Network


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

Hotspot


Dive into the research topics where Gregory S. Walker is active.

Publication


Featured researches published by Gregory S. Walker.


Journal of Medicinal Chemistry | 2014

Chemical and computational methods for the characterization of covalent reactive groups for the prospective design of irreversible inhibitors.

Mark Edward Flanagan; Joseph A. Abramite; Dennis P. Anderson; Ann Aulabaugh; Upendra P. Dahal; Adam M. Gilbert; Chao Li; Justin Ian Montgomery; Stacey R. Oppenheimer; Tim Ryder; Brandon P. Schuff; Daniel P. Uccello; Gregory S. Walker; Yan Wu; Matthew Frank Brown; Jinshan M. Chen; Matthew Merrill Hayward; Mark C. Noe; R. Scott Obach; Laurence Philippe; Veerabahu Shanmugasundaram; Michael J. Shapiro; Jeremy T. Starr; Justin G. Stroh; Ye Che

Interest in drugs that covalently modify their target is driven by the desire for enhanced efficacy that can result from the silencing of enzymatic activity until protein resynthesis can occur, along with the potential for increased selectivity by targeting uniquely positioned nucleophilic residues in the protein. However, covalent approaches carry additional risk for toxicities or hypersensitivity reactions that can result from covalent modification of unintended targets. Here we describe methods for measuring the reactivity of covalent reactive groups (CRGs) with a biologically relevant nucleophile, glutathione (GSH), along with kinetic data for a broad array of electrophiles. We also describe a computational method for predicting electrophilic reactivity, which taken together can be applied to the prospective design of thiol-reactive covalent inhibitors.


ACS Medicinal Chemistry Letters | 2011

Discovery of PF-04620110, a Potent, Selective, and Orally Bioavailable Inhibitor of DGAT-1.

Robert L. Dow; Jian-Cheng Li; Michael P. Pence; E. Michael Gibbs; Jennifer L. LaPerle; John Litchfield; David W. Piotrowski; Michael John Munchhof; Tara B. Manion; William J. Zavadoski; Gregory S. Walker; R. Kirk McPherson; Susan Tapley; Eliot Sugarman; Angel Guzman-Perez; Paul DaSilva-Jardine

Acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) catalyzes the final committed step in the biosynthesis of triglycerides. DGAT-1 knockout mice have been shown to be resistant to diet-induced obesity and have increased insulin sensitivity. Thus, inhibition of DGAT-1 may represent an attractive target for the treatment of obesity or type II diabetes. Herein, we report the discovery and characterization of a potent and selective DGAT-1 inhibitor PF-04620110 (3). Compound 3 inhibits DGAT-1 with an IC50 of 19 nM and shows high selectivity versus a broad panel of off-target pharmacologic end points. In vivo DGAT-1 inhibition has been demonstrated through reduction of plasma triglyceride levels in rodents at doses of ≥0.1 mg/kg following a lipid challenge. On the basis of this pharmacologic and pharmacokinetic profile, compound 3 has been advanced to human clinical studies.


Drug Metabolism and Disposition | 2014

The Pharmacokinetics, Metabolism, and Clearance Mechanisms of Tofacitinib, a Janus Kinase Inhibitor, in Humans

Martin E. Dowty; Jinyan Lin; Tim Ryder; Weiwei Wang; Gregory S. Walker; Alfin D. N. Vaz; Gary L. Chan; Sriram Krishnaswami; Chandra Prakash

Tofacitinib is a novel, oral Janus kinase inhibitor. The objectives of this study were to summarize the pharmacokinetics and metabolism of tofacitinib in humans, including clearance mechanisms. Following administration of a single 50-mg 14C-labeled tofacitinib dose to healthy male subjects, the mean (standard deviation) total percentage of administered radioactive dose recovered was 93.9% (±3.6), with 80.1% (±3.6) in the urine (28.8% parent), and 13.8% (±1.9) in feces (0.9% parent). Tofacitinib was rapidly absorbed, with plasma concentrations and total radioactivity peaking at around 1 hour after oral administration. The mean terminal phase half-life was approximately 3.2 hours for both parent drug and total radioactivity. Most (69.4%) circulating radioactivity in plasma was parent drug, with all metabolites representing less than 10% each of total circulating radioactivity. Hepatic clearance made up around 70% of total clearance, while renal clearance made up the remaining 30%. The predominant metabolic pathways of tofacitinib included oxidation of the pyrrolopyrimidine and piperidine rings, oxidation of the piperidine ring side-chain, N-demethylation and glucuronidation. Cytochrome P450 (P450) profiling indicated that tofacitinib was mainly metabolized by CYP3A4, with a smaller contribution from CYP2C19. This pharmacokinetic characterization of tofacitinib has been consistent with its clinical experience in drug-drug interaction studies.


