Network


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

Hotspot


Dive into the research topics where Brittney S. Bates is active.

Publication


Featured researches published by Brittney S. Bates.


Drug Metabolism and Disposition | 2012

The Role of Aldehyde Oxidase and Xanthine Oxidase in the Biotransformation of a Novel Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 5

Ryan D. Morrison; Blobaum Al; Frank W. Byers; Santomango Ts; Thomas M. Bridges; Stec D; Brewer Ka; Sanchez-Ponce R; Corlew Mm; Rush R; Andrew S. Felts; Jason Manka; Brittney S. Bates; Daryl F. Venable; Alice L. Rodriguez; Carrie K. Jones; Colleen M. Niswender; P.J. Conn; Craig W. Lindsley; Kyle A. Emmitte; Daniels Js

Negative allosteric modulation (NAM) of metabotropic glutamate receptor subtype 5 (mGlu5) represents a therapeutic strategy for the treatment of childhood developmental disorders, such as fragile X syndrome and autism. VU0409106 emerged as a lead compound within a biaryl ether series, displaying potent and selective inhibition of mGlu5. Despite its high clearance and short half-life, VU0409106 demonstrated efficacy in rodent models of anxiety after extravascular administration. However, lack of a consistent correlation in rat between in vitro hepatic clearance and in vivo plasma clearance for the biaryl ether series prompted an investigation into the biotransformation of VU0409106 using hepatic subcellular fractions. An in vitro appraisal in rat, monkey, and human liver S9 fractions indicated that the principal pathway was NADPH-independent oxidation to metabolite M1 (+16 Da). Both raloxifene (aldehyde oxidase inhibitor) and allopurinol (xanthine oxidase inhibitor) attenuated the formation of M1, thus implicating the contribution of both molybdenum hydroxylases in the biotransformation of VU0409106. The use of 18O-labeled water in the S9 experiments confirmed the hydroxylase mechanism proposed, because 18O was incorporated into M1 (+18 Da) as well as in a secondary metabolite (M2; +36 Da), the formation of which was exclusively xanthine oxidase-mediated. This unusual dual and sequential hydroxylase metabolism was confirmed in liver S9 and hepatocytes of multiple species and correlated with in vivo data because M1 and M2 were the principal metabolites detected in rats administered VU0409106. An in vitro-in vivo correlation of predicted hepatic and plasma clearance was subsequently established for VU0409106 in rats and nonhuman primates.


ChemMedChem | 2012

Discovery of 2‐(2‐Benzoxazoyl amino)‐4‐Aryl‐5‐Cyanopyrimidine as Negative Allosteric Modulators (NAMs) of Metabotropic Glutamate Receptor 5 (mGlu5): From an Artificial Neural Network Virtual Screen to an In Vivo Tool Compound

Ralf Mueller; Eric S. Dawson; Jens Meiler; Alice L. Rodriguez; Brian A. Chauder; Brittney S. Bates; Andrew S. Felts; Jeffrey P. Lamb; Usha N. Menon; Sataywan B. Jadhav; Alexander S. Kane; Carrie K. Jones; Karen J. Gregory; Colleen M. Niswender; P. Jeffrey Conn; Christopher M. Olsen; Danny G. Winder; Kyle A. Emmitte; Craig W. Lindsley

Glutamate, the major excitatory neurotransmitter, functions in the brain via activation of ligand gated cation channels and also the eight subtypes of Class C G protein-coupled metabotropic glutamate receptors (mGlus).[1] Selective allosteric modulation of mGlu5 has been shown to have potential for treatment of a variety of neurological disorders[2,3] including anxiety disorders[4,5], Parkinson’s disease[6–8], Fragile X syndrome[9] and schizophrenia.[10–14] The majority of mGlu5 negative allosteric modulators (NAMs) developed to date either contain an alkyne moiety 1–4 or employ the alkyne topology as basis for ligand design,[15] as in 5–8 (Figure 1). Only recently have mGlu5 NAM chemotypes been identified, through high-throughput screening (HTS) campaigns, that are structurally unrelated to the classical acetylenic derivatives, such as 9–12 (Figure 1).[16] Due to the prevalence of ‘molecular switch‘phenomenon in MPEP related scaffolds, our interest focused on the discovery and development of novel mGlu5 NAM chemotypes, by both HTS and Artificial Neural Network (ANN) virtual screens.


