Network


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

Hotspot


Dive into the research topics where Anton P. Porsteinsson is active.

Publication


Featured researches published by Anton P. Porsteinsson.


The New England Journal of Medicine | 2014

Two Phase 3 Trials of Bapineuzumab in Mild-to-Moderate Alzheimer's Disease

Stephen Salloway; Reisa A. Sperling; Nick C. Fox; Kaj Blennow; William E. Klunk; Murray A. Raskind; Marwan N. Sabbagh; Lawrence S. Honig; Anton P. Porsteinsson; Steven H. Ferris; Marcel Reichert; Nzeera Ketter; Bijan Nejadnik; Volkmar Guenzler; Maja Miloslavsky; Daniel Wang; Yuan Lu; Julia Lull; Iulia Cristina Tudor; Enchi Liu; Michael Grundman; Eric Yuen; Ronald S. Black; H. Robert Brashear

BACKGROUND Bapineuzumab, a humanized anti-amyloid-beta monoclonal antibody, is in clinical development for the treatment of Alzheimers disease. METHODS We conducted two double-blind, randomized, placebo-controlled, phase 3 trials involving patients with mild-to-moderate Alzheimers 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 Alzheimers 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 Alzheimers 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.).


Biological Psychiatry | 2010

Meta-analysis of Alzheimer's disease risk with obesity, diabetes, and related disorders.

Louis A. Profenno; Anton P. Porsteinsson; Stephen V. Faraone

BACKGROUND Late-onset Alzheimers disease (AD) is a multifactorial and heterogeneous disorder with major risk factors including advanced age, presence of an apolipoprotein E epsilon4 (APOE4) allele, and family history of AD. Other risk factors may be obesity and diabetes and related disorders, which are highly prevalent. METHODS We reviewed longitudinal epidemiological studies of body mass, diabetes, metabolic syndrome, and glucose and insulin levels on risk for AD. We conducted meta-analyses of the results from these studies. RESULTS For obesity assessed by body mass index, the pooled effect size for AD was 1.59 (95% confidence interval [CI] 1.02-2.5; z = 2.0; p = .042), and for diabetes, the pooled effect size for AD was 1.54 (95% CI 1.33-1.79; z = 5.7; p < .001). Eggers test did not find significant evidence for publication bias in the meta-analysis for obesity (t = -1.4, p = .21) or for diabetes (t = -.86, p = .42). Since these disorders are highly comorbid, we conducted a meta-analysis combining all studies of obesity, diabetes, and abnormal glucose or insulin levels, which yielded a highly significant pooled effect size for AD of 1.63 (95% CI 1.39-1.92; z = 5.9; p < .001). CONCLUSIONS Obesity and diabetes significantly and independently increase risk for AD. Though the level of risk is less than that with the APOE4 allele, the high prevalence of these disorders may result in substantial increases in future incidence of AD. Physiological changes common to obesity and diabetes plausibly promote AD.


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.


Lancet Neurology | 2012

Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis

Reisa A. Sperling; Stephen Salloway; David J. Brooks; Donatella Tampieri; Jerome Barakos; Nick C. Fox; Murray A. Raskind; Marwan N. Sabbagh; Lawrence S. Honig; Anton P. Porsteinsson; Ivan Lieberburg; H. Michael Arrighi; Kristen Morris; Yuan Lu; Enchi Liu; Keith M. Gregg; H. Robert Brashear; Gene G. Kinney; Ronald S. Black; Michael Grundman

