Network


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

Hotspot


Dive into the research topics where Nathan Bays is active.

Publication


Featured researches published by Nathan Bays.


Clinical Cancer Research | 2008

Control of cell growth and survival by enzymes of the fatty acid synthesis pathway in HCT-116 colon cancer cells.

Yanai Zhan; Nicole Ginanni; Michael R. Tota; Margaret Wu; Nathan Bays; Victoria M. Richon; Nancy E. Kohl; Eric Bachman; Peter Strack; Stefan Krauss

Purpose: For many tumor cells, de novo lipogenesis is a requirement for growth and survival. A considerable body of work suggests that inhibition of this pathway may be a powerful approach to antineoplastic therapy. It has recently been shown that inhibition of various steps in the lipogenic pathway individually can induce apoptosis or loss of viability in tumor cells. However, it is not clear whether quantitative differences exist in the ability of lipogenic enzymes to control tumor cell survival. We present a systematic approach that allows for a direct comparison of the control of lipogenic pathway enzymes over tumor cell growth and apoptosis using different cancer cells. Experimental Design: RNA interference-mediated, graded down-regulation of fatty acid synthase (FAS) pathway enzymes was employed in combination with measurements of lipogenesis, apoptosis, and cell growth. Results: In applying RNA interference titrations to two lipogenic enzymes, acetyl-CoA carboxylase 1 (ACC1) and FAS, we show that ACC1 and FAS both significantly control cell growth and apoptosis in HCT-116 cells. These results also extend to PC-3 and A2780 cancer cells. Conclusions: Control of tumor cell survival by different steps in de novo lipogenesis can be quantified. Because ACC1 and FAS both significantly control tumor cell growth and apoptosis, we propose that pharmacologic inhibitors of either enzyme might be useful agents in targeting cancer cells that critically rely on fatty acid synthesis. The experimental approach described here may be extended to other targets or disease-relevant pathways to identify steps suitable for therapeutic intervention.


ACS Medicinal Chemistry Letters | 2016

Discovery of 8-Amino-imidazo[1,5-a]pyrazines as Reversible BTK Inhibitors for the Treatment of Rheumatoid Arthritis.

Jian Liu; Deodial Guiadeen; Arto D. Krikorian; Xiaolei Gao; James Wang; Sobhana Babu Boga; Abdul-Basit Alhassan; Younong Yu; Henry A. Vaccaro; Shilan Liu; Chundao Yang; Hao Wu; Alan B. Cooper; Jos de Man; Allard Kaptein; Kevin M. Maloney; Viktor Hornak; Ying-Duo Gao; Thierry O. Fischmann; Hans C.A. Raaijmakers; Diep Vu-Pham; Jeremy Presland; My Mansueto; Zangwei Xu; Erica Leccese; Jie Zhang-Hoover; Ian Knemeyer; Charles G. Garlisi; Nathan Bays; Peter Stivers

Brutons tyrosine kinase (BTK) is a Tec family kinase with a well-defined role in the B cell receptor (BCR) pathway. It has become an attractive kinase target for selective B cell inhibition and for the treatment of B cell related diseases. We report a series of compounds based on 8-amino-imidazo[1,5-a]pyrazine that are potent reversible BTK inhibitors with excellent kinase selectivity. Selectivity is achieved through specific interactions of the ligand with the kinase hinge and driven by aminopyridine hydrogen bondings with Ser538 and Asp539, and by hydrophobic interaction of trifluoropyridine in the back pocket. These interactions are evident in the X-ray crystal structure of the lead compounds 1 and 3 in the complex with the BTK enzyme. Our lead compounds show desirable PK profiles and efficacy in the preclinical rat collagen induced arthritis model.


Bioorganic & Medicinal Chemistry Letters | 2011

Triazoles as γ-secretase modulators.

