- Review
- Open access
- Published:
Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers
Molecular Neurodegeneration volume 20, Article number: 33 (2025)
Abstract
Alzheimer’s disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Background
Alzheimer's disease (AD) is histopathologically characterized by the deposition of the amyloid-β peptide (Aβ) [1] as well as by the generation of intraneuronal neurofibrillary tangles (NFTs) consisting of abnormally phosphorylated tau (τ) protein (p-τ) [2]. To detect these hallmark lesions, neuropathological examination of tissue either from post-mortem brain autopsy or from ante-mortem brain biopsies is required [3]. Ante-mortem brain biopsies are currently not indicated for the in vivo diagnosis of AD because its result would not yield actionable clinical consequences. Therapeutic options are currently only provided to AD patients who exhibit at least mild symptoms. Treatment of asymptomatic patients can today only be considered in the context of clinical trials [4, 5]. Moreover, the impact of the currently available drugs on the course of AD is still limited [5, 6].
To allow an in vivo diagnosis of AD supported by biological evidence rather than solely clinical criteria, biomarkers have been developed to estimate the presence and severity of Aβ and p-τ pathology in patients ante-mortem. These biomarkers include positron emission tomography (PET) methods visualizing Aβ or p-τ in the brain with radiolabeled tracers that bind specifically to Aβ and p-τ aggregates [7,8,9,10,11] as well as measurement of Aβ or p-τ levels in cerebrospinal fluid (CSF) [12, 13] and blood [14, 15]. With these biomarkers it is now feasible to detect AD not only in symptomatic patients, but also in asymptomatic, also called preclinical, stages of the disease [16,17,18]. Moreover, imaging as well as fluid biomarkers have been developed that reflect downstream consequences of Aβ and p-τ pathology, including neuroinflammation and neurodegeneration, which are involved in AD pathophysiology but are not exclusive to AD. In addition to Aβ and p-τ pathology, most AD cases exhibit additional pathologies in the brain that may contribute to neurodegeneration, neuroinflammation and cognitive decline. Such co-pathologies are (1) the intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (pTDP-43) [19,20,21,22,23,24], (2) the accumulation of α-synuclein (αSyn) inclusions in neurons [19, 23, 25, 26] and (3) vascular lesions [27,28,29,30].
In this review article, we aim to clarify which neuropathologically-defined AD hallmark lesions can be detected with the current biomarkers and at what stages of the disease they become detectable. Second, we will highlight common co-pathologies identified by neuropathologists in AD patients and evaluate whether biomarkers can trace the presence or absence of these co-pathologies. Moreover, novel biomarker approaches will be discussed. Finally, we will illustrate possible concepts of using current and upcoming biomarkers for future clinical diagnosis and clinical trial-related stratifications as well as concepts on interpreting biomarker-based pathogenetic studies.
Neuropathological hallmark lesions
Since the first description of AD by Alois Alzheimer, NFTs and amyloid plaques have been indicated as the two neuropathological hallmark lesions [31]. NFTs were shown to represent fibrillar aggregates of the p-τ protein [32] whereas amyloid plaques mainly consist of fibrillar aggregates of Aβ [1]. Although Aβ1–40 is the most abundantly produced Aβ isoform, Aβ1–42 is the main constituent of extracellular amyloid plaques due to its propensity to aggregate [33, 34]. The Aβ peptide was also found to be the main constituent of vascular amyloid aggregates in cases with sporadic and some familiar forms of cerebral amyloid angiopathy (CAA) [35].
p-τ pathology in NFTs is accompanied by neuropil threads (i.e., p-τ accumulation in neurites) and pretangles (i.e., non-fibrillar accumulation of p-τ in the perikaryon of neurons) [36]. Precursor lesions in the form of initial cytoplasmic and neuropil τ have been described but it is not yet clear whether these lesions represent a still physiological, reversible status of τ phosphorylation or already indicate the start of an irreversible pathological process [37]. The evolution of cerebral p-τ pathology starts in the brain stem and extends into the transentorhinal (Braak NFT stage I), entorhinal cortex (Braak NFT stage II), limbic brain regions such as the hippocampus and the amygdala, the perirhinal cortex (Braak NFT stage III), the superior temporal (Braak NFT stage IV), tertiary and secondary neocortex (Braak NFT stage V), and ultimately primary cortical areas such as the primary visual cortex (Braak NFT stage VI) (Fig. 1) [38]. In this context, it is important to know that the development of p-τ pathology from Braak NFT stage I to VI can take 20 to 30 years [39]. Moreover, p-τ pathology is prevalent in nearly all individuals older than 40 years of age when considering brain stem p-τ pathology as the starting point [38].
Spreading of p-τ pathology in the human brain. a The first neurons exhibiting p-τ are detected in the brain stem in the locus coeruleus, raphe nuclei, and neurons of the basal forebrain [38, 40,41,42]. When p-τ pathology converts into argyrophilic NFT pathology, the transentorhinal and entorhinal cortex becomes involved, followed by limbic regions, the basal neocortex until the entire cortex is filled with NFT pathology including the primary cortical areas. This process is reflected by the Braak NFT-stages [38, 43]. Note that only the “argyrophilic” Braak NFT stages are covered in the neuropathological recommendations for the diagnosis of AD [44] as indicated by the B-scores B1-B3. Brain stem Braak NFT stages are considered as B0. b τ PET imaging with the 18F-MK6240 tracer reflects this process and shows also a stage like propagation of tracer positive regions starting in the (trans)entorhinal cortex, spreading to the amygdala and adjacent temporobasal cortex and finally into the neocortex [45]. Tracer retention is provided as distribution volume ratio (DVR). This is demonstrated in four 18F-MK6240 τ PET images from (1) a 79-year-old, cognitively normal control case (CDR-score = 0) without tracer retention, (2) a 68-year-old, non-demented, asymptomatic AD case (CDR-score = 0) with initial entorhinal τ tracer retention, (3) a 68 year old prodromal AD case (CDR-score = 0.5) with mild cognitive impairment and tracer retention extending to adjacent medial temporal lobe regions including amygdala and temporal neocortex, and (4) a 77-year-old symptomatic AD case (CDR-score = 1) with extensive neocortical τ tracer retention. The 18F-MK6240 τ tracer was chosen for depicting pathology progression in τ PET because it is a second generation τ tracer that does not show significant off-target binding especially in the medial temporal lobe. Schematic representations in a are modified from Thal & Tomé 2022 [46] with permission
Multiple types of Aβ plaques exist [47,48,49,50,51,52]. All of them contribute to the hierarchical sequence that characterizes the progression of Aβ pathology in the AD brain [49]. Diffuse non-neuritic plaques represent, in this context, an early-stage of Congo-red negative accumulations of Aβ that can—depending on the brain region of occurrence—develop into neuritic plaques [47, 48, 53]. This evolution occurs when amyloid aggregates associate with dystrophic neurites and reactive glial cells [47, 48, 53]. The Consortium to Establish a Registry for AD (CERAD) score represents a semi-quantitative measure based on the density of neuritic plaques, varying from none to frequent [54]. Given that neuritic plaques represent only Aβ plaques with dystrophic neurites usually containing p-τ, the CERAD score does not measure the extent of the Aβ pathology but rather represents the co-occurrence of Aβ and p-τ in plaques. The assessment of the entire extent of Aβ plaque pathology is done by assessing the phases of Aβ plaque distribution in the brain according to Thal et al. (Aβ phases) [55]. The first spot where amyloid plaques were found varies among places in the frontal, parietal, temporal, or occipital cortex [55]. After initiation in the neocortex (Aβ phase 1), Aβ plaques develop also in allocortical regions (Aβ phase 2), basal ganglia and diencephalon (Aβ phase 3), the midbrain and the inferior olivary nucleus (Aβ phase 4), and finally in the cerebellum and the pons (Aβ phase 5) (Fig. 2). The phases of Aβ plaque pathology correlate with the Braak NFT stages and confirm disease progression over a span of 20 to 30 years [55].
a Spreading of Aβ plaque pathology. Neuropathologically, the first plaques are found in the neocortex. The light-pink area describes, in a given phase, the area that the neuropathologist has to screen to find the first 2–3 amyloid plaques. Most of the area can be free of plaques at this stage. Full pink marked areas represent usually moderate amounts of plaques whereas full red marked areas show full-blown plaque pathology. The first area affected is the neocortex in phase 1. Propagation into allocortex (phase 2), basal ganglia & diencephalon (phase 3), midbrain & medulla oblongata (phase 4), and finally into pons & cerebellum in phase 5 is indicated by the respective Aβ phases and the A (amyloid)-scores [44, 55]. b18F-Flutemetamol amyloid PET images indicate that in Aβ phases 1–2 tracer retention does not obviously differ from Aβ phase 0 cases whereas Aβ phases 4 and 5 are easily identifiable by eye. Aβ phase 3 shows increased tracer retention compared to controls that can be measured by assessing SUVRs [7, 56]. Below the PET images SUVR thresholds are provided to distinguish Aβ phases 3, 4, and 5 from controls and from one another. The SUVRs were determined with pons as reference region [56]. Images in b are reproduced from Thal et al. 2015 [7] under CC BY-NC-ND license and with permission. The figure was produced in the context of a GE-Healthcare clinical trial: ClinicalTrials.gov identifier NCT01165554
The National Institute on Aging and the Alzheimer’s Association (NIA-AA) developed a scoring system that incorporates the Aβ phases, Braak NFT stages, and CERAD neuritic plaque scores into a global ABC score. In this score, A indicates the Aβ phase and ranges from A0 (no Aβ plaques) to A3 (final Aβ phases 4–5), B indicates the Braak NFT stage and ranges from B0 (no tangles) to B3 (final Braak stages V-VI) and C indicates the CERAD stage and ranges from C0 (no neuritic plaques) to C3 (frequent neuritic plaques). Based on this ABC score, the extent of AD neuropathological changes (ADNC) can be divided into “Low”, “Intermediate” and “High” categories [44] (for A- and B-score see also Figs. 1, 2). In this context, it is essential to note that the brain stem stages of p-τ pathology are not covered by the ABC score and technically fall under the category B0 = no NFT pathology.
Despite the typical hierarchical spreading patterns of Aβ and p-τ pathology, heterogeneity among the local severities of Aβ and p-τ pathology have been described [57]. Best known are the p-τ pathology distribution related subtypes of AD [58]. The “typical “ variant is characterized by a balanced severity of p-τ pathology in the cortex and the hippocampus as predicted by the Braak NFT stage. The limbic-predominant subtype shows predominant p-τ pathology in the hippocampus whereas other brain regions are less severely affected. This is in contrast with the hippocampal sparing subtype, which is characterized by less severe hippocampal involvement but severe cortical p-τ pathology [58]. In autosomal dominant AD cases, end-stage NFT and Aβ plaque pathology is usually seen at autopsy with specific plaque types such as cotton-wool plaques [59,60,61,62,63,64]. Coarse grained plaques were reported in early onset AD [65]. In patients with Down syndrome, amyloid plaque pathology starts to develop as early as 12 years of age [66]. Later, the pathology shows all features of AD even cotton wool plaques [67]. Only the inflammatory reaction in Down syndrome shows a spectrum of involved microglial cell types that differs from classical sporadic AD [68]. The development of Aβ plaques is frequently associated with that of CAA, which is present in 80% to 100% of symptomatic AD cases [69,70,71,72].
Aβ and p-τ are not restricted to the brain. They have also been reported in the retina [73,74,75,76,77,78,79,80]. p-τ pathology frequently occurs in the retina and follows a distinct sequence in which the layers of the retina become involved: stage 1 = affection of the outer plexiform layer; stage 2 = additional affection of the inner nuclear layer; stage 3 = additional affection of the inner plexiform layer [78]. Current studies see retinal p-τ pathology in AD but also in non-AD cases. This led to the idea that a primary retinal tauopathy (PReT) could represent a precursor lesion for retinal AD manifestation [78]. Interestingly, retinal p-τ pathology was also associated with a worsening of vision [78]. Experiments in τ-transgenic mice showed that the occurrence of p-τ pathology in retinal ganglion cells is associated with a reduced ganglion cell density [81]. In contrast, retinal amyloid pathology is less frequently observed and often represents very small lesions [74, 76, 78, 82]. Often, visualization of Aβ pathology in the retina requires specific staining techniques (flatmount technology that stains the retina or pieces of the retina as one flat mounted piece) [74]. The prevalence of retinal Aβ varies among different studies but is mainly observed in symptomatic AD cases in neuropathological studies [73, 74, 76, 78, 82]. Moreover, CAA-affected blood vessels in the retina were also reported [75].
Neuropathology of co-pathologies in AD brains
In addition to the AD hallmark lesions, other pathologies like vascular lesions, transactive-response DNA-binding protein 43 (TDP-43), αSyn pathology [19,20,21,22,23, 25,26,27,28,29,30], and granulovacuolar degeneration (GVD) [83,84,85] frequently occur in the elderly brain. Moreover, neuroinflammatory response to AD pathology impacts disease progression [86,87,88,89,90].
Vascular lesions, such as bleedings and infarcts are often found in the elderly brain [28, 91,92,93,94] and can have various causes. For example, atherosclerosis with thrombosis, thromboembolic occlusion of blood vessels and cardiogenic thromboembolic events are AD independent causes of brain infarcts [95,96,97,98]. Similarly, hypertensive arteriopathy is an AD-independent cause of intracerebral hemorrhage [95, 98, 99]. CAA, on the other hand, can also cause cerebral infarcts and bleedings [28, 70, 100, 101]. Given that CAA is the result of the accumulation of the same Aβ peptide in the vessel wall that also accumulates in amyloid plaques and given the strong association of CAA with AD-related β-amyloidosis, there is a potential link between CAA-related vascular lesions and AD [1, 35]. This is further supported by the fact that mouse models overexpressing transgenic amyloid precursor protein (APP) via a neuron-specific promoter produce not only Aβ plaques but also vascular Aβ deposits, i.e., CAA [102]. Moreover, CAA allows the distinction of two types of CAA which are associated with pathogenetically different forms of AD [103, 104]: Capillary CAA (CAA type 1) is strongly associated with the apolipoprotein E (APOE) ε4 allele whereas CAA cases lacking capillary amyloid deposits (CAA type 2) is not [105]. Moreover, CAA type 1 is associated with blood flow disturbances as shown in an animal model [106], and with the manifestation of allocortical microinfarcts in the human brain [107].
TDP-43 pathology in neuronal cytoplasmic inclusions and threads in elderly individuals with or without signs of amnestic dementia has recently been termed “limbic predominant age-related TDP-43 encephalopathy neuropathological changes “ (LATE-NC) [108]. LATE-NC occurs by definition either on its own or together with ADNC. Interestingly, TDP-43 aggregates are present in 20% to 74% of AD cases in which they can interact with p-τ in NFTs [21, 24, 109,110,111,112,113,114]. LATE-NC starts in the amygdala, hippocampus and entorhinal cortex (stage 1). From there it increases in severity, expands into the perirhinal cortex (stage 2) and later into the frontal cortex (stage 3) [20, 115]. With increasing age, the prevalence and stage of LATE-NC increases [22, 23]. TDP-43 pathology can also be seen in the retina. However, retinal TDP-43 pathology observed so far was either related to amyotrophic lateral sclerosis (ALS) or frontotemporal lobar degeneration (FTLD)-TDP [116,117,118]. About 43% to 63% of AD cases exhibit αSyn Lewy body pathology [119,120,121,122]. This pathology is characterized by Lewy bodies in neurons and Lewy neurites consisting of αSyn aggregates and is seen in AD cases with or without Parkinson motor symptoms. αSyn pathology in AD patients influences the clinical presentation (e.g., higher frequency of memory symptoms compared to pure dementia with Lewy bodies (DLB) and higher frequency of visual hallucinations, REM sleep behavior disorder or autonomic dysfunction compared to pure AD) and increases the rate of cognitive decline and mortality compared to pure AD or DLB [123,124,125,126]. The spread of αSyn pathology has been described to follow either a classical caudo-rostral distribution pattern [127] or alternatively the amygdala-predominant pattern [128,129,130,131,132]. The caudo-rostral distribution pattern starts with Lewy bodies in the dorsal nucleus of the vagal nerve (stage 1), extending into the locus coeruleus (stage 2), substantia nigra (stage 3), basal nucleus of Meynert and amygdala (stage 4), hippocampus (stage 5), and, finally, to the neocortex (stage 6) [127, 129, 133, 134]. In cases with the amygdala-predominant pattern, the amygdala is often affected without or with very mild accompanying involvement of the brain stem nuclei [128,129,130, 135, 136]. Interestingly, amygdala-predominant αSyn pathology has been frequently reported in AD cases [129, 136, 137]. At the cellular level, pathological αSyn accumulation has been reported in these cases within the same neurons that also bear NFTs [114, Ishizawa, 2003 #525, 129]. The prevalence of αSyn pathology in the general population is lower than that of AD hallmark lesions or LATE-NC [23]. Moreover, in the oldest old, the frequency of αSyn pathology decreases likely due to a lower life expectancy of cases with αSyn pathology compared to those without [23]. αSyn pathology has also been described in the retina of Parkinson’s disease patients and of patients with DLB [138,139,140].
GVD is neuropathologically characterized by large cytoplasmic vacuoles in neurons containing dense granules and a high number of proteins, which are often phosphorylated, including p-τ and pTDP-43 but also casein kinase 1δ/ε, charged multivesicular body protein 2B, and lysosome-associated membrane protein 1 occurring in the aging and demented brain [83, 141,142,143]. The latter proteins suggest that lysosomal degradation and autophagy may play a role in the development of GVD [143, 144]. GVD also contains the activated components of the necrosome, i.e., the executor complex for necroptosis, which is a distinct form of regulated cell death [145, 146]. The development of GVD starts in the CA1/subiculum region of the hippocampus, followed by expansion into the entorhinal and temporal cortex, into the amygdala and finally into the frontal cortex [85]. GVD has been reported in multiple neurodegenerative disorders including AD, ALS, Guam disease, etc. [85, 142, 143, 147]. In this context, the most advanced stages of GVD expansion were restricted to AD cases [85]. Interestingly, the GVD frequency correlated negatively with the neuronal density in the CA1 region of the hippocampus as well as in layer III of the frontal neocortex in AD cases [145]. Currently, no attempts have been made to detect GVD in vivo with biomarkers.
Finally, neuroinflammation is frequently found in AD cases and is associated with the disease development [48, 87, 90, 148,149,150,151]. Interestingly, late-stage AD cases showed a reduction of activated HLA-DR-positive microglial cells often exhibiting a conversion into the phagocytic phenotype [151]. With attention to single cell transcriptomic profiles, distinct types of microglial cells have been identified with a specific type of disease-associated microglial cells (DAMs) [152,153,154]. The inflammasome pathway had been identified as an important pathway in the context of AD [155,156,157]. Moreover, inflammasome activation is part of the pyroptosis pathway [158]. Pyroptosis is an inflammatory type of regulated necrosis and can cause cell death in multiple ways [158, 159]. In AD, the pyroptosis pathway is activated in microglial cells, astrocytes, and neurons. However, in each of the cell types, this activation is triggered via a different pathway [160]. Moreover, neuronal and microglial pyroptosis pathway activation both contribute independently to neuron death in AD [160, 161].
Sequence of neuropathological events throughout the pathogenesis of AD and its modification by co-pathologies (Fig. 3)
a Development of Aβ phases (0–5), Braak NFT stages (0-VI), GVD stages (indicative for necroptosis pathway-related neurodegeneration; 0–5), CAA stages (0–3), LATE-NC stages (representing TDP-43 pathology; 0–3), and Braak LBD stages (0–6) during the course of AD as represented by the Aβ phase in 323 autopsy brains using a locally weighted polynomial regression method based on the average pathology stages at each respective Aβ phase. The trajectory of each pathology is depicted by a line diagram with Aβ phase as x-axis. Note that not all AD cases exhibited LBD pathology or LATE-NC and that the resulting graphs include both cases with and without the respective pathologies. b To illustrate the sensitivity of 18F-flutemetamol amyloid PET compared to the neuropathological Aβ phases, we show a line diagram obtained through locally weighted polynomial regression based on the average Aβ PET stage at each respective Aβ phase. The Aβ phase is depicted by the x-axis and the y-axis shows the estimates for the Aβ phases dertermined with a given method. The black line represents the neuropathological Aβ phases whereas the red line represents the PET Aβ phase estimates according to Thal et al. [56] as measurement for 18F-flutemetamol tracer retention. The black diagonal line represents the neuropathological Aβ phase and was included as reference. Amyloid PET provides reliable detection of Aβ phase 3 and higher. The arrow in b indicates when amyloid PET detects the pathology. This figure is based on previously published results covering the proper box-plot diagrams of the illustration in b [56]. The statistically relevant information for a and b is depicted in Supplementary Fig. 1. c Schematic representation of a hypothetical cascade of neurodegeneration in AD and its key proteins Aβ, p-τ, and the co-pathology-related proteins pTDP-43 and αSyn. The first step in this hypothetical cascade is the development of p-τ as PART, which becomes further accelerated by Aβ (step 2) and can lead to the induction of GVD-necroptosis and neuron death as final steps in the neurodegenerative process. In this context, it is essential to note that necroptosis is only one of the cell death pathways that can execute neuron death in AD [161]. As a third step in the neurodegenerative process, co-pathologies can exacerbate p-τ pathology and neurodegeneration as shown for pTDP-43 and αSyn [19, 25, 136, 162]. At the moment p-τ and Aβ pathology can be detected with validated biomarkers whereas reliable biomarkers for the co-pathologies are not yet available
When analyzing the relationships among the pathological parameters in AD and the co-pathologies, it became apparent that p-τ pathology, as indicated by the Braak NFT-stages, usually precedes the appearance of Aβ plaques, CAA, GVD, LATE-NC and αSyn pathology (Fig. 3) [38, 163,164,165,166]. As AD progresses, Aβ pathology takes over the lead and p-τ pathology follows (for the data shown in Supplementary Fig. 1: Sign test, p = 0.004 (adjusted for multiple testing); n = 323). In a third step, GVD starts to occur and progresses through distinct stages of distribution (for the data shown in Supplementary Fig. 1: Sign test, p < 0.001 (adjusted for multiple testing); n = 323) followed by CAA stage, LATE-NC stage and Braak LBD stage (for the data shown in Supplementary Fig. 1: Sign test, p = 0.001 (adjusted for multiple testing); n = 323), which all three rise simultaneously (for the data shown in Supplementary Fig. 1: Sign test, p = 1.0 (adjusted for multiple testing); n = 323) (Fig. 3a). Results from studying the accelerating effect of Aβ on p-τ in animal models support the hypothesis that Aβ accelerates the propagation of p-τ pathology [167,168,169,170] possibly via an interaction of both proteins with the cellular prion protein [169, 171]. Downstream to Aβ and p-τ pathology, GVD develops (Fig. 3a, c) [172,173,174] containing components of the activated necrosome [145] and executing the regulated cell death pathway of necroptosis [158], which leads to neuron death [145, 175]. Pharmacological inhibition of the necroptosis pathway stopped the development of GVD and neuron loss in APPxτ-transgenic mice [175]. Interestingly, co-pathologies such as LATE-NC, αSyn pathology, and CAA most frequently follow the neurodegenerative process parallel to necroptosis pathway activation indicated by GVD (Fig. 3a, c). By doing so, co-pathologies are capable of accelerating the degenerative process as shown for pTDP-43 in LATE-NC, which interacts with p-τ [109] and is associated with increased p-τ seeding and propagation activity [176]. Additionally, accelerated neuron loss has been associated with an increase of GVD with necroptosis pathway activation [162]. These synergy effects were functionally demonstrated in model systems [177, 178]. Very recently, hippocampal and amygdala neurons were found to contain co-localized p-τ, pTDP-43 and αSyn [136] indicating that protein interactions in AD are not limited to bilateral interactions but can also be multilateral. Structural equation modelling indicated that also αSyn pathology and pyroptosis contribute to the severity of neuron loss in AD [136, 161].
To conclude, the pathological hallmark lesions in AD interact, at least indirectly, with one another. Aβ is, in this context, an accelerator for p-τ pathology. p-τ is capable of triggering neuron death by inducing GVD-linked necroptosis. Pyroptosis activation, TDP-43 and αSyn pathology presumably have additional effects on driving neurons into cell death.
In parallel, CAA, especially CAA type 1, also impacts neuronal survival as this type of CAA leads to the development of blood flow disturbances and microinfarcts [106, 107]. Other authors also reported an association of severe CAA with cerebral microinfarcts [101]. Accordingly, CAA can contribute to the degenerative process in AD via additional blood flow alterations.
Biomarkers in the clinics—relationship with neuropathology
The AD hallmark pathologies Aβ plaques and NFTs, as well as certain co-pathologies, can be estimated ante-mortem using specific biomarkers. An overview about the underlying pathologies and their related biomarkers is provided in Table 1. Specificity and sensitivity towards neuropathological ground truth has been estimated according to the current literature and these estimates are depicted in Fig. 4. In the following paragraphs we introduce the respective PET and fluid biomarkers, first for biomarkers of AD hallmark pathologies, second for disease progression, and third for frequent co-pathologies, and discuss their diagnostic potential. Although included in Fig. 4 for completeness, MRI for vascular lesions and CAA as well as αSyn and translocator protein (TSPO) PET will not be discussed in detail. We refer to the respective literature [179,180,181,182,183,184,185].
Estimation of biomarker performance (sensitivity, specificity, quantitative accuracy, and accessibility) targeting the estimation of the underlying neuropathology as ground truth. Imaging, CSF and blood biomarkers are compared for the detection of Aβ pathology, p-τ pathology, vascular pathology (CAA, infarcts and bleedings), αSyn pathology, neuroinflammation and neurodegeneration markers. The estimation was made in accordance with the literature cited in this article. Radar plots were generated using Plotly. Notes: Plasma p-τ is more specific for estimating Aβ pathology than p-τ pathology [15, 186]. The detection of CAA-related microbleeds is very specific and sensitive for symptomatic CAA cases. The bulk of the CAA cases without bleedings and siderosis that can be detected at autopsy could not be detected with MRI [187, 188]. Infarcts and hemorrhages can be excellently diagnosed by MRI. Only microinfarcts usually escape due to resolution restrictions in MRI imaging [179]. As neuroinflammation markers we included those markers that are used to determine AD-related neuroinflammation. These are the TSPO PET and measurements of glial fibrillary acidic protein (GFAP) in the CSF and blood plasma. Note that ground truth as the highest quality level does not apply to the accessibility parameter. For the determination of the degree of neurodegeneration we focus on CSF and plasma NfL as the best established fluid marker for this purpose and SV2A PET as synaptic marker to document synapse loss. MRI for determining the level of atrophy and total CSF τ are further markers for neurodegeneration but were not included in this figure as in our opinion NfL is superior to total CSF τ and synaptic PET provides a better view on the morphological correlate of neurodegeneration, i.e., a reduction of synaptic density even before substantial atrophy becomes visible
Racial disparity in the levels of some biomarkers have been described [189, 190] but comparison of neuropathology with imaging or fluid biomarkers in end-of-life studies is, today, mainly restricted to US or European cohorts [8, 9, 11, 191, 192]. Further studies comparing biomarker levels in individuals of Caucasian, African, Asian, American, and Australian origin to pathological ground truth in end-of-life studies are required to clarify whether racial disparity has influence on the quality of biomarker estimates. Given the lack of these data, we will focus on the results obtained mainly in Caucasian cohorts. This is a limitation of this review.
Biomarkers of AD hallmark pathologies
Amyloid PET
PET imaging is an opportunity to visualize specific pathological processes using radiolabeled ligands administered intravenously that bind specifically to their target. In the case of amyloid PET, Aβ aggregates in the brain represent the target. Pittsburgh compound B (PiB) was the first developed amyloid tracer but includes 11C as radioactive isotope whose short half-life (20 min) limits its use in clinical settings [193, 194]. Today, three amyloid tracers incorporating the 18F isotope (110 min half-life), namely 18F-florbetapir [9], 18F-flutemetamol [195], and 18F-florbetaben [8] are commercially available and have been approved for clinical use after undergoing phase 3 end-of-life trials [8, 9, 191]. Regulatory approval of amyloid PET imaging tracers is based on standardized procedures for visual interpretation of the scans by trained readers. These procedures result in positive or negative visual reads. All aforementioned amyloid tracers specifically label amyloid aggregates in the brain but their sensitivity only allows the detection of moderate to severe amyloid pathology whereas early phases (amyloid phases 1 and 2) of β amyloidosis could not be distinguished from age-matched control brains. These data have first been obtained for 18F-flutemetamol and 11C-PiB tracers [7, 11, 56] and later for all four tracers in a study in which standardized uptake value ratios (SUVRs) were converted to Centiloids allowing direct comparison of tracer measurements [196]. Definition of amyloid tracer retention tracer thresholds (in SUVRs or Centiloids) allow principally a distinction between the different Aβ phases from phases 0–2 vs. 3 to 5 [56, 196, 197]. Longitudinal follow-up studies comparing SUVR measurements or Centiloid values with the referring post-mortem Aβ phases determined in end-of-life studies, revealed that Aβ tracer retention increases in parallel with Aβ deposition in the brain, starting from Aβ phase 3 [56, 198,199,200,201,202]. In addition, to the neuropathological hierarchical pattern of Aβ plaque deposition, amyloid PET allowed the identification of several cortical areas that are more prominently affected than others [203]. Variation of the primarily affected neocortical areas among groups of cases was reported [204]. Today, the approved indication for amyloid PET is the evaluation of neuritic amyloid plaque density in individuals with cognitive impairment who are being evaluated for Alzheimer’s disease or other causes of cognitive decline [205]. For clinical trials, amyloid PET has played a critical role in confirmation of amyloid positivity for amyloid lowering drug trials and for evaluating the therapeutic effect of these drugs. Amyloid PET has also been instrumental in the definition of the asymptomatic stage of AD.
τ PET
Currently, the most widely used 18F-labeled τ PET ligands [206] are 18F-flortaucipir,18F-MK6240, 18F-PI2620, 18F-RO948, and 18F-GTP1. Other τ PET tracers have been abandoned based on company strategic decisions, such as 18F-JNJ067 [207], or because of lack of specificity for p-τ, such as 18F-THK5351 and 18F-THK5317. τ PET ligands are sometimes grouped as first (such as 18F-flortaucipir) versus second-generation tracers (such as 18F-MK6240, 18F-PI2620, 18F-RO948, and 18F-GTP1) [206]. The second-generation tracers show no off-target binding near the medial temporal cortex (choroid plexus) and less off-target binding in the striatum [208] (Fig. 1b). According to the end-of-life diagnostic phase 3 trial of 18F-flortaucipir [209], a binary visual read of 18F-flortaucipir PET as either “not consistent with AD” or “consistent with AD” can detect neuropathological Braak V-VI with high sensitivity and average-to-high specificity, meeting the primary endpoint. In this study, a positive visual read, i.e., the visual analysis indicated “consistent with AD”, could only be made if the tracer uptake extended beyond the medial and anterior lateral temporal cortex because nonspecific binding of the ligand to the choroid plexus renders evaluation of medial temporal binding of the 18F-flortaucipir tracer challenging. Around 75–80% of cases in Braak stage III or IV were read as negative. This pivotal study contributed to the Food and Drug Administration (FDA) approval of 18F-flortaucipir as a radioactive diagnostic agent for adult patients with cognitive impairment being investigated for AD. The ability of 18F-flortaucipir to detect neuropathological stage Braak IV or higher was confirmed in a second, academic study [210]. The degree of in vivo neocortical 18F-flortaucipir signal correlates closely with post-mortem p-τ load from anti-p-τS202/T205 immunostaining [10, 210], and less so in limbic regions [10]. τ PET positivity typically starts near symptom onset. Similar to 18F-fluorodeoxyglucose (FDG) PET, but in contrast to amyloid PET, the topographical extent of τ PET positivity holds prognostic value: more widespread τ PET positivity is associated with faster cognitive decline [211]. τ PET, therefore, contributes to biological staging of the disease [212].
Results obtained with first-generation τ PET tracers may not be generalizable to the second-generation tracers because of differences in signal amplitude, off-target binding and meningeal uptake. Based on a direct within-subjects comparison, accuracy based on visual reads, was similar between 18F-flortaucipir and 18F-MK6240 [208]. 18F-MK6240 retention had a dynamic range that was 1.7 to 1.9 times larger than that of 18F-flortaucipir. A higher dynamic range is advantageous for all research based on semi-quantitative assessments, such as longitudinal within-subject comparisons or correlational analyses. Given the absence of choroid plexus retention and the higher dynamic range, it is possible that the second-generation τ PET tracers may detect earlier stages of p-τ aggregation than 18F-flortaucipir but that remains to be demonstrated.
According to an in vivo 18F-flortaucipir τ PET study, the ‘classical’ distribution defined neuropathologically by Braak and Braak [43] was present in AD cases as depicted in Fig. 1 [45, 213] but only in 30%, while other cases showed medial temporal sparing on τ PET (20%), a posterior occipitotemporal pattern (30%) or a left-lateralized temporoparietal phenotype (20%) [214]. The prevalence of these patterns will probably strongly depend on the type of cohort, e.g. research and academic memory clinic based, and likely do not reflect the population-based prevalence. The topographical heterogeneity fits with the concept of different neuropathological patterns of p-τ distribution, limbic-predominant (more frequent in older cases) or hippocampal sparing [58]. The ability of τ PET to monitor the topographical pattern of increased τ aggregates renders them very attractive as surrogate markers for assessing the effect of study drugs targeting τ aggregation in AD.
In AD, p-τ occurs under different forms, as explained above. Furthermore, different stages of NFT maturity have been described based on staining affinities [37, 215]. The precise details of which AD-related p-τ aggregates are mostly responsible for τ PET positivity (neuropil threads, different types of NFT, maybe also τ astrogliopathy) is still unresolved. However, in vitro autoradiography binding assays proved the specific binding of the τ-PET tracers showing a similar distribution as the immunohistochemical staining with antibodies against p-τ although varying in sensitivity and specificity [216,217,218,219,220,221,222].
Neuropathologically, primary age-related tauopathy (PART) refers to the presence of NFTs in the absence of amyloid plaques. In a large study of more than 1,840 cases scanned with 18F-MK6240 [223], among the amyloid PET negative healthy controls, 8% showed increased τ PET signal in medial temporal cortex, while the remainder was τ PET negative. The opposite pattern, amyloid positivity without τ PET positivity is much more common and is almost the default pattern in the asymptomatic stage of AD. Among amyloid PET negative cases with suspected non-AD pathophysiology (SNAP) [224], 13% had increased medial temporal τ PET signal and 25% had increased medial temporal and neocortical τ. Clearly, amyloid PET negativity does not preclude τ positivity although the most typical sequence is that amyloid PET positivity precedes τ PET positivity. Possibly, PART accounts for these relatively rare cases of medial temporal τ PET signal in the absence of amyloid positivity, but a post-mortem study in patients who had received an 18F-flortaucipir study showed no tracer retention in PART [210]. Alternatively, the amyloid PET may be false negative.
Core CSF biomarkers for ADNC diagnosis
Given the relatively high costs for PET imaging and the limited access to this technology, cheaper and easier accessible biomarkers were needed. As body fluids can be taken easily and sent to laboratories that offer proper expertise, these fluid biomarkers are easier to access, and analysis is cheaper. Today, CSF biomarkers are well established whereas blood biomarkers are currently emerging and may replace the CSF biomarkers in the near future. CSF core AD biomarkers reflecting amyloid plaques and τ NFTs have been incorporated in the diagnostic criteria for supporting in vivo AD diagnosis since 2011, and include CSF Aβ1–42, total τ and phosphorylated τ at threonine181 (p-τ181) [212, 225].
CSF Aβ1–42 levels are typically lower in AD patients than in healthy elderly or patients with other dementia types (e.g. FTLD) as a result of sequestration to the brain, where it aggregates [226, 227]. However, low CSF Aβ1–42 levels have relatively low specificity as they can also be decreased in CAA, hydrocephalus, cerebral microangiopathy, Lewy body dementia and other neurological disorders (Fig. 4) [228,229,230,231,232,233,234,235]. Since CSF Aβ1–40 levels remain unchanged in AD, specificity can be substantially improved by using the Aβ1–42/Aβ1–40 ratio as it compensates for interindividual differences in total Aβ production [236,237,238,239]. In addition to Aβ, τ levels in CSF also serve as core diagnostic biomarkers for AD. In contrast to Aβ, τ is an intracellular protein that is thought to be released in the CSF through neurodegeneration, a process not specific to AD. CSF levels of p-τ181, on the other hand, are specifically increased upon the presence of AD pathology and are typically not altered in other neurodegenerative disorders [240]. Neuropathology as well as PET-based studies have shown that CSF-based changes in total τ as well as p-τ181 (e.g. at Aβ phase 4 or 5) follow both CSF-based Aβ1–42/Aβ1–40 changes and subsequent PET-based Aβ changes [237, 241,242,243,244]. Pathological levels of CSF total τ or p-τ181 may therefore not necessarily be present in all symptomatic cases with amyloid biomarker proven AD in a prodromal or early dementia phase (lack of sensitivity; Fig. 4) [212, 225].
Translation of core CSF AD biomarkers to blood – relationship to neuropathology
Although CSF- and PET-based methods have transformed the in vivo diagnosis of AD in expert centers who have access to these methods, their invasive nature, relatively high cost in the case of PET, and dependence on specialized facilities make them impractical for large scale testing. Blood-biomarkers provide a more accessible and scalable alternative, which may transform the diagnostic work-up of cognitive disorders at the population level. However, blood-based biomarker development has faced its own challenges. The up to 100 times lower levels in blood than in CSF, the contribution of peripheral biomarker production to blood, which is already a very complex matrix [245], as well as the influence of proteolytic degradation [246] and co-morbidities on blood-based biomarker levels all complicate the determination of their relationship with brain-level changes [247,248,249]. Nevertheless, technological advancements in the immunoassay field addressed the limitations of traditional first-generation enzyme-linked immunosorbent assays (ELISAs) typically used for biomarker quantification in CSF. These advancements include the development of ultrasensitive assays, which operate on the same principle as traditional ELISAs, where an antigen is recognized by an immunocomplex, and upon addition of a substrate, generates a detectable and quantifiable signal. However, these ultrasensitive methods reach higher sensitivity using advanced detection techniques such as (electro)chemiluminescence (Mesoscale Discovery (MSD), Lumipulse and Elecsys platforms) or bead-based single molecule digital detection (single molecule array (Simoa) platform) rather than relying on colorimetric signals. Additionally, improvements in ELISA design as well as increased attention for preanalytical parameters (e.g. centrifugation delay and parameters, sample tube, storage delay) have allowed quantification of AD-related proteins in blood.
Plasma Aβ1–42/Aβ1–40 ratio
Similar to CSF, the plasma Aβ1–42/Aβ1–40 ratio is decreased in individuals demonstrating ADNC at autopsy, regardless of clinical symptoms [192, 250, 251]. Several studies reported a decreased plasma Aβ1–42/Aβ1–40 ratio at early disease stages, i.e., intermediate ADNC and moderate plaque burden as determined by CERAD staging, after which it plateaued [192, 250]. This is comparable to the temporal trajectory observed for CSF Aβ1–42/Aβ1–40 [236, 252]. However, whereas the CSF Aβ1–42/Aβ1–40 ratio is reduced by more than 50% in AD [233], plasma-based reductions are less pronounced (10–20%) [253,254,255]. Importantly, the performance of plasma Aβ1–42/Aβ1–40 to detect AD is highly dependent on the assay used for its quantification. Whereas plasma Aβ measurements by first-generation Aβ ELISAs lacked performance as AD biomarkers, novel ELISAs that incorporate N-terminal – rather than midregion – antibodies, employ improved conjugation methods, and demonstrate comparable performance to equivalent ultrasensitive Simoa assays [253, 254]. Yet, highest performance of the Aβ1–42/Aβ1–40 ratio as an AD biomarker is obtained when it is quantified by mass-spectrometry based methods rather than immunoassays [253]. For example, classification accuracy for detecting amyloid status across the AD continuum – as determined by areas under the curve (AUC) in receiver operating characteristic (ROC) analyses—ranges between 0.83 and 0.97 for mass-spectrometry methods [253, 255,256,257] and between 0.62 and 0.79 for immunoassays (e.g., Simoa, ELISA) [253, 254, 258,259,260,261].
Plasma total and p-τ
Not all core CSF biomarkers have been successfully translated to blood. Blood-based total τ levels, for instance, are lower than those of Aβ1–42 and Aβ1–40, necessitating ultrasensitive methods for detection. However, plasma total τ measurements using either Simoa or Elecsys platforms have demonstrated poor correlation with CSF-based total τ and largely overlap between several neurodegenerative disorders as well as between clinical AD stages [262,263,264,265,266,267]. As a result, blood-based total τ has poor diagnostic and prognostic performance in AD [262,263,264,265,266,267,268]. In contrast, p-τ181, whose blood levels are even lower than those of total τ, has demonstrated consistent and specific elevations in plasma of patients with underlying ADNC [269, 270] when measured with ultrasensitive methods. These p-τ181 elevations did, however, occur in later stages than Aβ1–42/Aβ1–40 decreases (i.e., at high ADNC including frequent plaque burden according CERAD and Aβ phase 4 or 5) [192, 250, 271, 272]. Consequently, plasma Aβ1–42/Aβ1–40 correlates strongly with amyloid burden in early disease stages, whereas p-τ181 demonstrated a stronger correlation at later amyloid stages or once cognitive symptoms emerge than at earlier disease stages [192, 251]. Moreover, while Aβ1–42/Aβ1–40 mainly correlates with amyloid load [192, 254, 259], plasma p-τ181 is independently associated with both cerebral amyloid and p-τ pathology [192, 273]. Of note, the τ protein has over 40 possible phosphorylation sites and alternative p-τ species have also shown promise in AD diagnosis with different associations to cerebral p-τ pathology and Aβ pathology. p-τ231, for example, increases in earlier stages than p-τ181 in plasma as well as in CSF, yet has comparable performances to plasma p-τ181 beyond these earliest stages [250, 274,275,276]. p-τ217, on the other hand, has consistently shown superior performance to p-τ181 as well as p-τ231 and Aβ1–42/Aβ1–40 [277,278,279,280,281,282]. In CSF and/or plasma, p-τ111, p-τ153, p-τ208, and p-τ231 appear to be more strongly associated with amyloid, while other τ phosphorylation sites are more strongly associated with p-τ pathology, e.g., p-τ205 or with both to a comparable extent, e.g. p-τ181, and p-τ217 [192, 283].
When comparing p-τ plasma biomarkers, it is critically important to consider not only the measured species and employed platform, but also the assays that are used on the respective platforms. While plasma p-τ217 has repeatedly outperformed p-τ181, the Simoa and MSD assays used for p-τ181 quantification in these comparisons are typically based on the AT270 antibody, which cross-reacts with p-τ175 [273]. Since p-τ175 has not demonstrated AD-related changes in CSF [283], this cross-reactivity likely confounds performance of p-τ181. Alternatively, a p-τ181 Simoa immunoassay incorporating the more phospho-specific antibody ADx252 has demonstrated very comparable performances to p-τ217 in both CSF and plasma [268] with respect to detecting amyloid burden as well as predicting longitudinal amyloid accumulation in head-to-head comparisons (∆AUC = 0.007—0.045) [186, 284, 285]. While mass-spectrometric plasma p-τ217 assays did demonstrate higher performance than both p-τ217 and p-τ181 immunoassays, the difference was only minimal (∆AUC = 0.061 – 1.06, P < 0.027) [285], and immunoassays might therefore have higher clinical utility considering its higher throughput and ease of use. Fully-automated assays, such as p-τ217 and/or p-τ181 assays on Lumipulse and Elecsys platforms, which have recently become commercially available, will presumably further facilitate clinical implementation of blood-based biomarkers.
Advancements in blood-based biomarker diagnostics have contributed to a revision of the criteria for diagnosis and staging of AD in 2024 [212]. In addition to CSF and PET based biomarkers, blood-based levels of Aβ1–42/Aβ1–40, p-τ181, p-τ217 and p-τ231 have also been integrated as diagnostic biomarkers. Due to its limited diagnostic potential in blood, the use of total τ as a diagnostic AD biomarker is recommended in conjunction with Aβ1–42 in a hybrid ratio, rather than as a standalone biomarker [212, 226, 267].
Prognostic biomarkers
In addition to the well-established AD-specific diagnostic biomarkers, the revised criteria also incorporate prognostic biomarkers. Prognostic fluid biomarkers can be quantified in both blood and CSF [212]. In addition to τ-related AD-specific prognostic biomarkers, some reflect phenomena that are shared among several neurodegenerative disorders and are thus not specific for AD, but are predictive for future cognitive decline, pathogenic processes or dementia onset: Neuroinflammation-, neurodegeneration-related, and synaptic biomarkers. Estimates for the sensitivity, specificity, accessibility, and the potential for quantification of the most established and the most promising of these markers are depicted in Fig. 4 and included in Tab. 1. Given the information about the stage of the disease of a given patient provided by these markers, many of them may also be well suited for disease monitoring purposes under therapy.
τ-related prognostic biomarkers
In contrast to amyloid PET, τ PET has a high predictive value for subsequent cognitive decline over the next couple of years, both in the asymptomatic and the early symptomatic stage [211, 286,287,288].