Drug Metabolism and Disposition | 2012

Identifying a Selective Substrate and Inhibitor Pair for the Evaluation of CYP2J2 Activity

Caroline A. Lee; Jeffrey P. Jones; Jonathan Katayama; Rüdiger Kaspera; Ying Jiang; Sascha Freiwald; Evan Smith; Gregory S. Walker; Rheem A. Totah

CYP2J2, an arachidonic acid epoxygenase, is recognized for its role in the first-pass metabolism of astemizole and ebastine. To fully assess the role of CYP2J2 in drug metabolism, a selective substrate and potent specific chemical inhibitor are essential. In this study, we report amiodarone 4-hydoxylation as a specific CYP2J2-catalyzed reaction with no CYP3A4, or other drug-metabolizing enzyme, involvement. Amiodarone 4-hydroxylation enabled the determination of liver relative activity factor and intersystem extrapolation factor for CYP2J2. Amiodarone 4-hydroxylation correlated with astemizole O-demethylation but not with CYP2J2 protein content in a sample of human liver microsomes. To identify a specific CYP2J2 inhibitor, 138 drugs were screened using terfenadine and astemizole as probe substrates with recombinant CYP2J2. Forty-two drugs inhibited CYP2J2 activity by ≥50% at 30 μM, but inhibition was substrate-dependent. Of these, danazol was a potent inhibitor of both hydroxylation of terfenadine (IC50 = 77 nM) and O-demethylation of astemizole (Ki = 20 nM), and inhibition was mostly competitive. Danazol inhibited CYP2C9, CYP2C8, and CYP2D6 with IC50 values of 1.44, 1.95, and 2.74 μM, respectively. Amiodarone or astemizole were included in a seven-probe cocktail for cytochrome P450 (P450) drug-interaction screening potential, and astemizole demonstrated a better profile because it did not appreciably interact with other P450 probes. Thus, danazol, amiodarone, and astemizole will facilitate the ability to determine the metabolic role of CYP2J2 in hepatic and extrahepatic tissues.


Drug Metabolism and Disposition | 2011

Validation of isolated metabolites from drug metabolism studies as analytical standards by quantitative NMR.

Gregory S. Walker; Tim Ryder; Raman Sharma; Evan Smith; Amy Freund

In discovery and development, having a qualified metabolite standard is advantageous. Chemical synthesis of metabolite standards is often difficult and expensive. As an alternative, biological generation and isolation of metabolites in the nanomole range are readily feasible. However, without an accurately defined concentration, these isolates have limited utility as standards. There is a significant history of NMR as both a qualitative and a quantitative technique, and these concepts have been merged recently to provide both structural and quantitative information on biologically generated isolates from drug metabolism studies. Previous methodologies relied on either specialized equipment or the use of an internal standard to the isolate. We have developed a technique in which a mathematically generated signal can be inserted into a spectrum postacquisition and used as a quantitative reference: artificial signal insertion for calculation of concentration observed (aSICCO). This technique has several advantages over previous methodologies. Any region in the analyte spectra, free from interference, can be chosen for the reference signal. In addition, the magnitude of the inserted signal can be modified to appropriately match the intensity of the sample resonances. Because this is postacquisition quantification, no special equipment or pulse sequence is needed. Compared with quantitation via the addition of an internal standard (10 mM maleic acid), the signal insertion method produced similar results. For each method, precision and accuracy were within ±5%, stability of signal response over 8 days was ±5%, and the dynamic range was more than 3 orders of magnitude: 10 to 0.01 mM.


Drug Metabolism and Disposition | 2012

Selective Mechanism-Based Inactivation of CYP3A4 by CYP3cide (PF-04981517) and Its Utility as an In Vitro Tool for Delineating the Relative Roles of CYP3A4 versus CYP3A5 in the Metabolism of Drugs

Robert L. Walsky; Obach Rs; Ruth Hyland; Ping Kang; Sue Zhou; Mike West; Kieran F Geoghegan; Christopher J Helal; Gregory S. Walker; Theunis C. Goosen; Michael Zientek

