Network


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

Hotspot


Dive into the research topics where Stephen T. Wrobleski is active.

Publication


Featured researches published by Stephen T. Wrobleski.


Current Topics in Medicinal Chemistry | 2005

Structural comparison of p38 inhibitor-protein complexes: a review of recent p38 inhibitors having unique binding interactions.

Stephen T. Wrobleski; Arthur M. Doweyko

Small molecule inhibition of protein kinases in the treatment of significant diseases such as cancer, Alzheimers disease, diabetes, and rheumatoid arthritis has attracted significant attention over the past two decades and has clearly become one of the most significant challenges for drug discovery in the 21st century. While the recent identification of 518 different kinases in the human genome has offered a wealth of opportunities for drug intervention in the treatment of these diseases, it has also created a daunting challenge with respect to selective kinase inhibition as a viable strategy in target-based drug design. Over the past decade, the design and development of a small molecule that selectively inhibits the p38 mitogen activated protein (MAP) kinase has clearly emerged as one of these challenges within the industry. This review will focus on the comparison of the x-ray crystal structures and binding models of the most recent p38 inhibitor-enzyme complexes and the identification of the structural elements and interactions that may be important in providing inhibitor potency and selectivity toward the p38 MAP kinase.


Journal of Medicinal Chemistry | 2010

Discovery of 4-(5-(Cyclopropylcarbamoyl)-2-methylphenylamino)-5-methyl-N-propylpyrrolo[1,2-f][1,2,4]triazine-6-carboxamide (BMS-582949), a Clinical p38α MAP Kinase Inhibitor for the Treatment of Inflammatory Diseases

Chunjian Liu; James Lin; Stephen T. Wrobleski; Shuqun Lin; John Hynes; Hong Wu; Alaric J. Dyckman; Tianle Li; John Wityak; Kathleen M. Gillooly; Sidney Pitt; Ding Ren Shen; Rosemary Zhang; Kim W. McIntyre; Luisa Salter-Cid; David J. Shuster; Hongjian Zhang; Punit Marathe; Arthur M. Doweyko; John S. Sack; Susan E. Kiefer; Kevin Kish; John A. Newitt; Murray McKinnon; John H. Dodd; Joel C. Barrish; Gary L. Schieven; Katerina Leftheris

The discovery and characterization of 7k (BMS-582949), a highly selective p38α MAP kinase inhibitor that is currently in phase II clinical trials for the treatment of rheumatoid arthritis, is described. A key to the discovery was the rational substitution of N-cyclopropyl for N-methoxy in 1a, a previously reported clinical candidate p38α inhibitor. Unlike alkyl and other cycloalkyls, the sp(2) character of the cyclopropyl group can confer improved H-bonding characteristics to the directly substituted amide NH. Inhibitor 7k is slightly less active than 1a in the p38α enzymatic assay but displays a superior pharmacokinetic profile and, as such, was more effective in both the acute murine model of inflammation and pseudoestablished rat AA model. The binding mode of 7k with p38α was confirmed by X-ray crystallographic analysis.


Bioorganic & Medicinal Chemistry Letters | 2008

Synthesis and SAR of new pyrrolo[2,1-f][1,2,4]triazines as potent p38α MAP kinase inhibitors

Stephen T. Wrobleski; Shuqun Lin; John Hynes; Hong Wu; Sidney Pitt; Ding Ren Shen; Rosemary Zhang; Kathleen M. Gillooly; David J. Shuster; Kim W. McIntyre; Arthur M. Doweyko; Kevin Kish; Jeffrey Tredup; Gerald J. Duke; John S. Sack; Murray McKinnon; John H. Dodd; Joel C. Barrish; Gary L. Schieven; Katerina Leftheris

A novel series of compounds based on the pyrrolo[2,1-f][1,2,4]triazine ring system have been identified as potent p38 alpha MAP kinase inhibitors. The synthesis, structure-activity relationships (SAR), and in vivo activity of selected analogs from this class of inhibitors are reported. Additional studies based on X-ray co-crystallography have revealed that one of the potent inhibitors from this series binds to the DFG-out conformation of the p38 alpha enzyme.


Bioorganic & Medicinal Chemistry Letters | 2010

Utilization of a nitrogen–sulfur nonbonding interaction in the design of new 2-aminothiazol-5-yl-pyrimidines as p38α MAP kinase inhibitors

Shuqun Lin; Stephen T. Wrobleski; John Hynes; Sidney Pitt; Rosemary Zhang; Yi Fan; Arthur M. Doweyko; Kevin Kish; John S. Sack; Mary F. Malley; Susan E. Kiefer; John A. Newitt; Murray McKinnon; James M. Trzaskos; Joel C. Barrish; John H. Dodd; Gary L. Schieven; Katerina Leftheris

The design, synthesis, and structure-activity relationships (SAR) of a series of 2-aminothiazol-5-yl-pyrimidines as novel p38α MAP kinase inhibitors are described. These efforts led to the identification of 41 as a potent p38α inhibitor that utilizes a unique nitrogen-sulfur intramolecular nonbonding interaction to stabilize the conformation required for binding to the p38α active site. X-ray crystallographic studies that confirm the proposed binding mode of this class of inhibitors in p38 α and provide evidence for the proposed intramolecular nitrogen-sulfur interaction are discussed.


Annual Reports in Medicinal Chemistry | 2009

Chapter 12 Advances in the Discovery of Small Molecule JAK3 Inhibitors

Stephen T. Wrobleski; William J. Pitts

Publisher Summary JAnus Kinase 3 (JAK3) is a member of the JAK family of non-receptor protein tyrosine kinases (PTKs) that include the closely related isoforms—namely, JAK1, JAK2 and tyrosine kinase 2 (TYK2). The realization that human defects in JAK3 signaling result in the clinical manifestation of a severe combined immunodeficiency (SCID) phenotype has suggested that selective JAK3 inhibitors may be useful as therapeutic agents in the areas of organ transplantation and autoimmune diseases. In addition, the promising clinical efficacy reported for the JAK3 inhibitor CP-690,550 in rheumatoid arthritis patients is noteworthy and suggests that obtaining efficacy comparable to, or perhaps better than, the current marketed biologic therapies in this disease may be possible with a small molecule. Although the highly selective inhibition of JAK3 for immunosuppression is particularly attractive from a safety perspective, it remains to be convincingly demonstrated in the clinic. While CP-690,550 does potently inhibit JAK3, it has been shown to inhibit to some extent other JAK family members—namely, JAK1 and JAK2, which may contribute to enhance efficacy in the clinic relative to purely selective JAK3 inhibition.


Bioorganic & Medicinal Chemistry Letters | 2014

Discovery of pyrrolo[1,2-b]pyridazine-3-carboxamides as Janus kinase (JAK) inhibitors.

James J.-W. Duan; Zhonghui Lu; Bin Jiang; Bingwei V. Yang; Lidia M. Doweyko; David S. Nirschl; Lauren Haque; Shuqun Lin; Gregory D. Brown; John Hynes; John S. Tokarski; John S. Sack; Javed Khan; Jonathan Lippy; Rosemary Zhang; Sidney Pitt; Guoxiang Shen; William J. Pitts; Percy H. Carter; Joel C. Barrish; Steven G. Nadler; Luisa Salter-Cid; Murray McKinnon; Aberra Fura; Gary L. Schieven; Stephen T. Wrobleski

A new class of Janus kinase (JAK) inhibitors was discovered using a rationally designed pyrrolo[1,2-b]pyridazine-3-carboxamide scaffold. Preliminary studies identified (R)-(2,2-dimethylcyclopentyl)amine as a preferred C4 substituent on the pyrrolopyridazine core (3b). Incorporation of amino group to 3-position of the cyclopentane ring resulted in a series of JAK3 inhibitors (4g-4j) that potently inhibited IFNγ production in an IL2-induced whole blood assay and displayed high functional selectivity for JAK3-JAK1 pathway relative to JAK2. Further modifications led to the discovery of an orally bioavailable (2-fluoro-2-methylcyclopentyl)amino analogue 5g which is a nanomolar inhibitor of both JAK3 and TYK2, functionally selective for the JAK3-JAK1 pathway versus JAK2, and active in a human whole blood assay.


Bioorganic & Medicinal Chemistry Letters | 2013

The identification of novel p38α isoform selective kinase inhibitors having an unprecedented p38α binding mode

Stephen T. Wrobleski; Shuqun Lin; T. G. Murali Dhar; Alaric J. Dyckman; Tianle Li; Sidney Pitt; Rosemary Zhang; Yi Fan; Arthur M. Doweyko; John S. Tokarski; Kevin Kish; Susan E. Kiefer; John S. Sack; John A. Newitt; Mark R. Witmer; Murray McKinnon; Joel C. Barrish; John H. Dodd; Gary L. Schieven; Katerina Leftheris

A novel series of p38 MAP kinase inhibitors with high selectivity for the p38α isoform over the other family members including the highly homologous p38β isoform has been identified. X-ray co-crystallographic studies have revealed an unprecedented kinase binding mode in p38α for representative analogs, 5c and 9d, in which a Leu108/Met109 peptide flip occurs within the p38α hinge region. Based on these findings, a general strategy for the rational design of additional promising p38α isoform selective inhibitors by targeting this novel binding mode is proposed.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of potent and efficacious pyrrolopyridazines as dual JAK1/3 inhibitors

John Hynes; Hong Wu; James Kempson; James J.-W. Duan; Zhonghui Lu; Bin Jiang; Sylwia Stachura; John S. Tokarski; John S. Sack; Javed Khan; Jonathan Lippy; Rosemary Zhang; Sidney Pitt; Guoxiang Shen; Kate Gillooly; Kim W. McIntyre; Percy H. Carter; Joel C. Barrish; Steven G. Nadler; Luisa Salter-Cid; Aberra Fura; Gary L. Schieven; William J. Pitts; Stephen T. Wrobleski

A series of potent dual JAK1/3 inhibitors have been developed from a moderately selective JAK3 inhibitor. Substitution at the C6 position of the pyrrolopyridazine core with aryl groups provided exceptional biochemical potency against JAK1 and JAK3 while maintaining good selectivity against JAK2 and Tyk2. Translation to in vivo efficacy was observed in a murine model of chronic inflammation. X-ray co-crystal structure determination confirmed the presumed inhibitor binding orientation in JAK3. Efforts to reduce hERG channel inhibition will be described.


Journal of Medicinal Chemistry | 2015

Discovery of ((4-(5-(Cyclopropylcarbamoyl)-2-methylphenylamino)-5-methylpyrrolo[1,2-f][1,2,4]triazine-6-carbonyl)(propyl)carbamoyloxy)methyl-2-(4-(phosphonooxy)phenyl)acetate (BMS-751324), a Clinical Prodrug of p38α MAP Kinase Inhibitor.

Chunjian Liu; James Lin; John Hynes; Hong Wu; Stephen T. Wrobleski; Shuqun Lin; T. G. Murali Dhar; Jung-Hui Sun; Sam T. Chao; Rulin Zhao; Bei Wang; Bang-Chi Chen; Gerry Everlof; Christoph Gesenberg; Hongjian Zhang; Punit Marathe; Kim W. McIntyre; Tracy L. Taylor; Kathleen M. Gillooly; David J. Shuster; Murray McKinnon; John H. Dodd; Joel C. Barrish; Gary L. Schieven; Katerina Leftheris

In search for prodrugs to address the issue of pH-dependent solubility and exposure associated with 1 (BMS-582949), a previously disclosed phase II clinical p38α MAP kinase inhibitor, a structurally novel clinical prodrug, 2 (BMS-751324), featuring a carbamoylmethylene linked promoiety containing hydroxyphenyl acetic acid (HPA) derived ester and phosphate functionalities, was identified. Prodrug 2 was not only stable but also water-soluble under both acidic and neutral conditions. It was effectively bioconverted into parent drug 1 in vivo by alkaline phosphatase and esterase in a stepwise manner, providing higher exposure of 1 compared to its direct administration, especially within higher dose ranges. In a rat LPS-induced TNFα pharmacodynamic model and a rat adjuvant arthritis model, 2 demonstrated similar efficacy to 1. Most importantly, it was shown in clinical studies that prodrug 2 was indeed effective in addressing the pH-dependent absorption issue associated with 1.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of highly potent, selective, covalent inhibitors of JAK3

James Kempson; Damaso Ovalle; Junqing Guo; Stephen T. Wrobleski; Shuqun Lin; Steven H. Spergel; James J.-W. Duan; Bin Jiang; Zhonghui Lu; Jagabandhu Das; Bingwei V. Yang; John Hynes; Hong Wu; John S. Tokarski; John S. Sack; Javed Khan; Gary L. Schieven; Yuval Blatt; Charu Chaudhry; Luisa Salter-Cid; Aberra Fura; Joel C. Barrish; Percy H. Carter; William J. Pitts

A useful and novel set of tool molecules have been identified which bind irreversibly to the JAK3 active site cysteine residue. The design was based on crystal structure information and a comparative study of several electrophilic warheads.

Collaboration


Dive into the Stephen T. Wrobleski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Hynes

University of South Carolina

View shared research outputs
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

Hong Wu

Bristol-Myers Squibb

View shared research outputs
Researchain Logo
Decentralizing Knowledge