scispace - formally typeset
Search or ask a question

Showing papers by "Giovanni B. Frisoni published in 2019"


Journal ArticleDOI
Rosalinde E.R. Slot1, Sietske A.M. Sikkes1, Johannes Berkhof1, Henry Brodaty2, Rachel F. Buckley3, Rachel F. Buckley4, Enrica Cavedo, Efthimios Dardiotis5, Francoise Guillo-Benarous6, Harald Hampel, Nicole A. Kochan2, Simone Lista, Tobias Luck7, Paul Maruff, José Luis Molinuevo, Johannes Kornhuber8, Barry Reisberg6, Steffi G. Riedel-Heller7, Shannon L. Risacher9, Susanne Roehr7, Perminder S. Sachdev2, Nikolaos Scarmeas10, Nikolaos Scarmeas11, Philip Scheltens1, Melanie B. Shulman6, Andrew J. Saykin9, Sander C.J. Verfaillie1, Pieter Jelle Visser1, Pieter Jelle Visser12, Stephanie J.B. Vos12, Michael Wagner13, Michael Wagner14, Steffen Wolfsgruber13, Steffen Wolfsgruber14, Frank Jessen15, Frank Jessen13, Mercè Boada, Peter Paul De Deyn, Roy W. Jones, Giovanni B. Frisoni, L. Spiru1, Flavio Nobili, Yvonne Freund-Levi, Hilkka Soininen, Frans R.J. Verhey, Åsa K. Wallin, Jacques Touchon, Marcel G. M. Olde Rikkert, Anne-Sophie Rigaud, Roger Bullock, Magda Tsolaki, Bruno Vellas, Gordon K. Wilcock, Lutz Froelich, Hovagim Bakardjian, Habib Benali, Hugo Bertin, Joel Bonheur, Laurie Boukadida, Nadia Boukerrou, Patrizia A. Chiesa, Olivier Colliot, Bruno Dubois, Marion Dubois, Stéphane Epelbaum, Geoffroy Gagliardi, Remy Genthon, M.-O. Habert1, Marion Houot, Aurélie Kas, Foudil Lamari, Marcel Levy, Christiane Metzinger, Fanny Mochel, Francis Nyasse, Catherine Poisson, Marie-Claude Potier, Marie Revillon, Antonio Carlos dos Santos, Katia Andrade, Marine Sole, Mohmed Surtee, Michel Thiebaud de Schotten, Andrea Vergallo, Nadjia Younsi, Wiesje M. van der Flier1 
TL;DR: The incidence of Alzheimer's disease and non‐AD dementia and determinants of progression to dementia are assessed and subjective cognitive decline in community‐based and memory clinic settings is assessed.
Abstract: Introduction In this multicenter study on subjective cognitive decline (SCD) in community-based and memory clinic settings, we assessed the (1) incidence of Alzheimer's disease (AD) and non-AD dementia and (2) determinants of progression to dementia. Methods Eleven cohorts provided 2978 participants with SCD and 1391 controls. We estimated dementia incidence and identified risk factors using Cox proportional hazards models. Results In SCD, incidence of dementia was 17.7 (95% Poisson confidence interval 15.2-20.3)/1000 person-years (AD: 11.5 [9.6-13.7], non-AD: 6.1 [4.7-7.7]), compared with 14.2 (11.3-17.6) in controls (AD: 10.1 [7.7-13.0], non-AD: 4.1 [2.6-6.0]). The risk of dementia was strongly increased in SCD in a memory clinic setting but less so in a community-based setting. In addition, higher age (hazard ratio 1.1 [95% confidence interval 1.1-1.1]), lower Mini–Mental State Examination (0.7 [0.66-0.8]), and apolipoprotein E e4 (1.8 [1.3-2.5]) increased the risk of dementia. Discussion SCD can precede both AD and non-AD dementia. Despite their younger age, individuals with SCD in a memory clinic setting have a higher risk of dementia than those in community-based cohorts.

215 citations


Journal ArticleDOI
TL;DR: Plasma biomarkers for Alzheimer's disease diagnosis/stratification are a “Holy Grail” of AD research and intensively sought; however, there are no well‐established plasma markers.
Abstract: Introduction Plasma biomarkers for Alzheimer's disease (AD) diagnosis/stratification are a "Holy Grail" of AD research and intensively sought; however, there are no well-established plasma markers. Methods A hypothesis-led plasma biomarker search was conducted in the context of international multicenter studies. The discovery phase measured 53 inflammatory proteins in elderly control (CTL; 259), mild cognitive impairment (MCI; 199), and AD (262) subjects from AddNeuroMed. Results Ten analytes showed significant intergroup differences. Logistic regression identified five (FB, FH, sCR1, MCP-1, eotaxin-1) that, age/APOe4 adjusted, optimally differentiated AD and CTL (AUC: 0.79), and three (sCR1, MCP-1, eotaxin-1) that optimally differentiated AD and MCI (AUC: 0.74). These models replicated in an independent cohort (EMIF; AUC 0.81 and 0.67). Two analytes (FB, FH) plus age predicted MCI progression to AD (AUC: 0.71). Discussion Plasma markers of inflammation and complement dysregulation support diagnosis and outcome prediction in AD and MCI. Further replication is needed before clinical translation.

122 citations


Journal ArticleDOI
TL;DR: The findings show the value of blood NfL as a disease progression biomarker in genetic frontotemporal dementia and suggest that longitudinal N fL measurements could identify mutation carriers approaching symptom onset and capture rates of brain atrophy.
Abstract: Summary Background Neurofilament light chain (NfL) is a promising blood biomarker in genetic frontotemporal dementia, with elevated concentrations in symptomatic carriers of mutations in GRN, C9orf72, and MAPT. A better understanding of NfL dynamics is essential for upcoming therapeutic trials. We aimed to study longitudinal NfL trajectories in people with presymptomatic and symptomatic genetic frontotemporal dementia. Methods We recruited participants from 14 centres collaborating in the Genetic Frontotemporal Dementia Initiative (GENFI), which is a multicentre cohort study of families with genetic frontotemporal dementia done across Europe and Canada. Eligible participants (aged ≥18 years) either had frontotemporal dementia due to a pathogenic mutation in GRN, C9orf72, or MAPT (symptomatic mutation carriers) or were healthy at-risk first-degree relatives (either presymptomatic mutation carriers or non-carriers), and had at least two serum samples with a time interval of 6 months or more. Participants were excluded if they had neurological comorbidities that were likely to affect NfL, including cerebrovascular events. We measured NfL longitudinally in serum samples collected between June 8, 2012, and Dec 8, 2017, through follow-up visits annually or every 2 years, which also included MRI and neuropsychological assessments. Using mixed-effects models, we analysed NfL changes over time and correlated them with longitudinal imaging and clinical parameters, controlling for age, sex, and study site. The primary outcome was the course of NfL over time in the various stages of genetic frontotemporal dementia. Findings We included 59 symptomatic carriers and 149 presymptomatic carriers of a mutation in GRN, C9orf72, or MAPT, and 127 non-carriers. Nine presymptomatic carriers became symptomatic during follow-up (so-called converters). Baseline NfL was elevated in symptomatic carriers (median 52 pg/mL [IQR 24–69]) compared with presymptomatic carriers (9 pg/mL [6–13]; p Interpretation Our findings show the value of blood NfL as a disease progression biomarker in genetic frontotemporal dementia and suggest that longitudinal NfL measurements could identify mutation carriers approaching symptom onset and capture rates of brain atrophy. The characterisation of NfL over the course of disease provides valuable information for its use as a treatment effect marker. Funding ZonMw and the Bluefield project.

121 citations


Journal ArticleDOI
TL;DR: The proposed formulation of DPM provides a statistical reference for the accurate probabilistic assessment of the pathological stage of de‐novo individuals, and represents a valuable instrument for quantifying the variability and the diagnostic value of biomarkers across disease stages.

97 citations


Journal ArticleDOI
TL;DR: It is proposed that AD could be tackled not only using combination therapies including Aβ and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood–brain barrier targets that have been studied recently.
Abstract: Alzheimer's disease (AD), the most frequent cause of dementia, is escalating as a global epidemic, and so far, there is neither cure nor treatment to alter its progression. The most important feature of the disease is neuronal death and loss of cognitive functions, caused probably from several pathological processes in the brain. The main neuropathological features of AD are widely described as amyloid beta (Aβ) plaques and neurofibrillary tangles of the aggregated protein tau, which contribute to the disease. Nevertheless, AD brains suffer from a variety of alterations in function, such as energy metabolism, inflammation and synaptic activity. The latest decades have seen an explosion of genes and molecules that can be employed as targets aiming to improve brain physiology, which can result in preventive strategies for AD. Moreover, therapeutics using these targets can help AD brains to sustain function during the development of AD pathology. Here, we review broadly recent information for potential targets that can modify AD through diverse pharmacological and nonpharmacological approaches including gene therapy. We propose that AD could be tackled not only using combination therapies including Aβ and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood-brain barrier targets that have been studied recently. We also make a case for the role of gut microbiota in AD. Our hope is to promote the continuing research of diverse targets affecting AD and promote diverse targeting as a near-future strategy.

90 citations


Journal ArticleDOI
TL;DR: The models generated could aid clinicians in the interpretation of CSF biomarker and hippocampal volume results in individuals with MCI, and help research and clinical settings to prepare for a future of precision medicine in Alzheimer's disease.
Abstract: Summary Background Biomarker-based risk predictions of dementia in people with mild cognitive impairment are highly relevant for care planning and to select patients for treatment when disease-modifying drugs become available. We aimed to establish robust prediction models of disease progression in people at risk of dementia. Methods In this modelling study, we included people with mild cognitive impairment (MCI) from single-centre and multicentre cohorts in Europe and North America: the European Medical Information Framework for Alzheimer's Disease (EMIF-AD; n=883), Alzheimer's Disease Neuroimaging Initiative (ADNI; n=829), Amsterdam Dementia Cohort (ADC; n=666), and the Swedish BioFINDER study (n=233). Inclusion criteria were a baseline diagnosis of MCI, at least 6 months of follow-up, and availability of a baseline Mini-Mental State Examination (MMSE) and MRI or CSF biomarker assessment. The primary endpoint was clinical progression to any type of dementia. We evaluated performance of previously developed risk prediction models—a demographics model, a hippocampal volume model, and a CSF biomarkers model—by evaluating them across cohorts, incorporating different biomarker measurement methods, and determining prognostic performance with Harrell's C statistic. We then updated the models by re-estimating parameters with and without centre-specific effects and evaluated model calibration by comparing observed and expected survival. Finally, we constructed a model combining markers for amyloid deposition, tauopathy, and neurodegeneration (ATN), in accordance with the National Institute on Aging and Alzheimer's Association research framework. Findings We included all 2611 individuals with MCI in the four cohorts, 1007 (39%) of whom progressed to dementia. The validated demographics model (Harrell's C 0·62, 95% CI 0·59–0·65), validated hippocampal volume model (0·67, 0·62–0·72), and updated CSF biomarkers model (0·72, 0·68–0·74) had adequate prognostic performance across cohorts and were well calibrated. The newly constructed ATN model had the highest performance (0·74, 0·71–0·76). Interpretation We generated risk models that are robust across cohorts, which adds to their potential clinical applicability. The models could aid clinicians in the interpretation of CSF biomarker and hippocampal volume results in individuals with MCI, and help research and clinical settings to prepare for a future of precision medicine in Alzheimer's disease. Future research should focus on the clinical utility of the models, particularly if their use affects participants' understanding, emotional wellbeing, and behaviour. Funding ZonMW-Memorabel.

81 citations


Journal ArticleDOI
TL;DR: This work investigated relations between amyloid‐β status, apolipoprotein E (APOE) ε4, and cognition, with cerebrospinal fluid markers of neurogranin (Ng), neurofilament light (NFL), YKL‐40, and total tau (T‐tau).
Abstract: Introduction We investigated relations between amyloid-β (Aβ) status, apolipoprotein E (APOE) e4, and cognition, with cerebrospinal fluid markers of neurogranin (Ng), neurofilament light (NFL), YKL-40, and total tau (T-tau). Methods We included 770 individuals with normal cognition, mild cognitive impairment, and Alzheimer's disease (AD)-type dementia from the EMIF-AD Multimodal Biomarker Discovery study. We tested the association of Ng, NFL, YKL-40, and T-tau with Aβ status (Aβ− vs. Aβ+), clinical diagnosis APOE e4 carriership, baseline cognition, and change in cognition. Results Ng and T-tau distinguished between Aβ+ from Aβ− individuals in each clinical group, whereas NFL and YKL-40 were associated with Aβ+ in nondemented individuals only. APOE e4 carriership did not influence NFL, Ng, and YKL-40 in Aβ+ individuals. NFL was the best predictor of cognitive decline in Aβ+ individuals across the cognitive spectrum. Discussion Axonal degeneration, synaptic dysfunction, astroglial activation, and altered tau metabolism are involved already in preclinical AD. NFL may be a useful prognostic marker.

78 citations


Journal ArticleDOI
TL;DR: Blood levels of the candidate microRNAs investigated for association to AD conversion within 2 years in a group of 45 patients with MCI support a role for miR-146a and mi-181a in the mechanisms of AD progression.

63 citations


Journal ArticleDOI
TL;DR: This work sets out to test the performance of metabolites in blood to categorize AD when compared to CSF biomarkers to capture the metabolic complexity in Alzheimer Disease.

55 citations


Journal ArticleDOI
TL;DR: This data indicates that metabolites in blood associated with pathology as indexed by cerebrospinal fluid (CSF) AD biomarkers may be linked to brain pathology in patients with AD.
Abstract: Introduction A critical and as-yet unmet need in Alzheimer's disease (AD) is the discovery of peripheral small molecule biomarkers. Given that brain pathology precedes clinical symptom onset, we set out to test whether metabolites in blood associated with pathology as indexed by cerebrospinal fluid (CSF) AD biomarkers. Methods This study analyzed 593 plasma samples selected from the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery study, of individuals who were cognitively healthy (n = 242), had mild cognitive impairment (n = 236), or had AD-type dementia (n = 115). Logistic regressions were carried out between plasma metabolites (n = 883) and CSF markers, magnetic resonance imaging, cognition, and clinical diagnosis. Results Eight metabolites were associated with amyloid β and one with t-tau in CSF, these were primary fatty acid amides (PFAMs), lipokines, and amino acids. From these, PFAMs, glutamate, and aspartate also associated with hippocampal volume and memory. Discussion PFAMs have been found increased and associated with amyloid β burden in CSF and clinical measures.

52 citations


Journal ArticleDOI
01 Apr 2019-Brain
TL;DR: An inverse association between cerebral perfusion in frontotemporoparietal regions and expected age of onset is identified and Cerebral perfusion may be a promising imaging biomarker for presymptomatic genetic FTD.
Abstract: Genetic forms of frontotemporal dementia are most commonly due to mutations in three genes, C9orf72, GRN or MAPT, with presymptomatic carriers from families representing those at risk. While cerebral blood flow shows differences between frontotemporal dementia and other forms of dementia, there is limited evidence of its utility in presymptomatic stages of frontotemporal dementia. This study aimed to delineate the cerebral blood flow signature of presymptomatic, genetic frontotemporal dementia using a voxel-based approach. In the multicentre GENetic Frontotemporal dementia Initiative (GENFI) study, we investigated cross-sectional differences in arterial spin labelling MRI-based cerebral blood flow between presymptomatic C9orf72, GRN or MAPT mutation carriers (n = 107) and non-carriers (n = 113), using general linear mixed-effects models and voxel-based analyses. Cerebral blood flow within regions of interest derived from this model was then explored to identify differences between individual gene carrier groups and to estimate a timeframe for the expression of these differences. The voxel-based analysis revealed a significant inverse association between cerebral blood flow and the expected age of symptom onset in carriers, but not non-carriers. Regions included the bilateral insulae/orbitofrontal cortices, anterior cingulate/paracingulate gyri, and inferior parietal cortices, as well as the left middle temporal gyrus. For all bilateral regions, associations were greater on the right side. After correction for partial volume effects in a region of interest analysis, the results were found to be largely driven by the C9orf72 genetic subgroup. These cerebral blood flow differences first appeared approximately 12.5 years before the expected symptom onset determined on an individual basis. Cerebral blood flow was lower in presymptomatic mutation carriers closer to and beyond their expected age of symptom onset in key frontotemporal dementia signature regions. These results suggest that arterial spin labelling MRI may be a promising non-invasive imaging biomarker for the presymptomatic stages of genetic frontotemporal dementia.

Journal ArticleDOI
TL;DR: The ATN scheme identified different biomarker profiles with overlapping baseline features and patterns of cognitive decline, which may have different implications in patients with vs without dementia, poses a challenge to the application of the ATn scheme.
Abstract: OBJECTIVE: To apply the ATN scheme to memory clinic patients, to assess whether it discriminates patient populations with specific features. METHODS: We included 305 memory clinic patients (33% subjective cognitive decline [SCD]: 60 ± 9 years, 61% M; 19% mild cognitive impairment [MCI]: 68 ± 9 years, 68% M; 48% dementia: 66 ± 10 years, 58% M) classified for positivity (±) of amyloid (A) ([18F]Florbetaben PET), tau (T) (CSF p-tau), and neurodegeneration (N) (medial temporal lobe atrophy). We assessed ATN profiles' demographic, clinical, and cognitive features at baseline, and cognitive decline over time. RESULTS: The proportion of A+T+N+ patients increased with syndrome severity (from 1% in SCD to 14% in MCI and 35% in dementia), while the opposite was true for A-T-N- (from 48% to 19% and 6%). Compared to A-T-N-, patients with the Alzheimer disease profiles (A+T+N- and A+T+N+) were older (both p < 0.05) and had a higher prevalence of APOE e4 (both p < 0.05) and lower Mini-Mental State Examination (MMSE) (both p < 0.05), memory (both p < 0.05), and visuospatial abilities (both p < 0.05) at baseline. Non-Alzheimer profiles A-T-N+ and A-T+N+ showed more severe white matter hyperintensities (both p < 0.05) and worse language performance (both p < 0.05) than A-T-N-. A linear mixed model showed faster decline on MMSE over time in A+T+N- and A+T+N+ (p = 0.059 and p < 0.001 vs A-T-N-), attributable mainly to patients without dementia. CONCLUSIONS: The ATN scheme identified different biomarker profiles with overlapping baseline features and patterns of cognitive decline. The large number of profiles, which may have different implications in patients with vs without dementia, poses a challenge to the application of the ATN scheme.

Journal ArticleDOI
TL;DR: Current practice in dementia imaging is fairly homogeneous across Europe, in terms of image acquisition and image interpretation, and hurdles identified include training on the use of visual rating scales, implementation of volumetric assessment and structured reporting.
Abstract: Through a European-wide survey, we assessed the current clinical practice of imaging in the primary evaluation of dementia, with respect to standardised imaging, evaluation and reporting. An online questionnaire was emailed to all European Society of Neuroradiology (ESNR) members (n = 1662) and non-members who had expressed their interest in ESNR activities in the past (n = 6400). The questionnaire featured 42 individual items, divided into multiple choice, single best choice and free text answers. Information was gathered on the context of the practices, available and preferred imaging modalities, applied imaging protocols and standards for interpretation, reporting and communication. A total of 193 unique (non-duplicate) entries from the European academic and non-academic institutions were received from a total of 28 countries. Of these, 75% were neuroradiologists, 12% general radiologists and 11% (neuro) radiologists in training. Of responding centres, 38% performed more than five scans/week for suspected dementia. MRI was primarily used in 72% of centres. Over 90% of centres acquired a combination of T2w, FLAIR, T1w, DWI and T2*w sequences. Visual rating scales were used in 75% of centres, most often the Fazekas and medial temporal atrophy scale; 32% of respondents lacked full confidence in their use. Only 23% of centres performed volumetric analysis. A minority of centres (28%) used structured reports. Current practice in dementia imaging is fairly homogeneous across Europe, in terms of image acquisition and image interpretation. Hurdles identified include training on the use of visual rating scales, implementation of volumetric assessment and structured reporting.

Journal ArticleDOI
TL;DR: Plasma proteins have been widely studied as candidate biomarkers to predict brain amyloid deposition to increase recruitment efficiency in secondary prevention clinical trials for Alzheimer's disease but most studies are targeted to specific proteins or are biased toward high abundant proteins.
Abstract: Introduction Plasma proteins have been widely studied as candidate biomarkers to predict brain amyloid deposition to increase recruitment efficiency in secondary prevention clinical trials for Alzheimer's disease. Most such biomarker studies are targeted to specific proteins or are biased toward high abundant proteins. Methods 4001 plasma proteins were measured in two groups of participants (discovery group = 516, replication group = 365) selected from the European Medical Information Framework for Alzheimer's disease Multimodal Biomarker Discovery study, all of whom had measures of amyloid. Results A panel of proteins (n = 44), along with age and apolipoprotein E (APOE) e4, predicted brain amyloid deposition with good performance in both the discovery group (area under the curve = 0.78) and the replication group (area under the curve = 0.68). Furthermore, a causal relationship between amyloid and tau was confirmed by Mendelian randomization. Discussion The results suggest that high-dimensional plasma protein testing could be a useful and reproducible approach for measuring brain amyloid deposition.

Journal ArticleDOI
TL;DR: To identify brain regions whose metabolic impairment contributes to dementia with Lewy bodies clinical core features expression and to assess the influence of severity of global cognitive impairment on the DLB hypometabolic pattern.
Abstract: Objective To identify brain regions whose metabolic impairment contributes to dementia with Lewy bodies (DLB) clinical core features expression and to assess the influence of severity of global cognitive impairment on the DLB hypometabolic pattern. Methods Brain fluorodeoxyglucose positron emission tomography and information on core features were available in 171 patients belonging to the imaging repository of the European DLB Consortium. Principal component analysis was applied to identify brain regions relevant to the local data variance. A linear regression model was applied to generate core-feature-specific patterns controlling for the main confounding variables (Mini-Mental State Examination [MMSE], age, education, gender, and center). Regression analysis to the locally normalized intensities was performed to generate an MMSE-sensitive map. Results Parkinsonism negatively covaried with bilateral parietal, precuneus, and anterior cingulate metabolism; visual hallucinations (VH) with bilateral dorsolateral-frontal cortex, posterior cingulate, and parietal metabolism; and rapid eye movement sleep behavior disorder (RBD) with bilateral parieto-occipital cortex, precuneus, and ventrolateral-frontal metabolism. VH and RBD shared a positive covariance with metabolism in the medial temporal lobe, cerebellum, brainstem, basal ganglia, thalami, and orbitofrontal and sensorimotor cortex. Cognitive fluctuations negatively covaried with occipital metabolism and positively with parietal lobe metabolism. MMSE positively covaried with metabolism in the left superior frontal gyrus, bilateral-parietal cortex, and left precuneus, and negatively with metabolism in the insula, medial frontal gyrus, hippocampus in the left hemisphere, and right cerebellum. Interpretation Regions of more preserved metabolism are relatively consistent across the variegate DLB spectrum. By contrast, core features were associated with more prominent hypometabolism in specific regions, thus suggesting a close clinical-imaging correlation, reflecting the interplay between topography of neurodegeneration and clinical presentation in DLB patients. Ann Neurol 2019;85:715-725.

Journal ArticleDOI
Timothy Rittman1, Robin J Borchert1, Simon Jones1, John C. van Swieten2, Barbara Borroni3, Daniela Galimberti4, Mario Masellis5, Mario Masellis6, Mario Masellis7, Maria Carmela Tartaglia7, Caroline Graff8, Caroline Graff9, Fabrizio Tagliavini, Giovanni B. Frisoni10, Robert Laforce11, Elizabeth Finger12, Alexandre de Mendonça13, Sandro Sorbi14, Jonathan D. Rohrer15, James B. Rowe1, Sónia Afonso16, Maria Rosário Almeida16, Sarah Anderl-Straub17, Christin Andersson9, Anna Antonell, Silvana Archetti, Andrea Arighi4, Andrea Arighi18, Mircea Balasa, Myriam Barandiaran, Núria Bargalló, Robart Bartha12, Benjamin Bender19, Luisa Benussi, Valentina Bessi14, Giuliano Binetti, Sandra E. Black5, Martina Bocchetta15, Sergi Borrego-Écija, Jose Bras15, Rose Bruffaerts20, Paola Caroppo, David M. Cash15, Miguel Castelo-Branco16, Rhian S Convery15, Thomas E. Cope1, Maura Cosseddu3, Maria de Arriba, Giuseppe Di Fede, Zigor Diaz, Katrina M. Dick15, Diana Duro16, Chiara Fenoglio4, Chiara Fenoglio18, Camilla Ferrari14, C. Ferreira13, Toby Flanagana21, Nick C. Fox15, Morris Freedman7, Giorgio G. Fumagalli18, Giorgio G. Fumagalli4, Giorgio G. Fumagalli14, Alazne Gabilondo, Roberto Gasparotti3, Serge Gauthier22, Stefano Gazzina3, Roberta Ghidoni, Giorgio Giaccone, Ana Gorostidi, Caroline V. Greaves15, Rita Guerreiro15, Carolin Heller15, Tobias Hoegen23, Begoña Indakoetxea, Vesna Jelic9, Lize C. Jiskoot2, Hans-Otto Karnath19, Ron Keren24, Maria João Leitão16, Albert Lladó, Gemma Lombardi14, Sandra V. Loosli23, Carolina Maruta13, Simon Mead15, Lieke H.H. Meeter2, Gabriel Miltenberger13, Rick van Minkelen2, Sara Mitchell5, Benedetta Nacmias14, Mollie Neason15, Jennifer M. Nicholas25, Linn Öijerstedt8, Jaume Olives, Alessandro Padovani3, Jessica L. Panman2, Janne M. Papma2, Michela Pievani, Yolande A.L. Pijnenburg26, Enrico Premi3, Sara Prioni, Catharina Prix23, Rosa Rademakers27, Veronica Redaelli, Ekaterina Rogaeva7, Pedro Rosa-Neto22, Giacomina Rossi, Martin Rosser15, Beatriz Santiago16, Elio Scarpini18, Elio Scarpini4, Sonja Schönecker23, Elisa Semler17, Rachelle Shafei15, Christen Shoesmith12, Miguel Tábuas-Pereira16, Mikel Tainta, Ricardo Taipa, David F. Tang-Wai28, David L. Thomas15, Håkan Thonberg9, Carolyn Timberlake1, Pietro Tiraboschi, Philip Vandamme20, Mathieu Vandenbulcke20, Michele Veldsman29, Ana Verdelho13, Jorge Villanua, Jason D. Warren15, Carlo Wilke19, Ione O.C. Woollacott15, Elisabeth Wlasich23, Henrik Zetterberg15, Miren Zulaica 
TL;DR: It is proposed that maintaining the efficient organization of the brain's functional network supports cognitive health even as atrophy and connectivity decline presymptomatically.

Journal ArticleDOI
TL;DR: Inter-cohort transferability of two disease progression models and their robustness in detecting AD phases are demonstrated and this is an important step towards the adoption of data-driven statistical models into clinical domain.