ACS Chemical Neuroscience | 2014

Identification of specific ligand-receptor interactions that govern binding and cooperativity of diverse modulators to a common metabotropic glutamate receptor 5 allosteric site

Karen J. Gregory; Elizabeth Dong Nguyen; Chrysa Malosh; Jeffrey L. Mendenhall; Jessica Z. Zic; Brittney S. Bates; Meredith J. Noetzel; Emma F. Squire; Eric M. Turner; Jerri M. Rook; Kyle A. Emmitte; Shaun R. Stauffer; Craig W. Lindsley; Jens Meiler; P. Jeffrey Conn

A common metabotropic glutamate receptor 5 (mGlu5) allosteric site is known to accommodate diverse chemotypes. However, the structural relationship between compounds from different scaffolds and mGlu5 is not well understood. In an effort to better understand the molecular determinants that govern allosteric modulator interactions with mGlu5, we employed a combination of site-directed mutagenesis and computational modeling. With few exceptions, six residues (P654, Y658, T780, W784, S808, and A809) were identified as key affinity determinants across all seven allosteric modulator scaffolds. To improve our interpretation of how diverse allosteric modulators occupy the common allosteric site, we sampled the wealth of mGlu5 structure-activity relationship (SAR) data available by docking 60 ligands (actives and inactives) representing seven chemical scaffolds into our mGlu5 comparative model. To spatially and chemically compare binding modes of ligands from diverse scaffolds, the ChargeRMSD measure was developed. We found a common binding mode for the modulators that placed the long axes of the ligands parallel to the transmembrane helices 3 and 7. W784 in TM6 not only was identified as a key NAM cooperativity determinant across multiple scaffolds, but also caused a NAM to PAM switch for two different scaffolds. Moreover, a single point mutation in TM5, G747V, altered the architecture of the common allosteric site such that 4-nitro-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (VU29) was noncompetitive with the common allosteric site. Our findings highlight the subtleties of allosteric modulator binding to mGlu5 and demonstrate the utility in incorporating SAR information to strengthen the interpretation and analyses of docking and mutational data.


Journal of Pharmacology and Experimental Therapeutics | 2015

VU0477573: Partial Negative Allosteric Modulator of the Subtype 5 Metabotropic Glutamate Receptor with In Vivo Efficacy

Hilary Highfield Nickols; Joannes P. Yuh; Karen J. Gregory; Ryan D. Morrison; Brittney S. Bates; Shaun R. Stauffer; Kyle A. Emmitte; Michael Bubser; Weimin Peng; Michael T. Nedelcovych; Analisa D. Thompson; Xiaohui Lv; Zixiu Xiang; J. Scott Daniels; Colleen M. Niswender; Craig W. Lindsley; Carrie K. Jones; P. Jeffrey Conn

