Network


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

Hotspot


Dive into the research topics where Gopalan Sethuraman is active.

Publication


Featured researches published by Gopalan Sethuraman.


Annals of Neurology | 2006

Rapamycin causes regression of astrocytomas in tuberous sclerosis complex

David Neal Franz; Jennifer Leonard; Cynthia Tudor; Gail Chuck; Marguerite M. Caré; Gopalan Sethuraman; Argirios Dinopoulos; George Thomas; Kerry R. Crone

Tuberous sclerosis complex (TSC) is a genetic disorder characterized by the formation of hamartomas in multiple organs. Five to 15% of affected individuals display subependymal giant cell astrocytomas, which can lead to substantial neurological and postoperative morbidity due to the production of hydrocephalus, mass effect, and their typical location adjacent to the foramen of Monro. We sought to see whether therapy with oral rapamycin could affect growth or induce regression in astrocytomas associated with TSC.


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

BACKGROUNDnAlzheimers 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.nnnMETHODSnWe 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.nnnRESULTSnThe 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.nnnCONCLUSIONSnAs 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.)


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.


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.


Schizophrenia Research | 2005

A retrospective comparison of cumulative time spent in remission during treatment with olanzapine or risperidone among patients with schizophrenia

Gopalan Sethuraman; Cindy C. Taylor; Mark Enerson; Eduardo Dunayevich

BACKGROUNDnAvailable studies suggest comparable efficacy of olanzapine and risperidone for the treatment of schizophrenia over the short term.nnnMETHODnThis retrospective analysis of data from a 28-week, double-blind, schizophrenia trial compared the cumulative amount of time that patients met severity criteria for remission during olanzapine (10-20 mg/day) or risperidone (4-12 mg/day) treatment.nnnRESULTSnThe percentage cumulative time spent in remission was 40% for olanzapine- and 31% for risperidone-treated patients (P = 0.03) using Definition 1 (PANSS items P1, P2, P3, N1, N4, N6, G5, G9 < or = 3), and 18% and 11% (P = 0.01), respectively, using Definition 2 (BPRS Total reduced 50%, BPRS psychosis items < or = 3, CGI-severity < or = 3).nnnCONCLUSIONnDuring 28 weeks of treatment, olanzapine-treated patients spent more cumulative time in remission than risperidone-treated patients.


Journal of Alzheimer's Disease | 2012

Enriching Amnestic Mild Cognitive Impairment Populations for Clinical Trials: Optimal Combination of Biomarkers to Predict Conversion to Dementia

Peng Yu; Robert A. Dean; Stephen D. Hall; Yuan Qi; Gopalan Sethuraman; Brian A. Willis; Eric Siemers; Ferenc Martenyi; Johannes Tauscher; Adam J. Schwarz

The goal of this study was to identify the optimal combination of magnetic resonance imaging (MRI), [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET), and cerebrospinal fluid (CSF) biomarkers to predict conversion from amnestic mild cognitive impairment (aMCI) to Alzheimers disease (AD) dementia within two years, for enriching clinical trial populations. Data from 63 subjects in the Alzheimers Disease Neuroimaging Initiative aMCI cohort who had MRI and FDG-PET imaging along with CSF data at baseline and at least two years clinical follow-up were used. A Bayesian classification method was used to determine which combination of 31 variables (MRI, FDG-PET, CSF measurements, apolipoprotein E (ApoE) genotype, and cognitive scores) provided the most accurate prediction of aMCI to AD conversion. The cost and time trade-offs for the use of these biomarkers as inclusion criteria in clinical trials were evaluated. Using the combination of all biomarkers, ApoE genotype, and cognitive scores, we achieved an accuracy of 81% in predicting aMCI to AD conversion. With only ApoE genotype and cognitive scores, the prediction accuracy decreased to 62%. By comparing individual modalities, we found that MRI measures had the best predictive power (accuracy = 78%), followed by ApoE, FDG-PET, CSF, and the Alzheimers disease assessment scale-cognitive subscale. The combination of biomarkers from different modalities, measuring complementary aspects of AD pathology, provided the most accurate prediction of aMCI to AD conversion within two years. This was predominantly driven by MRI measures, which emerged as the single most powerful modality. Overall, the combination of MRI, ApoE, and cognitive scores provided the best trade-off between cost and time compared with other biomarker combinations for patient recruitment in clinical trial.


Alzheimer's & Dementia: Translational Research & Clinical Interventions | 2015

Delayed-start analysis: Mild Alzheimer's disease patients in solanezumab trials, 3.5 years

Hong Liu-Seifert; Eric Siemers; Karen C. Holdridge; Scott W. Andersen; Ilya Lipkovich; Christopher Carlson; Gopalan Sethuraman; Sharon L. Hoog; Roza Hayduk; Rachelle S. Doody; Paul S. Aisen

Solanezumab is an anti‐amyloid monoclonal antibody in clinical testing for treatment of Alzheimers disease (AD). Its mechanism suggests the possibility of slowing the progression of AD.


Journal of Psychiatric Research | 2009

In search of moderators and mediators of hyperglycemia with atypical antipsychotic treatment

Gerald M. Reaven; Jeffrey A. Lieberman; Gopalan Sethuraman; Helena C. Kraemer; John M. Davis; Christine Blasey; Ming T. Tsuang; Alan F. Schatzberg

Signal detection methods were used to identify values of metabolic variables that predict development of prediabetes or diabetes before (moderators) or associated with treatment (mediators), utilizing data from two multi-center clinical trials of patients with schizophrenia, treated for 6 months with olanzapine (OLZ) or ziprasidone (ZIP). At baseline, participants were often overweight/obese (63% with a body mass index >or=25.0kg/m(2)), dyslipidemic [more than one-third had elevated triglyceride (TG) and low high-density lipoprotein cholesterol (HDL-C) concentrations], and prediabetic (20%). Weight gain was significantly greater in OLZ-treated patients, as was accentuation of dyslipidemia. However, there were no significant correlations between weight gain and lipid changes from baseline to weeks 2, 4, 8 or to last observation. Type 2 diabetes developed in 4% and prediabetes in 18% of the population. Significant baseline predictors of diabetes were a HDL-C concentration <28mg/dL, or being >or=58-years-old if HDL-C concentration was >or=28mg/dL. Baseline plasma glucose concentration >or=92mg/dL was the only significant predictor of developing prediabetes, accounting for 60% of cases. Post-treatment increments in plasma TG concentrations >or=145mg/dL or >or=59mg/dL were significant predictors of diabetes (23%) or prediabetes (27%), respectively. If the increase in TG was <145mg/dL, rapid weight gain >or=6.1kg in 2 weeks predicted development of diabetes (18%). These findings provide a quantitative approach to identify those at greatest treatment-associated risk to develop glucose intolerance, and emphasize the need to address co-morbid medical disorders in these patients.


Journal of Alzheimer's Disease | 2011

Validation of ELISA Methods for Quantification of Total Tau and Phosphorylated-Tau181 in Human Cerebrospinal Fluid with Measurement in Specimens from Two Alzheimer's Disease Studies

D. Richard Lachno; Martin J. Romeo; Eric Siemers; Hugo Vanderstichele; Els Coart; Robert J. Konrad; Joseph J. Zajac; Jayne A. Talbot; Hans F. Jensen; Gopalan Sethuraman; Ronald B. DeMattos; Patrick May; Robert A. Dean

Tau measurements in cerebrospinal fluid (CSF) are gaining acceptance as aids to diagnosis of Alzheimers disease (AD) and differentiation from other dementias. Two ELISA assays, the INNOTEST® hTAU Ag and the INNOTEST® PHOSPHO-TAU(181P) for quantification of t-tau and p-tau181 respectively, have been validated to regulatory standards. Validation parameters were determined by repeated testing of human CSF pools. Specimens from Phase 2 studies of the γ-secretase inhibitor semagacestat and the therapeutic antibody solanezumab at baseline and following 12-14 weeks of treatment were also tested. Estimated intra-assay CV for repeated testing of 3 CSF pools were ≤11.5% and RE varied between -14.1% and +6.4%. Inter-assay CV for t-tau was <5% and RE was within ±8%. For p-tau181, inter-assay CV was <9% and RE was within ±2.5%. Total CV (intra-assay plus inter-assay) were below 10% for both analytes. Up to 20-fold dilutional linearity was demonstrated for both analytes. Stability of t-tau and p-tau181 was demonstrated in CSF during five freeze-thaw cycles at ≤-20 °C and ≤-70 °C and at 18-22 °C for up to 24 h. Neither semagacestat nor solanezumab interfered with either assay. Inter-individual t-tau and p-tau181 concentrations were highly variable but intra-individual variations were small. These assays are suitable for analysis of CSF t-tau and p-tau181 in a single laboratory supporting multi-center AD clinical trials. No effect of treatment with semagacestat or solanezumab was observed in response to three months of drug administration.


Alzheimer's Research & Therapy | 2015

Peripheral and central effects of γ-secretase inhibition by semagacestat in Alzheimer's disease

Rachelle S. Doody; Rema Raman; Reisa A. Sperling; Eric Seimers; Gopalan Sethuraman; Richard C. Mohs; Martin R. Farlow; Takeshi Iwatsubo; Bruno Vellas; Xiaoying Sun; Karin Ernstrom; Ronald G. Thomas; Paul S. Aisen

IntroductionThe negative efficacy study examining the γ-secretase inhibitor semagacestat in mild to moderate Alzheimer’s disease (AD) included a number of biomarkers of the disease as well as safety outcomes. We analyzed these data to explore relationships between drug exposure and pharmacodynamic effects and to examine the correlations among outcome measures.MethodsThe study was a multicenter, randomized, placebo-controlled trial of two dose regimens of semagacestat and a placebo administered for 18xa0months to individuals with mild to moderate AD. Changes in measures of central and peripheral drug activity were compared between the three treatment groups using one-way analysis of variance. The relationship between changes in each of the outcome measures and measures of drug exposure and peripheral pharmacodynamic effect were assessed using Spearman’s correlation coefficient.ResultsAssignment to the active treatment arms was associated with reduction in plasma amyloid-β (Aβ) peptides, increase in ventricular volume, decrease in cerebrospinal fluid phosphorylated tau (p-tau) and several other laboratory measures and adverse event categories. Within the active arms, exposure to drug, as indicated by area under the concentration curve (AUC) of blood concentration, was associated with reduction in plasma Aβ peptides and a subset of laboratory changes and adverse event rates. Ventricular volume increase, right hippocampal volume loss and gastrointestinal symptoms were related to change in plasma Aβ peptide but not AUC, supporting a link to inhibition of γ-secretase cleavage of the amyloid precursor protein. Cognitive decline correlated with ventricular expansion and reduction in p-tau.ConclusionThese findings may inform future studies of drugs targeting secretases involved in Aβ generation.Trial registrationClinicalTrials.gov Identifier: NCT00594568. Registered 11 January 2008.

Collaboration


Dive into the Gopalan Sethuraman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge