Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047224

ABSTRACT

There is a large unmet medical need to develop disease-modifying treatment options for individuals with age-related degenerative diseases of the central nervous system. The sigma-2 receptor (S2R), encoded by TMEM97, is expressed in brain and retinal cells, and regulates cell functions via its co-receptor progesterone receptor membrane component 1 (PGRMC1), and through other protein-protein interactions. Studies describing functions of S2R involve the manipulation of expression or pharmacological modulation using exogenous small-molecule ligands. These studies demonstrate that S2R modulates key pathways involved in age-related diseases including autophagy, trafficking, oxidative stress, and amyloid-ß and α-synuclein toxicity. Furthermore, S2R modulation can ameliorate functional deficits in cell-based and animal models of disease. This review summarizes the current evidence-based understanding of S2R biology and function, and its potential as a therapeutic target for age-related degenerative diseases of the central nervous system, including Alzheimer's disease, α-synucleinopathies, and dry age-related macular degeneration.


Subject(s)
Alzheimer Disease , Lewy Body Disease , Receptors, sigma , Animals , Alzheimer Disease/drug therapy , Receptors, sigma/metabolism , alpha-Synuclein/metabolism , Amyloid beta-Peptides , Biology
2.
Alzheimers Dement ; 17(8): 1365-1382, 2021 08.
Article in English | MEDLINE | ID: mdl-33559354

ABSTRACT

INTRODUCTION: Amyloid beta (Aß) oligomers are one of the most toxic structural forms of the Aß protein and are hypothesized to cause synaptotoxicity and memory failure as they build up in Alzheimer's disease (AD) patients' brain tissue. We previously demonstrated that antagonists of the sigma-2 receptor complex effectively block Aß oligomer toxicity. CT1812 is an orally bioavailable, brain penetrant small molecule antagonist of the sigma-2 receptor complex that appears safe and well tolerated in healthy elderly volunteers. We tested CT1812's effect on Aß oligomer pathobiology in preclinical AD models and evaluated CT1812's impact on cerebrospinal fluid (CSF) protein biomarkers in mild to moderate AD patients in a clinical trial (ClinicalTrials.gov NCT02907567). METHODS: Experiments were performed to measure the impact of CT1812 versus vehicle on Aß oligomer binding to synapses in vitro, to human AD patient post mortem brain tissue ex vivo, and in living APPSwe /PS1dE9 transgenic mice in vivo. Additional experiments were performed to measure the impact of CT1812 versus vehicle on Aß oligomer-induced deficits in membrane trafficking rate, synapse number, and protein expression in mature hippocampal/cortical neurons in vitro. The impact of CT1812 on cognitive function was measured in transgenic Thy1 huAPPSwe/Lnd+ and wild-type littermates. A multicenter, double-blind, placebo-controlled parallel group trial was performed to evaluate the safety, tolerability, and impact on protein biomarker expression of CT1812 or placebo given once daily for 28 days to AD patients (Mini-Mental State Examination 18-26). CSF protein expression was measured by liquid chromatography with tandem mass spectrometry or enzyme-linked immunosorbent assay in samples drawn prior to dosing (Day 0) and at end of dosing (Day 28) and compared within each patient and between pooled treated versus placebo-treated dosing groups. RESULTS: CT1812 significantly and dose-dependently displaced Aß oligomers bound to synaptic receptors in three independent preclinical models of AD, facilitated oligomer clearance into the CSF, increased synaptic number and protein expression in neurons, and improved cognitive performance in transgenic mice. CT1812 significantly increased CSF concentrations of Aß oligomers in AD patient CSF, reduced concentrations of synaptic proteins and phosphorylated tau fragments, and reversed expression of many AD-related proteins dysregulated in CSF. DISCUSSION: These preclinical studies demonstrate the novel disease-modifying mechanism of action of CT1812 against AD and Aß oligomers. The clinical results are consistent with preclinical data and provide evidence of target engagement and impact on fundamental disease-related signaling pathways in AD patients, supporting further development of CT1812.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognition/drug effects , Mice, Transgenic , Receptors, sigma/antagonists & inhibitors , Aged , Animals , Brain/metabolism , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , Humans , Male , Mice , Neurons/metabolism , Synapses/metabolism
3.
Mov Disord ; 33(8): 1287-1297, 2018 08.
Article in English | MEDLINE | ID: mdl-29701258

