Network


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

Hotspot


Dive into the research topics where James R. Gosset is active.

Publication


Featured researches published by James R. Gosset.


Xenobiotica | 2012

Application of PBPK modelling in drug discovery and development at Pfizer

Hannah M. Jones; Maurice Dickins; Kuresh Youdim; James R. Gosset; Neil Attkins; Tanya L. Hay; Ian Gurrell; Y. Raj Logan; Peter J. Bungay; Barry C. Jones; Iain Gardner

Early prediction of human pharmacokinetics (PK) and drug–drug interactions (DDI) in drug discovery and development allows for more informed decision making. Physiologically based pharmacokinetic (PBPK) modelling can be used to answer a number of questions throughout the process of drug discovery and development and is thus becoming a very popular tool. PBPK models provide the opportunity to integrate key input parameters from different sources to not only estimate PK parameters and plasma concentration-time profiles, but also to gain mechanistic insight into compound properties. Using examples from the literature and our own company, we have shown how PBPK techniques can be utilized through the stages of drug discovery and development to increase efficiency, reduce the need for animal studies, replace clinical trials and to increase PK understanding. Given the mechanistic nature of these models, the future use of PBPK modelling in drug discovery and development is promising, however, some limitations need to be addressed to realize its application and utility more broadly.


Expert Opinion on Drug Metabolism & Toxicology | 2013

Model-based approaches to predict drug–drug interactions associated with hepatic uptake transporters: preclinical, clinical and beyond

Hugh A. Barton; Yurong Lai; Theunis C. Goosen; Hannah M. Jones; Ayman El-Kattan; James R. Gosset; Jian Lin; Manthena V. Varma

Introduction: Membrane transporters have been recognized to play a key role in determining the absorption, distribution and elimination processes of drugs. The organic anion-transporting polypeptide (OATP)1B1 and OATP1B3 isoforms are selectively expressed in the human liver and are known to cause significant drug–drug interactions (DDIs), as observed with an increasing number of drugs. It is evident that DDIs involving hepatic transporters are capable of altering systemic, as well as tissue-specific, exposure of drug substrates resulting in marked differences in drug safety and/or efficacy. It is therefore essential to quantitatively predict such interactions early in the drug development to mitigate clinical risks. Areas covered: The role of hepatic uptake transporters in drug disposition and clinical DDIs has been reviewed with an emphasis on the current state of the models applicable for quantitative predictions. The readers will also gain insight into the in vitro experimental tools available to characterize transport kinetics, while appreciating the knowledge gaps in the in vitro–in vivo extrapolation (IVIVE), which warrant further investigation. Expert opinion: Static and dynamic models can be convincingly applied to quantitatively predict drug interactions, early in drug discovery, to mitigate clinical risks as well as to avoid unnecessary clinical studies. Compared to basic models, which focus on individual processes, mechanistic models provide the ability to assess DDI potential for compounds with systemic disposition determined by both transporters and metabolic enzymes. However, complexities in the experimental tools and an apparent disconnect in the IVIVE of transport kinetics have limited the physiologically based pharmacokinetic modeling strategies. Emerging data on the expression of transporter proteins and tissue drug concentrations are expected to help bridge these gaps. In addition, detailed characterization of substrate kinetics can facilitate building comprehensive mechanistic models.


Journal of Pharmacology and Experimental Therapeutics | 2014

Inhibition of TRPM8 channels reduces pain in the cold pressor test in humans.

Wendy J. Winchester; Katrina Gore; Sophie Glatt; Wendy Petit; Jennifer C. Gardiner; Kelly Conlon; Michael Postlethwaite; Pierre-Philippe Saintot; Sonia Roberts; James R. Gosset; Tomomi Matsuura; Mark David Andrews; Paul A. Glossop; Michael John Palmer; Nicola Clear; Susie Collins; Kevin Beaumont; David S. Reynolds