The microtubule-binding region of τ (MTBR-τ243) is a non-phosphorylated τ biomarker that can be quantified by mass spectrometry. MTBR-τ243 is the main component of insoluble τ aggregates and is increased in CSF of AD patients, but not other tauopathies, in which they have shown stronger associations with τ PET than p-τ species [289, 290]. Whereas p-τ species like p-τ181, p-τ217 and p-τ231 have shown the highest rate of change prior to τ-PET positivity, MTBR-τ243 increases more rapidly in τ-PET positive individuals, indicating it might provide additional value as a prognostic or staging marker in more advanced disease stages [290, 291]. Alternatively, the phosphorylated τ biomarker p-τ205 in CSF is also associated with τ PET in amyloid-positive individuals and increases earlier than MTBR-τ243, but later than the diagnostic p-τ181 and p-τ217 biomarkers and might thus also provide prognostic and staging information [290, 292]. To which extent these τ-related prognostic CSF biomarkers can be translated to blood remains to be investigated. Another novel approach is the use of antibodies specific to brain-derived τ to detect it in blood [293, 294].
Neuroinflammation-related biomarkers
Glial fibrillary acidic protein (GFAP), a marker of astrocytes and astrogliosis, is one of the “neuroinflammation-related markers” that has limited diagnostic utility for AD but has demonstrated prognostic potential. GFAP levels in both serum and plasma predict dementia onset as well as cognitive decline and grey matter loss in asymptomatic individuals [295,296,297,298]. Although not AD-specific, GFAP has demonstrated more pronounced elevations in plasma of AD patients than in other dementia types such as frontotemporal dementia (FTD), DLB or vascular dementia [299, 300]. Pereira et al. [301] reported that plasma GFAP levels associate with amyloid-PET, but not τ PET in non-demented individuals. Neuropathology studies, however, revealed that ante-mortem blood-based GFAP levels associate with both amyloid plaques and NFTs at autopsy [272, 302] which can be expected as both Aβ plaques and NFTs are associated with activated astrocytes [303, 304]. Moreover, when plaques and NFTs are included simultaneously in the statistical model, GFAP only associates with NFTs [192]. Remarkably, unlike other biomarkers, the magnitude of these blood-based GFAP increases is higher than those in CSF and blood-based GFAP has demonstrated higher performance to detect AD [305].
Soluble triggering receptor expression on myeloid cells 2 (sTREM2), a microglial marker, and Chitinase-3-like protein 1 (YKL-40), a marker of macrophage activity, as well as chitinase-enzyme activity are increased in AD patients, from preclinical stages, when measured in CSF, but not in plasma [306,307,308,309,310,311].
Another neuroinflammation-related biomarker is interleukin 18 (IL18) which is increased in the blood of patients with AD, ALS, FTD, or small vessel disease [312,313,314,315,316]. To what extent IL18 can serve as a biomarker for AD as suggested or more as an unspecific marker for neurodegeneration and small vessel disease-related neuroinflammation requires further research. Specific neuroinflammation-related pathways, such as pyroptosis, are not yet in the focus of the neuroinflammatory biomarkers for AD.
In addition to fluid biomarkers, TSPO PET as well as more novel PET tracers [317] have been used to trace neuroinflammation in the brain of patients with neurodegenerative disease including AD but this falls outside of the scope of this review [183,184,185].
Neurodegeneration-related biomarkers
Neurofilament light chain (NfL) is a cytoskeletal component of neurons that is essential for the growth and stability of axons and synapses [318]. Blood-based NfL shows a strong correlation with NfL in CSF [319] and is considered a general marker of neurodegeneration as its levels are elevated in a variety of neurodegenerative disorders [320, 321] considering its increased release from degenerating neurons in the form of extracellular vesicles [322]. In contrast to GFAP, the magnitude of plasma and CSF NfL elevations is not higher in AD than in other dementia types and does not associate with amyloid or p-τ pathology at autopsy [192, 234, 250,251,252, 269, 300, 302]. Similar to GFAP, blood-based NfL levels in asymptomatic individuals are predictive for subtle cognitive decline and onset of dementia due to AD or other causes [295, 297, 323, 324]. One important caveat is that the sensitivity of NfL for not only cognitive disorders but also numerous other neurological disorders or co-pathologies, might complicate interpretation of its prognostic and monitoring value in clinical settings where comorbidities are prevalent [325,326,327]. However, for the purpose of disease monitoring, reduced NfL levels may sufficiently indicate reduction of the degenerative process under therapy.
Synaptic biomarkers
In AD, but also other neurodegenerative disorders such as DLB and progressive supranuclear palsy, synaptic loss occurs early and precedes neurodegeneration, thereby marking one of the earliest pathological changes [328, 329]. This early involvement, combined with the strong correlation between synaptic density and cognitive decline, makes synaptic proteins particularly promising candidates as prognostic or even diagnostic markers in the early stages of the disease [330, 331]. CSF levels of several synaptic proteins have shown elevations in AD patients compared to controls [332, 333] and/or non-AD neurodegenerative disorders [333,334,335,336,337,338]. Some synaptic proteins have also shown AD biomarker potential in blood [339,340,341,342]. PET tracers targeting the synaptic vesicle protein 2A (SV2A) have been used for research purposes and showed decreased binding in several neurodegenerative disorders, including AD (reviewed in Carson et al. [343]). In neuropathological studies, the SV2A PET tracer UCB-J was shown to target SV2A with high specificity, and SV2A levels showed moderate to strong associations with the levels of the synaptic marker synaptophysin across diverse brain regions [344, 345]. Altogether this supports the utility of SV2A PET as a surrogate marker for synaptic loss in neurodegenerative disorders.
Retinal imaging
Multiple retinal imaging methods are currently explored for their potential to estimate brain AD pathology [346, 347]. Most of these methods aim to either determine degeneration of the retina as morphological parameters for degeneration of central nervous tissue, such as OCT [346,347,348,349,350,351,352], or focus on the detection of amyloid plaques or pathological protein accumulation in the retina by fluorescence imaging after dye application or hyperspectral imaging [353,354,355]. Recently, also p-τ pathology came in the focus of retinal imaging approaches [356]. With the current knowledge about the retinal manifestation of AD pathology, the detection of Aβ plaques appears to be a later event in the retina. Detection of neurodegeneration or even p-τ pathology in the retina would allow diagnosis of AD p-τ pathology at an earlier point in time. However, the first tauopathic manifestation in the retina appears to be a precursor lesion called PReT which has also been reported in young individuals without any evidence of AD. PReT has also been reported in the context of neuroinflammatory lesions in the retina as well as in cases with glaucoma [78, 79]. Given the convincing evidence of αSyn pathology and TDP-43 pathology in the retina, [116, 117, 138, 139] further research into a specific distinction between p-τ, αSyn, and TDP-43 pathologies in the retina could lead to a better diagnosis of co-pathologies and, by doing so, to a better stratification of AD allowing the development of personalized treatment strategies depending on the spectrum of co-pathologies.
Biomarkers for co-pathologies
Most AD patients are only given a single clinical diagnosis throughout life, but at autopsy, the majority demonstrates several co-pathologies [19, 23, 120, 357]. As mentioned earlier, αSyn aggregations are often present in patients with concomitant ADNC [19, 23, 357]. As a consequence of the common occurrence of Lewy body pathology in AD patients and vice versa AD pathology in DLB cases, DLB patients often demonstrate blood-based changes in AD-typical biomarkers like Aβ1–42/Aβ1–40, p-τ217 and p-τ181 [358]. However, neuropathological as well as imaging-based studies demonstrated that these AD biomarker changes in patients with mixed pathology are related to the underlying ADNC rather than αSyn pathology [234, 240, 250]. Given the possible influence of αSyn pathology on disease progression in AD patients [136], in vivo measures of αSyn co-pathology, when available, may provide an explanation for some of the inter-individual variability in the disease course or the treatment effects in clinical trials in AD.
In vivo measures of αSyn pathology may also be extremely useful in the diagnosis of synucleinopathies (e.g., DLB, Parkinson’s disease, multiple system’s atrophy (MSA)) and for more efficient drug trials for these disorders. αSyn PET tracers have been tested in first-in-human trials and show promise although the diagnostic accuracy may differ depending on the specific disease. One study revealed a high sensitivity in MSA with lower diagnostic performance in other synucleinopathies [359]. Very recently the αSyn PET ligand (18FC05-05) was presented to determine a specific signal in cases with Lewy body disease, i.e., Parkinson’s disease and DLB [360]. These reports need to be confirmed and may provide impetus for a novel field of research. Additionally, αSyn seed amplification assays (SAAs; also known as αSyn RT-QuIC test) detecting misfolded αSyn aggregates in CSF [361,362,363,364,365] and skin biopsies [366,367,368] have been validated. These SAAs leverage the intrinsic self-propagating nature of misfolded αSyn for its detection. This process involves addition of C-terminal truncated αSyn monomers to a patient sample. If misfolded αSyn seeds are present, the substrate is converted to a misfolded form, promoting aggregation through β-sheet formation [369]. Mechanical disruption of these aggregates causes fragmentation, generating new seeds for further polymerization. Repeated cycles of elongation and fragmentation lead to exponential amplification of the aggregates. The fluorescent dye thioflavin T binds β-sheets, enabling detection. In a direct comparison between ante-mortem and post-mortem CSF αSyn SAA, sensitivity and specificity to detect limbic or neocortical αSyn pathology in vivo was above 90%. However, amygdala predominant Lewy body pathology, which is common in AD, was detected with a sensitivity of only 14%, probably reflecting differences in biochemical characteristics of the different Lewy body subtypes [365, 370]. As a more accessible alternative, blood-based αSyn assays have recently been developed. Thereby extracellular vesicles (EVs) have been targeted, which are membrane-derived particles that are released into biological fluids and mediate intercellular communication or clearance of toxins [371]. In plasma of Parkinson’s disease (PD) patients, misfolded αSyn has been detected in such EVs, particularly in neuron-derived EVs [372]. Alternatively, addition of an immunoprecipitation (IP) step to RT-QuIC (IP/RT-QuIC) has allowed detection of even the smallest amounts of aggregated αSyn in native blood rather than in EVs. These blood-based αSyn levels – as determined by the IP/RT-QuIC method – are higher in PD, LBD and MSA patients compared to healthy controls and/or AD patients [373].
Other common co-pathologies like TDP-43 cytoplasmic inclusions [24, 374,375,376] also have a synergistic effect on cognitive impairment in AD. Development of pTDP-43 pathology-specific biomarkers would allow a more complete and pathology-based approach in clinical practice (precision-medicine) as well as subject stratification in therapeutic trials. The initial first-in-human studies of TDP-43 PET are currently starting. Preliminary evidence indicates high affinity and selectivity of TDP-43 PET tracers for FTLD-TDP pathology [377]. Hippocampal volume loss on structural MRI in older adults may also be partially explained by TDP43 pathology beyond what τ PET can explain [378]. This variance in hippocampal volume that cannot be explained by τ PET load is sometimes referred to as the “volume-uptake mismatch”. However many factors contribute to hippocampal volume beyond τ or TDP-43 aggregation.
TDP-43 fluid biomarkers are also still in an early development stage. Initial studies using ELISAs developed for determining TDP-43 [379] demonstrated elevated levels of total TDP-43 and pTDP-43 in CSF of patients with TDP-43 proteinopathies, including ALS, FTD and AD [380,381,382]. In blood, this assay had insufficient sensitivity to detect TDP-43 in the majority of patients, yet also demonstrated elevated TDP-43 or pTDP-43 levels in FTD, AD and/or amyotrohic lateral sclerosis (ALS) patients with detectable plasma levels [113, 379, 383]. In contrast, more recent blood-based studies using commercial assays on the ELISA or Simoa platform show decreased TDP-43 and pTDP-43 level in TDP-43 proteinopathies [384,385,386]. These discrepancies might be attributed to differences in assay sensitivities and TDP-43 contributions from peripheral tissues. Emerging approaches, such as detection of TDP-43 related cryptic exon neoepitopes [387,388,389] or quantification of TDP-43 in neuron-derived EVs [390, 391] show promise for more specific and early biomarkers. Recently, a Simoa assay of TDP-43 in small EVs (sEVs) from plasma combined with mass spectrometry measures of 3R/4R τ ratio in sEVs showed high sensitivity and specificity for distinguishing between TDP-43 proteinopathy and tauopathies (FTLD-TDP-43 and ALS versus FTLD-tau and progressive supranuclear palsy) [390]. Cryptic exon- and sEV-based assays may also hold promise for detection of TDP-43 and other co-pathologies in AD but that remains to be investigated. Of note, no interaction effect of TDP-43 pathology on plasma or CSF biomarkers reflecting AD pathology has been found in patients with mixed pathologies [234, 250, 392, 393].
The biomarker black box
As reported, the currently available biomarkers do not allow the diagnosis of very early stages of AD pathology, such as Aβ phases 1 or 2 (Fig. 3b), the non-argyrophilic stages of p-τ pathology [38] and even the Braak NFT stages I and II. This indicates a black box of cases with low AD pathology that biomarkers currently do not detect. This has important implications on pathogenetic conclusions drawn from biomarker studies. Biomarker studies showed that Aβ levels changed first, whereas p-τ levels followed [394]. Figure 3a illustrates that this fits well with the development of AD pathology outside the biomarker black box. Within the biomarker black box, however, it becomes evident that p-τ pathology prevails at very low levels, even in cases lacking Aβ pathology, confirming the interpretation of reports on the prevalence of the respective neuropathological changes [23, 38, 395]. When carefully comparing neuropathologically defined symptomatic and asymptomatic AD cases with non-AD controls from a previously published collection of cases [23], it becomes evident that especially in the earliest stages of Aβ pathology, p-τ emerges first, showing already an increase in non-AD cases without Aβ plaques (= Aβ phase 0) (Fig. 3).
Biomarkers in a clinical context
Given the updated clinical criteria for the diagnosis of AD, the determination of biological parameters with biomarkers is recommended [212]. At least one marker covering amyloid pathology and one covering p-τ pathology are suggested to be used for determining the biological diagnosis of AD [212]. Whether imaging, CSF, or blood-based markers of amyloid and p-τ will be used to reach this goal is left to the clinician [212]. The availability as well as cost aspects will, in this context, play a role for the individual decision. Based on these guidelines, the positivity for amyloid is already sufficient to diagnose AD [212]. Accordingly, one can discuss whether p-τ will add any additional diagnostic information. However, since p-τ markers may provide added value for determining the stage of the disease, this information may aid both differential diagnosis and disease prognosis [396,397,398].
In the clinic, a multimodal approach may be appropriate, e.g., based on a stepwise sequence of tests. Depending on the context of use, either specificity or sensitivity could be set at 90 or 95%, leading to a dual cut-off. With a dual cut-off a variable proportion of cases will have intermediate levels that do not allow strong diagnostic conclusions. In these selected cases subsequent amyloid or τ PET scanning may be useful to reach a higher degree of certainty. From a patient perspective, the dual cut-off approach with subsequent PET imaging in intermediate cases is attractive as it avoids excessive investigations in most cases and, at the same time, significantly reduces the rate of false-positive or false-negative diagnosis that may be seen when the investigation is limited to the fluid biomarker test [378]. If PET is not an option, patients with intermediate results will need to be informed about the degree of diagnostic uncertainty and a wait-and-see approach may be appropriate. Table 1 gives an overview about the current biomarkers and their use as diagnostic and prognostic/monitoring biomarkers. While fluid biomarkers are also in use for disease monitoring in the context of clinical trials, none of these markers have currently been validated for implementation in a clinical context.
An important ongoing effort relates to the establishment of a diagnostic interpretation algorithm that provides a standardized way of interpreting biomarkers for diagnosis of AD in its symptomatic and asymptomatic phase, which is commutable with respect to the different methods used across different laboratories. The Centiloid scale for amyloid imaging [399] is a highly successful example of such harmonization. A similar initiative is ongoing with the CenTauR scale for τ PET, however, this is hampered by the much higher heterogeneity between τ PET tracers than is the case for amyloid PET [400]. For biofluid markers, the commercialization of automated platform assays with universal thresholds as part of the regulatory approval package will also significantly enhance their diagnostic value on a very short term [401]. Artificial intelligence (AI)/machine learning may also help us to identify reproducible thresholds for diagnosis or disease staging. Especially for imaging techniques these approaches were already used in the past [197, 402,403,404,405]. AI approaches are also pursued to analyze complex biomarker datasets, e.g., for proteomics of CSF or blood samples or for analyzing genetic endophenotypes for predicting neuropathological diagnosis [401]. To conclude, fluid and PET biomarkers can significantly augment diagnostic accuracy in the clinic which is essential for disclosure of diagnosis and prognosis and the development of drug and nondrug management plans.
A critical perspective upon diagnostic and prognostic use of fluid or imaging biomarkers is the participant’s or patient’s view. Paradoxically, as a diagnosis of AD relies more and more on technical investigations, shared decision making has become more and more important so that the individual is informed about the possible outcomes of the test prior to testing and the options to choose from, including whether to perform a specific biomarker test or not. Shared decision making prior to testing is also advantageous at the time of test result disclosure as it helps the individual to prepare for the possible outcomes and avoids that information is disclosed which the individual would have preferred not to know.
Biomarkers for use in clinical trials and personalized treatment
For the use of biomarkers in clinical trials as well as for personalized treatment, it is essential to clarify the purpose of use. In clinical therapeutic trials in AD, biomarkers have been mainly used for (1) selection of the study target population and (2) for monitoring of target engagement and disease course. Given the disease heterogeneity of AD, co-pathologies as well as distinct subtypes may deserve more attention in the future as these may affect the therapeutic response. It is important to keep in mind that biomarkers do not identify individuals with initial ADNC. Since even such “subthreshold” initial ADNC are capable of inducing seeding, heterogeneity of the control group cannot be excluded. For example, lysates from brains with initial Aβ pathology (Aβ phase 1) can induce Aβ plaque seeding and propagation in mouse models as well as maturation of Aβ towards the additional accumulation of post-translationally modified forms of Aβ [406]. Likewise, Braak NFT-stage I brain lysates induced seeding in vitro [407] and after subretinal injection in the retina of τ-transgenic mice [81] whereas propagation into the brain was not observed in this study, except for AD brain lysates from symptomatic individuals [81]. The importance of subthreshold amounts of Aβ has been further demonstrated in APP knockout mice who received AD brain seed injections [408]. Although these mice did not develop Aβ deposits, when their brain lysates were injected into seeding competent APP transgenic mice, accelerated seeding of Aβ pathology was reported to be induced by persisting seeds [408].
For the identification of AD subtypes related to the regional pattern of neurodegeneration [58] τ-PET appears to be best suited as it reflects the density of tauopathic lesions in the different areas of the brain [409, 410]. However, atrophy related approaches using MRI for atrophy mapping have also been reported, often using AI-based methods for distinction [410, 411].
Given the high frequency of comorbidities in the AD brain [19, 23, 137, 357] it is likely that co-pathologies impact the disease course and treatment effects in clinical trials. Co-pathologies should, therefore, be considered in the stratification step and, during study follow-up. AD cases with αSyn-pathology could be identified by, e.g. an RT-QuIC test, although the sensitivity was not sufficient to detect LBD properly in demented individuals [363, 364]. Hence, more sensitive biomarkers for αSyn aggregates are required. A high frequency of pTDP-43 pathology (67%) in cases with the limbic-predominant type of AD has been reported [376]. For pTDP-43 pathology detection, an EV-based assay to estimate CNS TDP-43 pathology in the blood had been developed for FTLD-TDP and ALS and may also have a potential to estimate LATE-NC [390], which remains to be determined. Moreover, for αSyn and pTDP-43 biomarkers, the boundaries of the clinical black box still need to be delineated against neuropathological standards. Even for clinical practice, proper detection of co-pathologies that offer additional treatment opportunities makes sense when aiming for personalized treatment approaches. CAA is another co-pathology that becomes clinically evident by cerebral hemorrhages and microbleeds [187, 188]. In cases with at least microbleeds, CAA can be observed by MRI [180, 188, 412]. However, the largest number of AD cases with CAA co-pathology escape clinical detection as vascular Aβ deposition, which is characteristic for CAA, cannot be distinguished from amyloid plaques by amyloid-PET because the presence of plaques already causes tracer uptake [395]. The presence of an APOE ε4 allele is a very strong risk factor for capillary CAA (CAA type 1) and could be used to identify AD patients at risk for capillary CAA [103, 105]. Given the low sensitivity/specificity for detecting CAA in patients, better biomarkers for CAA would be important. However, 80% to 100% of the symptomatic AD patients showed CAA of any type at autopsy [69,70,71, 413], i.e. the biomarker-based diagnosis of symptomatic AD in combination with APOE genotypting may already allow for a guess that CAA will also be prevalent.
Biomarkers are also used in clinical trials and for personalized medicine for monitoring treated individuals. The first Aβ targeting drugs (lecanemab and donanemab) successfully lowered amyloid levels in the brain, reached the clinical outcome criteria [5, 6, 414,415,416], and have been approved by the FDA, and several other regulatory authorities worldwide. In addition to documenting target engagement (e.g., lowering of Aβ or p-τ), it is also necessary to document a disease modifying effect of a given drug with respect to neurodegeneration and neuroinflammation. To do so, we need biomarkers, which recapitulate the progression of amyloid and τ pathology in longitudinal studies, and others, which document the progress of the neurodegenerative process [198, 199, 250, 260, 301, 417,418,419,420]. In longitudinal studies, p-τ biomarkers documented the course of the pathological hallmarks well and especially τ PET provides information of the progressive destruction of the brain by p-τ pathology and potential sites of interaction with Aβ [198, 211, 262, 270, 276, 421,422,423]. The degenerative process can also be documented in a nonspecific manner, e.g., by means of synaptic biomarkers [250, 324]. The PET imaging for synapse densities with the SV2A tracer appears to be well suited to provide information about the result of the degenerative process and to what extent it continues or stops [343, 424,425,426,427]. To estimate the contribution of neuroinflammatory changes on AD progression and its modification under therapy, plasma GFAP has been shown to be helpful [297, 299, 301, 324, 420].
Conclusions
Current biomarkers are well suited to determine Aβ and p-τ pathology in symptomatic AD cases and in a subset of non-demented individuals with ADNC whereas early stages of ADNC escape detection and represent a “biomarker black box”. Although the currently available biomarkers represent an enormous progress for the clinical diagnosis of AD, we still need to improve their sensitivity, since the degenerative process already leads to the loss of neurons in non-demented cases demonstrating ADNC amounts below the sensitivity threshold of currently established biomarkers. Moreover, we need markers allowing a reliable detection of co-pathologies as they enable us to distinguish different subtypes or subforms of AD to improve personalized treatment approaches and possibly better stratification of patients participating in clinical trials.
Given the “biomarker black box” for the earliest disease stages, pathogenetic conclusions about the disease onset require gold standard neuropathological confirmation whereas the further course of the disease is similarly reflected by biomarkers as in neuropathological studies and can be longitudinally studied with the current biomarkers. For disease monitoring under therapy, documentation of target engagement as well as disease progression with neurodegeneration and inflammation is needed. Accordingly, different biomarkers are required that reflect the dynamics of the degenerative and inflammatory process (e.g., p-τ-, GFAP-, NfL-, or synapse-related biomarkers) rather than highly sensitive markers that reached already the plateau-phase when the clinical symptoms become evident (e.g., Aβ-related biomarkers). Biomarkers that reflect novel disease targets, such as regulated cell death pathways (e.g., necroptosis), and/or the accompanied inflammatory reaction may need to be developed in the future for designing personalized AD therapy concepts.
Data availability
Data reanalyzed for this review are provided in the supplementary material or were published elsewhere as indicated by respective citations. Further information can be requested from the corresponding author on reasonable request.
References
Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A. 1985;82:4245–9.
Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, Wisniewski M. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem. 1986;261:6084–9.
Montine TJ, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Dickson DW, Duyckaerts C, Frosch MP, Masliah E, Mirra SS, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease: a practical approach. Acta Neuropathol. 2012;123:1–11.
Aisen P, Touchon J, Amariglio R, Andrieu S, Bateman R, Breitner J, Donohue M, Dunn B, Doody R, Fox N, et al. EU/US/CTAD task force: lessons learned from recent and current Alzheimer’s prevention trials. J Prev Alzheimers Dis. 2017;4:116–24.
van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, Kanekiyo M, Li D, Reyderman L, Cohen S, et al. Lecanemab in early Alzheimer’s disease. N Engl J Med. 2023;388:9–21.
Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, Wessels AM, Shcherbinin S, Wang H, Monkul Nery ES, et al. Donanemab in early symptomatic Alzheimer disease: the TRAILBLAZER-ALZ 2 randomized clinical Trial. JAMA. 2023;330:512–27.
Thal DR, Beach TG, Zanette M, Heurling K, Chakrabarty A, Ismail A, Smith AP, Buckley C. [18F]flutemetamol amyloid PET in preclinical and symptomatic Alzheimer’s disease: specific detection of advanced phases of Aβ pathology. Alzheimers Dement. 2015;11:975–85.
Sabri O, Sabbagh MN, Seibyl J, Barthel H, Akatsu H, Ouchi Y, Senda K, Murayama S, Ishii K, Takao M, et al. Florbetaben PET imaging to detect amyloid beta plaques in Alzheimer’s disease: phase 3 study. Alzheimers Dement. 2015;11:964–74.
Clark CM, Pontecorvo MJ, Beach TG, Bedell BJ, Coleman RE, Doraiswamy PM, Fleisher AS, Reiman EM, Sabbagh MN, Sadowsky CH, et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-beta plaques: a prospective cohort study. Lancet Neurol. 2012;11:669–78.
Pontecorvo MJ, Keene CD, Beach TG, Montine TJ, Arora AK, Devous MD Sr, Navitsky M, Kennedy I, Joshi AD, Lu M, et al. Comparison of regional flortaucipir PET with quantitative tau immunohistochemistry in three subjects with Alzheimer’s disease pathology: a clinicopathological study. EJNMMI Res. 2020;10:65.
Murray ME, Lowe VJ, Graff-Radford NR, Liesinger AM, Cannon A, Przybelski SA, Rawal B, Parisi JE, Petersen RC, Kantarci K, et al. Clinicopathologic and 11C-Pittsburgh compound B implications of Thal amyloid phase across the Alzheimer’s disease spectrum. Brain. 2015;138:1370–81.
Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol. 2010;6:131–44.
Sjogren M, Vanderstichele H, Agren H, Zachrisson O, Edsbagge M, Wikkelso C, Skoog I, Wallin A, Wahlund LO, Marcusson J, et al. Tau and Abeta42 in cerebrospinal fluid from healthy adults 21–93 years of age: establishment of reference values. Clin Chem. 2001;47:1776–81.
Janelidze S, Mattsson N, Palmqvist S, Smith R, Beach TG, Serrano GE, Chai X, Proctor NK, Eichenlaub U, Zetterberg H, et al. Plasma P-tau181 in Alzheimer’s disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat Med. 2020;26:379–86.
Mila-Aloma M, Ashton NJ, Shekari M, Salvado G, Ortiz-Romero P, Montoliu-Gaya L, Benedet AL, Karikari TK, Lantero-Rodriguez J, Vanmechelen E, et al. Plasma p-tau231 and p-tau217 as state markers of amyloid-beta pathology in preclinical Alzheimer’s disease. Nat Med. 2022;28:1797–801.
Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, Holtzman DM, Jagust W, Jessen F, Karlawish J, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–62.
Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR Jr, Kaye J, Montine TJ, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:280–92.
Dubois B, Villain N, Schneider L, Fox N, Campbell N, Galasko D, Kivipelto M, Jessen F, Hanseeuw B, Boada M, et al. Alzheimer disease as a clinical-biological construct-an International Working Group recommendation. JAMA Neurol. 2024;81:1304–11.
Robinson JL, Richardson H, Xie SX, Suh E, Van Deerlin VM, Alfaro B, Loh N, Porras-Paniagua M, Nirschl JJ, Wolk D, et al. The development and convergence of co-pathologies in Alzheimer’s disease. Brain. 2021;144:953–62.
Josephs KA, Murray ME, Whitwell JL, Tosakulwong N, Weigand SD, Petrucelli L, Liesinger AM, Petersen RC, Parisi JE, Dickson DW. Updated TDP-43 in Alzheimer’s disease staging scheme. Acta Neuropathol. 2016;131:571–85.
Amador-Ortiz C, Lin WL, Ahmed Z, Personett D, Davies P, Duara R, Graff-Radford NR, Hutton ML, Dickson DW. TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol. 2007;61:435–45.
Nelson PT, Brayne C, Flanagan ME, Abner EL, Agrawal S, Attems J, Castellani RJ, Corrada MM, Cykowski MD, Di J, et al. Frequency of LATE neuropathologic change across the spectrum of Alzheimer’s disease neuropathology: combined data from 13 community-based or population-based autopsy cohorts. Acta Neuropathol. 2022;144:27–44.
Tome SO, Thal DR. Co-pathologies in Alzheimer’s disease: just multiple pathologies or partners in crime? Brain. 2021;144:706–8.
Uryu K, Nakashima-Yasuda H, Forman MS, Kwong LK, Clark CM, Grossman M, Miller BL, Kretzschmar HA, Lee VM, Trojanowski JQ, Neumann M. Concomitant TAR-DNA-binding protein 43 pathology is present in Alzheimer disease and corticobasal degeneration but not in other tauopathies. J Neuropathol Exp Neurol. 2008;67:555–64.
Toledo JB, Gopal P, Raible K, Irwin DJ, Brettschneider J, Sedor S, Waits K, Boluda S, Grossman M, Van Deerlin VM, et al. Pathological alpha-synuclein distribution in subjects with coincident Alzheimer’s and Lewy body pathology. Acta Neuropathol. 2016;131:393–409.
Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy body disease. Acta Neuropathol. 2008;115:427–36.
Jellinger KA, Attems J. Incidence of cerebrovascular lesions in Alzheimer’s disease: a postmortem study. Acta Neuropathol. 2003;105:14–7.
Thal DR, Grinberg LT, Attems J. Vascular dementia: different forms of vessel disorders contribute to the development of dementia in the elderly brain. Exp Gerontol. 2012;47:816–24.
Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, et al. Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2011;42:2672–713.
Toledo JB, Arnold SE, Raible K, Brettschneider J, Xie SX, Grossman M, Monsell SE, Kukull WA, Trojanowski JQ. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s Coordinating Centre. Brain. 2013;136:2697–706.
Alzheimer A. Ueber eine eigenartige Erkrankung der Hirnrinde. Allg Zschr Psych. 1907;64:146–8.
Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A. 1986;83:4913–7.
Jarrett JT, Berger EP, Lansbury PT Jr. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease. Biochemistry. 1993;32:4693–7.
Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43). Neuron. 1994;13:45–53.
Glenner GG, Wong CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 1984;120:885–90.
Bancher C, Brunner C, Lassmann H, Budka H, Jellinger K, Wiche G, Seitelberger F, Grundke-Iqbal I, Iqbal K, Wisniewski HM. Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer’s disease. Brain Res. 1989;477:90–9.
Aragao Gomes L, Uytterhoeven V, Lopez-Sanmartin D, Tome SO, Tousseyn T, Vandenberghe R, Vandenbulcke M, von Arnim CAF, Verstreken P, Thal DR. Maturation of neuronal AD-tau pathology involves site-specific phosphorylation of cytoplasmic and synaptic tau preceding conformational change and fibril formation. Acta Neuropathol. 2021;141:173–92.
Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol. 2011;70:960–9.
Ohm TG, Muller H, Braak H, Bohl J. Close-meshed prevalence rates of different stages as a tool to uncover the rate of Alzheimer’s disease-related neurofibrillary changes. Neuroscience. 1995;64:209–17.
Attems J, Thomas A, Jellinger K. Correlations between cortical and subcortical tau pathology. Neuropathol Appl Neurobiol. 2012;38:582–90.
Rub U, Del Tredici K, Schultz C, Thal DR, Braak E, Braak H. The evolution of Alzheimer’s disease-related cytoskeletal pathology in the human raphe nuclei. Neuropathol Appl Neurobiol. 2000;26:553–67.
Sassin I, Schultz C, Thal DR, Rub U, Arai K, Braak E, Braak H. Evolution of Alzheimer’s disease-related cytoskeletal changes in the basal nucleus of Meynert. Acta Neuropathol (Berl). 2000;100:259–69.
Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82:239–59.
Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC, Dickson DW, Duyckaerts C, Frosch MP, Masliah E, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement. 2012;8:1–13.
Schwarz AJ, Yu P, Miller BB, Shcherbinin S, Dickson J, Navitsky M, Joshi AD, Devous MD Sr, Mintun MS. Regional profiles of the candidate tau PET ligand 18F-AV-1451 recapitulate key features of Braak histopathological stages. Brain. 2016;139:1539–50.
Thal DR, Tome SO. The central role of tau in Alzheimer’s disease: from neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull. 2022;190:204–17.
Dickson DW. The pathogenesis of senile plaques. J Neuropathol Exp Neurol. 1997;56:321–39.
Griffin WS, Sheng JG, Roberts GW, Mrak RE. Interleukin-1 expression in different plaque types in Alzheimer’s disease: significance in plaque evolution. J Neuropathol Exp Neurol. 1995;54:276–81.
Thal DR, Rub U, Schultz C, Sassin I, Ghebremedhin E, Del Tredici K, Braak E, Braak H. Sequence of Abeta-protein deposition in the human medial temporal lobe. J Neuropathol Exp Neurol. 2000;59:733–48.
Wisniewski HM, Sadowski M, Jakubowska-Sadowska K, Tarnawski M, Wegiel J. Diffuse, lake-like amyloid-beta deposits in the parvopyramidal layer of the presubiculum in Alzheimer disease. J Neuropathol Exp Neurol. 1998;57:674–83.
Wisniewski HM, Terry RD. Reexamination of the pathogenesis of the senile plaque. In: Zimmerman HM, editor. Progress in neuropathology 2. New York: Grane & Stratton; 1973. p. 1–26.
Thal DR, Sassin I, Schultz C, Haass C, Braak E, Braak H. Fleecy amyloid deposits in the internal layers of the human entorhinal cortex are comprised of N-terminal truncated fragments of Abeta. J Neuropathol Exp Neurol. 1999;58:210–6.
Thal DR, Capetillo-Zarate E, Del Tredici K, Braak H. The development of amyloid beta protein deposits in the aged brain. Sci Aging Knowledge Environ. 2006;2006:re1.
Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, Vogel FS, Hughes JP, van Belle G, Berg L. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991;41:479–86.
Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791–800.
Thal DR, Beach TG, Zanette M, Lilja J, Heurling K, Chakrabarty A, Ismail A, Farrar G, Buckley C, Smith APL. Estimation of amyloid distribution by [(18)F]flutemetamol PET predicts the neuropathological phase of amyloid beta-protein deposition. Acta Neuropathol. 2018;136:557–67.
Galton CJ, Patterson K, Xuereb JH, Hodges JR. Atypical and typical presentations of Alzheimer’s disease: a clinical, neuropsychological, neuroimaging and pathological study of 13 cases. Brain. 2000;123(Pt 3):484–98.
Murray ME, Graff-Radford NR, Ross OA, Petersen RC, Duara R, Dickson DW. Neuropathologically defined subtypes of Alzheimer’s disease with distinct clinical characteristics: a retrospective study. Lancet Neurol. 2011;10:785–96.
Crook R, Ellis R, Shanks M, Thal LJ, Perez-Tur J, Baker M, Hutton M, Haltia T, Hardy J, Galasko D. Early-onset Alzheimer’s disease with a presenilin-1 mutation at the site corresponding to the Volga German presenilin-2 mutation. Ann Neurol. 1997;42:124–8.
Crook R, Verkkoniemi A, Perez-Tur J, Mehta N, Baker M, Houlden H, Farrer M, Hutton M, Lincoln S, Hardy J, et al. A variant of Alzheimer’s disease with spastic paraparesis and unusual plaques due to deletion of exon 9 of presenilin 1. Nat Med. 1998;4:452–5.
Murrell J, Farlow M, Ghetti B, Benson MD. A mutation in the amyloid precursor protein associated with hereditary Alzheimer’s disease. Science. 1991;254:97–9.
Reed LA, Grabowski TJ, Schmidt ML, Morris JC, Goate A, Solodkin A, Van Hoesen GW, Schelper RL, Talbot CJ, Wragg MA, Trojanowski JQ. Autosomal dominant dementia with widespread neurofibrillary tangles. Ann Neurol. 1997;42:564–72.
Daniels AJ, McDade E, Llibre-Guerra JJ, Xiong C, Perrin RJ, Ibanez L, Supnet-Bell C, Cruchaga C, Goate A, Renton AE, et al. 15 years of longitudinal genetic, clinical, cognitive, imaging, and biochemical measures in DIAN. medRxiv. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2024.08.08.24311689.
Ringman JM, Monsell S, Ng DW, Zhou Y, Nguyen A, Coppola G, Van Berlo V, Mendez MF, Tung S, Weintraub S, et al. Neuropathology of autosomal dominant Alzheimer disease in the national Alzheimer coordinating center database. J Neuropathol Exp Neurol. 2016;75:284–90.
Boon BDC, Bulk M, Jonker AJ, Morrema THJ, van den Berg E, Popovic M, Walter J, Kumar S, van der Lee SJ, Holstege H, et al. The coarse-grained plaque: a divergent Abeta plaque-type in early-onset Alzheimer’s disease. Acta Neuropathol. 2020;140:811–30.
Lemere CA, Blusztajn JK, Yamaguchi H, Wisniewski T, Saido TC, Selkoe DJ. Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol Dis. 1996;3:16–32.
Fortea J, Zaman SH, Hartley S, Rafii MS, Head E, Carmona-Iragui M. Alzheimer’s disease associated with Down syndrome: a genetic form of dementia. Lancet Neurol. 2021;20:930–42.
Wilcock DM, Hurban J, Helman AM, Sudduth TL, McCarty KL, Beckett TL, Ferrell JC, Murphy MP, Abner EL, Schmitt FA, Head E. Down syndrome individuals with Alzheimer’s disease have a distinct neuroinflammatory phenotype compared to sporadic Alzheimer’s disease. Neurobiol Aging. 2015;36:2468–74.
Joachim CL, Morris JH, Selkoe DJ. Clinically diagnosed Alzheimer’s disease: autopsy results in 150 cases. Ann Neurol. 1988;24:50–6.
Vinters HV, Gilbert JJ. Cerebral amyloid angiopathy: incidence and complications in the aging brain. II. The distribution of amyloid vascular changes. Stroke. 1983;14:924–8.
Mandybur TI. The incidence of cerebral amyloid angiopathy in Alzheimer’s disease. Neurology. 1975;25:120–6.
Thal DR, Griffin WS, Braak H. Parenchymal and vascular Abeta-deposition and its effects on the degeneration of neurons and cognition in Alzheimer’s disease. J Cell Mol Med. 2008;12:1848–62.
Koronyo Y, Biggs D, Barron E, Boyer DS, Pearlman JA, Au WJ, Kile SJ, Blanco A, Fuchs DT, Ashfaq A, et al. Retinal amyloid pathology and proof-of-concept imaging trial in Alzheimer’s disease. JCI Insight. 2017;2:e93621.
Koronyo-Hamaoui M, Koronyo Y, Ljubimov AV, Miller CA, Ko MK, Black KL, Schwartz M, Farkas DL. Identification of amyloid plaques in retinas from Alzheimer’s patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model. Neuroimage. 2011;54(Suppl 1):S204-217.
Shi H, Koronyo Y, Fuchs DT, Sheyn J, Jallow O, Mandalia K, Graham SL, Gupta VK, Mirzaei M, Kramerov AA, et al. Retinal arterial Abeta(40) deposition is linked with tight junction loss and cerebral amyloid angiopathy in MCI and AD patients. Alzheimers Dement. 2023;19:5185–97.
den Haan J, Morrema THJ, Verbraak FD, de Boer JF, Scheltens P, Rozemuller AJ, Bergen AAB, Bouwman FH, Hoozemans JJ. Amyloid-beta and phosphorylated tau in post-mortem Alzheimer’s disease retinas. Acta Neuropathol Commun. 2018;6:147.
Hart de Ruyter FJ, Morrema THJ, den Haan J, Netherlands Brain B, Twisk JWR, de Boer JF, Scheltens P, Boon BDC, Thal DR, Rozemuller AJ, et al. Phosphorylated tau in the retina correlates with tau pathology in the brain in Alzheimer’s disease and primary tauopathies. Acta Neuropathol. 2023;145:197–218.
Walkiewicz G, Ronisz A, Van Ginderdeuren R, Lemmens S, Bouwman FH, Hoozemans JJM, Morrema THJ, Rozemuller AJ, Hart de Ruyter FJ, De Groef L, et al. Primary retinal tauopathy: a tauopathy with a distinct molecular pattern. Alzheimers Dement. 2024;20:330–40.
Gupta N, Fong J, Ang LC, Yucel YH. Retinal tau pathology in human glaucomas. Can J Ophthalmol. 2008;43:53–60.
Hart de Ruyter FJ, Evers M, Morrema THJ, Dijkstra AA, den Haan J, Twisk JWR, de Boer JF, Scheltens P, Bouwman FH, Verbraak FD, et al. Neuropathological hallmarks in the post-mortem retina of neurodegenerative diseases. Acta Neuropathol. 2024;148:24.
Walkiewicz G, Ronisz A, Ospitalieri S, Tsaka G, Tomé SO, Vandenberghe R, von Arnim CAF, Rousseau F, Schymkowitz J, De Groef L, Thal DR. pTau pathology in the retina of TAU58 mice: association with ganglion cell degeneration and implications on seeding and propagation of pTau from human brain lysates. Acta Neuropathol Commun. 2024;12:194.
Qiu Y, Jin T, Mason E, Campbell MCW. Predicting thioflavin fluorescence of retinal amyloid deposits associated with Alzheimer’s disease from their polarimetric properties. Transl Vis Sci Technol. 2020;9: 47.
Simchowicz T. Histopathologische Studien über die senile Demenz. In: Nissl F, Alzheimer A, editors. Histologie und histopathologische Arbeiten über die Großhirnrinde, vol. 4. Jena: Fischer; 1911. p. 267–444.
Ball MJ. Neuronal loss, neurofibrillary tangles and granulovacuolar degeneration in the hippocampus with ageing and dementia. A quantitative study. Acta Neuropathol. 1977;37:111–8.
Thal DR, Del Tredici K, Ludolph AC, Hoozemans JJ, Rozemuller AJ, Braak H, Knippschild U. Stages of granulovacuolar degeneration: their relation to Alzheimer’s disease and chronic stress response. Acta Neuropathol. 2011;122:577–89.
Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21:383–421.
Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL 3rd, Araoz C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci U S A. 1989;86:7611–5.
McGeer PL, Akiyama H, Itagaki S, McGeer EG. Immune system response in Alzheimer’s disease. Can J Neurol Sci. 1989;16:516–27.
Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, et al. Neuroinflammation in Alzheimer disease. Nat Rev Immunol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-024-01104-7.
Fu C, Chute DJ, Farag ES, Garakian J, Cummings JL, Vinters HV. Comorbidity in dementia: an autopsy study. Arch Pathol Lab Med. 2004;128:32–8.
Vinters HV, Ellis WG, Zarow C, Zaias BW, Jagust WJ, Mack WJ, Chui HC. Neuropathologic substrates of ischemic vascular dementia. J Neuropathol Exp Neurol. 2000;59:931–45.
Launer LJ, Hughes TM, White LR. Microinfarcts, brain atrophy, and cognitive function: the Honolulu Asia aging study autopsy study. Ann Neurol. 2011;70:774–80.
White L, Petrovitch H, Hardman J, Nelson J, Davis DG, Ross GW, Masaki K, Launer L, Markesbery WR. Cerebrovascular pathology and dementia in autopsied Honolulu-Asia aging study participants. Ann N Y Acad Sci. 2002;977:9–23.
Grinberg LT, Thal DR. Vascular pathology in the aged human brain. Acta Neuropathol. 2010;119:277–90.
Fazio C. Red softening of the brain. J Neuropathol Exp Neurol. 1949;8:43–60.
Fisher CM. Lacunes: small, deep cerebral infarcts. Neurology. 1965;15:774–84.
Liberato B, Chong JY, Sacco RL. Focal brain ischemia. Clinical features, epidemiology, risk factors and outcome. In: Kalimo H, editor. Cerebrovascular diseases. Basel: ISN Neuropath Press; 2005. p. 176–185. Pathology & Genetics.
Lammie GA. Hypertensive cerebral small vessel disease and stroke. Brain Pathol. 2002;12:358–70.
Mandybur TI. Cerebral amyloid angiopathy: the vascular pathology and complications. J Neuropathol Exp Neurol. 1986;45:79–90.
Soontornniyomkij V, Lynch MD, Mermash S, Pomakian J, Badkoobehi H, Clare R, Vinters HV. Cerebral microinfarcts associated with severe cerebral beta-amyloid angiopathy. Brain Pathol. 2009;20:459–67.
Calhoun ME, Burgermeister P, Phinney AL, Stalder M, Tolnay M, Wiederhold KH, Abramowski D, Sturchler-Pierrat C, Sommer B, Staufenbiel M, Jucker M. Neuronal overexpression of mutant amyloid precursor protein results in prominent deposition of cerebrovascular amyloid. Proc Natl Acad Sci U S A. 1999;96:14088–93.
Thal DR, Papassotiropoulos A, Saido TC, Griffin WS, Mrak RE, Kolsch H, Del Tredici K, Attems J, Ghebremedhin E. Capillary cerebral amyloid angiopathy identifies a distinct APOE epsilon4-associated subtype of sporadic Alzheimer’s disease. Acta Neuropathol. 2010;120:169–83.
Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, et al. The probabilistic model of Alzheimer disease: the amyloid hypothesis revised. Nat Rev Neurosci. 2022;23:53–66.
Thal DR, Ghebremedhin E, Rüb U, Yamaguchi H, Del Tredici K, Braak H. Two types of sporadic cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2002;61:282–93.
Thal DR, Capetillo-Zarate E, Larionov S, Staufenbiel M, Zurbruegg S, Beckmann N. Capillary cerebral amyloid angiopathy is associated with vessel occlusion and cerebral blood flow disturbances. Neurobiol Aging. 2009;30:1936–48.
Hecht M, Kramer LM, von Arnim CAF, Otto M, Thal DR. Capillary cerebral amyloid angiopathy in Alzheimer’s disease: association with allocortical/hippocampal microinfarcts and cognitive decline. Acta Neuropathol. 2018;135:681–94.
Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, Rademakers R, Alafuzoff I, Attems J, Brayne C, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142:1503–27.
Tome SO, Gomes LA, Li X, Vandenberghe R, Tousseyn T, Thal DR. TDP-43 interacts with pathological tau protein in Alzheimer’s disease. Acta Neuropathol. 2021;141:795–9.
Josephs KA, Murray ME, Whitwell JL, Parisi JE, Petrucelli L, Jack CR, Petersen RC, Dickson DW. Staging TDP-43 pathology in Alzheimer’s disease. Acta Neuropathol. 2014;127:441–50.
Josephs KA, Dickson DW, Tosakulwong N, Weigand SD, Murray ME, Petrucelli L, Liesinger AM, Senjem ML, Spychalla AJ, Knopman DS, et al. Rates of hippocampal atrophy and presence of post-mortem TDP-43 in patients with Alzheimer’s disease: a longitudinal retrospective study. Lancet Neurol. 2017;16:917–24.
James BD, Wilson RS, Boyle PA, Trojanowski JQ, Bennett DA, Schneider JA. TDP-43 stage, mixed pathologies, and clinical Alzheimer’s-type dementia. Brain. 2016;139:2983–93.
Foulds PG, Davidson Y, Mishra M, Hobson DJ, Humphreys KM, Taylor M, Johnson N, Weintraub S, Akiyama H, Arai T, et al. Plasma phosphorylated-TDP-43 protein levels correlate with brain pathology in frontotemporal lobar degeneration. Acta Neuropathol. 2009;118:647–58.
McAleese KE, Walker L, Erskine D, Thomas AJ, McKeith IG, Attems J. TDP-43 pathology in Alzheimer’s disease, dementia with Lewy bodies and ageing. Brain Pathol. 2017;27:472–9.
Nelson PT, Lee EB, Cykowski MD, Alafuzoff I, Arfanakis K, Attems J, Brayne C, Corrada MM, Dugger BN, Flanagan ME, et al. LATE-NC staging in routine neuropathologic diagnosis: an update. Acta Neuropathol. 2023;145:159–73.
Dijkstra AA, Morrema THJ, Hart de Ruyter FJ, Gami-Patel P, Verbraak FD, de Boer JF, Bouwman FH, Pijnenburg YAL, den Haan J, Rozemuller AJ, Hoozemans JJM. TDP-43 pathology in the retina of patients with frontotemporal lobar degeneration. Acta Neuropathol. 2023;146:767–70.
Pediconi N, Gigante Y, Cama S, Pitea M, Mautone L, Ruocco G, Ghirga S, Di Angelantonio S. Retinal fingerprints of ALS in patients: Ganglion cell apoptosis and TDP-43/p62 misplacement. Front Aging Neurosci. 2023;15: 1110520.
Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.
Arai Y, Yamazaki M, Mori O, Muramatsu H, Asano G, Katayama Y. Alpha-synuclein-positive structures in cases with sporadic Alzheimer’s disease: morphology and its relationship to tau aggregation. Brain Res. 2001;888:287–96.
Hamilton RL. Lewy bodies in Alzheimer’s disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol. 2000;10:378–84.
Lippa CF, Fujiwara H, Mann DM, Giasson B, Baba M, Schmidt ML, Nee LE, O’Connell B, Pollen DA, St George-Hyslop P, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153:1365–70.
Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol. 2006;65:685–97.
Chatterjee A, Hirsch-Reinshagen V, Moussavi SA, Ducharme B, Mackenzie IR, Hsiung GR. Clinico-pathological comparison of patients with autopsy-confirmed Alzheimer’s disease, dementia with Lewy bodies, and mixed pathology. Alzheimers Dement (Amst). 2021;13:e12189.
Brenowitz WD, Keene CD, Hawes SE, Hubbard RA, Longstreth WT Jr, Woltjer RL, Crane PK, Larson EB, Kukull WA. Alzheimer’s disease neuropathologic change, Lewy body disease, and vascular brain injury in clinic- and community-based samples. Neurobiol Aging. 2017;53:83–92.
Irwin DJ, Grossman M, Weintraub D, Hurtig HI, Duda JE, Xie SX, Lee EB, Van Deerlin VM, Lopez OL, Kofler JK, et al. Neuropathological and genetic correlates of survival and dementia onset in synucleinopathies: a retrospective analysis. Lancet Neurol. 2017;16:55–65.
Kraybill ML, Larson EB, Tsuang DW, Teri L, McCormick WC, Bowen JD, Kukull WA, Leverenz JB, Cherrier MM. Cognitive differences in dementia patients with autopsy-verified AD, Lewy body pathology, or both. Neurology. 2005;64:2069–73.
Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.
Alafuzoff I, Ince PG, Arzberger T, Al-Sarraj S, Bell J, Bodi I, Bogdanovic N, Bugiani O, Ferrer I, Gelpi E, et al. Staging/typing of Lewy body related alpha-synuclein pathology: a study of the BrainNet Europe Consortium. Acta Neuropathol. 2009;117:635–52.
Attems J, Toledo JB, Walker L, Gelpi E, Gentleman S, Halliday G, Hortobagyi T, Jellinger K, Kovacs GG, Lee EB, et al. Neuropathological consensus criteria for the evaluation of Lewy pathology in post-mortem brains: a multi-centre study. Acta Neuropathol. 2021;141:159–72.
Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, Sasse J, Boyer S, Shirohi S, Brooks R, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–34.
Leverenz JB, Hamilton R, Tsuang DW, Schantz A, Vavrek D, Larson EB, Kukull WA, Lopez O, Galasko D, Masliah E, et al. Empiric refinement of the pathologic assessment of Lewy-related pathology in the dementia patient. Brain Pathol. 2008;18:220–4.
Jellinger KA. Lewy body-related alpha-synucleinopathy in the aged human brain. J Neural Transm (Vienna). 2004;111:1219–35.
Ulusoy A, Rusconi R, Perez-Revuelta BI, Musgrove RE, Helwig M, Winzen-Reichert B, Di Monte DA. Caudo-rostral brain spreading of alpha-synuclein through vagal connections. EMBO Mol Med. 2013;5:1119–27.
McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, Aarsland D, Galvin J, Attems J, Ballard CG, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89:88–100.
Irwin DJ, White MT, Toledo JB, Xie SX, Robinson JL, Van Deerlin V, Lee VM, Leverenz JB, Montine TJ, Duda JE, et al. Neuropathologic substrates of Parkinson disease dementia. Ann Neurol. 2012;72:587–98.
Gawor K, Tome SO, Vandenberghe R, Van Damme P, Vandenbulcke M, Otto M, von Arnim CAF, Ghebremedhin E, Ronisz A, Ospitalieri S, et al. Amygdala-predominant alpha-synuclein pathology is associated with exacerbated hippocampal neuron loss in Alzheimer’s disease. Brain Commun. 2024;6:fcae442.
Spina S, La Joie R, Petersen C, Nolan AL, Cuevas D, Cosme C, Hepker M, Hwang JH, Miller ZA, Huang EJ, et al. Comorbid neuropathological diagnoses in early versus late-onset Alzheimer’s disease. Brain. 2021;144:2186–98.
Beach TG, Carew J, Serrano G, Adler CH, Shill HA, Sue LI, Sabbagh MN, Akiyama H, Cuenca N, Arizona Parkinson’s Disease C. Phosphorylated alpha-synuclein-immunoreactive retinal neuronal elements in Parkinson’s disease subjects. Neurosci Lett. 2014;571:34–8.
Hart de Ruyter FJ, Morrema THJ, den Haan J, Gase G, Twisk JWR, de Boer JF, Scheltens P, Bouwman FH, Verbraak FD, Rozemuller AJM, Hoozemans JJM. alpha-Synuclein pathology in post-mortem retina and optic nerve is specific for alpha-synucleinopathies. NPJ Parkinsons Dis. 2023;9:124.
Veys L, Vandenabeele M, Ortuno-Lizaran I, Baekelandt V, Cuenca N, Moons L, De Groef L. Retinal alpha-synuclein deposits in Parkinson’s disease patients and animal models. Acta Neuropathol. 2019;137:379–95.
Ball MJ, Lo P. Granulovacuolar degeneration in the ageing brain and in dementia. J Neuropathol Exp Neurol. 1977;36:474–87.
Hirano A, Dembitzer HM, Kurland LT, Zimmerman HM. The fine structure of some intraganglionic alterations. Neurofibrillary tangles, granulovacuolar bodies and “rod-like” structures as seen in Guam amyotrophic lateral sclerosis and parkinsonism-dementia complex. J Neuropathol Exp Neurol. 1968;27:167–82.
Kohler C. Granulovacuolar degeneration: a neurodegenerative change that accompanies tau pathology. Acta Neuropathol. 2016;132:339–59.
Funk KE, Mrak RE, Kuret J. Granulovacuolar degeneration bodies of Alzheimer’s disease resemble late-stage autophagic organelles. Neuropathol Appl Neurobiol. 2011;37:295–306.
Koper MJ, Van Schoor E, Ospitalieri S, Vandenberghe R, Vandenbulcke M, von Arnim CAF, Tousseyn T, Balusu S, De Strooper B, Thal DR. Necrosome complex detected in granulovacuolar degeneration is associated with neuronal loss in Alzheimer’s disease. Acta Neuropathol. 2020;139:463–84.
Jayaraman A, Htike TT, James R, Picon C, Reynolds R. TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer’s disease hippocampus. Acta Neuropathol Commun. 2021;9:159.
Riku Y, Duyckaerts C, Boluda S, Plu I, Le Ber I, Millecamps S, Salachas F, Brainbank Neuro CEBNN, Yoshida M, Ando T, et al. Increased prevalence of granulovacuolar degeneration in C9orf72 mutation. Acta Neuropathol. 2019;138:783–93.
McGeer PL, Akiyama H, Itagaki S, McGeer EG. Activation of the classical complement pathway in brain tissue of Alzheimer patients. Neurosci Lett. 1989;107:341–6.
McGeer PL, Itagaki S, Tago H, McGeer EG. Reactive microglia in patients with senile dementia of the Alzheimer type are positive for the histocompatibility glycoprotein HLA-DR. Neurosci Lett. 1987;79:195–200.
Strauss S, Bauer J, Ganter U, Jonas U, Berger M, Volk B. Detection of interleukin-6 and alpha 2-macroglobulin immunoreactivity in cortex and hippocampus of Alzheimer’s disease patients. Lab Invest. 1992;66:223–30.
Thal DR, Arendt T, Waldmann G, Holzer M, Zedlick D, Rüb U, Schober R. Progression of neurofibrillary changes and PHF-tau in end-stage Alzheimer’s disease is different from plaque and cortical microglial pathology. Neurobiol Aging. 1998;19:517–25.
Mancuso R, Fattorelli N, Martinez-Muriana A, Davis E, Wolfs L, Van Den Daele J, Geric I, Premereur J, Polanco P, Bijnens B, et al. Xenografted human microglia display diverse transcriptomic states in response to Alzheimer’s disease-related amyloid-beta pathology. Nat Neurosci. 2024;27:886–900.
Hu Y, Fryatt GL, Ghorbani M, Obst J, Menassa DA, Martin-Estebane M, Muntslag TAO, Olmos-Alonso A, Guerrero-Carrasco M, Thomas D, et al. Replicative senescence dictates the emergence of disease-associated microglia and contributes to Abeta pathology. Cell Rep. 2021;35: 109228.
Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169:1276-1290 e1217.
Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674–8.
Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, Schwartz S, Albasset S, McManus RM, Tejera D, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–73.
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer’s disease. Acta Neuropathol. 2024;148:31.
Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29:347–64.
Bergsbaken T, Cookson BT. Macrophage activation redirects yersinia-infected host cell death from apoptosis to caspase-1-dependent pyroptosis. PLoS Pathog. 2007;3: e161.
Moonen S, Koper MJ, Van Schoor E, Schaeverbeke JM, Vandenberghe R, von Arnim CAF, Tousseyn T, De Strooper B, Thal DR. Pyroptosis in Alzheimer’s disease: cell type-specific activation in microglia, astrocytes and neurons. Acta Neuropathol. 2023;145:175–95.
Thal DR, Gawor K, Moonen S. Regulated cell death and its role in Alzheimer’s disease and amyotrophic lateral sclerosis. Acta Neuropathol. 2024;147:69.
Koper MJ, Tome SO, Gawor K, Belet A, Van Schoor E, Schaeverbeke J, Vandenberghe R, Vandenbulcke M, Ghebremedhin E, Otto M, et al. LATE-NC aggravates GVD-mediated necroptosis in Alzheimer’s disease. Acta Neuropathol Commun. 2022;10:128.
Braak H, Zetterberg H, Del Tredici K, Blennow K. Intraneuronal tau aggregation precedes diffuse plaque deposition, but amyloid-beta changes occur before increases of tau in cerebrospinal fluid. Acta Neuropathol. 2013;126:631–41.
Duyckaerts C, Braak H, Brion JP, Buee L, Del Tredici K, Goedert M, Halliday G, Neumann M, Spillantini MG, Tolnay M, Uchihara T. PART is part of Alzheimer disease. Acta Neuropathol. 2015;129:749–56.
Price JL, Davis PB, Morris JC, White DL. The distribution of tangles, plaques and related immunohistochemical markers in healthy aging and Alzheimer’s disease. Neurobiol Aging. 1991;12:295–312.
Spires-Jones TL, Attems J, Thal DR. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol. 2017;134:187–205.
Gotz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293:1491–5.
Lewis J, Dickson DW, Lin WL, Chisholm L, Corral A, Jones G, Yen SH, Sahara N, Skipper L, Yager D, et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science. 2001;293:1487–91.
Gomes LA, Hipp SA, Rijal Upadhaya A, Balakrishnan K, Ospitalieri S, Koper MJ, Largo-Barrientos P, Uytterhoeven V, Reichwald J, Rabe S, et al. Abeta-induced acceleration of Alzheimer-related tau-pathology spreading and its association with prion protein. Acta Neuropathol. 2019;138:913–41.
Kadamangudi S, Marcatti M, Zhang WR, Fracassi A, Kayed R, Limon A, Taglialatela G. Amyloid-beta oligomers increase the binding and internalization of tau oligomers in human synapses. Acta Neuropathol. 2024;149:2.
Corbett GT, Wang Z, Hong W, Colom-Cadena M, Rose J, Liao M, Asfaw A, Hall TC, Ding L, DeSousa A, et al. PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol. 2020;139:503–26.
Wiersma VI, van Ziel AM, Vazquez-Sanchez S, Nolle A, Berenjeno-Correa E, Bonaterra-Pastra A, Clavaguera F, Tolnay M, Musters RJP, van Weering JRT, et al. Granulovacuolar degeneration bodies are neuron-selective lysosomal structures induced by intracellular tau pathology. Acta Neuropathol. 2019;138:943–70.
Kohler C, Dinekov M, Gotz J. Granulovacuolar degeneration and unfolded protein response in mouse models of tauopathy and Abeta amyloidosis. Neurobiol Dis. 2014;71:169–79.
Balusu S, Horre K, Thrupp N, Craessaerts K, Snellinx A, Serneels L, T’Syen D, Chrysidou I, Arranz AM, Sierksma A, et al. MEG3 activates necroptosis in human neuron xenografts modeling Alzheimer’s disease. Science. 2023;381:1176–82.
Koper MJ, Moonen S, Ronisz A, Ospitalieri S, Callaerts-Vegh Z, T’Syen D, Rabe S, Staufenbiel M, De Strooper B, Balusu S, Thal DR. Inhibition of an Alzheimer’s disease-associated form of necroptosis rescues neuronal death in mouse models. Sci Transl Med. 2024;16:eadf5128.
Tome SO, Tsaka G, Ronisz A, Ospitalieri S, Gawor K, Gomes LA, Otto M, von Arnim CAF, Van Damme P, Van Den Bosch L, et al. TDP-43 pathology is associated with increased tau burdens and seeding. Mol Neurodegener. 2023;18:71.
Latimer CS, Liachko NF. Tau and TDP-43 synergy: a novel therapeutic target for sporadic late-onset Alzheimer’s disease. Geroscience. 2021;43:1627–34.
Latimer CS, Stair JG, Hincks JC, Currey HN, Bird TD, Keene CD, Kraemer BC, Liachko NF. TDP-43 promotes tau accumulation and selective neurotoxicity in bigenic Caenorhabditis elegans. Dis Model Mech. 2022;15:dmm049323.
van Veluw SJ, Charidimou A, van der Kouwe AJ, Lauer A, Reijmer YD, Costantino I, Gurol ME, Biessels GJ, Frosch MP, Viswanathan A, Greenberg SM. Microbleed and microinfarct detection in amyloid angiopathy: a high-resolution MRI-histopathology study. Brain. 2016;139:3151–62.
Greenberg SM, Vernooij MW, Cordonnier C, Viswanathan A, Al-Shahi Salman R, Warach S, Launer LJ, Van Buchem MA, Breteler MM. Cerebral microbleeds: a guide to detection and interpretation. Lancet Neurol. 2009;8:165–74.
Roman GC, Tatemichi TK, Erkinjuntti T, Cummings JL, Masdeu JC, Garcia JH, Amaducci L, Orgogozo JM, Brun A, Hofman A, et al. Vascular dementia: diagnostic criteria for research studies. Report of the NINDS-AIREN International Workshop. Neurology. 1993;43:250–60.
Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL, Black SE, Powers WJ, DeCarli C, Merino JG, Kalaria RN, et al. National Institute of Neurological Disorders and Stroke-Canadian stroke network vascular cognitive impairment harmonization standards. Stroke. 2006;37:2220–41.
Chauveau F, Winkeler A, Chalon S, Boutin H, Becker G. PET imaging of neuroinflammation: any credible alternatives to TSPO yet? Mol Psychiatry. 2025;30:213–28.
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: a transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun. 2023;113:415–31.
Van Weehaeghe D, Van Schoor E, De Vocht J, Koole M, Attili B, Celen S, Declercq L, Thal DR, Van Damme P, Bormans G, Van Laere K. TSPO versus P2X7 as a target for neuroinflammation: an in vitro and in vivo study. J Nucl Med. 2020;61:604–7.
De Meyer S, Schaeverbeke JM, Luckett ES, Reinartz M, Blujdea ER, Cleynen I, Dupont P, Van Laere K, Vanbrabant J, Stoops E, et al. Plasma pTau181 and pTau217 predict asymptomatic amyloid accumulation equally well as amyloid PET. Brain Commun. 2024;6:fcae162.
Thal DR, Gawor K. Cerebral amyloid angiopathy: neuropathological diagnosis, link to Alzheimer’s disease and impact on clinics. Clin Neuropathol. 2023;42:176–89.
Charidimou A, Boulouis G, Frosch MP, Baron JC, Pasi M, Albucher JF, Banerjee G, Barbato C, Bonneville F, Brandner S, et al. The Boston criteria version 2.0 for cerebral amyloid angiopathy: a multicentre, retrospective, MRI-neuropathology diagnostic accuracy study. Lancet Neurol. 2022;21:714–25.
Gleason CE, Zuelsdorff M, Gooding DC, Kind AJH, Johnson AL, James TT, Lambrou NH, Wyman MF, Ketchum FB, Gee A, et al. Alzheimer’s disease biomarkers in Black and non-Hispanic White cohorts: a contextualized review of the evidence. Alzheimers Dement. 2022;18:1545–64.
Morris JC, Schindler SE, McCue LM, Moulder KL, Benzinger TLS, Cruchaga C, Fagan AM, Grant E, Gordon BA, Holtzman DM, Xiong C. Assessment of racial disparities in biomarkers for Alzheimer disease. JAMA Neurol. 2019;76:264–73.
Curtis C, Gamez JE, Singh U, Sadowsky CH, Villena T, Sabbagh MN, Beach TG, Duara R, Fleisher AS, Frey KA, et al. Phase 3 trial of flutemetamol labeled with radioactive fluorine 18 imaging and neuritic plaque density. JAMA Neurol. 2015;72:287–94.
Salvado G, Ossenkoppele R, Ashton NJ, Beach TG, Serrano GE, Reiman EM, Zetterberg H, Mattsson-Carlgren N, Janelidze S, Blennow K, Hansson O. Specific associations between plasma biomarkers and postmortem amyloid plaque and tau tangle loads. EMBO Mol Med. 2023;15: e17123.
Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergstrom M, Savitcheva I, Huang GF, Estrada S, et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 2004;55:306–19.
Mathis CA, Wang Y, Holt DP, Huang GF, Debnath ML, Klunk WE. Synthesis and evaluation of 11C-labeled 6-substituted 2-arylbenzothiazoles as amyloid imaging agents. J Med Chem. 2003;46:2740–54.
Vandenberghe R, Van Laere K, Ivanoiu A, Salmon E, Bastin C, Triau E, Hasselbalch S, Law I, Andersen A, Korner A, et al. 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann Neurol. 2010;68:319–29.
La Joie R, Ayakta N, Seeley WW, Borys E, Boxer AL, DeCarli C, Dore V, Grinberg LT, Huang E, Hwang JH, et al. Multisite study of the relationships between antemortem [(11)C]PIB-PET Centiloid values and postmortem measures of Alzheimer’s disease neuropathology. Alzheimers Dement. 2019;15:205–16.
Reinartz M, Luckett ES, Schaeverbeke J, De Meyer S, Adamczuk K, Thal DR, Van Laere K, Dupont P, Vandenberghe R. Classification of (18)F-Flutemetamol scans in cognitively normal older adults using machine learning trained with neuropathology as ground truth. Eur J Nucl Med Mol Imaging. 2022;49:3772–86.
Hanseeuw BJ, Betensky RA, Jacobs HIL, Schultz AP, Sepulcre J, Becker JA, Cosio DMO, Farrell M, Quiroz YT, Mormino EC, et al. Association of amyloid and tau with cognition in preclinical Alzheimer disease: a longitudinal study. JAMA Neurol. 2019;76:915–24.
Hanseeuw BJ, Betensky RA, Mormino EC, Schultz AP, Sepulcre J, Becker JA, Jacobs HIL, Buckley RF, LaPoint MR, Vannini P, et al. PET staging of amyloidosis using striatum. Alzheimers Dement. 2018;14:1281–92.
Luckett ES, Schaeverbeke J, De Meyer S, Adamczuk K, Van Laere K, Dupont P, Vandenberghe R. Longitudinal changes in (18)F-Flutemetamol amyloid load in cognitively intact APOE4 carriers versus noncarriers: methodological considerations. Neuroimage Clin. 2023;37:103321.
Su Y, Flores S, Wang G, Hornbeck RC, Speidel B, Joseph-Mathurin N, Vlassenko AG, Gordon BA, Koeppe RA, Klunk WE, et al. Comparison of Pittsburgh compound B and florbetapir in cross-sectional and longitudinal studies. Alzheimers Dement (Amst). 2019;11:180–90.
Villemagne VL, Pike KE, Chetelat G, Ellis KA, Mulligan RS, Bourgeat P, Ackermann U, Jones G, Szoeke C, Salvado O, et al. Longitudinal assessment of Abeta and cognition in aging and Alzheimer disease. Ann Neurol. 2011;69:181–92.
Grothe MJ, Barthel H, Sepulcre J, Dyrba M, Sabri O, Teipel SJ, Alzheimer’s Disease Neuroimaging I. In vivo staging of regional amyloid deposition. Neurology. 2017;89:2031–8.
Collij LE, Salvado G, Wottschel V, Mastenbroek SE, Schoenmakers P, Heeman F, Aksman L, Wink AM, Berckel BNM, van de Flier WM, et al. Spatial-temporal patterns of beta-amyloid accumulation: a subtype and stage inference model analysis. Neurology. 2022;98:e1692–703.
Johnson KA, Minoshima S, Bohnen NI, Donohoe KJ, Foster NL, Herscovitch P, Karlawish JH, Rowe CC, Carrillo MC, Hartley DM, et al. Appropriate use criteria for amyloid PET: a report of the Amyloid Imaging Task Force, the Society of Nuclear Medicine and Molecular Imaging, and the Alzheimer’s Association. Alzheimers Dement. 2013;9:e-1-16.
Leuzy A, Chiotis K, Lemoine L, Gillberg PG, Almkvist O, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging in neurodegenerative tauopathies-still a challenge. Mol Psychiatry. 2019;24:1112–34.
Declercq L, Rombouts F, Koole M, Fierens K, Marien J, Langlois X, Andres JI, Schmidt M, Macdonald G, Moechars D, et al. Preclinical evaluation of (18)F-JNJ64349311, a novel PET tracer for tau imaging. J Nucl Med. 2017;58:975–81.
Gogola A, Minhas DS, Villemagne VL, Cohen AD, Mountz JM, Pascoal TA, Laymon CM, Mason NS, Ikonomovic MD, Mathis CA, et al. Direct comparison of the tau PET tracers (18)F-Flortaucipir and (18)F-MK-6240 in human subjects. J Nucl Med. 2022;63:108–16.
Fleisher AS, Pontecorvo MJ, Devous MD Sr, Lu M, Arora AK, Truocchio SP, Aldea P, Flitter M, Locascio T, Devine M, et al. Positron emission tomography imaging with [18F]flortaucipir and postmortem assessment of Alzheimer disease neuropathologic changes. JAMA Neurol. 2020;77:829-39.
Lowe VJ, Lundt ES, Albertson SM, Min HK, Fang P, Przybelski SA, Senjem ML, Schwarz CG, Kantarci K, Boeve B, et al. Tau-positron emission tomography correlates with neuropathology findings. Alzheimers Dement. 2020;16:561–71.
Ossenkoppele R, Pichet Binette A, Groot C, Smith R, Strandberg O, Palmqvist S, Stomrud E, Tideman P, Ohlsson T, Jogi J, et al. Amyloid and tau PET-positive cognitively unimpaired individuals are at high risk for future cognitive decline. Nat Med. 2022;28:2381–7.
Jack CR Jr, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, et al. Revised criteria for diagnosis and staging of Alzheimer’s disease: Alzheimer’s Association Workgroup. Alzheimers Dement. 2024;20:5143–69.
Ossenkoppele R, Schonhaut DR, Scholl M, Lockhart SN, Ayakta N, Baker SL, O’Neil JP, Janabi M, Lazaris A, Cantwell A, et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain. 2016;139:1551–67.
Vogel JW, Young AL, Oxtoby NP, Smith R, Ossenkoppele R, Strandberg OT, La Joie R, Aksman LM, Grothe MJ, Iturria-Medina Y, et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat Med. 2021;27:871–81.
Moloney CM, Lowe VJ, Murray ME. Visualization of neurofibrillary tangle maturity in Alzheimer’s disease: a clinicopathologic perspective for biomarker research. Alzheimers Dement. 2021;17:1554–74.
Schaeverbeke J, Celen S, Cornelis J, Ronisz A, Serdons K, Van Laere K, Thal DR, Tousseyn T, Bormans G, Vandenberghe R. Binding of [(18)F]AV1451 in post mortem brain slices of semantic variant primary progressive aphasia patients. Eur J Nucl Med Mol Imaging. 2020;47:1949–60.
Aliaga A, Therriault J, Quispialaya KM, Aliaga A, Hopewell R, Rahmouni N, Macedo AC, Kunach P, Soucy JP, Massarweh G, et al. Comparison between brain and cerebellar autoradiography using [(18)F]Flortaucipir, [(18)F]MK6240, and [(18)F]PI2620 in postmortem human brain tissue. J Nucl Med. 2025;66:123–9.
Lowe VJ, Curran G, Fang P, Liesinger AM, Josephs KA, Parisi JE, Kantarci K, Boeve BF, Pandey MK, Bruinsma T, et al. An autoradiographic evaluation of AV-1451 Tau PET in dementia. Acta Neuropathol Commun. 2016;4:58.
Lemoine L, Saint-Aubert L, Marutle A, Antoni G, Eriksson JP, Ghetti B, Okamura N, Nennesmo I, Gillberg PG, Nordberg A. Visualization of regional tau deposits using (3)H-THK5117 in Alzheimer brain tissue. Acta Neuropathol Commun. 2015;3:40.
Ono M, Sahara N, Kumata K, Ji B, Ni R, Koga S, Dickson DW, Trojanowski JQ, Lee VM, Yoshida M, et al. Distinct binding of PET ligands PBB3 and AV-1451 to tau fibril strains in neurodegenerative tauopathies. Brain. 2017;140:764–80.
Marquie M, Normandin MD, Meltzer AC, Siao Tick Chong M, Andrea NV, Anton-Fernandez A, Klunk WE, Mathis CA, Ikonomovic MD, Debnath M, et al. Pathological correlations of [F-18]-AV-1451 imaging in non-Alzheimer tauopathies. Ann Neurol. 2017;81:117–28.
Wren MC, Lashley T, Arstad E, Sander K. Large inter- and intra-case variability of first generation tau PET ligand binding in neurodegenerative dementias. Acta Neuropathol Commun. 2018;6:34.
Shuping JL, Matthews DC, Adamczuk K, Scott D, Rowe CC, Kreisl WC, Johnson SC, Lukic AS, Johnson KA, Rosa-Neto P, et al. Development, initial validation, and application of a visual read method for [(18)F]MK-6240 tau PET. Alzheimers Dement (N Y). 2023;9: e12372.
Vos SJB, Delvenne A, Jack CR Jr, Thal DR, Visser PJ. The clinical importance of suspected non-Alzheimer disease pathophysiology. Nat Rev Neurol. 2024;20:337–46.
McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, 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;7:263–9.
Clark CM, Xie S, Chittams J, Ewbank D, Peskind E, Galasko D, Morris JC, McKeel DW Jr, Farlow M, Weitlauf SL, et al. Cerebrospinal fluid tau and beta-amyloid: how well do these biomarkers reflect autopsy-confirmed dementia diagnoses? Arch Neurol. 2003;60:1696–702.
Shaw LM, Vanderstichele H, Knapik-Czajka M, Clark CM, Aisen PS, Petersen RC, Blennow K, Soares H, Simon A, Lewczuk P, et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol. 2009;65:403–13.
Banerjee G, Ambler G, Keshavan A, Paterson RW, Foiani MS, Toombs J, Heslegrave A, Dickson JC, Fraioli F, Groves AM, et al. Cerebrospinal fluid biomarkers in cerebral amyloid angiopathy. J Alzheimers Dis. 2020;74:1189–201.
Irwin DJ, McMillan CT, Toledo JB, Arnold SE, Shaw LM, Wang LS, Van Deerlin V, Lee VM, Trojanowski JQ, Grossman M. Comparison of cerebrospinal fluid levels of tau and Abeta 1–42 in Alzheimer disease and frontotemporal degeneration using 2 analytical platforms. Arch Neurol. 2012;69:1018–25.
Ewers M, Mattsson N, Minthon L, Molinuevo JL, Antonell A, Popp J, Jessen F, Herukka SK, Soininen H, Maetzler W, et al. CSF biomarkers for the differential diagnosis of Alzheimer’s disease: a large-scale international multicenter study. Alzheimers Dement. 2015;11:1306–15.
Kapaki EN, Paraskevas GP, Tzerakis NG, Sfagos C, Seretis A, Kararizou E, Vassilopoulos D. Cerebrospinal fluid tau, phospho-tau181 and beta-amyloid1-42 in idiopathic normal pressure hydrocephalus: a discrimination from Alzheimer’s disease. Eur J Neurol. 2007;14:168–73.
Jeppsson A, Zetterberg H, Blennow K, Wikkelso C. Idiopathic normal-pressure hydrocephalus: pathophysiology and diagnosis by CSF biomarkers. Neurology. 2013;80:1385–92.
Janelidze S, Zetterberg H, Mattsson N, Palmqvist S, Vanderstichele H, Lindberg O, van Westen D, Stomrud E, Minthon L, Blennow K, et al. CSF Abeta42/Abeta40 and Abeta42/Abeta38 ratios: better diagnostic markers of Alzheimer disease. Ann Clin Transl Neurol. 2016;3:154–65.
Grothe MJ, Moscoso A, Ashton NJ, Karikari TK, Lantero-Rodriguez J, Snellman A, Zetterberg H, Blennow K, Scholl M. Alzheimer’s Disease Neuroimaging I: Associations of fully automated CSF and novel plasma biomarkers with Alzheimer disease neuropathology at autopsy. Neurology. 2021;97:e1229–42.
Kurihara M, Matsubara T, Morimoto S, Arakawa A, Ohse K, Kanemaru K, Iwata A, Murayama S, Saito Y. Neuropathological changes associated with aberrant cerebrospinal fluid p-tau181 and Abeta42 in Alzheimer’s disease and other neurodegenerative diseases. Acta Neuropathol Commun. 2024;12:48.
Baiardi S, Abu-Rumeileh S, Rossi M, Zenesini C, Bartoletti-Stella A, Polischi B, Capellari S, Parchi P. Antemortem CSF Abeta42/Abeta40 ratio predicts Alzheimer’s disease pathology better than Abeta42 in rapidly progressive dementias. Ann Clin Transl Neurol. 2019;6:263–73.
Lewczuk P, Matzen A, Blennow K, Parnetti L, Molinuevo JL, Eusebi P, Kornhuber J, Morris JC, Fagan AM. Cerebrospinal fluid Abeta42/40 corresponds better than Abeta42 to amyloid PET in Alzheimer’s disease. J Alzheimers Dis. 2017;55:813–22.
Shoji M, Matsubara E, Kanai M, Watanabe M, Nakamura T, Tomidokoro Y, Shizuka M, Wakabayashi K, Igeta Y, Ikeda Y, et al. Combination assay of CSF tau, A beta 1–40 and A beta 1–42(43) as a biochemical marker of Alzheimer’s disease. J Neurol Sci. 1998;158:134–40.
Pannee J, Portelius E, Minthon L, Gobom J, Andreasson U, Zetterberg H, Hansson O, Blennow K. Reference measurement procedure for CSF amyloid beta (Abeta)(1–42) and the CSF Abeta(1–42) /Abeta(1–40) ratio - a cross-validation study against amyloid PET. J Neurochem. 2016;139:651–8.
Tapiola T, Alafuzoff I, Herukka SK, Parkkinen L, Hartikainen P, Soininen H, Pirttila T. Cerebrospinal fluid beta-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Arch Neurol. 2009;66:382–9.
Luo J, Agboola F, Grant E, Masters CL, Albert MS, Johnson SC, McDade EM, Voglein J, Fagan AM, Benzinger T, et al. Sequence of Alzheimer disease biomarker changes in cognitively normal adults: a cross-sectional study. Neurology. 2020;95:e3104–16.
Palmqvist S, Mattsson N, Hansson O. Alzheimer’s Disease Neuroimaging I: Cerebrospinal fluid analysis detects cerebral amyloid-beta accumulation earlier than positron emission tomography. Brain. 2016;139:1226–36.
Fagan AM, Mintun MA, Shah AR, Aldea P, Roe CM, Mach RH, Marcus D, Morris JC, Holtzman DM. Cerebrospinal fluid tau and ptau(181) increase with cortical amyloid deposition in cognitively normal individuals: implications for future clinical trials of Alzheimer’s disease. EMBO Mol Med. 2009;1:371–80.
Mattsson N, Insel PS, Donohue M, Landau S, Jagust WJ, Shaw LM, Trojanowski JQ, Zetterberg H, Blennow K, Weiner MW, Alzheimer’s Disease Neuroimaging I. Independent information from cerebrospinal fluid amyloid-beta and florbetapir imaging in Alzheimer’s disease. Brain. 2015;138:772–83.
Reiber H. Dynamics of brain-derived proteins in cerebrospinal fluid. Clin Chim Acta. 2001;310:173–86.
Xin SH, Tan L, Cao X, Yu JT, Tan L. Clearance of amyloid beta and tau in Alzheimer’s disease: from mechanisms to therapy. Neurotox Res. 2018;34:733–48.
De Meyer S, Blujdea ER, Schaeverbeke JM, Adamczuk K, Vandenberghe R, Poesen K, Teunissen CE. Serum biomarkers as prognostic markers for Alzheimer’s disease in a clinical setting. Alzheimers Dement (Amst). 2025;17:e70071.
Roher AE, Esh CL, Kokjohn TA, Castano EM, Van Vickle GD, Kalback WM, Patton RL, Luehrs DC, Daugs ID, Kuo YM, et al. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 2009;5:18–29.
Blennow K, Zetterberg H. Understanding biomarkers of neurodegeneration: ultrasensitive detection techniques pave the way for mechanistic understanding. Nat Med. 2015;21:217–9.
Smirnov DS, Ashton NJ, Blennow K, Zetterberg H, Simren J, Lantero-Rodriguez J, Karikari TK, Hiniker A, Rissman RA, Salmon DP, Galasko D. Plasma biomarkers for Alzheimer’s disease in relation to neuropathology and cognitive change. Acta Neuropathol. 2022;143:487–503.
Bermudez C, Graff-Radford J, Syrjanen JA, Stricker NH, Algeciras-Schimnich A, Kouri N, Kremers WK, Petersen RC, Jack CR Jr, Knopman DS, et al. Plasma biomarkers for prediction of Alzheimer’s disease neuropathologic change. Acta Neuropathol. 2023;146:13–29.
Mattsson-Carlgren N, Grinberg LT, Boxer A, Ossenkoppele R, Jonsson M, Seeley W, Ehrenberg A, Spina S, Janelidze S, Rojas-Martinex J, et al. Cerebrospinal fluid biomarkers in autopsy-confirmed Alzheimer disease and frontotemporal lobar degeneration. Neurology. 2022;98:e1137–50.
Janelidze S, Teunissen CE, Zetterberg H, Allue JA, Sarasa L, Eichenlaub U, Bittner T, Ovod V, Verberk IMW, Toba K, et al. Head-to-head comparison of 8 plasma amyloid-beta 42/40 assays in Alzheimer disease. JAMA Neurol. 2021;78:1375–82.
De Meyer S, Schaeverbeke JM, Verberk IMW, Gille B, De Schaepdryver M, Luckett ES, Gabel S, Bruffaerts R, Mauroo K, Thijssen EH, et al. Comparison of ELISA- and SIMOA-based quantification of plasma Abeta ratios for early detection of cerebral amyloidosis. Alzheimers Res Ther. 2020;12:162.
Ovod V, Ramsey KN, Mawuenyega KG, Bollinger JG, Hicks T, Schneider T, Sullivan M, Paumier K, Holtzman DM, Morris JC, et al. Amyloid beta concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement. 2017;13:841–9.
Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Dore V, Fowler C, Li QX, Martins R, Rowe C, et al. High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature. 2018;554:249–54.
Schindler SE, Bollinger JG, Ovod V, Mawuenyega KG, Li Y, Gordon BA, Holtzman DM, Morris JC, Benzinger TLS, Xiong C, et al. High-precision plasma beta-amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019;93:e1647–59.
Janelidze S, Stomrud E, Palmqvist S, Zetterberg H, van Westen D, Jeromin A, Song L, Hanlon D, Tan Hehir CA, Baker D, et al. Plasma beta-amyloid in Alzheimer’s disease and vascular disease. Sci Rep. 2016;6:26801.
Verberk IMW, Slot RE, Verfaillie SCJ, Heijst H, Prins ND, van Berckel BNM, Scheltens P, Teunissen CE, van der Flier WM. Plasma amyloid as prescreener for the earliest Alzheimer pathological changes. Ann Neurol. 2018;84:648–58.
Vergallo A, Megret L, Lista S, Cavedo E, Zetterberg H, Blennow K, Vanmechelen E, De Vos A, Habert MO, Potier MC, et al. Plasma amyloid beta 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer’s disease. Alzheimers Dement. 2019;15:764–75.
Fandos N, Perez-Grijalba V, Pesini P, Olmos S, Bossa M, Villemagne VL, Doecke J, Fowler C, Masters CL, Sarasa M, Group AR. Plasma amyloid beta 42/40 ratios as biomarkers for amyloid beta cerebral deposition in cognitively normal individuals. Alzheimers Dement (Amst). 2017;8:179–87.
Illan-Gala I, Lleo A, Karydas A, Staffaroni AM, Zetterberg H, Sivasankaran R, Grinberg LT, Spina S, Kramer JH, Ramos EM, et al. Plasma tau and neurofilament light in frontotemporal lobar degeneration and Alzheimer disease. Neurology. 2021;96:e671–83.
Pase MP, Beiser AS, Himali JJ, Satizabal CL, Aparicio HJ, DeCarli C, Chene G, Dufouil C, Seshadri S. Assessment of plasma total tau level as a predictive biomarker for dementia and related endophenotypes. JAMA Neurol. 2019;76:598–606.
Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreasson U, Stomrud E, Palmqvist S, Baker D, Tan Hehir CA, Jeromin A, et al. Plasma tau in Alzheimer disease. Neurology. 2016;87:1827–35.
Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J, Hansson O. Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther. 2013;5:9.
Palmqvist S, Insel PS, Zetterberg H, Blennow K, Brix B, Stomrud E, Alzheimer’s Disease Neuroimaging I, Swedish Bio Fs, Mattsson N, Hansson O. Accurate risk estimation of beta-amyloid positivity to identify prodromal Alzheimer’s disease: cross-validation study of practical algorithms. Alzheimers Dement. 2019;15:194–204.
Kovacs GG, Andreasson U, Liman V, Regelsberger G, Lutz MI, Danics K, Keller E, Zetterberg H, Blennow K. Plasma and cerebrospinal fluid tau and neurofilament concentrations in rapidly progressive neurological syndromes: a neuropathology-based cohort. Eur J Neurol. 2017;24:1326-e1377.
Ashton NJ, Puig-Pijoan A, Mila-Aloma M, Fernandez-Lebrero A, Garcia-Escobar G, Gonzalez-Ortiz F, Kac PR, Brum WS, Benedet AL, Lantero-Rodriguez J, et al. Plasma and CSF biomarkers in a memory clinic: Head-to-head comparison of phosphorylated tau immunoassays. Alzheimers Dement. 2023;19:1913–24.
Thijssen EH, La Joie R, Wolf A, Strom A, Wang P, Iaccarino L, Bourakova V, Cobigo Y, Heuer H, Spina S, et al. Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration. Nat Med. 2020;26:387–97.
Lantero Rodriguez J, Karikari TK, Suarez-Calvet M, Troakes C, King A, Emersic A, Aarsland D, Hye A, Zetterberg H, Blennow K, Ashton NJ. Plasma p-tau181 accurately predicts Alzheimer’s disease pathology at least 8 years prior to post-mortem and improves the clinical characterisation of cognitive decline. Acta Neuropathol. 2020;140:267–78.
Palmqvist S, Insel PS, Stomrud E, Janelidze S, Zetterberg H, Brix B, Eichenlaub U, Dage JL, Chai X, Blennow K, et al. Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer’s disease. EMBO Mol Med. 2019;11: e11170.
Cousins KAQ, Irwin DJ, Chen-Plotkin A, Shaw LM, Arezoumandan S, Lee EB, Wolk DA, Weintraub D, Spindler M, Deik A, et al. Plasma GFAP associates with secondary Alzheimer’s pathology in Lewy body disease. Ann Clin Transl Neurol. 2023;10:802–13.
De Meyer S, Vanbrabant J, Schaeverbeke JM, Reinartz M, Luckett ES, Dupont P, Van Laere K, Stoops E, Vanmechelen E, Poesen K, Vandenberghe R. Phospho-specific plasma p-tau181 assay detects clinical as well as asymptomatic Alzheimer’s disease. Ann Clin Transl Neurol. 2022;9:734–46.
Ashton NJ, Pascoal TA, Karikari TK, Benedet AL, Lantero-Rodriguez J, Brinkmalm G, Snellman A, Scholl M, Troakes C, Hye A, et al. Plasma p-tau231: a new biomarker for incipient Alzheimer’s disease pathology. Acta Neuropathol. 2021;141:709–24.
Ashton NJ, Janelidze S, Mattsson-Carlgren N, Binette AP, Strandberg O, Brum WS, Karikari TK, Gonzalez-Ortiz F, Di Molfetta G, Meda FJ, et al. Differential roles of Abeta42/40, p-tau231 and p-tau217 for Alzheimer’s trial selection and disease monitoring. Nat Med. 2022;28:2555–62.
Suarez-Calvet M, Karikari TK, Ashton NJ, Lantero Rodriguez J, Mila-Aloma M, Gispert JD, Salvado G, Minguillon C, Fauria K, Shekari M, et al. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Abeta pathology are detected. EMBO Mol Med. 2020;12: e12921.
Barthelemy NR, Bateman RJ, Hirtz C, Marin P, Becher F, Sato C, Gabelle A, Lehmann S. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer’s disease and PET amyloid-positive patient identification. Alzheimers Res Ther. 2020;12:26.
Janelidze S, Stomrud E, Smith R, Palmqvist S, Mattsson N, Airey DC, Proctor NK, Chai X, Shcherbinin S, Sims JR, et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat Commun. 2020;11:1683.
Hanes J, Kovac A, Kvartsberg H, Kontsekova E, Fialova L, Katina S, Kovacech B, Stevens E, Hort J, Vyhnalek M, et al. Evaluation of a novel immunoassay to detect p-tau Thr217 in the CSF to distinguish Alzheimer disease from other dementias. Neurology. 2020;95:e3026–35.
Palmqvist S, Janelidze S, Quiroz YT, Zetterberg H, Lopera F, Stomrud E, Su Y, Chen Y, Serrano GE, Leuzy A, et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA. 2020;324:772–81.
Mielke MM, Frank RD, Dage JL, Jeromin A, Ashton NJ, Blennow K, Karikari TK, Vanmechelen E, Zetterberg H, Algeciras-Schimnich A, et al. Comparison of plasma phosphorylated tau species with amyloid and tau positron emission tomography, neurodegeneration, vascular pathology, and cognitive outcomes. JAMA Neurol. 2021;78:1108–17.
Karikari TK, Emersic A, Vrillon A, Lantero-Rodriguez J, Ashton NJ, Kramberger MG, Dumurgier J, Hourregue C, Cucnik S, Brinkmalm G, et al. Head-to-head comparison of clinical performance of CSF phospho-tau T181 and T217 biomarkers for Alzheimer’s disease diagnosis. Alzheimers Dement. 2021;17:755–67.
Barthelemy NR, Saef B, Li Y, Gordon BA, He Y, Horie K, Stomrud E, Salvado G, Janelidze S, Sato C, et al. CSF tau phosphorylation occupancies at T217 and T205 represent improved biomarkers of amyloid and tau pathology in Alzheimer’s disease. Nat Aging. 2023;3:391–401.
Bayoumy S, Verberk IMW, den Dulk B, Hussainali Z, Zwan M, van der Flier WM, Ashton NJ, Zetterberg H, Blennow K, Vanbrabant J, et al. Clinical and analytical comparison of six Simoa assays for plasma P-tau isoforms P-tau181, P-tau217, and P-tau231. Alzheimers Res Ther. 2021;13:198.
Janelidze S, Bali D, Ashton NJ, Barthelemy NR, Vanbrabant J, Stoops E, Vanmechelen E, He Y, Dolado AO, Triana-Baltzer G, et al. Head-to-head comparison of 10 plasma phospho-tau assays in prodromal Alzheimer’s disease. Brain. 2023;146:1592–601.
Soleimani-Meigooni DN, Iaccarino L, La Joie R, Baker S, Bourakova V, Boxer AL, Edwards L, Eser R, Gorno-Tempini ML, Jagust WJ, et al. 18F-flortaucipir PET to autopsy comparisons in Alzheimer’s disease and other neurodegenerative diseases. Brain. 2020;143:3477–94.
Schaeverbeke JM, Gabel S, Meersmans K, Luckett ES, De Meyer S, Adamczuk K, Nelissen N, Goovaerts V, Radwan A, Sunaert S, et al. Baseline cognition is the best predictor of 4-year cognitive change in cognitively intact older adults. Alzheimers Res Ther. 2021;13:75.
Ossenkoppele R, Smith R, Mattsson-Carlgren N, Groot C, Leuzy A, Strandberg O, Palmqvist S, Olsson T, Jogi J, Stormrud E, et al. Accuracy of tau positron emission tomography as a prognostic marker in preclinical and prodromal Alzheimer disease: a head-to-head comparison against amyloid positron emission tomography and magnetic resonance imaging. JAMA Neurol. 2021;78:961–71.
Horie K, Barthelemy NR, Sato C, Bateman RJ. CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer’s disease. Brain. 2021;144:515–27.
Horie K, Salvado G, Barthelemy NR, Janelidze S, Li Y, He Y, Saef B, Chen CD, Jiang H, Strandberg O, et al. CSF MTBR-tau243 is a specific biomarker of tau tangle pathology in Alzheimer’s disease. Nat Med. 2023;29:1954–63.
Salvado G, Horie K, Barthelemy NR, Vogel JW, Pichet Binette A, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Holtzman DM, et al. Disease staging of Alzheimer’s disease using a CSF-based biomarker model. Nat Aging. 2024;4:694–708.
Barthelemy NR, Li Y, Joseph-Mathurin N, Gordon BA, Hassenstab J, Benzinger TLS, Buckles V, Fagan AM, Perrin RJ, Goate AM, et al. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat Med. 2020;26:398–407.
Gonzalez-Ortiz F, Turton M, Kac PR, Smirnov D, Premi E, Ghidoni R, Benussi L, Cantoni V, Saraceno C, Rivolta J, et al. Brain-derived tau: a novel blood-based biomarker for Alzheimer’s disease-type neurodegeneration. Brain. 2023;146:1152–65.
Gonzalez-Ortiz F, Kirsebom BE, Contador J, Tanley JE, Selnes P, Gisladottir B, Palhaugen L, Suhr Hemminghyth M, Jarholm J, Skogseth R, et al. Plasma brain-derived tau is an amyloid-associated neurodegeneration biomarker in Alzheimer’s disease. Nat Commun. 2024;15:2908.
De Meyer S, Blujdea ER, Schaeverbeke J, Reinartz M, Luckett ES, Adamczuk K, Van Laere K, Dupont P, Teunissen CE, Vandenberghe R, Poesen K. Longitudinal associations of serum biomarkers with early cognitive, amyloid and grey matter changes. Brain. 2024;147:936–48.
Chatterjee P, Pedrini S, Stoops E, Goozee K, Villemagne VL, Asih PR, Verberk IMW, Dave P, Taddei K, Sohrabi HR, et al. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Transl Psychiatry. 2021;11:27.
Verberk IMW, Laarhuis MB, van den Bosch KA, Ebenau JL, van Leeuwenstijn M, Prins ND, Scheltens P, Teunissen CE, van der Flier WM. Serum markers glial fibrillary acidic protein and neurofilament light for prognosis and monitoring in cognitively normal older people: a prospective memory clinic-based cohort study. Lancet Healthy Longev. 2021;2:e87–95.
Guo Y, You J, Zhang Y, Liu WS, Huang YY, Zhang YR, Zhang W, Dong Q, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict future dementia in healthy adults. Nat Aging. 2024;4:247–60.
Oeckl P, Anderl-Straub S, Von Arnim CAF, Baldeiras I, Diehl-Schmid J, Grimmer T, Halbgebauer S, Kort AM, Lima M, Marques TM, et al. Serum GFAP differentiates Alzheimer’s disease from frontotemporal dementia and predicts MCI-to-dementia conversion. J Neurol Neurosurg Psychiatry. 2022;93:659–67.
Zhu N, Santos-Santos M, Illan-Gala I, Montal V, Estelles T, Barroeta I, Altuna M, Arranz J, Munoz L, Belbin O, et al. Plasma glial fibrillary acidic protein and neurofilament light chain for the diagnostic and prognostic evaluation of frontotemporal dementia. Transl Neurodegener. 2021;10:50.
Pereira JB, Janelidze S, Smith R, Mattsson-Carlgren N, Palmqvist S, Teunissen CE, Zetterberg H, Stomrud E, Ashton NJ, Blennow K, Hansson O. Plasma GFAP is an early marker of amyloid-beta but not tau pathology in Alzheimer’s disease. Brain. 2021;144:3505–16.
Cousins KAQ, Phillips JS, Das SR, O’Brien K, Tropea TF, Chen-Plotkin A, Shaw LM, Nasrallah IM, Mechanic-Hamilton D, McMillan CT, et al. Pathologic and cognitive correlates of plasma biomarkers in neurodegenerative disease. Alzheimers Dement. 2024;20:3889–905.
Dickson DW, Farlo J, Davies P, Crystal H, Fuld P, Yen SH. Alzheimer’s disease. A double-labeling immunohistochemical study of senile plaques. Am J Pathol. 1988;132:86–101.
Sheng JG, Mrak RE, Griffin WS. Glial-neuronal interactions in Alzheimer disease: progressive association of IL-1alpha+ microglia and S100beta+ astrocytes with neurofibrillary tangle stages. J Neuropathol Exp Neurol. 1997;56:285–90.
Benedet AL, Mila-Aloma M, Vrillon A, Ashton NJ, Pascoal TA, Lussier F, Karikari TK, Hourregue C, Cognat E, Dumurgier J, et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 2021;78:1471–83.
Ashton NJ, Suarez-Calvet M, Heslegrave A, Hye A, Razquin C, Pastor P, Sanchez-Valle R, Molinuevo JL, Visser PJ, Blennow K, et al. Plasma levels of soluble TREM2 and neurofilament light chain in TREM2 rare variant carriers. Alzheimers Res Ther. 2019;11:94.
Piccio L, Deming Y, Del-Aguila JL, Ghezzi L, Holtzman DM, Fagan AM, Fenoglio C, Galimberti D, Borroni B, Cruchaga C. Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol. 2016;131:925–33.
Mila-Aloma M, Salvado G, Gispert JD, Vilor-Tejedor N, Grau-Rivera O, Sala-Vila A, Sanchez-Benavides G, Arenaza-Urquijo EM, Crous-Bou M, Gonzalez-de-Echavarri JM, et al. Amyloid beta, tau, synaptic, neurodegeneration, and glial biomarkers in the preclinical stage of the Alzheimer’s continuum. Alzheimers Dement. 2020;16:1358–71.
Villar-Pique A, Schmitz M, Hermann P, Goebel S, Bunck T, Varges D, Ferrer I, Riggert J, Llorens F, Zerr I. Plasma YKL-40 in the spectrum of neurodegenerative dementia. J Neuroinflammation. 2019;16:145.
Schaeverbeke J, Gille B, Adamczuk K, Vanderstichele H, Chassaing E, Bruffaerts R, Neyens V, Stoops E, Tournoy J, Vandenberghe R, Poesen K. Cerebrospinal fluid levels of synaptic and neuronal integrity correlate with gray matter volume and amyloid load in the precuneus of cognitively intact older adults. J Neurochem. 2019;149:139–57.
Watabe-Rudolph M, Song Z, Lausser L, Schnack C, Begus-Nahrmann Y, Scheithauer MO, Rettinger G, Otto M, Tumani H, Thal DR, et al. Chitinase enzyme activity in CSF is a powerful biomarker of Alzheimer disease. Neurology. 2012;78:569–77.
Martirosian RA, Wiedner CD, Sanchez J, Mun KT, Marla K, Teran C, Thirion M, Liebeskind DS, McGrath ER, Zucker JM, et al. Association of incident stroke risk with an IL-18-centered inflammatory network biomarker composite. Stroke. 2024;55:1601–8.
Altendahl M, Maillard P, Harvey D, Cotter D, Walters S, Wolf A, Singh B, Kakarla V, Azizkhanian I, Sheth SA, et al. An IL-18-centered inflammatory network as a biomarker for cerebral white matter injury. PLoS One. 2020;15: e0227835.
Italiani P, Carlesi C, Giungato P, Puxeddu I, Borroni B, Bossu P, Migliorini P, Siciliano G, Boraschi D. Evaluating the levels of interleukin-1 family cytokines in sporadic amyotrophic lateral sclerosis. J Neuroinflammation. 2014;11: 94.
Lok HC, Katzeff JS, Hodges JR, Piguet O, Fu Y, Halliday GM, Kim WS. Elevated GRO-alpha and IL-18 in serum and brain implicate the NLRP3 inflammasome in frontotemporal dementia. Sci Rep. 2023;13:8942.
Corbo RM, Businaro R, Scarabino D. Leukocyte telomere length and plasma interleukin-1beta and interleukin-18 levels in mild cognitive impairment and Alzheimer’s disease: new biomarkers for diagnosis and disease progression? Neural Regen Res. 2021;16:1397–8.
Villemagne VL, Harada R, Dore V, Furumoto S, Mulligan R, Kudo Y, Burnham S, Krishnadas N, Bozinovski S, Huang K, et al. First-in-humans evaluation of (18)F-SMBT-1, a novel (18)F-labeled monoamine oxidase-B PET tracer for imaging reactive astrogliosis. J Nucl Med. 2022;63:1551–9.
Gafson AR, Barthelemy NR, Bomont P, Carare RO, Durham HD, Julien JP, Kuhle J, Leppert D, Nixon RA, Weller RO, et al. Neurofilaments: neurobiological foundations for biomarker applications. Brain. 2020;143:1975–98.
Gaetani L, Blennow K, Calabresi P, Di Filippo M, Parnetti L, Zetterberg H. Neurofilament light chain as a biomarker in neurological disorders. J Neurol Neurosurg Psychiatry. 2019;90:870–81.
Kuhle J, Barro C, Andreasson U, Derfuss T, Lindberg R, Sandelius A, Liman V, Norgren N, Blennow K, Zetterberg H. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa. Clin Chem Lab Med. 2016;54:1655–61.
Khalil M, Teunissen CE, Otto M, Piehl F, Sormani MP, Gattringer T, Barro C, Kappos L, Comabella M, Fazekas F, et al. Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol. 2018;14:577–89.
Yuan A, Nixon RA. Neurofilament proteins as biomarkers to monitor neurological diseases and the efficacy of therapies. Front Neurosci. 2021;15: 689938.
de Wolf F, Ghanbari M, Licher S, McRae-McKee K, Gras L, Weverling GJ, Wermeling P, Sedaghat S, Ikram MK, Waziry R, et al. Plasma tau, neurofilament light chain and amyloid-beta levels and risk of dementia; a population-based cohort study. Brain. 2020;143:1220–32.
Wang X, Shi Z, Qiu Y, Sun D, Zhou H. Peripheral GFAP and NfL as early biomarkers for dementia: longitudinal insights from the UK Biobank. BMC Med. 2024;22:192.
Graff-Radford J, Mielke MM, Hofrenning EI, Kouri N, Lesnick TG, Moloney CM, Rabinstein A, Cabrera-Rodriguez JN, Rothberg DM, Przybelski SA, et al. Association of plasma biomarkers of amyloid and neurodegeneration with cerebrovascular disease and Alzheimer’s disease. Neurobiol Aging. 2022;119:1–7.
Dittrich A, Ashton NJ, Zetterberg H, Blennow K, Zettergren A, Simren J, Skillback T, Shams S, Machado A, Westman E, et al. Association of chronic kidney disease with plasma NfL and other biomarkers of neurodegeneration: the H70 birth cohort study in Gothenburg. Neurology. 2023;101:e277–88.
Fitzgerald KC, Sotirchos ES, Smith MD, Lord HN, DuVal A, Mowry EM, Calabresi PA. Contributors to serum NfL levels in people without neurologic disease. Ann Neurol. 2022;92:688–98.
Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007;68:1501–8.
Bereczki E, Francis PT, Howlett D, Pereira JB, Hoglund K, Bogstedt A, Cedazo-Minguez A, Baek JH, Hortobagyi T, Attems J, et al. Synaptic proteins predict cognitive decline in Alzheimer’s disease and Lewy body dementia. Alzheimers Dement. 2016;12:1149–58.
Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA, Katzman R. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–80.
DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol. 1990;27:457–64.
Nilsson J, Cousins KAQ, Gobom J, Portelius E, Chen-Plotkin A, Shaw LM, Grossman M, Irwin DJ, Trojanowski JQ, Zetterberg H, et al. Cerebrospinal fluid biomarker panel of synaptic dysfunction in Alzheimer’s disease and other neurodegenerative disorders. Alzheimers Dement. 2023;19:1775–84.
Das S, van Engelen ME, Goossens J, Jacobs D, Bongers B, Fieldhouse JLP, Pijnenburg YAL, Teunissen CE, Vanmechelen E, Verberk IMW. The use of synaptic biomarkers in cerebrospinal fluid to differentiate behavioral variant of frontotemporal dementia from primary psychiatric disorders and Alzheimer’s disease. Alzheimers Res Ther. 2024;16:34.
Nilsson J, Pichet Binette A, Palmqvist S, Brum WS, Janelidze S, Ashton NJ, Spotorno N, Stomrud E, Gobom J, Zetterberg H, et al. Cerebrospinal fluid biomarker panel for synaptic dysfunction in a broad spectrum of neurodegenerative diseases. Brain. 2024;147:2414–27.
Das S, Goossens J, Jacobs D, Dewit N, Pijnenburg YAL, In ’t Veld S, Teunissen CE, Vanmechelen E. Synaptic biomarkers in the cerebrospinal fluid associate differentially with classical neuronal biomarkers in patients with Alzheimer’s disease and frontotemporal dementia. Alzheimers Res Ther. 2023;15:62.
Clarke MTM, Brinkmalm A, Foiani MS, Woollacott IOC, Heller C, Heslegrave A, Keshavan A, Fox NC, Schott JM, Warren JD, et al. CSF synaptic protein concentrations are raised in those with atypical Alzheimer’s disease but not frontotemporal dementia. Alzheimers Res Ther. 2019;11:105.
Barba L, Abu-Rumeileh S, Halbgebauer S, Bellomo G, Paolini Paoletti F, Gaetani L, Oeckl P, Steinacker P, Massa F, Parnetti L, Otto M. CSF synaptic biomarkers in AT(N)-based subgroups of Lewy body disease. Neurology. 2023;101:e50–62.
Kivisakk P, Carlyle BC, Sweeney T, Quinn JP, Ramirez CE, Trombetta BA, Mendes M, Brock M, Rubel C, Czerkowicz J, et al. Increased levels of the synaptic proteins PSD-95, SNAP-25, and neurogranin in the cerebrospinal fluid of patients with Alzheimer’s disease. Alzheimers Res Ther. 2022;14:58.
Oeckl P, Bluma M, Bucci M, Halbgebauer S, Chiotis K, Sandebring-Matton A, Ashton NJ, Molfetta GD, Grotschel L, Kivipelto M, et al. Blood beta-synuclein is related to amyloid PET positivity in memory clinic patients. Alzheimers Dement. 2023;19:4896–907.
Oeckl P, Janelidze S, Halbgebauer S, Stomrud E, Palmqvist S, Otto M, Hansson O. Higher plasma beta-synuclein indicates early synaptic degeneration in Alzheimer’s disease. Alzheimers Dement. 2023;19:5095–102.
Vrillon A, Mouton-Liger F, Martinet M, Cognat E, Hourregue C, Dumurgier J, Bouaziz-Amar E, Brinkmalm A, Blennow K, Zetterberg H, et al. Plasma neuregulin 1 as a synaptic biomarker in Alzheimer’s disease: a discovery cohort study. Alzheimers Res Ther. 2022;14:71.
Chang KA, Shin KY, Nam E, Lee YB, Moon C, Suh YH, Lee SH. Plasma soluble neuregulin-1 as a diagnostic biomarker for Alzheimer’s disease. Neurochem Int. 2016;97:1–7.
Carson RE, Naganawa M, Toyonaga T, Koohsari S, Yang Y, Chen MK, Matuskey D, Finnema SJ. Imaging of synaptic density in neurodegenerative disorders. J Nucl Med. 2022;63:60S-67S.
Shanaki Bavarsad M, Spina S, Oehler A, Allen IE, Suemoto CK, Leite REP, Seeley WS, Green A, Jagust W, Rabinovici GD, Grinberg LT. Comprehensive mapping of synaptic vesicle protein 2A (SV2A) in health and neurodegenerative diseases: a comparative analysis with synaptophysin and ground truth for PET-imaging interpretation. Acta Neuropathol. 2024;148:58.
Kumar A, Scarpa M, Nordberg A. Tracing synaptic loss in Alzheimer’s brain with SV2A PET-tracer UCB-J. Alzheimers Dement. 2024;20:2589–605.
Alber J, Bouwman F, den Haan J, Rissman RA, De Groef L, Koronyo-Hamaoui M, Lengyel I, Thal DR, Alzheimer’s Association ITEaaBfADPIA. Retina pathology as a target for biomarkers for Alzheimer’s disease: current status, ophthalmopathological background, challenges, and future directions. Alzheimers Dement. 2024;20:728–40.
Snyder PJ, Alber J, Alt C, Bain LJ, Bouma BE, Bouwman FH, DeBuc DC, Campbell MCW, Carrillo MC, Chew EY, et al. Retinal imaging in Alzheimer’s and neurodegenerative diseases. Alzheimers Dement. 2021;17:103–11.
Alber J, Arthur E, Sinoff S, DeBuc DC, Chew EY, Douquette L, Hatch WV, Hudson C, Kashani A, Lee CS, et al. A recommended “minimum data set” framework for SD-OCT retinal image acquisition and analysis from the Atlas of Retinal Imaging in Alzheimer’s Study (ARIAS). Alzheimers Dement (Amst). 2020;12:e12119.
Coppola G, Di Renzo A, Ziccardi L, Martelli F, Fadda A, Manni G, Barboni P, Pierelli F, Sadun AA, Parisi V. Optical coherence tomography in Alzheimer’s disease: a meta-analysis. PLoS One. 2015;10:e0134750.
Doustar J, Torbati T, Black KL, Koronyo Y, Koronyo-Hamaoui M. Optical coherence tomography in Alzheimer’s disease and other neurodegenerative diseases. Front Neurol. 2017;8:701.
O’Bryhim BE, Apte RS, Kung N, Coble D, Van Stavern GP. Association of preclinical Alzheimer disease with optical coherence tomographic angiography findings. JAMA Ophthalmol. 2018;136:1242–8.
Wisely CE, Wang D, Henao R, Grewal DS, Thompson AC, Robbins CB, Yoon SP, Soundararajan S, Polascik BW, Burke JR, et al. Convolutional neural network to identify symptomatic Alzheimer’s disease using multimodal retinal imaging. Br J Ophthalmol. 2022;106:388–95.
Hadoux X, Hui F, Lim JKH, Masters CL, Pebay A, Chevalier S, Ha J, Loi S, Fowler CJ, Rowe C, et al. Non-invasive in vivo hyperspectral imaging of the retina for potential biomarker use in Alzheimer’s disease. Nat Commun. 2019;10:4227.
Lemmens S, Van Craenendonck T, Van Eijgen J, De Groef L, Bruffaerts R, de Jesus DA, Charle W, Jayapala M, Sunaric-Megevand G, Standaert A, et al. Combination of snapshot hyperspectral retinal imaging and optical coherence tomography to identify Alzheimer’s disease patients. Alzheimers Res Ther. 2020;12:144.
More SS, Beach JM, McClelland C, Mokhtarzadeh A, Vince R. In vivo assessment of retinal biomarkers by hyperspectral imaging: early detection of Alzheimer’s disease. ACS Chem Neurosci. 2019;10:4492–501.
Du X, Koronyo Y, Mirzaei N, Yang C, Fuchs DT, Black KL, Koronyo-Hamaoui M, Gao L. Label-free hyperspectral imaging and deep-learning prediction of retinal amyloid beta-protein and phosphorylated tau. PNAS Nexus. 2022;1:pgac164.
Attems J, Jellinger K. Neuropathological correlates of cerebral multimorbidity. Curr Alzheimer Res. 2013;10:569–77.
Bolsewig K, van Unnik A, Blujdea ER, Gonzalez MC, Ashton NJ, Aarsland D, Zetterberg H, Padovani A, Bonanni L, Mollenhauer B, et al. Association of plasma amyloid, P-Tau, GFAP, and NfL with CSF, clinical, and cognitive features in patients with dementia with Lewy bodies. Neurology. 2024;102: e209418.
Smith R, Capotosti F, Schain M, Ohlsson T, Vokali E, Molette J, Touilloux T, Hliva V, Dimitrakopoulos IK, Puschmann A, et al. The alpha-synuclein PET tracer [18F] ACI-12589 distinguishes multiple system atrophy from other neurodegenerative diseases. Nat Commun. 2023;14:6750.
Endo H, Ono M, Takado Y, Matsuoka K, Takahashi M, Tagai K, Kataoka Y, Hirata K, Takahata K, Seki C, et al. Imaging alpha-synuclein pathologies in animal models and patients with Parkinson’s and related diseases. Neuron. 2024;112(2540–2557): e2548.
Fernandes Gomes B, Farris CM, Ma Y, Concha-Marambio L, Lebovitz R, Nellgard B, Dalla K, Constantinescu J, Constantinescu R, Gobom J, et al. alpha-Synuclein seed amplification assay as a diagnostic tool for parkinsonian disorders. Parkinsonism Relat Disord. 2023;117: 105807.
Samudra N, Fischer DL, Lenio S, Lario Lago A, Ljubenkov PA, Rojas JC, Seeley WW, Spina S, Staffaroni AM, Tablante J, et al. Clinicopathological correlation of cerebrospinal fluid alpha-synuclein seed amplification assay in a behavioral neurology autopsy cohort. Alzheimers Dement. 2024;20:3334–41.
Rossi M, Candelise N, Baiardi S, Capellari S, Giannini G, Orru CD, Antelmi E, Mammana A, Hughson AG, Calandra-Buonaura G, et al. Ultrasensitive RT-QuIC assay with high sensitivity and specificity for Lewy body-associated synucleinopathies. Acta Neuropathol. 2020;140:49–62.
Bongianni M, Ladogana A, Capaldi S, Klotz S, Baiardi S, Cagnin A, Perra D, Fiorini M, Poleggi A, Legname G, et al. alpha-Synuclein RT-QuIC assay in cerebrospinal fluid of patients with dementia with Lewy bodies. Ann Clin Transl Neurol. 2019;6:2120–6.
Arnold MR, Coughlin DG, Brumbach BH, Smirnov DS, Concha-Marambio L, Farris CM, Ma Y, Kim Y, Wilson EN, Kaye JA, et al. alpha-Synuclein seed amplification in CSF and brain from patients with different brain distributions of pathological alpha-Synuclein in the context of co-pathology and non-LBD diagnoses. Ann Neurol. 2022;92:650–62.
Manne S, Kondru N, Jin H, Serrano GE, Anantharam V, Kanthasamy A, Adler CH, Beach TG, Kanthasamy AG. Blinded RT-QuIC analysis of alpha-Synuclein biomarker in skin tissue from Parkinson’s disease patients. Mov Disord. 2020;35:2230–9.
Mammana A, Baiardi S, Quadalti C, Rossi M, Donadio V, Capellari S, Liguori R, Parchi P. RT-QuIC detection of pathological alpha-Synuclein in skin punches of patients with Lewy body disease. Mov Disord. 2021;36:2173–7.
Iranzo A, Mammana A, Munoz-Lopetegi A, Dellavalle S, Maya G, Rossi M, Serradell M, Baiardi S, Arqueros A, Quadalti C, et al. Misfolded alpha-Synuclein assessment in the skin and CSF by RT-QuIC in isolated REM sleep behavior disorder. Neurology. 2023;100:e1944–54.
Sorrentino ZA, Vijayaraghavan N, Gorion KM, Riffe CJ, Strang KH, Caldwell J, Giasson BI. Physiological C-terminal truncation of alpha-synuclein potentiates the prion-like formation of pathological inclusions. J Biol Chem. 2018;293:18914–32.
Hall S, Orru CD, Serrano GE, Galasko D, Hughson AG, Groveman BR, Adler CH, Beach TG, Caughey B, Hansson O. Performance of alphaSynuclein RT-QuIC in relation to neuropathological staging of Lewy body disease. Acta Neuropathol Commun. 2022;10:90.
van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.
Kluge A, Bunk J, Schaeffer E, Drobny A, Xiang W, Knacke H, Bub S, Luckstadt W, Arnold P, Lucius R, et al. Detection of neuron-derived pathological alpha-synuclein in blood. Brain. 2022;145:3058–71.
Okuzumi A, Hatano T, Matsumoto G, Nojiri S, Ueno SI, Imamichi-Tatano Y, Kimura H, Kakuta S, Kondo A, Fukuhara T, et al. Propagative alpha-synuclein seeds as serum biomarkers for synucleinopathies. Nat Med. 2023;29:1448–55.
Robinson JL, Lee EB, Xie SX, Rennert L, Suh E, Bredenberg C, Caswell C, Van Deerlin VM, Yan N, Yousef A, et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain. 2018;141:2181–93.
Josephs KA, Whitwell JL, Weigand SD, Murray ME, Tosakulwong N, Liesinger AM, Petrucelli L, Senjem ML, Knopman DS, Boeve BF, et al. TDP-43 is a key player in the clinical features associated with Alzheimer’s disease. Acta Neuropathol. 2014;127:811–24.
Josephs KA, Whitwell JL, Tosakulwong N, Weigand SD, Murray ME, Liesinger AM, Petrucelli L, Senjem ML, Ivnik RJ, Parisi JE, et al. TAR DNA-binding protein 43 and pathological subtype of Alzheimer’s disease impact clinical features. Ann Neurol. 2015;78:697–709.
Seredenina T, Vokali E, Dreyfus N, Chevallier E, Afroz T, Jaquier T, Serra AM, Clavel M, Rathnam M, Melly T, et al. Discovery and optimization of the first-in-class TDP-43 PET tracer. Alzheimers Dement. 2023;19: e075525.
Carlos AF, Tosakulwong N, Weigand SD, Senjem ML, Schwarz CG, Knopman DS, Boeve BF, Petersen RC, Nguyen AT, Reichard RR, et al. TDP-43 pathology effect on volume and flortaucipir uptake in Alzheimer’s disease. Alzheimers Dement. 2023;19:2343–54.
Foulds P, McAuley E, Gibbons L, Davidson Y, Pickering-Brown SM, Neary D, Snowden JS, Allsop D, Mann DM. TDP-43 protein in plasma may index TDP-43 brain pathology in Alzheimer’s disease and frontotemporal lobar degeneration. Acta Neuropathol. 2008;116:141–6.
Kasai T, Tokuda T, Ishigami N, Sasayama H, Foulds P, Mitchell DJ, Mann DM, Allsop D, Nakagawa M. Increased TDP-43 protein in cerebrospinal fluid of patients with amyotrophic lateral sclerosis. Acta Neuropathol. 2009;117:55–62.
Noto Y, Shibuya K, Sato Y, Kanai K, Misawa S, Sawai S, Mori M, Uchiyama T, Isose S, Nasu S, et al. Elevated CSF TDP-43 levels in amyotrophic lateral sclerosis: specificity, sensitivity, and a possible prognostic value. Amyotroph Lateral Scler. 2011;12:140–3.
Hosokawa M, Arai T, Yamashita M, Tsuji H, Nonaka T, Masuda-Suzukake M, Tamaoka A, Hasegawa M, Akiyama H. Differential diagnosis of amyotrophic lateral sclerosis from Guillain-Barre syndrome by quantitative determination of TDP-43 in cerebrospinal fluid. Int J Neurosci. 2014;124:344–9.
Verstraete E, Kuiperij HB, van Blitterswijk MM, Veldink JH, Schelhaas HJ, van den Berg LH, Verbeek MM. TDP-43 plasma levels are higher in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2012;13:446–51.
Suarez-Calvet M, Dols-Icardo O, Llado A, Sanchez-Valle R, Hernandez I, Amer G, Anton-Aguirre S, Alcolea D, Fortea J, Ferrer I, et al. Plasma phosphorylated TDP-43 levels are elevated in patients with frontotemporal dementia carrying a C9orf72 repeat expansion or a GRN mutation. J Neurol Neurosurg Psychiatry. 2014;85:684–91.
Ren Y, Li S, Chen S, Sun X, Yang F, Wang H, Li M, Cui F, Huang X. TDP-43 and phosphorylated TDP-43 levels in paired plasma and CSF samples in amyotrophic lateral sclerosis. Front Neurol. 2021;12: 663637.
Katisko K, Huber N, Kokkola T, Hartikainen P, Kruger J, Heikkinen AL, Paananen V, Leinonen V, Korhonen VE, Helisalmi S, et al. Serum total TDP-43 levels are decreased in frontotemporal dementia patients with C9orf72 repeat expansion or concomitant motoneuron disease phenotype. Alzheimers Res Ther. 2022;14:151.
Irwin KE, Jasin P, Braunstein KE, Sinha IR, Garret MA, Bowden KD, Chang K, Troncoso JC, Moghekar A, Oh ES, et al. A fluid biomarker reveals loss of TDP-43 splicing repression in presymptomatic ALS-FTD. Nat Med. 2024;30:382–93.
Seddighi S, Qi YA, Brown AL, Wilkins OG, Bereda C, Belair C, Zhang YJ, Prudencio M, Keuss MJ, Khandeshi A, et al. Mis-spliced transcripts generate de novo proteins in TDP-43-related ALS/FTD. Sci Transl Med. 2024;16:eadg7162.
Costantino I, Meng A, Ravits J. Alternatively spliced ELAVL3 cryptic exon 4a causes ELAVL3 downregulation in ALS TDP-43 proteinopathy. Acta Neuropathol. 2024;147:93.
Chatterjee M, Ozdemir S, Fritz C, Mobius W, Kleineidam L, Mandelkow E, Biernat J, Dogdu C, Peters O, Cosma NC, et al. Plasma extracellular vesicle tau and TDP-43 as diagnostic biomarkers in FTD and ALS. Nat Med. 2024;30:1771–83.
Hook V, Podvin S, Mosier C, Boyarko B, Seyffert L, Stringer H, Rissman RA. Emerging evidence for dysregulated proteome cargoes of tau-propagating extracellular vesicles driven by familial mutations of tau and presenilin. Extracell Vesicles Circ Nucl Acids. 2023;4:588–98.
Murray ME, Moloney CM, Kouri N, Syrjanen JA, Matchett BJ, Rothberg DM, Tranovich JF, Sirmans TNH, Wiste HJ, Boon BDC, et al. Global neuropathologic severity of Alzheimer’s disease and locus coeruleus vulnerability influences plasma phosphorylated tau levels. Mol Neurodegener. 2022;17:85.
Yu L, Boyle PA, Janelidze S, Petyuk VA, Wang T, Bennett DA, Hansson O, Schneider JA. Plasma p-tau181 and p-tau217 in discriminating PART, AD and other key neuropathologies in older adults. Acta Neuropathol. 2023;146:1–11.
Jack CR Jr, Holtzman DM. Biomarker modeling of Alzheimer’s disease. Neuron. 2013;80:1347–58.
Thal DR, Ronisz A, Tousseyn T, Rijal Upadhaya A, Balakrishnan K, Vandenberghe R, Vandenbulcke M, von Arnim CAF, Otto M, Beach TG, et al. Different aspects of Alzheimer’s disease-related amyloid beta-peptide pathology and their relationship to amyloid positron emission tomography imaging and dementia. Acta Neuropathol Commun. 2019;7:178.
Hoglinger GU, Adler CH, Berg D, Klein C, Outeiro TF, Poewe W, Postuma R, Stoessl AJ, Lang AE. A biological classification of Parkinson’s disease: the SynNeurGe research diagnostic criteria. Lancet Neurol. 2024;23:191–204.
Simuni T, Chahine LM, Poston K, Brumm M, Buracchio T, Campbell M, Chowdhury S, Coffey C, Concha-Marambio L, Dam T, et al. A biological definition of neuronal alpha-synuclein disease: towards an integrated staging system for research. Lancet Neurol. 2024;23:178–90.
Kovacs GG, Grinberg LT, Halliday G, Alafuzoff I, Dugger BN, Murayama S, Forrest SL, Martinez-Valbuena I, Tanaka H, Kon T, et al. Biomarker-based approach to alpha-Synucleinopathies: lessons from neuropathology. Mov Disord. 2024;39:2173–9.
Klunk WE, Koeppe RA, Price JC, Benzinger TL, Devous MD Sr, Jagust WJ, Johnson KA, Mathis CA, Minhas D, Pontecorvo MJ, et al. The Centiloid Project: standardizing quantitative amyloid plaque estimation by PET. Alzheimers Dement. 2015;11:1-15 e11-14.
Leuzy A, Raket LL, Villemagne VL, Klein G, Tonietto M, Olafson E, Baker S, Saad ZS, Bullich S, Lopresti B, et al. Harmonizing tau positron emission tomography in Alzheimer’s disease: the CenTauR scale and the joint propagation model. Alzheimers Dement. 2024;20:5833–48.
Thompson PM, Raj T. Artificial Intelligence and Machine Learning (AIML) approaches to empower genomics, drug, and biomarker discovery in ADRD. Alzheimers Dement. 2023;19: e077714.
Momota Y, Bun S, Hirano J, Kamiya K, Ueda R, Iwabuchi Y, Takahata K, Yamamoto Y, Tezuka T, Kubota M, et al. Amyloid-beta prediction machine learning model using source-based morphometry across neurocognitive disorders. Sci Rep. 2024;14:7633.
Tiwari VK, Indic P, Tabassum S. A study on machine learning models in detecting cognitive impairments in Alzheimer’s patients using cerebrospinal fluid biomarkers. Am J Alzheimers Dis Other Demen. 2024;39: 15333175241308645.
Dyrba M, Barkhof F, Fellgiebel A, Filippi M, Hausner L, Hauenstein K, Kirste T, Teipel SJ, group Es. Predicting prodromal Alzheimer’s disease in subjects with mild cognitive impairment using machine learning classification of multimodal multicenter diffusion-tensor and magnetic resonance imaging data. J Neuroimaging. 2015;25:738–47.
Eke CS, Jammeh E, Li X, Carroll C, Pearson S, Ifeachor E. Early detection of Alzheimer’s disease with blood plasma proteins using support vector machines. IEEE J Biomed Health Inform. 2021;25:218–26.
Li X, Ospitalieri S, Robberechts T, Hofmann L, Schmid C, Rijal Upadhaya A, Koper MJ, von Arnim CAF, Kumar S, Willem M, et al. Seeding, maturation and propagation of amyloid beta-peptide aggregates in Alzheimer’s disease. Brain. 2022;145:3558–70.
Kaufman SK, Del Tredici K, Thomas TL, Braak H, Diamond MI. Tau seeding activity begins in the transentorhinal/entorhinal regions and anticipates phospho-tau pathology in Alzheimer’s disease and PART. Acta Neuropathol. 2018;136:57–67.
Ye L, Fritschi SK, Schelle J, Obermuller U, Degenhardt K, Kaeser SA, Eisele YS, Walker LC, Baumann F, Staufenbiel M, Jucker M. Persistence of Abeta seeds in APP null mouse brain. Nat Neurosci. 2015;18:1559–61.
Charil A, Shcherbinin S, Southekal S, Devous MD, Mintun M, Murray ME, Miller BB, Schwarz AJ. Tau subtypes of Alzheimer’s disease determined in vivo using flortaucipir PET imaging. J Alzheimers Dis. 2019;71:1037–48.
Ferreira D, Mohanty R, Murray ME, Nordberg A, Kantarci K, Westman E. The hippocampal sparing subtype of Alzheimer’s disease assessed in neuropathology and in vivo tau positron emission tomography: a systematic review. Acta Neuropathol Commun. 2022;10:166.
Young AL, Marinescu RV, Oxtoby NP, Bocchetta M, Yong K, Firth NC, Cash DM, Thomas DL, Dick KM, Cardoso J, et al. Uncovering the heterogeneity and temporal complexity of neurodegenerative diseases with subtype and stage inference. Nat Commun. 2018;9:4273.
Gilbert JJ, Vinters HV. Cerebral amyloid angiopathy: incidence and complications in the aging brain. I. Cerebral hemorrhage. Stroke. 1983;14:915–23.
Thal DR, Griffin WST, De Vos RAI, Ghebremedhin E. Cerebral amyloid angiopathy and its relationship to Alzheimer’s disease. Acta Neuropathol. 2008;115:599–609.
Castellani RJ, Shanes ED, McCord M, Reish NJ, Flanagan ME, Mesulam MM, Jamshidi P. Neuropathology of anti-amyloid-beta immunotherapy: a case report. J Alzheimers Dis. 2023;93:803–13.
Jucker M, Walker LC. Alzheimer’s disease: from immunotherapy to immunoprevention. Cell. 2023;186:4260–70.
Travis J. Latest Alzheimer’s antibody is “not a miracle drug.” Science. 2023;380:571.
Morris JC, Weiner M, Xiong C, Beckett L, Coble D, Saito N, Aisen PS, Allegri R, Benzinger TLS, Berman SB, et al. Autosomal dominant and sporadic late onset Alzheimer’s disease share a common in vivo pathophysiology. Brain. 2022;145:3594–607.
Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367:795–804.
Hanseeuw BJ, Malotaux V, Dricot L, Quenon L, Sznajer Y, Cerman J, Woodard JL, Buckley C, Farrar G, Ivanoiu A, Lhommel R. Defining a centiloid scale threshold predicting long-term progression to dementia in patients attending the memory clinic: an [(18)F] flutemetamol amyloid PET study. Eur J Nucl Med Mol Imaging. 2021;48:302–10.
Cicognola C, Janelidze S, Hertze J, Zetterberg H, Blennow K, Mattsson-Carlgren N, Hansson O. 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;13:68.
Utianski RL, Martin PR, Botha H, Schwarz CG, Duffy JR, Petersen RC, Knopman DS, Clark HM, Butts AM, Machulda MM, et al. Longitudinal flortaucipir ([(18)F]AV-1451) PET imaging in primary progressive apraxia of speech. Cortex. 2019;124:33–43.
Lee WJ, Brown JA, Kim HR, La Joie R, Cho H, Lyoo CH, Rabinovici GD, Seong JK, Seeley WW, Alzheimer’s Disease Neuroimaging I. Regional Abeta-tau interactions promote onset and acceleration of Alzheimer’s disease tau spreading. Neuron. 2022;110(1932–1943):e1935.
Karikari TK, Pascoal TA, Ashton NJ, Janelidze S, Benedet AL, Rodriguez JL, Chamoun M, Savard M, Kang MS, Therriault J, 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;19:422–33.
Bastin C, Bahri MA, Meyer F, Manard M, Delhaye E, Plenevaux A, Becker G, Seret A, Mella C, Giacomelli F, et al. In vivo imaging of synaptic loss in Alzheimer’s disease with [18F]UCB-H positron emission tomography. Eur J Nucl Med Mol Imaging. 2020;47:390–402.
Mecca AP, Chen MK, O’Dell RS, Naganawa M, Toyonaga T, Godek TA, Harris JE, Bartlett HH, Zhao W, Nabulsi NB, et al. In vivo measurement of widespread synaptic loss in Alzheimer’s disease with SV2A PET. Alzheimers Dement. 2020;16:974–82.
Naganawa M, Li S, Nabulsi N, Henry S, Zheng MQ, Pracitto R, Cai Z, Gao H, Kapinos M, Labaree D, et al. First-in-human evaluation of (18)F-SynVesT-1, a radioligand for PET imaging of synaptic vesicle glycoprotein 2A. J Nucl Med. 2021;62:561–7.
Vanderlinden G, Ceccarini J, Vande Casteele T, Michiels L, Lemmens R, Triau E, Serdons K, Tournoy J, Koole M, Vandenbulcke M, Van Laere K. Spatial decrease of synaptic density in amnestic mild cognitive impairment follows the tau build-up pattern. Mol Psychiatry. 2022;27:4244–51.
Acknowledgements
The authors would like to thank the participants of their brain donation programs for their post-mortem brain donation.
Funding
The results reviewed here were funded by Fonds Wetenschappelijk Onderzoek (FWO, Belgium): G0F8516N, G065721N, G024925N (DRT), 11M0522N, 11M0524N (SDM), 18B2622N (KP); KU Leuven internal funds (Belgium): C3/20/057 (DRT), C14/21/109 (RV); Stichting Alzheimer Onderzoek, Belgium: SAO/FRA 2020/017, SAO/FRA 2023/0009 (DRT); SAO/FRA 2021/0007 (KP); SAO/FRA 2022/009 (RV); and Alzheimer’s Association (USA): 22-AAIIA-963171 (DRT); VLAIO ICON grant HBC.2019.2523 (RV).
Author information
Authors and Affiliations
Contributions
DRT: Concept and structure of this review and first draft on Background, Neuropathology, Biomarker black box, Biomarkers in a clinical context, Biomarkers for use in clinical trials and personalized medicine, and Conclusions, Figs. 1, 2, and 3, final adaptation of the different manuscript parts; KP: Concept and manuscript draft for fluid biomarkers; RV: Concept and first draft on Imaging biomarkers, PET and images for Fig. 1, contributions to the manuscript draft (other chapters); SDM: Concept and first draft on fluid biomarkers, Fig. 4, contributions to the manuscript draft (other chapters).
Corresponding author
Ethics declarations
Ethics approval and consent to participate
All autopsies were carried out according to local legislation with appropriate consent. Ethical approval for the use of cases was granted by the ethical committee of Ulm University (Germany, study 54/08) and UZ/KU-Leuven ethical committee (Belgium). The studies reviewed here covering the retrospective analysis of samples and data were approved by the UZ/KU-Leuven ethical committee (S-59295, S-65147, S-66705) (Belgium). The samples obtained from the UK were received from the brainbank donated for research by GE Healthcare after the phase III flutemetamol autopsy study had concluded (ClinicalTrials. gov identifiers NCT01165554, and NCT02090855).
Consent for publication
Not applicable for this study, which did not use person’s data. Only anonymized or pseudonymized data were processed.
Competing interests
DRT collaborated with Novartis Pharma AG (Switzerland), Probiodrug (Germany), and GE-Healthcare (UK) and received consultant honoraria from Muna Therapeutics (Belgium). DRT is vice chair of the Alzheimer's Association ISTAART professional interest area “The eye as a biomarker for AD”. RV’s institution has clinical trial agreements (RV as PI) with Alector, AviadoBio, Biogen, Denali, J&J, Eli Lilly and UCB. RV’s institution has consultancy agreements (RV as member of DSMB) with AC Immune. RV was global PI of the industry-sponsored pivotal phase 2 flutemetamol trial. KP has a consultancy agreement with Eisai. KP’s institution has biomarker trial agreements with ADx NeuroSciences, Euroimmun and Fujirebio. KP is member of the scientific advisory board of Stichting Alzheimer Onderzoek/Fondation Recherche Alzheimer, Belgium. SDM reported no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Thal, D.R., Poesen, K., Vandenberghe, R. et al. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegeneration 20, 33 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00819-y
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00819-y