ABSTRACT

BACKGROUND: Ubiquitous digital technologies such as smartphone sensors promise to fundamentally change biomedical research and treatment monitoring in neurological diseases such as PD, creating a new domain of digital biomarkers. OBJECTIVES: The present study assessed the feasibility, reliability, and validity of smartphone-based digital biomarkers of PD in a clinical trial setting. METHODS: During a 6-month, phase 1b clinical trial with 44 Parkinson participants, and an independent, 45-day study in 35 age-matched healthy controls, participants completed six daily motor active tests (sustained phonation, rest tremor, postural tremor, finger-tapping, balance, and gait), then carried the smartphone during the day (passive monitoring), enabling assessment of, for example, time spent walking and sit-to-stand transitions by gyroscopic and accelerometer data. RESULTS: Adherence was acceptable: Patients completed active testing on average 3.5 of 7 times/week. Sensor-based features showed moderate-to-excellent test-retest reliability (average intraclass correlation coefficient = 0.84). All active and passive features significantly differentiated PD from controls with P < 0.005. All active test features except sustained phonation were significantly related to corresponding International Parkinson and Movement Disorder Society-Sponsored UPRDS clinical severity ratings. On passive monitoring, time spent walking had a significant (P = 0.005) relationship with average postural instability and gait disturbance scores. Of note, for all smartphone active and passive features except postural tremor, the monitoring procedure detected abnormalities even in those Parkinson participants scored as having no signs in the corresponding International Parkinson and Movement Disorder Society-Sponsored UPRDS items at the site visit. CONCLUSIONS: These findings demonstrate the feasibility of smartphone-based digital biomarkers and indicate that smartphone-sensor technologies provide reliable, valid, clinically meaningful, and highly sensitive phenotypic data in Parkinson's disease. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/therapeutic use , Motor Activity/physiology , Outcome Assessment, Health Care/methods , Parkinson Disease/diagnosis , Parkinson Disease/physiopathology , Smartphone , Aged , Case-Control Studies , Feasibility Studies , Female , Humans , Male , Middle Aged , Neurologic Examination , Parkinson Disease/psychology , Patient Compliance/psychology , Psychomotor Performance , Reproducibility of Results , Severity of Illness Index , Time Factors
4.
N Engl J Med ; 370(4): 322-33, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24450891

ABSTRACT

BACKGROUND: Bapineuzumab, a humanized anti-amyloid-beta monoclonal antibody, is in clinical development for the treatment of Alzheimer's disease. METHODS: We conducted two double-blind, randomized, placebo-controlled, phase 3 trials involving patients with mild-to-moderate Alzheimer's disease--one involving 1121 carriers of the apolipoprotein E (APOE) ε4 allele and the other involving 1331 noncarriers. Bapineuzumab or placebo, with doses varying by study, was administered by intravenous infusion every 13 weeks for 78 weeks. The primary outcome measures were scores on the 11-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog11, with scores ranging from 0 to 70 and higher scores indicating greater impairment) and the Disability Assessment for Dementia (DAD, with scores ranging from 0 to 100 and higher scores indicating less impairment). A total of 1090 carriers and 1114 noncarriers were included in the efficacy analysis. Secondary outcome measures included findings on positron-emission tomographic amyloid imaging with the use of Pittsburgh compound B (PIB-PET) and cerebrospinal fluid phosphorylated tau (phospho-tau) concentrations. RESULTS: There were no significant between-group differences in the primary outcomes. At week 78, the between-group differences in the change from baseline in the ADAS-cog11 and DAD scores (bapineuzumab group minus placebo group) were -0.2 (P=0.80) and -1.2 (P=0.34), respectively, in the carrier study; the corresponding differences in the noncarrier study were -0.3 (P=0.64) and 2.8 (P=0.07) with the 0.5-mg-per-kilogram dose of bapineuzumab and 0.4 (P=0.62) and 0.9 (P=0.55) with the 1.0-mg-per-kilogram dose. The major safety finding was amyloid-related imaging abnormalities with edema among patients receiving bapineuzumab, which increased with bapineuzumab dose and APOE ε4 allele number and which led to discontinuation of the 2.0-mg-per-kilogram dose. Between-group differences were observed with respect to PIB-PET and cerebrospinal fluid phospho-tau concentrations in APOE ε4 allele carriers but not in noncarriers. CONCLUSIONS: Bapineuzumab did not improve clinical outcomes in patients with Alzheimer's disease, despite treatment differences in biomarkers observed in APOE ε4 carriers. (Funded by Janssen Alzheimer Immunotherapy and Pfizer; Bapineuzumab 301 and 302 ClinicalTrials.gov numbers, NCT00575055 and NCT00574132, and EudraCT number, 2009-012748-17.).


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Antibodies, Monoclonal, Humanized/adverse effects , Apolipoproteins E/genetics , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Brain/pathology , Cognition/drug effects , Double-Blind Method , Edema/chemically induced , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Neuropsychological Tests , Phosphorylation , Positron-Emission Tomography , Severity of Illness Index , Treatment Failure , tau Proteins/cerebrospinal fluid
5.
Mov Disord ; 32(2): 211-218, 2017 02.
Article in English | MEDLINE | ID: mdl-27886407

ABSTRACT

BACKGROUND: α-Synuclein is a major component of pathologic inclusions that characterize Parkinson's disease. PRX002 is an antibody that targets α-synuclein, and its murine parent antibody 9E4 has been shown in preclinical studies to reduce α-synuclein pathology and to protect against cognitive and motor deteriorations and progressive neurodegeneration in human α-synuclein transgenic mice. METHODS: This first-in-human, randomized, double-blind, placebo-controlled, phase 1 study assessed the impact of PRX002 administered to 40 healthy participants in 5 ascending-dose cohorts (n = 8/cohort) in which participants were randomly assigned to receive a single intravenous infusion of study drug (0.3, 1, 3, 10, or 30 mg/kg; n = 6/cohort) or placebo (n = 2/cohort). RESULTS: PRX002 demonstrated favorable safety, tolerability, and pharmacokinetic profiles at all doses tested, with no immunogenicity. No serious adverse events, discontinuations as a result of adverse events, or dose-limiting toxicities were reported. Serum PRX002 exposure was dose proportional; the average terminal half-life across all doses was 18.2 days. A significant dose-dependent reduction in free serum α-synuclein (unbound to PRX002) was apparent within 1 hour after PRX002 administration, whereas total α-synuclein (free plus bound) increased dose-dependently, presumably because of the expected change in kinetics following antibody binding. CONCLUSIONS: This study demonstrates that serum α-synuclein can be safely modulated in a dose-dependent manner after single intravenous infusions of an anti-α-synuclein antibody. These findings support continued development of PRX002, including further characterization of its safety, tolerability, pharmacokinetics, and pharmacodynamic effects in the central nervous system in patients with Parkinson's disease. © 2016 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , alpha-Synuclein/blood , alpha-Synuclein/drug effects , alpha-Synuclein/immunology , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Double-Blind Method , Female , Healthy Volunteers , Humans , Immunoglobulin G/immunology , Infusions, Intravenous , Male , Middle Aged , Young Adult
6.
Dement Geriatr Cogn Disord ; 44(3-4): 129-143, 2017.
Article in English | MEDLINE | ID: mdl-28787712

ABSTRACT

AIMS: To evaluate the impact of amyloid PET imaging on diagnosis and patient management in a multicenter, randomized, controlled study. METHODS: Physicians identified patients seeking a diagnosis for mild cognitive impairment or dementia, possibly due to Alzheimer disease (AD), and recorded a working diagnosis and a management plan. The patients underwent florbetapir PET scanning and were randomized to either immediate or delayed (1-year) feedback regarding amyloid status. At the 3-month visit, the physician updated the diagnosis and recorded a summary of the actual patient management since the post-scan visit. The study examined the impact of immediate versus delayed feedback on patient diagnosis/management at 3 and 12 months. RESULTS: A total of 618 subjects were randomized (1:1) to immediate or delayed feedback arms, and 602 subjects completed the 3-month primary endpoint visit. A higher proportion of patients in the immediate feedback arm showed a change in diagnosis compared to the controls (32.6 vs. 6.4%; p = 0.0001). Similarly, a higher proportion of patients receiving immediate feedback had a change in management plan (68 vs. 55.5%; p < 0.002), mainly driven by changes in AD medication. Specifically, acetylcholinesterase inhibitors were prescribed to 67% of the amyloid-positive and 27% of the amyloid-negative subjects in the information group compared with 56 and 43%, respectively, in the control group (p < 0.0001). These between-group differences persisted until the 12-month visit. CONCLUSION: Knowledge of the amyloid status affects the diagnosis and alters patient management.


Subject(s)
Alzheimer Disease/diagnostic imaging , Aniline Compounds , Cognitive Dysfunction/diagnostic imaging , Ethylene Glycols , Plaque, Amyloid/metabolism , Positron-Emission Tomography/methods , Aged , Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid/metabolism , Amyloidogenic Proteins/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/psychology , Feedback , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies
7.
Dement Geriatr Cogn Disord ; 41(1-2): 80-92, 2016.
Article in English | MEDLINE | ID: mdl-26745445

ABSTRACT

BACKGROUND: Published appropriate use criteria (AUC) describe patients for whom amyloid positron emission tomography (PET) might be most useful. This study compared the impact of amyloid PET on diagnosis and management in subjects likely to either meet or not meet AUC. METHODS: Physicians provided a provisional diagnosis and management plan for patients presenting with cognitive decline before and after amyloid PET imaging with florbetapir F 18. Participants were classified as AUC-like or not, based on the prescan diagnosis and demographic features. RESULTS: In all, 125 of 229 participants (55%) were classified as AUC-like. Sixty-two percent of the AUC-like subjects had a change in diagnosis after scanning compared with 45% of the non-AUC subjects (p = 0.011). Both groups demonstrated high rates of change in their management plans after scanning (88.0% for AUC-like cases, 85.6% for non-AUC cases). CONCLUSIONS: The impact of amyloid imaging on diagnosis and planned management was maintained and, if anything, amplified in AUC-like patients.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid/analysis , Cognitive Dysfunction/diagnosis , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/analysis , Aniline Compounds , Brain/diagnostic imaging , Cognition , Cognitive Dysfunction/drug therapy , Ethylene Glycols , Female , Humans , Male , Neuropsychological Tests , Positron-Emission Tomography , Radiopharmaceuticals
8.
Qual Life Res ; 23(6): 1813-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24448684

ABSTRACT

PURPOSE: To estimate the relative importance that Alzheimer's disease (AD) caregivers in the United States and Germany place on preserving patients' ability to perform activities of daily living. METHODS: US and German residents providing care for a person with AD completed an online preference survey. Each respondent completed five best-worst scaling questions. Each question related to five of 10 activities from the Disability Assessment for Dementia scale. Preference weights, indicating the relative importance of preserving the ability to perform these 10 activities for 36 months, were estimated using maximum-difference scaling. A separate model was estimated for each country. RESULTS: Four hundred and three US and 400 German caregivers completed the survey. In both countries, preserving a patients' ability to use the toilet without accidents was the most important activity and handling money was the least important activity. There were few differences between US and German caregivers in the relative importance across activities. CONCLUSIONS: Caregivers generally placed greater importance on preserving basic activities of daily living than on preserving instrumental activities of daily living. Understanding differences in the relative importance of functional items in the DAD may contribute to a better understanding of the benefits of different AD treatment and support measures.


Subject(s)
Activities of Daily Living/psychology , Alzheimer Disease/therapy , Caregivers/psychology , Quality of Life , Surveys and Questionnaires , Caregivers/statistics & numerical data , Cross-Sectional Studies , Disability Evaluation , Educational Status , Female , Germany , Humans , Interviews as Topic , Male , Middle Aged , Social Class , United States
9.
Alzheimers Dement ; 10(6): 853-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25458309

ABSTRACT

Several lines of evidence from Alzheimer's disease (AD) research continue to support the notion that the biological changes associated with AD are occurring possibly several decades before an individual will experience the cognitive and functional changes associated with the disease. The National Institute on Aging-Alzheimer's Association revised criteria for AD provided a framework for this new thinking. As a result of this growing understanding, several research efforts have launched or will be launching large secondary prevention trials in AD. These and other efforts have clearly demonstrated a need for better measures of cognitive and functional change in people with the earliest changes associated with AD. Recent draft guidance from the US Food and Drug Administration further elevated the importance of cognitive and functional assessments in early stage clinical trials by proposing that even in the pre-symptomatic stages of the disease, approval will be contingent on demonstrating clinical meaningfulness. The Alzheimer's Association's Research Roundtable addressed these issues at its fall meeting October 28-29, 2013, in Washington, D.C. The focus of the discussion included the need for improved cognitive and functional outcome measures for clinical of participants with preclinical AD and those diagnosed with Mild Cognitive Impairment due to AD.


Subject(s)
Alzheimer Disease , Clinical Trials as Topic , Cognition Disorders/diagnosis , Cognition Disorders/etiology , Neuropsychological Tests , Alzheimer Disease/complications , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Humans
10.
Alzheimers Res Ther ; 16(1): 20, 2024 01 25.
Article in English | MEDLINE | ID: mdl-38273408

ABSTRACT

BACKGROUND: Effective, disease-modifying therapeutics for the treatment of Alzheimer's disease (AD) remain a large unmet need. Extensive evidence suggests that amyloid beta (Aß) is central to AD pathophysiology, and Aß oligomers are among the most toxic forms of Aß. CT1812 is a novel brain penetrant sigma-2 receptor ligand that interferes with the binding of Aß oligomers to neurons. Preclinical studies of CT1812 have demonstrated its ability to displace Aß oligomers from neurons, restore synapses in cell cultures, and improve cognitive measures in mouse models of AD. CT1812 was found to be generally safe and well tolerated in a placebo-controlled phase 1 clinical trial in healthy volunteers and phase 1a/2 clinical trials in patients with mild to moderate dementia due to AD. The unique objective of this study was to incorporate synaptic positron emission tomography (PET) imaging as an outcome measure for CT1812 in AD patients. METHODS: The present phase 1/2 study was a randomized, double-blind, placebo-controlled, parallel-group trial conducted in 23 participants with mild to moderate dementia due to AD to primarily evaluate the safety of CT1812 and secondarily its pharmacodynamic effects. Participants received either placebo or 100 mg or 300 mg per day of oral CT1812 for 24 weeks. Pharmacodynamic effects were assessed using the exploratory efficacy endpoints synaptic vesicle glycoprotein 2A (SV2A) PET, fluorodeoxyglucose (FDG) PET, volumetric MRI, cognitive clinical measures, as well as cerebrospinal fluid (CSF) biomarkers of AD pathology and synaptic degeneration. RESULTS: No treatment differences relative to placebo were observed in the change from baseline at 24 weeks in either SV2A or FDG PET signal, the cognitive clinical rating scales, or in CSF biomarkers. Composite region volumetric MRI revealed a trend towards tissue preservation in participants treated with either dose of CT1812, and nominally significant differences with both doses of CT1812 compared to placebo were found in the pericentral, prefrontal, and hippocampal cortices. CT1812 was safe and well tolerated. CONCLUSIONS: The safety findings of this 24-week study and the observed changes on volumetric MRI with CT1812 support its further clinical development. TRIAL REGISTRATION: The clinical trial described in this manuscript is registered at clinicaltrials.gov (NCT03493282).


Subject(s)
Alzheimer Disease , Mice , Animals , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Pilot Projects , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Biomarkers/cerebrospinal fluid
11.
Alzheimer Dis Assoc Disord ; 27(1): 4-15, 2013.
Article in English | MEDLINE | ID: mdl-23203162

ABSTRACT

Florbetapir F18 has been approved by the Food and Drug Administration for in vivo assessment of amyloid pathology in patients undergoing evaluation for Alzheimer disease (AD). The aim of this study was to determine the impact of amyloid imaging on the diagnoses and management of patients undergoing evaluation for cognitive decline. Patients were recruited to participate at 19 clinical sites. The site physician provided a provisional diagnosis, an estimate of their diagnostic confidence, and their plan for diagnostic evaluation and management both before and after receiving the results from amyloid imaging with florbetapir F18. Analyses compared the frequency of AD and non-AD diagnoses, plans for ancillary testing, and intended patient management before and after florbetapir imaging. A total of 229 patients participated in the trial (113 amyloid positive, 116 amyloid negative). After receiving the results of the florbetapir scan, diagnosis changed in 125/229, or 54.6% [95% confidence intervals (CI), 48.1%-60.9%], of cases, and diagnostic confidence increased by an average of 21.6% (95% CI, 18.3%-24.8%). A total of 199/229 or 86.9% (95% CI, 81.9%-90.7%) of cases had at least 1 change in their management plan. Intended cholinesterase inhibitor or memantine treatment increased by 17.7% (95% CI, 11.8%-25.8%) of all cases with positive scans and decreased by 23.3% (95% CI, 16.5%-31.8%) of all those with negative scans. Among subjects who had not yet undergone a completed work up, planned brain structural imaging (computed tomographic/magnetic resonance imaging) decreased by 24.4% (95% CI, 17.5%-32.8%) and planned neuropsychological testing decreased by 32.8% (95% CI, 25.0%-41.6%). In summary, amyloid imaging results altered physician's diagnostic thinking, intended testing, and management of patients undergoing evaluation for cognitive decline.


Subject(s)
Alzheimer Disease/therapy , Plaque, Amyloid/diagnostic imaging , Practice Patterns, Physicians' , Aged , Alzheimer Disease/diagnostic imaging , Aniline Compounds , Ethylene Glycols , Female , Humans , Male , Neuropsychological Tests , Positron-Emission Tomography/methods , Radiopharmaceuticals
12.
Alzheimers Dement ; 9(1 Suppl): S45-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22658286

ABSTRACT

BACKGROUND: We used the database of the Alzheimer's Disease Neuroimaging Initiative (ADNI) to explore the psychometric properties of the Clinical Dementia Rating Sum of Boxes (CDR-SB) to consider its utility as an outcome measure for clinical trials in early and mild, as well as later, stages of Alzheimer's disease (AD). METHODS: We assessed internal consistency, structural validity, convergent validity, and 2-year internal and external responsiveness of the CDR-SB using data from 382 subjects with early or mild AD at entry into the ADNI study. RESULTS: The CDR-SB assesses both cognitive and functional domains of AD disability. Mean scores declined nearly linearly; CDR-SB cognitive and functional subsums contributed equally to total scores at both very mild (early) and mild stages of the disease. CONCLUSIONS: The CDR-SB has psychometric properties that make it attractive as a primary outcome measure that comprehensively assesses both cognitive and functional disability in AD patients. It may prove particularly useful for studies in early, predementia stages of AD.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/psychology , Neuropsychological Tests , Aged , Aged, 80 and over , Clinical Trials as Topic , Female , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Psychometrics , Research Design
13.
Alzheimers Dement ; 9(1 Suppl): S21-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23127469

ABSTRACT

BACKGROUND: The Alzheimer's Disease Assessment Scale-Cognitive (ADAS-Cog) has been used widely as a cognitive end point in Alzheimer's Disease (AD) clinical trials. Efforts to treat AD pathology at earlier stages have also used ADAS-Cog, but failure in these trials can be difficult to interpret because the scale has well-known ceiling effects that limit its use in mild cognitive impairment (MCI) and early AD. A wealth of data exists in ADAS-Cog from both historical trials and contemporary longitudinal natural history studies that can provide insights about parts of the scale that may be better suited for MCI and early AD trials. METHODS: Using Alzheimer's Disease Neuroimaging Initiative study data, we identified the most informative cognitive measures from the ADAS-Cog and other available scales. We used cross-sectional analyses to characterize trajectories of ADAS-Cog and its individual subscales, as well as other cognitive, functional, or global measures across disease stages. Informative measures were identified based on standardized mean of 2-year change from baseline and were combined into novel composite endpoints. We assessed performance of the novel endpoints based on sample size requirements for a 2-year clinical trial. A bootstrap validation procedure was also undertaken to assess the reproducibility of the standardized mean changes of the selected measures and the corresponding composites. RESULTS: All proposed novel endpoints have improved standardized mean changes and thus improved statistical power compared with the ADAS-Cog 11. Further improvements were achieved by using cognitive-functional composites. Combining the novel composites with an enrichment strategy based on cerebral spinal fluid beta-amyloid (Aß(1-42)) in a 2-year trial yielded gains in power of 20% to 40% over ADAS-Cog 11, regardless of the novel measure considered. CONCLUSION: An empirical, data-driven approach with existing instruments was used to derive novel composite scales based on ADAS-Cog 11 with improved performance characteristics for MCI and early AD clinical trials. Together with patient enrichment based on Aß(1-42) pathology, these modified endpoints may allow more efficient clinical trials in these populations and can be assessed without modifying current test administration procedures in ongoing trials.


Subject(s)
Alzheimer Disease/diagnosis , Cognitive Dysfunction/diagnosis , Early Diagnosis , Aged , Alzheimer Disease/psychology , Clinical Trials as Topic , Female , Humans , Male , Neuropsychological Tests
14.
Alzheimers Dement ; 9(1): 39-49, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23164548

ABSTRACT

Over the past 30 years, many drugs have been studied as possible treatments for Alzheimer's disease, but only four have demonstrated sufficient efficacy to be approved as treatments, of which three are in the same class. This lack of success has raised questions both in the pharmaceutical industry and academia about the future of Alzheimer's disease therapy. The high cost and low success rate of drug development across many disease areas can be attributed, in large part, to late-stage clinical failures (Schachter and Ramoni, Nat Rev Drug Discov 2007;6:107-8). Thus, identifying in phase II, or preferably phase I, drugs that are likely to fail would have a dramatic impact on the costs associated with developing new drugs. With this in mind, the Alzheimer's Association convened a Research Roundtable on June 23 and 24, 2011, in Washington, DC, bringing together scientists from academia, industry, and government regulatory agencies to discuss strategies for improving the probability of phase II trial results predicting success when considering the go/no-go decision-making process leading to the initiation of phase III.


Subject(s)
Alzheimer Disease/drug therapy , Clinical Trials, Phase II as Topic , Neuroprotective Agents/economics , Neuroprotective Agents/pharmacology , Cost-Benefit Analysis , Humans
15.
Alzheimers Dement ; 9(4): 438-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23809364

ABSTRACT

An international task force of investigators from academia, industry, nonprofit foundations, and regulatory agencies met in Monte Carlo, Monaco, on October 31, 2012, to review lessons learned from the recent bapineuzumab and solanezumab trials, and to incorporate insights gained from these trials into future clinical studies. Although there is broad consensus that Alzheimer's disease (AD) should be treated during its earliest stages, the concept of secondary prevention has evolved to be described more accurately as treatment of preclinical, presymptomatic, or early AD. There continues to be a strong emphasis on biomarkers and a need for new biomarkers; however, there has also been a realization, based on completed trials, that the most reliable indicator of clinical efficacy across the entire spectrum of disease from asymptomatic to AD dementia is likely a measure of cognition. The task force made many recommendations that should improve the likelihood of success in future trials, including larger phase 2 or combined phase 2/phase 3 studies, clear evidence of target engagement in the central nervous system, evidence of downstream effects on biomarkers before initiating phase 3 studies, consideration of adaptive and targeted trial designs, and use of sensitive measures of cognition as the most robust indicator of treatment benefit.


Subject(s)
Alzheimer Disease/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Clinical Trials, Phase III as Topic/methods , Multicenter Studies as Topic/methods , Nootropic Agents/therapeutic use , Randomized Controlled Trials as Topic/methods , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/analysis , Apolipoproteins E/analysis , Apolipoproteins E/genetics , Atrophy/etiology , Biomarkers , Brain/pathology , Brain Chemistry , Brain Edema/etiology , Clinical Trials, Phase II as Topic/statistics & numerical data , Cognitive Dysfunction/drug therapy , Disease Progression , Endpoint Determination/methods , Humans , Neuroimaging , Patient Selection , Treatment Failure
16.
Transl Neurodegener ; 12(1): 54, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37968718

ABSTRACT

TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT05821153, Registered April 20 2023, Retrospectively registered, https://classic. CLINICALTRIALS: gov/ct2/show/NCT05821153.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Pilot Projects , Treatment Outcome , Immunotherapy
17.
Alzheimers Dement ; 7(4): 367-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21784348

ABSTRACT

Amyloid imaging related abnormalities (ARIA) have now been reported in clinical trials with multiple therapeutic avenues to lower amyloid-ß burden in Alzheimer's disease (AD). In response to concerns raised by the Food and Drug Administration, the Alzheimer's Association Research Roundtable convened a working group to review the publicly available trial data, attempts at developing animal models, and the literature on the natural history and pathology of related conditions. The spectrum of ARIA includes signal hyperintensities on fluid attenuation inversion recoverysequences thought to represent "vasogenic edema" and/or sulcal effusion (ARIA-E), as well as signal hypointensities on GRE/T2* thought to represent hemosiderin deposits (ARIA-H), including microhemorrhage and superficial siderosis. The etiology of ARIA remains unclear but the prevailing data support vascular amyloid as a common pathophysiological mechanism leading to increased vascular permeability. The workgroup proposes recommendations for the detection and monitoring of ARIA in ongoing AD clinical trials, as well as directions for future research.


Subject(s)
Alzheimer Disease , Amyloid/metabolism , Clinical Trials as Topic/methods , Magnetic Resonance Imaging , Societies, Medical/organization & administration , Alzheimer Disease/complications , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid/immunology , Animals , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/pathology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Clinical Trials as Topic/standards , Disease Models, Animal , Humans
18.
Nat Med ; 27(8): 1451-1457, 2021 08.
Article in English | MEDLINE | ID: mdl-34385707

ABSTRACT

A randomized, double-blind, placebo-controlled, 52-week study (no. NCT03068468) evaluated gosuranemab, an anti-tau monoclonal antibody, in the treatment of progressive supranuclear palsy (PSP). In total, 486 participants dosed were assigned to either gosuranemab (n = 321) or placebo (n = 165). Efficacy was not demonstrated on adjusted mean change of PSP Rating Scale score at week 52 between gosuranemab and placebo (10.4 versus 10.6, P = 0.85, primary endpoint), or at secondary endpoints, resulting in discontinuation of the open-label, long-term extension. Unbound N-terminal tau in cerebrospinal fluid decreased by 98% with gosuranemab and increased by 11% with placebo (P < 0.0001). Incidences of adverse events and deaths were similar between groups. This well-powered study suggests that N-terminal tau neutralization does not translate into clinical efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Supranuclear Palsy, Progressive/drug therapy , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Double-Blind Method , Female , Humans , Male , Pneumonia/etiology , Treatment Outcome , tau Proteins/immunology
19.
Neurology ; 96(20): 944-954, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33674360

ABSTRACT

Drug development for Alzheimer disease and other neurodegenerative dementias, including frontotemporal dementia, has experienced a long history of phase 2 and phase 3 clinical trials that failed to show efficacy of investigational drugs. Despite differences in clinical and behavioral characteristics, these disorders have shared pathologies and face common challenges in designing early-phase trials that are predictive of late-stage success. Here, we discuss exploratory clinical trials in neurodegenerative dementias. These are generally phase 1b or phase 2a trials that are designed to assess pharmacologic effects and rely on biomarker outcomes, with shorter treatment durations and fewer patients than traditional phase 2 studies. Exploratory trials can establish go/no-go decision points, support proof of concept and dose selection, and terminate drugs that fail to show target engagement with suitable exposure and acceptable safety profiles. Early failure saves valuable resources including opportunity costs. This is especially important for programs in academia and small biotechnology companies but may be applied to high-risk projects in large pharmaceutical companies to achieve proof of concept more rapidly at lower costs than traditional approaches. Exploratory studies in a staged clinical development program may provide promising data to warrant the substantial resources needed to advance compounds through late-stage development. To optimize the design and application of exploratory trials, the Alzheimer's Drug Discovery Foundation and the Association for Frontotemporal Degeneration convened an advisory panel to provide recommendations on outcome measures and statistical considerations for these types of studies and study designs that can improve efficiency in clinical development.


Subject(s)
Alzheimer Disease/drug therapy , Clinical Trials as Topic/methods , Drug Development/methods , Frontotemporal Dementia/drug therapy , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Dementia/drug therapy , Humans , Neurodegenerative Diseases/drug therapy , Outcome Assessment, Health Care , Proof of Concept Study , Research Design , Treatment Failure , Treatment Outcome
20.
Alzheimer Dis Assoc Disord ; 24(2): 198-203, 2010.
Article in English | MEDLINE | ID: mdl-20505438

ABSTRACT

The safety, tolerability, and pharmacokinetics (PKs) of bapineuzumab (AAB-001), a humanized monoclonal antibody to amyloid beta, were evaluated in patients with mild-to-moderate Alzheimer disease in a phase 1, randomized, third-party unblinded, placebo-controlled, single ascending dose trial. Thirty patients received bapineuzumab infusion of 0.5, 1.5, or 5 mg/kg or placebo (6 active, 2 placebo for 0.5 and 1.5-mg/kg cohorts; 10 active, 4 placebo for 5.0-mg/kg cohort). Three patients in the highest dose cohort (5.0 mg/kg) developed magnetic resonance imaging abnormalities consistent with vasogenic edema, predominantly high signal abnormalities on fluid-attenuated inversion recovery sequences, all of which resolved over time. Plasma amyloid beta was elevated from baseline, peaking approximately 24 hours after infusion. PK analysis demonstrated a half-life of 21 to 26 days, supporting a 13-week dosing interval for bapineuzumab. This small, single-dose study demonstrated the safety profile and PK characteristics of bapineuzumab and was used to design later safety and efficacy trials.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/administration & dosage , Nootropic Agents/administration & dosage , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Dose-Response Relationship, Drug , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Nootropic Agents/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL