Network


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

Hotspot


Dive into the research topics where Barbara Tate is active.

Publication


Featured researches published by Barbara Tate.


Journal of Pharmacology and Experimental Therapeutics | 2006

Concentration-Dependent Modulation of Amyloid-β in Vivo and in Vitro Using the γ-Secretase Inhibitor, LY-450139

Thomas A. Lanz; Michael J. Karmilowicz; Kathleen M. Wood; Nikolay Pozdnyakov; Ping Du; Mary A. Piotrowski; Tracy M. Brown; Charles E. Nolan; Karl E.G. Richter; James E. Finley; Qing Fei; Charles F. Ebbinghaus; Yuhpyng L. Chen; Douglas K. Spracklin; Barbara Tate; Kieran F. Geoghegan; Lit-Fui Lau; David D. Auperin; Joel B. Schachter

LY-450139 is a γ-secretase inhibitor shown to have efficacy in multiple cellular and animal models. Paradoxically, robust elevations of plasma amyloid-β (Aβ) have been reported in dogs and humans after administration of subefficacious doses. The present study sought to further evaluate Aβ responses to LY-450139 in the guinea pig, a nontransgenic model that has an Aβ sequence identical to that of human. Male guinea pigs were treated with LY-450139 (0.2–60 mg/kg), and brain, cerebrospinal fluid, and plasma Aβ levels were characterized at 1, 3, 6, 9, and 14 h postdose. Low doses significantly elevated plasma Aβ levels at early time points, with return to baseline within hours. Higher doses inhibited Aβ levels in all compartments at early time points, but elevated plasma Aβ levels at later time points. To determine whether this phenomenon occurs under steady-state drug exposure, guinea pigs were implanted with subcutaneous minipumps delivering LY-450139 (0.3–30 mg/kg/day) for 5 days. Plasma Aβ was significantly inhibited at 10–30 mg/kg/day, but significantly elevated at 1 mg/kg/day. To further understand the mechanism of Aβ elevation by LY-450139, H4 cells overexpressing the Swedish mutant of amyloid-precursor protein and a mouse embryonic stem cell-derived neuronal cell line were studied. In both cellular models, elevated levels of secreted Aβ were observed at subefficacious concentrations, whereas dose-responsive inhibition was observed at higher concentrations. These results suggest that LY-450139 modulates the γ-secretase complex, eliciting Aβ lowering at high concentrations but Aβ elevation at low concentrations.


Journal of Pharmacology and Experimental Therapeutics | 2007

An Inhibitor of Casein Kinase Iϵ Induces Phase Delays in Circadian Rhythms under Free-Running and Entrained Conditions

Lori Badura; Terri Swanson; Wendy O. Adamowicz; Jessica Adams; Julie Cianfrogna; Katherine Fisher; Janice Holland; Robin J. Kleiman; Frederick R. Nelson; Linda Reynolds; Kristin St. Germain; Eric Schaeffer; Barbara Tate; Jeffrey Sprouse

Casein kinase Iϵ (CKIϵ) is an essential component of the biological clock, phosphorylating PER proteins, and in doing so regulating their turnover and nuclear entry in oscillator cells of the suprachiasmatic nucleus (SCN). Although hereditary decreases in PER phosphorylation have been well characterized, little is known about the consequences of acute enzyme inhibition by pharmacological means. A novel reagent, 4-[3-cyclohexyl-5-(4-fluoro-phenyl)-3H-imidazol-4-yl]-pyrimidin-2-ylamine (PF-670462), proved to be both a potent (IC50 = 7.7 ± 2.2 nM) and selective (>30-fold with respect to 42 additional kinases) inhibitor of CKIϵ in isolated enzyme preparations; in transfected whole cell assays, it caused a concentration-related redistribution of nuclear versus cytosolic PER. When tested in free-running animals, 50 mg/kg s.c. PF-670462 produced robust phase delays when dosed at circadian time (CT)9 (–1.97 ± 0.17 h). Entrained rats dosed in normal light-dark (LD) and then released to constant darkness also experienced phase delays that were dose- and time of dosing-dependent. PF-670462 yielded only phase delays across the circadian cycle with the most sensitive time at CT12 when PER levels are near their peak in the SCN. Most importantly, these drug-induced phase delays persisted in animals entrained and maintained in LD throughout the entire experiment; re-entrainment to the prevailing LD required days in contrast to the rapid elimination of the drug (t1/2 = 0.46 ± 0.04 h). Together, these results suggest that inhibition of CKIϵ yields a perturbation of oscillator function that forestalls light as a zeitgeber, and they demonstrate that pharmacological tools such as PF-670462 may yield valuable insight into clock function.


Journal of Medicinal Chemistry | 2012

Optimization of a natural product-based class of γ-secretase modulators.

Jed Hubbs; Nathan O. Fuller; Wesley F. Austin; Ruichao Shen; Steffen P. Creaser; Timothy D. McKee; Robyn Loureiro; Barbara Tate; Weiming Xia; Jeffrey L. Ives; Brian Scott Bronk

A series of triterpene-based γ-secretase modulators is optimized. An acetate present at the C24 position of the natural product was replaced with either carbamates or ethers to provide compounds with better metabolic stability. With one of those pharmacophores in place at C24, morpholines or carbamates were installed at the C3 position to refine the physicochemical properties of the analogues. This strategy gave compounds with low clearance and good distribution into the central nervous system (CNS) of CD-1 mice. Two of these compounds, 100 and 120, were tested for a pharmacodynamic effect in the strain and lowered brain Aβ42 levels.


Journal of Pharmacology and Experimental Therapeutics | 2010

Pharmacodynamics and Pharmacokinetics of the γ-Secretase Inhibitor PF-3084014

Thomas A. Lanz; Kathleen M. Wood; Karl E.G. Richter; Charles E. Nolan; Stacey L. Becker; Nikolay Pozdnyakov; Barbara-Anne Martin; Ping Du; Christine E. Oborski; Douglas E. Wood; Tracy M. Brown; James E. Finley; Sharon A. Sokolowski; Carol D. Hicks; Karen J. Coffman; Kieran F. Geoghegan; Michael Aaron Brodney; Dane Liston; Barbara Tate

PF-3084014 [(S)-2-((S)-5,7-difluoro-1,2,3,4-tetrahydronaphthalen-3-ylamino)-N-(1-(2-methyl-1-(neopentylamino)propan-2-yl)-1H-imidazol-4-yl)pentanamide] is a novel γ-secretase inhibitor that reduces amyloid-β (Aβ) production with an in vitro IC50 of 1.2 nM (whole-cell assay) to 6.2 nM (cell-free assay). This compound inhibits Notch-related T- and B-cell maturation in an in vitro thymocyte assay with an EC50 of 2.1 μM. A single acute dose showed dose-dependent reduction in brain, cerebrospinal fluid (CSF), and plasma Aβ in Tg2576 mice as measured by enzyme-linked immunosorbent assay and immunoprecipitation (IP)/mass spectrometry (MS). Guinea pigs were dosed with PF-3084014 for 5 days via osmotic minipump at 0.03 to 3 mg/kg/day and exhibited dose-dependent reduction in brain, CSF, and plasma Aβ. To further characterize Aβ dynamics in brain, CSF, and plasma in relation to drug exposure and Notch-related toxicities, guinea pigs were dosed with 0.03 to 10 mg/kg PF-3084014, and tissues were collected at regular intervals from 0.75 to 30 h after dose. Brain, CSF, and plasma all exhibited dose-dependent reductions in Aβ, and the magnitude and duration of Aβ lowering exceeded those of the reductions in B-cell endpoints. Other γ-secretase inhibitors have shown high potency at elevating Aβ in the conditioned media of whole cells and the plasma of multiple animal models and humans. Such potentiation was not observed with PF-3084014. IP/MS analysis, however, revealed dose-dependent increases in Aβ11-40 and Aβ1-43 at doses that potently inhibited Aβ1-40 and Aβ1-42. PF-3084014, like previously described γ-secretase inhibitors, preferentially reduced Aβ1-40 relative to Aβ1-42. Potency at Aβ relative to Notch-related endpoints in vitro and in vivo suggests that a therapeutic index can be achieved with this compound.


Journal of Pharmacology and Experimental Therapeutics | 2006

DOMINANCE OF AMYLOID PRECURSOR PROTEIN SEQUENCE OVER HOST CELL SECRETASES IN DETERMINING β-AMYLOID PROFILES STUDIES OF INTERSPECIES VARIATION AND DRUG ACTION BY INTERNALLY STANDARDIZED IMMUNOPRECIPITATION/MASS SPECTROMETRY

Ping Du; Kathleen M. Wood; Michele H. Rosner; David Cunningham; Barbara Tate; Kieran F. Geoghegan

β-Amyloid peptides, tentatively regarded as the principal neurotoxins responsible for Alzheimers Disease, make up a set of products that varies significantly among different biological systems. The full implications of this complexity and its variations have yet to be defined. In this work, Aβ peptide populations were extracted from animal brain tissue or cell-conditioned media, immunoprecipitated with specific antibodies, and analyzed by matrix-assisted laser desorption time-of-flight mass spectrometry. 15N-Substituted Aβ internal standards were added to gauge variations in the profile of captured peptides. Results from a range of species, including guinea pig, dog, rabbit, and wild-type and transgenic mice, showed that the Aβ peptide population in each system was mainly determined by the species of origin of the amyloid precursor protein (APP) and not by the host tissue or cell line. The same method was used to gauge the effect on the Aβ peptide profile of an inhibitor of γ-secretase, one of the two proteinases that excises Aβ peptides from the precursor protein with different effects on specific peptides. Overall, the results demonstrate that the species of origin of the APP substrate dictates the outcome of APP processing to a greater extent than the origin of the processing enzymes, an important consideration in rationalizing the properties of different model systems.


