Network


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

Hotspot


Dive into the research topics where Jessica Y. Mancuso is active.

Publication


Featured researches published by Jessica Y. Mancuso.


Neurology | 2014

Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease

Douglas Galasko; Joanne Bell; Jessica Y. Mancuso; James W. Kupiec; Marwan N. Sabbagh; Christopher H. van Dyck; Ronald G. Thomas; Paul S. Aisen

Objective: To examine safety, tolerability, and efficacy of PF-04494700, an inhibitor of the receptor for advanced glycation end products (RAGE), in mild to moderate Alzheimer disease (AD). Methods: Double-blind, placebo-controlled trial at 40 academic centers (United States). Subjects with AD and Mini-Mental State Examination score 14–26 were randomized to PF-04494700 60 mg/day × 6 days, then 20 mg daily (high dose); 15 mg/day × 6 days, then 5 mg daily (low dose); or placebo, for 18 months. Clinical and laboratory measures were used to evaluate safety and tolerability. The primary efficacy measure was the Alzheimers Disease Assessment Scale–cognitive (ADAS-cog). Secondary measures assessed clinical stage, function, behavior, MRI, and CSF biomarkers. Results: A total of 399 subjects were randomized. In a prespecified interim analysis, when 50% of subjects had completed the 6-month visit, the high dose was associated with confusion, falls, and greater ADAS-cog decline and was discontinued. A second prespecified analysis compared low-dose and placebo groups for futility and safety approximately 12 months after all subjects were randomized. This analysis met criteria for futility, and treatment was discontinued. There were no safety concerns in the low-dose group. Analyses including post-futility data showed decreased decline on the ADAS-cog in the low-dose group at month 18. Other clinical and biomarker measures showed no differences between low-dose treatment and placebo. Conclusions: PF-04494700 at 20 mg/d was associated with increased adverse events and cognitive decline. At 5 mg/d, PF-04494700 had a good safety profile. A potential benefit for this low dose on the ADAS-cog is not conclusive, because of high dropout and discontinuation rates subsequent to the interim analyses. Classification of evidence: This study provides Class I evidence that in patients with AD high-dose PF-04494700 increased cognitive decline at 6 months and Class IV evidence that low-dose PF-04494700 slowed cognitive decline at 18 months.


Journal of Medicinal Chemistry | 2014

Discovery and preclinical characterization of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo-[3,4-b]pyrazine (PF470): a highly potent, selective, and efficacious metabotropic glutamate receptor 5 (mGluR5) negative allosteric modulator.

Lei Zhang; Gayatri Balan; Gabriela Barreiro; Brian P. Boscoe; Lois K. Chenard; Julie Cianfrogna; Michelle Marie Claffey; Laigao Chen; Karen J. Coffman; Susan E. Drozda; Joshua R. Dunetz; Kari R. Fonseca; Paul Galatsis; Sarah Grimwood; John T. Lazzaro; Jessica Y. Mancuso; Emily L. Miller; Matthew R. Reese; Bruce N. Rogers; Isao Sakurada; Marc B. Skaddan; Deborah L. Smith; Antonia F. Stepan; Patrick Trapa; Jamison B. Tuttle; Patrick Robert Verhoest; Daniel P. Walker; Ann S. Wright; Margaret M. Zaleska; Kenneth Zasadny

A novel series of pyrazolopyrazines is herein disclosed as mGluR5 negative allosteric modulators (NAMs). Starting from a high-throughput screen (HTS) hit (1), a systematic structure-activity relationship (SAR) study was conducted with a specific focus on balancing pharmacological potency with physicochemical and pharmacokinetic (PK) properties. This effort led to the discovery of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo[3,4-b]pyrazine (PF470, 14) as a highly potent, selective, and orally bioavailable mGluR5 NAM. Compound 14 demonstrated robust efficacy in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-rendered Parkinsonian nonhuman primate model of l-DOPA-induced dyskinesia (PD-LID). However, the progression of 14 to the clinic was terminated because of a potentially mechanism-mediated finding consistent with a delayed-type immune-mediated type IV hypersensitivity in a 90-day NHP regulatory toxicology study.


