Network


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

Hotspot


Dive into the research topics where Paul D. Bonin is active.

Publication


Featured researches published by Paul D. Bonin.


Bioorganic & Medicinal Chemistry Letters | 2009

Synthesis and SAR of 1,2,3,4-tetrahydroisoquinolin-1-ones as novel G-protein-coupled receptor 40 (GPR40) antagonists.

Paul S. Humphries; John William Benbow; Paul D. Bonin; David Boyer; Shawn D. Doran; Richard K. Frisbie; David W. Piotrowski; Gayatri Balan; Bruce M. Bechle; Edward L. Conn; Kenneth J. DiRico; Robert M. Oliver; Walter C. Soeller; James A. Southers; Xiaojing Yang

The development of a series of novel 1,2,3,4-tetrahydroisoquinolin-1-ones as antagonists of G protein-coupled receptor 40 (GPR40) is described. The synthesis, in vitro inhibitory values for GPR40, in vitro microsomal clearance and rat in vivo clearance data are discussed. Initial hits displayed high rat in vivo clearances that were higher than liver blood flow. Optimization of rat in vivo clearance was achieved and led to the identification of 15i, whose rat oral pharmacokinetic data is reported.


Journal of Medicinal Chemistry | 2011

Activation of the G-protein-coupled receptor 119: a conformation-based hypothesis for understanding agonist response.

Kim F. McClure; Etzer Darout; Cristiano R. W. Guimarães; Michael Paul Deninno; Vincent Mascitti; Michael John Munchhof; Ralph P. Robinson; Jeffrey T. Kohrt; Anthony R. Harris; Dianna E. Moore; Bryan Li; Lacey Samp; Bruce Allen Lefker; Kentaro Futatsugi; Daniel Kung; Paul D. Bonin; Peter Cornelius; Ruduan Wang; Eben Salter; Sam Hornby; Amit S. Kalgutkar; Yue Chen

The synthesis and properties of the bridged piperidine (oxaazabicyclo) compounds 8, 9, and 11 are described. A conformational analysis of these structures is compared with the representative GPR119 ligand 1. These results and the differences in agonist pharmacology are used to formulate a conformation-based hypothesis to understand activation of the GPR119 receptor. We also show for these structures that the agonist pharmacology in rat masks the important differences in human pharmacology.


Journal of Immunology | 2016

Efficacy and Pharmacology of the NLRP3 Inflammasome Inhibitor CP-456,773 (CRID3) in Murine Models of Dermal and Pulmonary Inflammation

Primiano Mj; Lefker Ba; Bowman Mr; Bree Ag; Hubeau C; Paul D. Bonin; Matthew Mangan; Dower K; Brian G. Monks; Cushing L; Wang S; Guzova J; Jiao A; Lih-Ling Lin; Eicke Latz; David Hepworth; Hall Jp

A critical component of innate immune response to infection and tissue damage is the NACHT, LRR, and PYD domains–containing protein 3 (NLRP3) inflammasome, and this pathway and its activation products have been implicated in the pathophysiology of a variety of diseases. NLRP3 inflammasome activation leads to the cleavage of pro–IL-1β and pro–IL-18, as well as the subsequent release of biologically active IL-1β, IL-18, and other soluble mediators of inflammation. In this study, we further define the pharmacology of the previously reported NLRP3 inflammasome–selective, IL-1β processing inhibitor CP-456,773 (also known as MCC950), and we demonstrate its efficacy in two in vivo models of inflammation. Specifically, we show that in human and mouse innate immune cells CP-456,773 is an inhibitor of the cellular release of IL-1β, IL-1α, and IL-18, that CP-456,773 prevents inflammasome activation induced by disease-relevant soluble and crystalline NLRP3 stimuli, and that CP-456,773 inhibits R848- and imiquimod-induced IL-1β release. In mice, CP-456,773 demonstrates potent inhibition of the release of proinflammatory cytokines following acute i.p. challenge with LPS plus ATP in a manner that is proportional to the free/unbound concentrations of the drug, thereby establishing an in vivo pharmacokinetic/pharmacodynamic model for CP-456,773. Furthermore, CP-456,773 reduces ear swelling in an imiquimod cream–induced mouse model of skin inflammation, and it reduces airway inflammation in mice following acute challenge with house dust mite extract. These data implicate the NLRP3 inflammasome in the pathogenesis of dermal and airway inflammation, and they highlight the utility of CP-456,773 for interrogating the contribution of the NLRP3 inflammasome and its outputs in preclinical models of inflammation and disease.


Journal of Pharmacology and Experimental Therapeutics | 2015

PF-1355, a Mechanism-Based Myeloperoxidase Inhibitor, Prevents Immune Complex Vasculitis and Anti–Glomerular Basement Membrane Glomerulonephritis

Wei Zheng; Roscoe L. Warner; Roger Benjamin Ruggeri; Chunyan Su; Christian Cortes; Athanasia Skoura; Jessica Ward; Kay Ahn; Amit S. Kalgutkar; Dexue Sun; Tristan S. Maurer; Paul D. Bonin; Carlin Okerberg; Walter F. Bobrowski; Thomas T. Kawabe; Yanwei Zhang; Timothy M. Coskran; Sammy Bell; Bhupesh Kapoor; Kent J. Johnson; Leonard Buckbinder

Small vessel vasculitis is a life-threatening condition and patients typically present with renal and pulmonary injury. Disease pathogenesis is associated with neutrophil accumulation, activation, and oxidative damage, the latter being driven in large part by myeloperoxidase (MPO), which generates hypochlorous acid among other oxidants. MPO has been associated with vasculitis, disseminated vascular inflammation typically involving pulmonary and renal microvasculature and often resulting in critical consequences. MPO contributes to vascular injury by 1) catabolizing nitric oxide, impairing vasomotor function; 2) causing oxidative damage to lipoproteins and endothelial cells, leading to atherosclerosis; and 3) stimulating formation of neutrophil extracellular traps, resulting in vessel occlusion and thrombosis. Here we report a selective 2-thiouracil mechanism-based MPO inhibitor (PF-1355 [2-(6-(2,5-dimethoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide) and demonstrate that MPO is a critical mediator of vasculitis in mouse disease models. A pharmacokinetic/pharmacodynamic response model of PF-1355 exposure in relation with MPO activity was derived from mouse peritonitis. The contribution of MPO activity to vasculitis was then examined in an immune complex model of pulmonary disease. Oral administration of PF-1355 reduced plasma MPO activity, vascular edema, neutrophil recruitment, and elevated circulating cytokines. In a model of anti–glomerular basement membrane disease, formerly known as Goodpasture disease, albuminuria and chronic renal dysfunction were completely suppressed by PF-1355 treatment. This study shows that MPO activity is critical in driving immune complex vasculitis and provides confidence in testing the hypothesis that MPO inhibition will provide benefit in treating human vasculitic diseases.


Chemistry & Biology | 2015

Transcriptional Profiling of a Selective CREB Binding Protein Bromodomain Inhibitor Highlights Therapeutic Opportunities.

Eugene Lvovich Piatnitski Chekler; Jessica A. Pellegrino; Thomas A. Lanz; R. Aldrin Denny; Andrew C. Flick; Jotham Wadsworth Coe; Jonathan Langille; Arindrajit Basak; Shenping Liu; Ingrid A. Stock; Parag Sahasrabudhe; Paul D. Bonin; Kevin Lee; Mathew T. Pletcher; Lyn H. Jones

Bromodomains are involved in transcriptional regulation through the recognition of acetyl lysine modifications on diverse proteins. Selective pharmacological modulators of bromodomains are lacking, although the largely hydrophobic nature of the pocket makes these modules attractive targets for small-molecule inhibitors. This work describes the structure-based design of a highly selective inhibitor of the CREB binding protein (CBP) bromodomain and its use in cell-based transcriptional profiling experiments. The inhibitor downregulated a number of inflammatory genes in macrophages that were not affected by a selective BET bromodomain inhibitor. In addition, the CBP bromodomain inhibitor modulated the mRNA level of the regulator of G-protein signaling 4 (RGS4) gene in neurons, suggesting a potential therapeutic opportunity for CBP inhibitors in the treatment of neurological disorders.


Bioorganic & Medicinal Chemistry Letters | 2011

Design and evaluation of a 2-(2,3,6-trifluorophenyl)acetamide derivative as an agonist of the GPR119 receptor.

Vincent Mascitti; Benjamin D. Stevens; Chulho Choi; Kim F. McClure; Cristiano R. W. Guimarães; Kathleen A. Farley; Michael John Munchhof; Ralph P. Robinson; Kentaro Futatsugi; Sophie Y. Lavergne; Bruce Allen Lefker; Peter Cornelius; Paul D. Bonin; Amit S. Kalgutkar; Raman Sharma; Yue Chen

The design and synthesis of a GPR119 agonist bearing a 2-(2,3,6-trifluorophenyl)acetamide group is described. The design capitalized on the conformational restriction found in N-β-fluoroethylamide derivatives to help maintain good levels of potency while driving down both lipophilicity and oxidative metabolism in human liver microsomes. The chemical stability and bioactivation potential are discussed.


Chemical Research in Toxicology | 2011

Oxidative metabolism of a quinoxaline derivative by xanthine oxidase in rodent plasma.

Raman Sharma; Heather Eng; Gregory S. Walker; Gabriela Barreiro; Antonia F. Stepan; Kim F. McClure; Angela Wolford; Paul D. Bonin; Peter Cornelius; Amit S. Kalgutkar

As part of efforts directed at the G protein-coupled receptor 119 agonist program for type 2 diabetes, a series of cyanopyridine derivatives exemplified by isopropyl-4-(3-cyano-5-(quinoxalin-6-yl)pyridine-2-yl)piperazine-1-carboxylate (1) were identified as novel chemotypes worthy of further hit-to-lead optimization. Compound 1, however, was found to be unstable in plasma (37 °C, pH 7.4) from rat (T(1/2) = 16 min), mouse (T(1/2) = 61 min), and guinea pig (T(1/2) = 4 min). Lowering the temperature of plasma incubations (4-25 °C) attenuated the degradation of 1, implicating the involvement of an enzyme-mediated process. Failure to detect any appreciable amount of 1 in plasma samples from protein binding and pharmacokinetic studies in rats was consistent with its labile nature in plasma. Instability noted in rodent plasma was not observed in plasma from dogs, monkeys, and humans (T(1/2) > 370 min at 37 °C, pH 7.4). Metabolite identification studies in rodent plasma revealed the formation of a single metabolite (M1), which was 16 Da higher than the molecular weight of 1 (compound 1, MH(+) = 403; M1, MH(+) = 419). Pretreatment of rat plasma with allopurinol, but not raloxifene, abolished the conversion of 1 to M1, suggesting that xanthine oxidase (XO) was responsible for the oxidative instability. Consistent with the known catalytic mechanism of XO, the source of oxygen incorporated in M1 was derived from water rather than molecular oxygen. The formation of M1 was also demonstrated in incubations of 1 with purified bovine XO. The structure of M1 was determined by NMR analysis to be isopropyl-4-(3-cyano-5-(3-oxo-3,4-dihydroquinoxalin-6-yl)pyridine-2-yl)piperazine-1-carboxylate. The regiochemistry of quinoxaline ring oxidation in 1 was consistent with ab initio calculations and molecular docking studies using a published crystal structure of bovine XO. A close-in analogue of 1, which lacked the quinoxaline motif (e.g., 5-(4-cyano-3-methylphenyl)-2-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperidin-1-yl)nicotinitrile (2)) was stable in rat plasma and possessed substantially improved GPR119 agonist properties. To the best of our knowledge, our studies constitute the first report on the involvement of rodent XO in oxidative drug metabolism in plasma.


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.


Bioorganic & Medicinal Chemistry Letters | 2013

From partial to full agonism: Identification of a novel 2,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole as a full agonist of the human GPR119 receptor

Kentaro Futatsugi; Vincent Mascitti; Cristiano R. W. Guimarães; Nao Morishita; Cuiman Cai; Michael Paul Deninno; Hua Gao; Michael Hamilton; Richard F. Hank; Anthony R. Harris; Daniel W. Kung; Sophie Y. Lavergne; Bruce Allen Lefker; Michael G. Lopaze; Kim F. McClure; Michael John Munchhof; Cathy Préville; Ralph P. Robinson; Stephen W. Wright; Paul D. Bonin; Peter Cornelius; Yue Chen; Amit S. Kalgutkar

A novel GPR119 agonist based on the 2,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole scaffold was designed through lead optimization starting from pyrazole-based GPR119 agonist 1. The design is centered on the conformational restriction of the core scaffold, while minimizing the change in spatial relationships of two key pharmacophoric elements (piperidine-carbamate and aryl sulfone).


MedChemComm | 2014

Design and chemoproteomic functional characterization of a chemical probe targeted to bromodomains of BET family proteins

Jiang Wu; Julia Shin; Cara Williams; Kieran F. Geoghegan; Stephen W. Wright; David Limburg; Parag Sahasrabudhe; Paul D. Bonin; Bruce Allen Lefker; Simeon Ramsey

Bromodomain-containing proteins form the signal-reading element of a principal system for the control of gene expression in eukaryotes. Their potential as targets for selective drug action is increasingly being assessed and exploited. Deep characterization of the specificity, potency and other attributes of prototypical agents is an essential element of this process. Continuing studies of a dihydroquinazolinone-based series (prototype: PFI-1) with specificity for members of the BET (bromodomain and extra terminal) family led to the discovery of quinolin-2(1H)-one inhibitors with similar potency and selectivity, but increased chemical stability. Structure-guided design then led to the elaboration of a desthiobiotinylated analog retaining a high fraction of the potency of its parent compound and therefore suitable for chemoproteomic affinity capture experiments. These experiments, conducted using nuclear extracts of THP-1 cells, extended confidence in the selectivity of the series as first proposed. An additional and subsequent evaluation of specificity performed with a panel of recombinant bromodomains (BROMOscan™, DiscoveRx) supported the BET family specificity of the dihydroquinazolinone and quinolin-2(1H)-one series while adding appreciation of weaker effects shown at other bromodomains.

Researchain Logo
Decentralizing Knowledge