Christian Fischer; Susan L. Zultanski; Hua Zhou; Joey L. Methot; W. Colby Brown; Dawn M. Mampreian; Adam J. Schell; Sanjiv Shah; Hugh Nuthall; Bethany Hughes; Nadja Smotrov; Candia M. Kenific; Jonathan C. Cruz; Deborah Walker; Melanie Bouthillette; George Nikov; Dan Savage; Valentina V. Jeliazkova-Mecheva; Damaris Diaz; Alexander A. Szewczak; Nathan Bays; Richard E. Middleton; Benito Munoz; Mark S. Shearman

Synthesis, SAR, and evaluation of aryl triazoles as novel gamma secretase modulators (GSMs) are presented in this communication. Starting from the literature and in-house leads, we evaluated a range of five-membered heterocycles as replacements for olefins commonly found in non-acid GSMs. 1,2,3-C-aryl-triazoles were identified as suitable replacements which exhibited good modulation of γ-secretase activity, excellent pharmacokinetics and good central lowering of Aβ42 in Sprague-Dawley rats.


Bioorganic & Medicinal Chemistry Letters | 2012

Triazoloamides as potent γ-secretase modulators with reduced hERG liability.

Christian Fischer; Susan L. Zultanski; Hua Zhou; Joey L. Methot; Sanjiv Shah; Hugh Nuthall; Bethany Hughes; Nadja Smotrov; Armetta D. Hill; Alexander A. Szewczak; Christopher M. Moxham; Nathan Bays; Richard E. Middleton; Benito Munoz; Mark S. Shearman

Synthesis and SAR studies of novel aryl triazoles as gamma secretase modulators (GSMs) are presented in this communication. Starting from our aryl triazole leads, optimization studies were continued and the series progressed towards novel amides and lactams. Triazole 57 was identified as the most potent analog in this series, displaying single-digit nanomolar Aβ42 IC(50) in cell-based assays and reduced affinity for the hERG channel.


Journal of Biomolecular Screening | 2009

A Simplified Scintillation Proximity Assay for Fatty Acid Synthase Activity: Development and Comparison with Other FAS Activity Assays

Nathan Bays; Armetta D. Hill; Ilona Kariv

Fatty acid synthase (FAS), an essential enzyme for de novo lipogenesis, has been implicated in a number of disease states, including obesity, dyslipidemia, and cancer. To identify small-molecule inhibitors of FAS, the authors developed a bead-based scintillation proximity assay (SPA) to detect the fatty acid products of FAS enzymatic activity. This homogeneous SPA assay discriminates between a radiolabeled hydrophilic substrate of FAS (acetyl-coenzyme A) and the labeled lipophilic products of FAS (fatty acids), generating signal only when labeled fatty acids are present. The assay requires a single addition of unmodified polystyrene imaging SPA beads and can be miniaturized to 384- or 1536-well density with appropriate assay statistics for high-throughput screening. High-potency FAS inhibitors were used to compare the sensitivity of the SPA bead assay with previously described assays that measure FAS reaction intermediates (CoA-SH and NADP +). The advantages and disadvantages of these different FAS assays in small-molecule inhibitor discovery are discussed. (Journal of Biomolecular Screening 2009:636-642)


Journal of Pharmacology and Experimental Therapeutics | 2017

Evaluation of JAK3 biology in autoimmune disease using a highly selective, irreversible JAK3 inhibitor

Fiona Elwood; David J. Witter; Jennifer Piesvaux; Brian Kraybill; Nathan Bays; Carla Alpert; Peter Goldenblatt; Yujie Qu; Irena Ivanovska; Hyun-Hee Lee; Chi-Sung Chiu; Hao Tang; Mark E. Scott; Sujal V. Deshmukh; Mark Zielstorff; Alan Byford; Kalyan Chakravarthy; Lauren Dorosh; Alexey Rivkin; Joel A. Klappenbach; Bo-Sheng Pan; Ilona Kariv; Christopher J. Dinsmore; Deborah Slipetz; Peter J. Dandliker