Alzheimer's Research & Therapy | 2013

Efficacy of SPI-1865, a novel gamma-secretase modulator, in multiple rodent models

Robyn Loureiro; Jo Ann Dumin; Timothy D. McKee; Wesley F. Austin; Nathan O. Fuller; Jed L. Hubbs; Ruichao Shen; Jeff Jonker; Jeff Ives; Brian Scott Bronk; Barbara Tate

IntroductionModulation of the gamma-secretase enzyme, which reduces the production of the amyloidogenic Aβ42 peptide while sparing the production of other Aβ species, is a promising therapeutic approach for the treatment of Alzheimers disease. Satori has identified a unique class of small molecule gamma-secretase modulators (GSMs) capable of decreasing Aβ42 levels in cellular and rodent model systems. The compound class exhibits potency in the nM range in vitro and is selective for lowering Aβ42 and Aβ38 while sparing Aβ40 and total Aβ levels. In vivo, a compound from the series, SPI-1865, demonstrates similar pharmacology in wild-type CD1 mice, Tg2576 mice and Sprague Dawley rats.MethodsAnimals were orally administered either a single dose of SPI-1865 or dosed for multiple days. Aβ levels were measured using a sensitive plate-based ELISA system (MSD) and brain and plasma exposure of drug were assessed by LC/MS/MS.ResultsIn wild-type mice using either dosing regimen, brain Aβ42 and Aβ38 levels were decreased upon treatment with SPI-1865 and little to no statistically meaningful effect on Aβ40 was observed, reflecting the changes observed in vitro. In rats, brain Aβ levels were examined and similar to the mouse studies, brain Aβ42 and Aβ38 were lowered. Comparable changes were also observed in the Tg2576 mice, where Aβ levels were measured in brain as well as plasma and CSF.ConclusionsTaken together, these data indicate that SPI-1865 is orally bioavailable, brain penetrant, and effective at lowering Aβ42 in a dose responsive manner. With this unique profile, the class of compounds represented by SPI-1865 may be a promising new therapy for Alzheimers disease.


ACS Medicinal Chemistry Letters | 2012

Initial Optimization of a New Series of γ-Secretase Modulators Derived from a Triterpene Glycoside

Nathan O. Fuller; Jed L. Hubbs; Wesley F. Austin; Steffen P. Creaser; Timothy D. McKee; Robyn Loureiro; Barbara Tate; Weiming Xia; Jeffrey L. Ives; Mark A. Findeis; Brian Scott Bronk

The discovery of a new series of γ-secretase modulators is disclosed. Starting from a triterpene glycoside γ-secretase modulator that gave a very low brain-to-plasma ratio, initial SAR and optimization involved replacement of a pendant sugar with a series of morpholines. This modification led to two compounds with significantly improved central nervous system (CNS) exposure.


Journal of Pharmacology and Experimental Therapeutics | 2010

Pharmacodynamics and pharmacokinetics of the gamma-secretase inhibitor PF-3084014.

Thomas A. Lanz; Kathleen M. Wood; Karl E.G. Richter; Charles E. Nolan; Stacey L. Becker; Nikolay Pozdnyakov; Barbara-Anne Martin; Ping Du; Christine E. Oborski; Douglas E. Wood; Tracy M. Brown; James E. Finley; Sharon A. Sokolowski; Carol D. Hicks; Karen J. Coffman; Kieran F. Geoghegan; Michael Aaron Brodney; Dane Liston; Barbara Tate

PF-3084014 [(S)-2-((S)-5,7-difluoro-1,2,3,4-tetrahydronaphthalen-3-ylamino)-N-(1-(2-methyl-1-(neopentylamino)propan-2-yl)-1H-imidazol-4-yl)pentanamide] is a novel γ-secretase inhibitor that reduces amyloid-β (Aβ) production with an in vitro IC50 of 1.2 nM (whole-cell assay) to 6.2 nM (cell-free assay). This compound inhibits Notch-related T- and B-cell maturation in an in vitro thymocyte assay with an EC50 of 2.1 μM. A single acute dose showed dose-dependent reduction in brain, cerebrospinal fluid (CSF), and plasma Aβ in Tg2576 mice as measured by enzyme-linked immunosorbent assay and immunoprecipitation (IP)/mass spectrometry (MS). Guinea pigs were dosed with PF-3084014 for 5 days via osmotic minipump at 0.03 to 3 mg/kg/day and exhibited dose-dependent reduction in brain, CSF, and plasma Aβ. To further characterize Aβ dynamics in brain, CSF, and plasma in relation to drug exposure and Notch-related toxicities, guinea pigs were dosed with 0.03 to 10 mg/kg PF-3084014, and tissues were collected at regular intervals from 0.75 to 30 h after dose. Brain, CSF, and plasma all exhibited dose-dependent reductions in Aβ, and the magnitude and duration of Aβ lowering exceeded those of the reductions in B-cell endpoints. Other γ-secretase inhibitors have shown high potency at elevating Aβ in the conditioned media of whole cells and the plasma of multiple animal models and humans. Such potentiation was not observed with PF-3084014. IP/MS analysis, however, revealed dose-dependent increases in Aβ11-40 and Aβ1-43 at doses that potently inhibited Aβ1-40 and Aβ1-42. PF-3084014, like previously described γ-secretase inhibitors, preferentially reduced Aβ1-40 relative to Aβ1-42. Potency at Aβ relative to Notch-related endpoints in vitro and in vivo suggests that a therapeutic index can be achieved with this compound.


International Journal of Alzheimer's Disease | 2012

Modulation of Gamma-Secretase for the Treatment of Alzheimer's Disease

Barbara Tate; Timothy D. McKee; Robyn Loureiro; Jo Ann Dumin; Weiming Xia; Kevin Pojasek; Wesley F. Austin; Nathan O. Fuller; Jed L. Hubbs; Ruichao Shen; Jeff Jonker; Jeff Ives; Brian Scott Bronk

The Amyloid Hypothesis states that the cascade of events associated with Alzheimers disease (AD)—formation of amyloid plaques, neurofibrillary tangles, synaptic loss, neurodegeneration, and cognitive decline—are triggered by Aβ peptide dysregulation (Kakuda et al., 2006, Sato et al., 2003, Qi-Takahara et al., 2005). Since γ-secretase is critical for Aβ production, many in the biopharmaceutical community focused on γ-secretase as a target for therapeutic approaches for Alzheimers disease. However, pharmacological approaches to control γ-secretase activity are challenging because the enzyme has multiple, physiologically critical protein substrates. To lower amyloidogenic Aβ peptides without affecting other γ-secretase substrates, the epsilon (ε) cleavage that is essential for the activity of many substrates must be preserved. Small molecule modulators of γ-secretase activity have been discovered that spare the ε cleavage of APP and other substrates while decreasing the production of Aβ 42. Multiple chemical classes of γ-secretase modulators have been identified which differ in the pattern of Aβ peptides produced. Ideally, modulators will allow the ε cleavage of all substrates while shifting APP cleavage from Aβ 42 and other highly amyloidogenic Aβ peptides to shorter and less neurotoxic forms of the peptides without altering the total Aβ pool. Here, we compare chemically distinct modulators for effects on APP processing and in vivo activity.


Bioorganic & Medicinal Chemistry Letters | 2011

Design, synthesis, and in vivo characterization of a novel series of tetralin amino imidazoles as γ-secretase inhibitors: Discovery of PF-3084014

Michael Aaron Brodney; David D. Auperin; Stacey L. Becker; Brian Scott Bronk; Tracy M. Brown; Karen J. Coffman; James E. Finley; Carol D. Hicks; Michael J. Karmilowicz; Thomas A. Lanz; Dane Liston; Xingrong Liu; Barbara-Anne Martin; Robert B. Nelson; Charles E. Nolan; Christine E. Oborski; Christine P. Parker; Karl E.G. Richter; Nikolay Pozdnyakov; Barbara G. Sahagan; Joel B. Schachter; Sharon A. Sokolowski; Barbara Tate; Douglas E. Wood; Kathleen M. Wood; Jeffrey Van Deusen; Lei Zhang

A novel series of tetralin containing amino imidazoles, derived from modification of the corresponding phenyl acetic acid derivatives is described. Replacement of the amide led to identification of a potent series of tetralin-amino imidazoles with robust central efficacy. The reduction of brain Aβ in guinea pigs in the absence of changes in B-cells suggested a potential therapeutic index with respect to APP processing compared with biomarkers of notch related toxicity. Optimization of the FTOC to plasma concentrations at the brain Aβ EC(50) lead to the identification of compound 14f (PF-3084014) which was selected for clinical development.

Collaboration


Dive into the Barbara Tate's collaboration.

Researchain Logo
Decentralizing Knowledge