Plasma p-tau217 in Alzheimer’s disease: Lumipulse and ALZpath SIMOA head-to-head comparison ============================================================================================= * Andrea Pilotto * Virginia Quaresima * Chiara Trasciatti * Chiara Tolassi * Diego Bertoli * Cristina Mordenti * Alice Galli * Andrea Rizzardi * Salvatore Caratozzolo * Andrea Zancanaro * José Contador * Oskar Hansson * Sebastian Palmqvist * Giovanni De Santis * Henrik Zetterberg * Kaj Blennow * Duilio Brugnoni * Marc Suárez-Calvet * Nicholas J Ashton * Alessandro Padovani ## ABSTRACT **Background** Plasma phosphorylated-tau217 (p-tau217) has been shown to be one of the most accurate diagnostic markers for Alzheimer’s disease (AD). No studies have compared the clinical performance of p-tau217 as assessed by the fully automated Lumipulse and SIMOA ALZpath p-tau217. **Aim** To evaluate the diagnostic accuracy of Lumipulse and SIMOA plasma p-tau217 assays for AD. **Methods** The study included 392 participants, 162 with AD, 70 with other neurodegenerative diseases (NDD) with CSF biomarkers and 160 healthy controls. Plasma p-tau217 levels were measured using the Lumipulse and ALZpath SIMOA assays. The ability of p-tau217 assessed by both techniques to discriminate AD from NDD and controls was investigated using ROC analyses. **Results** Both techniques showed high internal consistency of p-tau217 with similar correlation with CSF p-tau181 levels. In head-to-head comparison, Lumipulse and SIMOA showed similar diagnostic accuracy for differentiating AD from NDD (area under the curve [AUC] 0.952, 95%CI 0.927–0.978 vs 0.955, 95%CI 0.928–0.982, respectively) and HC (AUC 0.938, 95%CI 0.910-0.966 and 0.937, 95% CI0.907-0.967 for both assays). **Conclusions** This study demonstrated the high precision and diagnostic accuracy of p-tau217 for the clinical diagnosis of Alzheimer’s disease using either fully automated or semi-automated techniques. Keywords * plasma markers * Alzheimer’s disease * p-tau217 * Lumipulse * ALZPath * SIMOA ## INTRODUCTION Cerebrospinal fluid (CSF) biomarkers are informative, sensitive and specific for the detection of Alzheimer’s disease (AD) in clinical and research settings from early stages of the disease.1,2 The recent development of plasma biomarkers is dramatically changing the AD scenario, as they are scalable tools to aid clinical evaluation and trial recruitment.3,4 Phosphorylated tau species (p-tau) stand at the forefront of emerging AD blood tests, exhibiting superior accuracy in diagnosis and specificity for the disease compared to the Aβ42/40 ratio or other suggested biomarkers.5–9 To date, phosphorylated tau at threonine 217 (p-tau217) appeared to be one of the most sensitive and specific AD markers compared to other phosphorylated tau species for differentiating AD from other neurodegenerative disorders.6,10–16 In addition, p-tau217 exhibits a unique longitudinal trajectory in preclinical AD amyloid-positive individuals, with increases over time being significantly associated with worsening cortical atrophy and declining cognitive performance.4,6,13,17,18 Most published studies focusing on p-tau species have used immunoassays on either the Meso Scale Discovery (MSD) or SIMOA platforms.5,10,11,15,19 The recent development of similar assays using chemiluminescent enzyme immunoassay (CLEIA) technology (including the fully automated Lumipulse platform) represents an attractive further step for their easier use and wider consistent applicability in clinical practice. The fully automation produces more consistent results between laboratories, and overtime in the same laboratory. For Lumipulse p-tau217, only one preliminary study suggested a high discrimination accuracy for AD diagnosis, thought without a head-to-head comparison available to date.20 Despite the growing number of unpublished data available, there is an urgent need for high quality technical and clinical validation of newly developed p-tau217 markers. The objective of the study was therefore to compare the diagnostic accuracy performance of Lumipulse vs SIMOA plasma p-tau217 in a large real-world memory clinic scenario with clinically approved CSF AD biomarkers as the reference standard. ## MATERIAL AND METHODS ### Study population The study included participants with mild cognitive impairment (MCI) or mild dementia who underwent CSF assessment at the outpatient Neurodegenerative clinic of the Brescia University Hospital, Italy and matched healthy control subjects. A standardized full cognitive and behavioural assessment, including Mini-Mental State Examination (MMSE), Neuropsychiatric Inventory (NPI) and Clinical Dementia Rating Scale (CDR) was performed in each participant. Each patient underwent lumbar puncture in fasting condition according to the standardized protocol of the outpatient Neurodegenerative clinic. The CSF specimens were collected in 15 mL polypropylene sterile tubes, gently mixed to avoid gradient effects and sent directly to the hospital laboratory for routine assessments and Lumipulse CSF core AD markers.21 Patients with cognitive impairment or dementia were further classified in AD vs neurodegenerative disorders (NDD) according to the Internal cut-off value of Lumipulse Aβ42/p-tau181 ratio >11.1.22,23 A group of neurologically and cognitively normal individuals (healthy controls, HC) were recruited from participants’ caregivers and was included as reference group for plasma analyses. The study was approved by the local ethics committee (NP 1471, DMA, Brescia) and was performed in conformity with the Helsinki Declaration; informed consent was obtained from each study participant or their legally authorized representative. ### Plasma collection and analysis Blood samples were collected from each participant using 7.5 mL tubes containing K2-ethylenediaminetetraacetic acid (K2-EDTA). The tubes were gently inverted 5 to 10 times to mix the blood and then centrifuged at 2500×g for 10 minutes at room temperature (RT). Next, 0.5mL plasma aliquots were pipetted into polypropylene cryotubes and stored at ultra-low temperature freezing (ULTF) −80°C. On the day of analysis, the plasma samples were brought to RT (21–23 °C). Following the manufacturer’s instructions, plasma samples were centrifuged at 2000g for 5 minutes. The plasma was then transferred to the instrument cuvettes for testing on Lumipulse using the Lumipulse G p-tau217 - Plasma Immunoreaction Cartridges RUO (for research use only). The commercial ALZpath p-tau217 assay uses a proprietary monoclonal p-tau217 specific capture antibody, an N-terminal detector antibody, and a peptide calibrator.5 It has been validated as a fit-for-purpose assay24 with a limit of detection of 0.0052 to 0.0074 pg/mL, a functional lower limit of quantification of 0.06 pg/mL, and a dynamic range of 0.007 to 30 pg/mL. The spike recovery for the endogenous analyte was 80%, and intrarun and interrun precision was 0.5% to 13% and 9.2% to 15.7%, respectively. SIMOA analyses were performed on HD-X with commercially available p-tau217 ALZpath Simoa pTau-217 V2 Kits (Quanterix) at the Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.5 ### Statistical and precision analyses The study investigated the within-lab precision of the Lumipulse plasma Immunoreaction Cartridges RUO through repeated inter-day testing schemes 3X5 and 5×5. For the 3×5 testing, three plasma aliquots from a healthy control and three plasma aliquots from an AD patient were used. Two commercial QC samples, namely the high and the low-level controls provided by the company, were tested five times a day for five days. The Lumipulse testing precision and between-day repeatability has been assessed in 15 and 25 runs, respectively. The calculation of the intermediate precision and the between-run precision were calculated following the CLSI EP15.25 The 15 independent plasma samples were stored at −80°C during the 5 days of the assessments. The 25 commercial QC of the p-tau217 kit were kept at −20°C as per the manufacturer’s instructions. Normality distribution was evaluated using the Shapiro-Wilk test and Q-Q plots, outliers were defined based on single values higher/lower than 3 standard deviations compared to the mean of the group. To compare clinical and demographic characteristics as well as cognitive assessments and CSF and plasma markers between diagnostic groups (AD, HC, NDD), Kruskal-Wallis test was conducted. Post-hoc comparisons between AD, HC and NDD was performed using the Mann-Whitney U test. The comparability between the two analytical platforms was assessed using Passing-Bablok regression, while their imprecision was assessed by calculating the laboratory’s coefficient of variation (CV). The association between plasma and CSF biomarkers was determined using Pearson’s correlation coefficient within a correlation matrix. The accuracy in discriminating between AD and NDD/HC using plasma biomarkers, in terms of specificity and sensibility, was assessed using a receiver operating characteristic (ROC) approach. Area under the ROC curve (AUCs) were computed using the pROC package in R. The same statistical analyses were performed only considering AD-MCI and NDD-MCI subgroups (i.e. CDR <1). All analyses were conducted using R Statistical Software ([https://www.r-project.org/](https://www.r-project.org/)). Statistical significance was defined at α=0.05, and all tests were two-tailed. ## RESULTS ### Precision and repeatability of p-tau217 Lumipulse G600II testing Fifteen different specimens were aliquoted from plasma samples collected from one AD CSF-confirmed patient (Positive control, PC) and a healthy control subject (Negative control, NC), both tested as independent samples to perform the between-day repeatability and calculate the testing precision. The Clinical Laboratory’s CV and the between-run CV(%) for PC and NC for p-tau217 resulted in 2.340 and 1.310 for the PC, 3.749 and 2.280 for the NC, respectively (Suppl. Tables 1,2 and 5). Likewise, commercial Quality controls (QC) materials were also used (Suppl. Table 3 and 4). The commercial samples resulted in a CV within laboratory and between run of 5.080 and 5.340, for the L1, and 3.387 and 3.490 for the L2, respectively (Suppl. Table 5). ### Clinical validation and SIMOA head-to-head comparison The clinical study included 392 subjects, namely 232 patients and 160 controls. The clinical assessment and CSF AD markers allowed the classification of patients in 162 AD (of which 112 had MCI) and 70 other neurodegenerative disorders (of which 45 had MCI). No outliers were detected and all SIMOA and Lumipulse values were included in the final analyses. In the whole cohort and AD/NDD/HC subgroups, no correlations between age and p-tau217 levels (tested by Lumipulse and SIMOA) were detected. Clinical and demographics data and CSF core biomarkers are indicated in Table 1. P-tau217 values showed a constant, systematic and proportional error between the two detection methods as highlighted by the Passing-Bablok regression (Figure 1). The intercept is 0.067 (95%CI 0.046 – 0.084) and the slope=1.552 (95%CI 1.433 – 1.703). AD showed higher levels of plasma p-tau217 assessed with both techniques compared to both NDD and HC (Table 1 and Figure 1). The correlation analyses demonstrated a positive relationship between plasma p-tau217 analysed by Lumipulse testing and CSF p-tau181 and t-tau (respectively r=0.578, p<0.001 and r=0.431, p<0.001). A similar positive correlation was found for plasma p-tau217 tested by SIMOA (r=0.595, p<0.001 and r=0.659, p<0.001), being p-tau217 Lumipulse/SIMOA levels highly correlated (r=0.830, p<0.001). ![Figure1](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/04/2024.05.02.24306780/F1.medium.gif) [Figure1](http://medrxiv.org/content/early/2024/05/04/2024.05.02.24306780/F1) View this table: [Table 1](http://medrxiv.org/content/early/2024/05/04/2024.05.02.24306780/T1) Table 1 Participants’ characteristics and Plasma biomarkers assessed by Lumipulse and SIMOA platforms. ### Discriminant analyses for AD diagnosis The discrimination accuracy of plasma biomarkers analysed with Lumipulse and SIMOA techniques for the diagnosis of AD with respect to both HC and NDD was separately evaluated using AUC-ROC analysis (Figure 2). Plasma p-tau217 analysed on the Lumipulse system, resulted in an AUC for AD vs NDD of 0.952 (95%CI 0.927–0.978) and 0.938 (95%CI 0.910–0.966) vs HC. Plasma p-tau217 tested on SIMOA yielded similar diagnostic accuracy, with an AUC of 0.955 (95%CI 0.928–0.982) for the discrimination of AD from NDD and 0.937 (95%CI 0.907–0.967) from HC. The calculated best cutoffs (i.e highest Youden index) for AD vs HC and AD vs NDD were 0.291 pg/mL and 0.276 pg/mL (Figure 2), respectively, for Lumipulse. The computed best cutoffs considering p-tau217 levels in SIMOA, for AD vs HC and AD vs NDD were 0.542 pg/ml and 0.518 pg/ml, respectively, (highest Youden index). ![Figure2](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/04/2024.05.02.24306780/F2.medium.gif) [Figure2](http://medrxiv.org/content/early/2024/05/04/2024.05.02.24306780/F2) In the MCI subset, including 112 AD-MCI and 45 NDD-MCI, the AUC and the cutoffs were similar to the whole cohort (Suppl. Tables 6 and 7). Specifically, Lumipulse p-tau217 yielded an AUC of 0.946 (95%CI 0.911–0.981) for the discrimination between AD-MCI and NDD-MCI and of 0.960 (95%CI 0.936–0.985) for the differentiation with HC. SIMOA ALZpath p-tau217 exhibited a similar accuracy with AUCs of 0.934 (95%CI 0.893–0.976) vs NDD-MCI and 0.960 (95%CI 0.936–0.985) for HC (Suppl. Table 7). ## DISCUSSION This study demonstrated the excellent clinical accuracy of plasma p-tau217 for AD detected using Lumipulse and SIMOA techniques. These suggest that both techniques are valid, solid and comparable alternatives for the assessment of plasma p-tau217 levels, potentially broadening the accessibility of this biomarker in clinical settings. The technical validation of Lumipulse p-tau217 assessment showed a CV within laboratory around 5% for p-tau217 lower levels, otherwise below 3.5% for higher level plasma samples. These values are in line with the precision levels observed for both SIMOA and MSD techniques.5,15 The method comparison analysis (Passing-Bablok) showed that the two testing platforms identified different but highly related p-tau217 concentrations. Therefore, two distinct cut-offs (or conversion methods) for p-tau217 are required for Lumipulse and SIMOA techniques. This is consistent with previous data evaluating p-tau181 assays across techniques in clinical settings.21 When applied in a clinical setting, the p-tau217 plasma assay confirmed its high biological validity, with a high discrimination accuracy of more than 93% for AD compared to other CSF-confirmed neurodegenerative diseases and age-matched healthy controls. These results are consistent with the greater fold change of p-tau217 compared to other p-tau species, namely p-tau231 and p-tau181 recently demonstrated.4,5,10–16 Of note, the cutoffs resulting in the highest Youden index in the ROC analyses for discriminating AD versus NDD and controls resulted in very similar cutoff values across assays, suggesting the possible adoption of a single value for AD diagnosis, ideally to be established by multi-centre validation studies. The head-to-head comparison with ALZpath p-tau217 showed a slightly higher fold-change for Lumipulse with a comparable discrimination accuracy compared to SIMOA, even in the subcohort of MCI subjects at earlier stages of the disease. The strong correlation between plasma p-tau217 and CSF p-tau181 standard levels further supports its utility as a non-invasive alternative for diagnosis and monitoring of Alzheimer’s disease, potentially limiting CSF analysis to a subset of subjects with borderline levels.26 Noteworthy, the study included subjects with different diseases and ages not accounting on *a priori* selection, thus confirming the broad applicability of such techniques in real-life settings. Nevertheless, further technical validations of the testing methods are warranted to challenge the stability of biomarkers *in vivo* in different settings, as testing immediately after −80°C storage is not always available. This is particularly important when considering the transition from research to clinical use of such assays.21,27,28 While our study demonstrates high concordance between the Lumipulse and SIMOA techniques, further validation efforts are warranted to confirm the biological relevance of plasma p-tau217 as a reliable biomarker for Alzheimer’s disease in different patient populations and disease stages, even using different standard-of-truth methods. Future research should focus on addressing the remaining validation gaps by using predefined cut-offs values and optimising the clinical utility of plasma p-tau217 assays. Furthermore, longitudinal studies are needed to establish the stability over short and long-term time window and the prognostic value of plasma p-tau217 in predicting disease progression and treatment response in AD patients, even in combination with other existing plasma biomarkers. In addition, efforts should be made to standardise assay protocols and establish reference ranges for plasma p-tau217 levels to facilitate its integration into routine clinical practice for early detection and monitoring of AD continuum. In conclusion, our study adds to the growing body of evidence supporting the utility of plasma p-tau217 as a reliable biomarker for the diagnosis of Alzheimer’s disease. The validation of Lumipulse p-tau217 highlights its potential to complement existing diagnostic approaches and improve the accuracy of AD detection in clinical practice. ## Supporting information Supplementary tables 1-7 [[supplements/306780_file04.docx]](pending:yes) ## DATA AVAILABILITY The datasets used and analyzed during the current study are available from the corresponding author on reasonable request. ## FUNDING The research is supported by Airalzh Foundation AGYR2021 Life-Bio Grant, the Italian Ministry of University and Research PRIN COCOON (2017MYJ5TH) and PRIN 2021 RePlast (20202THZAW), the Italian Ministry of Health, Grant/Award Number: RF-2018-12366209 and PNRR-Health PNRR-MAD-2022-12376110. ### Other fundings APi has been supported by grants of Airalzh Foundation AGYR2021 Life-Bio Grant, The LIMPE-DISMOV Foundation Segala Grant 2021, the Italian Ministry of University and Research PRIN COCOON (2017MYJ5TH) and PRIN 2021 RePlast (20202THZAW), the H2020 IMI IDEA-FAST (ID853981), Italian Ministry of Health, Grant/Award Number: RF-2018-12366209 and PNRR-Health PNRR-MAD-2022-12376110. VQ is supported by the H2020 IMI IDEA-FAST (ID853981) CTra is supported by the Italian Ministry of University and Research CTo is supported by the Ministry of Health PRIN 2021 RePlast. AG is supported by the Ministry of Health PRIN 2021 RePlast and the H2020 IMI IDEA-FAST (ID853981). AR is supported by the H2020 IMI IDEA-FAST (ID853981) and the PNRR-Health PNRR-MAD-2022-12376110. O.H. was supported by the European Research Council (ADG-101096455), Alzheimer’s Association (ZEN24-1069572, SG-23-1061717), GHR Foundation, Swedish Research Council (2022-00775), ERA PerMed (ERAPERMED2021-184), Knut and Alice Wallenberg foundation (2022-0231), Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, Swedish Alzheimer Foundation (AF-980907), Swedish Brain Foundation (FO2021-0293), Parkinson foundation of Sweden (1412/22), Cure Alzheimer’s fund, Rönström Family Foundation, Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2022-1259) and Swedish federal government under the ALF agreement (2022-Projekt0080). SP is supported by the National Institute of Aging (USA; #R01AG083740), the Swedish Research Council (#2018-02052), the Swedish Alzheimer Foundation (#AF-994075), the Swedish Brain Foundation (#FO2022-0204), and the Family Rönström’s Foundation (#FRS-0004). HZ is a Wallenberg Scholar and a Distinguished Professor at the Swedish Research Council supported by grants from the Swedish Research Council (#2023-00356; #2022-01018 and #2019-02397), the European Union’s Horizon Europe research and innovation program under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, #ADSF-21-831377-C, and #ADSF-24-1284328-C), the Bluefield Project, Cure Alzheimer’s Fund, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), the National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL (UKDRI-1003). Kaj Blennov is supported by the Swedish Research Council (2017-00915 and 2022-00732), Swedish Alzheimer Foundation (AF-930351, AF-939721, and AF-968270), Hjärnfonden, Sweden (FO2017-0243 and ALZ2022-0006), Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement (ALFGBG-715986 and ALFGBG-965240), European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236), Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495), and Alzheimer’s Association 2022-2025 grant (SG-23-1038904 QC). Dr Zetterberg is a Wallenberg Scholar supported by grants from the Swedish Research Council (2022-01018 and 2019-02397), European Union’s Horizon Europe research and innovation programme (grant agreement 101053962), Swedish State Support for Clinical Research (ALFGBG-71320), Alzheimer Drug Discovery Foundation (201809-2016862), AD Strategic Fund, and Alzheimer’s Association (ADSF-21-831376-C, ADSF-21-831381-C, and ADSF-21-831377-C), Bluefield Project, Olav Thon Foundation, Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (FO2022-0270), European Union’s Horizon 2020 research and innovation programme (Marie Skłodowska-Curie grant agreement 860197, MIRIADE), European Union Joint Programme– Neurodegenerative Disease Research (JPND2021-00694), National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and UK Dementia Research Institute at UCL (UKDRI-1003). The Wisconsin Registry for Alzheimer’s Prevention is supported by NIH grants AG027161 and AG021155. MSC receives funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (Grant agreement No. 948677); ERA PerMed (ERAPERMED2021-184); Project “PI19/00155” and “PI22/00456, funded by Instituto de Salud Carlos III (ISCIII) and co-funded by the European Union; and from a fellowship from “la Caixa” Foundation (ID 100010434) and from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 847648 (LCF/BQ/PR21/11840004). APa has been supported by grants of the Italian Ministry of University and Research PRIN COCOON (2017MYJ5TH) and PRIN 2021 RePlast (20202THZAW), the H2020 IMI IDEA-FAST (ID853981), Italian Ministry of Health, Grant/Award Number: RF-2018-12366209, RF-2019-12369272 and PNRR-Health PNRR-MAD-2022-12376110. ## COMPETING INTEREST APi received consultancy/speaker fees from Abbvie, Bial, Lundbeck, Roche and Zambon pharmaceuticals. OH has acquired research support (for the institution) from AVID Radiopharmaceuticals, Biogen, C2N Diagnostics, Eli Lilly, Eisai, Fujirebio, GE Healthcare, and Roche. In the past 2 years, he has received consultancy/speaker fees from AC Immune, Alzpath, BioArctic, Biogen, Bristol Meyer Squibb, Cerveau, Eisai, Eli Lilly, Fujirebio, Merck, Novartis, Novo Nordisk, Roche, Sanofi and Siemens. SP has acquired research support (for the institution) from ki elements / ADDF and Avid. In the past 2 years, he has received consultancy/speaker fees from Bioartic, Biogen, Esai, Lilly, and Roche. HZ has served at scientific advisory boards and/or as a consultant for Abbvie, Acumen, Alector, Alzinova, ALZPath, Amylyx, Annexon, Apellis, Artery Therapeutics, AZTherapies, Cognito Therapeutics, CogRx, Denali, Eisai, Merry Life, Nervgen, Novo Nordisk, Optoceutics, Passage Bio, Pinteon Therapeutics, Prothena, Red Abbey Labs, reMYND, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures in symposia sponsored by Alzecure, Biogen, Cellectricon, Fujirebio, Lilly, and Roche, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). KB reported having served as a consultant and at advisory boards for Acumen, ALZpath, BioArctic, Biogen, Eisai, Lilly, Moleac, Novartis, Ono Pharma, Prothena, Roche Diagnostics, and Siemens Healthineers; having served at data monitoring committees for Julius Clinical and Novartis; having given lectures, produced educational materials, and participated in educational programs for AC Immune, Biogen, Celdara Medical, Eisai, and Roche Diagnostics; and being a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. MSC has given lectures in symposia sponsored by Almirall, Eli Lilly, Novo Nordisk, Roche Diagnostics, and Roche Farma; received consultancy fees (paid to the institution) from Roche Diagnostics; and served on advisory boards of Roche Diagnostics and Grifols. He was granted a project and is a site investigator of a clinical trial (funded to the institution) by Roche Diagnostics. In-kind support for research (to the institution) was received from ADx Neurosciences, Alamar Biosciences, Avid Radiopharmaceuticals, Eli Lilly, Fujirebio, Janssen Research & Development, and Roche Diagnostics. NJA has received consultancy/speaker fees from Bioartic, Biogen, Lilly, Quanterix and Alamar Biosciences. APa received grant support from Ministry of Health (MINSAL) and Ministry of Education, Research and University (MIUR), from CARIPLO Foundation; personal compensation as a consultant/scientific advisory board member for Biogen, Lundbeck, Roche, Nutricia, General Healthcare (GE). ## AUTHORS’ CONTRIBUTION Api, VQ: contributed to design the work, acquisition, analysis, interpretation of data. CTo, MP, DB, CM, AG, AR, AZ: data acquisition and analysis. CTr, VQ: data analysis and statistics. APa, DBr: have drafted the work and substantively revised it. SC, AB, KB, HZ, SG, NJA, MSC, JC, OH, SP, GDS: revised and approved the submitted version. ## SUPPLEMENTARY MATERIAL Supplementary material include 6 tables and 3 figures. View this table: [Table 1](http://medrxiv.org/content/early/2024/05/04/2024.05.02.24306780/T2) Table 1 Diagnostic accuracy of Lumipulse (L) and Simoa (S) plasma p-tau217 ## ACKNOWLEDGEMENTS The authors wish to thank the study participants who took part in this research. We are very grateful for the assistance received from all the nurses of the Neurology department of the Spedali Civili of Brescia. We want to thank especially the laboratory technicians of the Central Clinical Laboratory for their amazing availability. ## Footnotes * updated corrected author list (in line wih manuscript) * Received May 2, 2024. * Revision received May 4, 2024. * Accepted May 4, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## REFERENCES 1. 1.Leuzy A, Cullen NC, Mattsson-Carlgren N, Hansson O. Current advances in plasma and cerebrospinal fluid biomarkers in Alzheimer’s disease. Curr Opin Neurol. 2021;34(2):266–274. doi:10.1097/WCO.0000000000000904 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/WCO.0000000000000904&link_type=DOI) 2. 2.Hansson O. Biomarkers for neurodegenerative diseases. Nat Med. 2021;27(6):954–963. doi:10.1038/s41591-021-01382-x [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01382-x&link_type=DOI) 3. 3.Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer’s disease in clinical practice and trials. Nat Aging. 2023;3(5):506–519. doi:10.1038/s43587-023-00403-3 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s43587-023-00403-3&link_type=DOI) 4. 4.Ashton NJ, Janelidze S, Mattsson-Carlgren N, et al. Differential roles of Aβ42/40, p-tau231 and p-tau217 for Alzheimer’s trial selection and disease monitoring. Nat Med. 2022;28(12):2555–2562. doi:10.1038/s41591-022-02074-w [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-02074-w&link_type=DOI) 5. 5.Ashton NJ, Brum WS, Molfetta G Di, et al. Diagnostic Accuracy of a Plasma Phosphorylated Tau 217 Immunoassay for Alzheimer Disease Pathology. JAMA Neurol. 2024;81(3):255–263. doi:10.1001/JAMANEUROL.2023.5319 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/JAMANEUROL.2023.5319&link_type=DOI) 6. 6.Ashton NJ, Puig-Pijoan A, Milà-Alomà M, et al. Plasma and CSF biomarkers in a memory clinic: Head-to-head comparison of phosphorylated tau immunoassays. Alzheimers Dement. 2023;19(5):1913–1924. doi:10.1002/ALZ.12841 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ALZ.12841&link_type=DOI) 7. 7.Therriault J, Ashton NJ, Pola I, et al. Comparison of two plasma p-tau217 assays to detect and monitor Alzheimer’s pathology. EBioMedicine. 2024;102. doi:10.1016/J.EBIOM.2024.105046 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.EBIOM.2024.105046&link_type=DOI) 8. 8.Thijssen EH, La Joie R, Strom A, et al. Association of Plasma P-tau217 and P-tau181 with clinical phenotype, neuropathology, and imaging markers in Alzheimer’s disease and frontotemporal lobar degeneration: a retrospective diagnostic performance study. Lancet Neurol. 2021;20(9):739. doi:10.1016/S1474-4422(21)00214-3 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(21)00214-3&link_type=DOI) 9. 9.Janelidze S, Bali D, Ashton NJ, et al. Head-to-head comparison of 10 plasma phospho-tau assays in prodromal Alzheimer’s disease. Brain. 2023;146(4):1592–1601. doi:10.1093/BRAIN/AWAC333 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/BRAIN/AWAC333&link_type=DOI) 10. 10.Janelidze S, Stomrud E, Smith R, et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat Commun. 2020;11(1). doi:10.1038/s41467-020-15436-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-020-15436-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F04%2F2024.05.02.24306780.atom) 11. 11.Gonzalez-Ortiz F, Ferreira PCL, González-Escalante A, et al. A novel ultrasensitive assay for plasma p-tau217: Performance in individuals with subjective cognitive decline and early Alzheimer’s disease. Alzheimers Dement. 2024;20(2):1239–1249. doi:10.1002/ALZ.13525 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ALZ.13525&link_type=DOI) 12. 12.Karikari TK, Ashton NJ, Brinkmalm G, et al. Blood phospho-tau in Alzheimer disease: analysis, interpretation, and clinical utility. 7:17. doi:10.1038/s41582-022-00665-2 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41582-022-00665-2&link_type=DOI) 13. 13.Mattsson-Carlgren N, Janelidze S, Palmqvist S, et al. Longitudinal plasma p-tau217 is increased in early stages of Alzheimer’s disease. Brain. 2020;143(11):3234–3241. doi:10.1093/BRAIN/AWAA286 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awaa286&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F04%2F2024.05.02.24306780.atom) 14. 14.Mielke MM, Dage JL, Frank RD, et al. Performance of plasma phosphorylated tau 181 and 217 in the community. Nat Med. 2022;28(7):1398. doi:10.1038/S41591-022-01822-2 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/S41591-022-01822-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F04%2F2024.05.02.24306780.atom) 15. 15.Mielke MM, Aakre JA, Algeciras-Schimnich A, et al. Comparison of CSF phosphorylated tau 181 and 217 for cognitive decline. Alzheimers Dement. 2022;18(4):602. doi:10.1002/ALZ.12415 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ALZ.12415&link_type=DOI) 16. 16.Palmqvist S, Janelidze S, Quiroz YT, et al. Discriminative Accuracy of Plasma Phospho-tau217 for Alzheimer Disease vs Other Neurodegenerative Disorders. JAMA. 2020;324(8):772–781. doi:10.1001/JAMA.2020.12134 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/JAMA.2020.12134&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F04%2F2024.05.02.24306780.atom) 17. 17.Jonaitis EM, Janelidze S, Cody KA, et al. Plasma phosphorylated tau 217 in preclinical Alzheimer’s disease. Brain Commun. 2023;5(2). doi:10.1093/BRAINCOMMS/FCAD057 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/BRAINCOMMS/FCAD057&link_type=DOI) 18. 18.Aguillon D, Langella S, Chen Y, et al. Plasma p-tau217 predicts in vivo brain pathology and cognition in autosomal dominant Alzheimer’s disease. Alzheimer’s & Dementia. 2023;19(6):2585–2594. doi:10.1002/ALZ.12906 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ALZ.12906&link_type=DOI) 19. 19.Montoliu-Gaya L, Benedet AL, Tissot C, et al. Mass spectrometric simultaneous quantification of tau species in plasma shows differential associations with amyloid and tau pathologies. Nat Aging. 2023;3(6):661–669. doi:10.1038/s43587-023-00405-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s43587-023-00405-1&link_type=DOI) 20. 20.Arranz J, Zhu N, Rubio-Guerra S, et al. Diagnostic performance of plasma pTau 217, pTau 181, Aβ 1–42 and Aβ 1–40 in the LUMIPULSE automated platform for the detection of Alzheimer disease. Res Sq. Published online December 13, 2023. doi:10.21203/RS.3.RS-3725688/V1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.21203/RS.3.RS-3725688/V1&link_type=DOI) 21. 21.Quaresima V, Pilotto A, Trasciatti C, et al. Plasma P-Tau181 and Amyloid Markers in Alzheimer’s Disease: A Method Comparison between Simoa and Lumipulse. SSRN Electronic Journal. Published online 2024. doi:10.2139/SSRN.4774535 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2139/SSRN.4774535&link_type=DOI) 22. 22.Pilotto A, Parigi M, Bonzi G, et al. Differences Between Plasma and Cerebrospinal Fluid p-tau181 and p-tau231 in Early Alzheimer’s Disease. J Alzheimers Dis. 2022;87(3):991–997. doi:10.3233/JAD-215646 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-215646&link_type=DOI) 23. 23.Jack CR, Bennett DA, Blennow K, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535–562. doi:10.1016/J.JALZ.2018.02.018 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.JALZ.2018.02.018&link_type=DOI) 24. 24.Jani D, Allinson J, Berisha F, et al. Recommendations for Use and Fit-for-Purpose Validation of Biomarker Multiplex Ligand Binding Assays in Drug Development. AAPS J. 2016;18(1):1–14. doi:10.1208/S12248-015-9820-Y [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1208/S12248-015-9820-Y&link_type=DOI) 25. 25.Neill Carey R, Durham FAP, Hauck WW, et al. User Verification of Precision and Estimation of Bias; Approved Guideline-Third Edition. 2014;34:12. Accessed January 30, 2024. [www.clsi.org](http://www.clsi.org). 26. 26.Telser J, Risch L, Saely CH, Grossmann K, Werner P. P-tau217 in Alzheimer’s disease. Clinica Chimica Acta. 2022;531:100–111. doi:10.1016/J.CCA.2022.03.018 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.CCA.2022.03.018&link_type=DOI) 27. 27.Bellomo G, Bayoumy S, Megaro A, et al. Fully automated measurement of plasma Aβ42/40 and p-tau181: Analytical robustness and concordance with cerebrospinal fluid profile along the Alzheimer’s disease continuum in two independent cohorts. Alzheimer’s and Dementia. Published online 2024. doi:10.1002/alz.13687 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/alz.13687&link_type=DOI) 28. 28.Wilson EN, Young CB, Ramos Benitez J, et al. Performance of a fully-automated Lumipulse plasma phospho-tau181 assay for Alzheimer’s disease. Alzheimers Res Ther. 2022;14(1). doi:10.1186/s13195-022-01116-2 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13195-022-01116-2&link_type=DOI)