Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Eric Siemers is active.

Publication


Featured researches published by Eric Siemers.


Alzheimers & Dementia | 2011

Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease

Reisa A. Sperling; Paul S. Aisen; Laurel Beckett; David A. Bennett; Suzanne Craft; Anne M. Fagan; Takeshi Iwatsubo; Clifford R. Jack; Jeffrey Kaye; Thomas J. Montine; Denise C. Park; Eric M. Reiman; Christopher C. Rowe; Eric Siemers; Yaakov Stern; Kristine Yaffe; Maria C. Carrillo; Bill Thies; Marcelle Morrison-Bogorad; Molly V. Wagster; Creighton H. Phelps

The pathophysiological process of Alzheimers disease (AD) is thought to begin many years before the diagnosis of AD dementia. This long “preclinical” phase of AD would provide a critical opportunity for therapeutic intervention; however, we need to further elucidate the link between the pathological cascade of AD and the emergence of clinical symptoms. The National Institute on Aging and the Alzheimers Association convened an international workgroup to review the biomarker, epidemiological, and neuropsychological evidence, and to develop recommendations to determine the factors which best predict the risk of progression from “normal” cognition to mild cognitive impairment and AD dementia. We propose a conceptual framework and operational research criteria, based on the prevailing scientific evidence to date, to test and refine these models with longitudinal clinical research studies. These recommendations are solely intended for research purposes and do not have any clinical implications at this time. It is hoped that these recommendations will provide a common rubric to advance the study of preclinical AD, and ultimately, aid the field in moving toward earlier intervention at a stage of AD when some disease‐modifying therapies may be most efficacious.


Annals of Neurology | 2009

Cerebrospinal Fluid Biomarker Signature in Alzheimer’s Disease Neuroimaging Initiative Subjects

Leslie M. Shaw; Hugo Vanderstichele; Malgorzata Knapik-Czajka; Christopher M. Clark; Paul S. Aisen; Ronald C. Petersen; Kaj Blennow; Holly Soares; Adam J. Simon; Piotr Lewczuk; Robert A. Dean; Eric Siemers; William Z. Potter; Virginia M.-Y. Lee; John Q. Trojanowski

Develop a cerebrospinal fluid biomarker signature for mild Alzheimers disease (AD) in Alzheimers Disease Neuroimaging Initiative (ADNI) subjects.


The New England Journal of Medicine | 2014

Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer's Disease

Rachelle S. Doody; Ronald G. Thomas; Martin R. Farlow; Takeshi Iwatsubo; Bruno Vellas; Steven Joffe; Karl Kieburtz; Rema Raman; Xiaoying Sun; Paul S. Aisen; Eric Siemers; Hong Liu-Seifert; Richard C. Mohs

BACKGROUND Alzheimers disease is characterized by amyloid-beta plaques, neurofibrillary tangles, gliosis, and neuronal loss. Solanezumab, a humanized monoclonal antibody, preferentially binds soluble forms of amyloid and in preclinical studies promoted its clearance from the brain. METHODS In two phase 3, double-blind trials (EXPEDITION 1 and EXPEDITION 2), we randomly assigned 1012 and 1040 patients, respectively, with mild-to-moderate Alzheimers disease to receive placebo or solanezumab (administered intravenously at a dose of 400 mg) every 4 weeks for 18 months. The primary outcomes were the changes from baseline to week 80 in scores on the 11-item cognitive subscale of the Alzheimers Disease Assessment Scale (ADAS-cog11; range, 0 to 70, with higher scores indicating greater cognitive impairment) and the Alzheimers Disease Cooperative Study-Activities of Daily Living scale (ADCS-ADL; range, 0 to 78, with lower scores indicating worse functioning). After analysis of data from EXPEDITION 1, the primary outcome for EXPEDITION 2 was revised to the change in scores on the 14-item cognitive subscale of the Alzheimers Disease Assessment Scale (ADAS-cog14; range, 0 to 90, with higher scores indicating greater impairment), in patients with mild Alzheimers disease. RESULTS Neither study showed significant improvement in the primary outcomes. The modeled difference between groups (solanezumab group minus placebo group) in the change from baseline was -0.8 points for the ADAS-cog11 score (95% confidence interval [CI], -2.1 to 0.5; P=0.24) and -0.4 points for the ADCS-ADL score (95% CI, -2.3 to 1.4; P=0.64) in EXPEDITION 1 and -1.3 points (95% CI, -2.5 to 0.3; P=0.06) and 1.6 points (95% CI, -0.2 to 3.3; P=0.08), respectively, in EXPEDITION 2. Between-group differences in the changes in the ADAS-cog14 score were -1.7 points in patients with mild Alzheimers disease (95% CI, -3.5 to 0.1; P=0.06) and -1.5 in patients with moderate Alzheimers disease (95% CI, -4.1 to 1.1; P=0.26). In the combined safety data set, the incidence of amyloid-related imaging abnormalities with edema or hemorrhage was 0.9% with solanezumab and 0.4% with placebo for edema (P=0.27) and 4.9% and 5.6%, respectively, for hemorrhage (P=0.49). CONCLUSIONS Solanezumab, a humanized monoclonal antibody that binds amyloid, failed to improve cognition or functional ability. (Funded by Eli Lilly; EXPEDITION 1 and 2 ClinicalTrials.gov numbers, NCT00905372 and NCT00904683.).


