Network


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

Hotspot


Dive into the research topics where Jamison B. Tuttle is active.

Publication


Featured researches published by Jamison B. Tuttle.


ACS Medicinal Chemistry Letters | 2012

Discovery of Brain-Penetrant, Irreversible Kynurenine Aminotransferase II Inhibitors for Schizophrenia.

Amy B. Dounay; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Michelle Marie Claffey; Artem G. Evdokimov; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Matthew Merrill Hayward; Weldon Horner; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vanessa Paradis; Vinod D. Parikh; Matthew R. Reese; Suobao Rong; Michelle A. Salafia; Katherine Schuyten; Christine A. Strick; Jamison B. Tuttle; James Valentine; Hong Wang; Laura E. Zawadzke; Patrick Robert Verhoest

Kynurenine aminotransferase (KAT) II has been identified as a potential new target for the treatment of cognitive impairment associated with schizophrenia and other psychiatric disorders. Following a high-throughput screen, cyclic hydroxamic acid PF-04859989 was identified as a potent and selective inhibitor of human and rat KAT II. An X-ray crystal structure and (13)C NMR studies of PF-04859989 bound to KAT II have demonstrated that this compound forms a covalent adduct with the enzyme cofactor, pyridoxal phosphate (PLP), in the active site. In vivo pharmacokinetic and efficacy studies in rat show that PF-04859989 is a brain-penetrant, irreversible inhibitor and is capable of reducing brain kynurenic acid by 50% at a dose of 10 mg/kg (sc). Preliminary structure-activity relationship investigations have been completed and have identified the positions on this scaffold best suited to modification for further optimization of this novel series of KAT II inhibitors.


Journal of Medicinal Chemistry | 2012

Design and Discovery of 6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a Selective Brain Penetrant PDE9A Inhibitor for the Treatment of Cognitive Disorders

Patrick Robert Verhoest; Kari R. Fonseca; Xinjun Hou; Caroline Proulx-Lafrance; Michael Corman; Christopher John Helal; Michelle Marie Claffey; Jamison B. Tuttle; Karen J. Coffman; Shenpinq Liu; Frederick R. Nelson; Robin J. Kleiman; Frank S. Menniti; Christopher J. Schmidt; Michelle Vanase-Frawley; Spiros Liras

6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943) is a novel PDE9A inhibitor identified using parallel synthetic chemistry and structure-based drug design (SBDD) and has advanced into clinical trials. Selectivity for PDE9A over other PDE family members was achieved by targeting key residue differences between the PDE9A and PDE1C catalytic site. The physicochemical properties of the series were optimized to provide excellent in vitro and in vivo pharmacokinetics properties in multiple species including humans. It has been reported to elevate central cGMP levels in the brain and CSF of rodents. In addition, it exhibits procognitive activity in several rodent models and synaptic stabilization in an amyloid precursor protein (APP) transgenic mouse model. Recent disclosures from clinical trials confirm that it is well tolerated in humans and elevates cGMP in cerebral spinal fluid of healthy volunteers, confirming that it is a quality pharmacological tool for testing clinical hypotheses in disease states associated with impairment of cGMP signaling or cognition.


Journal of Medicinal Chemistry | 2015

Challenges and Opportunities in the Discovery of New Therapeutics Targeting the Kynurenine Pathway

Amy B. Dounay; Jamison B. Tuttle; Patrick Robert Verhoest

The kynurenine pathway is responsible for the metabolism of more than 95% of dietary tryptophan (TRP) and produces numerous bioactive metabolites. Recent studies have focused on three enzymes in this pathway: indoleamine dioxygenase (IDO1), kynurenine monooxygenase (KMO), and kynurenine aminotransferase II (KAT II). IDO1 inhibitors are currently in clinical trials for the treatment of cancer, and these agents may also have therapeutic utility in neurological disorders, including multiple sclerosis. KMO inhibitors are being investigated as potential treatments for neurodegenerative diseases, such as Huntingtons and Alzheimers diseases. KAT II inhibitors have been proposed in new therapeutic approaches toward psychiatric and cognitive disorders, including cognitive impairment associated with schizophrenia. Numerous medicinal chemistry studies are currently aimed at the design of novel, potent, and selective inhibitors for each of these enzymes. The emerging opportunities and significant challenges associated with pharmacological modulation of these enzymes will be explored in this review.


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.


Journal of Organic Chemistry | 2011

A general strategy for the synthesis of cyclic N-aryl hydroxamic acids via partial nitro group reduction.

Laura A. McAllister; Bruce M. Bechle; Amy B. Dounay; Edelweiss Evrard; Xinmin Gan; Somraj Ghosh; Ji-Young Kim; Vinod D. Parikh; Jamison B. Tuttle; Patrick Robert Verhoest

We describe a generalized approach to stereocontrolled synthesis of substituted cyclic hydroxamic acids (3-amino-1-hydroxy-3,4-dihydroquinolinones) by selective reduction of substituted 2-nitrophenylalanine substrates. Compounds in this series have antibacterial properties and have also recently been reported as KAT II inhibitors. The key nitrophenyl alanine intermediates are prepared enantioselectively in excellent yield by phase transfer catalyzed alkylation of the corresponding nitrobenzyl bromides. The scope and limitations of the reductive cyclization transformation have been explored with attention to the effects of substitution pattern and electronics on reaction efficiency and byproduct formation. In addition, a novel activated trifluoroethyl ester cyclization strategy has been developed as an alternate approach to the most sterically demanding systems in this series.