Journal ArticleDOI
TL;DR: The Amyloid Imaging to Prevent Alzheimer's Disease–Diagnostic and Patient Management Study (AMYPAD‐DPMS) is designed to fill the gap in reimbursement of amyloid–positron emission tomography.
Abstract: Introduction Reimbursement of amyloid–positron emission tomography (PET) is lagging due to the lack of definitive evidence on its clinical utility and cost-effectiveness. The Amyloid Imaging to Prevent Alzheimer's Disease–Diagnostic and Patient Management Study (AMYPAD-DPMS) is designed to fill this gap. Methods AMYPAD-DPMS is a phase 4, multicenter, prospective, randomized controlled study. Nine hundred patients with subjective cognitive decline plus, mild cognitive impairment, and dementia possibly due to Alzheimer's disease will be randomized to ARM1, amyloid-PET performed early in the diagnostic workup; ARM2, amyloid-PET performed after 8 months; and ARM3, amyloid-PET performed whenever the physician chooses to do so. Endpoints The primary endpoint is the difference between ARM1 and ARM2 in the proportion of patients receiving a very-high-confidence etiologic diagnosis after 3 months. Secondary endpoints address diagnosis and diagnostic confidence, diagnostic/therapeutic management, health economics and patient-related outcomes, and methods for image quantitation. Expected Impacts AMYPAD-DPMS will supply physicians and health care payers with real-world data to plan management decisions.

Journal ArticleDOI
TL;DR: This study suggests that EOD patients without full penetrant mutations are characterized by higher probability to carry polygenic risk alleles that patients with LOAD forms, suggesting that the genetic risk factors identified in LOAD might modulate the risk also in EOAD.
Abstract: Author(s): Bonvicini, Cristian; Scassellati, Catia; Benussi, Luisa; Di Maria, Emilio; Maj, Carlo; Ciani, Miriam; Fostinelli, Silvia; Mega, Anna; Bocchetta, Martina; Lanzi, Gaetana; Giacopuzzi, Edoardo; Ferraboli, Sergio; Pievani, Michela; Fedi, Virginia; Defanti, Carlo Alberto; Giliani, Silvia; Alzheimer’s Disease Neuroimaging Initiative; Frisoni, Giovanni Battista; Ghidoni, Roberta; Gennarelli, Massimo | Abstract: BackgroundEarly onset dementias (EOD) are rare neurodegenerative dementias that present before 65 years. Genetic factors have a substantially higher pathogenetic contribution in EOD patients than in late onset dementia.ObjectiveTo identify known and/or novel rare variants in major candidate genes associated to EOD by high-throughput sequencing. Common-risk variants of apolipoprotein E (APOE) and prion protein (PRNP) genes were also assessed.MethodsWe studied 22 EOD patients recruited in Memory Clinics, in the context of studies investigating genetic forms of dementia. Two methodological approaches were applied for the target-Next Generation Sequencing (NGS) analysis of these patients. In addition, we performed progranulin plasma dosage, C9Orf72 hexanucleotide repeat expansion analysis, and APOE genotyping.ResultsWe detected three rare known pathogenic mutations in the GRN and PSEN2 genes and eleven unknown-impact mutations in the GRN, VCP, MAPT, FUS, TREM2, and NOTCH3 genes. Six patients were carriers of only common risk variants (APOE and PRNP), and one did not show any risk mutation/variant. Overall, 69% (n = 9) of our early onset Alzheimer's disease (EAOD) patients, compared with 34% (n = 13) of sporadic late onset Alzheimer's disease (LOAD) patients and 27% (n = 73) of non-affected controls (ADNI, whole genome data), were carriers of at least two rare/common risk variants in the analyzed candidate genes panel, excluding the full penetrant mutations.ConclusionThis study suggests that EOD patients without full penetrant mutations are characterized by higher probability to carry polygenic risk alleles that patients with LOAD forms. This finding is in line with recently reported evidence, thus suggesting that the genetic risk factors identified in LOAD might modulate the risk also in EOAD.

Journal ArticleDOI
Carole H. Sudre1, Carole H. Sudre2, Carole H. Sudre3, Martina Bocchetta3, Carolin Heller3, Rhian S Convery3, Mollie Neason3, Katrina M. Moore3, David M. Cash2, David M. Cash3, David L. Thomas3, Ione O.C. Woollacott3, Martha S. Foiani3, Amanda Heslegrave3, Rachelle Shafei1, Caroline V. Greaves3, John C. van Swieten4, Fermin Moreno, Raquel Sánchez-Valle5, Barbara Borroni6, Robert Laforce7, Mario Masellis8, Maria Carmela Tartaglia9, Caroline Graff10, Daniela Galimberti11, James B. Rowe12, Elizabeth Finger13, Matthis Synofzik14, Rik Vandenberghe15, Alexandre de Mendonça16, Fabrizio Tagliavini, Isabel Santana17, Simon Ducharme18, Christopher C Butler19, Alexander Gerhard20, Johannes Levin21, Adrian Danek21, Giovanni B. Frisoni, Sandro Sorbi22, Markus Otto23, Henrik Zetterberg3, Sebastien Ourselin1, M. Jorge Cardoso1, M. Jorge Cardoso2, M. Jorge Cardoso3, Jonathan D. Rohrer3, Martin N. Rossor, Jason D. Warren, Nick C. Fox, Rita Guerreiro, Jose Bras, Jennifer M. Nicholas, Simon Mead, Lize C. Jiskoot, Lieke H.H. Meeter, Jessica L. Panman, Janne M. Papma, Rick van Minkelen, Y.A.L. Pijnenburg, Myriam Barandiaran, Begoña Indakoetxea, Alazne Gabilondo, Mikel Tainta, Maria de Arriba, Ana Gorostidi, Miren Zulaica, Jorge Villanua, Zigor Diaz, Sergi Borrego-Écija, Jaume Olives, Albert Lladó, Mircea Balasa, Anna Antonell, Núria Bargalló, Enrico Premi, Maura Cosseddu, Stefano Gazzina, Alessandro Padovani, Roberto Gasparotti, Silvana Archetti, Sandra E. Black, Sara Mitchell, Ekaterina Rogaeva, Morris Freedman, Ron Keren, David F. Tang-Wai, Linn Öijerstedt, Christin Andersson, Vesna Jelic, Håkan Thonberg, Andrea Arighi, Chiara Fenoglio, Elio Scarpini, Giorgio G. Fumagalli, Thomas E. Cope, Carolyn Timberlake, Timothy Rittman, Christen Shoesmith, Robart Bartha, Rosa Rademakers, Carlo Wilke, Hans-Otto Karnarth, Benjamin Bender, Rose Bruffaerts, Philip Vandamme, Mathieu Vandenbulcke, C. Ferreira, Gabriel Miltenberger, Carolina Maruta, Ana Verdelho, Sónia Afonso, Ricardo Taipa, Paola Caroppo, Giuseppe Di Fede, Giorgio Giaccone, Sara Prioni, Veronica Redaelli, Giacomina Rossi, Pietro Tiraboschi, Diana Duro, Maria Rosário Almeida, Miguel Castelo-Branco, Maria João Leitão, Miguel Tábuas-Pereira, Beatriz Santiago, Serge Gauthier, Pedro Rosa-Neto, Michele Veldsman, Toby Flanagan, Catharina Prix, Tobias Hoegen, Elisabeth Wlasich, Sandra V. Loosli, Sonja Schönecker, Elisa Semler, Sarah Anderl-Straub, Luisa Benussi, Giuliano Binetti, Roberta Ghidoni, Michela Pievani, Gemma Lombardi, Benedetta Nacmias, Camilla Ferrari, Valentina Bessi 
TL;DR: Investigation of longitudinal change in WMH and the associations of WMH burden with grey matter (GM) loss, markers of neurodegeneration and cognitive function in GRN mutation carriers revealed that WMH load was higher in both symptomatic and presymptomatic groups compared with controls and this load increased over time.

Journal ArticleDOI
TL;DR: Functional connectivity in BPD patients and its association with BPD clinical features was assessed, with patients showing significant lower mean FC in the DMN and SN, while, at the local level, a cluster of lower functional connectivity emerged in the posterior cingulate cortex of theDMN.
Abstract: A few studies reported functional abnormalities at rest in borderline personality disorder (BPD), but their relationship with clinical aspect is unclear. We aimed to assess functional connectivity (FC) in BPD patients and its association with BPD clinical features. Twenty-one BPD patients and 14 healthy controls (HC) underwent a multidimensional assessment and resting-state fMRI. Independent component analysis was performed to identify three resting-state networks: default mode network (DMN), salience network (SN), and executive control network (ECN). FC differences between BPD and HC were assessed with voxel-wise two-sample t-tests. Additionally, we investigated the mean FC within each network and the relationship between connectivity measures and BPD clinical features. Patients showed significant lower mean FC in the DMN and SN, while, at the local level, a cluster of lower functional connectivity emerged in the posterior cingulate cortex of the DMN. The DMN connectivity was positively correlated with the anger-state intensity and expression, while the SN connectivity was positively correlated with metacognitive abilities and a negative correlation emerged with the interpersonal aggression. The dysfunctional connectivity within these networks might explain clinical features of BPD patients.

Journal ArticleDOI
TL;DR: Results show that presymptomatic FTD exhibits diminished dynamic fluidity, visiting less meta‐states, shifting less often across them, and travelling through a narrowed meta‐state distance, as compared to HC.

Posted ContentDOI
22 Nov 2019-medRxiv
TL;DR: The findings suggest that maintenance of functional network connectivity enables carriers to maintain cognitive performance, and coupling between functional connectivity and cognition was stronger for carriers than for non-carriers, and increased with proximity to the expected onset of disease.
Abstract: INTRODUCTION: The presymptomatic phase of neurodegenerative disease can last many years, with sustained cognitive function despite progressive atrophy. We investigate this phenomenon in familial Frontotemporal dementia (FTD). METHODS: We studied 121 presymptomatic FTD mutation carriers and 134 family members without mutations, using multivariate data-driven approach to link cognitive performance with both structural and functional magnetic resonance imaging. Atrophy and brain network connectivity were compared between groups, in relation to the time from expected symptom onset. RESULTS: There were group differences in brain structure and function, in the absence of differences in cognitive performance. Specifically, we identified behaviourally-relevant structural and functional network differences. Structure-function relationships were similar in both groups, but coupling between functional connectivity and cognition was stronger for carriers than for non-carriers, and increased with proximity to the expected onset of disease. DISCUSSION: Our findings suggest that maintenance of functional network connectivity enables carriers to maintain cognitive performance.

Journal ArticleDOI
TL;DR: High educational attainment was associated with slower loss of grey matter over time in mutation carriers, demonstrating that even in presence of ongoing pathological processes, education may facilitate both brain reserve and brain maintenance in the presymptomatic phase of genetic FTD.
Abstract: Objective Cognitively engaging lifestyles have been associated with reduced risk of conversion to dementia. Multiple mechanisms have been advocated, including increased brain volumes (ie, brain reserve) and reduced disease progression (ie, brain maintenance). In cross-sectional studies of presymptomatic frontotemporal dementia (FTD), higher education has been related to increased grey matter volume. Here, we examine the effect of education on grey matter loss over time. Methods Two-hundred twenty-nine subjects at-risk of carrying a pathogenic mutation leading to FTD underwent longitudinal cognitive assessment and T1-weighted MRI at baseline and at 1 year follow-up. The first principal component score of the graph-Laplacian Principal Component Analysis on 112 grey matter region-of-interest volumes was used to summarise the grey matter volume (GMV). The effects of education on cognitive performances and GMV at baseline and on the change between 1 year follow-up and baseline (slope) were tested by Structural Equation Modelling. Results Highly educated at-risk subjects had better cognition and higher grey matter volume at baseline; moreover, higher educational attainment was associated with slower loss of grey matter over time in mutation carriers. Conclusions This longitudinal study demonstrates that even in presence of ongoing pathological processes, education may facilitate both brain reserve and brain maintenance in the presymptomatic phase of genetic FTD.

Journal ArticleDOI
TL;DR: Findings disclose similar abnormalities in ADMCI and DLBMCI patients as revealed by alpha source connectivity, which can be speculated that source connectivity mostly reflects common cholinergic impairment in prodromal state of both AD andDLB, before a substantial dopaminergic derangement in the dementia stage of DLB.

Journal ArticleDOI
TL;DR: In PD patients in quiet wakefulness, alpha cortical source activations may reflect an excitatory effect of dopamine neuromodulation, as revealed by resting state eyes-closed electroencephalographic (rsEEG) rhythms at alpha frequencies.

Journal ArticleDOI
TL;DR: Plasma Aβ42 showed disease progression-related features in amnesic mild cognitive impairment patients with prodromal AD, and correlated with the ADAS-Cog13 score both in aMCI patients as a whole and the prodROMal AD group alone.
Abstract: It is an open issue whether blood biomarkers serve to diagnose Alzheimer's disease (AD) or monitor its progression over time from prodromal stages. Here, we addressed this question starting from data of the European FP7 IMI-PharmaCog/E-ADNI longitudinal study in amnesic mild cognitive impairment (aMCI) patients including biological, clinical, neuropsychological (e.g., ADAS-Cog13), neuroimaging, and electroencephalographic measures. PharmaCog/E-ADNI patients were classified as "positive" (i.e., "prodromal AD" n = 76) or "negative" (n = 52) based on a diagnostic cut-off of Aβ42/P-tau in cerebrospinal fluid as well as APOE e 4 genotype. Blood was sampled at baseline and at two follow-ups (12 and 18 months), when plasma amyloid peptide 42 and 40 (Aβ42, Aβ40) and apolipoprotein J (clusterin, CLU) were assessed. Linear Mixed Models found no significant differences in plasma molecules between the "positive" (i.e., prodromal AD) and "negative" groups at baseline. In contrast, plasma Aβ42 showed a greater reduction over time in the prodromal AD than the "negative" aMCI group (p = 0.048), while CLU and Aβ40 increased, but similarly in the two groups. Furthermore, plasma Aβ42 correlated with the ADAS-Cog13 score both in aMCI patients as a whole and the prodromal AD group alone. Finally, CLU correlated with the ADAS-Cog13 only in the whole aMCI group, and no association with ADAS-Cog13 was found for Aβ40. In conclusion, plasma Aβ42 showed disease progression-related features in aMCI patients with prodromal AD.

Journal ArticleDOI
TL;DR: MRI structural biomarkers can enrich prodromal AD with fast progressors and significantly decrease group size in clinical trials of disease modifying drugs.
Abstract: Background: Early Alzheimer's disease (AD) detection using cerebrospinal fluid (CSF) biomarkers has been recommended as enrichment strategy for trials involving mild cognitive impairment (MCI) patients. Objective: To model a prodromal AD trial for identifying MRI structural biomarkers to improve subject selection and to be used as surrogate outcomes of disease progression. Methods: APOE ϵ4 specific CSF Aβ 42 /P-tau cut-offs were used to identify MCI with prodromal AD (Aβ 42 /P-tau positive) in the WP5-PharmaCog (E-ADNI) cohort. Linear mixed models were performed 1) with baseline structural biomarker, time, and biomarker×time interaction as factors to predict longitudinal changes in ADAS-cog13, 2) with Aβ 42 /P-tau status, time, and Aβ 42 /P-tau status×time interaction as factors to explain the longitudinal changes in MRI measures, and 3) to compute sample size estimation for a trial implemented with the selected biomarkers. Results: Only baseline lateral ventricle volume was able to identify a subgroup of prodromal AD patients who declined faster (interaction, p = 0.003). Lateral ventricle volume and medial temporal lobe measures were the biomarkers most sensitive to disease progression (interaction, p≤0.042). Enrichment through ventricular volume reduced the sample size that a clinical trial would require from 13 to 76%, depending on structural outcome variable. The biomarker needing the lowest sample size was the hippocampal subfield GC-ML-DG (granule cells of molecular layer of the dentate gyrus) (n = 82 per arm to demonstrate a 20% atrophy reduction). Conclusion: MRI structural biomarkers can enrich prodromal AD with fast progressors and significantly decrease group size in clinical trials of disease modifying drugs.

Journal ArticleDOI
TL;DR: A spatiotemporal analysis pipeline based on Large Diffeomorphic Deformation Metric Mapping (LDDMM) to detect shape changes from volumetric MRI scans is proposed and found statistical differences in shape in the anterior region of the thalamus at least five years before the mutation carrier subjects develop any clinical symptoms.

Journal ArticleDOI
TL;DR: This EU Joint Programme‐Neurodegenerative Diseases Research–funded project surveyed the neuroim imaging community to assess perceived barriers in multicentric neuroimaging harmonization and actions to overcome them.