BACKGROUND Amyloid-related imaging abnormalities (ARIA) have been reported in patients with Alzheimers disease treated with bapineuzumab, a humanised monoclonal antibody against amyloid β. ARIA include MRI signal abnormalities suggestive of vasogenic oedema and sulcal effusions (ARIA-E) and microhaemorrhages and haemosiderin deposits (ARIA-H). Our aim was to investigate the incidence of ARIA during treatment with bapineuzumab, and evaluate associated risk factors. METHODS Two neuroradiologists independently reviewed 2572 fluid-attenuated inversion recovery (FLAIR) MRI scans from 262 participants in two phase 2 studies of bapineuzumab and an open-label extension study. Readers were masked to the patients treatment, APOE ɛ4 genotype, medical history, and demographics. Patients were included in risk analyses if they had no evidence of ARIA-E in their pre-treatment MRI, had received bapineuzumab, and had at least one MRI scan after treatment. We used Kaplan-Meier survival analysis to examine the distribution of incident ARIA-E from the start of bapineuzumab treatment and proportional hazards regression models to assess risk factors associated with ARIA. FINDINGS 210 patients were included in the risk analyses. 36 patients (17%) developed ARIA-E during treatment with bapineuzumab; 15 of these ARIA-E cases (42%) had not been detected previously. 28 of these patients (78%) did not report associated symptoms. Adverse events, reported in eight symptomatic patients, included headache, confusion, and neuropsychiatric and gastrointestinal symptoms. Incident ARIA-H occurred in 17 of the patients with ARIA-E (47%), compared with seven of 177 (4%) patients without ARIA-E. 13 of the 15 patients in whom ARIA were detected in our study received additional treatment infusions while ARIA-E were present, without any associated symptoms. Occurrence of ARIA-E increased with bapineuzumab dose (hazard ratio [HR] 2·24 per 1 mg/kg increase in dose, 95% CI 1·40-3·62; p=0·0008) and presence of APOE ɛ4 alleles (HR 2·55 per allele, 95% CI 1·57-4·12; p=0·0001). INTERPRETATION ARIA consist of a spectrum of imaging findings with variable clinical correlates, and some patients with ARIA-E remain asymptomatic even if treatment is continued. The increased risk of ARIA among APOE ɛ4 carriers, its association with high bapineuzumab dose, and its timecourse in relation to dosing suggest an association between ARIA and alterations in vascular amyloid burden. FUNDING Elan Corporation, Janssen Alzheimer Immunotherapy, Wyeth Pharmaceuticals, and Pfizer.


JAMA | 2014

Effect of Citalopram on Agitation in Alzheimer Disease: The CitAD Randomized Clinical Trial

Anton P. Porsteinsson; Lea T. Drye; Bruce G. Pollock; D.P. Devanand; Constantine Frangakis; Zahinoor Ismail; Christopher Marano; Curtis L. Meinert; Jacobo Mintzer; Cynthia A. Munro; Gregory H. Pelton; Peter V. Rabins; Paul B. Rosenberg; Lon S. Schneider; David M. Shade; Daniel Weintraub; Jerome A. Yesavage; Constantine G. Lyketsos

IMPORTANCE Agitation is common, persistent, and associated with adverse consequences for patients with Alzheimer disease. Pharmacological treatment options, including antipsychotics are not satisfactory. OBJECTIVE The primary objective was to evaluate the efficacy of citalopram for agitation in patients with Alzheimer disease. Key secondary objectives examined effects of citalopram on function, caregiver distress, safety, cognitive safety, and tolerability. DESIGN, SETTING, AND PARTICIPANTS The Citalopram for Agitation in Alzheimer Disease Study (CitAD) was a randomized, placebo-controlled, double-blind, parallel group trial that enrolled 186 patients with probable Alzheimer disease and clinically significant agitation from 8 academic centers in the United States and Canada from August 2009 to January 2013. INTERVENTIONS Participants (n = 186) were randomized to receive a psychosocial intervention plus either citalopram (n = 94) or placebo (n = 92) for 9 weeks. Dosage began at 10 mg per day with planned titration to 30 mg per day over 3 weeks based on response and tolerability. MAIN OUTCOMES AND MEASURES Primary outcome measures were based on scores from the 18-point Neurobehavioral Rating Scale agitation subscale (NBRS-A) and the modified Alzheimer Disease Cooperative Study-Clinical Global Impression of Change (mADCS-CGIC). Other outcomes were based on scores from the Cohen-Mansfield Agitation Inventory (CMAI) and the Neuropsychiatric Inventory (NPI), ability to complete activities of daily living (ADLs), caregiver distress, cognitive safety (based on scores from the 30-point Mini Mental State Examination [MMSE]), and adverse events. RESULTS Participants who received citalopram showed significant improvement compared with those who received placebo on both primary outcome measures. The NBRS-A estimated treatment difference at week 9 (citalopram minus placebo) was -0.93 (95% CI, -1.80 to -0.06), P = .04. Results from the mADCS-CGIC showed 40% of citalopram participants having moderate or marked improvement from baseline compared with 26% of placebo recipients, with estimated treatment effect (odds ratio [OR] of being at or better than a given CGIC category) of 2.13 (95% CI, 1.23-3.69), P = .01. Participants who received citalopram showed significant improvement on the CMAI, total NPI, and caregiver distress scores but not on the NPI agitation subscale, ADLs, or in less use of rescue lorazepam. Worsening of cognition (-1.05 points; 95% CI, -1.97 to -0.13; P = .03) and QT interval prolongation (18.1 ms; 95% CI, 6.1-30.1; P = .01) were seen in the citalopram group. CONCLUSIONS AND RELEVANCE Among patients with probable Alzheimer disease and agitation who were receiving psychosocial intervention, the addition of citalopram compared with placebo significantly reduced agitation and caregiver distress; however, cognitive and cardiac adverse effects of citalopram may limit its practical application at the dosage of 30 mg per day. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00898807.


Current Alzheimer Research | 2008

Memantine Treatment in Patients with Mild to Moderate Alzheimers Disease Already Receiving a Cholinesterase Inhibitor: A Randomized, Double-Blind, Placebo-Controlled Trial

Anton P. Porsteinsson; George T. Grossberg; Jacobo Mintzer; Jason T. Olin

OBJECTIVE To evaluate the efficacy and safety of memantine in patients with mild to moderate Alzheimers disease (AD) receiving cholinesterase inhibitor (ChEI) treatment. METHODS Participants (N= 433) with probable AD, Mini-Mental State Exam (MMSE) scores between 10-22 (inclusive), and concurrent stable use of ChEIs (donepezil, rivastigmine, galantamine) were randomized to placebo or memantine (20 mg once daily) for 24 weeks. Primary outcomes were changes from baseline on the Alzheimers Disease Assessment Scale-cognitive subscale (ADAS-cog) and on Clinicians Interview-Based Impression of Change Plus Caregiver Input (CIBIC-Plus) score. Secondary measures comprised the 23-item Alzheimer Disease Cooperative Study-Activities of Daily Living Scale (ADCS-ADL(23)), Neuropsychiatric Inventory (NPI), and MMSE. RESULTS At the end of the trial, there were no statistically significant differences between the memantine- and placebo group on primary and secondary outcome measures. The incidence of adverse events (AEs) was similar between the two groups, with no AE occurring in more than 5% of memantine-treated patients and at a rate twice that of the placebo group. CONCLUSIONS In this trial, memantine did not show an advantage over placebo based on protocol-specified primary or secondary analyses in patients with mild to moderate AD on stable ChEI regimens. There were no significant differences in tolerability and safety between the memantine- and placebo groups.


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.


American Journal of Geriatric Psychiatry | 2001

Placebo-Controlled Study of Divalproex Sodium for Agitation in Dementia

Anton P. Porsteinsson; Pierre N. Tariot; Rosemary Erb; Christopher Cox; Elizabeth Smith; Laura Jakimovich; John Noviasky; Nancy Kowalski; Connie Holt; Carrie Irvine

The authors assessed the efficacy, tolerability, and safety of divalproex sodium for the treatment of agitation associated with dementia in a 6-week, randomized study of 56 nursing home patients with agitation and dementia treated with either placebo or individualized doses of divalproex sodium. Participants were blinded to treatment except for a physician-monitor and a pharmacist. When several covariates were taken into account, the drug/placebo difference in Brief Psychiatric Rating Scale Agitation scores became statistically significant (P=0.05). Sixty-eight percent of patients on divalproex were rated as showing reduced agitation on the Clinical Global Impression scale, vs. 52% on placebo (P=0.06 in the adjusted analysis). Side effects occurred in 68% of the divalproex group vs. 33% of the placebo group (P=0.03) and were generally rated as mild. This placebo-controlled study, despite some limitations, suggests possible short-term efficacy, tolerability, and safety of divalproex for agitation in dementia and supports further placebo-controlled studies.


Neurology | 2011

A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease

Stephen Salloway; Reisa A. Sperling; Ron Keren; Anton P. Porsteinsson; C.H. van Dyck; Pierre N. Tariot; Sid Gilman; Douglas L. Arnold; Susan Abushakra; C. Hernandez; Gerald Crans; Earvin Liang; G. Quinn; Menghis Bairu; Aleksandra Pastrak; Jesse M. Cedarbaum

Objective: This randomized, double-blind, placebo-controlled, dose-ranging phase 2 study explored safety, efficacy, and biomarker effects of ELND005 (an oral amyloid anti-aggregation agent) in mild to moderate Alzheimer disease (AD). Methods: A total of 353 patients were randomized to ELND005 (250, 1,000, or 2,000 mg) or placebo twice daily for 78 weeks. Coprimary endpoints were the Neuropsychological Test Battery (NTB) and Alzheimers Disease Cooperative Study–Activities of Daily Living (ADCS-ADL) scale. The primary analysis compared 250 mg (n =84) to placebo (n =82) after an imbalance of infections and deaths led to early discontinuation of the 2 higher dose groups. Results: The 250 mg dose demonstrated acceptable safety. The primary efficacy analysis at 78 weeks revealed no significant differences between the treatment groups on the NTB or ADCS-ADL. Brain ventricular volume showed a small but significant increase in the overall 250 mg group (p =0.049). At the 250 mg dose, scyllo-inositol concentrations increased in CSF and brain and CSF Aβx-42 was decreased significantly compared to placebo (p =0.009). Conclusions: Primary clinical efficacy outcomes were not significant. The safety and CSF biomarker results will guide selection of the optimal dose for future studies, which will target earlier stages of AD. Classification of evidence: Due to the small sample sizes, this Class II trial provides insufficient evidence to support or refute a benefit of ELND005.


Alzheimers & Dementia | 2013

Neuropsychiatric symptoms in Alzheimer's disease: Past progress and anticipation of the future

Yonas E. Geda; Lon S. Schneider; Laura N. Gitlin; David S. Miller; Gwenn S. Smith; Joanne Bell; Jovier D. Evans; Michael K. Lee; Anton P. Porsteinsson; Krista L. Lanctôt; Paul B. Rosenberg; David L. Sultzer; Paul T. Francis; Henry Brodaty; Prasad P. Padala; Chiadikaobi Onyike; Luis Agüera Ortiz; Sonia Ancoli-Israel; Donald L. Bliwise; Jennifer L. Martin; Michael V. Vitiello; Kristine Yaffe; Phyllis C. Zee; Nathan Herrmann; Robert A. Sweet; Clive Ballard; Ni A. Khin; Cara Alfaro; Patrick S. Murray; Susan K. Schultz

Neuropsychiatric symptoms (NPS) in Alzheimers disease (AD) are widespread and disabling. This has been known since Dr. Alois Alzheimers first case, Frau Auguste D., presented with emotional distress and delusions of infidelity/excessive jealousy, followed by cognitive symptoms. Being cognizant of this, in 2010 the Alzheimers Association convened a research roundtable on the topic of NPS in AD. A major outcome of the roundtable was the founding of a Professional Interest Area (PIA) within the International Society to Advance Alzheimers Research and Treatment (ISTAART). The NPS‐PIA has prepared a series of documents that are intended to summarize the literature and provide more detailed specific recommendations for NPS research. This overview paper is the first of these living documents that will be updated periodically as the science advances. The overview is followed by syndrome‐specific synthetic reviews and recommendations prepared by NPS‐PIA workgroups on depression, apathy, sleep, agitation, and psychosis.

Collaboration


Dive into the Anton P. Porsteinsson's collaboration.

Top Co-Authors

Avatar

Jacobo Mintzer

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Constantine G. Lyketsos

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lon S. Schneider

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Paul B. Rosenberg

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Weintraub

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Cynthia A. Munro

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lea T. Drye

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge