Abstract
INTRODUCTION
Blood biomarkers have proven useful in Alzheimer's disease (AD) research. However, little is known about their biological variation (BV), which improves the interpretation of individual‐level data.
METHODS
We measured plasma amyloid beta (Aβ42, Aβ40), phosphorylated tau (p‐tau181, p‐tau217, p‐tau231), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL) in plasma samples collected weekly over 10 weeks from 20 participants aged 40 to 60 years from the European Biological Variation Study. We estimated within‐ (CVI) and between‐subject (CVG) BV, analytical variation, and reference change values (RCV).
RESULTS
Biomarkers presented considerable variability in CVI and CVG. Aβ42/Aβ40 had the lowest CVI (≈ 3%) and p‐tau181 the highest (≈ 16%), while others ranged from 6% to 10%. Most RCVs ranged from 20% to 30% (decrease) and 25% to 40% (increase).
DISCUSSION
BV estimates for AD plasma biomarkers can potentially refine their clinical and research interpretation. RCVs might be useful for detecting significant changes between serial measurements when monitoring early disease progression or interventions.
Highlights
Plasma amyloid beta (Aβ42/Aβ40) presents the lowest between‐ and within‐subject biological variation, but also changes the least in Alzheimer's disease (AD) patients versus controls.
Plasma phosphorylated tau variants significantly vary in their within‐subject biological variation, but their substantial fold‐changes in AD likely limits the impact of their variability.
Plasma neurofilament light chain and glial fibrillary acidic protein demonstrate high between‐subject variation, the impact of which will depend on clinical context.
Reference change values can potentially be useful in monitoring early disease progression and the safety/efficacy of interventions on an individual level.
Serial sampling revealed that unexpectedly high values in heathy individuals can be observed, which urges caution when interpreting AD plasma biomarkers based on a single test result.
Keywords: amyloid, analytical variation, biological variation, glial fibrillary acidic protein, neurofilament light, plasma biomarkers, phosphorylated tau, reference change values
1. BACKGROUND
Novel technologies to measure brain pathophysiological processes in the blood have revolutionized the Alzheimer's disease (AD) research landscape. 1 , 2 Established and highly accurate methods for tracking such processes face barriers to their large‐scale implementation, such as the high costs, radiation exposure, and limited availability of positron emission tomography (PET) scans, as well as the relative invasiveness of lumbar punctures, required for measuring AD biomarkers in the cerebrospinal fluid (CSF). 3 Blood‐based AD biomarkers have demonstrated great promise so far, and are particularly promising for scalable implementation due to their minimally invasive and cost‐effective nature. 1 , 2
Among blood‐based biomarkers so far investigated, plasma phosphorylated tau (p‐tau) variants, such as p‐tau181, p‐tau231, and p‐tau217, have demonstrated the greatest potential to identify AD‐specific processes, showing high accuracy for identifying neuropathological or biomarker‐confirmed AD and predicting cognitive decline. 4 , 5 , 6 , 7 , 8 While p‐tau231 may be more sensitive to incipient amyloid beta (Aβ) pathology, plasma p‐tau217 seems the most well suited for clinical implementation, presenting the highest fold increases in cognitively impaired patients with AD‐type pathology, and it can dynamically track longitudinal AD clinical progression. 4 , 6 , 7 , 9 , 10 , 11 Plasma Aβ, in the form of the Aβ42/Aβ40 ratio, has also shown good performance in detecting Aβ pathology, but its modest fold change (reduced by 8% to 14% in AD compared to Aβ‐negative controls, when in the CSF it is reduced by > 50%) 12 , 13 makes it more vulnerable to analytical fluctuations normally observed in a day‐to‐day clinical chemistry routine. 14 , 15 , 16 Plasma levels of glial fibrillary acidic protein (GFAP), a cytoskeletal protein highly expressed in reactive astrocytes, 17 have been positively associated with early Aβ pathology. 18 , 19 , 20 , 21 Neurofilament light chain (NfL), a marker for axonal damage, has gained increasingly clinical significance with robust evidence for its diagnostic and prognostic utility in a wide range of neurodegenerative diseases (AD, frontotemporal dementia, atypical parkinsonian disorders) and in acute neurological conditions, such as stroke and traumatic brain injury. 22 , 23 , 24 , 25 , 26 Furthermore, all of these biomarker candidates have been evaluated as potential surrogate endpoints in disease‐modifying clinical trials in AD, with a recent example being reductions in plasma p‐tau217 as early as after 12 weeks of treatment with a promising anti‐Aβ monoclonal antibody, donanemab. 27
RESEARCH IN CONTEXT
Systematic review: We reviewed PubMed for articles and conference abstracts that evaluated the biological variation (BV) of novel Alzheimer's disease (AD) blood biomarkers. Two previous studies had reported BV estimates for serum glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL). Thus, we aimed to provide the first robust BV estimates for plasma amyloid beta (Aβ) and phosphorylated tau (p‐tau) biomarkers, as well as for plasma GFAP and NfL in in the same population.
Interpretation: Plasma biomarkers of key pathological features of AD demonstrate heterogeneity in their within‐ and between‐subject variation. Plasma Aβ42/Aβ40 generally shows lower variability but also changes very modestly in AD patients versus controls. While plasma p‐tau variants demonstrate higher variability, the clinical impact is likely limited due to large fold increases in AD. Plasma NfL and GFAP had the largest between‐subject variability, which may impact their application in certain contexts. Most research on blood biomarkers so far has been done using either single measurements or repeated measurements over longer (e.g., yearly) time frames; the weekly serial sampling in our study revealed that unexpected outlier values may occur, urging caution in clinical and research interpretation.
Future directions: Future studies should evaluate the potential clinical impact of the application of BV knowledge upon clinical and research interpretation of AD plasma biomarkers, especially in disease monitoring and in the evaluation of safety and efficacy of novel therapeutic interventions.
Nevertheless, several research questions must be addressed before large‐scale implementation of blood‐based AD biomarkers. 28 While most studies have focused on their diagnostic and prognostic properties, little is known about their biological variation (BV), a foundational concept in clinical chemistry, crucial to ensure the safe implementation of diagnostic markers and to minimize misclassification risks in laboratory medicine. 29 BV refers to the variation observed in clinical laboratory measurements determined by patients’ physiology, and a strict guideline‐defined methodology must be followed by BV studies to ensure robust results. 30 , 31 Such studies require the serial, tightly controlled collection of samples from healthy individuals with a regular sampling rate, and that analytes should be quantified, at least, in duplicate. 30 , 31 The key BV components are the within‐subject biological variation (CVI), which informs how much the concentration of a biomarker fluctuates around each individual's homeostatic setpoint, and the between‐subject biological variation (CVG), which informs on the variability between the homeostatic setpoints between different individuals. These parameters, alongside known assay‐dependent analytical variation (CVA), can provide highly clinically useful information for biomarker implementation. These include the reference change value (RCV), 32 , 33 which enumerates the change needed between consecutive measurements to exceed biological and analytical variation; the analytical performance specifications (APS) that clinical‐grade assays should meet; 34 , 35 and the index of individuality (II), which evaluates the utility of population based reference intervals. 33 , 36 Thus, high‐quality BV data are needed in this rapidly developing area of AD diagnostics, in which specific biomarkers and assays are being considered for clinical implementation and therapeutic trial use.
Here, we aimed to determine BV estimates for plasma Aβ42, Aβ40, Aβ42/Aβ40, p‐tau181, p‐tau217, p‐tau231, GFAP, and NfL (and associated APS and RCVs) in healthy adults between 40 and 60 years from the European Biological Variation Study (EuBIVAS), led by the European Federation of Clinical Chemistry and Laboratory Medicine (EFLM) Working Group on Biological Variation. 37 , 38 The EuBIVAS is a highly powered multi‐center study that included weekly blood sampling over 10 weeks from presumably healthy volunteers from five European countries and that has delivered high‐quality BV estimates for many measurands widely used in diverse medical areas. 38 , 39 , 40 , 41 , 42
2. METHODS
2.1. Study participants and sample collection
In this study, we quantified biomarkers in plasma‐citrate samples from a subset of 20 individuals aged between 40 and 60 years within the EuBIVAS, 37 , 38 which originally enrolled 91 healthy volunteers (53 females, 38 males; ages 21 to 69 years), from six European laboratories located in five different countries (Italy, Norway, Spain, Turkey and the Netherlands). We chose to include in the current study those in the older EuBIVAS age range that had sufficient sample material for analyses. Information on the participants’ health status and lifestyle was collected with an enrollment questionnaire, and participants were screened at enrollment with a selection of laboratory tests to further confirm compatibility with inclusion criteria. Fasting blood samples were collected weekly over 10 consecutive weeks for each study participant (April–June 2015), always in the morning. At each center, samples were centrifuged at 3000 × g for 10 minutes at room temperature within 1 hour of the blood draw, aliquoted, and frozen rapidly by immersion in a bowl with methanol and dry ice, and sent to the coordinating center (San Raffaele Hospital in Milan, Italy), where they were stored at −80°C. In November 2021, the samples included in this study were sent to the Clinical Neurochemistry Laboratory (Sahlgrenska University Hospital, Gothenburg, Sweden), where the AD blood biomarkers were measured (April 2022, except p‐tau217, analyzed December 2022). Further details regarding the inclusion/exclusion criteria; health status; and sample collection, processing, and storage protocol used in EuBIVAS have been previously reported. 37 While there was a considerable gap between sample collection and biochemical analyses (≈ 7 years), the evaluated AD blood biomarkers are known to be stable under the storage conditions used for the samples tested in this project. 4 , 5 , 18 , 23
The protocol for EuBIVAS received approval from the institutional ethical review board of San Raffaele Hospital, in compliance with the World Medical Association Declaration of Helsinki, as well as the ethical board/regional ethics committee for each participating center.
2.2. Biomarker quantification
Biomarker quantification was conducted using single molecule array (Simoa) HD‐X Analyzers from Quanterix at the Clinical Neurochemistry Laboratory of Sahlgrenska University Hospital in Sweden. A commercially available assay (Quanterix Neurology‐4 Plex E) was used to simultaneously quantify for Aβ42, Aβ40 (and Aβ42/Aβ40, consequently), NfL, and GFAP. 27 P‐tau231 and p‐tau181 were analyzed using Simoa assays developed at the University of Gothenburg, which have been validated as described elsewhere. 5 , 6 To measure p‐tau217, a novel commercially available assay from ALZpath (ALZpathDX) was used. 43 All samples from the same participant were analyzed in the same analytical run, and each sample was quantified in duplicate. Internal quality controls (iQC) at three different concentrations, for each measurand, were analyzed in duplicate in the beginning and end of each run. Before analysis, blood samples were thawed, vortexed, and centrifuged at 4000 × g for 10 minutes as suggested in recent studies. 44 , 45
2.3. Statistical analysis
Our statistical analyses followed a series of well‐established and guideline‐defined steps for deriving BV data, as set out by the Biological Variation Data Critical Appraisal Checklist (BIVAC), a standard for the executing and reporting of BV studies. 31 Outlier detection procedures were performed on three levels, including analytical (between replicates), within‐subject (among 10 collections for CVI calculation), and between‐subject level (for CVG calculation). 46 , 47 , 48 , 49 , 50 For obtaining CVI and CVA estimates, we initially performed CV‐transformation of the data in which each person's data are “normalized” by dividing by that person's mean value, so as to later perform analysis of variance (ANOVA) on these CV‐transformed values. 51 After CV transformation, we performed outlier identification and removal on the analytical levels (between replicates) by assessing the homogeneity of CVA with the Bartlett test. In case of heterogeneity for the analytical component, we first excluded the replicate value of the measurement that most deviated from that participant's mean. If the heterogeneity persisted, we then also excluded the second measurement result of the time point showing abnormal analytical variation. After ensuring analytical homogeneity, we evaluated the presence of outliers on the within‐individual variation level by assessing the homogeneity of the within‐individual CVI with the Cochran test. Then, we evaluated for each biomarker whether the results were consistent with steady state (i.e., no trend for increase or decrease during study) by fitting a linear regression model with the mean blood drawing value (pooled mean of the duplicate concentration measurements of each participant) as the dependent variable, with blood drawing number (from 1 to 10) as the independent variable. Individuals were considered in a steady state if the 95% confidence interval (CI) of the blood draw term (i.e., the slope) included 0. 52 Finally, the CVI was estimated with CV‐ANOVA, the “Røraas method,” a validated and recommended ANOVA method for estimating CVI and CVA. 51 , 53 To calculate the between‐subject biological variation (CVG), we first applied the Dixon Q test to detect outliers in mean biomarker concentrations between subjects, and the Shapiro–Wilk test to verify the normality assumption on mean concentrations. If the latter tests detected a non‐normal distribution, concentration data were natural log‐transformed, prior to obtaining the CVG by ANOVA. 46 , 47 , 48 , 49 , 50 First, CVI and CVG estimates were calculated for the whole study population, and also secondarily separately for males and females, for all measurands. Confidence intervals for BV estimates were calculated as previously described, 54 and the lack of overlap of the 95% CI of estimates was used to indicate significant differences between subgroups.
Other relevant metrics were computed based on the above‐mentioned BV estimates calculated as follows. Desirable APS were calculated for imprecision (CVAPS = 0.5xCVI) and for bias (BiasAPS = 0.25x√[CVI 2 + CVG 2]). The RCV was calculated at a 95% bidirectional alpha (z = 1.65) as RCV = 100*(exp[± zx√2xσ]–1), where σ = √ln(σ2 CVi + σ2 CVi), with σ2 CVi = ln(CVi 2 + 1) and σ2 CVa = ln(CVa 2 + 1). The II was calculated as the ratio of CVI and CVG for each biomarker, indicating whether population‐based reference intervals can be useful for evaluating results. 33 , 36 We also calculated the number of samples needed to be collected to estimate an individual's homeostatic point (NHSP) with a “D” absolute percentage proximity to the individual's true value with the equation n = (zx√[CVI 2 + CVA 2]/D) 2 , in which z = 1.96, corresponding to a 95% alpha. NHSP was calculated based on 5%, 10%, and 20% deviations from the homeostatic setpoint. Metrics such as RCVs and APS were always derived based on CVI and CVG of all participants. All analyses were performed with R Statistical Software (version 4.2.1; www.r‐project.com), and statistical significance was set as alpha = 0.05.
3. RESULTS
3.1. Participant characteristics
We included data analyzed from a total of 196 plasma samples, collected weekly over 10 weeks from 20 participants, with a mean number of 9.8 samples per participant. Key demographic information is described in Table 1. The age range of the included participants was 40 to 60 years, with a mean (standard deviation [SD]) age of 46.4 years (6.20) for the whole study population. Half of the participants were female, and key demographic characteristics were generally similar between sexes. The study population came from five centers in four European countries (Italy [n = 7], Netherlands [n = 5], Norway [n = 5], Spain [n = 3]), and all participants were White. Participants were healthy, with a mean (SD) body mass index of 23.3 kg/m2 (2.85 kg/m2), did not have hypertension, and the majority (55%) engaged in physical activity for more than 3 hours per week. Only one participant was a smoker (5%), and 11 reported consuming 1 to 2 units of alcohol per week.
TABLE 1.
Demographic characteristics.
|
Female (n = 10) |
Male (N = 10) |
Overall (N = 20) |
|
|---|---|---|---|
| Age, years, mean (SD) | 47.0 (5.98) | 45.8 (6.68) | 46.4 (6.20) |
| BMI, kg/m2, mean (SD) | 22.3 (2.50) | 24.2 (2.98) | 23.3 (2.85) |
| Hypertension, n (%) | 0 | 0 | 0 |
| Alcohol consumption, units/week, n (%) | |||
| 0 | 3 (30.0) | 3 (30.0) | 6 (30.0) |
| 1–2 | 6 (60.0) | 5 (50.0) | 11 (55.0) |
| ≥ 3 | 1 (10.0) | 2 (20.0) | 4 (15.0) |
| Smokers, n (%) | 1 (10) | 0 | 1 (5) |
| Physical exercise, n (%) | 8 (80) | 5 (50) | 13 (65) |
| No physical exercise | 2 (20) | 5 (50) | 7 (35) |
| < 3 hours per week | 1 (10) | 1 (10) | 2 (10) |
| ≥ 3 hours per week | 7 (70) | 4 (40) | 11 (55) |
| Study center, n (%) | |||
| Italy (Milan) | 2 (20) | 2 (20) | 4 (20) |
| Italy (Padua) | 3 (30) | 0 | 3 (15) |
| Netherlands | 3 (30) | 2 (20) | 5 (25) |
| Norway | 2 (20) | 3 (30) | 5 (25) |
| Spain | 0 | 3 (30) | 3 (15) |
Note: The table summarizes key demographic information for the included participants. Data are described as mean (SD) or n (%).
Abbreviations: BMI, body mass index; SD, standard deviation.
TABLE 2.
Homogeneity analyses and number of results included for calculation of biological variation estimates.
| Number of excluded results/subjects | ||||||||||
|---|---|---|---|---|---|---|---|---|---|---|
| Homogeneity (Bartlett and Cochran tests) |
Dixon Q test |
|||||||||
| Biomarker | Subjects, n | Total measurements, n |
Mean number of samples/ individual |
Mean number of replicates/ sample |
Replicates (analytical homogeneity) |
Samples (within homogeneity) |
Subjects (within homogeneity) |
Subjects (between) |
Data used to estimate CVI, n | Total % of outliers |
| Aβ40 | 20 | 385 | 9.80 | 1.96 | 0 | 8 | 0 | 0 | 377 | 2.08% |
| Aβ42 | 20 | 385 | 9.80 | 1.96 | 2 | 8 | 0 | 0 | 375 | 2.60% |
| Aβ42/Aβ40 | 20 | 385 | 9.80 | 1.96 | 0 | 4 | 0 | 2 | 381 | 1.04% |
| GFAP | 20 | 385 | 9.80 | 1.96 | 0 | 7 | 0 | 0 | 378 | 1.82% |
| NfL | 20 | 385 | 9.80 | 1.96 | 0 | 8 | 0 | 1 | 377 | 2.08% |
| P‐tau181 | 20 | 392 | 9.80 | 2.00 | 4 | 18 | 0 | 0 | 370 | 5.61% |
| P‐tau217 | 20 | 355 | 9.45 | 1.88 | 9 | 14 | 0 | 0 | 332 | 6.48% |
| P‐tau231 | 20 | 384 | 9.60 | 2.00 | 4 | 22 | 0 | 0 | 358 | 6.77% |
Note: The table displays the overall number of samples included and biomarker results produced, as well as the results of the homogeneity analyses carried to detect the presence of outliers on the replicate, sample, and subject levels.
Abbreviations: Aβ, amyloid beta; CVI, within‐individual biological variation; GFAP, glial fibrillary acidic protein; NfL, neurofilament light; p‐tau, phosphorylated tau.
3.2. Homogeneity analyses and outliers
Table 2 displays results of the homogeneity analyses for outlier detection and the final number of results included for each of the biomarkers. All samples were always analyzed in duplicate, except in very few cases with insufficient volume left, resulting in a mean of 1.97 replicate quantification per sample per biomarker. When evaluating the analytical homogeneity with the Bartlett test, no outliers for the replicate measurement were identified for Aβ40, Aβ42/Aβ40, GFAP, and NfL, while a few replicates were excluded for Aβ42, p‐tau181, p‐tau217, and p‐tau231. When assessing the variance homogeneity for within‐subject variation, outlier time points were identified for all biomarkers, but no subject had to be fully excluded. For the total study population, the mean percentage of results identified as outliers at the homogeneity analyses was 3.56% (range, 1.0% to 6.8%), which left a mean of 369 results (range, 332 to 381) used per biomarker to estimate the CVI.
In Figure 1 the 10‐week biological variation, in concentrations, of each plasma biomarker, stratified by sex and ordered by increasing age, is displayed. In a separate outlier detection procedure before the CVG estimation, the Dixon Q test identified one outlier subject for NfL, and two outlier subjects for Aβ42/Aβ40 (indicated in Figure 1). No trend was identified for any of the included biomarkers in the overall study population or in male or female subgroups. No biomarker measurement for any analyte was below the lower limit of detection or the lower limit of quantification.
FIGURE 1.

Participant‐level plasma biomarker concentrations over 10 weeks. The figure displays median (dots) and range (error bars) of biomarker concentrations over 10 weeks. Females are represented in dark green, and males in orange, and participants are shown with increasing age (subject 1 is the youngest female participant, and subject 20 the oldest male). Triangles represent the concentration outliers detected with the Dixon Q test before the CVG calculation. Aβ, amyloid beta; CVG, between‐subject biological variation; GFAP, glial fibrillary acidic protein; NfL, neurofilament light; p‐tau, phosphorylated tau.
3.3. Analytical performance (CVA)
The CVA for each biomarker, which indicates the imprecision between duplicate measurements, and associated 95% CIs, are graphically displayed in Figure 2A and numerically represented in Table 3. The CVA ranged from ≈ 3% for all Aβ biomarkers (Aβ42: 2.8%; Aβ40: 2.6%; Aβ42/Aβ40: 3.0%), to ≈ 6% for GFAP (6.4%) and NfL (6.3%), and to ≈ 5.5% for all p‐tau biomarkers (p‐tau181: 5.6%; p‐tau217: 5.7%; p‐tau231: 5.6%). Analytical variability of internal quality controls presented similar CVs to those estimated with CV‐ANOVA, and no systematic trends in concentration change between runs were observed by visual inspection.
FIGURE 2.

Biological variation estimates in the whole study population. The forest plot graphically summarizes the biological variation estimates obtained in this study, with dots corresponding to point estimates and error bars to 95% CIs. A, Analytical variation (CVA), (B) estimates for within‐individual biological variation, (C) between‐individual biological variation. Aβ, amyloid beta; CI, confidence interval; CVA, analytical variation; CVG, between‐subject biological variation; CVI, within‐subject biological variation; GFAP, glial fibrillary acidic protein; NfL, neurofilament light; p‐tau, phosphorylated tau.
TABLE 3.
Biological variation estimates for the whole study population and according to sex.
| Biomarker | Study population | Mean concentration, (pg/mL*, 95% CI) | CVA (%, 95% CI) | CVI (%, 95% CI) | CVG, (%, 95% CI) |
|---|---|---|---|---|---|
| Aβ42 | All participants | 6.24 (6.13–6.35) | 2.84 (2.57–3.15) | 6.50 (5.82–7.27) | 15.3 (11.1–21.1) |
| Male | 6.21 (6.06–6.36) | 6.26 (5.34–7.35) | 15.6 (9.56–25.4) | ||
| Female | 6.26 (6.11–6.42) | 6.76 (5.79–7.89) | 15.8 (9.97–25.1) | ||
| Aβ40 | All participants | 88.0 (86.8–89.3) | 2.63 (2.38–2.91) | 6.39 (5.73–7.13) | 12.5 (9.03–17.4) |
| Male | 88.9 (87.2–90.6) | 6.26 (5.36–7.31) | 11.6 (7.18‐18.7) | ||
| Female | 87.2 (85.3–89.2) | 6.76 (5.80–7.88) | 14.1 (8.76–22.6) | ||
| Aβ42/Aβ40 | All participants | 0.0714 (0.0704–0.0724) | 2.95 (2.67–3.26) | 3.33 (2.88–3.85) | 6.58 (4.64–9.34) |
| Male | 0.0707 (0.0689–0.0726) | 3.54 (2.90–4.32) | 6.22 (3.59–10.8) | ||
| Female | 0.0720 (0.0712–0.0729) | 3.13 (2.53–3.88) | 7.20 (4.47–11.6) | ||
| GFAP | All participants | 59.9 (58–61.7) | 6.40 (5.78–7.08) | 8.01 (6.99–9.18) | 30.1 (21.8–41.6) |
| Male | 54.9 (52.6–57.2) | 8.12 (6.74–9.77) | 28.5 (17.8–45.5) | ||
| Female | 64.9 (62.2–67.7) | 7.95 (6.5–9.72) | 29.7 (18.6–47.5) | ||
| NfL | All participants | 10 (9.7–10.3) | 6.32 (5.71–7.00) | 7.39 (6.4–8.52) | 21.2 (15.1–29.8) |
| Male | 8.8 (8.6–9.1) | 7.16 (5.89–8.71) | 15.9 (9.8–25.8) | ||
| Female | 11.2 (10.6–11.8) | 7.67 (6.23–9.44) | 24.9 (15.0–42.1) | ||
| P‐tau181 | All participants | 4.56 (4.41–4.70) | 5.62 (5.07–6.23) | 16.7 (15.0–18.6) | 25.7 (18.3–36.1) |
| Male | 4.97 (4.77–5.18) | 13.3 (11.4–15.5) | 24.8 (15.3–40.1) | ||
| Female | 4.13 (3.95–4.31) | 19.7 (16.9–22.9) | 23.7 (14.0–40.4) | ||
| P‐tau217 | All participants | 0.232 (0.226–0.238) | 5.67 (5.07–6.35) | 10.3 (9.15–11.7) | 21.1 (15.1–29.3) |
| Male | 0.236 (0.226–0.245) | 10.5 (8.92–12.4) | 24.8 (15.4–39.9) | ||
| Female | 0.229 (0.222–0.236) | 10.3 (8.58–12.3) | 17.9 (11.0–29.0) | ||
| P‐tau231 | All participants | 6.06 (5.94–6.18) | 5.55 (5.0–6.15) | 6.33 (5.46–7.35) | 17.2 (12.5–23.6) |
| Male | 6.31 (6.12–6.5) | 5.49 (4.32–6.98) | 19.7 (12.3–31.4) | ||
| Female | 5.82 (5.68–5.95) | 7.1 (5.86–8.61) | 13.9 (8.63–22.4) |
Note: The table displays the biological variation estimates for each biomarker and mean concentrations for the whole participant population in sex‐stratified sub‐groups and their 95% CIs.
Abbreviations: Aβ, amyloid beta; CI, confidence interval; CVA, analytical variation; CVG, between‐individual biological variation; CVI, within‐individual biological variation; GFAP, glial fibrillary acidic protein; NfL, neurofilament light; p‐tau, phosphorylated tau.
3.4. Within‐subject biological variation (CVI)
Figure 2B graphically represents the CVI values and their associated 95% CIs, that is, how much biomarker concentrations fluctuate around each individual's homeostatic setpoint. Plasma Aβ42 and Aβ40 demonstrated low and very similar CVI’=s (Aβ42: 6.5%, 95% CI: 5.8–7.3; Aβ40: 6.4%, 95% CI: 5.7–7.1), and the plasma Aβ42/Aβ40 ratio demonstrated the lowest CVI among all evaluated biomarkers (3.3%, 95% CI: 2.9–3.9). Among plasma p‐tau variants, p‐tau231 demonstrated the lowest CVI (6.3%, 95% CI: 5.5–7.4), followed by p‐tau217 (10.3%, 95% CI: 9.2–11.7), and by p‐tau181 with a considerably higher CVI (16.7%, 95% CI: 15.0–18.6). Plasma GFAP also demonstrated a relatively low CVI (8.0%, 95% CI: 7.0–9.2), comparable to that observed for NfL (7.4%, 95% CI: 6.4–8.5). In Table 3, the CVIs are also shown separately for males and females, important and needed subgroup analyses in BV studies. Except for p‐tau181, no differences in CVI were observed for the evaluated biomarkers, with overlapping 95% CIs for male and female CVIs. For plasma p‐tau181, females (19.7%, 95% CI: 16.9–22.9) demonstrated a higher CVI than males (13.3%, 95% CI: 11.4–15.5).
3.5. Between‐subject biological variation (CVG)
Figure 2C graphically represents the CVG values and their associated 95% CIs, that is, how much biomarker levels vary among healthy individuals. Among Aβ biomarkers, plasma Aβ42/Aβ40 demonstrated the lowest CVG (6.6%, 95% CI: 4.6–9.3), with higher and similar estimates for Aβ42 (15.3%, 95% CI: 11.1–21.1) and Aβ40 (12.5%, 95% CI: 9.0–17.4). For the other biomarkers, CVGs were generally higher than those for Aβ biomarkers. GFAP demonstrated the highest CVG among all biomarkers (30.1%, 95% CI: 21.8–41.6), and slightly higher than that of NfL (21.2%, 95% CI: 15.1–29.8). Among p‐tau biomarkers, p‐tau231 (17.2%, 95% CI: 17.2–19.7) demonstrated the lowest CVG, followed by p‐tau217 (21.1%, 95% CI: 15.1–29.3) and p‐tau181 (25.7%, 95% CI: 18.3–36.1%). Table 3 indicates the CVGs separately for males and females. No differences in CVG estimates were found for the evaluated biomarkers, with overlapping 95% CIs between males and females for all measurands. Table 3 also shows the mean concentrations and their 95% CIs for males and females separately, with slightly higher concentrations in females observed for plasma GFAP and NfL, and slightly higher concentrations in males for plasma p‐tau181 and p‐tau231.
3.6. Analytical performance specifications and other metrics
Table 4 shows APSs based on the desirable criteria (intermediate stringency), for imprecision, bias. In terms of desirable assay imprecision, the highest demand was for plasma Aβ42/Aβ40 (CVAPS < 1.7%), with the lowest demands for plasma p‐tau217 (CVAPS < 5.2%) and p‐tau181 (CVAPS < 8.4%). Table 4 shows the estimated RCVs, as well as the number of samples needed to estimate the homeostatic point. Plasma Aβ42/Aβ40 demonstrated the lowest RCVs needed for a significant decrease (11%) and for an increase (13%). Similar RCVs for both decrease (−20% to –28%) and increase (26% to 38%) were observed for GFAP, NfL, p‐tau217, and p‐tau231, with the highest RCVs for p‐tau181 (decrease: −38.3%; increase: 62.2%).
TABLE 4.
Metrics derived from BV estimates.
| Analytical performance specifications | Reference change value, % | Serial samples needed to estimate each individual's homeostatic setpoint within varying proximities to the true value | ||||||
|---|---|---|---|---|---|---|---|---|
| Imprecision, % CVAPS | Bias, % BAPS | Decrease | Increase | Index of individuality | ± 5% | ± 10% | ± 20% | |
| Aβ42 | 3.25 | 8.31 | –17.76 | 21.60 | 0.46 | 8 | 2 | 1 |
| Aβ40 | 3.25 | 7.06 | –17.57 | 21.31 | 0.56 | 7 | 2 | 1 |
| Aβ42/Aβ40 | 1.66 | 3.69 | –11.54 | 13.04 | 0.68 | 3 | 1 | 1 |
| GFAP | 4.00 | 15.59 | –24.60 | 32.62 | 0.34 | 16 | 4 | 1 |
| NfL | 3.69 | 11.21 | –23.49 | 30.71 | 0.43 | 15 | 4 | 1 |
| P‐tau181 | 8.36 | 15.34 | –38.34 | 62.18 | 0.69 | 48 | 12 | 3 |
| P‐tau217 | 5.17 | 11.73 | –27.71 | 38.33 | 0.56 | 21 | 5 | 1 |
| P‐tau231 | 3.17 | 9.17 | –20.70 | 26.10 | 0.49 | 11 | 3 | 1 |
Note: The table displays each biomarker's desirable analytical performance specifications (APS) for replicate precision and bias. Also, the table displays the reference change values (RCV), which indicate the percentage change needed between two consecutive measurements so that such an increase or decrease significantly overcomes analytical and biological variation. The index of individuality and number of samples needed to estimate the homeostatic point with a given proximity are also shown.
Abbreviations: Aβ, amyloid beta; BAPS, bias analytical performance specification; CVAPS, imprecision analytical performance specification; GFAP, glial fibrillary acidic protein; NfL, neurofilament light; p‐tau, phosphorylated tau.
4. DISCUSSION
We report BV estimates for AD plasma biomarkers generated based on a high number of weekly samples per individual, for a comprehensive biomarker panel measured within the same participants. These are the first reported BV estimates for blood Aβ42, Aβ40, Aβ42/Aβ40, and p‐tau, but also for plasma NfL and GFAP (previously evaluated in serum). 55 , 56 , 57 We found that within‐ and between‐subject biological variation can be considerably different for AD biomarker classes, which may impact biomarkers differently according to each application context.
Beyond improving the interpretation of laboratory tests, reliable BV data enable the determination of the APS needed for each biomarker. Assay imprecision (i.e., CVA) should be considerably lower than the biomarker's CVI, with the desirable analytical performance being that CVA ≤ CVI/2. 58 Here, CVAs were slightly higher than desired for most biomarkers. Plasma Aβ42 and Aβ40 (but not Aβ42/Aβ40) and p‐tau181 were within the desirable range, with p‐tau217 showing a very close to desirable analytical performance (CVA = 5.7%; CVAPS ≤ 5.2%). Nevertheless, it is important to emphasize that BV estimates are not standalone criteria to determine APS for assays, but rather a complementary tool to refine the determination of analytical goals given each analyte's clinical application context. For instance, the higher‐than‐desirable observed CVAs are likely not a cause of concern in light of the main clinical applications of NfL and p‐tau variants, whereas Aβ42/Aβ40 might be more affected, as discussed below.
Plasma p‐tau is an AD‐specific biomarker envisioned to be implemented as a screening tool to classify patients seeking medical advice for cognitive symptoms into high, intermediate, and low risk of having AD pathology. 59 , 60 In our study, CVA was remarkably similar for all three plasma p‐tau variants, but they demonstrated considerably different CVIs. Interestingly, p‐tau231 demonstrated the lowest CVI (6.3%, 95% CI: 5.5–7.4%), followed by p‐tau217 (10.3%, 95% CI: 9.2–11.7%) and p‐tau181 (16.7%, 95% CI: 12.5%–23.6%), and this could suggest possible differences in release, clearance, or transportation of plasma p‐tau species. Recent head‐to‐head comparisons of plasma p‐tau variants showed the most promising candidates were increased between 100% and 360% in the presence of AD pathology. 9 , 10 Considering the magnitude of these increases, currently available p‐tau assays demonstrate satisfactory analytical performance for clinical applications, making their diagnostic ability less vulnerable to biological and analytical variation.
Plasma NfL has been successfully introduced in some clinical routine laboratories, being useful in a range of neurodegenerative diseases and acute neurological conditions. 22 , 24 , 26 , 61 Plasma NfL showed a relatively low CVI (7.4%, 95% CI: 6.4–8.5%), and a higher CVG (21.2%, 95% CI: 15.1–29.8%). This is in accordance with the ≈ 10% CVI and CVGs reported in a previous BV study evaluating serum NfL in a Turkish cohort with 10 weekly collections. 57 Our RCVs for NfL (increase: +30.7; decrease: −23.4%) also closely agreed with those in that study (increase: +32.7; decrease: −24.7%). A study in a Danish cohort reported a lower CVI for serum NfL with non‐overlapping CIs (CVI = 3%, 95% CI: 1.2–5.0%), and lower RCVs (increase: +24.3; decrease: −19.5%). 56 These lower estimates could be attributed to the shorter sampling period (three consecutive days vs. 10 weeks), which may underestimate BV. 62 The relatively high CVG seen for plasma NfL (reflecting higher inter‐individual variability) may pose a challenge to its clinical interpretation in conditions with modest NfL fold changes such as AD, where the diagnostic utility of NfL remains limited. However, in certain clinical scenarios, the magnitude of NfL increases is much larger compared to this higher between‐subject variation, including differentiating primary psychiatric disorders from frontotemporal dementia, 63 or in prognostic evaluation of acute conditions such as cardiac arrest, stroke, and traumatic brain injury. 24 , 26 , 64
While it is not yet clear what GFAP in the blood reflects (showing unexpected differences against CSF GFAP), 65 it has been associated with Aβ pathology, showing promising diagnostic performance, 18 , 20 even though increased GFAP levels have been reported in Aβ‐negative neurodegenerative conditions. 66 We observed a relatively low CVI for plasma GFAP (8.0%, 95% CI: 7.0–9.2), and the highest CVG among all biomarkers (30.1%, 95% CI: 21.8–41.6%). A previous study with serum GFAP reported a similar CVI (9.7%, 95% CI: 7.6–11.8), and also a high CVG (39.5%, 95% CI: 31.7–47.3), with RCVs also agreeing between studies. The high between‐individual variability of plasma GFAP, alongside its poorly understood clinical meaning, may pose difficulties for its individual‐level interpretation.
Plasma Aβ42/Aβ40 is a widely investigated biomarker associated with Aβ pathology currently in clinical use to support an AD diagnosis. 12 , 67 , 68 However, in Aβ‐positive versus Aβ‐negative individuals, plasma Aβ42/Aβ40 is only decreased by 8% to 14%. 13 This clinical context places, per se, an issue for this biomarker, because the modest disease‐related fold changes are in a similar magnitude to that of common analytical variation figures seen in clinical chemistry. 2 , 14 , 15 , 16 Our BV findings further support that plasma Aβ42/Aβ40 will likely face long‐term difficulties if introduced in clinical practice. Plasma Aβ42/Aβ40 demonstrated a considerably low CVI (3.3%, 95% CI: 2.9–3.9), and a relatively low CVG (6.6%, 95% CI: 4.6–9.3). The low CVI introduces a very high demand on analytical performance, because, desirably, the CVA should be less than half of the CVI (CVAPS = 1.7%; Table 4), and, optimally, less or equal to a quarter of CVI (CVI/4 = 0.7%). In contrast, plasma p‐tau presents larger disease‐related increases, making the ≈ 5.5% CVAs acceptable. Additionally, while there are several different plasma Aβ assays currently in use, they present similarities in their biochemical target and in small disease changes. 13 Considering previous BV studies showing different assay versions for the same analyte present indistinguishable CVIs, it is expected that the CVI for plasma Aβ42/Aβ40 would be similar across assays. 47 , 52 Further, the desirable bias is also considerably low for plasma Aβ42/Aβ40 (BAPS = 3.7%; Table 4), and it is unlikely that batch‐to‐batch variations could be kept low enough to meet this APS.
We report RCVs for AD plasma biomarkers, which can be potentially clinically valuable when monitoring individuals over time by enumerating the change that can be explained by biological and analytical variation, becoming especially relevant with novel anti‐Aβ immunotherapies such as lecanemab and donanemab. 27 , 69 These drugs substantially reduced plasma p‐tau217 levels, on a group level, as early as at 12 weeks of treatment (with our RCVs derived in a similar time frame), during the initial phase of Aβ‐plaque reduction. 27 RCVs could be potentially used to identify whether a reduction in plasma p‐tau217 after treatment initiation could indeed be related to a positive treatment response. On the other hand, this class of drugs can cause amyloid‐related imaging abnormalities (ARIA) of the hemorrhage or edema type, which can be very harmful. 70 If NfL proves capable of tracking such changes, RCVs could also be potentially useful to monitor ARIA emergence. However, it is important to consider that our RCVs were obtained using the presumably healthy cognitively unimpaired sample and assays herein described, and, for this reason, cannot be considered universal values, and each laboratory has to determine their own RCVs based on their CVA estimates.
Our study provides a unique opportunity to evaluate shorter term fluctuations of AD blood biomarkers. Most published studies have collected samples either cross‐sectionally or over longer periods of time (e.g., 6 months, yearly), and little is known about their shorter term variability. Figure 3 shows plasma p‐tau181 and p‐tau217 for two male subjects with similar ages. Subject “A” shows minimal fluctuation around the homeostatic point, while subject “B” experiences two spikes in plasma p‐tau. These fluctuations were not analytical outliers, and because a chronic disease like AD is unlikely to manifest an oscillatory progression from week to week in middle‐aged adults, there might be yet uncharacterized factors influencing biomarker readings. Clinical decisions made on a single sample collected on a p‐tau “spike” day could erroneously classify patients as “abnormal,” as exemplified by two previously described cut‐offs. 43 , 71 Such high‐value outliers are not uncommon in Aβ‐negative groups when examining data points from recent cross‐sectional studies, 4 , 5 , 6 and we recommend caution for researchers and clinicians when interpreting AD blood biomarker results from a single sample. While this has also happened to the other biomarkers in our study, we chose to highlight this phenomenon for p‐tau to demonstrate that even the most promising biomarker classes may be subject to unexpected variations that need to be characterized.
FIGURE 3.

Example of 10‐week variability in plasma p‐tau181 and p‐tau217 in two study subjects. The figure shows the variability in plasma p‐tau181 and p‐tau217 levels over 10 weeks in two study subjects. Dots correspond to the mean concentration of the two duplicate measurements, and all of these subjects’ measurements demonstrated acceptable agreement between replicates, indicating that any deviation observed does not come from analytical imprecision. Dashed lines represent previously published cut‐offs for plasma p‐tau181 and p‐tau217 as illustrative examples of the potential impacts of these fluctuations over decision making. Of note, the outlier data points were excluded from CVI calculations in the homogeneity analyses as they do not reflect the expected homeostatic fluctuation. It is likely that these outlier data points instead correspond to a yet unknown factor that affects biomarker readings. CVI, within‐individual biological variation; p‐tau, phosphorylated tau.
Additionally, knowledge of CVI enables calculating the number of samples needed to estimate the individual's homeostatic point (HSP) within a certain proximity of the true value. To estimate the true HSPs of all analytes with a deviation of ± 20% (α < 0.05), a reasonable margin for most analytes here (given clinical contexts discussed above), one sample suffices for all biomarkers but p‐tau181 (NHSP = 3). Reducing this deviation to 5% (likely needed for plasma Aβ42/Aβ40 but not for others), three samples would be required (Table 4). The II evaluates the utility of reference intervals (RIs). For analytes with pronounced individuality and a relatively low CVI compared to CVG (II < 0.6), RCVs are more useful than RIs for accurate interpretation of sequential results, with each individual serving as the optimal reference point for assessing serial results. However, RIs remain suitable for analytes with high II (particularly when II > 1.4). 33 , 36 Here, all II values were below 0.6, except for p‐tau181 and Aβ42/Aβ40, which had slightly higher values, indicating marked individuality for these analytes. 33 , 36 , 72
This study has limitations. Although well powered in individual‐level serial sampling, the number of participants was relatively small, possibly affecting CVG more than CVI estimates. We found some concentration differences between males and females, and an unexpected sex difference in CVI for p‐tau181, warranting further studies. The relatively younger population studied here may not capture biomarker fluctuations related to factors such as co‐morbidities and medication use. 73 Lack of confirmatory CSF or PET biomarkers prevented us from evaluating the effects of AD pathology over BV estimates. Further evaluation on ethnically diverse populations is also needed. The use of citrate plasma does not affect the obtained CV values, but biomarker concentrations (such as those in Figure 1) should be interpreted bearing in mind the matrix type and collection tube. For most of the evaluated AD biomarkers, citrate plasma has shown very similar concentrations compared to paired ethylenediaminetetraacetic acid, and slightly lower levels in citrate plasma for NfL and GFAP. 44 Our study has a number of strengths, involving quantifying a comprehensive panel of AD biomarkers in a dataset following all EFLM recommendations for BV studies, 30 , 31 which has generated reliable BV data for many other analytes. 37 , 38
CONFLICT OF INTEREST STATEMENT
K.B. has served as a consultant and on advisory boards for Acumen, ALZPath, BioArctic, Biogen, Eisai, Lilly, Moleac Pte. Ltd, Novartis, Ono Pharma, Prothena, Roche Diagnostics, and Siemens Healthineers; has served on data monitoring committees for Julius Clinical and Novartis; has given lectures, produced educational materials, and participated in educational programs for AC Immune, Biogen, Celdara Medical, Eisai, and Roche Diagnostics; and is a co‐founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program, outside the work presented in this paper. N.J.A. has given lectures for BioArtic, Eli Lily, and Quanterix. H.Z. has served on scientific advisory boards and/or as a consultant for Abbvie, Acumen, Alector, Alzinova, ALZPath, Annexon, Apellis, Artery Therapeutics, AZTherapies, CogRx, Denali, Eisai, Nervgen, Novo Nordisk, Optoceutics, Passage Bio, Pinteon Therapeutics, Prothena, Red Abbey Labs, reMYND, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave; has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, Biogen, and Roche; and is a co‐founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). A.J. is employed by ALZPath. All other authors declare no conflicts of interest. Author disclosures are available in the supporting information.
CONSENT STATEMENT
All volunteers in the EuBIVAS study provided informed consent. The EuBIVAS study received approval from the institutional ethical review board of San Raffaele Hospital, in compliance with the World Medical Association Declaration of Helsinki, as well as the ethical board/regional ethics committee for each participating center.
Supporting information
Supporting information.
ACKNOWLEDGMENTS
The authors thank all volunteers who participated in the EuBIVAS study and all the staff involved. HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2022‐01018 and #2019‐02397), the European Union's Horizon Europe research and innovation programme under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG‐71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809‐2016862), the AD Strategic Fund and the Alzheimer's Association (#ADSF‐21‐831376‐C, #ADSF‐21‐831381‐C, and #ADSF‐21‐831377‐C), the Bluefield Project, the Olav Thon Foundation, the Erling‐Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022‐0270), the European Union's Horizon 2020 research and innovation programme under the Marie Skłodowska‐Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021‐00694), the National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL (UKDRI‐1003). TKK was supported by the NIH (P30 AG066468, RF1 AG052525‐01A1, R01 AG053952‐05, R37 AG023651‐17, RF1 AG025516‐12A1, R01 AG073267‐02, R01 AG075336‐01, R01 AG072641‐02, P01 AG025204‐16), the Swedish Research Council (Vetenskåpradet; #2021‐03244), the Alzheimer's Association (#AARF‐21‐850325), the Swedish Alzheimer Foundation (Alzheimerfonden), the Aina (Ann) Wallströms and Mary‐Ann Sjöbloms stiftelsen, and the Emil och Wera Cornells stiftelsen. KB is supported by the Swedish Research Council (#2017‐00915 and #2022‐00732), the Swedish Alzheimer Foundation (#AF‐930351, #AF‐939721 and #AF‐968270), Hjärnfonden, Sweden (#FO2017‐0243 and #ALZ2022‐0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF‐agreement (#ALFGBG‐715986 and #ALFGBG‐965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019‐466‐236), the Alzheimer's Association 2021 Zenith Award (ZEN‐21‐848495), and the Alzheimer's Association 2022‐2025 Grant (SG‐23‐1038904 QC).
Brum WS, Ashton NJ, Simrén J, et al. Biological variation estimates of Alzheimer's disease plasma biomarkers in healthy individuals. Alzheimer's Dement. 2024;20:1284–1297. 10.1002/alz.13518
Anna Carobene and Kaj Blennow contributed equally as senior authors.
Contributor Information
Wagner S. Brum, Email: wagner.scheeren.brum@gu.se.
Anna Carobene, Email: carobene.anna@hsr.it.
Kaj Blennow, Email: kaj.blennow@neuro.gu.se.
REFERENCES
- 1. Hansson O. Biomarkers for neurodegenerative diseases. Nat Med. 2021;27(6):954‐963. doi: 10.1038/s41591-021-01382-x [DOI] [PubMed] [Google Scholar]
- 2. Karikari TK, Ashton NJ, Brinkmalm G, et al. Blood phospho‐tau in Alzheimer disease: analysis, interpretation, and clinical utility. Nat Rev Neurol. 2022;18(7):400‐418. doi: 10.1038/s41582-022-00665-2 [DOI] [PubMed] [Google Scholar]
- 3. Jack CR, Bennett DA, Blennow K, et al. NIA‐AA research framework: toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14(4):535‐562. doi: 10.1016/j.jalz.2018.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Palmqvist S, Janelidze S, Quiroz YT, et al. Discriminative accuracy of plasma phospho‐tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA, J Am Med Assoc. 2020;324(8):772‐781. doi: 10.1001/jama.2020.12134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Karikari TK, Pascoal TA, Ashton NJ, et al. Blood phosphorylated tau 181 as a biomarker for Alzheimer's disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol. 2020;19(5):422‐433. doi: 10.1016/S1474-4422(20)30071-5 [DOI] [PubMed] [Google Scholar]
- 6. Ashton NJ, Pascoal TA, Karikari TK, et al. Plasma p‐tau231: a new biomarker for incipient Alzheimer's disease pathology. Acta Neuropathol. 2021;141(5):709‐724. doi: 10.1007/s00401-021-02275-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Ashton NJ, Janelidze S, Mattsson‐Carlgren N, et al. Differential roles of Aβ42/40, p‐tau231 and p‐tau217 for Alzheimer's trial selection and disease monitoring. Nat Med. 2022:1‐8. doi: 10.1038/s41591-022-02074-w. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Montoliu‐Gaya L, Benedet AL, Tissot C, et al. Mass spectrometric simultaneous quantification of tau species in plasma shows differential associations with amyloid and tau pathologies. Nat Aging. 2023:1‐9. doi: 10.1038/s43587-023-00405-1. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Janelidze S, Bali D, Ashton NJ, et al. Head‐to‐head comparison of 10 plasma phospho‐tau assays in prodromal Alzheimer's disease. Brain. doi: 10.1093/brain/awac333. Published online September 10, 2022:awac333.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Ashton NJ, Puig‐Pijoan A, Milà‐Alomà M, et al. Plasma and CSF biomarkers in a memory clinic: head‐to‐head comparison of phosphorylated tau immunoassays. Alzheimer's & Dementia. doi: 10.1002/alz.12841. n/a(n/a). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Milà‐Alomà M, Ashton NJ, Shekari M, et al. Plasma p‐tau231 and p‐tau217 as state markers of amyloid‐β pathology in preclinical Alzheimer's disease. Nat Med. 2022;28(9):1797‐1801. doi: 10.1038/s41591-022-01925-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Schindler SE, Bollinger JG, Ovod V, et al. High‐precision plasma β‐amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019;93(17):e1647‐e1659. doi: 10.1212/WNL.0000000000008081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Janelidze S, Teunissen CE, Zetterberg H, et al. Head‐to‐head comparison of 8 plasma amyloid‐β 42/40 assays in Alzheimer disease. JAMA Neurol. 2021;78(11):1375‐1382. doi: 10.1001/jamaneurol.2021.3180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Benedet AL, Brum WS, Hansson O, et al. The accuracy and robustness of plasma biomarker models for amyloid PET positivity. Alzheimers Res Ther. 2022;14(1):26. doi: 10.1186/s13195-021-00942-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Rabe C, Bittner T, Jethwa A, et al. Clinical performance and robustness evaluation of plasma amyloid‐β42/40 prescreening. Alzheimers Dement. 2023;19(4):1393‐1402. doi: 10.1002/alz.12801 [DOI] [PubMed] [Google Scholar]
- 16. Cullen NC, Janelidze S, Mattsson‐Carlgren N, et al. Test‐retest variability of plasma biomarkers in Alzheimer's disease and its effects on clinical prediction models. Alzheimers Dement. 2022. doi: 10.1002/alz.12706. Published online June 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50(4):427‐434. doi: 10.1002/glia.20207 [DOI] [PubMed] [Google Scholar]
- 18. Benedet AL, Milà‐Alomà M, Vrillon A, et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 2021;78(12):1471‐1483. doi: 10.1001/jamaneurol.2021.3671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Pereira JB, Janelidze S, Smith R, et al. Plasma GFAP is an early marker of amyloid‐β but not tau pathology in Alzheimer's disease. Brain. 2021;144(11):3505‐3516. doi: 10.1093/brain/awab223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Shen XN, Huang SY, Cui M, et al. Plasma glial fibrillary acidic protein in the Alzheimer disease continuum: relationship to other biomarkers, differential diagnosis, and prediction of clinical progression. Clin Chem. doi: 10.1093/clinchem/hvad018. Published online March 2, 2023:hvad018.. [DOI] [PubMed] [Google Scholar]
- 21. Montoliu‐Gaya L, Alcolea D, Ashton NJ, et al. Plasma and cerebrospinal fluid glial fibrillary acidic protein levels in adults with down syndrome: a longitudinal cohort study. EBioMedicine. 2023;90:104547. doi: 10.1016/j.ebiom.2023.104547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Ashton NJ, Janelidze S, Al Khleifat A, et al. A multicentre validation study of the diagnostic value of plasma neurofilament light. Nat Commun. 2021;12(1):3400. doi: 10.1038/s41467-021-23620-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. 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). doi: 10.1001/jamaneurol.2019.0765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Gendron TF, Badi MK, Heckman MG, et al. Plasma neurofilament light predicts mortality in patients with stroke. Sci Transl Med. 2020;12(569):eaay1913. doi: 10.1126/scitranslmed.aay1913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Michaëlsson I, Hallén T, Carstam L, et al. Circulating brain injury biomarkers: a novel method for quantification of the impact on the brain after tumor surgery. Neurosurgery. 2023. doi: 10.1227/neu.0000000000002510. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Ashton NJ, Moseby‐Knappe M, Benedet AL, et al. Alzheimer disease blood biomarkers in patients with out‐of‐hospital cardiac arrest. JAMA Neurol. 2023. doi: 10.1001/jamaneurol.2023.0050. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Pontecorvo MJ, Lu M, Burnham SC, et al. Association of donanemab treatment with exploratory plasma biomarkers in early symptomatic Alzheimer disease: a secondary analysis of the TRAILBLAZER‐ALZ randomized clinical trial. JAMA Neurol. 2022. doi: 10.1001/jamaneurol.2022.3392. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Hansson O, Edelmayer RM, Boxer AL, et al. The Alzheimer's association appropriate use recommendations for blood biomarkers in Alzheimer's disease. Alzheimers Dement. 2022;18(12):2669‐2686. doi: 10.1002/alz.12756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Fraser GG, Harris EK. Generation and application of data on biological variation in clinical chemistry. Crit Rev Clin Lab Sci. 1989;27(5):409‐437. doi: 10.3109/10408368909106595 [DOI] [PubMed] [Google Scholar]
- 30. Bartlett WA, Braga F, Carobene A, et al. A checklist for critical appraisal of studies of biological variation. Clin Chem Lab Med. 2015;53(6):879‐885. doi: 10.1515/cclm-2014-1127 [DOI] [PubMed] [Google Scholar]
- 31. Aarsand AK, Røraas T, Fernandez‐Calle P, et al. The biological variation data critical appraisal checklist: a standard for evaluating studies on biological variation. Clin Chem. 2018;64(3):501‐514. doi: 10.1373/clinchem.2017.281808 [DOI] [PubMed] [Google Scholar]
- 32. Fraser CG. Reference change values: the way forward in monitoring. Ann Clin Biochem. 2009;46(Pt 3):264‐265. doi: 10.1258/acb.2009.009006 [DOI] [PubMed] [Google Scholar]
- 33. Coşkun A, Sandberg S, Unsal I, et al. Personalized reference intervals in laboratory medicine: a new model based on within‐subject biological variation. Clin Chem. 2021;67(2):374‐384. doi: 10.1093/clinchem/hvaa233 [DOI] [PubMed] [Google Scholar]
- 34. Fraser CG, Petersen PH. Analytical performance characteristics should be judged against objective quality specifications. Clin Chem. 1999;45(3):321‐323. [PubMed] [Google Scholar]
- 35. Haeckel R, Wosniok W, Kratochvila J, Carobene A. A pragmatic proposal for permissible limits in external quality assessment schemes with a compromise between biological variation and the state of the art. Clin Chem Lab Med. 2012;50(5):833‐839. doi: 10.1515/cclm-2011-0862 [DOI] [PubMed] [Google Scholar]
- 36. Carobene A, Banfi G, Locatelli M, Vidali M. Within‐person biological variation estimates from the European Biological Variation Study (EuBIVAS) for serum potassium and creatinine used to obtain personalized reference intervals. Clin Chim Acta. 2021;523:205‐207. doi: 10.1016/j.cca.2021.09.018 [DOI] [PubMed] [Google Scholar]
- 37. Carobene A, Strollo M, Jonker N, et al. Sample collections from healthy volunteers for biological variation estimates’ update: a new project undertaken by the working group on biological variation established by the European Federation of Clinical Chemistry and Laboratory Medicine. Clin Chem Lab Med. 2016;54(10):1599‐1608. doi: 10.1515/cclm-2016-0035 [DOI] [PubMed] [Google Scholar]
- 38. Carobene A, Aarsand AK, Bartlett WA, et al. The European Biological Variation Study (EuBIVAS): a summary report. Clin Chem Lab Med. 2022;60(4):505‐517. doi: 10.1515/cclm-2021-0370 [DOI] [PubMed] [Google Scholar]
- 39. Cavalier E, Lukas P, Bottani M, et al. European Biological Variation Study (EuBIVAS): within‐ and between‐subject biological variation estimates of β‐isomerized C‐terminal telopeptide of type I collagen (β‐CTX), N‐terminal propeptide of type I collagen (PINP), osteocalcin, intact fibroblast growth factor 23 and uncarboxylated‐unphosphorylated matrix‐Gla protein‐a cooperation between the EFLM Working Group on Biological Variation and the International Osteoporosis Foundation‐International Federation of Clinical Chemistry Committee on Bone Metabolism. Osteoporos Int. 2020;31(8):1461‐1470. doi: 10.1007/s00198-020-05362-8 [DOI] [PubMed] [Google Scholar]
- 40. Clouet‐Foraison N, Marcovina SM, Guerra E, et al. Analytical performance specifications for Lipoprotein(a), Apolipoprotein B‐100, and Apolipoprotein A‐I using the biological variation model in the EuBIVAS population. Clin Chem. 2020;66(5):727‐736. doi: 10.1093/clinchem/hvaa054 [DOI] [PubMed] [Google Scholar]
- 41. Aarsand AK, Kristoffersen AH, Sandberg S, et al. The European Biological Variation Study (EuBIVAS): biological variation data for coagulation markers estimated by a bayesian model. Clin Chem. 2021;67(9):1259‐1270. doi: 10.1093/clinchem/hvab100 [DOI] [PubMed] [Google Scholar]