CYP3cide (PF-4981517; 1-methyl-3-[1-methyl-5-(4-methylphenyl)-1H-pyrazol-4-yl]-4-[(3S)-3-piperidin-1-ylpyrrolidin-1-yl]-1H-pyrazolo[3,4-d]pyrimidine) is a potent, efficient, and specific time-dependent inactivator of human CYP3A4. When investigating its inhibitory properties, an extreme metabolic inactivation efficiency (kinact/KI) of 3300 to 3800 ml · min−1 · μmol−1 was observed using human liver microsomes from donors of nonfunctioning CYP3A5 (CYP3A5 *3/*3). This observed efficiency equated to an apparent KI between 420 and 480 nM with a maximal inactivation rate (kinact) equal to 1.6 min−1. Similar results were achieved with testosterone, another CYP3A substrate, and other sources of the CYP3A4 enzyme. To further illustrate the abilities of CYP3cide, its partition ratio of inactivation was determined with recombinant CYP3A4. These studies produced a partition ratio approaching unity, thus underscoring the inactivation capacity of CYP3cide. When CYP3cide was tested at a concentration and preincubation time to completely inhibit CYP3A4 in a library of genotyped polymorphic CYP3A5 microsomes, the correlation of the remaining midazolam 1′-hydroxylase activity to CYP3A5 abundance was significant (R2 value equal to 0.51, p value of <0.0001). The work presented here supports these findings by fully characterizing the inhibitory properties and exploring CYP3cides mechanism of action. To aid the researcher, multiple commercially available sources of CYP3cide were established, and a protocol was developed to quantitatively determine CYP3A4 contribution to the metabolism of an investigational compound. Through the establishment of this protocol and the evidence provided here, we believe that CYP3cide is a very useful tool for understanding the relative roles of CYP3A4 versus CYP3A5 and the impact of CYP3A5 genetic polymorphism on a compounds pharmacokinetics.


Drug Metabolism and Disposition | 2014

Biosynthesis of Drug Metabolites and Quantitation Using NMR Spectroscopy for Use in Pharmacologic and Drug Metabolism Studies

Gregory S. Walker; Jonathan N. Bauman; Tim Ryder; Evan B. Smith; Douglas K. Spracklin; Obach Rs

The contribution of drug metabolites to the pharmacologic and toxicologic activity of a drug can be important; however, for a variety of reasons metabolites can frequently be difficult to synthesize. To meet the need of having samples of drug metabolites for further study, we have developed biosynthetic methods coupled with quantitative NMR spectroscopy (qNMR) to generate solutions of metabolites of known structure and concentration. These quantitative samples can be used in a variety of ways when a synthetic sample is unavailable, including pharmacologic assays, standards for in vitro work to help establish clearance pathways, and/or as analytical standards for bioanalytical work to ascertain exposure, among others. We illustrate five examples of metabolite biosynthesis and qNMR. The types of metabolites include one glucuronide and four oxidative products. Concentrations of the isolated metabolite stock solutions ranged from 0.048 to 8.3 mM, with volumes from approximately 0.04 to 0.150 ml in hexadeutarated dimethylsulfoxide. These specific quantified isolates were used as standards in the drug discovery setting as substrates in pharmacology assays, for bioanalytical assays to establish exposure, and in variety of routine absorption, distribution, metabolism, and excretion assays, such as protein binding and determining blood-to-plasma ratios. The methods used to generate these materials are described in detail with the objective that these methods can be generally used for metabolite biosynthesis and isolation.


Expert Opinion on Drug Metabolism & Toxicology | 2008

Comparison of LC-NMR and conventional NMR for structure elucidation in drug metabolism studies.

Gregory S. Walker; Thomas N. O'Connell

Liquid chromatography-nuclear magnetic resonance (LC-NMR) has proven to be a useful technique for the structure elucidation of novel metabolites from pharmaceutical compounds. Proponents of LC-NMR tout the advantage of eliminating the step of a separate chromatographic isolation. However, the advantages of directly coupling NMR and HPLC instrumentation must be weighed against compromises in performance made to each technique to achieve a hyphenated system. While significant advances have been made in LC-NMR technology, a strong case can be made that HPLC purification of metabolites followed by conventional tube NMR is equally useful. It is relatively rare that one approach will be successful and the other not. The fundamental consideration is whether there is sufficient chromatographic expertise in the NMR laboratory to adequately design and execute appropriate experiments such that a pure chromatographic peak will be produced in the hyphenated system. Due to speed and sensitivity differences between NMR spectroscopy and mass spectrometry, liquid chromatography/mass spectrometry (LC/MS) continues to be the front-line approach for the structure elucidation of metabolites.


Magnetic Resonance in Chemistry | 1997

Long‐range two‐dimensional 1H–15N heteronuclear shift correlation at natural abundance using GHNMQC. A study of the reverse transcriptase inhibitor delavirdine

Kathleen A. Farley; Gregory S. Walker; Gary E. Martin

Delavirdine is a novel BHAP [bis(heteroaryl)piperazine] RT (reverse transcriptase) inhibitor in the final stages of development for treatment of human AIDS (acquired immuno deficiency syndrome). The assignment of the six unique nitrogen resonances in the molecule and its 5‐aminoindole precursor are reported at natural abundance using the two‐dimensional GHNMQC (gradient hydrogen–nitrogen multiple quantum coherence) experiment. The molecule contains three protonated secondary nitrogens and three tertiary nitrogens in its structure. The protonated nitrogen of the acidic sulfonamidomethyl moiety at the 5‐position of the indole substituent of the molecule is shown to be sufficiently acidic in DMSO solutions at ambient temperature to preclude the observation of a direct response for this 1H–15N heteronuclear pair; in contrast, direct responses for the indole and isopropyl amino nitrogens were readily observable under these conditions. Variable‐temperature studies in pyridine demonstrated that it was possible to slow autoprotonation sufficiently to allow the direct response to be observed with full intensity at ‐35°C. Similar, although less pronounced behavior was observed for the 5‐amino group contained in the structure of the 5‐aminoindole precursor of delavirdine. The broad proton multiplet for the H‐2′/H‐6′ methylene proton resonance of the piperazine portion of the molecule failed to afford a direct response to the piperazine N‐4′ resonance in the conventional GHNMQC experiment. Modifying the pulse sequence to apply a selective pulse to the H‐2′/H‐6′ proton in a manner analogous to that recently reported for the selective HMBC experiment readily facilitated the observation of the otherwise absent long‐range coupling to N‐4′.


Chemical Research in Toxicology | 2011

Oxidative metabolism of a quinoxaline derivative by xanthine oxidase in rodent plasma.

Raman Sharma; Heather Eng; Gregory S. Walker; Gabriela Barreiro; Antonia F. Stepan; Kim F. McClure; Angela Wolford; Paul D. Bonin; Peter Cornelius; Amit S. Kalgutkar

As part of efforts directed at the G protein-coupled receptor 119 agonist program for type 2 diabetes, a series of cyanopyridine derivatives exemplified by isopropyl-4-(3-cyano-5-(quinoxalin-6-yl)pyridine-2-yl)piperazine-1-carboxylate (1) were identified as novel chemotypes worthy of further hit-to-lead optimization. Compound 1, however, was found to be unstable in plasma (37 °C, pH 7.4) from rat (T(1/2) = 16 min), mouse (T(1/2) = 61 min), and guinea pig (T(1/2) = 4 min). Lowering the temperature of plasma incubations (4-25 °C) attenuated the degradation of 1, implicating the involvement of an enzyme-mediated process. Failure to detect any appreciable amount of 1 in plasma samples from protein binding and pharmacokinetic studies in rats was consistent with its labile nature in plasma. Instability noted in rodent plasma was not observed in plasma from dogs, monkeys, and humans (T(1/2) > 370 min at 37 °C, pH 7.4). Metabolite identification studies in rodent plasma revealed the formation of a single metabolite (M1), which was 16 Da higher than the molecular weight of 1 (compound 1, MH(+) = 403; M1, MH(+) = 419). Pretreatment of rat plasma with allopurinol, but not raloxifene, abolished the conversion of 1 to M1, suggesting that xanthine oxidase (XO) was responsible for the oxidative instability. Consistent with the known catalytic mechanism of XO, the source of oxygen incorporated in M1 was derived from water rather than molecular oxygen. The formation of M1 was also demonstrated in incubations of 1 with purified bovine XO. The structure of M1 was determined by NMR analysis to be isopropyl-4-(3-cyano-5-(3-oxo-3,4-dihydroquinoxalin-6-yl)pyridine-2-yl)piperazine-1-carboxylate. The regiochemistry of quinoxaline ring oxidation in 1 was consistent with ab initio calculations and molecular docking studies using a published crystal structure of bovine XO. A close-in analogue of 1, which lacked the quinoxaline motif (e.g., 5-(4-cyano-3-methylphenyl)-2-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperidin-1-yl)nicotinitrile (2)) was stable in rat plasma and possessed substantially improved GPR119 agonist properties. To the best of our knowledge, our studies constitute the first report on the involvement of rodent XO in oxidative drug metabolism in plasma.

Collaboration


Dive into the Gregory S. Walker's collaboration.

Researchain Logo
Decentralizing Knowledge