Network


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

Hotspot


Dive into the research topics where Scott W. Bagley is active.

Publication


Featured researches published by Scott W. Bagley.


Angewandte Chemie | 2014

Application of Fundamental Organometallic Chemistry to the Development of a Gold‐Catalyzed Synthesis of Sulfinate Derivatives

Miles W. Johnson; Scott W. Bagley; Neal P. Mankad; Robert G. Bergman; Vincent Mascitti; F. Dean Toste

The development of a gold(I)-catalyzed sulfination of aryl boronic acids is described. This transformation proceeds through an unprecedented mechanism which exploits the reactivity of gold(I)-heteroatom bonds to form sulfinate anions. Further in situ elaboration of the sulfinate intermediates leads to the corresponding sulfones and sulfonamides, two pharmacophores routinely encountered in drug discovery.


Journal of Medicinal Chemistry | 2014

Decreasing the Rate of Metabolic Ketone Reduction in the Discovery of a Clinical Acetyl-CoA Carboxylase Inhibitor for the Treatment of Diabetes

David A. Griffith; Daniel W. Kung; William Esler; Paul Amor; Scott W. Bagley; Carine Beysen; Santos Carvajal-Gonzalez; Shawn D. Doran; Chris Limberakis; Alan M. Mathiowetz; Kirk McPherson; David A. Price; Eric Ravussin; Gabriele Sonnenberg; James A. Southers; Laurel Sweet; Scott M. Turner; Felix Vajdos

Acetyl-CoA carboxylase (ACC) inhibitors offer significant potential for the treatment of type 2 diabetes mellitus (T2DM), hepatic steatosis, and cancer. However, the identification of tool compounds suitable to test the hypothesis in human trials has been challenging. An advanced series of spirocyclic ketone-containing ACC inhibitors recently reported by Pfizer were metabolized in vivo by ketone reduction, which complicated human pharmacology projections. We disclose that this metabolic reduction can be greatly attenuated through introduction of steric hindrance adjacent to the ketone carbonyl. Incorporation of weakly basic functionality improved solubility and led to the identification of 9 as a clinical candidate for the treatment of T2DM. Phase I clinical studies demonstrated dose-proportional increases in exposure, single-dose inhibition of de novo lipogenesis (DNL), and changes in indirect calorimetry consistent with increased whole-body fatty acid oxidation. This demonstration of target engagement validates the use of compound 9 to evaluate the role of DNL in human disease.


Journal of Medicinal Chemistry | 2015

Discovery of 2-(6-(5-Chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide (PF-06282999): A Highly Selective Mechanism-Based Myeloperoxidase Inhibitor for the Treatment of Cardiovascular Diseases.

Roger Benjamin Ruggeri; Leonard Buckbinder; Scott W. Bagley; Philip A. Carpino; Edward L. Conn; Matthew S. Dowling; Dilinie P. Fernando; Wenhua Jiao; Daniel W. Kung; Suvi T. M. Orr; Yingmei Qi; Benjamin N. Rocke; Aaron Smith; Joseph Scott Warmus; Yan Zhang; Daniel Bowles; Daniel W. Widlicka; Heather Eng; Tim Ryder; Raman Sharma; Angela Wolford; Carlin Okerberg; Karen Walters; Tristan S. Maurer; Yanwei Zhang; Paul D. Bonin; Samantha N. Spath; Gang Xing; David Hepworth; Kay Ahn

Myeloperoxidase (MPO) is a heme peroxidase that catalyzes the production of hypochlorous acid. Clinical evidence suggests a causal role for MPO in various autoimmune and inflammatory disorders including vasculitis and cardiovascular and Parkinsons diseases, implying that MPO inhibitors may represent a therapeutic treatment option. Herein, we present the design, synthesis, and preclinical evaluation of N1-substituted-6-arylthiouracils as potent and selective inhibitors of MPO. Inhibition proceeded in a time-dependent manner by a covalent, irreversible mechanism, which was dependent upon MPO catalysis, consistent with mechanism-based inactivation. N1-Substituted-6-arylthiouracils exhibited low partition ratios and high selectivity for MPO over thyroid peroxidase and cytochrome P450 isoforms. N1-Substituted-6-arylthiouracils also demonstrated inhibition of MPO activity in lipopolysaccharide-stimulated human whole blood. Robust inhibition of plasma MPO activity was demonstrated with the lead compound 2-(6-(5-chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide (PF-06282999, 8) upon oral administration to lipopolysaccharide-treated cynomolgus monkeys. On the basis of its pharmacological and pharmacokinetic profile, PF-06282999 has been advanced to first-in-human pharmacokinetic and safety studies.


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 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

The synthesis of 4,6-dihydrospiro[piperidine-4,5-pyrazolo[3,4-c]pyridin]-7(2H)-one-based acetyl-CoA carboxylase inhibitors is reported. The hitherto unknown N-2 tert-butyl pyrazolospirolactam core was synthesized from ethyl 3-amino-1H-pyrazole-4-carboxylate in a streamlined 10-step synthesis requiring only one chromatography procedure. The described synthetic strategy provides pyrazolo-fused spirolactams from halogenated benzylic arenes and cyclic carboxylates. Key steps include a regioselective pyrazole alkylation providing the N-2 tert-butyl pyrazole and a Curtius rearrangement under both conventional and flow conditions to install the hindered amine via a stable and isolable isocyanate. Finally, a Parham-type cyclization was used to furnish the desired spirolactam. An analogous route provided efficient access to the related N-1 isopropyl lactam series. Elaboration of the lactam cores via amidation enabled synthesis of novel ACC inhibitors and the identification of potent analogues.


Chemical Research in Toxicology | 2010

Discovery Tactics To Mitigate Toxicity Risks Due to Reactive Metabolite Formation with 2-(2-Hydroxyaryl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one Derivatives, Potent Calcium-Sensing Receptor Antagonists and Clinical Candidate(s) for the Treatment of Osteoporosis

Amit S. Kalgutkar; David A. Griffith; Tim Ryder; Hao Sun; Zhuang Miao; Jonathan N. Bauman; Mary Theresa Didiuk; Kosea S. Frederick; Sabrina X. Zhao; Chandra Prakash; John R. Soglia; Scott W. Bagley; Bruce M. Bechle; Ryan M. Kelley; Kenneth J. DiRico; Michael P. Zawistoski; Jianke Li; Robert M. Oliver; Angel Guzman-Perez; Kevin K.-C. Liu; Daniel P. Walker; John William Benbow; Joel Morris

The synthesis and structure-activity relationship studies on 5-trifluoromethylpyrido[4,3-d]pyrimidin-4(3H)-ones as antagonists of the human calcium receptor (CaSR) have been recently disclosed [ Didiuk et al. ( 2009 ) Bioorg. Med. Chem. Lett. 19 , 4555 - 4559 ). On the basis of its pharmacology and disposition attributes, (R)-2-(2-hydroxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one (1) was considered for rapid advancement to first-in-human (FIH) trials to mitigate uncertainty surrounding the pharmacokinetic/pharmacodynamic (PK/PD) predictions for a short-acting bone anabolic agent. During the course of metabolic profiling, however, glutathione (GSH) conjugates of 1 were detected in human liver microsomes in an NADPH-dependent fashion. Characterization of the GSH conjugate structures allowed insight(s) into the bioactivation pathway, which involved CYP3A4-mediated phenol ring oxidation to the catechol, followed by further oxidation to the electrophilic ortho-quinone species. While the reactive metabolite (RM) liability raised concerns around the likelihood of a potential toxicological outcome, a more immediate program goal was establishing confidence in human PK predictions in the FIH study. Furthermore, the availability of a clinical biomarker (serum parathyroid hormone) meant that PD could be assessed side by side with PK, an ideal scenario for a relatively unprecedented pharmacologic target. Consequently, progressing 1 into the clinic was given a high priority, provided the compound demonstrated an adequate safety profile to support FIH studies. Despite forming identical RMs in rat liver microsomes, no clinical or histopathological signs prototypical of target organ toxicity were observed with 1 in in vivo safety assessments in rats. Compound 1 was also devoid of metabolism-based mutagenicity in in vitro (e.g., Salmonella Ames) and in vivo assessments (micronuclei induction in bone marrow) in rats. Likewise, metabolism-based studies (e.g., evaluation of detoxicating routes of clearance and exhaustive PK/PD studies in animals to prospectively predict the likelihood of a low human efficacious dose) were also conducted, which mitigated the risks of idiosyncratic toxicity to a large degree. In parallel, medicinal chemistry efforts were initiated to identify additional compounds with a complementary range of human PK predictions, which would maximize the likelihood of achieving the desired PD effect in the clinic. The back-up strategy also incorporated an overarching goal of reducing/eliminating reactive metabolite formation observed with 1. Herein, the collective findings from our discovery efforts in the CaSR program, which include the incorporation of appropriate derisking steps when dealing with RM issues are summarized.


Molecular Pharmacology | 2009

Molecular Characterization of Novel and Selective Peroxisome Proliferator-Activated Receptor α Agonists with Robust Hypolipidemic Activity in Vivo

Christopher D. Kane; Kimberly A. Stevens; James E Fischer; Mehrdad Haghpassand; Lori Royer; Charles E. Aldinger; Katherine T. Landschulz; Panayiotis Zagouras; Scott W. Bagley; William A. Hada; Robert Dullea; Cheryl Myers Hayward; Omar L. Francone

The nuclear receptor peroxisome proliferator-activated receptor α (PPARα) is recognized as the primary target of the fibrate class of hypolipidemic drugs and mediates lipid lowering in part by activating a transcriptional cascade that induces genes involved in the catabolism of lipids. We report here the characterization of three novel PPARα agonists with therapeutic potential for treating dyslipidemia. These structurally related compounds display potent and selective binding to human PPARα and support robust recruitment of coactivator peptides in vitro. These compounds markedly potentiate chimeric transcription systems in cell-based assays and strikingly lower serum triglycerides in vivo. The transcription networks induced by these selective PPARα agonists were assessed by transcriptional profiling of mouse liver after short- and long-term treatment. The induction of several known PPARα target genes involved with fatty acid metabolism were observed, reflecting the expected pharmacology associated with PPARα activation. We also noted the down-regulation of a number of genes related to immune cell function, the acute phase response, and glucose metabolism, suggesting that these compounds may have anti-inflammatory action in the mammalian liver. Whereas these compounds are efficacious in acute preclinical models, extended safety studies and further clinical testing will be required before the full therapeutic promise of a selective PPARα agonist is realized.


Journal of Organic Chemistry | 2012

Synthesis of 7-Oxo-dihydrospiro[indazole-5,4′-piperidine] Acetyl-CoA Carboxylase Inhibitors

Scott W. Bagley; James A. Southers; Shawn Cabral; Colin R. Rose; David J. Bernhardson; David J. Edmonds; Jana Polivkova; Xiaojing Yang; Daniel W. Kung; David A. Griffith; Scott Bader

Synthesis of oxo-dihydrospiroindazole-based acetyl-CoA carboxylase (ACC) inhibitors is reported. The dihydrospiroindazoles were assembled in a regioselective manner in six steps from substituted hydrazines and protected 4-formylpiperidine. Enhanced regioselectivity in the condensation between a keto enamine and substituted hydrazines was observed when using toluene as the solvent, leading to selective formation of 1-substituted spiroindazoles. The 2-substituted spiroindazoles were formed selectively from alkyl hydrazones by ring closure with Vilsmeier reagent. The key step in the elaboration to the final products is the conversion of an intermediate olefin to the desired ketone through elimination of HBr from an O-methyl bromohydrin. This methodology enabled the synthesis of each desired regioisomer on 50-75 g scale with minimal purification. Acylation of the resultant spirocyclic amines provided potent ACC inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2009

Short-acting 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as orally-active calcium-sensing receptor antagonists.

Mary Theresa Didiuk; David A. Griffith; John William Benbow; Kevin K.-C. Liu; Daniel P. Walker; F. Christopher Bi; Joel Morris; Angel Guzman-Perez; Hua Gao; Bruce M. Bechle; Ryan M. Kelley; Xiaojing Yang; Kenneth J. DiRico; Syed Ahmed; William M. Hungerford; Joseph DiBrinno; Michael P. Zawistoski; Scott W. Bagley; Jianke Li; Yuan Zeng; Stephanie Santucci; Robert M. Oliver; Matthew Corbett; Thanh V. Olson; Chiliu Chen; Mei Li; Vishwas M. Paralkar; Keith Riccardi; David R. Healy; Amit S. Kalgutkar

Synthesis and structure-activity relationship (SAR) studies on 5-trifluoromethylpyrido[4,3-d]pyrimidin-4(3H)-ones, a novel class of calcium receptor antagonists is described with particular emphasis on optimization of the pharmacokinetic/pharmacodynamic parameters required for a short duration of action compound. Orally-active compounds were identified which displayed the desired animal pharmacology (rapid and transient stimulation of parathyroid hormone) essential for bone anabolic effects.


ACS Medicinal Chemistry Letters | 2015

Discovery of an in Vivo Tool to Establish Proof-of-Concept for MAP4K4-Based Antidiabetic Treatment

Mark Ammirati; Scott W. Bagley; Samit Kumar Bhattacharya; Leonard Buckbinder; Anthony A. Carlo; Rebecca Conrad; Christian Cortes; Robert L. Dow; Matthew S. Dowling; Ayman El-Kattan; Kristen Ford; Cristiano R. W. Guimarães; David Hepworth; Wenhua Jiao; Jennifer L. LaPerle; Shenping Liu; Allyn T. Londregan; Paula M. Loria; Alan M. Mathiowetz; Michael John Munchhof; Suvi T. M. Orr; Donna N. Petersen; David A. Price; Athanasia Skoura; Aaron Smith; Jian Wang

Recent studies in adipose tissue, pancreas, muscle, and macrophages suggest that MAP4K4, a serine/threonine protein kinase may be a viable target for antidiabetic drugs. As part of the evaluation of MAP4K4 as a novel antidiabetic target, a tool compound, 16 (PF-6260933) and a lead 17 possessing excellent kinome selectivity and suitable properties were delivered to establish proof of concept in vivo. The medicinal chemistry effort that led to the discovery of these lead compounds is described herein together with in vivo pharmacokinetic properties and activity in a model of insulin resistance.

Researchain Logo
Decentralizing Knowledge