Reversible janus associated kinase (JAK) inhibitors such as tofacitinib and decernotinib block cytokine signaling and are efficacious in treating autoimmune diseases. However, therapeutic doses are limited due to inhibition of other JAK/signal transducer and activator of transcription pathways associated with hematopoiesis, lipid biogenesis, infection, and immune responses. A selective JAK3 inhibitor may have a better therapeutic index; however, until recently, no compounds have been described that maintain JAK3 selectivity in cells, as well as against the kinome, with good physicochemical properties to test the JAK3 hypothesis in vivo. To quantify the biochemical basis for JAK isozyme selectivity, we determined that the apparent Km value for each JAK isozyme ranged from 31.8 to 2.9 μM for JAK1 and JAK3, respectively. To confirm compound activity in cells, we developed a novel enzyme complementation assay that read activity of single JAK isozymes in a cellular context. Reversible JAK3 inhibitors cannot achieve sufficient selectivity against other isozymes in the cellular context due to inherent differences in enzyme ATP Km values. Therefore, we developed irreversible JAK3 compounds that are potent and highly selective in vitro in cells and against the kinome. Compound 2, a potent inhibitor of JAK3 (0.15 nM) was 4300-fold selective for JAK3 over JAK1 in enzyme assays, 67-fold [interleukin (IL)-2 versus IL-6] or 140-fold [IL-2 versus erythropoietin or granulocyte-macrophage colony-stimulating factor (GMCSF)] selective in cellular reporter assays and >35-fold selective in human peripheral blood mononuclear cell assays (IL-7 versus IL-6 or GMCSF). In vivo, selective JAK3 inhibition was sufficient to block the development of inflammation in a rat model of rheumatoid arthritis, while sparing hematopoiesis.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of novel triazolobenzazepinones as γ-secretase modulators with central Aβ42 lowering in rodents and rhesus monkeys.

Christian Fischer; Susan L. Zultanski; Hua Zhou; Joey L. Methot; Sanjiv Shah; Ikuo Hayashi; Bethany Hughes; Christopher M. Moxham; Nathan Bays; Nadya Smotrov; Armetta D. Hill; Bo-Sheng Pan; Zhenhua Wu; Lily Y. Moy; Flobert Tanga; Candia M. Kenific; Jonathan C. Cruz; Deborah Walker; Melanie Bouthillette; George Nikov; Sujal V. Deshmukh; Valentina V. Jeliazkova-Mecheva; Damaris Diaz; Maria S. Michener; Jacquelynn J. Cook; Benito Munoz; Mark S. Shearman

Synthesis and SAR studies of novel triazolobenzazepinones as gamma secretase modulators (GSMs) are presented in this communication. Starting from our azepinone leads, optimization studies toward improving central lowering of Aβ42 led to the discovery of novel benzo-fused azepinones. Several benzazepinones were profiled in vivo and found to lower brain Aβ42 levels in Sprague Dawley rats and transgenic APP-YAC mice in a dose-dependent manner after a single oral dose. Compound 34 was further progressed into a pilot study in our cisterna-magna-ported rhesus monkey model, where we observed robust lowering of CSF Aβ42 levels.


Journal of Biomolecular Screening | 2009

Development of a Cell-Based Assay for Measurement of c-Met Phosphorylation Using AlphaScreenTM Technology and High-Content Imaging Analysis

Nadya Smotrov; Anjili Mathur; Ilona Kariv; Christopher M. Moxham; Nathan Bays

c-Met is a receptor tyrosine kinase (RTK) with a critical role in many fundamental cellular processes, including cell proliferation and differentiation. Deregulated c-Met signaling has been implicated in both the initiation and progression of human cancers and therefore represents an attractive target for anticancer therapy. Monitoring the phosphorylation status of relevant tyrosine residues provides an important method of assessing c-Met kinase activity. This report describes a novel assay to monitor c-Met phosphorylation in cells using Amplified Luminescent Proximity Homogeneous Assay (AlphaScreen™) technology. Using AlphaScreen™, the authors were able to detect both global and site-specific phosphorylation of c-Met in transformed cell lines. Data obtained from the AlphaScreen™ assay were compared to data obtained from a high-content imaging (HCI) method developed in parallel to monitor c-Met phosphorylation at the single cell level. The AlphaScreen™ assay was miniaturized to a 384-well format with acceptable signal-to-background ratio (S/B) and Z′ statistics and was employed to measure c-Met kinase activity in situ after treatment with potent c-Met-specific kinase inhibitors. The authors discuss the utility of quantifying endogenous cellular c-Met phosphorylation in lead optimization and how the modular design of the AlphaScreen™ assay allows its adaptation to measure cellular activity of other kinases. (Journal of Biomolecular Screening 2009:404-411)


Journal of Medicinal Chemistry | 2017

The Discovery of 3-((4-Chloro-3-methoxyphenyl)amino)-1-((3R,4S)-4-cyanotetrahydro-2H-pyran-3-yl)-1H-pyrazole-4-carboxamide, a Highly Ligand Efficient and Efficacious Janus Kinase 1 Selective Inhibitor with Favorable Pharmacokinetic Properties

Tony Siu; Jason Brubaker; Peter Fuller; Luis Torres; Hongbo Zeng; Joshua Close; Dawn M. Mampreian; Feng Shi; Duan Liu; Xavier Fradera; Kevin Johnson; Nathan Bays; Elma Kadic; Fang He; Peter Goldenblatt; Lynsey Shaffer; Sangita B. Patel; Charles A. Lesburg; Carla Alpert; Lauren Dorosh; Sujal V. Deshmukh; Hongshi Yu; Joel A. Klappenbach; Fiona Elwood; Christopher J. Dinsmore; Rafael Fernández; Lily Y. Moy; Jonathan R. Young

The discovery of a potent selective low dose Janus kinase 1 (JAK1) inhibitor suitable for clinical evaluation is described. As part of an overall goal to minimize dose, we pursued a medicinal chemistry strategy focused on optimization of key parameters that influence dose size, including lowering human Clint and increasing intrinsic potency, bioavailability, and solubility. To impact these multiple parameters simultaneously, we used lipophilic ligand efficiency as a key metric to track changes in the physicochemical properties of our analogs, which led to improvements in overall compound quality. In parallel, structural information guided advancements in JAK1 selectivity by informing on new vector space, which enabled the discovery of a unique key amino acid difference between JAK1 (Glu966) and JAK2 (Asp939). This difference was exploited to consistently produce analogs with the best balance of JAK1 selectivity, efficacy, and projected human dose, ultimately culminating in the discovery of compound 28.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of novel BTK inhibitors with carboxylic acids

Xiaolei Gao; James C. Wang; Jian Liu; Deodial Guiadeen; Arto D. Krikorian; Sobhana Babu Boga; Abdul-Basit Alhassan; Oleg Selyutin; Wensheng Yu; Younong Yu; Rajan Anand; Shilan Liu; Chundao Yang; Hao Wu; Jiaqiang Cai; Alan B. Cooper; Hugh Y. Zhu; Kevin M. Maloney; Ying-Duo Gao; Thierry O. Fischmann; Jeremy Presland; My Mansueto; Zangwei Xu; Erica Leccese; Jie Zhang-Hoover; Ian Knemeyer; Charles G. Garlisi; Nathan Bays; Peter Stivers; Philip E. Brandish

We report the design and synthesis of a series of novel Brutons Tyrosine Kinase (BTK) inhibitors with a carboxylic acid moiety in the ribose pocket. This series of compounds has demonstrated much improved off-target selectivities including adenosine uptake (AdU) inhibition compared to the piperidine amide series. Optimization of the initial lead compound 4 based on BTK enzyme inhibition, and human peripheral blood mononuclear cell (hPBMC) and human whole blood (hWB) activity led to the discovery of compound 40, with potent BTK inhibition, reduced off target activities, as well as favorable pharmacokinetic profile in both rat and dog.

Researchain Logo
Decentralizing Knowledge