. Plasma N-terminal containing tau fragments (NTA-tau): a biomarker of tau deposition in Alzheimer's Disease. Mol Neurodegener. 2024 Feb 17;19(1):19. PubMed.

Recommends

Please login to recommend the paper.

Comments

  1. Lantero-Rodriguez and colleagues have conducted a comprehensive characterization of the NTA-tau assay, which employs anti-tau mAb (HT7) for capture and anti-tau (Tau12) for detection. This assay was developed and validated by the Clinical Neurochemistry Laboratory in Sweden. Through two studies, BioFINDER-1 and BioFINDER-2, they have demonstrated associations with Amyloid PET, Tau PET, and Neurodegeneration (cortical thickness) in various ways. They identify a stronger association with tau PET than with amyloid in amyloid-positive subjects.

    These findings are largely consistent with our 2018 findings (Mielke et al. 2018) evaluating plasma tTau and P-tau181 versus amyloid PET, tau PET, and cortical thickness.   

    The authors acknowledge that this is not the first NTA-tau assay. It would be beneficial to compare this assay with the one developed by my team at Lilly (Ashton et al. 2022), where the epitopes are even closer together. It would also be worthwhile to examine these assays in clinical trial samples from amyloid removal therapy trials to compare the different responses between tTau and P-tau, thereby further investigating the differential relationship with amyloid.

    Interestingly, the levels in BioFINDER-1 differ significantly from those in BioFINDER-2, as shown in Table 1. Given the similarities in results, this bias might be uniform and due to pre-analytical differences between the studies. However, further investigation into this observation is warranted.

    This work once again underscores the complexity of soluble tau forms in disease. Moreover, it provides additional evidence that exploring post-translational modifications, such as differential proteolytic cleavage, offers additional opportunities for novel biomarker discovery for Alzheimer’s disease and related dementias.

    References:

    . Plasma phospho-tau181 increases with Alzheimer's disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 2018 Aug;14(8):989-997. Epub 2018 Apr 5 PubMed.

    . Plasma and CSF biomarkers in a memory clinic: Head-to-head comparison of phosphorylated tau immunoassays. Alzheimers Dement. 2022 Nov 12; PubMed.

    View all comments by Jeffrey Dage
  2. This study tests the performance of another blood test from the UGOT team in the well-characterized Biofinder cohort. The authors showed convincing results that support that the variance of their new NTA-tau assay is much better explained by tau PET than by amyloid PET levels.

    Interestingly, the assay characteristics, when correlated with other biomarkers, were similar to what has been demonstrated for tau PET in previous studies. For example, NTA-tau levels increased across the disease spectrum but were particularly elevated in later stages. Its longitudinal changes were associated with Aβ status in asymptomatic and symptomatic and parallel atrophy and cognitive dysfunction. This new assay appears to perform similarly to other new fluid markers postulated to better represent tau tangles, such as MTBR-tau243.

    View all comments by Tharick Pascoal
  3. As most phosphorylated-tau biomarkers available in blood or CSF reflect mainly Aβ-related pathology, there is an urgent need for the identification of cost-effective and scalable fluid biomarkers specific for tau aggregate pathology in Alzheimer’s disease. Plasma NTA-tau discussed in this paper increased across the AD continuum, especially during mid- to late-AD stages, and was associated with brain tau aggregates in terms of tau-PET. These findings will make waves in the AD biomarker research field.

    Here, it is important to understand the biology behind the characteristics of the biomarker, i.e., why plasma NTA-tau increases in the late stage of AD. The authors have discussed the possibility that plasma NTA-tau may serve as an indicator of AD tau pathology by showing a correlation between plasma NTA-tau and tau-PET, but whether this relationship is truly causal requires careful attention. Since the NTA-tau is unlikely enriched in neurofibrillary tangles (NFT), it is possible that NTA-tau is not released directly from NFT but is increased in extracellular spaces by passive secretion of soluble tau due to neurodegeneration induced by NFTs accumulation. The exact stage in which plasma NTA-tau levels are altered should be further investigated.

    We have recently reported CSF MTBR-tau243 as a specific biomarker for AD tau pathology through validation by large clinical cohorts analyses (Horie et al., 2023). The MTBR-tau243 fragment may have a direct causal relationship with AD tau pathology, since this tau species has been found to be significantly enriched in AD brain tau tangles (Horie et al., 2021; Wesseling et al., 2020). 

    Regarding plasma NTA-tau, it is not only a potential biomarker reflecting AD tau pathology, but also a promising biomarker for subsequent neurodegeneration. Recently, we proposed a strategy for staging AD by measuring a panel of fluid biomarkers from one aliquot of CSF (Salvadó et al., 2023). The biomarker panel includes Aβ42/40 and p-tau217 for early continuum prediction, p-tau205 and MTBR-tau243 for mid- to late-stage continuum, and total tau for the latest neurodegeneration stage. In the future, plasma NTA-tau may be promising as a marker of neurodegeneration stage to construct the plasma biomarker panel.

    References:

    . CSF MTBR-tau243 is a specific biomarker of tau tangle pathology in Alzheimer's disease. Nat Med. 2023 Aug;29(8):1954-1963. Epub 2023 Jul 13 PubMed.

    . Corrigendum to: CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer's disease. Brain. 2021 Oct 22;144(9):e82. PubMed.

    . Tau PTM Profiles Identify Patient Heterogeneity and Stages of Alzheimer's Disease. Cell. 2020 Dec 10;183(6):1699-1713.e13. Epub 2020 Nov 13 PubMed.

    . Novel CSF tau biomarkers can be used for disease staging of sporadic Alzheimer's disease. 2023 Jul 16 10.1101/2023.07.14.23292650 (version 1) medRxiv.

    View all comments by Kanta Horie
  4. The characterization of tau in CSF and blood has marked a milestone in the field of biomarkers for AD, and we now have a range of assays targeting different tau forms. However, most tau biomarkers are highly correlated among each other and this prompts the question: Do they provide unique insights into the disease?

    In my view, three critical aspects need to be considered. Firstly, Alzheimer's as a continuum involves sequential biomarker changes. We know that changes in p-tau231 and p-tau217 occur early, while p-tau235 changes in later stages of preclinical AD (Lantero‐Rodriguez et al., 2021; Milà-Alomà et al., 2022). Second, we realized that the "T" in the ATN classification not only reflects soluble tau changes (mainly driven by Aβ), but also insoluble tau deposition. The former may be more related to Aβ biomarkers, while the latter to Tau PET. Lastly, beyond early detection, there is a need for biomarkers that can effectively stage and monitor the disease's progression across all the stages of the disease.

    An innovative NTA-tau assay by Rodriguez-Lantero and Salvadó et al. offers additional information, particularly in intermediate and later disease stages. Plasma NTA-tau is characterized by its increase throughout the continuum of AD but, unlike other tau biomarkers, that increase mostly occurs in later stages of the disease (as shown by its higher levels in the A+T--, Braak V-VI, and A+MTL+N++ groups). Plasma NTA-tau variation is mainly explained by Tau PET, not amyloid pathology biomarkers, suggesting that it reflects tau insoluble aggregates. Moreover, plasma NTA-tau predicts tau accumulation, brain atrophy and cognitive decline, and longitudinal changes in plasma NTA-tau are correlated with disease progression.

    Plasma NTA-tau is not the optimal plasma biomarker for detecting AD, neither in early nor later stages, yet it offers valuable insights. Firstly, it aids in disease staging, particularly tracking insoluble tau deposition. This has therapeutic implications, as certain novel disease-modifying treatments may be indicated at specific stages. Additionally, in intermediate and late disease stages, plasma NTA-tau serves as a prognostic biomarker, supporting clinical trials, and may provide prognosis information to patients. Longitudinal changes in plasma NTA-tau also suggest its potential use in monitoring insoluble tau deposition.

    An essential consideration involves comparing plasma NTA-tau with other tau biomarkers related to tau pathology (such as MTBR-tau234), markers of neurodegeneration linked to AD pathology (like BD-tau or t-tau), or those independent of AD pathology (such as NfL).

    Another crucial aspect is the apparent specificity of plasma NTA-tau for AD. As observed in most tau biomarkers, increases in CSF NTA-tau have been reported during acute or rapidly progressive neurological diseases like stroke or CJD (Snellman et al., 2022). Investigating the capacity of plasma NTA-tau to indicate neurodegeneration beyond AD is important.

    In conclusion, plasma NTA-tau emerges as a promising fluid biomarker for in vivo tracking of tau aggregation, and could be used for disease staging and progression monitoring.

    View all comments by Marc Suárez-Calvet

Make a Comment

To make a comment you must login or register.

This paper appears in the following:

News

  1. Tau Fragments in Plasma Track with Tangles, Cognitive Decline