ACS Medicinal Chemistry Letters | 2013

Structure-Based Design of Irreversible Human KAT II Inhibitors: Discovery of New Potency-Enhancing Interactions

Jamison B. Tuttle; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Cheng Chang; Amy B. Dounay; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Weldon Horner; Larry C. James; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vinod D. Parikh; Brian Rago; Michelle A. Salafia; Christine A. Strick; Laura E. Zawadzke; Patrick Robert Verhoest

A series of aryl hydroxamates recently have been disclosed as irreversible inhibitors of kynurenine amino transferase II (KAT II), an enzyme that may play a role in schizophrenia and other psychiatric and neurological disorders. The utilization of structure-activity relationships (SAR) in conjunction with X-ray crystallography led to the discovery of hydroxamate 4, a disubstituted analogue that has a significant potency enhancement due to a novel interaction with KAT II. The use of k inact/K i to assess potency was critical for understanding the SAR in this series and for identifying compounds with improved pharmacodynamic profiles.


Bioorganic & Medicinal Chemistry Letters | 2013

PF-04859989 as a template for structure-based drug design: Identification of new pyrazole series of irreversible KAT II inhibitors with improved lipophilic efficiency

Amy B. Dounay; Marie Anderson; Bruce M. Bechle; Edelweiss Evrard; Xinmin Gan; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Suobao Rong; Michelle A. Salafia; Jamison B. Tuttle; Laura E. Zawadzke; Patrick Robert Verhoest

The structure-based design, synthesis, and biological evaluation of a new pyrazole series of irreversible KAT II inhibitors are described herein. The modification of the inhibitor scaffold of 1 and 2 from a dihydroquinolinone core to a tetrahydropyrazolopyridinone core led to discovery of a new series of potent KAT II inhibitors with excellent physicochemical properties. Compound 20 is the most potent and lipophilically efficient of these new pyrazole analogs, with a k(inact)/K(i) value of 112,000 M(-1)s(-1) and lipophilic efficiency (LipE) of 8.53. The X-ray crystal structure of 20 with KAT II demonstrates key features that contribute to this remarkable potency and binding efficiency.


MedChemComm | 2013

Discovery of hydroxamate bioisosteres as KAT II inhibitors with improved oral bioavailability and pharmacokinetics

Jaclyn Louise Henderson; Aarti Sawant-Basak; Jamison B. Tuttle; Amy B. Dounay; Laura A. McAllister; Jayvardhan Pandit; Suobao Rong; Xinjun Hou; Bruce M. Bechle; Ji-Young Kim; Vinod D. Parikh; Somraj Ghosh; Edelweiss Evrard; Laura E. Zawadzke; Michelle A. Salafia; Brian Rago; Obach Rs; Alan J Clark; Kari R. Fonseca; Cheng Chang; Patrick Robert Verhoest

A series of kynurenine aminotransferase II (KAT II) inhibitors has been developed replacing the hydroxamate motif with a bioisostere. Triazolinones or triazoles have proven to be effective replacements with significantly improved pharmacokinetics including reduced clearance and increased bioavailability. An X-ray crystal structure of an inhibitor bound in KAT II confirms that the irreversible binding to the co-factor is maintained and that the heterocycles make productive hydrogen bonds to the arginine-399.


Molecular Pharmacology | 2018

Quantitative Translational Analysis of Brain Kynurenic Acid Modulation via Irreversible Kynurenine Aminotransferase II Inhibition

Cheng Chang; Kari R. Fonseca; Cheryl Li; Weldon Horner; Laura E. Zawadzke; Michelle A. Salafia; Kathryn Welch; Christine A. Strick; Brian M. Campbell; Steve S. Gernhardt; Haojing Rong; Aarti Sawant-Basak; Jennifer Liras; Amy B. Dounay; Jamison B. Tuttle; Patrick Robert Verhoest; Tristan S. Maurer

Kynurenic acid (KYNA) plays a significant role in maintaining normal brain function, and abnormalities in KYNA levels have been associated with various central nervous system disorders. Confirmation of its causality in human diseases requires safe and effective modulation of central KYNA levels in the clinic. The kynurenine aminotransferases (KAT) II enzyme represents an attractive target for pharmacologic modulation of central KYNA levels; however, KAT II and KYNA turnover kinetics, which could contribute to the duration of pharmacologic effect, have not been reported. In this study, the kinetics of central KYNA-lowering effect in rats and nonhuman primates (NHPs, Cynomolgus macaques) was investigated using multiple KAT II irreversible inhibitors as pharmacologic probes. Mechanistic pharmacokinetic-pharmacodynamic analysis of in vivo responses to irreversible inhibition quantitatively revealed that 1) KAT II turnover is relatively slow [16–76 hours’ half-life (t1/2)], whereas KYNA is cleared more rapidly from the brain (<1 hour t1/2) in both rats and NHPs, 2) KAT II turnover is slower in NHPs than in rats (76 hours vs. 16 hours t1/2, respectively), and 3) the percent contribution of KAT II to KYNA formation is constant (∼80%) across rats and NHPs. Additionally, modeling results enabled establishment of in vitro-in vivo correlation for both enzyme turnover rates and drug potencies. In summary, quantitative translational analysis confirmed the feasibility of central KYNA modulation in humans. Model-based analysis, where system-specific properties and drug-specific properties are mechanistically separated from in vivo responses, enabled quantitative understanding of the KAT II-KYNA pathway, as well as assisted development of promising candidates to test KYNA hypothesis in humans.


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.

Collaboration


Dive into the Jamison B. Tuttle's collaboration.

Researchain Logo
Decentralizing Knowledge