Network


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

Hotspot


Dive into the research topics where Judith L. Treadway is active.

Publication


Featured researches published by Judith L. Treadway.


Expert Opinion on Investigational Drugs | 2001

Glycogen phosphorylase inhibitors for treatment of type 2 diabetes mellitus

Judith L. Treadway; Phil Mendys; Dennis J. Hoover

Type 2 diabetes mellitus is a severe disease with large economic consequences, which is significantly under-diagnosed and incompletely treated in the general population. Control of blood glucose levels is a key objective in treating diabetic patients, who are most often prescribed one or more oral hypoglycaemic agents in addition to diet and exercise modification as well as insulin. In spite of the availability of different classes of hypoglycaemic drugs, treatment regimens are often unable to achieve an intensive degree of glucose control known to most effectively reduce the incidence and severity of diabetic complications. Hepatic glucose output is elevated in type 2 diabetic patients and current evidence indicates that glycogenolysis (release of monomeric glucose from the glycogen polymer storage form) is an important contributor to the abnormally high production of glucose by the liver. Glycogen phosphorylase is the enzyme that catalyses this release and recent advances in new inhibitors of this structurally and kinetically well studied enzyme have enabled work which further delineate the pharmacological and physiological consequences of inhibiting glucose production by this pathway. Most notably, these agents lower glucose in diabetic animal models, both acutely and chronically, appear to affect both gluconeogenic and glycogenolytic pathways and demonstrate potential for a beneficial effect on cardiovascular risk factors. Cumulatively, this information has bolstered interest and promise in glycogen phosphorylase inhibitors (GPIs) as potential new hypoglycaemic agents for treatment of type 2 diabetes mellitus.


Chemistry & Biology | 2000

Human liver glycogen phosphorylase inhibitors bind at a new allosteric site

Virginia L. Rath; Mark Ammirati; Dennis E. Danley; Jennifer L Ekstrom; E. Michael Gibbs; Thomas R. Hynes; Alan M. Mathiowetz; R. Kirk McPherson; Thanh V. Olson; Judith L. Treadway; Dennis J. Hoover

BACKGROUND Glycogen phosphorylases catalyze the breakdown of glycogen to glucose-1-phosphate for glycolysis. Maintaining control of blood glucose levels is critical in minimizing the debilitating effects of diabetes, making liver glycogen phosphorylase a potential therapeutic target. RESULTS The binding site in human liver glycogen phosphorylase (HLGP) for a class of promising antidiabetic agents was identified crystallographically. The site is novel and functions allosterically by stabilizing the inactive conformation of HLGP. The initial view of the complex revealed key structural information and inspired the design of a new class of inhibitors which bind with nanomolar affinity and whose crystal structure is also described. CONCLUSIONS We have identified the binding site of a new class of allosteric HLGP inhibitors. The crystal structure revealed the details of inhibitor binding, led to the design of a new class of compounds, and should accelerate efforts to develop therapeutically relevant molecules for the treatment of diabetes.


Journal of Medicinal Chemistry | 2012

Discovery of (S)-6-(3-Cyclopentyl-2-(4-(trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic Acid as a Hepatoselective Glucokinase Activator Clinical Candidate for Treating Type 2 Diabetes Mellitus

Jeffrey A. Pfefferkorn; Angel Guzman-Perez; John Litchfield; Robert J. Aiello; Judith L. Treadway; John C. Pettersen; Martha L. Minich; Kevin J. Filipski; Christopher S. Jones; Meihua Tu; Gary E. Aspnes; Hud Risley; Jianwei Bian; Benjamin D. Stevens; Patricia Bourassa; Theresa D’Aquila; Levenia Baker; Nicole Barucci; Alan Robertson; Francis Bourbonais; David R. Derksen; Margit MacDougall; Over Cabrera; Jing Chen; Amanda Lee Lapworth; James A. Landro; William J. Zavadoski; Karen Atkinson; Nahor Haddish-Berhane; Beijing Tan

Glucokinase is a key regulator of glucose homeostasis, and small molecule allosteric activators of this enzyme represent a promising opportunity for the treatment of type 2 diabetes. Systemically acting glucokinase activators (liver and pancreas) have been reported to be efficacious but in many cases present hypoglycaemia risk due to activation of the enzyme at low glucose levels in the pancreas, leading to inappropriately excessive insulin secretion. It was therefore postulated that a liver selective activator may offer effective glycemic control with reduced hypoglycemia risk. Herein, we report structure-activity studies on a carboxylic acid containing series of glucokinase activators with preferential activity in hepatocytes versus pancreatic β-cells. These activators were designed to have low passive permeability thereby minimizing distribution into extrahepatic tissues; concurrently, they were also optimized as substrates for active liver uptake via members of the organic anion transporting polypeptide (OATP) family. These studies lead to the identification of 19 as a potent glucokinase activator with a greater than 50-fold liver-to-pancreas ratio of tissue distribution in rodent and non-rodent species. In preclinical diabetic animals, 19 was found to robustly lower fasting and postprandial glucose with no hypoglycemia, leading to its selection as a clinical development candidate for treating type 2 diabetes.


Journal of Pharmacology and Experimental Therapeutics | 2010

The Application of Target Information and Preclinical Pharmacokinetic/Pharmacodynamic Modeling in Predicting Clinical Doses of a Dickkopf-1 Antibody for Osteoporosis

Alison Betts; Tracey Clark; Jianxin Yang; Judith L. Treadway; Mei Li; Michael A. Giovanelli; Yasmina Noubia Abdiche; Donna Marie Stone; Vishwas M. Paralkar

PF-04840082 is a humanized prototype anti-Dickkopf-1 (Dkk-1) immunoglobulin isotype G2 (IgG2) antibody for the treatment of osteoporosis. In vitro, PF-04840082 binds to human, monkey, rat, and mouse Dkk-1 with high affinity. After administration of PF-04840082 to rat and monkey, free Dkk-1 concentrations decreased rapidly and returned to baseline in a dose-dependent manner. In rat and monkey, PF-04840082 exhibited nonlinear pharmacokinetics (PK) and a target-mediated drug disposition (TMDD) model was used to characterize PF-04840082 versus Dkk-1 concentration response relationship. PK/pharmacodynamic (PK/PD) modeling enabled estimation of antibody non-target-mediated elimination, Dkk-1 turnover, complex formation, and complex elimination. The TMDD model was translated to human to predict efficacious dose and minimum anticipated biological effect level (MABEL) by incorporating information on typical IgG2 human PK, antibody-target association/dissociation rates, Dkk-1 expression, and turnover rates. The PK/PD approach to MABEL was compared with the standard “no adverse effect level” (NOAEL) approach to calculating clinical starting doses and a pharmacological equilibrium method. The NOAEL method gave estimates of dose that were too high to ensure safety of clinical trials. The pharmacological equilibrium approach calculated receptor occupancy (RO) based on equilibrium dissociation constant alone and did not take into account rate of turnover of the target or antibody–target complex kinetics and, as a result, it likely produced a substantial overprediction of RO at a given dose. It was concluded that the calculation of MABEL according to the TMDD model was the most appropriate means for ensuring safety and efficacy in clinical studies.


The Journal of Nuclear Medicine | 2012

In Vivo Imaging of Endogenous Pancreatic β-Cell Mass in Healthy and Type 1 Diabetic Subjects Using 18F-Fluoropropyl-Dihydrotetrabenazine and PET

Marc D. Normandin; Kitt Falk Petersen; Yu-Shin Ding; Shu-fei Lin; Sarita Naik; Krista Fowles; Daniel Skovronsky; Kevan C. Herold; Timothy J. McCarthy; Roberto A. Calle; Richard E. Carson; Judith L. Treadway; Gary W. Cline

The ability to noninvasively measure endogenous pancreatic β-cell mass (BCM) would accelerate research on the pathophysiology of diabetes and revolutionize the preclinical development of new treatments, the clinical assessment of therapeutic efficacy, and the early diagnosis and subsequent monitoring of disease progression. The vesicular monoamine transporter type 2 (VMAT2) is coexpressed with insulin in β-cells and represents a promising target for BCM imaging. Methods: We evaluated the VMAT2 radiotracer 18F-fluoropropyl-dihydrotetrabenazine (18F-FP-(+)-DTBZ, also known as 18F-AV-133) for quantitative PET of BCM in healthy control subjects and patients with type 1 diabetes mellitus. Standardized uptake value was calculated as the net tracer uptake in the pancreas normalized by injected dose and body weight. Total volume of distribution, the equilibrium ratio of tracer concentration in tissue relative to plasma, was estimated by kinetic modeling with arterial input functions. Binding potential, the steady-state ratio of specific binding to nondisplaceable uptake, was calculated using the renal cortex as a reference tissue devoid of specific VMAT2 binding. Results: Mean pancreatic standardized uptake value, total volume of distribution, and binding potential were reduced by 38%, 20%, and 40%, respectively, in type 1 diabetes mellitus. The radiotracer binding parameters correlated with insulin secretion capacity as determined by arginine-stimulus tests. Group differences and correlations with β-cell function were enhanced for total pancreas binding parameters that accounted for tracer binding density and organ volume. Conclusion: These findings demonstrate that quantitative evaluation of islet β-cell density and aggregate BCM can be performed clinically with 18F-FP-(+)-DTBZ PET.


BMC Genomics | 2011

Polyomic profiling reveals significant hepatic metabolic alterations in glucagon-receptor (GCGR) knockout mice: implications on anti-glucagon therapies for diabetes

Jianxin Yang; Margit MacDougall; Michael T. McDowell; Li Xi; Ru Wei; William J. Zavadoski; Mark P. Molloy; John D Baker; Max Kuhn; Over Cabrera; Judith L. Treadway

BackgroundGlucagon is an important hormone in the regulation of glucose homeostasis, particularly in the maintenance of euglycemia and prevention of hypoglycemia. In type 2 Diabetes Mellitus (T2DM), glucagon levels are elevated in both the fasted and postprandial states, which contributes to inappropriate hyperglycemia through excessive hepatic glucose production. Efforts to discover and evaluate glucagon receptor antagonists for the treatment of T2DM have been ongoing for approximately two decades, with the challenge being to identify an agent with appropriate pharmaceutical properties and efficacy relative to potential side effects. We sought to determine the hepatic & systemic consequence of full glucagon receptor antagonism through the study of the glucagon receptor knock-out mouse (Gcgr-/-) compared to wild-type littermates.ResultsLiver transcriptomics was performed using Affymetric expression array profiling, and liver proteomics was performed by iTRAQ global protein analysis. To complement the transcriptomic and proteomic analyses, we also conducted metabolite profiling (~200 analytes) using mass spectrometry in plasma. Overall, there was excellent concordance (R = 0.88) for changes associated with receptor knock-out between the transcript and protein analysis. Pathway analysis tools were used to map the metabolic processes in liver altered by glucagon receptor ablation, the most notable being significant down-regulation of gluconeogenesis, amino acid catabolism, and fatty acid oxidation processes, with significant up-regulation of glycolysis, fatty acid synthesis, and cholesterol biosynthetic processes. These changes at the level of the liver were manifested through an altered plasma metabolite profile in the receptor knock-out mice, e.g. decreased glucose and glucose-derived metabolites, and increased amino acids, cholesterol, and bile acid levels.ConclusionsIn sum, the results of this study suggest that the complete ablation of hepatic glucagon receptor function results in major metabolic alterations in the liver, which, while promoting improved glycemic control, may be associated with adverse lipid changes.


Molecular Imaging and Biology | 2011

Pancreatic beta cell mass PET imaging and quantification with [11C]DTBZ and [18F]FP-(+)-DTBZ in rodent models of diabetes.

Tarun Singhal; Yu-Shin Ding; David Weinzimmer; Marc D. Normandin; David Labaree; Jim Ropchan; Nabeel Nabulsi; Shu-fei Lin; Marc B. Skaddan; Walter C. Soeller; Yiyun Huang; Richard E. Carson; Judith L. Treadway; Gary W. Cline

PurposeThe aim of this study is to compare the utility of two positron emission tomography (PET) imaging ligands ((+)-[11C]dihydrotetrabenazine ([11C]DTBZ) and the fluoropropyl analog ([18F]FP-(+)-DTBZ)) that target islet β-cell vesicular monoamine transporter type II to measure pancreatic β-cell mass (BCM).Procedures[11C]DTBZ or [18F]FP-(+)-DTBZ was injected, and serial PET images were acquired in rat models of diabetes (streptozotocin-treated and Zucker diabetic fatty) and β-cell compensation (Zucker fatty). Radiotracer standardized uptake values (SUV) were correlated to pancreas insulin content measured biochemically and histomorphometrically.ResultsOn a group level, a positive correlation of [11C]DTBZ pancreatic SUV with pancreas insulin content and BCM was observed. In the STZ diabetic model, both [18F]FP-(+)-DTBZ and [11C]DTBZ correlated positively with BCM, although only ∼25% of uptake could be attributed to β-cell uptake. [18F]FP-(+)-DTBZ displacement studies indicate that there is a substantial fraction of specific binding that is not to pancreatic islet β cells.ConclusionsPET imaging with [18F]FP-(+)-DTBZ provides a noninvasive means to quantify insulin-positive BCM and may prove valuable as a diagnostic tool in assessing treatments to maintain or restore BCM.


Bioorganic & Medicinal Chemistry Letters | 2003

3-(2-carboxyethyl)-4,6-dichloro-1H-indole-2-carboxylic acid: an allosteric inhibitor of fructose-1,6-bisphosphatase at the AMP site.

Stephen W. Wright; Anthony A. Carlo; Dennis E. Danley; David L. Hageman; George A. Karam; Mahmoud N. Mansour; Lester D. McClure; Jayvardhan Pandit; Gayle K. Schulte; Judith L. Treadway; Ing-Kae Wang; Paul H. Bauer

3-(2-Carboxyethyl)-4,6-dichloro-1H-indole-2-carboxylic acid (MDL-29951), an antagonist of the glycine site of the NMDA receptor, has been found to be an allosteric inhibitor of the enzyme fructose 1,6-bisphosphatase. The compound binds at the AMP regulatory site by X-ray crystallography. This represents a new approach to inhibition of fructose 1,6-bisphosphatase and serves as a lead for further drug design.


MedChemComm | 2011

Designing glucokinase activators with reduced hypoglycemia risk: discovery of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as a clinical candidate for the treatment of type 2 diabetes mellitus

Jeffrey A. Pfefferkorn; Angel Guzman-Perez; Peter J. Oates; John Litchfield; Gary E. Aspnes; Arindrajit Basak; John William Benbow; Martin A. Berliner; Jianwei Bian; Chulho Choi; Kevin Daniel Freeman-Cook; Jeffrey W. Corbett; Mary Theresa Didiuk; Joshua R. Dunetz; Kevin J. Filipski; William M. Hungerford; Christopher S. Jones; Kapil Karki; Anthony Lai Ling; Jian-Cheng Li; Leena Patel; Christian Perreault; Hud Risley; James Saenz; Wei Song; Meihua Tu; Robert J. Aiello; Karen Atkinson; Nicole Barucci; David A. Beebe

Glucokinase is a key regulator of glucose homeostasis and small molecule activators of this enzyme represent a promising opportunity for the treatment of Type 2 diabetes. Several glucokinase activators have advanced to clinical studies and demonstrated promising efficacy; however, many of these early candidates also revealed hypoglycemia as a key risk. In an effort to mitigate this hypoglycemia risk while maintaining the promising efficacy of this mechanism, we have investigated a series of substituted 2-methylbenzofurans as “partial activators” of the glucokinase enzyme leading to the identification of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as an early development candidate.


Bioorganic & Medicinal Chemistry Letters | 2001

Allosteric inhibition of fructose-1,6-bisphosphatase by anilinoquinazolines

Stephen W. Wright; David L. Hageman; Lester D. McClure; Anthony A. Carlo; Judith L. Treadway; Alan M. Mathiowetz; Jane M. Withka; Paul H. Bauer

Anilinoquinazolines currently of interest as inhibitors of tyrosine kinases have been found to be allosteric inhibitors of the enzyme fructose 1,6-bisphosphatase. These represent a new approach to inhibition of F16BPase and serve as leads for further drug design. Enzyme inhibition is achieved by binding at an unidentified allosteric site.

Collaboration


Dive into the Judith L. Treadway's collaboration.

Researchain Logo
Decentralizing Knowledge