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor subtype 5 (mGlu5) have potential applications in the treatment of fragile X syndrome, levodopa-induced dyskinesia in Parkinson disease, Alzheimer disease, addiction, and anxiety; however, clinical and preclinical studies raise concerns that complete blockade of mGlu5 and inverse agonist activity of current mGlu5 NAMs contribute to adverse effects that limit the therapeutic use of these compounds. We report the discovery and characterization of a novel mGlu5 NAM, N,N-diethyl-5-((3-fluorophenyl)ethynyl)picolinamide (VU0477573) that binds to the same allosteric site as the prototypical mGlu5 NAM MPEP but displays weak negative cooperativity. Because of this weak cooperativity, VU0477573 acts as a “partial NAM” so that full occupancy of the MPEP site does not completely inhibit maximal effects of mGlu5 agonists on intracellular calcium mobilization, inositol phosphate (IP) accumulation, or inhibition of synaptic transmission at the hippocampal Schaffer collateral-CA1 synapse. Unlike previous mGlu5 NAMs, VU0477573 displays no inverse agonist activity assessed using measures of effects on basal [3H]inositol phosphate (IP) accumulation. VU0477573 acts as a full NAM when measuring effects on mGlu5-mediated extracellular signal-related kinases 1/2 phosphorylation, which may indicate functional bias. VU0477573 exhibits an excellent pharmacokinetic profile and good brain penetration in rodents and provides dose-dependent full mGlu5 occupancy in the central nervous system (CNS) with systemic administration. Interestingly, VU0477573 shows robust efficacy, comparable to the mGlu5 NAM MTEP, in models of anxiolytic activity at doses that provide full CNS occupancy of mGlu5 and demonstrate an excellent CNS occupancy-efficacy relationship. VU0477573 provides an exciting new tool to investigate the efficacy of partial NAMs in animal models.


European Journal of Organic Chemistry | 2013

Towards the Total Synthesis of Marineosin A: Construction of the Macrocyclic Pyrrole and an Advanced, Functionalized Spiroaminal Model.

Leslie N. Aldrich; Cynthia B. Berry; Brittney S. Bates; Leah C. Konkol; Miranda So; Craig W. Lindsley

Herein, we describe the enantioselective construction of the 12-membered macrocyclic pyrrole core 4 of marineosin A in 5.1% overall yield from (S)-propylene oxide. The route features a key Stetter reaction to install a 1,4-diketone, which is then subjected to Paal-Knorr pyrrole synthesis and ring closing metathesis (RCM) to afford macrocycle 4. A divergence point in the synthetic scheme also enabled access to a highly functionalized spiroaminal model system 8 via an acid-mediated hydroxyketoamide cyclization strategy.


European Journal of Organic Chemistry | 2013

Towards the total synthesis of marineosin A

Leslie N. Aldrich; Cynthia B. Berry; Brittney S. Bates; Leah C. Konkol; Miranda So; Craig W. Lindsley

Herein, we describe the enantioselective construction of the 12-membered macrocyclic pyrrole core 4 of marineosin A in 5.1% overall yield from (S)-propylene oxide. The route features a key Stetter reaction to install a 1,4-diketone, which is then subjected to Paal-Knorr pyrrole synthesis and ring closing metathesis (RCM) to afford macrocycle 4. A divergence point in the synthetic scheme also enabled access to a highly functionalized spiroaminal model system 8 via an acid-mediated hydroxyketoamide cyclization strategy.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of VU0409106: A negative allosteric modulator of mGlu5 with activity in a mouse model of anxiety.

Brittney S. Bates; Alice L. Rodriguez; Andrew S. Felts; Ryan D. Morrison; Daryl F. Venable; Anna L. Blobaum; Frank W. Byers; Kera P. Lawson; J. Scott Daniels; Colleen M. Niswender; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley; Kyle A. Emmitte

Development of SAR in an aryl ether series of mGlu5 NAMs leading to the identification of tool compound VU0409106 is described in this Letter. VU0409106 is a potent and selective negative allosteric modulator of mGlu5 that binds at the known allosteric binding site and demonstrates good CNS exposure following intraperitoneal dosing in mice. VU0409106 also proved efficacious in a mouse marble burying model of anxiety, an assay known to be sensitive to mGlu5 antagonists as well as clinically efficacious anxiolytics.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of a New Molecular Probe ML228: An Activator of the Hypoxia Inducible Factor (HIF) Pathway

Jimmy R. Theriault; Andrew S. Felts; Brittney S. Bates; Jose R. Perez; Michelle Palmer; Shawn Gilbert; Eric S. Dawson; Julie L. Engers; Craig W. Lindsley; Kyle A. Emmitte

Hypoxia and ischemia are linked to several serious public health problems that affect most major organ systems. Specific examples include diseases of the cardiovascular, pulmonary, renal, neurologic, and musculoskeletal systems. The most significant pathway for cellular response to hypoxia is the hypoxia inducible factor (HIF) pathway. HIFs are transcription factors responsible for the activation of genes which encode proteins that mediate adaptive responses to reduced oxygen availability. A high-throughput cell-based HIF-mediated gene reporter screen was carried out using the NIHs Molecular Libraries Small Molecule Repository to identify activators of the HIF pathway. This communication describes the subsequent medicinal chemistry optimization of a triazine scaffold that led to the identification of the new molecular probe ML228. A discussion of HIF activation SAR within this chemotype as well as detailed in vitro characterization of the probe molecule is presented here.


Journal of Medicinal Chemistry | 2017

Discovery of N-(5-Fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (VU0424238): A Novel Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 5 Selected for Clinical Evaluation

Andrew S. Felts; Alice L. Rodriguez; Anna L. Blobaum; Ryan D. Morrison; Brittney S. Bates; Analisa Thompson Gray; Jerri M. Rook; M. N. Tantawy; Frank W. Byers; Sichen Chang; Daryl F. Venable; Vincent B. Luscombe; Gilles Tamagnan; Colleen M. Niswender; J. Scott Daniels; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley; Kyle A. Emmitte

Preclinical evidence in support of the potential utility of mGlu5 NAMs for the treatment of a variety of psychiatric and neurodegenerative disorders is extensive, and multiple such molecules have entered clinical trials. Despite some promising results from clinical studies, no small molecule mGlu5 NAM has yet to reach market. Here we present the discovery and evaluation of N-(5-fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (27, VU0424238), a compound selected for clinical evaluation. Compound 27 is more than 900-fold selective for mGlu5 versus the other mGlu receptors, and binding studies established a Ki value of 4.4 nM at a known allosteric binding site. Compound 27 had a clearance of 19.3 and 15.5 mL/min/kg in rats and cynomolgus monkeys, respectively. Imaging studies using a known mGlu5 PET ligand demonstrated 50% receptor occupancy at an oral dose of 0.8 mg/kg in rats and an intravenous dose of 0.06 mg/kg in baboons.


ChemMedChem | 2016

Identification and Optimization of Anthranilic Acid Based Inhibitors of Replication Protein A.

James D. Patrone; Nicholas F. Pelz; Brittney S. Bates; Elaine M. Souza-Fagundes; Bhavatarini Vangamudi; DeMarco V. Camper; Alexey G. Kuznetsov; Carrie F. Browning; Michael D. Feldkamp; Andreas O. Frank; Benjamin A. Gilston; Edward T. Olejniczak; Olivia W. Rossanese; Alex G. Waterson; Walter J. Chazin; Stephen W. Fesik

Replication protein A (RPA) is an essential single‐stranded DNA (ssDNA)‐binding protein that initiates the DNA damage response pathway through protein–protein interactions (PPIs) mediated by its 70N domain. The identification and use of chemical probes that can specifically disrupt these interactions is important for validating RPA as a cancer target. A high‐throughput screen (HTS) to identify new chemical entities was conducted, and 90 hit compounds were identified. From these initial hits, an anthranilic acid based series was optimized by using a structure‐guided iterative medicinal chemistry approach to yield a cell‐penetrant compound that binds to RPA70N with an affinity of 812 nm. This compound, 2‐(3‐ (N‐(3,4‐dichlorophenyl)sulfamoyl)‐4‐methylbenzamido)benzoic acid (20 c), is capable of inhibiting PPIs mediated by this domain.

Collaboration


Dive into the Brittney S. Bates'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
Top Co-Authors

Avatar

Shawn Gilbert

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Stuart L Schreiber

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Craig W. Lindsley

Office of Technology Transfer

View shared research outputs
Researchain Logo
Decentralizing Knowledge