Network


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

Hotspot


Dive into the research topics where Lynn M. Abell is active.

Publication


Featured researches published by Lynn M. Abell.


Journal of Medicinal Chemistry | 2013

Discovery of 2-(phenoxypyridine)-3-phenylureas as small molecule P2Y1 antagonists.

Hannguang Chao; Huji Turdi; Timothy F. Herpin; Jacques Y. Roberge; Yalei Liu; Dora M. Schnur; Michael A. Poss; Robert Rehfuss; Ji Hua; Qimin Wu; Laura A. Price; Lynn M. Abell; William A. Schumacher; Jeffrey S. Bostwick; Thomas E. Steinbacher; Anne B. Stewart; Martin L. Ogletree; Christine Huang; Ming Chang; Angela Cacace; Maredith J. Arcuri; Deborah Celani; Ruth R. Wexler; R. Michael Lawrence

Two distinct G protein-coupled purinergic receptors, P2Y1 and P2Y12, mediate ADP-driven platelet activation. The clinical effectiveness of P2Y12 blockade is well established. Recent preclinical data suggest that P2Y1 and P2Y12 inhibition provide equivalent antithrombotic efficacy, while targeting P2Y1 has the potential for reduced bleeding liability. In this account, the discovery of a 2-(phenoxypyridine)-3-phenylurea chemotype that inhibited ADP-mediated platelet aggregation in human blood samples is described. Optimization of this series led to the identification of compound 16, 1-(2-(2-tert-butylphenoxy)pyridin-3-yl)-3-4-(trifluoromethoxy)phenylurea, which demonstrated a 68 ± 7% thrombus weight reduction in an established rat arterial thrombosis model (10 mg/kg plus 10 mg/kg/h) while only prolonging cuticle and mesenteric bleeding times by 3.3- and 3.1-fold, respectively, in provoked rat bleeding time models. These results suggest that a P2Y1 antagonist could potentially provide a safe and efficacious antithrombotic profile.


Journal of Medicinal Chemistry | 2013

Conformationally Constrained ortho-Anilino Diaryl Ureas: Discovery of 1-(2-(1′-Neopentylspiro[indoline-3,4′-piperidine]-1-yl)phenyl)-3-(4-(trifluoromethoxy)phenyl)urea, a Potent, Selective, and Bioavailable P2Y1 Antagonist

Jennifer X. Qiao; Tammy C. Wang; Rejean Ruel; Carl Thibeault; Alexandre L’Heureux; William A. Schumacher; Steven A. Spronk; Sheldon Hiebert; Gilles Bouthillier; John Lloyd; Zulan Pi; Dora M. Schnur; Lynn M. Abell; Ji Hua; Laura A. Price; Eddie C.-K. Liu; Qimin Wu; Thomas E. Steinbacher; Jeffrey S. Bostwick; Ming Chang; Joanna Zheng; Qi Gao; Baoqing Ma; Patricia A. McDonnell; Christine Huang; Robert Rehfuss; Ruth R. Wexler; Patrick Y. S. Lam

Preclinical antithrombotic efficacy and bleeding models have demonstrated that P2Y1 antagonists are efficacious as antiplatelet agents and may offer a safety advantage over P2Y12 antagonists in terms of reduced bleeding liabilities. In this article, we describe the structural modification of the tert-butyl phenoxy portion of lead compound 1 and the subsequent discovery of a novel series of conformationally constrained ortho-anilino diaryl ureas. In particular, spiropiperidine indoline-substituted diaryl ureas are described as potent, orally bioavailable small-molecule P2Y1 antagonists with improved activity in functional assays and improved oral bioavailability in rats. Homology modeling and rat PK/PD studies on benchmark compound 3l will also be presented. Compound 3l was our first P2Y1 antagonist to demonstrate a robust oral antithrombotic effect with mild bleeding liability in the rat thrombosis and hemostasis models.


International Journal of Toxicology | 2013

Nonclinical Toxicology Assessments Support the Chronic Safety of Dapagliflozin, a First-in-Class Sodium-Glucose Cotransporter 2 Inhibitor

Mark Tirmenstein; Thomas E. Dorr; Evan B. Janovitz; Deborah Hagan; Lynn M. Abell; Joelle M. Onorato; Jean M. Whaley; Michael Graziano; Timothy P. Reilly

Dapagliflozin, a first-in-class, selective inhibitor of sodium-glucose cotransporter 2 (SGLT2), promotes urinary glucose excretion to reduce hyperglycemia for the treatment of type 2 diabetes. A series of nonclinical studies were undertaken to evaluate dapagliflozin in species where it was shown to have pharmacologic activity comparable with that in humans at doses that resulted in supratherapeutic exposures. In vitro screening (>300 targets; 10 μmol/L) indicated no significant off-target activities for dapagliflozin or its primary human metabolite. Once daily, orally administered dapagliflozin was evaluated in Sprague-Dawley rats (≤6 months) and in beagle dogs (≤1 year) at exposures >5000-fold those observed at the maximum recommended human clinical dose (MRHD; 10 mg). Anticipated, pharmacologically mediated effects of glucosuria, osmotic diuresis, and mild electrolyte loss were observed, but there were no adverse effects at clinically relevant exposures, including in the kidneys or urogenital tract. The SGLT2−/− mice, which show chronic glucosuria, and dapagliflozin-treated, wild-type mice exhibited similar safety profiles. In rats but not dogs, dapagliflozin at >2000-fold MRHD exposures resulted in tissue mineralization and trabecular bone accretion. Investigative studies suggested that the effect was not relevant to human safety, since it was partially related to off-target inhibition of SGLT1, which was observed only at high doses of dapagliflozin and resulted in intestinal glucose malabsorption and increased intestinal calcium absorption. The rigorous assessment of supra- and off-target dapagliflozin pharmacology in nonclinical species allowed for a thorough evaluation of potential toxicity, providing us with confidence in its safety in patients with diabetes.


Journal of Pharmacology and Experimental Therapeutics | 2011

Dissecting the Activation of Thienopyridines by Cytochromes P450 Using a Pharmacodynamic Assay In Vitro

Lynn M. Abell; Eddie C.-K. Liu

The thienopyridine antiplatelet drugs, such as ticlopidine, clopidogrel, and prasugrel, require activation by cytochromes P450 in vivo to effectively block platelet aggregation. The study of the metabolic activation of these compounds has been hampered by the lability and reactivity of the ring-opened active metabolite (AM) and by the numerous metabolites that can be formed in such a transformation. We have developed a novel method whereby platelets are incubated with the cytochrome P450 and the thienopyridine of interest for various amounts of time, and the effects on ADP-driven platelet aggregation are directly examined. In this way, the platelet is used as a biosensor for detection of the AM. Using this method, cytochromes P450 capable of converting clopidogrel, prasugrel, and 2-oxo-clopidogrel to metabolites that inhibit ADP-induced platelet aggregation were identified as well as which cytochromes P450 were capable of catalyzing partial reactions (e.g., conversion of 2-oxo-clopidogrel to the AM). These studies show that, in vitro, CYP3A4/5, 2C19, and 2B6 are individually capable of converting clopidogrel and prasugrel to the AM and that the cytochrome P450 preference for these two thienopyridines is very similar.


Journal of Biological Chemistry | 2014

Mechanism of Inhibition for BMS-791325, a Novel Non-nucleoside Inhibitor of Hepatitis C Virus NS5B Polymerase

Karen Rigat; Hao Lu; Ying-Kai Wang; Argyrides Argyrou; Caroline Fanslau; Brett R. Beno; Yi Wang; Jovita Marcinkeviciene; Min Ding; Robert G. Gentles; Min Gao; Lynn M. Abell; Susan B. Roberts

Background: BMS-791325, a non-nucleoside inhibitor of HCV NS5B, has robust clinical efficacy. Results: Biochemical and biophysical methods revealed a non-competitive time-dependent inhibition mechanism and permitted complete parameterization of inhibitor binding kinetics. Conclusion: Thumb and finger variants affect BMS-791325 association rates. Significance: The impact of NS5B variants on BMS-791325 binding provides insight into the basis of inhibitor resistance and the process of replication complex formation. HCV infection is an urgent global health problem that has triggered a drive to discover therapies that specifically target the virus. BMS-791325 is a novel direct antiviral agent specifically targeting HCV NS5B, an RNA-dependent RNA polymerase. Robust viral clearance of HCV was observed in infected patients treated with BMS-791325 in combination with other anti-HCV agents in Phase 2 clinical studies. Biochemical and biophysical studies revealed that BMS-791325 is a time-dependent, non-competitive inhibitor of the polymerase. Binding studies with NS5B genetic variants (WT, L30S, and P495L) exposed a two-step, slow binding mechanism, but details of the binding mechanism differed for each of the polymerase variants. For the clinically relevant resistance variant (P495L), the rate of initial complex formation and dissociation is similar to WT, but the kinetics of the second step is significantly faster, showing that this variant impacts the final tight complex. The resulting shortened residence time translates into the observed decrease in inhibitor potency. The L30S variant has a significantly different profile. The rate of initial complex formation and dissociation is 7–10 times faster for the L30S variant compared with WT; however, the forward and reverse rates to form the final complex are not significantly different. The impact of the L30S variant on the inhibition profile and binding kinetics of BMS-791325 provides experimental evidence for the dynamic interaction of fingers and thumb domains in an environment that supports the formation of active replication complexes and the initiation of RNA synthesis.


Analytical Biochemistry | 2016

Development of a RapidFire mass spectrometry assay and a fluorescence assay for the discovery of kynurenine aminotransferase II inhibitors to treat central nervous system disorders.

Hao Lu; Lisa M. Kopcho; Kaushik Ghosh; Mark R. Witmer; Michael F. Parker; Sumit Gupta; Marilyn Paul; Prasad Krishnamurthy; Basanth Laksmaiah; Dianlin Xie; Jeffrey Tredup; Litao Zhang; Lynn M. Abell

Kynurenine aminotransferases convert kynurenine to kynurenic acid and play an important role in the tryptophan degradation pathway. Kynurenic acid levels in brain have been hypothesized to be linked to a number of central nervous system (CNS) disorders. Kynurenine aminotransferase II (KATII) has proven to be a key modulator of kynurenic acid levels in brain and, thus, is an attractive target to treat CNS diseases. A sensitive, high-throughput, label-free RapidFire mass spectrometry assay has been developed for human KATII. Unlike other assays, this method is directly applicable to KATII enzymes from different animal species, which allows us to select proper animal model(s) to evaluate human KATII inhibitors. We also established a coupled fluorescence assay for human KATII. The short assay time and kinetic capability of the fluorescence assay provide a useful tool for orthogonal inhibitor validation and mechanistic studies.


Bioorganic & Medicinal Chemistry Letters | 2018

Discovery and synthesis of tetrahydropyrimidinedione-4-carboxamides as endothelial lipase inhibitors

Carol Hui Hu; Tammy C. Wang; Jennifer X. Qiao; Lauren Haque; Alice Y.A. Chen; David S. Taylor; Xiaohong Ying; Joelle M. Onorato; Michael A. Galella; Hong Shen; Christine Huang; Nathalie Toussaint; Yi-Xin Li; Lynn M. Abell; Leonard P. Adam; David A. Gordon; Ruth R. Wexler; Heather Finlay

Endothelial lipase (EL) inhibitors have been shown to elevate HDL-C levels in pre-clinical murine models and have potential benefit in prevention and treatment of cardiovascular diseases. Modification of the 1-ethyl-3-hydroxy-1,5-dihydro-2H-pyrrol-2-one (DHP) lead, 1, led to the discovery of a series of potent tetrahydropyrimidinedione (THP) EL inhibitors. Synthesis and SAR studies including modification of the amide group, together with changes on the pyrimidinone core led to a series of arylcycloalkyl, indanyl, and tetralinyl substituted 5-amino or 5-hydroxypyrimidinedione-4-carboxamides. Several compounds were advanced to PK evaluation. Among them, compound 4a was one of the most potent with measurable ELHDL hSerum potency and compound 3g demonstrated the best overall pharmacokinetic parameters.


ACS Medicinal Chemistry Letters | 2018

PK/PD Disconnect Observed with a Reversible Endothelial Lipase Inhibitor

Jon J. Hangeland; Lynn M. Abell; Leonard P. Adam; Ji Jiang; Todd J. Friends; Lauren Haque; James Neels; Joelle M. Onorato; Alice Ye A. Chen; David S. Taylor; Xiaohong Yin; Thomas Harrity; Michael D. Basso; Richard Yang; Paul G. Sleph; David A. Gordon; Christine Huang; Ruth R. Wexler; Heather Finlay; R. Michael Lawrence

Screening of a small set of nonselective lipase inhibitors against endothelial lipase (EL) identified a potent and reversible inhibitor, N-(3-(3,4-dichlorophenyl)propyl)-3-hydroxy-1-methyl-2-oxo-1,2-dihydropyridine-4-carboxamide (5; EL IC50 = 61 nM, ELHDL IC50 = 454 nM). Deck mining identified a related hit, N-(3-(3,4-dichlorophenyl)propyl)-4-hydroxy-1-methyl-5-oxo-2,5-dihydro-1H-pyrrole-3-carboxamide (6a; EL IC50 = 41 nM, ELHDL IC50 = 1760 nM). Both compounds were selective against lipoprotein lipase (LPL) but nonselective versus hepatic lipase (HL). Optimization of compound 6a for EL inhibition using HDL as substrate led to N-(4-(3,4-dichlorophenyl)butan-2-yl)-1-ethyl-4-hydroxy-5-oxo-2,5-dihydro-1H-pyrrole-3-carboxamide (7c; EL IC50 = 148 nM, ELHDL IC50 = 218 nM) having improved PK over compound 6a, providing a tool molecule to test for the ability to increase HDL-cholesterol (HDL-C) levels in vivo using a reversible EL inhibitor. Compound 7c did not increase HDL-C in vivo despite achieving plasma exposures targeted on the basis of enzyme activity and protein binding demonstrating the need to develop more physiologically relevant in vitro assays to guide compound progression for in vivo evaluation.


Analytical Biochemistry | 2006

Development and validation of a platelet calcium flux assay using a fluorescent imaging plate reader.

Eddie C.-K. Liu; Lynn M. Abell


Archive | 2012

Pyrrolinone carboxamide compounds useful as endothelial lipase inhibitors

Lynn M. Abell; Leonard P. Adam; Cullen L. Cavallaro; Heather Finlay; Todd J. Friends; Jon J. Hangeland; Ji Jiang; R. Michael Lawrence; Jennifer X. Qiao; Carol Hui Hu; Tammy C. Wang

Collaboration


Dive into the Lynn M. Abell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ji Jiang

Bristol-Myers Squibb

View shared research outputs
Researchain Logo
Decentralizing Knowledge