Drug Metabolism and Disposition | 2012

An Evaluation of Using Rat-derived Single-dose Neuropharmacokinetic Parameters to Project Accurately Large Animal Unbound Brain Drug Concentrations

Angela C. Doran; Sarah Osgood; Jessica Y. Mancuso; Christopher L. Shaffer

Previous publications suggest that interstitial fluid compound concentrations (CISF) best determine quantitative neurotherapeutic pharmacology relationships, although confirming large animal CISF remains elusive. Therefore, this work primarily evaluated using respective acute dose, rat-derived unbound brain compound concentration-to-unbound plasma compound concentration ratios (Cb,u/Cp,u) to project accurately dog and nonhuman primate (nhp) Cb,u, a CISF surrogate, from measured Cp,u for the highly permeable non-P-glycoprotein substrates N-{(3R,4S)-3-[4-(5-cyano-2-thienyl)phenyl]tetrahydro-2H-pyran-4-yl}propane-2-sulfonamide (PF-4778574) and [4-chloro-5-fluoro-2-(3-methoxy-2-methyl-phenoxy)-benzyl]-methylamine (CE-157119) and the P-glycoprotein substrates risperidone and 9-hydroxyrisperidone. First, in rats, it was determined for eight of nine commercial compounds that their single-dose-derived Cb,u/Cp,u were ≤2.5-fold different from their steady-state values; for all nine drugs, their Cb,u/Cp,u were ≤2.5-fold different from their steady-state CISF/Cp,u (Drug Metab Dispos 37:787–793, 2009). Subsequently, PF-4778574, CE-157119 and risperidone underwent rat, dog, and nhp neuropharmacokinetics studies. In large animals at each measured Cp,u, the methodology adequately predicted [estimated mean (95% confidence interval) of 1.02 (0.80, 1.29)] the observed Cb,u for PF-4778574 and CE-157119 but underpredicted [0.17 (0.12, 0.22)] Cb,u for risperidone and 9-hydroxyrisperidone. The data imply that forecasting higher species Cb,u from a measured Cp,u and rat acute dose-determined Cb,u:Cp,u is of high confidence for nonefflux transporter substrates that show net passive diffusion (PF-4778574) or net active influx (CE-157119) at the blood-brain barrier in rats. However, this methodology appears ineffective for correctly predicting large animal Cb,u for P-glycoprotein substrates (risperidone and 9-hydroxyrisperidone) because of their apparently much greater Cp,u-favoring Cb,u:Cp,u asymmetry in rats versus dogs or nhp. Instead, for such P-glycoprotein substrates, large animal-specific cerebrospinal fluid compound concentrations (CCSF) seemingly best represent Cb,u.


Drug Information Journal | 2006

A Confidence Interval for the Maximal Mean QT Interval Change Caused by Drug Effect

Morris L. Eaton; Robb J. Muirhead; Jessica Y. Mancuso; Sheela Kolluri

A statistical problem of primary interest in a thorough QT/QTc study is that of deciding if a drug is noninferior to placebo in terms of QT/QTc prolongation. A standard way of approaching this problem is to construct a 90% two-sided (or a 95% one-sided) confidence interval, using the t distribution, at each time point in the study for the difference in mean QTc between drug and placebo and to conclude that the drug is noninferior to placebo if the upper end points of all of these confidence intervals is less than a prespecified constant, such as 10 ms. Under standard normality assumptions, this procedure corresponds to both an intersection-union test and the likelihood ratio test of size .05. It is not without its drawbacks, however. It is conservative in that the probability of a type I error may be smaller than the intended level .05. It is also biased, which means that the power function, for some values of parameters in the alternative space, takes values less than .05. The May 12, 2005, draft of the International Conference on Harmonisation E14 guidance states: “a negative ‘thorough QT/QTc study’ is one in which the upper bound of the 95% one-sided confidence interval for the largest time-matched mean effect of the drug on the QTc interval excludes 10 ms.” In this article, we show how an approximate confidence interval can be constructed for the largest difference in population mean QT/QTc between drug and placebo. The interval is approximate in the sense that, as sample sizes increase, the asymptotic probability of coverage is at least as large as intended. The results of simulations on a proposed one-sided 95% confidence interval are provided and discussed. Situations in which this interval works well, and does not work well, are delineated.


Journal of Pharmaceutical Sciences | 2014

Diphenhydramine has Similar Interspecies Net Active Influx at the Blood–Brain Barrier

Christopher L. Shaffer; Sarah Osgood; Jessica Y. Mancuso; Angela C. Doran

In rats, oxycodone, diphenhydramine, and [4-chloro-5-fluoro-2-(3-methoxy-2-methyl-phenoxy)-benzyl]-methylamine (CE-157119) undergo net active influx at the blood-brain barrier (BBB) based on significantly greater interstitial fluid compound concentrations (CISF ) than unbound plasma compound concentrations (Cp,u ). Oxycodone and diphenhydramine have CISF :Cp,u of 3.0 and 5.5, respectively, while CE-157119 has an unbound brain compound concentration (Cb,u ):Cp,u of 3.90; Cb,u is a high-confidence CISF surrogate. However, only CE-157119 has published dog and nonhuman primate (nhp) neuropharmacokinetics, which show similar Cb,u :Cp,u (4.61 and 2.04, respectively) as rats. Thus, diphenhydramine underwent identical interspecies neuropharmacokinetics studies to determine if its net active BBB influx in rats replicated in dogs and/or nhp. The single-dose-derived rat Cb,u :Cp,u (3.90) was consistent with prior steady-state-derived CISF :Cp,u and similar to those in dogs (4.88) and nhp (4.51-5.00). All large animal interneurocompartmental ratios were ≤1.8-fold different than their rat values, implying that diphenhydramine has constant and substantial Cb,u -favoring disequilibria in these mammals. Accordingly, the applied Cb,u -forecasting methodology accurately predicted [estimated mean (95% confidence interval) of 0.84 (0.68, 1.05)] Cb,u from each measured Cp,u in large animals. The collective datasets suggest these Cb,u -preferring asymmetries are mediated by a species-independent BBB active uptake system whose identification, full characterization, and structure-activity relationships should be prioritized for potential exploitation.


Journal of Stroke & Cerebrovascular Diseases | 2016

Phosphodiesterase-5 Inhibitor PF-03049423 Effect on Stroke Recovery: A Double-Blind, Placebo-Controlled Randomized Clinical Trial

Franco Di Cesare; Jessica Y. Mancuso; Phil Woodward; Martin M. Bednar; Peter T. Loudon

BACKGROUND The therapeutic potential of phosphodiesterase-5 inhibitor PF-03049423 was evaluated in a phase 2, multicenter, randomized, double-blind, placebo-controlled study of subjects with acute ischemic stroke ( CLINICAL TRIAL REGISTRATION INFORMATION http://www.clinicaltrials.gov, unique identifier: NCT01208233; http://www.clinicaltrialsregister.eu, EudraCT number: 2010-021414-32). MATERIALS AND METHODS Subjects (N = 70) received PF-03049423 6 mg (or placebo, N = 67) once daily, orally, commencing between 24 and 78 hours of stroke onset, and continuing for 90 days. Postbaseline efficacy assessments were performed on Days 7, 14, 30, 60, and 90. Modified Rankin Scale (mRS), Barthel Index, National Institutes of Health Stroke Scale, Box and Blocks Test, Hand-Grip Strength Test, 10-Meter Walk Test, Repeatable Battery Assessment of Neuropsychological Status Naming and Coding Subtests, Line Cancellation Test, and Recognition Memory Test were administered to evaluate poststroke recovery. The primary endpoint was the mRS responder rate (score 0-2 at Day 90). The study included a planned interim analysis of efficacy data. RESULTS The primary efficacy analysis using logistic regression showed no statistically significant difference between PF-03049423 6 mg and placebo (responder rate of 42.6% and 46.2%, respectively). Although PF-03049423 showed a satisfactory safety and tolerability profile, no signal of efficacy emerged from any of the outcome measures. CONCLUSIONS PF-03049423 showed no therapeutic potential for acute ischemic stroke.


