PT - JOURNAL ARTICLE AU - Ferrari-Souza, João Pedro AU - Brum, Wagner S. AU - Hauschild, Lucas A. AU - Da Ros, Lucas U. AU - Lukasewicz Ferreira, Pâmela C. AU - Bellaver, Bruna AU - Leffa, Douglas T. AU - Bieger, Andrei AU - Tissot, Cécile AU - De Bastiani, Marco Antônio AU - Povala, Guilherme AU - Benedet, Andréa L. AU - Therriault, Joseph AU - Wang, Yi-Ting AU - Ashton, Nicholas J. AU - Zetterberg, Henrik AU - Blennow, Kaj AU - Martins, Sheila O. AU - Souza, Diogo O. AU - Rosa-Neto, Pedro AU - Karikari, Thomas AU - Pascoal, Tharick A. AU - Zimmer, Eduardo R. AU - , TI - Vascular risk burden is a key player in the early progression of Alzheimer’s disease AID - 10.1101/2021.12.18.21267994 DP - 2021 Jan 01 TA - medRxiv PG - 2021.12.18.21267994 4099 - http://medrxiv.org/content/early/2021/12/19/2021.12.18.21267994.short 4100 - http://medrxiv.org/content/early/2021/12/19/2021.12.18.21267994.full AB - Understanding whether vascular risk factors synergistically potentiate Alzheimer’s disease progression is important in the context of emerging treatments for preclinical Alzheimer’s disease. The existence of a synergistic relationship could suggest that the combination of therapies targeting Alzheimer’s disease pathophysiology and vascular risk factors might potentiate treatment outcomes. In the present retrospective cohort study, we tested whether vascular risk factor burden interacts with Alzheimer’s disease pathophysiology to accelerate neurodegeneration and cognitive decline in cognitively unimpaired subjects. We evaluated 503 cognitively unimpaired participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) study. Baseline vascular risk factor burden was calculated considering the history of cardiovascular disease, hypertension, diabetes mellitus, hyperlipidemia, stroke or transient ischemic attack, smoking, atrial fibrillation, and left ventricular hypertrophy. Alzheimer’s disease pathophysiology was evaluated using cerebrospinal fluid (CSF) amyloid-β1-42 (Aβ1-42) reflecting brain amyloidosis (A) and tau phosphorylated at threonine 181 (p-tau181) reflecting brain tau pathology (T). Individuals were dichotomized as having an elevated vascular risk factor burden (V+ if having two or more vascular risk factors) and as presenting preclinical Alzheimer’s disease [(AT)+ if having abnormal CSF p-tau181 and Aβ1-42 levels]. Neurodegeneration was assessed with plasma neurofilament light (NfL) and global cognition with the modified version of the Preclinical Alzheimer’s Cognitive Composite. Linear mixed-effects models revealed that an elevated vascular risk factor burden synergistically interacted with Alzheimer’s disease pathophysiology to drive longitudinal increases in plasma NfL levels (β = 5.08, P = 0.016) and cognitive decline (β = −0.43, P = 0.020). Additionally, we observed that vascular risk factor burden was not associated with CSF Aβ1-42 or p-tau181 changes over time. Survival analysis demonstrated that individuals with preclinical Alzheimer’s disease and elevated vascular risk factor burden [(AT)+V+] had a significantly greater risk of clinical progression to cognitive impairment (adjusted Hazard Ratio = 3.5, P < 0.001). Our results support the notion that vascular risk factor burden and Alzheimer’s disease pathophysiology are independent processes; however, they synergistically lead to neurodegeneration and cognitive decline. These findings can help in providing the blueprints for the combination of vascular risk factor management and Alzheimer’s disease pathophysiology treatment in preclinical stages. Moreover, we observed plasma NfL as a robust marker of disease progression that may be used to track therapeutic response in future trials.Competing Interest StatementH.Z. has served at scientific advisory boards and/or as a consultant for Abbvie, Alector, Annexon, AZTherapies, CogRx, Denali, Eisai, Nervgen, Pinteon Therapeutics, Red Abbey Labs, Passage Bio, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure and Biogen, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. K.B. has served as a consultant, at advisory boards, or at data monitoring committees for Abcam, Axon, Biogen, JOMDD/Shimadzu. Julius Clinical, Lilly, MagQu, Novartis, Prothena, Roche Diagnostics, and Siemens Healthineers, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program, all unrelated to the work presented in this paper. S.O.M. reports receiving speaker fees from Medtronic, Novartis, Novo Nordisk, Pfizer, Bayer and advisory board fees from Boehringer Ingelheim. All other authors declare no competing interests.Funding StatementJ.P.F-S. receives financial support from CAPES (#88887.627297/2021-00). W.S.B is supported by CAPES (#88887.372371/2019-00 and #88887.596742/2020-00). B.B. receives financial support from CAPES (#88887.336490/2019-00). D.T.L. is supported by a NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation (#29486). A.B. receives financial support from CAPES (#88882.345554/2019-01). C.T. receives funding from Faculty of Medicine McGill and IPN McGill. M.A.D.B. receives financial support from CNPq (#150293/2019-4). G.P. receives financial support from CAPES (#88882.345577/2019-01). A.L.B. is supported by the Swedish Alzheimer Foundation, Stiftelsen för Gamla Tjänarinnor, and Stohne Stiftelsen. J.T is supported by the Canadian Institutes of Health Research & McGill Healthy Brains Healthy Lives initiative. H.Z. is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712), Swedish State Support for Clinical Research (#ALFGBG-720931), the Alzheimer Drug Discovery Foundation (#201809-2016862), the AD Strategic Fund and the Alzheimer's Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden (#FO2019-0228), the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 860197 (MIRIADE), and the UK Dementia Research Institute at UCL. K.B. is supported by the Swedish Research Council (#2017-00915), the Alzheimer Drug Discovery Foundation (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden (#FO2017-0243), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986), the European Union Joint Program for Neurodegenerative Disorders (#JPND2019-466-236), the NIH (#1R01AG068398-01), and the Alzheimer's Association 2021 Zenith Award (#ZEN-21-848495). S.O.M. is supported by the Research Grant for Resilient Study (DECIT/CNPq #401821/2015-3), for Trident Study (PROADI-SUS Hospital Moinhos de Vento #NUP 25000.209767/2018-61), for Polypill and Stroke Riskometer for Prevention of Stroke and Cognitive Decline (PROADI-SUS HMV #NUP 25000.013007/2021-55), for Resilient Direct TNK and Extend IV (PROADI-SUS HMV #NUP 25000.0334940/2021-66). D.O.S. is supported by CAPES (#88887.185806/2018-00, #88887.507218/2020-00, and #88887.507161/ 2020-00), CNPQ/INCT (#465671/2014-4), CNPQ/ZIKA (#440763/ 2016-9), CNPQ/FAPERGS/PRONEX (#16/2551- 0000475-7), and FAPERGS (#19/2551-0000700-0). P.R-N. is supported by the Canadian Institutes of Health Research (#MOP-11-51-31), the Alzheimer's Association (#NIRG-12-92090 and #NIRP-12-259245), Fonds de Recherche du Québec - Santé (Chercheur Boursier; #2020-VICO-279314). T.K. is funded by the Swedish Research Council's career establishment fellowship (#2021-03244), the Alzheimer's Association Research Fellowship (#850325), the BrightFocus Foundation (#A2020812F), the International Society for Neurochemistry's Career Development Grant, the Swedish Alzheimer Foundation (Alzheimerfonden; #AF-930627), the Swedish Brain Foundation (Hjärnfonden; #FO2020-0240), the Swedish Dementia Foundation (Demensförbundet), the Swedish Parkinson Foundation (Parkinsonfonden), Gamla Tjänarinnor Foundation, the Aina (Ann) Wallströms and Mary-Ann Sjöbloms Foundation, the Agneta Prytz-Folkes & Gösta Folkes Foundation (#2020-00124), the Gun and Bertil Stohnes Foundation, and the Anna Lisa and Brother Björnsson's Foundation. T.A.P. is supported by the NIH (#R01AG075336 and #R01AG073267) and the Alzheimer's Association (#AACSF-20-648075). E.R.Z. receives financial support from CNPq (#435642/2018-9 and #312410/2018- 2), Instituto Serrapilheira (#Serra-1912-31365), Brazilian National Institute of Science and Technology in Excitotoxicity and Neuroprotection (#465671/2014-4), FAPERGS/MS/CNPq/SESRS-PPSUS (#30786.434.24734.231120170), and ARD/FAPERGS (#54392.632.30451.05032021).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Data used in the preparation of the present manuscript is publicly available for download at http://adni.loni.usc.edu.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesData used in the preparation of the present manuscript is publicly available for download at http://adni.loni.usc.edu. If needed for replication purposes, derived data used in our analysis will be made available upon reasonable request.ADAS-CogAlzheimer’s Disease Assessment Scale – Cognitive SubscaleADNIAlzheimer’s Disease Neuroimaging InitiativeAPOE ε4Apolipoprotein E ε4Aβ1-42amyloid-β1-42BBBblood-brain barrierBIOCARDBiomarkers for Older Controls at Risk for Dementia(CDR-SB)clinical dementia rating scale sum of boxesCUcognitively unimpairedFINGERFinnish Geriatric Intervention Study to Prevent Cognitive Impairment and DisabilityHABSHarvard Aging Brain StudyHRHazard RatioLMElinear mixed-effectsMCImild cognitive impairmentMMSEMini-Mental State ExaminationmPACCmodified version of Preclinical Alzheimer’s Cognitive CompositeNfLneurofilament lightNIA-AANational Institute on Aging and the Alzheimer’s Associationp-tau181tau phosphorylated at threonine 181SDstandard deviationSimoasingle molecule arrayt-tautotal tauTIAtransient ischemic attackVRFvascular risk factorVIFvariance inflation factorWMHwhite matter hyperintensities.