The transient receptor potential (subfamily M, member 8; TRPM8) is a nonselective cation channel localized in primary sensory neurons, and is a candidate for cold thermosensing, mediation of cold pain, and bladder overactivity. Studies with TRPM8 knockout mice and selective TRPM8 channel blockers demonstrate a lack of cold sensitivity and reduced cold pain in various rodent models. Furthermore, TRPM8 blockers significantly lower body temperature. We have identified a moderately potent (IC50 = 103 nM), selective TRPM8 antagonist, PF-05105679 [(R)-3-[(1-(4-fluorophenyl)ethyl)(quinolin-3-ylcarbonyl)amino]methylbenzoic acid]. It demonstrated activity in vivo in the guinea pig bladder ice water and menthol challenge tests with an IC50 of 200 nM and reduced core body temperature in the rat (at concentrations >1219 nM). PF-05105679 was suitable for acute administration to humans and was evaluated for effects on core body temperature and experimentally induced cold pain, using the cold pressor test. Unbound plasma concentrations greater than the IC50 were achieved with 600- and 900-mg doses. The compound displayed a significant inhibition of pain in the cold pressor test, with efficacy equivalent to oxycodone (20 mg) at 1.5 hours postdose. No effect on core body temperature was observed. An unexpected adverse event (hot feeling) was reported, predominantly periorally, in 23 and 36% of volunteers (600- and 900-mg dose, respectively), which in two volunteers was nontolerable. In conclusion, this study supports a role for TRPM8 in acute cold pain signaling at doses that do not cause hypothermia.


Scientific Reports | 2015

Discovery and characterization of novel inhibitors of the sodium-coupled citrate transporter (NaCT or SLC13A5)

Kim Huard; Janice A. Brown; Jessica E. C. Jones; Shawn Cabral; Kentaro Futatsugi; Matthew Gorgoglione; Adhiraj Lanba; Nicholas B. Vera; Yimin Zhu; Qingyun Yan; Yingjiang Zhou; Cecile Vernochet; Keith Riccardi; Angela Wolford; David Pirman; Mark Niosi; Gary E. Aspnes; Michael Herr; Nathan E. Genung; Thomas V. Magee; Daniel P. Uccello; Paula M. Loria; Li Di; James R. Gosset; David Hepworth; Timothy P. Rolph; Jeffrey A. Pfefferkorn; Derek M. Erion

Citrate is a key regulatory metabolic intermediate as it facilitates the integration of the glycolysis and lipid synthesis pathways. Inhibition of hepatic extracellular citrate uptake, by blocking the sodium-coupled citrate transporter (NaCT or SLC13A5), has been suggested as a potential therapeutic approach to treat metabolic disorders. NaCT transports citrate from the blood into the cell coupled to the transport of sodium ions. The studies herein report the identification and characterization of a novel small dicarboxylate molecule (compound 2) capable of selectively and potently inhibiting citrate transport through NaCT, both in vitro and in vivo. Binding and transport experiments indicate that 2 specifically binds NaCT in a competitive and stereosensitive manner, and is recognized as a substrate for transport by NaCT. The favorable pharmacokinetic properties of 2 permitted in vivo experiments to evaluate the effect of inhibiting hepatic citrate uptake on metabolic endpoints.


Journal of Medicinal Chemistry | 2015

Discovery and Optimization of Imidazopyridine-Based Inhibitors of Diacylglycerol Acyltransferase 2 (DGAT2)

Kentaro Futatsugi; Daniel W. Kung; Suvi T. M. Orr; Shawn Cabral; David Hepworth; Gary E. Aspnes; Scott Bader; Jianwei Bian; Markus Boehm; Philip A. Carpino; Steven B. Coffey; Matthew S. Dowling; Michael Herr; Wenhua Jiao; Sophie Y. Lavergne; Qifang Li; Ronald W. Clark; Derek M. Erion; Kou Kou; Kyuha Lee; Brandon Pabst; Sylvie Perez; Julie Purkal; Csilla C. Jorgensen; Theunis C. Goosen; James R. Gosset; Mark Niosi; John C. Pettersen; Jeffrey A. Pfefferkorn; Kay Ahn

The medicinal chemistry and preclinical biology of imidazopyridine-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2) is described. A screening hit 1 with low lipophilic efficiency (LipE) was optimized through two key structural modifications: (1) identification of the pyrrolidine amide group for a significant LipE improvement, and (2) insertion of a sp(3)-hybridized carbon center in the core of the molecule for simultaneous improvement of N-glucuronidation metabolic liability and off-target pharmacology. The preclinical candidate 9 (PF-06424439) demonstrated excellent ADMET properties and decreased circulating and hepatic lipids when orally administered to dyslipidemic rodent models.


Bioorganic & Medicinal Chemistry | 2012

The discovery of UK-369003, a novel PDE5 inhibitor with the potential for oral bioavailability and dose-proportional pharmacokinetics

David James Rawson; Stephen Ballard; Christopher Gordon Barber; Laura Barker; Kevin Beaumont; Mark Edward Bunnage; Susan Cole; Martin Corless; Stephen Martin Denton; David Ellis; Marion Floc’h; Laura Foster; James R. Gosset; Frances Holmwood; Charlotte Alice Louise Lane; David Leahy; John Paul Mathias; Graham Nigel Maw; William A. Million; Cedric Poinsard; Jenny Price; Rachel Russel; Stephen Derek Albert Street; Lesa Watson

This paper describes our recent efforts to design and synthesise potent and selective PDE5 inhibitors and the use of in vitro predictors of clearance, absorption and permeability to maximise the potential for dose-proportional pharmacokinetics and good oral bioavailability in man. Optimisation of the preclinical profile resulted in the identification of UK-369003 (19a) and its nomination as a clinical candidate. The clinical pharmacokinetic and safety profile has enabled us to progress the compound to test its efficacy in patients with lower urinary tract symptoms (LUTS) associated with benign prostatic hyperplasia (BPH) and a paper describing its efficacy has recently been published.


Journal of Medicinal Chemistry | 2016

Optimization of a Dicarboxylic Series for in Vivo Inhibition of Citrate Transport by the Solute Carrier 13 (SLC13) Family

Kim Huard; James R. Gosset; Justin Ian Montgomery; Adam M. Gilbert; Matthew Merrill Hayward; Thomas V. Magee; Shawn Cabral; Daniel P. Uccello; Kevin B. Bahnck; Janice A. Brown; Julie Purkal; Matthew Gorgoglione; Adhiraj Lanba; Kentaro Futatsugi; Michael Herr; Nathan E. Genung; Gary E. Aspnes; Jana Polivkova; Carmen N. Garcia-Irizarry; Qifang Li; Daniel Canterbury; Mark Niosi; Nicholas B. Vera; Zhenhong Li; Bhagyashree Khunte; Jaclyn Siderewicz; Timothy P. Rolph; Derek M. Erion

Inhibition of the sodium-coupled citrate transporter (NaCT or SLC13A5) has been proposed as a new therapeutic approach for prevention and treatment of metabolic diseases. In a previous report, we discovered dicarboxylate 1a (PF-06649298) which inhibits the transport of citrate in in vitro and in vivo settings via a specific interaction with NaCT. Herein, we report the optimization of this series leading to 4a (PF-06761281), a more potent inhibitor with suitable in vivo pharmacokinetic profile for assessment of in vivo pharmacodynamics. Compound 4a was used to demonstrate dose-dependent inhibition of radioactive [(14)C]citrate uptake in liver and kidney in vivo, resulting in modest reductions in plasma glucose concentrations.


Drug Metabolism and Disposition | 2016

Determination of Unbound Partition Coefficient and in Vitro–in Vivo Extrapolation for SLC13A Transporter–Mediated Uptake

Keith Riccardi; Zhenhong Li; Janice A. Brown; Matthew Gorgoglione; Mark Niosi; James R. Gosset; Kim Huard; Derek M. Erion; Li Di

Unbound partition coefficient (Kpuu) is important to an understanding of the asymmetric free drug distribution of a compound between cells and medium in vitro, as well as between tissue and plasma in vivo, especially for transporter-mediated processes. Kpuu was determined for a set of compounds from the SLC13A family that are inhibitors and substrates of transporters in hepatocytes and transporter-transfected cell lines. Enantioselectivity was observed, with (R)-enantiomers achieving much higher Kpuu (>4) than the (S)-enantiomers (<1) in human hepatocytes and SLC13A5-transfected human embryonic 293 cells. The intracellular free drug concentration correlated directly with in vitro pharmacological activity rather than the nominal concentration in the assay because of the high Kpuu mediated by SLC13A5 transporter uptake. Delivery of the diacid PF-06649298 directly or via hydrolysis of the ethyl ester prodrug PF-06757303 resulted in quite different Kpuu values in human hepatocytes (Kpuu of 3 for diacid versus 59 for prodrug), which was successfully modeled on the basis of passive diffusion, active uptake, and conversion rate from ester to diacid using a compartmental model. Kpuu values changed with drug concentrations; lower values were observed at higher concentrations possibly owing to a saturation of transporters. Michaelis-Menten constant (Km) of SLC13A5 was estimated to be 24 μM for PF-06649298 in human hepatocytes. In vitro Kpuu obtained from rat suspension hepatocytes supplemented with 4% fatty acid free bovine serum albumin showed good correlation with in vivo Kpuu of liver-to-plasma, illustrating the potential of this approach to predict in vivo Kpuu from in vitro systems.


Xenobiotica | 2013

Elucidation of the biochemical basis for a clinical drug–drug interaction between atorvastatin and 5-(N-(4-((4-ethylbenzyl)thio)phenyl)sulfamoyl)-2-methyl benzoic acid (CP-778 875), a subtype selective agonist of the peroxisome proliferator-activated receptor alpha

Amit S. Kalgutkar; Danny Chen; Manthena V. Varma; Bo Feng; Steven G. Terra; Renato J. Scialis; Charles J. Rotter; Kosea S. Frederick; Mark A. West; Theunis C. Goosen; James R. Gosset; Robert L. Walsky; Omar L. Francone

Abstract 1. 5-(N-(4-((4-ethylbenzyl)thio)phenyl)sulfamoyl)-2-methyl benzoic acid (CP-778 875), an agonist of the peroxisome proliferator-activated receptor alpha, has been evaluated in the clinic to treat dyslipidemia and type 2 diabetes mellitus. Herein, we investigate the effect of CP-778 875 on the pharmacokinetics of atorvastatin acid and its metabolites in humans. 2. The study incorporated a fixed-sequence design conducted in two groups. Group A was designed to estimate the effects of multiple doses of CP-778 875 on the single dose pharmacokinetics of atorvastatin. Subjects in group A (n = 26) received atorvastatin (40 mg) on days 1 and 9 and CP-778 875 (1.0 mg QD) on days 5–12. Group B was designed to examine the effects of multiple doses of atorvastatin on the single dose pharmacokinetics of CP-778 875. Subjects in group B (n = 29) received CP-778 875 (0.3 mg) on days 1 and 9 and atorvastatin (40 mg QD) on days 5–12. 3. Mean maximum serum concentration (Cmax) and area under the curve of atorvastatin were increased by 45% and 20%, respectively, upon co-administration with CP-778 875. Statistically significant increases in the systemic exposure of ortho- and para-hydroxyatorvastatin were also observed upon concomitant dosing with CP-778 875. CP-778 875 pharmacokinetics, however, were not impacted upon concomitant dosing with atorvastatin. 4. Inhibition of organic anion transporting polypeptide 1B1 by CP-778 875 (IC50 = 2.14 ±0.40 μM) could be the dominant cause of the pharmacokinetic interaction as CP-778 875 did not exhibit significant inhibition of cytochrome P450 3A4/3A5, multidrug resistant protein 1 or breast cancer resistant protein, which are also involved in the hepatobiliary disposition of atorvastatin.


Journal of Medicinal Chemistry | 2015

Discovery of Selective Small Molecule Inhibitors of Monoacylglycerol Acyltransferase 3.

Kim Huard; Allyn T. Londregan; Gregory Tesz; Kevin B. Bahnck; Thomas V. Magee; David Hepworth; Jana Polivkova; Steven B. Coffey; Brandon Pabst; James R. Gosset; Anu Nigam; Kou Kou; Hao Sun; Kyuha Lee; Michael Herr; Markus Boehm; Philip A. Carpino; Bryan Goodwin; Christian Perreault; Qifang Li; Csilla C. Jorgensen; George T. Tkalcevic; Timothy A. Subashi; Kay Ahn

Inhibition of triacylglycerol (TAG) biosynthetic enzymes has been suggested as a promising strategy to treat insulin resistance, diabetes, dyslipidemia, and hepatic steatosis. Monoacylglycerol acyltransferase 3 (MGAT3) is an integral membrane enzyme that catalyzes the acylation of both monoacylglycerol (MAG) and diacylglycerol (DAG) to generate DAG and TAG, respectively. Herein, we report the discovery and characterization of the first selective small molecule inhibitors of MGAT3. Isoindoline-5-sulfonamide (6f, PF-06471553) selectively inhibits MGAT3 with high in vitro potency and cell efficacy. Because the gene encoding MGAT3 (MOGAT3) is found only in higher mammals and humans, but not in rodents, a transgenic mouse model expressing the complete human MOGAT3 was used to characterize the effects of 6f in vivo. In the presence of a combination of diacylglycerol acyltransferases 1 and 2 (DGAT1 and DGAT2) inhibitors, an oral administration of 6f exhibited inhibition of the incorporation of deuterium-labeled glycerol into TAG in this mouse model. The availability of a potent and selective chemical tool and a humanized mouse model described in this report should facilitate further dissection of the physiological function of MGAT3 and its role in lipid homeostasis.

Collaboration


Dive into the James R. Gosset's collaboration.

Researchain Logo
Decentralizing Knowledge