554 results on '"Paul S. Aisen"'
Search Results
2. Evaluation of Blood-Based Plasma Biomarkers as Potential Markers of Amyloid Burden in Preclinical Alzheimer’s Disease
- Author
-
Charisse N. Winston, Oliver Langford, Natalie Levin, Rema Raman, Kevin Yarasheski, Tim West, Sara Abdel-Latif, Michael Donohue, Akinori Nakamura, Kenji Toba, Colin L. Masters, James Doecke, Reisa A. Sperling, Paul S. Aisen, and Robert A. Rissman
- Subjects
Psychiatry and Mental health ,Clinical Psychology ,General Neuroscience ,General Medicine ,Geriatrics and Gerontology - Abstract
Background: Participant eligibility for the A4 Study was determined by amyloid PET imaging. Given the disadvantages of amyloid PET imaging in accessibility and cost, blood-based biomarkers may serve as a sufficient biomarker and more cost-effective screening tool for patient enrollment into preclinical AD trials. Objective: To determine if a blood-based screening test can adequately identify amyloid burden in participants screened into a preclinical AD trial. Methods: In this cross-sectional study, 224 participants from the A4 Study received an amyloid PET scan (18Florbetapir) within 90 days of blood sample collection. Blood samples from all study participants were processed within 2 h after phlebotomy. Plasma amyloid measures were quantified by Shimazdu and C2 N Diagnostics using mass spectrometry-based platforms. A corresponding subset of blood samples (n = 100) was processed within 24 h after phlebotomy and analyzed by C2 N. Results: Plasma Aβ42/Aβ40 demonstrated the highest association for Aβ accumulation in the brain with an AUC 0.76 (95%CI = 0.69, 0.82) at C2 N and 0.80 (95%CI = 0.75, 0.86) at Shimadzu. Blood samples processed to plasma within 2 h after phlebotomy provided a better prediction of amyloid PET status than blood samples processed within 24 h (AUC 0.80 versus 0.64; p
- Published
- 2023
- Full Text
- View/download PDF
3. The AHEAD 3‐45 Study: Design of a prevention trial for Alzheimer's disease
- Author
-
Michael S, Rafii, Reisa A, Sperling, Michael C, Donohue, Jin, Zhou, Claire, Roberts, Michael C, Irizarry, Shobha, Dhadda, Gopalan, Sethuraman, Lynn D, Kramer, Chad J, Swanson, David, Li, Stephen, Krause, Robert A, Rissman, Sarah, Walter, Rema, Raman, Keith A, Johnson, and Paul S, Aisen
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Abstract
The Alzheimer's disease (AD) continuum begins with a long asymptomatic or preclinical stage, during which amyloid beta (Aβ) is accumulating for more than a decade prior to widespread cortical tauopathy, neurodegeneration, and manifestation of clinical symptoms. The AHEAD 3-45 Study (BAN2401-G000-303) is testing whether intervention with lecanemab (BAN2401), a humanized immunoglobulin 1 (IgG1) monoclonal antibody that preferentially targets soluble aggregated Aβ, initiated during this asymptomatic stage can slow biomarker changes and/or cognitive decline. The AHEAD 3-45 Study is conducted as a Public-Private Partnership of the Alzheimer's Clinical Trial Consortium (ACTC), funded by the National Institute on Aging, National Institutes of Health (NIH), and Eisai Inc.The AHEAD 3-45 Study was launched on July 14, 2020, and consists of two sister trials (A3 and A45) in cognitively unimpaired (CU) individuals ages 55 to 80 with specific dosing regimens tailored to baseline brain amyloid levels on screening positron emission tomography (PET) scans: intermediate amyloid (≈20 to 40 Centiloids) for A3 and elevated amyloid (40 Centiloids) for A45. Both trials are being conducted under a single protocol, with a shared screening process and common schedule of assessments. A3 is a Phase 2 trial with PET-imaging end points, whereas A45 is a Phase 3 trial with a cognitive composite primary end point. The treatment period is 4 years. The study utilizes innovative approaches to enriching the sample with individuals who have elevated brain amyloid. These include recruiting from the Trial-Ready Cohort for Preclinical and Prodromal Alzheimer's disease (TRC-PAD), the Australian Dementia Network (ADNeT) Registry, and the Japanese Trial Ready Cohort (J-TRC), as well as incorporation of plasma screening with the C2N mass spectrometry platform to quantitate the Aβ 42/40 ratio (Aβ 42/40), which has been shown previously to reliably identify cognitively normal participants not likely to have elevated brain amyloid levels. A blood sample collected at a brief first visit is utilized to "screen out" individuals who are less likely to have elevated brain amyloid, and to determine the participant's eligibility to proceed to PET imaging. Eligibility to randomize into the A3 Trial or A45 Trial is based on the screening PET imaging results.The focus of this article is on the innovative design of the study.The AHEAD 3-45 Study will test whether with lecanemab (BAN2401) can slow the accumulation of tau and prevent the cognitive decline associated with AD during its preclinical stage. It is specifically targeting both the preclinical and the early preclinical (intermediate amyloid) stages of AD and is the first secondary prevention trial to employ plasma-based biomarkers to accelerate the screening process and potentially substantially reduce the number of screening PET scans.
- Published
- 2022
- Full Text
- View/download PDF
4. Tau positron emission tomography in preclinical Alzheimer’s disease
- Author
-
Philip S Insel, Christina B Young, Paul S Aisen, Keith A Johnson, Reisa A Sperling, Elizabeth C Mormino, and Michael C Donohue
- Subjects
Original Article ,Neurology (clinical) - Abstract
Rates of tau accumulation in cognitively unimpaired older adults are subtle, with magnitude and spatial patterns varying in recent reports. Regional accumulation also likely varies in the degree to which accumulation is amyloid-β-dependent. Thus, there is a need to evaluate the pattern and consistency of tau accumulation across multiple cognitively unimpaired cohorts and how these patterns relate to amyloid burden, in order to design optimal tau end points for clinical trials. Using three large cohorts of cognitively unimpaired older adults, the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s and companion study, Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (n = 447), the Alzheimer’s Disease Neuroimaging Initiative (n = 420) and the Harvard Aging Brain Study (n = 190), we attempted to identify regions with high rates of tau accumulation and estimate how these rates evolve over a continuous spectrum of baseline amyloid deposition. Optimal combinations of regions, tailored to multiple ranges of baseline amyloid burden as hypothetical clinical trial inclusion criteria, were tested and validated. The inferior temporal cortex, fusiform gyrus and middle temporal cortex had the largest effect sizes of accumulation in both longitudinal cohorts when considered individually. When tau regions of interest were combined to find composite weights to maximize the effect size of tau change over time, both longitudinal studies exhibited a similar pattern—inferior temporal cortex, almost exclusively, was optimal for participants with mildly elevated amyloid β levels. For participants with highly elevated baseline amyloid β levels, combined optimal composite weights were 53% inferior temporal cortex, 31% amygdala and 16% fusiform. At mildly elevated levels of baseline amyloid β, a sample size of 200/group required a treatment effect of 0.40–0.45 (40–45% slowing of tau accumulation) to power an 18-month trial using the optimized composite. Neither a temporal lobe composite nor a global composite reached 80% power with 200/group with an effect size under 0.5. The focus of early tau accumulation on the medial temporal lobe has resulted from the observation that the entorhinal cortex is the initial site to show abnormal levels of tau with age. However, these abnormal levels do not appear to be the result of a high rate of accumulation in the short term, but possibly a more moderate rate occurring early with respect to age. While the entorhinal cortex plays a central role in the early appearance of tau, it may be the inferior temporal cortex that is the critical region for rapid tau accumulation in preclinical Alzheimer’s disease.
- Published
- 2022
- Full Text
- View/download PDF
5. Recommendations to address key recruitment challenges of Alzheimer's disease clinical trials
- Author
-
Jessica B, Langbaum, Julie, Zissimopoulos, Rhoda, Au, Niranjan, Bose, Chris J, Edgar, Evan, Ehrenberg, Howard, Fillit, Carl V, Hill, Lynne, Hughes, Michael, Irizarry, Sarah, Kremen, Darius, Lakdawalla, Nancy, Lynn, Kristina, Malzbender, Tetsuyuki, Maruyama, Holly A, Massett, Deep, Patel, Desi, Peneva, Eric M, Reiman, Klaus, Romero, Carol, Routledge, Michael W, Weiner, Stacie, Weninger, and Paul S, Aisen
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Abstract
Clinical trials for Alzheimer's disease (AD) are slower to enroll study participants, take longer to complete, and are more expensive than trials in most other therapeutic areas. The recruitment and retention of a large number of qualified, diverse volunteers to participate in clinical research studies remain among the key barriers to the successful completion of AD clinical trials. An advisory panel of experts from academia, patient-advocacy organizations, philanthropy, non-profit, government, and industry convened in 2020 to assess the critical challenges facing recruitment in Alzheimer's clinical trials and develop a set of recommendations to overcome them. This paper briefly reviews existing challenges in AD clinical research and discusses the feasibility and implications of the panel's recommendations for actionable and inclusive solutions to accelerate the development of novel therapies for AD.
- Published
- 2022
- Full Text
- View/download PDF
6. Early-stage Alzheimer disease: getting trial-ready
- Author
-
Paul S. Aisen, Gustavo A. Jimenez-Maggiora, Michael S. Rafii, Sarah Walter, and Rema Raman
- Subjects
Cellular and Molecular Neuroscience ,Neurology (clinical) - Published
- 2022
- Full Text
- View/download PDF
7. Associations of Stages of Objective Memory Impairment With Amyloid PET and Structural MRI
- Author
-
Ellen Grober, Richard B. Lipton, Reisa A. Sperling, Kathryn V. Papp, Keith A. Johnson, Dorene M. Rentz, Amy E. Veroff, Paul S. Aisen, and Ali Ezzati
- Subjects
Male ,Memory Disorders ,Amyloid beta-Peptides ,Alzheimer Disease ,Positron-Emission Tomography ,Brain ,Humans ,Cognitive Dysfunction ,tau Proteins ,Neurology (clinical) ,Magnetic Resonance Imaging ,Aged - Abstract
Background and ObjectivesThe goal of this work was to investigate the neuroimaging correlates of the Stages of Objective Memory Impairment (SOMI) system operationalized with the Free and Cued Selective Reminding Test (FCSRT), a widely used episodic memory measure.MethodsThe FCSRT begins with a study phase in which items (e.g., grapes) are identified in response to unique semantic cues (e.g., fruit) that are used in the test phase to prompt recall of items not retrieved by free recall. There are 3 test trials of the 16 items (maximum 48). Data from 4,484 cognitively unimpaired participants from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were used. All participants had amyloid PET imaging, and a subset of 1,262 β-amyloid (Aβ)–positive had structural MRIs. We compared the Aβ mean cortical standardized uptake value ratio (SUVR) and volumetric measures of hippocampus, parahippocampal gyrus, entorhinal cortex, and inferior temporal cortex between the 5 SOMI stages.ResultsParticipants had a mean age of 71.3 (SD 4.6) years; 40.6% were male; and 34.6% were APOE ε4 positive. Half had no memory impairment; the other half had retrieval deficits, storage limitations, or both. Analysis of covariance in the entire sample while controlling for age, sex, education, and APOE ε4 showed that individuals in higher SOMI stages had higher global amyloid SUVR (p < 0.001). Both SOMI-4 and -3 subgroups had higher amyloid SUVR than SOMI-0 and SOMI-1 subgroups. Individuals in higher SOMI stages had smaller hippocampal volume (p = 0.003), entorhinal cortex (p < 0.05), and inferior temporal lobes (p < 0.05), but there was no difference between parahippocampal gyrus volume of different SOMI stages. Pairwise comparison of SOMI subgroups showed that the SOMI-4, -3, and -2 subgroups had smaller hippocampal volume than the SOMI-0 and -1 subgroup. The SOMI-4 subgroup had significantly smaller entorhinal cortex and smaller inferior temporal lobe compared to all other groups.DiscussionPresence of Alzheimer disease pathology is closely related to memory impairment according to SOMI stages in the cognitively unimpaired sample of A4. Results from structural MRIs suggest that memory storage impairment (SOMI-3 and -4) is present when there is widespread medial temporal lobe atrophy.Trial Registration InformationClinicalTrials.gov identifier: NCT02008357.Classification of EvidenceThis study provides Class I evidence that, in normal older individuals, higher stages of memory impairment assessed with FCSRT were associated with higher amyloid imaging burden and lower volume of hippocampus, entorhinal cortex, and inferior temporal lobes.
- Published
- 2022
- Full Text
- View/download PDF
8. Expectations and clinical meaningfulness of randomized controlled trials
- Author
-
Ronald C. Petersen, Paul S. Aisen, J. Scott Andrews, Alireza Atri, Brandy R. Matthews, Dorene M. Rentz, Eric R. Siemers, Christopher J. Weber, and Maria C. Carrillo
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2023
- Full Text
- View/download PDF
9. Contribution of Alzheimer's biomarkers and risk factors to cognitive impairment and decline across the Alzheimer's disease continuum
- Author
-
William J. Jagust, Leslie M. Shaw, Andrew J. Saykin, John Q. Trojanowski, Alzheimer’s Disease Neuroimaging Initiative, Ronald C. Petersen, Dallas P. Veitch, Daniel Weintraub, Clifford R. Jack, Michael W. Weiner, Paul S. Aisen, Zeynep Demir, and Duygu Tosun
- Subjects
Oncology ,Apolipoprotein E ,medicine.medical_specialty ,Epidemiology ,Amyloid beta ,tau Proteins ,Cellular and Molecular Neuroscience ,Atrophy ,Developmental Neuroscience ,Alzheimer Disease ,Risk Factors ,Internal medicine ,medicine ,Humans ,Dementia ,Cognitive Dysfunction ,Cognitive decline ,Aged ,Amyloid beta-Peptides ,biology ,business.industry ,Health Policy ,Neurodegeneration ,Brain ,Cognition ,medicine.disease ,Hyperintensity ,Psychiatry and Mental health ,Cross-Sectional Studies ,Positron-Emission Tomography ,biology.protein ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,Biomarkers - Abstract
Introduction Amyloid beta (Aβ), tau, and neurodegeneration jointly with the Alzheimer's disease (AD) risk factors affect the severity of clinical symptoms and disease progression. Methods Within 248 Aβ-positive elderly with and without cognitive impairment and dementia, partial least squares structural equation pathway modeling was used to assess the direct and indirect effects of imaging biomarkers (global Aβ-positron emission tomography [PET] uptake, regional tau-PET uptake, and regional magnetic resonance imaging-based atrophy) and risk-factors (age, sex, education, apolipoprotein E [APOE], and white-matter lesions) on cross-sectional cognitive impairment and longitudinal cognitive decline. Results Sixteen percent of variance in cross-sectional cognitive impairment was accounted for by Aβ, 46% to 47% by tau, and 25% to 29% by atrophy, although 53% to 58% of total variance in cognitive impairment was explained by incorporating mediated and direct effects of AD risk factors. The Aβ-tau-atrophy pathway accounted for 50% to 56% of variance in longitudinal cognitive decline while Aβ, tau, and atrophy independently explained 16%, 46% to 47%, and 25% to 29% of the variance, respectively. Discussion These findings emphasize that treatments that remove Aβ and completely stop downstream effects on tau and neurodegeneration would only be partially effective in slowing of cognitive decline or reversing cognitive impairment.
- Published
- 2021
- Full Text
- View/download PDF
10. Using the Alzheimer's Disease Neuroimaging Initiative to improve early detection, diagnosis, and treatment of Alzheimer's disease
- Author
-
Paul S. Aisen, Michael W. Weiner, Danielle J Harvey, William J. Jagust, John Q. Trojanowski, Robert C. Green, Alzheimer’s Disease Neuroimaging Initiative, Susan M. Landau, Arthur W. Toga, Leslie M. Shaw, Ronald C. Petersen, Monica Rivera-Mindt, Duygu Tosun, Richard J. Perrin, John C. Morris, Laurel A. Beckett, Andrew J. Saykin, Clifford R. Jack, Ozioma C. Okonkwo, Dallas P. Veitch, and Charles DeCarli
- Subjects
Oncology ,Aging ,Epidemiology ,Disease ,Neurodegenerative ,Alzheimer's Disease ,tau ,screening and diagnosis ,biology ,Health Policy ,AV1541 tau positron emission tomography ,amyloid ,plasma biomarker ,Detection ,Psychiatry and Mental health ,Neurological ,Cohort ,Disease Progression ,Biomedical Imaging ,Alzheimer's Disease Neuroimaging Initiative ,medicine.medical_specialty ,Amyloid beta ,Clinical Sciences ,Neuroimaging ,tau Proteins ,Cellular and Molecular Neuroscience ,disease progression ,mild cognitive impairment ,Developmental Neuroscience ,Alzheimer Disease ,Internal medicine ,mental disorders ,Acquired Cognitive Impairment ,medicine ,Humans ,Cognitive Dysfunction ,Amyloid beta-Peptides ,Vascular disease ,business.industry ,Prevention ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,medicine.disease ,Brain Disorders ,4.1 Discovery and preclinical testing of markers and technologies ,Biomarker (cell) ,Clinical trial ,Geriatrics ,biology.protein ,Dementia ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,Biomarkers - Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) has accumulated 15 years of clinical, neuroimaging, cognitive, biofluid biomarker and genetic data, and biofluid samples available to researchers, resulting in more than 3500 publications. This review covers studies from 2018 to 2020. METHODS We identified 1442 publications using ADNI data by conventional search methods and selected impactful studies for inclusion. RESULTS Disease progression studies supported pivotal roles for regional amyloid beta (Aβ) and tau deposition, and identified underlying genetic contributions to Alzheimer's disease (AD). Vascular disease, immune response, inflammation, resilience, and sex modulated disease course. Biologically coherent subgroups were identified at all clinical stages. Practical algorithms and methodological changes improved determination of Aβ status. Plasma Aβ, phosphorylated tau181, and neurofilament light were promising noninvasive biomarkers. Prognostic and diagnostic models were externally validated in ADNI but studies are limited by lack of ethnocultural cohort diversity. DISCUSSION ADNI has had a profound impact in improving clinical trials for AD.
- Published
- 2021
- Full Text
- View/download PDF
11. Pilot Evaluation of the Unsupervised, At-Home Cogstate Brief Battery in ADNI-2
- Author
-
Bruce Albala, Chris J. Edgar, Ronald C. Petersen, Eric Siemers, Paul S. Aisen, Michael W. Weiner, and Paul Maruff
- Subjects
cognition ,Male ,medicine.medical_specialty ,Concordance ,healthcare research ,Neuroimaging ,Pilot Projects ,Neuropsychological Tests ,Physical medicine and rehabilitation ,Medicine ,Humans ,Cognitive Dysfunction ,Cognitive impairment ,Aged ,business.industry ,General Neuroscience ,Cognition ,Usability ,General Medicine ,Telemedicine ,Test (assessment) ,Clinical trial ,Psychiatry and Mental health ,Clinical Psychology ,clinical trials as a topic ,Female ,Geriatrics and Gerontology ,digital technology ,business ,Alzheimer’s disease ,Research Article - Abstract
Background: There is a need for feasible, scalable assessments to detect cognitive impairment and decline. The Cogstate Brief Battery (CBB) is validated for Alzheimer’s disease (AD) and in unsupervised and bring your own device contexts. The CBB has shown usability for self-completion in the home but has not been employed in this way in a multisite clinical trial in AD. Objective: The objective of the pilot was to evaluate feasibility of at-home, self-completion of the CBB in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) over 24 months. Methods: The CBB was included as a pilot for cognitively normal (CN) and mild cognitive impairment (MCI) participants in ADNI-2, invited to take the assessment in-clinic, then at at-home over a period of 24 months follow-up. Data were analyzed to explore acceptability/usability, concordance of in-clinic and at-home assessment, and validity. Results: Data were collected for 104 participants (46 CN, 51 MCI, and 7 AD) who consented to provide CBB data. Subsequent analyses were performed for the CN and MCI groups only. Test completion rates were 100%for both the first in-clinic supervised and first at-home unsupervised assessments, with few repeat performances required. However, available follow-up data declined sharply over time. Good concordance was seen between in-clinic and at-home assessments, with non-significant and small effect size differences (Cohen’s d between -0.04 and 0.28) and generally moderate correlations (r = 0.42 to 0.73). Known groups validity was also supported (11/16 comparisons with Cohen’s d≥0.3). Conclusion: These data demonstrate the feasibility of use for the CBB for unsupervised at-home, testing, including MCI groups. Optimal approaches to the application of assessments to support compliance over time remain to be determined.
- Published
- 2021
12. A trial of gantenerumab or solanezumab in dominantly inherited Alzheimer’s disease
- Author
-
Christopher H. van Dyck, Roger Clarnette, Susan Mills, John C. Morris, Carlos Cruchaga, Anna Santacruz, Ging-Yuek Robin Hsiung, Roy Yaari, Suman Jayadev, Caroline Giacobino, William S. Brooks, Robert A. Koeppe, Raquel Sánchez-Valle, Anne M. Fagan, Eric McDade, Sarah B. Berman, Catherine J. Mummery, Florence Pasquier, Scott M. Berry, Randall J. Bateman, Brian A. Gordon, Jorge J. Llibre-Guerra, Maïté Formaglio, Paul S. Aisen, Paulo Fontoura, Mark A. Mintun, Bruno Dubois, Erik D. Roberson, Kelley Coalier, Ronald G. Thomas, Martin R. Farlow, John R. Sims, Serge Gauthier, Douglas Galasko, Mario Masellis, G. Mustafa Surti, Barbara A. Wendelberger, Guoqiao Wang, James J. Lah, Yan Li, David B. Clifford, David Wallon, Paul Delmar, Alison Goate, Rachelle S. Doody, Didier Hannequin, Stephen Salloway, Geoffrey A. Kerchner, Karen C. Holdridge, Ivonne Z. Jimenez-Velazquez, Janice M. Hitchcock, Monika Baudler, Lawrence S. Honig, Tammie L.S. Benzinger, Clifford R. Jack, Peter J. Snyder, Scott W. Andersen, J. Pariente, Andrew J. Aschenbrenner, Jason Hassenstab, Richard J. Perrin, Colin L. Masters, Chengjie Xiong, Jared R. Brosch, and B. Joy Snider
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Disease ,Antibodies, Monoclonal, Humanized ,Placebo ,Asymptomatic ,Article ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Placebos ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Alzheimer Disease ,law ,Internal medicine ,medicine ,Humans ,Solanezumab ,Cognitive decline ,Amyloid beta-Peptides ,Dose-Response Relationship, Drug ,business.industry ,General Medicine ,Middle Aged ,030104 developmental biology ,030220 oncology & carcinogenesis ,Disease Progression ,Biomarker (medicine) ,Female ,medicine.symptom ,Gantenerumab ,business ,Biomarkers ,medicine.drug - Abstract
Dominantly inherited Alzheimer's disease (DIAD) causes predictable biological changes decades before the onset of clinical symptoms, enabling testing of interventions in the asymptomatic and symptomatic stages to delay or slow disease progression. We conducted a randomized, placebo-controlled, multi-arm trial of gantenerumab or solanezumab in participants with DIAD across asymptomatic and symptomatic disease stages. Mutation carriers were assigned 3:1 to either drug or placebo and received treatment for 4-7 years. The primary outcome was a cognitive end point; secondary outcomes included clinical, cognitive, imaging and fluid biomarker measures. Fifty-two participants carrying a mutation were assigned to receive gantenerumab, 52 solanezumab and 40 placebo. Both drugs engaged their Aβ targets but neither demonstrated a beneficial effect on cognitive measures compared to controls. The solanezumab-treated group showed a greater cognitive decline on some measures and did not show benefits on downstream biomarkers. Gantenerumab significantly reduced amyloid plaques, cerebrospinal fluid total tau, and phospho-tau181 and attenuated increases of neurofilament light chain. Amyloid-related imaging abnormalities edema was observed in 19.2% (3 out of 11 were mildly symptomatic) of the gantenerumab group, 2.5% of the placebo group and 0% of the solanezumab group. Gantenerumab and solanezumab did not slow cognitive decline in symptomatic DIAD. The asymptomatic groups showed no cognitive decline; symptomatic participants had declined before reaching the target doses.
- Published
- 2021
- Full Text
- View/download PDF
13. The Longitudinal Early‐onset Alzheimer's Disease Study (LEADS): Framework and methodology
- Author
-
Steve Salloway, Anne M. Fagan, Neill R. Graff-Radford, Robert A. Koeppe, Ani Eloyan, Lea T. Grinberg, Bradford C. Dickerson, Constantine Gatsonis, Walter A. Kukull, Thomas S. Wingo, Amy Trullinger, Gregory S. Day, Emily Rogalski, David A. Wolk, Paul S. Aisen, Prashanthi Vemuri, Joel H. Kramer, Gil D. Rabinovici, Erik S. Musiek, Tatiana Foroud, Arthur W. Toga, Clifford R. Jack, Mario F. Mendez, Leonardo Iaccarino, Joseph C. Masdeu, Malia Rumbaugh, Keith N. Fargo, Liana G. Apostolova, Chiadi U. Onyike, Kelly N.H. Nudelman, Lawrence S. Honig, Maria C. Carrillo, Melissa E. Murray, and David T.W. Jones
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Epidemiology ,Disease ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Alzheimer Disease ,Internal medicine ,Stilbenes ,National Institute on Aging (U.S.) ,medicine ,Humans ,Dementia ,Cognitive Dysfunction ,Early-onset Alzheimer's disease ,Longitudinal Studies ,Florbetaben ,Aniline Compounds ,medicine.diagnostic_test ,business.industry ,Health Policy ,Brain ,Magnetic resonance imaging ,Cognition ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,United States ,Clinical trial ,Psychiatry and Mental health ,Early Diagnosis ,030104 developmental biology ,Positron-Emission Tomography ,Biomarker (medicine) ,Female ,Autopsy ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Patients with early-onset Alzheimer's disease (EOAD) are commonly excluded from large-scale observational and therapeutic studies due to their young age, atypical presentation, or absence of pathogenic mutations. The goals of the Longitudinal EOAD Study (LEADS) are to (1) define the clinical, imaging, and fluid biomarker characteristics of EOAD; (2) develop sensitive cognitive and biomarker measures for future clinical and research use; and (3) establish a trial-ready network. LEADS will follow 400 amyloid beta (Aβ)-positive EOAD, 200 Aβ-negative EOnonAD that meet National Institute on Aging-Alzheimer's Association (NIA-AA) criteria for mild cognitive impairment (MCI) or AD dementia, and 100 age-matched controls. Participants will undergo clinical and cognitive assessments, magnetic resonance imaging (MRI), [18 F]Florbetaben and [18 F]Flortaucipir positron emission tomography (PET), lumbar puncture, and blood draw for DNA, RNA, plasma, serum and peripheral blood mononuclear cells, and post-mortem assessment. To develop more effective AD treatments, scientists need to understand the genetic, biological, and clinical processes involved in EOAD. LEADS will develop a public resource that will enable future planning and implementation of EOAD clinical trials.
- Published
- 2021
- Full Text
- View/download PDF
14. The Institute on Methods and Protocols for Advancement of Clinical Trials in ADRD (IMPACT-AD): A Novel Clinical Trials Training Program
- Author
-
Tyler Berkness, Paul S. Aisen, Joshua D. Grill, C. Flournoy, Maria C. Carrillo, Rema Raman, R. C. Petersen, Reisa A. Sperling, and Heather M. Snyder
- Subjects
Male ,medicine.medical_specialty ,Aging ,Acceptance rate ,Clinical Trials and Supportive Activities ,education ,IMPACT-AD ,Neurodegenerative ,Alzheimer's Disease ,Ethics, Research ,diversity ,s disease ,Multidisciplinary approach ,Alzheimer Disease ,Clinical Research ,Acquired Cognitive Impairment ,Medicine ,Humans ,Fellowships and Scholarships ,Alzheimer’ ,Original Research ,Ethics ,Clinical Trials as Topic ,clinical trials ,training ,business.industry ,clinical Trials ,Teaching ,Research ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Cultural Diversity ,ADRD ,Brain Disorders ,Clinical trial ,First person ,Research Design ,Informatics ,Family medicine ,Cohort ,Female ,Dementia ,Biostatistics ,Training program ,business ,Alzheimer’s disease - Abstract
Background: Alzheimer’s Disease and Related Dementias (ADRD) clinical trials require multidisciplinary expertise in medicine, biostatistics, trial design, biomarkers, ethics, and informatics. Objectives: To provide focused interactive training in ADRD clinical trials to a diverse cadre of investigators. Design: The Institute on Methods and Protocols for Advancement of Clinical Trials in ADRD (IMPACT-AD) is a novel multidisciplinary clinical trial training program funded by the National Institute on Aging and the Alzheimer’s Association with two educational tracks. The Professionals track includes individuals who fill a broad variety of roles including clinicians, study coordinators, psychometricians, and other study professionals who wish to further their knowledge and advance their careers in ADRD trials. The Fellowship track includes current and future principal investigators and focuses on the design, conduct and analysis of ADRD clinical trials. Setting: The 2020 inaugural iteration of IMPACT-AD was held via Zoom. Participants: Thirty-five trainees (15 Fellowship track; 20 Professionals track) were selected from 104 applications (34% acceptance rate). Most (n=25, 71%) identified as female. Fifteen (43%) were of a non-white race; six (18%) were of Hispanic ethnicity; eight (23%) indicated they were the first person in their family to attend college. Measurements: Participants completed daily evaluations as well as pre- and post-course assessments of learning. Results: Across topic areas, >90% of trainees evaluated their change in knowledge based on the lectures as “very much” or “somewhat increased.” The mean proportion correct responses in pre- and post-course assessments increased from 55% to 75% for the Professionals track and from 54% to 78% for the Fellowship track. Conclusions: IMPACT-AD successfully launched a new training opportunity amid a global pandemic that preliminarily achieved the goals of attracting a diverse cohort and providing meaningful training. The course is funded through 2025.
- Published
- 2021
15. Aducanumab: Appropriate Use Recommendations
- Author
-
Paul S. Aisen, Alireza Atri, Liana G. Apostolova, Steven Salloway, Jeffrey L. Cummings, and M. Weiner
- Subjects
Amyloid ,medicine.medical_specialty ,education.field_of_study ,Neurology ,Drug-Related Side Effects and Adverse Reactions ,business.industry ,Incidence (epidemiology) ,Population ,Guidelines as Topic ,Disease ,Antibodies, Monoclonal, Humanized ,medicine.disease ,Appropriate use ,Magnetic Resonance Imaging ,United States ,Discontinuation ,Alzheimer Disease ,Humans ,Medicine ,Dementia ,Aducanumab ,Infusions, Intravenous ,business ,Intensive care medicine ,education - Abstract
Aducanumab has been approved by the US Food and Drug Administration for treatment of Alzheimer’s disease (AD). Clinicians require guidance on the appropriate use of this new therapy. An Expert Panel was assembled to construct Appropriate Use Recommendations based on the participant populations, conduct of the pivotal trials of aducanumab, updated Prescribing Information, and expert consensus. Aducanumab is an amyloid-targeting monoclonal antibody delivered by monthly intravenous infusions. The pivotal trials included patients with early AD (mild cognitive impairment due to AD and mild AD dementia) who had confirmed brain amyloid using amyloid positron tomography. The Expert Panel recommends that use of aducanumab be restricted to this population in which efficacy and safety have been studied. Aducanumab is titrated to a dose of 10 mg/kg over a 6-month period. The Expert Panel recommends that the aducanumab be titrated to the highest dose to maximize the opportunity for efficacy. Aducanumab can substantially increase the incidence of amyloid-related imaging abnormalities (ARIA) with brain effusion or hemorrhage. Dose interruption or treatment discontinuation is recommended for symptomatic ARIA and for moderate-severe ARIA. The Expert Panel recommends MRIs prior to initiating therapy, during the titration of the drug, and at any time the patient has symptoms suggestive of ARIA. Recommendations are made for measures less cumbersome than those used in trials for the assessment of effectiveness in the practice setting. The Expert Panel emphasized the critical importance of engaging in a process of patient-centered informed decision-making that includes comprehensive discussions and clear communication with the patient and care partner regarding the requirements for therapy, the expected outcome of therapy, potential risks and side effects, and the required safety monitoring, as well as uncertainties regarding individual responses and benefits.
- Published
- 2021
- Full Text
- View/download PDF
16. Platform Trials to Expedite Drug Development in Alzheimer’s Disease: A Report from the EU/US CTAD Task Force
- Author
-
Randall J. Bateman, Reisa S. Sperling, John R. Sims, Paul S. Aisen, Bruno Vellas, Rachelle Doody, Keith A. Johnson, and Maria C. Carrillo
- Subjects
medicine.medical_specialty ,Advisory Committees ,Psychological intervention ,tau Proteins ,Disease ,Antibodies, Monoclonal, Humanized ,adaptive trial design ,Physical medicine and rehabilitation ,Drug Development ,Alzheimer Disease ,Interim ,Outcome Assessment, Health Care ,Humans ,Medicine ,anti-amyloid therapies ,master protocols ,Amyloid beta-Peptides ,business.industry ,Treatment regimen ,Task force ,platform trials ,Outcome measures ,CTAD Task Force Paper ,shared placebos ,Clinical trial ,Drug development ,Research Design ,Asymptomatic Diseases ,anti-tau therapies ,business ,Alzheimer’s disease ,Biomarkers ,secondary prevention - Abstract
A diverse range of platforms has been established to increase the efficiency and speed of clinical trials for Alzheimer’s disease (AD). These platforms enable parallel assessment of multiple therapeutics, treatment regimens, or participant groups; use uniform protocols and outcome measures; and may allow treatment arms to be added or dropped based on interim analyses of outcomes. The EU/US CTAD Task Force discussed the lessons learned from the Dominantly Inherited Alzheimer’s Network Trials Unit (DIAN-TU) platform trial and the challenges addressed by other platform trials that have launched or are in the planning stages. The landscape of clinical trial platforms in the AD space includes those testing experimental therapies such as DIAN-TU, platforms designed to test multidomain interventions, and those designed to streamline trial recruitment by building trial-ready cohorts. The heterogeneity of the AD patient population, AD drugs, treatment regimens, and analytical methods complicates the design and execution of platform trials, yet Task Force members concluded that platform trials are essential to advance the search for effective AD treatments, including combination therapies.
- Published
- 2021
- Full Text
- View/download PDF
17. The Ups and Downs of Amyloid in Alzheimer’s
- Author
-
Maria C. Carrillo, Paul S. Aisen, and E. Siemers
- Subjects
Amyloid ,Amyloid beta-Peptides ,Alzheimer Disease ,Geriatrics gerontology ,business.industry ,Humans ,Medicine ,business ,Neuroscience - Abstract
Very recently, the Food and Drug Administration (FDA) in the United States gave an “accelerated approval” to aducanumab, the first new drug to be available to patients with Alzheimer’s disease (AD) in nearly two decades and the first ever that targets the underlying neuropathology. The accelerated approval pathway is based on a biomarker effect, in this case reduction in brain amyloid as measured by PET scan, that is “reasonably likely” to predict clinical efficacy. While there were numerous complexities surrounding the approval, this event was nevertheless seminal for the treatment of AD and for the amyloid hypothesis.
- Published
- 2021
- Full Text
- View/download PDF
18. Intranasal Insulin Reduces White Matter Hyperintensity Progression in Association with Improvements in Cognition and CSF Biomarker Profiles in Mild Cognitive Impairment and Alzheimer’s Disease
- Author
-
James B. Brewer, Suzanne Craft, Paul S. Aisen, Rema Raman, Derek Kellar, Robert A. Rissman, and Samuel N. Lockhart
- Subjects
Male ,medicine.medical_specialty ,Neurology ,medicine.medical_treatment ,Disease ,Neuropsychological Tests ,Placebo ,Gastroenterology ,White matter ,Cognition ,Alzheimer Disease ,Insulin, Regular, Human ,Internal medicine ,Activities of Daily Living ,Image Processing, Computer-Assisted ,medicine ,Humans ,Hypoglycemic Agents ,Insulin ,Cognitive Dysfunction ,Administration, Intranasal ,Aged ,business.industry ,Brain ,Magnetic Resonance Imaging ,White Matter ,Hyperintensity ,Clinical trial ,medicine.anatomical_structure ,Biomarker (medicine) ,Female ,business ,Biomarkers - Abstract
Background: Intranasally administered insulin has shown promise in both rodent and human studies in Alzheimer’s disease; however, both effects and mechanisms require elucidation. Objective: We assessed the effects of intranasally administered insulin on white matter health and its association with cognition and cerebral spinal fluid biomarker profiles in adults with mild cognitive impairment or Alzheimer’s disease in secondary analyses from a prior phase 2 clinical trial (NCT01767909). Design: A randomized (1:1) double-blind clinical trial. Setting: Twelve sites across the United States. Participants: Adults with mild cognitive impairment or Alzheimer’s disease. Intervention: Participants received either twice daily placebo or insulin (20 IU Humulin R U-100 b.i.d.) intranasally for 12 months. Seventy-eight participants were screened, of whom 49 (32 men) were enrolled. Measurements: Changes from baseline in global and regional white matter hyperintensity volume and gray matter volume were analyzed and related to changes in cerebral spinal fluid biomarkers, Alzheimer’s Disease Assessment Scale-Cognition, Clinical Disease Rating-Sum of Boxes, Alzheimer’s Disease Cooperative Study–Activities of Daily Living Scale, and a memory composite. Results: The insulin-treated group demonstrated significantly reduced changes in white matter hyperintensity volume in deep and frontal regions after 12 months, with a similar trend for global volume. White matter hyperintensity volume progression correlated with worsened Alzheimer’s disease cerebral spinal fluid biomarker profile and cognitive function; however, patterns of correlations differed by treatment group. Conclusion: Intranasal insulin treatment for 12 months reduced white matter hyperintensity volume progression and supports insulin’s potential as a therapeutic option for Alzheimer’s disease.
- Published
- 2021
- Full Text
- View/download PDF
19. Traumatic brain injury and post-traumatic stress disorder are not associated with Alzheimer's disease pathology measured with biomarkers
- Author
-
Michael W, Weiner, Danielle, Harvey, Susan M, Landau, Dallas P, Veitch, Thomas C, Neylan, Jordan H, Grafman, Paul S, Aisen, Ronald C, Petersen, Clifford R, Jack, Duygu, Tosun, Leslie M, Shaw, John Q, Trojanowski, Andrew J, Saykin, Jacqueline, Hayes, and Charles, De Carli
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Abstract
Epidemiological studies report an association between traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD) and clinically diagnosed Alzheimer's disease (AD). We examined the association between TBI/PTSD and biomarker-defined AD.We identified 289 non-demented veterans with TBI and/or PTSD and controls who underwent clinical evaluation, cerebrospinal fluid (CSF) collection, magnetic resonance imaging (MRI), amyloid beta (Aβ) and tau positron emission tomography, and apolipoprotein E testing. Participants were followed for up to 5.2 years.Exposure groups (TBI, PTSD, and TBI + PTSD) had higher prevalence of mild cognitive impairment (MCI: P .0001) and worse Mini-Mental State Examination scores (PTSD: P = .008; TBIPTSD: P = .009) than controls. There were no significant differences in other cognitive scores, MRI volumes, Aβ or tau accumulation, or in most longitudinal measures.TBI and/or PTSD were not associated with elevated AD biomarkers. The poorer cognitive status of exposed veterans may be due to other comorbid pathologies.
- Published
- 2022
20. Randomised controlled trials for the prevention of cognitive decline or dementia: A systematic review
- Author
-
Nicola Coley, Caroline Giulioli, Paul S. Aisen, Bruno Vellas, and Sandrine Andrieu
- Subjects
Aging ,Cognition ,Neurology ,Cognitive Behavioral Therapy ,Alzheimer Disease ,Humans ,Cognitive Dysfunction ,Molecular Biology ,Biochemistry ,Life Style ,Biotechnology ,Randomized Controlled Trials as Topic - Abstract
Dementia prevention research has progressed rapidly in recent years, with publication of several large lifestyle intervention trials, and renewed interest in pharmacological interventions, notably for individuals with Alzheimer's disease biomarkers, warranting an updated review of results and methodology. We identified 112 completed trials testing the efficacy of single-domain pharmacological (n = 33, 29%), nutritional (n = 27, 24%), physical activity (n = 18, 16%) and cognitive stimulation (n = 13, 12%), or multidomain (n = 22, 20%) interventions on incident dementia, or a relevant intermediate marker (e.g. cognitive function, biomarkers or dementia risk scores) in people without dementia. The earliest trials tested pharmacological interventions or nutritional supplements, but lifestyle interventions predominated in the last decade. In total, 21 (19%) trials demonstrated a clear beneficial effect on the pre-specified primary outcome (or all co-primary outcomes), but only two (10%) were large-scale (testing blood pressure lowering (Syst-Eur) or multidomain (FINGER) interventions on incident dementia and cognitive change in cognitive function, respectively). Of the 116 ongoing trials, 40% (n = 46) are testing multidomain interventions. Recent methodological shifts concern target populations, primary outcome measures, and intervention design, but study design remains constant (parallel group randomised controlled trial). Future trials may consider using adaptive trials or interventions, and more targeted approaches, since certain interventions may be more effective in certain subgroups of the population, and at specific times in the life-course. Efforts should also be made to increase the representativeness and diversity of prevention trial populations.
- Published
- 2022
21. Meaningful benefits: a framework to assess disease-modifying therapies in preclinical and early Alzheimer's disease
- Author
-
Sheila Seleri, Assunção, Reisa A, Sperling, Craig, Ritchie, Diana R, Kerwin, Paul S, Aisen, Claire, Lansdall, Alireza, Atri, and Jeffrey, Cummings
- Subjects
Cognitive Neuroscience ,Alzheimer's disease ,Preclinical ,Cognition ,Clinical trials ,Neurology ,Alzheimer Disease ,Clinical meaningfulness ,Disease Progression ,Humans ,Cognitive Dysfunction ,Dementia ,Meaningful benefit ,Neurology (clinical) ,Mild cognitive impairment due to Alzheimer's disease ,Biomarkers - Abstract
Background The need for preventive therapies that interrupt the progression of Alzheimer’s disease (AD) before the onset of symptoms or when symptoms are emerging is urgent and has spurred the ongoing development of disease-modifying therapies (DMTs) in preclinical and early AD (mild cognitive impairment [MCI] to mild dementia). Assessing the meaningfulness of what are likely small initial treatment effects in these earlier stages of the AD patho-clinical disease continuum is a major challenge and warrants further consideration. Body To accommodate a shift towards earlier intervention in AD, we propose meaningful benefits as a new umbrella concept that encapsulates the spectrum of potentially desirable outcomes that may be demonstrated in clinical trials and other studies across the AD continuum, with an emphasis on preclinical AD and early AD (i.e., MCI due to AD and mild AD dementia). The meaningful benefits framework applies to data collection, assessment, and communication across three dimensions: (1) multidimensional clinical outcome assessments (COAs) including not only core disease outcomes related to cognition and function but also patient- and caregiver-reported outcomes, health and economic outcomes, and neuropsychiatric symptoms; (2) complementary analyses that help contextualize and expand the understanding of COA-based assessments, such as number-needed-to-treat or time-to-event analyses; and (3) assessment of both cumulative benefit and predictive benefit, where early changes on cognitive, functional, or biomarker assessments predict longer-term clinical benefit. Conclusion The concept of meaningful benefits emphasizes the importance of multidimensional reporting of clinical trial data while, conceptually, it advances our understanding of treatment effects in preclinical AD and mild cognitive impairment due to AD. We propose that such an approach will help bridge the gap between the emergence of DMTs and their clinical use, particularly now that a DMT is available for patients diagnosed with MCI due to AD and mild AD dementia.
- Published
- 2022
- Full Text
- View/download PDF
22. Initiation of symptomatic medication in Alzheimer's disease clinical trials: Hypothetical versus treatment policy approach
- Author
-
Fabian Model, Michael C. Donohue, Nicola Volye, Alzheimer’s Disease Neuroimaging Initiative, Michael S. Rafii, Paul S. Aisen, Paul Delmar, and Hong Liu-Seifert
- Subjects
Male ,medicine.medical_specialty ,Randomization ,Epidemiology ,Neuroimaging ,Disease ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Cognition ,0302 clinical medicine ,Developmental Neuroscience ,Alzheimer Disease ,Experimental therapy ,Humans ,Medicine ,Cognitive Dysfunction ,Prospective Studies ,030212 general & internal medicine ,Intensive care medicine ,Cognitive impairment ,Aged ,Clinical Trials as Topic ,business.industry ,Health Policy ,Clinical trial ,Psychiatry and Mental health ,Disease Progression ,Female ,Observational study ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,030217 neurology & neurosurgery - Abstract
In clinical trials in populations with mild cognitive impairment, it is common for participants to initiate concurrent symptomatic medications for Alzheimer's disease after randomization to the experimental therapy. One strategy for addressing this occurrence is to exclude any observations that occur after the concurrent medication is initiated. The rationale for this approach is that these observations might reflect a symptomatic benefit of the concurrent medication that would adversely bias efficacy estimates for an effective experimental therapy. We interrogate the assumptions underlying such an approach by estimating the effect of newly prescribed concurrent medications in an observational study, the Alzheimer's Disease Neuroimaging Initiative.
- Published
- 2020
- Full Text
- View/download PDF
23. Non-Amyloid Approaches to Disease Modification for Alzheimer’s Disease: An EU/US CTAD Task Force Report
- Author
-
Frank M. Longo, Rudolph E. Tanzi, Pierre N. Tariot, Bruno Vellas, Rema Raman, M. J. Detke, M. Weiner, Serge Gauthier, Jacques Touchon, Marwan N. Sabbagh, Lon S. Schneider, Paul S. Aisen, and Jeffrey L. Cummings
- Subjects
Combination therapy ,Amyloid ,Geroscience ,geroscience ,business.industry ,tauopathy ,lifestyle intervention ,Disease ,CTAD Task Force Paper ,medicine.disease ,neurotrophins ,neuroinflammation ,Drug development ,Disease modification ,photobiomodulation ,Medicine ,Dementia ,Tauopathy ,tau ,business ,Neuroscience ,Alzheimer’s disease ,dementia ,neurostimulation - Abstract
While amyloid-targeting therapies continue to predominate in the Alzheimer’s disease (AD) drug development pipeline, there is increasing recognition that to effectively treat the disease it may be necessary to target other mechanisms and pathways as well. In December 2019, The EU/US CTAD Task Force discussed these alternative approaches to disease modification in AD, focusing on tau-targeting therapies, neurotrophin receptor modulation, anti-microbial strategies, and the innate immune response; as well as vascular approaches, aging, and non-pharmacological approaches such as lifestyle intervention strategies, photobiomodulation and neurostimulation. The Task Force proposed a general strategy to accelerate the development of alternative treatment approaches, which would include increased partnerships and collaborations, improved trial designs, and further exploration of combination therapy strategies.
- Published
- 2020
24. RECRUITMENT INTO THE ALZHEIMER PREVENTION TRIALS (APT) WEBSTUDY FOR A TRIAL-READY COHORT FOR PRECLINICAL AND PRODROMAL ALZHEIMER’S DISEASE (TRCPAD)
- Author
-
Michael S. Rafii, Paul S. Aisen, E. J. Shaffer, Oliver Langford, Reisa A. Sperling, Jeffrey L. Cummings, Joshua D. Grill, Gustavo Jimenez-Maggiora, Sarah Walter, and T. B. Clanton
- Subjects
Male ,Aging ,medicine.medical_specialty ,Referral ,Clinical Trials and Supportive Activities ,Population ,web-based ,Prodromal Symptoms ,Disease ,Neurodegenerative ,Alzheimer's Disease ,Article ,prevention ,Clinical Research ,Alzheimer Disease ,Acquired Cognitive Impairment ,preclinical ,medicine ,Humans ,Longitudinal Studies ,Registries ,education ,Aged ,Internet ,education.field_of_study ,business.industry ,Patient Selection ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Cognition ,online registry ,Middle Aged ,Brain Disorders ,remote recruitment ,Clinical trial ,Observational Studies as Topic ,Trial-ready cohort ,Family medicine ,Cohort ,Biomarker (medicine) ,Dementia ,Female ,Observational study ,business ,Alzheimer’s disease - Abstract
BACKGROUND: The Alzheimer Prevention Trials (APT) Webstudy is the first stage in establishing a Trial-ready Cohort for Preclinical and Prodromal Alzheimer’s disease (TRC-PAD). This paper describes recruitment approaches for the APT Webstudy. Objectives: To remotely enroll a cohort of individuals into a web-based longitudinal observational study. Participants are followed quarterly with brief cognitive and functional assessments, and referred to Sites for in-clinic testing and biomarker confirmation prior to enrolling in the Trial-ready Cohort (TRC). Design: Participants are referred to the APT Webstudy from existing registries of individuals interested in brain health and Alzheimer’s disease research, as well as through central and site recruitment efforts. The study team utilizes Urchin Tracking Modules (UTM) codes to better understand the impact of electronic recruitment methods. Setting: A remotely enrolled online study. Participants: Volunteers who are at least 50 years old and interested in Alzheimer’s research. Measurements: Demographics and recruitment source of participant where measured by UTM. Results: 30,650 participants consented to the APT Webstudy as of April 2020, with 69.7% resulting from referrals from online registries. Emails sent by the registry to participants were the most effective means of recruitment. Participants are distributed across the US, and the demographics of the APT Webstudy reflect the referral registries, with 73.1% female, 85.0% highly educated, and 92.5% Caucasian. Conclusions: We have demonstrated the feasibility of enrolling a remote web-based study utilizing existing registries as a primary referral source. The next priority of the study team is to engage in recruitment initiatives that will improve the diversity of the cohort, towards the goal of clinical trials that better represent the US population.
- Published
- 2020
- Full Text
- View/download PDF
25. PREDICTING AMYLOID BURDEN TO ACCELERATE RECRUITMENT OF SECONDARY PREVENTION CLINICAL TRIALS
- Author
-
Oliver Langford, Reisa A. Sperling, Chung-Kai Sun, Paul S. Aisen, Jeffrey L. Cummings, Michael C. Donohue, Gustavo Jimenez-Maggiora, and Rema Raman
- Subjects
Male ,Oncology ,Apolipoprotein E ,medicine.medical_specialty ,Neurology ,Amyloid ,Prodromal Symptoms ,Disease ,Article ,Alzheimer Disease ,Internal medicine ,Secondary Prevention ,medicine ,Humans ,Mass Screening ,Cognitive Dysfunction ,Longitudinal Studies ,Aged ,Amyloid beta-Peptides ,Receiver operating characteristic ,business.industry ,Patient Selection ,Mental Status and Dementia Tests ,Cognitive test ,Clinical trial ,machine learning ,Cross-Sectional Studies ,Trial-ready cohort ,Cohort ,Female ,business ,Alzheimer’s disease - Abstract
BACKGROUND: Screening to identify individuals with elevated brain amyloid (Aβ+) for clinical trials in Preclinical Alzheimer’s Disease (PAD), such as the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s disease (A4) trial, is slow and costly. The Trial-Ready Cohort in Preclinical/Prodromal Alzheimer’s Disease (TRC-PAD) aims to accelerate and reduce costs of AD trial recruitment by maintaining a web-based registry of potential trial participants, and using predictive algorithms to assess their likelihood of suitability for PAD trials. OBJECTIVES: Here we describe how algorithms used to predict amyloid burden within TRC-PAD project were derived using screening data from the A4 trial. DESIGN: We apply machine learning techniques to predict amyloid positivity. Demographic variables, APOE genotype, and measures of cognition and function are considered as predictors. Model data were derived from the A4 trial. SETTING: TRC-PAD data are collected from web-based and in-person assessments and are used to predict the risk of elevated amyloid and assess eligibility for AD trials. PARTICIPANTS: Pre-randomization, cross-sectional data from the ongoing A4 trial are used to develop statistical models. MEASUREMENTS: Models use a range of cognitive tests and subjective memory assessments, along with demographic variables. Amyloid positivity in A4 was confirmed using positron emission tomography (PET). RESULTS: The A4 trial screened N=4,486 participants, of which N=1323 (29%) were classified as Aβ+ (SUVR ≥ 1.15). The Area under the Receiver Operating Characteristic curves for these models ranged from 0.60 (95% CI 0.56 to 0.64) for a web-based battery without APOE to 0.74 (95% CI 0.70 to 0.78) for an in-person battery. The number needed to screen to identify an Aβ+ individual is reduced from 3.39 in A4 to 2.62 in the remote setting without APOE, and 1.61 in the remote setting with APOE. CONCLUSIONS: Predictive algorithms in a web-based registry can improve the efficiency of screening in future secondary prevention trials. APOE status contributes most to predictive accuracy with cross-sectional data. Blood-based assays of amyloid will likely improve the prediction of amyloid PET positivity.
- Published
- 2020
- Full Text
- View/download PDF
26. THE TRIAL-READY COHORT FOR PRECLINICAL/PRODROMAL ALZHEIMER’S DISEASE (TRC-PAD) PROJECT: AN OVERVIEW
- Author
-
Paul S. Aisen, M. S. Rafii, Robert A. Rissman, S. Walter, Michael C. Donohue, O. Langford, T. B. Clanton, Reisa A. Sperling, Rema Raman, G. A. Jimenez-Maggiora, and Jeffrey L. Cummings
- Subjects
Apolipoprotein E ,medicine.medical_specialty ,Neurology ,Trial-Ready Cohort ,Prodromal Symptoms ,Disease ,Article ,Apolipoproteins E ,Drug Development ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,Longitudinal Studies ,Stage (cooking) ,Randomized Controlled Trials as Topic ,Amyloid beta-Peptides ,medicine.diagnostic_test ,Lumbar puncture ,business.industry ,Patient Selection ,Cognition ,Cohort ,Risk assessment ,business ,Alzheimer’s disease - Abstract
The Trial-Ready Cohort for Preclinical/prodromal Alzheimer’s Disease (TRC-PAD) project is a collaborative effort to establish an efficient mechanism for recruiting participants into very early stage Alzheimer’s disease trials. Clinically normal and mildly symptomatic individuals are followed longitudinally in a web-based component called the Alzheimer’s Prevention Trial Webstudy (APT Webstudy), with quarterly assessment of cognition and subjective concerns. The Webstudy data is used to predict the likelihood of brain amyloid elevation; individuals at relatively high risk are invited for in-person assessment in the TRC screeing phase, during which a cognitive battery is administered and Apolipoprotein E genotype is obtained followed by reassessment of risk of amyloid elevation. After an initial validation study, plasma amyloid peptide ratios will be included in this risk assessment. Based on this second risk calculation, individuals may have amyloid testing by PET scan or lumbar puncture, with those potentially eligible for trials followed in the TRC, while the rest are invited to remain in the APT Webstudy. To date, over 30,000 individuals have participated in the Webstudy; enrollment in the TRC is in its early stage.
- Published
- 2020
- Full Text
- View/download PDF
27. DATA-DRIVEN PARTICIPANT RECRUITMENT: FINDINGS FROM THE ALZHEIMER’S DISEASE NEUROIMAGING INITIATIVE 3
- Author
-
S. Moore, Juliet Fockler, Michael W. Weiner, Derek Flenniken, A. Ulbricht, Charissa Barger, W. Kwang, and Paul S. Aisen
- Subjects
Medical education ,Biomedical Research ,business.industry ,Patient Selection ,Neuroimaging ,Disease ,Data-driven ,Clinical trial ,Observational Studies as Topic ,Advertising ,Alzheimer Disease ,Phone ,Recruitment Activity ,Analytics ,Surveys and Questionnaires ,Humans ,Longitudinal Studies ,Recruitment methods ,Psychology ,business ,Alzheimer's Disease Neuroimaging Initiative - Abstract
Background: Effective and measurable participant recruitment methods are urgently needed for clinical studies in Alzheimer’s disease. Objectives: To develop methods for measuring recruitment tactics and evaluating effectiveness. Methods: Recruitment tactics for the Alzheimer’s Disease Neuroimaging Initiative (ADNI3) were measured using web and phone analytics, campaign metrics and survey responses. Results: A total of 462 new participants were enrolled into ADNI3 through recruitment efforts. We collected metrics on recruitment activities including 82,003 unique visitors to the recruitment website and 3,335 calls to study phone numbers. The recruitment sources that produced the most screening and enrollment included online advertisements, local radio and newspaper coverage and emails and referrals from registries. Conclusions: Analysis of recruitment activity obtained through tracking methods provided some insight for effective recruitment. ADNI3 can serve as an example of how a data-driven approach to centralized participant recruitment can be utilized to facilitate clinical research.
- Published
- 2020
- Full Text
- View/download PDF
28. TRC-PAD: Accelerating Recruitment of AD Clinical Trials through Innovative Information Technology
- Author
-
Jeffrey L. Cummings, Michael S. Rafii, Paul S. Aisen, Rema Raman, Oliver Langford, Reisa A. Sperling, Michael C. Donohue, S Bruschi, and Gustavo Jimenez-Maggiora
- Subjects
Information management ,Male ,medicine.medical_specialty ,Prodromal Symptoms ,Risk Assessment ,Article ,stomatognathic system ,Alzheimer Disease ,medicine ,informatics ,Humans ,Medical physics ,Effects of sleep deprivation on cognitive performance ,Longitudinal Studies ,Registries ,Aged ,clinical trials ,Clinical Trials as Topic ,business.industry ,Patient Selection ,Cognition ,Middle Aged ,Clinical trial ,Workflow ,recruitment ,Informatics ,Biomarker (medicine) ,Information technology architecture ,Female ,business ,Information Technology ,Alzheimer’s - Abstract
BACKGROUND: The Trial-Ready Cohort for Preclinical/Prodromal Alzheimer’s Disease (TRC-PAD) Informatics Platform (TRC-PAD IP) was developed to facilitate the efficient selection, recruitment, and assessment of study participants in support of the TRC-PAD program. Objectives: Describe the innovative architecture, workflows, and components of the TRC-PAD IP. Design: The TRC-PAD IP was conceived as a secure, scalable, multi-tiered information management platform designed to facilitate high-throughput, cost-effective selection, recruitment, and assessment of TRC-PAD study participants and to develop a learning algorithm to select amyloid-bearing participants to participate in trials of early-stage Alzheimer’s disease. Setting: TRC-PAD participants were evaluated using both web-based and in-person assessments to predict their risk of amyloid biomarker abnormalities and eligibility for preclinical and prodromal clinical trials. Participant data were integrated across multiple stages to inform the prediction of amyloid biomarker elevation. Participants: TRC-PAD participants were age 50 and above, with an interest in participating in Alzheimer’s research. Measurements: TRC-PAD participants’ cognitive performance and subjective memory concerns were remotely assessed on a longitudinal basis to predict participant risk of biomarker abnormalities. Those participants determined to be at the highest risk were invited to an in-clinic screening visit for a full battery of clinical and cognitive assessments and amyloid biomarker confirmation using positron emission tomography (PET) or lumbar puncture (LP). Results: The TRC-PAD IP supported growth in recruitment, screening, and enrollment of TRC-PAD participants by leveraging a secure, scalable, cost-effective cloud-based information technology architecture. Conclusions: The TRC-PAD program and its underlying information management infrastructure, TRC-PAD IP, have demonstrated feasibility concerning the program aims. The flexible and modular design of the TRC-PAD IP will accommodate the introduction of emerging diagnostic technologies.
- Published
- 2020
29. The Computerized Cognitive Composite (C3) in A4, an Alzheimer’s Disease Secondary Prevention Trial
- Author
-
Roy Yaari, Chung-Kai Sun, Kathryn V. Papp, Karen C. Holdridge, Reisa A. Sperling, Paul Maruff, Dorene M. Rentz, Michael A. Yassa, Adrian Schembri, Rema Raman, Michael C. Donohue, Paul S. Aisen, Alette M. Wessels, and Craig E.L. Stark
- Subjects
cognition ,Male ,Change over time ,medicine.medical_specialty ,Pattern separation ,Neurology ,Disease ,Audiology ,Asymptomatic ,Article ,Alzheimer Disease ,preclinical Alzheimer’s disease ,Secondary Prevention ,Humans ,Medicine ,Qualitative Research ,Aged ,Secondary prevention ,Clinical Trials as Topic ,Amyloid beta-Peptides ,Computers ,business.industry ,Cognition ,Mental Status and Dementia Tests ,Cross-Sectional Studies ,Quartile ,computerized testing ,Feasibility Studies ,Female ,Digital biomarkers ,medicine.symptom ,business ,Biomarkers - Abstract
Background: Computerized cognitive assessments may improve Alzheimer’s disease (AD) secondary prevention trial efficiency and accuracy. However, they require validation against standard outcomes and relevant biomarkers. Objective: To assess the feasibility and validity of the tablet-based Computerized Cognitive Composite (C3). Design: Cross-sectional analysis of cognitive screening data from the A4 study (Anti-Amyloid in Asymptomatic AD). Setting: Multi-center international study. Participants: Clinically normal (CN) older adults (65-85; n=4486). Measurements: Participants underwent florbetapir-Positron Emission Tomography for Aβ+/- classification. They completed the C3 and standard paper and pencil measures included in the Preclinical Alzheimer’s Cognitive Composite (PACC). The C3 combines memory measures sensitive to change over time (Cogstate Brief Battery-One Card Learning) and measures shown to be declining early in AD including pattern separation (Behavioral Pattern Separation Test- Object- Lure Discrimination Index) and associative memory (Face Name Associative Memory Exam- Face-Name Matching). C3 acceptability and completion rates were assessed using qualitative and quantitative methods. C3 performance was explored in relation to Aβ+/- groups (n=1323/3163) and PACC. Results: C3 was feasible for CN older adults to complete. Rates of incomplete or invalid administrations were extremely low, even in the bottom quartile of cognitive performers (PACC). C3 was moderately correlated with PACC (r=0.39). Aβ+ performed worse on C3 compared with Aβ- [unadjusted Cohen’s d=-0.22 (95%CI: -0.31,-0.13) p
- Published
- 2020
- Full Text
- View/download PDF
30. THE TRIAL-READY COHORT FOR PRECLINICAL AND PRODROMAL ALZHEIMER’S DISEASE (TRC-PAD): EXPERIENCE FROM THE FIRST 3 YEARS
- Author
-
T. B. Clanton, E. J. Shaffer, Gustavo Jimenez-Maggiora, Rema Raman, Michael S. Rafii, Paul S. Aisen, Jeffrey L. Cummings, Oliver Langford, Reisa A. Sperling, and Sarah Walter
- Subjects
Male ,Wechsler Memory Scale ,medicine.medical_specialty ,webstudy ,Prodromal Symptoms ,Neuropsychological Tests ,Article ,prevention ,Alzheimer Disease ,medicine ,Humans ,Longitudinal Studies ,Program Development ,Family history ,Aged ,Aged, 80 and over ,Clinical Trials as Topic ,Amyloid beta-Peptides ,Mini–Mental State Examination ,medicine.diagnostic_test ,business.industry ,Patient Selection ,remote study ,Wechsler Adult Intelligence Scale ,Middle Aged ,Clinical trial ,Cohort ,Physical therapy ,Female ,business ,Risk assessment ,Alzheimer’s disease ,Biomarkers ,Cohort study - Abstract
BACKGROUND: The Trial-Ready Cohort for Preclinical and Prodromal Alzheimer’s disease (TRC-PAD) aims to accelerate enrollment for Alzheimer’s disease (AD) clinical trials by remotely identifying and tracking individuals who are at high risk for developing symptoms of AD, and referring these individuals to in-person cognitive and biomarker evaluation with the purpose of engaging them in clinical trials. A risk algorithm using statistical modeling to predict brain amyloidosis will be refined as TRC-PAD advances with a maturing data set. Objectives: To provide a summary of the steps taken to build this Trial-Ready cohort (TRC) and share results of the first 3 years of enrollment into the program. Design: Participants are remotely enrolled in the Alzheimer Prevention Trials (APT) Webstudy with quarterly assessments, and through an algorithm identified as potentially at high risk, referred to clinical sites for biomarker confirmation, and enrolled into the TRC. Setting: Both an online study and in-clinic non-interventional cohort study. Participants: APT Webstudy participants are aged 50 or older, with an interest in participation in AD therapeutic trials. TRC participants must have a study partner, stable medical condition, and elevated brain amyloid, as measured by amyloid positron emission tomography or cerebrospinal fluid analysis. Additional risk assessments include apolipoprotein E genotyping. Measurements: In the APT Webstudy, participants complete the Cognitive Function Index and Cogstate Brief Battery. The TRC includes the Preclinical Alzheimer’s Cognitive Composite, comprised of the Free and Cued Selective Reminding Test, the Delayed Paragraph Recall score on the Logical Memory IIa test from the Wechsler Memory Scale, the Digit-Symbol Substitution test from the Wechsler Adult Intelligence Scale-Revised, and the Mini Mental State Examination total score (1). Results: During the first 3 years of this program, the APT Webstudy has 30,650 consented participants, with 23 sites approved for in person screening, 112 participants have been referred for in-clinic screening visits with eighteen enrolled to the TRC. The majority of participants consented to APT Webstudy have a family history of AD (62%), identify as Caucasian (92.5%), have over twelve years of formal education (85%), and are women (73%). Follow up rates for the first quarterly assessment were 38.2% with 29.5% completing the follow up Cogstate Battery. Conclusions: After successfully designing and implementing this program, the study team’s priority is to improve diversity of participants both in the APT Webstudy and TRC, to continue enrollment into the TRC to our target of 2,000, and to improve longitudinal retention, while beginning the process of referring TRC participants into clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
31. ATRI and ACTC: Academic Programs to Accelerate Alzheimer’s Disease Drug Development
- Author
-
Paul S. Aisen, Rema Raman, Michael S. Rafii, Reisa A. Sperling, and Ronald C. Petersen
- Published
- 2022
- Full Text
- View/download PDF
32. Early-stage Alzheimer disease: getting trial-ready
- Author
-
Paul S, Aisen, Gustavo A, Jimenez-Maggiora, Michael S, Rafii, Sarah, Walter, and Rema, Raman
- Subjects
Alzheimer Disease ,Brain ,Humans ,Prodromal Symptoms ,Longitudinal Studies ,Biomarkers - Abstract
Slowing the progression of Alzheimer disease (AD) might be the greatest unmet medical need of our time. Although one AD therapeutic has received a controversial accelerated approval from the FDA, more effective and accessible therapies are urgently needed. Consensus is growing that for meaningful disease modification in AD, therapeutic intervention must be initiated at very early (preclinical or prodromal) stages of the disease. Although the methods for such early-stage clinical trials have been developed, identification and recruitment of the required asymptomatic or minimally symptomatic study participants takes many years and requires substantial funds. As an example, in the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease Trial (the first phase III trial to be performed in preclinical AD), 3.5 years and more than 5,900 screens were required to recruit and randomize 1,169 participants. A new clinical trials infrastructure is required to increase the efficiency of recruitment and accelerate therapeutic progress. Collaborations in North America, Europe and Asia are now addressing this need by establishing trial-ready cohorts of individuals with preclinical and prodromal AD. These collaborations are employing innovative methods to engage the target population, assess risk of brain amyloid accumulation, select participants for biomarker studies and determine eligibility for trials. In the future, these programmes could provide effective tools for pursuing the primary prevention of AD. Here, we review the lessons learned from the AD trial-ready cohorts that have been established to date, with the aim of informing ongoing and future efforts towards efficient, cost-effective trial recruitment.
- Published
- 2022
33. Screening and enrollment of underrepresented ethnocultural and educational populations in the Alzheimer's Disease Neuroimaging Initiative (ADNI)
- Author
-
Miriam T, Ashford, Rema, Raman, Garrett, Miller, Michael C, Donohue, Ozioma C, Okonkwo, Monica Rivera, Mindt, Rachel L, Nosheny, Godfrey A, Coker, Ronald C, Petersen, Paul S, Aisen, and Michael W, Weiner
- Abstract
An analysis of the ethnocultural and socioeconomic composition of Alzheimer's Disease Neuroimaging Initiative (ADNI) participants is needed to assess the generalizability of ADNI data to diverse populations.ADNI data collected between October 2004 and November 2020 were used to determine ethnocultural and educational composition of the sample and differences in the following metrics: screening, screen fails, enrollment, biomarkers.Of 3739 screened individuals, 11% identified as being from ethnoculturally underrepresented populations (e.g., Black, Latinx) and 16% had12 years of education. Of 2286 enrolled participants, 11% identified as ethnoculturally underrepresented individuals and 15% had12 years of education. This participation is considerably lower than US Census data for adults 60+ (ethnoculturally underrepresented populations: 25%;12 years of education: 4%). Individuals with12 years of education failed screening at a higher rate.Our findings suggest that ADNI results may not be entirely generalizable to ethnoculturally diverse and low education populations.
- Published
- 2022
34. Corrigendum to: ATRI EDC: a novel cloud-native remote data capture system for large multicenter Alzheimer’s disease and Alzheimer’s disease-related dementias clinical trials
- Author
-
Gustavo A Jimenez-Maggiora, Stefania Bruschi, Hongmei Qiu, Jia-Shing So, and Paul S Aisen
- Subjects
Health Informatics - Published
- 2022
- Full Text
- View/download PDF
35. Impact of sex and APOE ε4 on the association of cognition and hippocampal volume in clinically normal, amyloid positive adults
- Author
-
Kellen K. Petersen, Ellen Grober, Richard B. Lipton, Reisa A. Sperling, Rachel F. Buckley, Paul S. Aisen, and Ali Ezzati
- Subjects
Psychiatry and Mental health ,Neurology (clinical) - Published
- 2022
- Full Text
- View/download PDF
36. AHEAD 3‐45 study: Preliminary screening and baseline characteristics from a placebo‐controlled, double‐blind study evaluating lecanemab in participants with preclinical Alzheimer’s disease and elevated (A45 trial) and intermediate (A3 trial) amyloid
- Author
-
Jin Zhou, Michael C Irizarry, Lynn D Kramer, Chad J Swanson, Claire Roberts, Shobha Dhadda, David JianJun Li, Martin Rabe, Stephen Krause, Rema Raman, Michael C Donohue, Gopalan Sethuraman, Keith A. Johnson, Reisa A. Sperling, and Paul S Aisen
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
37. The screening and enrollment of underrepresented ethnoracial and educational populations in the Alzheimer's Disease Neuroimaging Initiative
- Author
-
Miriam T Ashford, Garrett Miller, Rema Raman, Michael C Donohue, Ozioma C Okonkwo, Monica Rivera Mindt, Rachel L Nosheny, Ronald C Petersen, Paul S Aisen, and Mike W Weiner
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
38. Individual patient data meta‐analysis assessing the adverse event profile of Alzheimer dementia randomized clinical trials
- Author
-
Oliver Langford, Jeffrey L Cummings, Lon S Schneider, Michael S Rafii, Karin Ernstrom, Paul S Aisen, and Rema Raman
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
39. Cognitive, neuropsychiatric and imaging comparisons between early‐onset and late‐onset Alzheimer’s disease participants from LEADS and ADNI3
- Author
-
Liana G. Apostolova, Ani Eloyan, Sujuan Gao, Leonardo Iaccarino, Alexandra Touroutoglou, Paul S Aisen, Laurel Beckett, Bret J Borowski, Michael C Donohue, Anne M Fagan, Tatiana M. Foroud, Constantine Gatsonis, Clifford R. Jack, Joel H Kramer, Robert A. Koeppe, Andrew J. Saykin, Arthur W. Toga, Prashanthi Vemuri, Gregory S Day, Neill R. Graff‐Radford, Lawrence S Honig, David T. Jones, Joseph C Masdeu, Mario Mendez, Chiadi U Onyike, Emily J Rogalski, Stephen P. Salloway, David A. Wolk, Thomas S. Wingo, Maria C. Carrillo, Gil D. Rabinovici, and Brad C. Dickerson
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
40. Using Digital Tools to Advance Alzheimer's Drug Trials During a Pandemic: The EU/US CTAD Task Force
- Author
-
Jeffrey Kaye, Maria C. Carrillo, Gustavo Jimenez-Maggiora, Simon Lovestone, Bruno Vellas, Kathryn V. Papp, Paul S. Aisen, Howard Fillit, Maria Soto, Rhoda Au, Takeshi Iwatsubo, Rebecca E. Amariglio, F. Natanegara, M. Weiner, and Clive Ballard
- Subjects
Clinical Trials as Topic ,Digital Technology ,Biomedical Research ,Standardization ,Computer science ,Advisory Committees ,COVID-19 ,Cognition ,Disease ,CTAD Task Force Paper ,Data science ,remote assessments ,clinical outcomes ,United States ,Clinical trial ,Data sharing ,Clinical research ,Alzheimer Disease ,Pandemic ,Humans ,Generalizability theory ,European Union ,Alzheimer’s disease ,digital tools - Abstract
The 2020 COVID-19 pandemic has disrupted Alzheimer’s disease (AD) clinical studies worldwide. Digital technologies may help minimize disruptions by enabling remote assessment of subtle cognitive and functional changes over the course of the disease. The EU/US Clinical Trials in Alzheimer’s Disease (CTAD) Task Force met virtually in November 2020 to explore the opportunities and challenges associated with the use of digital technologies in AD clinical research. While recognizing the potential of digital tools to accelerate clinical trials, improve the engagement of diverse populations, capture clinically meaningful data, and lower costs, questions remain regarding the stability, validity, generalizability, and reproducibility of digital data. Substantial concerns also exist regarding regulatory acceptance and privacy. Nonetheless, the Task Force supported further exploration of digital technologies through collaboration and data sharing, noting the need for standardization of digital readouts. They also concluded that while it may be premature to employ remote assessments for trials of novel experimental medications, remote studies of non-invasive, multi-domain approaches may be feasible at this time.
- Published
- 2021
41. ATRI EDC: a novel cloud-native remote data capture system for large multicenter Alzheimer's disease and Alzheimer's disease-related dementias clinical trails
- Author
-
Gustavo A Jimenez-Maggiora, Stefania Bruschi, Hongmei Qiu, Jia-Shing So, and Paul S Aisen
- Subjects
Health Informatics - Abstract
Objective The Alzheimer’s Therapeutic Research Institute (ATRI) developed a novel clinical data management system, the ATRI electronic data capture system (ATRI EDC), to address the complex regulatory, operational, and data requirements that arise in the conduct of multicenter Alzheimer’s disease and Alzheimer’s disease-related dementias (AD/ADRDs) clinical trials. We describe the system, its utility, and the broader implications for the field of clinical trials and clinical research informatics. Materials and Methods The ATRI EDC system was developed, tested, and validated using community-based agile software development methods and cloud-native single-page application design principles. It offers an increasing number of application modules, supports a high degree of study-specific configuration, and empowers study teams to effectively communicate and collaborate on the accurate and timely completion of study activities. Results To date, the ATRI EDC system supports 10 clinical studies, collecting study data for 4596 participants. Three case descriptions further illustrate how the system’s capabilities support diverse study-specific requirements. Discussion The ATRI EDC system has several advantages: its modular capabilities can accommodate rapidly evolving research designs and technologies; its community-based agile development approach and community-friendly licensing model encourage collaboration per the principles of open science; finally, with continued development and community building efforts, the system has the potential to facilitate the effective conduct of clinical studies beyond the field of AD/ADRD. Conclusion By effectively addressing the requirements of multicenter AD/ADRD studies, the ATRI EDC system supports ATRI’s scientific mission of rigorously testing new AD/ADRD therapies and facilitating the effective conduct of multicenter clinical studies.
- Published
- 2021
42. Impact of sex and
- Author
-
Kellen K, Petersen, Ellen, Grober, Richard B, Lipton, Reisa A, Sperling, Rachel F, Buckley, Paul S, Aisen, and Ali, Ezzati
- Abstract
Cognitive decline follows pathological changes including neurodegeneration on the Alzheimer's disease continuum. However, it is unclear which cognitive domains first become affected by neurodegeneration in amyloid-positive individuals and if sex or apolipoprotein (Data from 1233 cognitively unimpaired, amyloid-positive individuals 65 to 85 years of age were studied to assess the effect of hippocampal volume (HV) on cognition and to evaluate differences due to sex andLower HV was linked with worse performance on measures of memory (free recall, total recall, logical memory delayed recall, Mini-Mental State Examination [MMSE]), executive functioning (digit symbol substitution, DSS), and the Preclinical Alzheimer's Cognitive Composite (PACC). Among both women andDespite all cognitive measures being associated with HV, cognitive tests show differences in detecting early or late signs of neurodegeneration. Differences exist in association between cognition and neurodegeneration based on sex and
- Published
- 2021
43. Item-Level Investigation of Participant and Study Partner Report on the Cognitive Function Index from the A4 Study Screening Data
- Author
-
John R. Sims, Jennifer R. Gatchel, Gad A. Marshall, Rema Raman, Roy Yaari, Rebecca E. Amariglio, Paul S. Aisen, Karen C. Holdridge, Chung-Kai Sun, Dorene M. Rentz, Rachel F. Buckley, Sietske A.M. Sikkes, Joshua D. Grill, Keith A. Johnson, Michael C. Donohue, Reisa A. Sperling, Neurology, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
Male ,Amyloid ,Population ,Amyloid pet ,Neuropsychological Tests ,Article ,Cognition ,Subjective cognitive cecline ,Alzheimer Disease ,Surveys and Questionnaires ,Cognitive Changes ,Medicine ,Humans ,Cognitive skill ,education ,Spouses ,Aged ,Aged, 80 and over ,education.field_of_study ,Aniline Compounds ,business.industry ,Subjective report ,clinical trial ,Odds ratio ,Middle Aged ,Healthy Volunteers ,Clinical trial ,Positron-Emission Tomography ,Ethylene Glycols ,Female ,Self Report ,business ,Alzheimer’s disease ,Clinical psychology - Abstract
Background: Greater subjective cognitive changes on the Cognitive Function Index (CFI) was previously found to be associated with elevated amyloid (Aß) status in participants screening for the A4 Study, reported by study partners and the participants themselves. While the total score on the CFI related to amyloid for both sources respectively, potential differences in the specific types of cognitive changes reported by either participants or their study partners was not investigated. Objectives: To determine the specific types of subjective cognitive changes endorsed by participants and their study partners that are associated with amyloid status in individuals screening for an AD prevention trial. Design, Setting, Participants: Four thousand four hundred and eighty-six cognitively unimpaired (CDR=0; MMSE 25-30) participants (ages 65-85) screening for the A4 Study completed florbetapir (Aß) Positron Emission Tomography (PET) imaging. Participants were classified as elevated amyloid (Aß+; n=1323) or non-elevated amyloid (Aß-; n=3163). Measurements: Prior to amyloid PET imaging, subjective report of changes in cognitive functioning were measured using the CFI (15 item questionnaire; Yes/Maybe/No response options) and administered separately to both participants and their study partners (i.e., a family member or friend in regular contact with the participant). The impact of demographic factors on CFI report was investigated. For each item of the CFI, the relationship between Aß and CFI response was investigated using an ordinal mixed effects model for participant and study partner report. Results: Independent of Aß status, participants were more likely to report ‘Yes’ or ‘Maybe’ compared to the study partners for nearly all CFI items. Older age (r= 0.06, p
- Published
- 2021
- Full Text
- View/download PDF
44. A randomized clinical trial to evaluate home‐based assessment of people over 75 years old
- Author
-
Carolyn W. Zhu, Mary Sano, Steven H. Ferris, James C. Mundt, Lon S. Schneider, Jeffrey Kaye, Yanxin Jiang, Howard Feldman, Michael C. Donohue, Ronald G. Thomas, Tamara L. Hayes, Paul S. Aisen, Chung-Kai Sun, and Susan Egelko
- Subjects
Male ,Gerontology ,business.product_category ,Epidemiology ,education ,Neuropsychological Tests ,Interactive kiosk ,law.invention ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Randomized controlled trial ,law ,Surveys and Questionnaires ,Clinical endpoint ,Humans ,Medicine ,Dementia ,030212 general & internal medicine ,Stable group ,Geriatric Assessment ,Dropout (neural networks) ,Aged ,Aged, 80 and over ,business.industry ,Health Policy ,Cognition ,medicine.disease ,Home Care Services ,Healthy Volunteers ,Telephone ,Psychiatry and Mental health ,Feasibility Studies ,Female ,The Internet ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,030217 neurology & neurosurgery - Abstract
Introduction There is an unmet need for effective methods for conducting dementia prevention trials. Methods Home-based assessment study compared feasibility and efficiency, ability to capture change over time using in-home instruments, and ability to predict cognitive conversion using predefined triggers in a randomized clinical trial in (1) mail-in questionnaire/live telephone interviews, (2) automated telephone/interactive voice recognition, and (3) internet-based computer Kiosk technologies. Primary endpoint was defined as cognitive conversion. Results Analysis followed a modified intent-to-treat principle. Dropout rates were low and similar across technologies but participants in Kiosk were more likely to dropout earlier. Staff resources needed were higher in Kiosk. In-home instruments distinguished conversion and stable groups. Cognitively stable group showed improvement in cognitive measures. Triggering was associated with higher likelihood of conversion but statistically significant only in mail-in questionnaire/live telephone interviews. Discussion Relatively low efficiency of internet-based assessment compared with testing by live-assessors has implications for internet-based recruitment and assessment efforts currently proposed for diverse populations.
- Published
- 2019
- Full Text
- View/download PDF
45. RANDOMIZED, PLACEBO CONTROLLED TRIAL OF NPT088, A PHAGE-DERIVED, AMYLOID-TARGETED TREATMENT FOR ALZHEIMER’S DISEASE
- Author
-
Jonathan M. Levenson, Michael Grundman, Richard Fisher, Paul S. Aisen, K. Magnuson, Franz Hefti, D. Michelson, K. Marek, and Martha L. Gray
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Neurology ,Amyloid ,Recombinant Fusion Proteins ,Placebo-controlled study ,Plaque, Amyloid ,tau Proteins ,Disease ,Clinical study ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,In patient ,Aged ,Aged, 80 and over ,Amyloid beta-Peptides ,Aniline Compounds ,Dose-Response Relationship, Drug ,medicine.diagnostic_test ,business.industry ,Brain ,Middle Aged ,Immunoglobulin Fc Fragments ,Tolerability ,Positron emission tomography ,Positron-Emission Tomography ,Ethylene Glycols ,Female ,business ,Bacteriophage M13 - Abstract
The engineered fusion protein NPT088 targets amyloid in vitro and in animal models of Alzheimer’s disease. Previous studies showed that NPT088 treatment reduced β-amyloid plaque and tau aggregate loads in mouse disease models. Here, we present the results from an initial clinical study of NPT088 in patients with mild to moderate Alzheimer’s disease. Patients were treated with 4 dose levels of NPT088 for 6 months to evaluate its safety and tolerability. Exploratory measurements included measurement of change in β-amyloid plaque and tau burden utilizing Positron Emission Tomography imaging as well as measures of Alzheimer’s disease symptoms. At endpoint NPT088 was generally safe and well-tolerated with the most prominent finding being infusion reactions in a minority of patients. No effect of NPT088 on brain plaques, tau aggregates or Alzheimer’s disease symptoms was observed.
- Published
- 2019
- Full Text
- View/download PDF
46. PLASMA BIOMARKERS OF AD EMERGING AS ESSENTIAL TOOLS FOR DRUG DEVELOPMENT: AN EU/US CTAD TASK FORCE REPORT
- Author
-
Susan L. Hendrix, Henrik Zetterberg, Kaj Blennow, Bruno Vellas, Simon Lovestone, Rachel Schindler, Paul S. Aisen, Stephen Salloway, Randall J. Bateman, Rachelle Doody, and M. Weiner
- Subjects
medicine.medical_specialty ,Amyloid beta-Peptides ,Neurology ,medicine.diagnostic_test ,biology ,business.industry ,Amyloid beta ,Advisory Committees ,Drug Evaluation, Preclinical ,tau Proteins ,Disease ,Omics ,Bioinformatics ,Clinical trial ,Drug Development ,Drug development ,Alzheimer Disease ,Neurofilament Proteins ,biology.protein ,Humans ,Medicine ,Blood test ,Biomarker (medicine) ,business ,Biomarkers - Abstract
There is an urgent need to develop reliable and sensitive blood-based biomarkers of Alzheimer’s disease (AD) that can be used for screening and to increase the efficiency of clinical trials. The European Union-North American Clinical Trials in Alzheimer’s Disease Task Force (EU/US CTAD Task Force) discussed the current status of blood-based AD biomarker development at its 2018 annual meeting in Barcelona, Spain. Recent improvements in technologies to assess plasma levels of amyloid beta indicate that a single sample of blood could provide an accurate estimate of brain amyloid positivity. Plasma neurofilament light protein appears to provide a good marker of neurodegeneration, although not specific for AD. Plasma tau shows some promising results but weak or no correlation with CSF tau levels, which may reflect rapid clearance of tau in the bloodstream. Blood samples analyzed using -omics and other approaches are also in development and may provide important insight into disease mechanisms as well as biomarker profiles for disease prediction. To advance these technologies, international multidisciplinary, multi-stakeholder collaboration is essential.
- Published
- 2019
- Full Text
- View/download PDF
47. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering
- Author
-
Alireza Atri, Jeffrey L. Cummings, Stephen Salloway, Cynthia A. Lemere, Paul S. Aisen, and Marwan N. Sabbagh
- Subjects
medicine.medical_specialty ,Amyloid ,Neurology ,medicine.drug_class ,Donanemab ,Cognitive Neuroscience ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Amyloidogenic Proteins ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Lecanemab ,Viewpoint ,Clinical trials ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,Aducanumab ,RC346-429 ,Amyloid beta-Peptides ,Geriatrics gerontology ,business.industry ,Clinical trial ,Gantenerumab ,Monoclonal antibodies ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,business ,Geriatric psychiatry ,FDA ,RC321-571 ,medicine.drug - Published
- 2021
48. The Impact of Amyloid Burden and APOE on Rates of Cognitive Impairment in Late Life Depression
- Author
-
Arthur W. Toga, Philip S. Insel, Craig Nelson, Howie Rosen, R. Scott Mackin, Susan M. Landau, Paul S. Aisen, Duygu Tosun, Ruth T. Morin, Alzheimer’s Disease Neuroimaging Initiative, Adni Depression, Andrew J. Saykin, Meryl A. Butters, Devon Gessert, Michael W. Weiner, Emma Rhodes, Rema Raman, David Bickford, and Clifford R. Jack
- Subjects
Male ,medicine.medical_specialty ,Neurology ,Trail Making Test ,Apolipoprotein E4 ,Disease ,Audiology ,Verbal learning ,Article ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Memory impairment ,Humans ,Cognitive Dysfunction ,Depression (differential diagnoses) ,Aged ,Depressive Disorder, Major ,Amyloid beta-Peptides ,business.industry ,General Neuroscience ,Cognition ,General Medicine ,Late life depression ,Middle Aged ,030227 psychiatry ,Psychiatry and Mental health ,Clinical Psychology ,Female ,Geriatrics and Gerontology ,business ,030217 neurology & neurosurgery - Abstract
Background: Cognitive impairment (CI) is a key feature of late life depression (LLD), but the contribution of underlying neurodegenerative pathology remains unclear. Objective: To evaluate cognitive dysfunction in LLD relative to a sample of nondepressed (ND) older adults with matched levels of memory impairment and amyloid-β (Aβ) burden. Methods: Participants included 120 LLD and 240 ND older adults matched on age, education, sex, Mini-Mental State Exam, mild cognitive impairment diagnosis, and PET Aβ burden. Results: LLD showed higher rates of impairment relative to ND with 54.6% of the LLD sample demonstrating impairment in at least one cognitive domain compared to 42.9% of controls (H = 7.13, p = 0.008). LLD had poorer performance and higher rates of impairment on Rey Auditory Verbal Learning Test learning and memory compared to controls. In the overall sample, Aβ positivity was associated with worse performance on Logical Memory I (p = 0.044), Logical Memory II (p = 0.011), and Trail Making Test –B (p = 0.032), and APOE ɛ4 genotype was associated with worse performance on Logical Memory I (p = 0.022); these relationships did not differ between LLD and ND. Conclusion: LLD showed higher rates of CI driven by focal deficits in verbal learning and memory. Alzheimer’s disease (AD) biomarkers were associated with worse performance on timed set-shifting and story learning and memory, and these relationships were not impacted by depression status. These findings suggest that AD may account for a portion of previously reported multi-domain CI in LLD and highlight the potential for AD to confound studies of cognition in LLD.
- Published
- 2021
49. Brain amyloid burden, sleep, and 24-hour rest/activity rhythms: screening findings from the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration Studies
- Author
-
Paul S. Aisen, Gad A. Marshall, Mark N. Wu, Vadim Zipunnikov, John J. Kim, Jiawei Bai, Roy Yaari, Paul B. Rosenberg, Anton P. Porsteinsson, Cynthia M. Carlsson, Jiyoon Choi, Adam P. Spira, Sarah K. Wanigatunga, Jacobo Mintzer, Junrui Di, Reisa A. Sperling, and Rema Raman
- Subjects
Oncology ,medicine.medical_specialty ,Amyloid ,Disease ,rhythm ,Asymptomatic ,cognitive ,Internal medicine ,wrist ,medicine ,Circadian rhythm ,AcademicSubjects/MED00385 ,sleep ,Morning ,AcademicSubjects/SCI01870 ,business.industry ,activity ,Neurodegeneration ,amyloid ,Actigraphy ,Original Articles ,General Medicine ,medicine.disease ,Clinical trial ,circadian ,AcademicSubjects/MED00310 ,medicine.symptom ,business ,Alzheimer’s disease ,AcademicSubjects/MED00370 ,actigraphy - Abstract
Study Objectives To examine in a subsample at the screening phase of a clinical trial of a β-amyloid (Aβ) antibody whether disturbed sleep and altered 24-hour rest/activity rhythms (RARs) may serve as markers of preclinical Alzheimer’s disease (AD). Methods Overall, 26 Aβ-positive (Aβ+) and 33 Aβ-negative (Aβ−) cognitively unimpaired participants (mean age = 71.3 ± 4.6 years, 59% women) from the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s (A4) and the Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) studies, respectively, wore actigraphs for 5.66 ± 0.88 24-hour periods. We computed standard sleep parameters, standard RAR metrics (mean estimating statistic of rhythm, amplitude, acrophase, interdaily stability, intradaily variability, relative amplitude), and performed a novel RAR analysis (function-on-scalar regression [FOSR]). Results We were unable to detect any differences between Aβ+ and Aβ− participants in standard sleep parameters or RAR metrics with our sample size. When we used novel FOSR methods, however, Aβ+ participants had lower activity levels than Aβ− participants in the late night through early morning (11:30 pm to 3:00 am), and higher levels in the early morning (4:30 am to 8:30 am) and from midday through late afternoon (12:30 pm to 5:30 pm; all p < .05). Aβ+ participants also had higher variability in activity across days from 9:30 pm to 1:00 am and 4:30 am to 8:30 am, and lower variability from 2:30 am to 3:30 am (all p < .05). Conclusions Although we found no association of preclinical AD with standard actigraphic sleep or RAR metrics, a novel data-driven analytic method identified temporally “local” RAR alterations in preclinical AD.
- Published
- 2021
- Full Text
- View/download PDF
50. The Amyloid-β Pathway in Alzheimer's Disease
- Author
-
Michele Vendruscolo, Paul S. Aisen, Takeshi Iwatsubo, George Perry, John Hardy, Lars Lannfelt, Jeffrey L. Cummings, Min Cho, Andrea Vergallo, Kaj Blennow, Colin L. Masters, Victor L. Villemagne, Harald Hampel, Christopher Chen, and Seung Hyun Kim
- Subjects
Amyloid β ,Neurologi ,tau Proteins ,Diseases ,Disease ,Review Article ,Clinical onset ,Cellular and Molecular Neuroscience ,Neurochemical ,Basic research ,Alzheimer Disease ,Medicine ,Humans ,Molecular Biology ,Amyloid beta-Peptides ,business.industry ,Neurodegeneration ,Diagnostic markers ,medicine.disease ,Molecular medicine ,Psychiatry and Mental health ,Neurology ,Biomarker (medicine) ,business ,Neuroscience ,Biomarkers - Abstract
Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer’s disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.