Neurology | 2003

Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD

John G. Nutt; Kim J. Burchiel; Cynthia L. Comella; Joseph Jankovic; Anthony E. Lang; Edward R. Laws; Andres M. Lozano; Richard D. Penn; Richard K. Simpson; Mark Stacy; G. F. Wooten; J. Lopez; M. Harrigan; F. F. Marciano; Julie H. Carter; Stone C; Joel M. Trugman; Elke Rost-Ruffner; Christopher O'Brien; J. H. McVicker; Thomas L. Davis; David Charles; G. Allen; William J. Weiner; H. J. Landy; J. Bronstein; William C. Koller; Rajesh Pahwa; Steve Wilkinson; Eric Siemers

Objective: To assess the safety, tolerability, and biological activity of glial cell line-derived neurotrophic factor (GDNF) administered by an implanted intracerebroventricular (ICV) catheter and access port in advanced PD. Background: GDNF is a peptide that promotes survival of dopamine neurons. It improved 6-OHDA- or MPTP-induced behavioral deficits in rodents and monkeys. Methods: A multicenter, randomized, double-blind, placebo-controlled, sequential cohort study compared the effects of monthly ICV administration of placebo and 25, 75, 150, 300, and 500 to 4,000 μg of GDNF in 50 subjects with PD for 8 months. An open-label study extended exposure up to an additional 20 months and maximum single doses of up to 4,000 μg in 16 subjects. Laboratory testing, adverse events (AE), and Unified Parkinson’s Disease Rating Scale (UPDRS) scoring were obtained at 1- to 4-week intervals throughout the studies. Results: Twelve subjects received placebo and seven or eight subjects were assigned to each of the other GDNF dose groups. “On” and “off” total and motor UPDRS scores were not improved by GDNF at any dose. Nausea, anorexia, and vomiting were common hours to several days after injections of GDNF. Weight loss occurred in the majority of subjects receiving 75 μg or larger doses of GDNF. Paresthesias, often described as electric shocks (Lhermitte sign), were common in GDNF-treated subjects, were not dose related, and resolved on discontinuation of GDNF. Asymptomatic hyponatremia occurred in over half of subjects receiving 75 μg or larger doses of GDNF; it was symptomatic in several subjects. The open-label extension study had similar AE and lack of therapeutic efficacy. Conclusions: GDNF administered by ICV injection is biologically active as evidenced by the spectrum of AE encountered in this study. GDNF did not improve parkinsonism, possibly because GDNF did not reach the target tissues—putamen and substantia nigra.


The New England Journal of Medicine | 2013

A Phase 3 Trial of Semagacestat for Treatment of Alzheimer's Disease

Rachelle S. Doody; Rema Raman; Martin R. Farlow; Takeshi Iwatsubo; Bruno Vellas; Steven Joffe; Karl Kieburtz; Feng He; Xiaoying Sun; Ronald G. Thomas; Paul S. Aisen; Eric Siemers; Gopalan Sethuraman; Richard C. Mohs

BACKGROUND Alzheimers disease is characterized by the presence of cortical amyloid-beta (Aβ) protein plaques, which result from the sequential action of β-secretase and γ-secretase on amyloid precursor protein. Semagacestat is a small-molecule γ-secretase inhibitor that was developed as a potential treatment for Alzheimers disease. METHODS We conducted a double-blind, placebo-controlled trial in which 1537 patients with probable Alzheimers disease underwent randomization to receive 100 mg of semagacestat, 140 mg of semagacestat, or placebo daily. Changes in cognition from baseline to week 76 were assessed with the use of the cognitive subscale of the Alzheimers Disease Assessment Scale for cognition (ADAS-cog), on which scores range from 0 to 70 and higher scores indicate greater cognitive impairment, and changes in functioning were assessed with the Alzheimers Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) scale, on which scores range from 0 to 78 and higher scores indicate better functioning. A mixed-model repeated-measures analysis was used. RESULTS The trial was terminated before completion on the basis of a recommendation by the data and safety monitoring board. At termination, there were 189 patients in the group receiving placebo, 153 patients in the group receiving 100 mg of semagacestat, and 121 patients in the group receiving 140 mg of semagacestat. The ADAS-cog scores worsened in all three groups (mean change, 6.4 points in the placebo group, 7.5 points in the group receiving 100 mg of the study drug, and 7.8 points in the group receiving 140 mg; P=0.15 and P=0.07, respectively, for the comparison with placebo). The ADCS-ADL scores also worsened in all groups (mean change at week 76, -9.0 points in the placebo group, -10.5 points in the 100-mg group, and -12.6 points in the 140-mg group; P=0.14 and P<0.001, respectively, for the comparison with placebo). Patients treated with semagacestat lost more weight and had more skin cancers and infections, treatment discontinuations due to adverse events, and serious adverse events (P<0.001 for all comparisons with placebo). Laboratory abnormalities included reduced levels of lymphocytes, T cells, immunoglobulins, albumin, total protein, and uric acid and elevated levels of eosinophils, monocytes, and cholesterol; the urine pH was also elevated. CONCLUSIONS As compared with placebo, semagacestat did not improve cognitive status, and patients receiving the higher dose had significant worsening of functional ability. Semagacestat was associated with more adverse events, including skin cancers and infections. (Funded by Eli Lilly; ClinicalTrials.gov number, NCT00594568.)


Neurology | 2006

Effects of a γ-secretase inhibitor in a randomized study of patients with Alzheimer disease

Eric Siemers; Joseph F. Quinn; J. Kaye; Martin R. Farlow; Anton P. Porsteinsson; Pierre N. Tariot; P. Zoulnouni; James E. Galvin; David M. Holtzman; D. S. Knopman; J. Satterwhite; C. Gonzales; Robert A. Dean; P. C. May

LY450139 dihydrate, a γ-secretase inhibitor, was studied in a randomized, controlled trial of 70 patients with Alzheimer disease. Subjects were given 30 mg for 1 week followed by 40 mg for 5 weeks. Treatment was well tolerated. Aβ1-40 in plasma decreased by 38.2%; in CSF, Aβ1-40 decreased by 4.42 ± 9.55% (p = not significant). Higher drug doses may result in additional decreases in plasma Aβ concentrations and a measurable decrease in CSF Aβ.


Alzheimers & Dementia | 2010

The Alzheimer’s Disease Neuroimaging Initiative: Progress report and future plans

Michael W. Weiner; Paul S. Aisen; Clifford R. Jack; William J. Jagust; John Q. Trojanowski; Leslie M. Shaw; Andrew J. Saykin; John C. Morris; Nigel J. Cairns; Laurel Beckett; Arthur W. Toga; Robert C. Green; Sarah Walter; Holly Soares; Peter J. Snyder; Eric Siemers; William Z. Potter; Patricia E. Cole; Mark E. Schmidt

The Alzheimers Disease Neuroimaging Initiative (ADNI) beginning in October 2004, is a 6‐year research project that studies changes of cognition, function, brain structure and function, and biomarkers in elderly controls, subjects with mild cognitive impairment, and subjects with Alzheimers disease (AD). A major goal is to determine and validate MRI, PET images, and cerebrospinal fluid (CSF)/blood biomarkers as predictors and outcomes for use in clinical trials of AD treatments. Structural MRI, FDG PET, C‐11 Pittsburgh compound B (PIB) PET, CSF measurements of amyloid β (Aβ) and species of tau, with clinical/cognitive measurements were performed on elderly controls, subjects with mild cognitive impairment, and subjects with AD. Structural MRI shows high rates of brain atrophy, and has high statistical power for determining treatment effects. FDG PET, C‐11 Pittsburgh compound B PET, and CSF measurements of Aβ and tau were significant predictors of cognitive decline and brain atrophy. All data are available at UCLA/LONI/ADNI, without embargo. ADNI‐like projects started in Australia, Europe, Japan, and Korea. ADNI provides significant new information concerning the progression of AD.


JAMA Neurology | 2008

Phase 2 Safety Trial Targeting Amyloid β Production With a γ-Secretase Inhibitor in Alzheimer Disease

Adam S. Fleisher; Rema Raman; Eric Siemers; Lida M. Becerra; Christopher M. Clark; Robert A. Dean; Martin R. Farlow; James E. Galvin; Elaine R. Peskind; Joseph F. Quinn; Abdullah Sherzai; B. Brooke Sowell; Paul S. Aisen; Leon J. Thal

OBJECTIVE To evaluate the safety, tolerability, and amyloid beta (Abeta) response to the gamma-secretase inhibitor LY450139 in Alzheimer disease. DESIGN Multicenter, randomized, double-blind, dose-escalation, placebo-controlled trial. SETTING Community-based clinical research centers. Patients Fifty-one individuals with mild to moderate Alzheimer disease were randomized to receive placebo (n=15) or LY450139 (100 mg [n=22] or 140 mg [n=14]), with 43 completing the treatment phase. Intervention The LY450139 groups received 60 mg/d for 2 weeks, then 100 mg/d for 6 weeks, and then either 100 or 140 mg/d for 6 additional weeks. MAIN OUTCOME MEASURES Primary outcome measures were adverse events, plasma and cerebrospinal fluid Abeta levels, vital signs, electrocardiographic data, and laboratory safety test results. Secondary outcome measures included the Alzheimers Disease Assessment Scale cognitive subscale and the Alzheimers Disease Cooperative Study Activities of Daily Living Scale. RESULTS Group differences were seen in skin and subcutaneous tissue concerns (P=.05), including 3 possible drug rashes and 3 reports of hair color change in the treatment groups. There were 3 adverse event-related discontinuations, including 1 transient bowel obstruction. The plasma Abeta(40) concentration was reduced by 58.2% for the 100-mg group and 64.6% for the 140-mg group (P<.001). No significant reduction was seen in cerebrospinal fluid Abeta levels. No group differences were seen in cognitive or functional measures. CONCLUSIONS LY450139 was generally well tolerated at doses of up to 140 mg/d for 14 weeks, with several findings indicating the need for close clinical monitoring in future studies. Decreases in plasma Abeta concentrations were consistent with inhibition of gamma-secretase. Trial Registration clinicaltrials.gov Identifier: NCT00244322.


Neurology | 2000

Rate of functional decline in Huntington’s disease

Karen Marder; Hongwei Zhao; Richard H. Myers; Merit Cudkowicz; Elise Kayson; Karl Kieburtz; Constance Orme; Jane S. Paulsen; John B. Penney; Eric Siemers; Ira Shoulson

Objective: To determine the rate of functional decline in a large cohort of patients with Huntington’s disease (HD) followed at 43 sites by the Huntington Study Group (HSG). Methods: The annual rate of functional decline was measured using the Total Functional Capacity Scale (TFC) and the Independence Scale (IS) in 960 patients with definite HD followed prospectively for a mean of 18.3 months. All patients were rated with the Unified Huntington’s Disease Rating Scale (UHDRS). Sample size calculations for hypothetical clinical trials were calculated. Results: A factor analysis of the UHDRS at baseline yielded 15 factors accounting for 77% of the variance. The TFC score declined at a rate of 0.72 units/year (standard error [SE] 0.04) and the IS score declined at a rate of 4.52 units/year (SE 0.23). Lower TFC score at baseline, indicating more severe impairment, was associated with less rapid annual decline in TFC score, perhaps reflecting the floor effect of the scale. The annual rate of decline for 575 patients with baseline TFC scores of 7 to 13 was 0.97 (SE 0.06), was 0.38 (SE 0.08) for 270 patients with baseline TFC scores of 3 to 6, and was 0.06 (SE 0.1) for 101 patients with TFC scores of 0 to 2. In multivariate analysis (n = 960), longer disease duration and better cognitive status at baseline were associated with a less rapid rate of decline in TFC score, whereas depressive symptomatology was the only factor associated with more rapid decline on the IS score. Age at onset of HD, sex, weight, and education did not affect decline on either score. Conclusions: The comparable rates of decline on the TFC and the IS scores with other published studies suggest that these estimates of functional decline are representative of HD patients who are evaluated at HSG research sites. In longitudinal analysis, longer disease duration and better neuropsychological performance at baseline were associated with a less rapid rate of decline in TFC score, whereas depressive symptomatology at baseline was associated with a more rapid decline in the IS score. These rates of functional decline and the covariates that modify them should be considered in estimating statistical power and designing future therapeutic trials involving HD patients with early or moderately severe disease.


Alzheimers & Dementia | 2011

Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: Recommendations from the Alzheimer’s Association Research Roundtable Workgroup

Reisa A. Sperling; Clifford R. Jack; Sandra E. Black; Matthew P. Frosch; Steven M. Greenberg; Bradley T. Hyman; Philip Scheltens; Maria C. Carrillo; William Thies; Martin M. Bednar; Ronald S. Black; H. Robert Brashear; Michael Grundman; Eric Siemers; Howard Feldman; Rachel Schindler

Amyloid imaging related abnormalities (ARIA) have now been reported in clinical trials with multiple therapeutic avenues to lower amyloid‐β burden in Alzheimers disease (AD). In response to concerns raised by the Food and Drug Administration, the Alzheimers 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.

Collaboration


Dive into the Eric Siemers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul S. Aisen

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronald B. DeMattos

State University of New York System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge