Network


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

Hotspot


Dive into the research topics where Robert A. Dean is active.

Publication


Featured researches published by Robert A. Dean.


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.


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.


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.


The Journal of Neuroscience | 2011

Robust Central Reduction of Amyloid-β in Humans with an Orally Available, Non-Peptidic β-Secretase Inhibitor

Patrick May; Robert A. Dean; Stephen L. Lowe; Ferenc Martenyi; S.M Sheehan; L.N Boggs; Scott A. Monk; B.M Mathes; D.J Mergott; B.M Watson; S.L Stout; D.E Timm; E Smith Labell; Celedon Gonzales; Masako Nakano; Stanford Jhee; Mark Yen; Larry Ereshefsky; T.D Lindstrom; D.O Calligaro; P.J Cocke; D Greg Hall; Stuart Friedrich; Martin Citron; James E. Audia

According to the amyloid cascade hypothesis, cerebral deposition of amyloid-β peptide (Aβ) is critical for Alzheimers disease (AD) pathogenesis. Aβ generation is initiated when β-secretase (BACE1) cleaves the amyloid precursor protein. For more than a decade, BACE1 has been a prime target for designing drugs to prevent or treat AD. However, development of such agents has turned out to be extremely challenging, with major hurdles in cell penetration, oral bioavailability/metabolic clearance, and brain access. Using a fragment-based chemistry strategy, we have generated LY2811376 [(S)-4-(2,4-difluoro-5-pyrimidin-5-yl-phenyl)-4-methyl-5,6-dihydro-4H-[1,3]thiazin-2-ylamine], the first orally available non-peptidic BACE1 inhibitor that produces profound Aβ-lowering effects in animals. The biomarker changes obtained in preclinical animal models translate into man at doses of LY2811376 that were safe and well tolerated in healthy volunteers. Prominent and long-lasting Aβ reductions in lumbar CSF were measured after oral dosing of 30 or 90 mg of LY2811376. This represents the first translation of BACE1-driven biomarker changes in CNS from preclinical animal models to man. Because of toxicology findings identified in longer-term preclinical studies, this compound is no longer progressing in clinical development. However, BACE1 remains a viable target because the adverse effects reported here were recapitulated in LY2811376-treated BACE1 KO mice and thus are unrelated to BACE1 inhibition. The magnitude and duration of central Aβ reduction obtainable with BACE1 inhibition positions this protease as a tractable small-molecule target through which to test the amyloid hypothesis in man.


Alzheimers & Dementia | 2010

Update on the biomarker core of the Alzheimer's Disease Neuroimaging Initiative subjects

John Q. Trojanowski; Hugo Vandeerstichele; Magdalena Korecka; 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; Michael W. Weiner; Clifford R. Jack; William J. Jagust; Arthur W. Toga; Virginia M.-Y. Lee; Leslie M. Shaw

Here, we review progress by the Penn Biomarker Core in the Alzheimers Disease Neuroimaging Initiative (ADNI) toward developing a pathological cerebrospinal fluid (CSF) and plasma biomarker signature for mild Alzheimers disease (AD) as well as a biomarker profile that predicts conversion of mild cognitive impairment (MCI) and/or normal control subjects to AD. The Penn Biomarker Core also collaborated with other ADNI Cores to integrate data across ADNI to temporally order changes in clinical measures, imaging data, and chemical biomarkers that serve as mileposts and predictors of the conversion of normal control to MCI as well as MCI to AD, and the progression of AD. Initial CSF studies by the ADNI Biomarker Core revealed a pathological CSF biomarker signature of AD defined by the combination of Aβ1‐42 and total tau (T‐tau) that effectively delineates mild AD in the large multisite prospective clinical investigation conducted in ADNI. This signature appears to predict conversion from MCI to AD. Data fusion efforts across ADNI Cores generated a model for the temporal ordering of AD biomarkers which suggests that Aβ amyloid biomarkers become abnormal first, followed by changes in neurodegenerative biomarkers (CSF tau, F‐18 fluorodeoxyglucose‐positron emission tomography, magnetic resonance imaging) with the onset of clinical symptoms. The timing of these changes varies in individual patients due to genetic and environmental factors that increase or decrease an individuals resilience in response to progressive accumulations of AD pathologies. Further studies in ADNI will refine this model and render the biomarkers studied in ADNI more applicable to routine diagnosis and to clinical trials of disease modifying therapies.


Clinical Neuropharmacology | 2005

Safety, tolerability, and changes in amyloid β concentrations after administration of a γ-secretase inhibitor in volunteers

Eric Siemers; Michael Skinner; Robert A. Dean; Celedon Gonzales; Julie Satterwhite; Martin R. Farlow; Daniel K. Ness; Patrick C. May

Amyloid β (Aβ) may play a central role in the pathogenesis of Alzheimer disease. A functional γ-secretase inhibitor, LY450139, was developed that inhibits Aβ formation in whole cell assays, transgenic mice, and beagle dogs. The authors wished to determine the safety and tolerability of this drug, and the reduction of Aβ in plasma and cerebrospinal fluid (CSF) after multiple doses. Volunteer subjects (N = 37) were studied using doses from 5 to 50 mg/day given for 14 days. Plasma and CSF concentrations of LY450139, Aβ1-40 and Aβ1-X (“Aβtotal”) were determined, and safety and tolerability were assessed. The plasma half-life of LY450139 was approximately 2.5 hours. Pharmacokinetic analyses showed a linear relationship between dose and plasma concentrations, with a Cmax of 828 ± 19.2 ng/mL after a 50-mg dose. Plasma Aβ concentrations decreased in a dose-dependent manner over a 6-hour interval following drug administration, with a maximum decrease of approximately 40% relative to baseline. After returning to baseline, Aβ concentrations were transiently increased. CSF Aβ concentrations were unchanged. Adverse events reported by subjects taking 5-mg, 20-mg, or 40-mg doses were similar to those reported by subjects taking placebo. Two of 7 subjects taking 50 mg/day experienced adverse events that may have been drug related. In this phase 1 volunteer study, reported adverse events after taking LY450139 were manageable. A dose-dependent reduction in plasma Aβ was demonstrated, and changes in plasma Aβ concentrations were temporally related to the pharmacokinetic characteristics of LY450139.


The FASEB Journal | 1991

Human liver cocaine esterases: ethanol-mediated formation of ethylcocaine.

Robert A. Dean; C D Christian; R H Sample; William F. Bosron

A new, pharmacologically active metabolite of cocaine, ethylcocaine, has been reported in individuals after concurrent use of cocaine and ethanol. Formation of ethylcocaine may contribute to the common coabuse of these two drugs and the apparent danger of this practice. We have identified a nonspecific carboxylesterase that catalyzes the ethyl transesterification of cocaine to ethylcocaine in the presence of ethanol. In the absence of ethanol, this human liver esterase catalyzes the hydrolysis of cocaine to benzoylecgonine, a metabolite that is inactive as a psychomotor stimulant. A second human liver esterase is also described. This enzyme catalyzes hydrolysis of cocaine to ecgonine methyl ester, also inactive as a stimulant. These two liver esterases may play important roles in regulating the metabolic inactivation of cocaine.—Dean, R. A.; Christian, C. D.; Sample, R. H. B.; Bosron, W. F. Human liver cocaine enterases: ethanol‐mediated formation of ethylcocaine. FASEB J. 5: 2735‐2739; 1991.


Alzheimers & Dementia | 2012

Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease

Martin R. Farlow; Steven E. Arnold; Christopher H. van Dyck; Paul S. Aisen; B. Joy Snider; Anton P. Porsteinsson; Stuart Friedrich; Robert A. Dean; Celedon Gonzales; Gopalan Sethuraman; Ronald B. DeMattos; Richard C. Mohs; Steven M. Paul; Eric Siemers

To assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of 12 weekly infusions of solanezumab, an anti‐β‐amyloid (Aβ) antibody, in patients with mild‐to‐moderate Alzheimers disease. Cognitive measures were also obtained.


Clinical Neuropharmacology | 2007

Safety, Tolerability, and Effects on Plasma and Cerebrospinal Fluid Amyloid-β After Inhibition of γ-Secretase

Eric Siemers; Robert A. Dean; Stuart Friedrich; Lisa Ferguson-Sells; Celedon Gonzales; Martin R. Farlow; Patrick C. May

Objectives: &ggr;-Secretase inhibitors may be useful as disease-modifying drugs for the treatment of Alzheimer disease. LY450139 is a &ggr;-secretase inhibitor currently in clinical development, with doses being optimized through the use of biomarkers. Methods: To further characterize biomarker responses to LY450139, single oral doses of 60, 100, or 140 mg were administered to volunteers without neuropsychiatric disease. Extensive safety assessments were obtained along with measures of changes in amyloid-&bgr; (A&bgr;) in plasma and cerebrospinal fluid (CSF). A measure of the change in plasma A&bgr;1-40 was derived (area above the curve), which was determined by both the magnitude and duration of A&bgr;1-40 reduction. Results: A total of 31 subjects (ages 49-53 years, 19 men) were enrolled. With the possible exception of headache, no clinically significant adverse events or laboratory changes were observed. A dose-proportional increase in drug exposure was present in plasma and in CSF. A dose-dependent change in plasma A&bgr;1-40 area above the curve was also demonstrated. Using the 140-mg dose, a maximum 72.6% reduction in plasma A&bgr;1-40 was demonstrated that did not return to baseline for more than 12 hours. Cerebrospinal fluid concentrations of A&bgr; were unchanged 4 hours after drug administration. Conclusions: These data show that single doses of LY450139 up to 140 mg are accompanied by a dose-dependent plasma A&bgr; response. No response in CSF A&bgr; was apparent 4 hours after dosing.


Alzheimers & Dementia | 2016

Phase 3 solanezumab trials: Secondary outcomes in mild Alzheimer's disease patients

Eric Siemers; Karen Sundell; Christopher Carlson; Michael Case; Gopalan Sethuraman; Hong Liu-Seifert; Sherie A. Dowsett; Michael J. Pontecorvo; Robert A. Dean; Ronald B. DeMattos

EXPEDITION and EXPEDITION2 were identically designed placebo‐controlled phase 3 studies assessing effects of solanezumab, an antiamyloid monoclonal antibody binding soluble amyloid‐β peptide, on cognitive and functional decline over 80 weeks in patients with mild‐to‐moderate Alzheimers disease (AD). Primary findings for both studies have been published.

Collaboration


Dive into the Robert A. Dean's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leslie M. Shaw

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kaj Blennow

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge