Network


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

Hotspot


Dive into the research topics where Kim Huard is active.

Publication


Featured researches published by Kim Huard.


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 | 2013

Spirolactam-Based Acetyl-CoA Carboxylase Inhibitors: Toward Improved Metabolic Stability of a Chromanone Lead Structure

David A. Griffith; Robert L. Dow; Kim Huard; David J. Edmonds; Scott W. Bagley; Jana Polivkova; Dongxiang Zeng; Carmen N. Garcia-Irizarry; James A. Southers; William Esler; Paul Amor; Kathrine Loomis; Kirk McPherson; Kevin B. Bahnck; Cathy Préville; Tereece Banks; Dianna E. Moore; Alan M. Mathiowetz; Elnaz Menhaji-Klotz; Aaron Smith; Shawn D. Doran; David A. Beebe; Matthew F. Dunn

Acetyl-CoA carboxylase (ACC) catalyzes the rate-determining step in de novo lipogenesis and plays a crucial role in the regulation of fatty acid oxidation. Alterations in lipid metabolism are believed to contribute to insulin resistance; thus inhibition of ACC offers a promising option for intervention in type 2 diabetes mellitus. Herein we disclose a series of ACC inhibitors based on a spirocyclic pyrazololactam core. The lactam series has improved chemical and metabolic stability relative to our previously reported pyrazoloketone series, while retaining potent inhibition of ACC1 and ACC2. Optimization of the pyrazole and amide substituents led to quinoline amide 21, which was advanced to preclinical development.


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.


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.


Bioorganic & Medicinal Chemistry | 2017

A multi-endpoint matched molecular pair (MMP) analysis of 6-membered heterocycles

George Chang; Kim Huard; Gregory W. Kauffman; Antonia F. Stepan; Christopher E. Keefer

Aromatic rings, ubiquitous in pharmaceutical compounds, are often exchanged with another ring during the optimization process of drug discovery. Inevitably, the preferred ring system for one endpoint may prove detrimental to another, thus necessitating a holistic, multiple endpoint optimization approach for finding the ideal replacement. Accordingly, we conducted an extensive matched molecular pair (MMP) analysis of common 6-membered aromatic rings across 4 endpoints critical for drug discovery (logD lipophilicity, microsomal metabolism, P-gp efflux and passive permeability). We also investigated the effect of context by considering the connecting atom. Heat maps were created as a simple yet comprehensive way to view and analyze the vast amount of interrelated data. Paired difference statistical tests were used to identify transforms with changes that were significantly different from zero. We conclude that the heat maps of transforms provide a unique and powerful approach for multiparameter optimization.


Molecular Pharmacology | 2016

Molecular basis for inhibition of the Na+/citrate transporter, NaCT (SLC13A5) by dicarboxylate inhibitors.

Ana M. Pajor; César Augusto F. de Oliveira; Kun Song; Kim Huard; Veerabahu Shanmugasundaram; Derek M. Erion

The Na+/citrate transporter, NaCT (SLC13A5), is a therapeutic target for metabolic diseases. Citrate is an important signaling molecule that regulates the activity of lipid- and glucose-metabolizing enzymes in cells. Previous studies identified two compounds, PF-06649298 (compound 2) and PF-06678419 (compound 4), that inhibit human NaCT with high affinity, and one of the compounds demonstrated specificity relative to other SLC13 family members. Here we use molecular modeling and site-directed mutagenesis of hNaCT followed by transport characterization and cell-surface biotinylation to examine the residues involved in inhibitor binding and transport. The results indicate that residues located near the putative citrate binding site, G228, V231, V232, and G409, affect both citrate transport and inhibition of citrate uptake by compounds 2 and 4. V231 appears to distinguish between compounds 2 and 4 as inhibitors. Furthermore, residues located outside of the putative citrate binding site, Q77 and T86, may also play a role in NaCT inhibition by compounds 2 and 4. Our results provide new insight into the mechanism of transport and inhibition in NaCT and the SLC13 family. These findings should provide a basis for future drug design of SLC13 inhibitors.


Journal of Medicinal Chemistry | 2017

Discovery of Fragment-Derived Small Molecules for in Vivo Inhibition of Ketohexokinase (KHK)

Kim Huard; Kay Ahn; Paul Amor; David A. Beebe; Kris A. Borzilleri; Boris A. Chrunyk; Steven B. Coffey; Yang Cong; Edward L. Conn; Jeffrey S. Culp; Matthew S. Dowling; Matthew Gorgoglione; Jemy A. Gutierrez; John D. Knafels; Erik LaChapelle; Jayvardhan Pandit; Kevin D. Parris; Sylvie Perez; Jeffrey A. Pfefferkorn; David A. Price; Brian Raymer; Trenton T. Ross; Andre Shavnya; Aaron Smith; Timothy A. Subashi; Gregory Tesz; Benjamin A. Thuma; Meihua Tu; John D. Weaver; Yan Weng

Increased fructose consumption and its subsequent metabolism have been implicated in hepatic steatosis, dyslipidemia, obesity, and insulin resistance in humans. Since ketohexokinase (KHK) is the principal enzyme responsible for fructose metabolism, identification of a selective KHK inhibitor may help to further elucidate the effect of KHK inhibition on these metabolic disorders. Until now, studies on KHK inhibition with small molecules have been limited due to the lack of viable in vivo pharmacological tools. Herein we report the discovery of 12, a selective KHK inhibitor with potency and properties suitable for evaluating KHK inhibition in rat models. Key structural features interacting with KHK were discovered through fragment-based screening and subsequent optimization using structure-based drug design, and parallel medicinal chemistry led to the identification of pyridine 12.


Journal of Organic Chemistry | 2012

Synthesis of Spiropiperidine Lactam Acetyl-CoA Carboxylase Inhibitors

Kim Huard; Scott W. Bagley; Elnaz Menhaji-Klotz; Cathy Préville; James A. Southers; Aaron Smith; David J. Edmonds; John C. Lucas; Matthew F. Dunn; Nigel M. Allanson; Emma L. Blaney; Carmen N. Garcia-Irizarry; Jeffrey T. Kohrt; David A. Griffith; Robert L. Dow


MedChemComm | 2017

Small structural changes of the imidazopyridine diacylglycerol acyltransferase 2 (DGAT2) inhibitors produce an improved safety profile

Kentaro Futatsugi; Kim Huard; Daniel W. Kung; John C. Pettersen; Declan Flynn; James R. Gosset; Gary E. Aspnes; R. J. Barnes; Shawn Cabral; Matthew S. Dowling; Dilinie P. Fernando; Theunis C. Goosen; W. P. Gorczyca; David Hepworth; Michael Herr; Sophie Y. Lavergne; Qifang Li; Mark Niosi; Suvi T. M. Orr; I. D. Pardo; Sylvie Perez; Julie Purkal; T. J. Schmahai; N. Shirai; A. M. Shoieb; J. Zhou; Bryan Goodwin

Collaboration


Dive into the Kim Huard's collaboration.

Researchain Logo
Decentralizing Knowledge