Alzheimer's Research & Therapy | 2018

A Phase 2 clinical trial of PF-05212377 (SAM-760) in subjects with mild to moderate Alzheimer’s disease with existing neuropsychiatric symptoms on a stable daily dose of donepezil

Terence Fullerton; Brendon Binneman; William David; Marielle Delnomdedieu; James W. Kupiec; Peter Lockwood; Jessica Y. Mancuso; Jeffrey Miceli; Joanne Bell

BackgroundSymptomatic benefits have been reported for 5-HT6 receptor antagonists in Alzheimer’s disease (AD) trials. SAM-760 is a potent and selective 5-HT6 receptor antagonist that has demonstrated central 5-HT6 receptor saturation in humans at a dose of 30 mg.MethodsThis was a randomized, double-blind, placebo-controlled, parallel-group, multicenter trial evaluating the efficacy and safety of SAM-760 30 mg once daily (QD) for 12 weeks in subjects with AD on a stable regimen of donepezil 5 to 10 mg QD. The study included an interim analysis with stopping rules for futility or efficacy after 180 subjects completed the week 12 visit. Up to 342 subjects with AD (Mini-Mental State Examination (MMSE) score 10–24) and neuropsychiatric symptoms (Neuropsychiatric Inventory (NPI) total score ≥ 10) were to be enrolled if the study continued after the interim analysis. After a 4-week, single-blind, placebo run-in period, subjects entered the 12-week double-blind period and were randomized to either SAM-760 or placebo. The primary and key secondary efficacy endpoints were the change from baseline in Alzheimer’s Disease Assessment Scale-cognitive subscale (ADAS-cog13) and NPI total scores. Mixed models for repeated measures were used to analyze the data.ResultsAt the interim analysis, when 186 subjects had been randomized and 163 had completed the week 12 visit, the study met futility criteria and was stopped. The mean week 12 treatment difference was 0.70 points (P = 0.43) for ADAS-cog13 and 2.19 points (P = 0.20) for NPI score, both of which were numerically in favor of placebo. Other secondary endpoints did not demonstrate any significant benefit for SAM-760. In total, 46.2% of SAM-760 subjects reported adverse events (AE) versus 44.7% for placebo, and there were 5 (5.5%) serious AEs in the SAM-760 group versus 3 (3.2%) for placebo. There were two deaths, one prior to randomization and one in the SAM-760 group (due to a traffic accident during washout of active treatment).ConclusionsSAM-760 was safe and well tolerated, but there was no benefit of SAM-760 on measures of cognition, neuropsychiatric symptoms, or daily function. Differences in trial design, study population, region, or pharmacological profile may explain differences in outcome compared with other 5-HT6 receptor antagonists.Trial registrationClinicaltrials.gov, NCT01712074. Registered 19 October 2012.


Drug Metabolism and Disposition | 2018

Metabolism of a 5HT6 antagonist, 2-methyl-1-(phenylsulfonyl)-4-(piperazin-1-yl)-1H-benzo[d]imidazole (SAM-760): impact of sulfonamide metabolism on diminution of a ketoconazole mediated clinical drug-drug interaction

Aarti Sawant-Basak; R. Scott Obach; Angela C. Doran; Peter Lockwood; Klaas Schildknegt; Hongying Gao; Jessica Y. Mancuso; Tse S; Thomas A. Comery

SAM-760 [(2-methyl-1-(phenylsulfonyl)-4-(piperazin-1-yl)-1H-benzo[d]imidazole)], a 5HT6 antagonist, was investigated in humans for the treatment of Alzheimer’s disease. In liver microsomes and recombinant cytochrome P450 (P450) isozymes, SAM-760 was predominantly metabolized by CYP3A (∼85%). Based on these observations and an expectation of a 5-fold magnitude of interaction with moderate to strong CYP3A inhibitors, a clinical DDI study was performed. In the presence of ketoconazole, the mean Cmax and area under the plasma concentration-time curve from time zero extrapolated to infinite time values of SAM-760 showed only a modest increase by 30% and 38%, respectively. In vitro investigation of this unexpectedly low interaction was undertaken using [14C]SAM-760. Radiometric profiling in human hepatocytes confirmed all oxidative metabolites previously observed with unlabeled SAM-760; however, the predominant radiometric peak was an unexpected polar metabolite that was insensitive to the pan-P450 inhibitor 1-aminobenzotriazole. In human hepatocytes, radiometric integration attributed 43% of the total metabolism of SAM-760 to this non-P450 pathway. Using an authentic standard, this predominant metabolite was confirmed as benzenesulfinic acid. Additional investigation revealed that the benzenesulfinic acid metabolite may be a novel, nonenzymatic, thiol-mediated reductive cleavage of an aryl sulfonamide group of SAM-760. We also determined the relative contribution of P450 to the metabolism of SAM-760 in human hepatocytes by following the rate of formation of oxidative metabolites in the presence and absence of P450 isoform–specific inhibitors. The P450-mediated oxidative metabolism of SAM-760 was still primarily attributed to CYP3A (33%), with minor contributions from P450 isoforms CYP2C19 and CYP2D6. Thus, the disposition of [14C]SAM-760 in human hepatocytes via novel sulfonamide metabolism and CYP3A verified the lower than expected clinical DDI when SAM-760 was coadministered with ketoconazole.


Journal of Medicinal Chemistry | 2017

Discovery and Characterization of (R)-6-Neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894), an Alkyne-Lacking Metabotropic Glutamate Receptor 5 Negative Allosteric Modulator Profiled in both Rat and Nonhuman Primates

Antonia F. Stepan; Michelle Marie Claffey; Matthew R. Reese; Gayatri Balan; Gabriela Barreiro; Jason Barricklow; Michael John Bohanon; Brian P. Boscoe; Gregg D. Cappon; Lois K. Chenard; Julie Cianfrogna; Laigao Chen; Karen J. Coffman; Susan E. Drozda; Joshua R. Dunetz; Somraj Ghosh; Xinjun Hou; Christopher Houle; Kapil Karki; John T. Lazzaro; Jessica Y. Mancuso; John M. Marcek; Emily L. Miller; Mark A. Moen; Steven V. O’Neil; Isao Sakurada; Marc B. Skaddan; Vinod D. Parikh; Deborah L. Smith; Patrick Trapa

We previously observed a cutaneous type IV immune response in nonhuman primates (NHP) with the mGlu5 negative allosteric modulator (NAM) 7. To determine if this adverse event was chemotype- or mechanism-based, we evaluated a distinct series of mGlu5 NAMs. Increasing the sp3 character of high-throughput screening hit 40 afforded a novel morpholinopyrimidone mGlu5 NAM series. Its prototype, (R)-6-neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894, 8), possessed favorable properties and a predicted low clinical dose (2 mg twice daily). Compound 8 did not show any evidence of immune activation in a mouse drug allergy model. Additionally, plasma samples from toxicology studies confirmed that 8 did not form any reactive metabolites. However, 8 caused the identical microscopic skin lesions in NHPs found with 7, albeit with lower severity. Holistically, this work supports the hypothesis that this unique toxicity may be mechanism-based although additional work is required to confirm this and determine clinical relevance.


Statistics in Medicine | 2001

Order-restricted dose-related trend tests.

Jessica Y. Mancuso; Hongshik Ahn; James J. Chen

Collaboration


Dive into the Jessica Y. Mancuso's collaboration.

Researchain Logo
Decentralizing Knowledge