex-specific biomarkers in alzheimer’s disease progression: framingham heart study
.
Alzheimers Dement
.
2022
;
14
(
1
):
e12369
.
44.
Barthélemy
NR
,
Salvadó
G
,
Schindler
SE
,
He
Y
,
Janelidze
S
,
Collij
LE
, et al
.
Highly accurate blood test for Alzheimer’s disease is similar or superior to clinical cerebrospinal fluid tests
.
Nat Med
.
2024
;
30
(
4
):
1085
95
.
45.
Janelidze
S
,
Bali
D
,
Ashton
NJ
,
Barthélemy
NR
,
Vanbrabant
J
,
Stoops
E
, et al
.
Head-to-head comparison of 10 plasma phospho-tau assays in prodromal Alzheimer’s disease
.
Brain
.
2023
;
146
(
4
):
1592
601
.
46.
Janelidze
S
,
Berron
D
,
Smith
R
,
Strandberg
O
,
Proctor
NK
,
Dage
JL
, et al
.
Associations of plasma phospho-tau217 levels with tau positron emission tomography in early alzheimer disease
.
JAMA Neurol
.
2021
;
78
(
2
):
149
56
.
47.
Hansson
O
,
Blennow
K
,
Zetterberg
H
,
Dage
J
.
Blood biomarkers for Alzheimer’s disease in clinical practice and trials
.
Nat Aging
.
2023
;
3
(
5
):
506
19
.
48.
Mattsson-Carlgren
N
,
Andersson
E
,
Janelidze
S
,
Ossenkoppele
R
,
Insel
P
,
Strandberg
O
, et al
.
Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive Tau PET in Alzheimer’s disease
.
Sci Adv
.
2020
;
6
(
16
):
eaaz2387
.
49.
Mielke
MM
,
Dage
JL
,
Frank
RD
,
Algeciras-Schimnich
A
,
Knopman
DS
,
Lowe
VJ
, et al
.
Performance of plasma phosphorylated tau 181 and 217 in the community
.
Nat Med
.
2022
;
28
(
7
):
1398
405
.
50.
Mielke
MM
,
Frank
RD
,
Dage
JL
,
Jeromin
A
,
Ashton
NJ
,
Blennow
K
, et al
.
Comparison of plasma phosphorylated tau species with amyloid and tau positron emission tomography, neurodegeneration, vascular pathology, and cognitive outcomes
.
JAMA Neurol
.
2021
;
78
(
9
):
1108
17
.
51.
Tsiknia
AA
,
Edland
SD
,
Sundermann
EE
,
Reas
ET
,
Brewer
JB
,
Galasko
D
, et al
.
Sex differences in plasma p-tau181 associations with Alzheimer’s disease biomarkers, cognitive decline, and clinical progression
.
Mol Psychiatry
.
2022
;
27
(
10
):
4314
22
.
52.
Thijssen
EH
,
La Joie
R
,
Wolf
A
,
Strom
A
,
Wang
P
,
Iaccarino
L
, et al
.
Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration
.
Nat Med
.
2020
;
26
(
3
):
387
97
.
53.
Ramanan
VK
,
Graff-Radford
J
,
Syrjanen
J
,
Shir
D
,
Algeciras-Schimnich
A
,
Lucas
J
, et al
.
Association of plasma biomarkers of alzheimer disease with cognition and medical comorbidities in a biracial cohort
.
Neurology
.
2023
;
101
(
14
):
e1402
11
.
54.
Lussier
FZ
,
Benedet
AL
,
Therriault
J
,
Pascoal
TA
,
Tissot
C
,
Chamoun
M
, et al
.
Plasma levels of phosphorylated tau 181 are associated with cerebral metabolic dysfunction in cognitively impaired and amyloid-positive individuals
.
Brain Commun
.
2021
;
3
(
2
):
fcab073
.
55.
Saloner
R
,
VandeVrede
L
,
Asken
BM
,
Paolillo
EW
,
Gontrum
EQ
,
Wolf
A
, et al
.
Plasma phosphorylated tau-217 exhibits sex-specific prognostication of cognitive decline and brain atrophy in cognitively unimpaired adults
.
Alzheimers Dement
.
2024
;
20
(
1
):
376
87
.
56.
Vila-Castelar
C
,
Chen
Y
,
Langella
S
,
Lopera
F
,
Zetterberg
H
,
Hansson
O
, et al
.
Sex differences in blood biomarkers and cognitive performance in individuals with autosomal dominant Alzheimer’s disease
.
Alzheimers Dement
.
2023
;
19
(
9
):
4127
38
.
57.
Johnson
ECB
,
Dammer
EB
,
Duong
DM
,
Ping
L
,
Zhou
M
,
Yin
L
, et al
.
Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation
.
Nat Med
.
2020
;
26
(
5
):
769
80
.
58.
Baldacci
F
,
Lista
S
,
Manca
ML
,
Chiesa
PA
,
Cavedo
E
,
Lemercier
P
, et al
.
Age and sex impact plasma NFL and t-Tau trajectories in individuals with subjective memory complaints: a 3-year follow-up study
.
Alzheimers Res Ther
.
2020
;
12
(
1
):
147
.
59.
Pase
MP
,
Beiser
AS
,
Himali
JJ
,
Satizabal
CL
,
Aparicio
HJ
,
DeCarli
C
, et al
.
Assessment of plasma total tau level as a predictive biomarker for dementia and related endophenotypes
.
JAMA Neurol
.
2019
;
76
(
5
):
598
606
.
60.
Mattsson
N
,
Zetterberg
H
,
Janelidze
S
,
Insel
PS
,
Andreasson
U
,
Stomrud
E
, et al
.
Plasma tau in Alzheimer disease
.
Neurology
.
2016
;
87
(
17
):
1827
35
.
61.
Dage
JL
,
Wennberg
AMV
,
Airey
DC
,
Hagen
CE
,
Knopman
DS
,
Machulda
MM
, et al
.
Levels of tau protein in plasma are associated with neurodegeneration and cognitive function in a population-based elderly cohort
.
Alzheimers Dement
.
2016
;
12
(
12
):
1226
34
.
62.
Mielke
MM
,
Syrjanen
JA
,
Blennow
K
,
Zetterberg
H
,
Vemuri
P
,
Skoog
I
, et al
.
Plasma and CSF neurofilament light: relation to longitudinal neuroimaging and cognitive measures
.
Neurology
.
2019
;
93
(
3
):
e252
60
.
63.
Mattsson
N
,
Insel
PS
,
Palmqvist
S
,
Portelius
E
,
Zetterberg
H
,
Weiner
M
, et al
.
Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer’s disease
.
EMBO Mol Med
.
2016
;
8
(
10
):
1184
96
.
64.
Mielke
MM
,
Syrjanen
JA
,
Blennow
K
,
Zetterberg
H
,
Skoog
I
,
Vemuri
P
, et al
.
Comparison of variables associated with cerebrospinal fluid neurofilament, total-tau, and neurogranin
.
Alzheimers Dement
.
2019
;
15
(
11
):
1437
47
.
65.
Bridel
C
,
van Wieringen
WN
,
Zetterberg
H
,
Tijms
BM
,
Teunissen
CE
,
and the NFL Group
, et al
.
Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: a systematic review and meta-analysis
.
JAMA Neurol
.
2019
;
76
(
9
):
1035
48
.
66.
Pelkmans
W
,
Shekari
M
,
Brugulat-Serrat
A
,
Sánchez-Benavides
G
,
Minguillón
C
,
Fauria
K
, et al
.
Astrocyte biomarkers GFAP and YKL-40 mediate early Alzheimer’s disease progression
.
Alzheimers Dement
.
2024
;
20
(
1
):
483
93
.
67.
Mattsson
N
,
Cullen
NC
,
Andreasson
U
,
Zetterberg
H
,
Blennow
K
.
Association between longitudinal plasma neurofilament light and neurodegeneration in patients with alzheimer disease
.
JAMA Neurol
.
2019
;
76
(
7
):
791
9
.
68.
Walker
KA
,
Windham
BG
,
Brown
CH
,
Knopman
DS
,
Jack
CR
,
Mosley
TH
, et al
.
The association of mid- and late-life systemic inflammation with brain amyloid deposition: the ARIC-PET study
.
J Alzheimers Dis
.
2018
;
66
(
3
):
1041
52
.
69.
Knapskog
AB
,
Edwin
TH
,
Ueland
PM
,
Ulvik
A
,
Fang
EF
,
Eldholm
RS
, et al
.
Sex-specific associations of kynurenic acid with neopterin in Alzheimer’s disease
.
Alzheimers Res Ther
.
2024
;
16
(
1
):
167
.
70.
Aksnes
M
,
Edwin
TH
,
Saltvedt
I
,
Eldholm
RS
,
Chaudhry
FA
,
Halaas
NB
, et al
.
Sex-specific associations of matrix metalloproteinases in Alzheimer’s disease
.
Biol Sex Differ
.
2023
;
14
(
1
):
35
.
71.
Casaletto
KB
,
Nichols
E
,
Aslanyan
V
,
Simone
SM
,
Rabin
JS
,
La Joie
R
, et al
.
Sex-specific effects of microglial activation on Alzheimer’s disease proteinopathy in older adults
.
Brain
.
2022
;
145
(
10
):
3536
45
.
72.
Benedet
AL
,
Milà-Alomà
M
,
Vrillon
A
,
Ashton
NJ
,
Pascoal
TA
,
Lussier
F
, et al
.
Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the alzheimer disease continuum
.
JAMA Neurol
.
2021
;
78
(
12
):
1471
83
.
73.
Chatterjee
P
,
Pedrini
S
,
Stoops
E
,
Goozee
K
,
Villemagne
VL
,
Asih
PR
, et al
.
Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease
.
Transl Psychiatry
.
2021
;
11
(
1
):
27
.
74.
Cicognola
C
,
Janelidze
S
,
Hertze
J
,
Zetterberg
H
,
Blennow
K
,
Mattsson-Carlgren
N
, et al
.
Plasma glial fibrillary acidic protein detects Alzheimer pathology and predicts future conversion to Alzheimer dementia in patients with mild cognitive impairment
.
Alzheimers Res Ther
.
2021
;
13
(
1
):
68
.
75.
Sundermann
EE
,
Panizzon
MS
,
Chen
X
,
Andrews
M
,
Galasko
D
,
Banks
SJ
, et al
.
Sex differences in Alzheimer’s-related Tau biomarkers and a mediating effect of testosterone
.
Biol Sex Differ
.
2020
;
11
(
1
):
33
.
76.
Mosconi
L
,
Berti
V
,
Quinn
C
,
McHugh
P
,
Petrongolo
G
,
Varsavsky
I
, et al
.
Sex differences in Alzheimer risk: brain imaging of endocrine vs chronologic aging
.
Neurology
.
2017
;
89
(
13
):
1382
90
.
77.
Nebel
RA
,
Aggarwal
NT
,
Barnes
LL
,
Gallagher
A
,
Goldstein
JM
,
Kantarci
K
, et al
.
Understanding the impact of sex and gender in Alzheimer’s disease: a call to action
.
Alzheimers Dement
.
2018
;
14
(
9
):
1171
83
.
78.
Subramaniapillai
S
,
Am Galea
L
,
Einstein
G
,
de Lange
AM
.
Sex and gender in health research: intersectionality matters
.
Front Neuroendocrinol
.
2023
;
72
:
101104
.
79.
Misiura
MB
,
Butts
B
,
Hammerschlag
B
,
Munkombwe
C
,
Bird
A
,
Fyffe
M
, et al
.
Intersectionality in alzheimer’s disease: the role of female sex and black American race in the development and prevalence of alzheimer’s disease
.
Neurotherapeutics
.
2023
;
20
(
4
):
1019
36
.
80.
van der Velpen
IF
,
Yaqub
A
,
Vernooij
MW
,
Perry
M
,
Vernooij-Dassen
MJF
,
Ghanbari
M
, et al
.
Sex-differences in the association of social health and marital status with blood-based immune and neurodegeneration markers in a cohort of community-dwelling older adults
.
Brain Behav Immun
.
2024
;
120
:
71
81
.
81.
Levinsson
A
,
de Denus
S
,
Sandoval
J
,
Lemieux Perreault
LP
,
Rouleau
J
,
Tardif
JC
, et al
.
Construction of a femininity score in the UK Biobank and its association with angina diagnosis prior to myocardial infarction
.
Sci Rep
.
2022
;
12
(
1
):
1780
.