- 42. Bottani M, Aarsand AK, Banfi G, et al. European Biological Variation Study (EuBIVAS): within‐ and between‐subject biological variation estimates for serum thyroid biomarkers based on weekly samplings from 91 healthy participants. Clin Chem Lab Med. 2022;60(4):523‐532. doi: 10.1515/cclm-2020-1885 [DOI] [PubMed] [Google Scholar]
- 43. Ashton NJ, Brum WS, di MolfettaG, et al. Diagnostic accuracy of the plasma ALZpath pTau217 immunoassay to identify Alzheimers disease pathology. medRxiv. 2023. doi: 10.1101/2023.07.11.23292493. Published online July 12. 2023.07.11.23292493. [DOI] [Google Scholar]
- 44. Ashton NJ, Suárez‐Calvet M, Karikari TK, et al. Effects of pre‐analytical procedures on blood biomarkers for Alzheimer's pathophysiology, glial activation, and neurodegeneration. Alzheimers Dement (Amst). 2021;13(1):e12168. doi: 10.1002/dad2.12168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Verberk IMW, Misdorp EO, Koelewijn J, et al. Characterization of pre‐analytical sample handling effects on a panel of Alzheimer's disease–related blood‐based biomarkers: results from the Standardization of Alzheimer's Blood Biomarkers (SABB) working group. Alzheimer's & Dementia. 2022;18(8):1484‐1497. doi: 10.1002/alz.12510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Carobene A, Røraas T, Sølvik UØ, et al. Biological Variation estimates obtained from 91 healthy study participants for 9 enzymes in serum. Clin Chem. 2017;63(6):1141‐1150. doi: 10.1373/clinchem.2016.269811 [DOI] [PubMed] [Google Scholar]
- 47. Carobene A, Marino I, Coşkun A, et al. The EuBIVAS project: within‐ and between‐subject biological variation data for serum creatinine using enzymatic and alkaline picrate methods and implications for monitoring. Clin Chem. 2017;63(9):1527‐1536. doi: 10.1373/clinchem.2017.275115 [DOI] [PubMed] [Google Scholar]
- 48. Aarsand AK, Díaz‐Garzón J, Fernandez‐Calle P, et al. The EuBIVAS: within‐ and between‐subject biological variation data for electrolytes, lipids, urea, uric acid, total protein, total bilirubin, direct bilirubin, and glucose. Clin Chem. 2018;64(9):1380‐1393. doi: 10.1373/clinchem.2018.288415 [DOI] [PubMed] [Google Scholar]
- 49. Carobene A, Guerra E, Locatelli M, et al. Providing correct estimates of biological variation‐not an easy task. the example of S100‐β protein and neuron‐specific enolase. Clin Chem. 2018;64(10):1537‐1539. doi: 10.1373/clinchem.2018.292169 [DOI] [PubMed] [Google Scholar]
- 50. Carobene A, Guerra E, Locatelli M, et al. Biological variation estimates for prostate specific antigen from the European Biological Variation Study; consequences for diagnosis and monitoring of prostate cancer. Clin Chim Acta. 2018;486:185‐191. doi: 10.1016/j.cca.2018.07.043 [DOI] [PubMed] [Google Scholar]
- 51. Røraas T, Støve B, Petersen PH, Sandberg S. Biological variation: the effect of different distributions on estimated within‐person variation and reference change values. Clin Chem. 2016;62(5):725‐736. doi: 10.1373/clinchem.2015.252296 [DOI] [PubMed] [Google Scholar]
- 52. Ceriotti F, Díaz‐Garzón Marco J, Fernández‐Calle P, et al. The European Biological Variation Study (EuBIVAS): weekly biological variation of cardiac troponin I estimated by the use of two different high‐sensitivity cardiac troponin I assays. Clin Chem Lab Med. 2020;58(10):1741‐1747. doi: 10.1515/cclm-2019-1182 [DOI] [PubMed] [Google Scholar]
- 53. Røraas T, Støve B, Petersen PH, Sandberg S. Biological variation: evaluation of methods for constructing confidence intervals for estimates of within‐person biological variation for different distributions of the within‐person effect. Clin Chim Acta. 2017;468:166‐173. doi: 10.1016/j.cca.2017.02.021 [DOI] [PubMed] [Google Scholar]
- 54. Graybill RKB, Franklin A. Confidence Intervals on Variance Components. CRC Press; 2014. doi: 10.1201/9781482277142 [DOI] [Google Scholar]
- 55. Christensen SH, Hviid CVB, Madsen AT, Parkner T, Winther‐Larsen A. Short‐term biological variation of serum glial fibrillary acidic protein. Clin Chem Lab Med. 2022;60(11):1813‐1819. doi: 10.1515/cclm-2022-0480 [DOI] [PubMed] [Google Scholar]
- 56. Hviid CVB, Madsen AT, Winther‐Larsen A. Biological variation of serum neurofilament light chain. Clin Chem Lab Med. 2022;60(4):569‐575. doi: 10.1515/cclm-2020-1276 [DOI] [PubMed] [Google Scholar]
- 57. Carobene A, Maiese K, Aboud‐Diwan C, et al. Biological variation estimates for serum neurofilament light chain in healthy subjects. Clin Chim Acta. 2023;551:117608. doi: 10.1016/j.cca.2023.117608 [DOI] [PubMed] [Google Scholar]
- 58. Sandberg S, Fraser CG, Horvath AR, et al. Defining analytical performance specifications: consensus statement from the 1st strategic conference of the european federation of clinical chemistry and laboratory medicine. Clin Chem Lab Med. 2015;53(6):833‐835. doi: 10.1515/cclm-2015-0067 [DOI] [PubMed] [Google Scholar]
- 59. Brum WS, Cullen NC, Janelidze S, et al. A two‐step workflow based on plasma p‐tau217 to screen for amyloid β positivity with further confirmatory testing only in uncertain cases. Nat Aging. 2023:1‐12. doi: 10.1038/s43587-023-00471-5. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. Nat Aging. 2023;3(5):506‐519. doi: 10.1038/s43587-023-00403-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Simrén J, Andreasson U, Gobom J, et al. Establishment of reference values for plasma neurofilament light based on healthy individuals aged 5‐90 years. Brain Commun. 2022;4(4):fcac174. doi: 10.1093/braincomms/fcac174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Coskun A, Braga F, Carobene A, et al. Systematic review and meta‐analysis of within‐subject and between‐subject biological variation estimates of 20 haematological parameters. Clin Chem Lab Med. 2019;58(1):25‐32. doi: 10.1515/cclm-2019-0658 [DOI] [PubMed] [Google Scholar]
- 63. Al Shweiki MR, Steinacker P, Oeckl P, et al. Neurofilament light chain as a blood biomarker to differentiate psychiatric disorders from behavioural variant frontotemporal dementia. J Psychiatr Res. 2019;113:137‐140. doi: 10.1016/j.jpsychires.2019.03.019 [DOI] [PubMed] [Google Scholar]
- 64. Shahim P, Politis A, van der Merwe A, et al. Neurofilament light as a biomarker in traumatic brain injury. Neurology. 2020;95(6):e610‐e622. doi: 10.1212/WNL.0000000000009983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Simrén J, Weninger H, Brum WS, et al. Differences between blood and cerebrospinal fluid glial fibrillary Acidic protein levels: the effect of sample stability. Alzheimers Dement. 2022;18(10):1988‐1992. doi: 10.1002/alz.12806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Benussi A, Cantoni V, Rivolta J, et al. Classification accuracy of blood‐based and neurophysiological markers in the differential diagnosis of Alzheimer's disease and frontotemporal lobar degeneration. Alzheimers Res Ther. 2022;14(1):155. doi: 10.1186/s13195-022-01094-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Nakamura A, Kaneko N, Villemagne VL, et al. High performance plasma amyloid‐β biomarkers for Alzheimer's disease. Nature. 2018;554(7691):249‐254. doi: 10.1038/nature25456 [DOI] [PubMed] [Google Scholar]
- 68. Monane M, Johnson KG, Snider BJ, et al. A blood biomarker test for brain amyloid impacts the clinical evaluation of cognitive impairment. Ann Clin Transl Neurol. 2023. doi: 10.1002/acn3.51863. Published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in Early Alzheimer's Disease. N Engl J Med. 2022;0(0). doi: 10.1056/NEJMoa2212948 [DOI] [PubMed] [Google Scholar]
- 70. Villain N, Planche V, Levy R. High‐clearance anti‐amyloid immunotherapies in Alzheimer's disease. Part 1: meta‐analysis and review of efficacy and safety data, and medico‐economical aspects. Rev Neurol. 2022;178(10):1011‐1030. doi: 10.1016/j.neurol.2022.06.012 [DOI] [PubMed] [Google Scholar]
- 71. Clark C, Lewczuk P, Kornhuber J, et al. Plasma neurofilament light and phosphorylated tau 181 as biomarkers of Alzheimer's disease pathology and clinical disease progression. Alzheimer's Research & Therapy. 2021;13(1):65. doi: 10.1186/s13195-021-00805-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Carobene A, Banfi G, Locatelli M, Vidali M. Personalized reference intervals: from the statistical significance to the clinical usefulness. Clin Chim Acta. 2022;524:203‐204. doi: 10.1016/j.cca.2021.10.036 [DOI] [PubMed] [Google Scholar]
- 73. Mielke MM, Dage JL, Frank RD, et al. Performance of plasma phosphorylated tau 181 and 217 in the community. Nat Med. 2022;28(7):1398‐1405. doi: 10.1038/s41591-022-01822-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supporting information.
