Network


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

Hotspot


Dive into the research topics where Susan Kephart is active.

Publication


Featured researches published by Susan Kephart.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Small-molecule p21-activated kinase inhibitor PF-3758309 is a potent inhibitor of oncogenic signaling and tumor growth

Brion W. Murray; Chuangxing Guo; Joseph Piraino; John K. Westwick; Cathy Zhang; Jane E. Lamerdin; Eleanor Dagostino; Daniel R. Knighton; Cho‐Ming Loi; Michael Zager; Eugenia Kraynov; Ian Popoff; James G. Christensen; Ricardo N. Martínez; Susan Kephart; Joseph Timothy Marakovits; Shannon Marie Karlicek; Simon Bergqvist; Tod Smeal

Despite abundant evidence that aberrant Rho-family GTPase activation contributes to most steps of cancer initiation and progression, there is a dearth of inhibitors of their effectors (e.g., p21-activated kinases). Through high-throughput screening and structure-based design, we identify PF-3758309, a potent (Kd = 2.7 nM), ATP-competitive, pyrrolopyrazole inhibitor of PAK4. In cells, PF-3758309 inhibits phosphorylation of the PAK4 substrate GEF-H1 (IC50 = 1.3 nM) and anchorage-independent growth of a panel of tumor cell lines (IC50 = 4.7 ± 3 nM). The molecular underpinnings of PF-3758309 biological effects were characterized using an integration of traditional and emerging technologies. Crystallographic characterization of the PF-3758309/PAK4 complex defined determinants of potency and kinase selectivity. Global high-content cellular analysis confirms that PF-3758309 modulates known PAK4-dependent signaling nodes and identifies unexpected links to additional pathways (e.g., p53). In tumor models, PF-3758309 inhibits PAK4-dependent pathways in proteomic studies and regulates functional activities related to cell proliferation and survival. PF-3758309 blocks the growth of multiple human tumor xenografts, with a plasma EC50 value of 0.4 nM in the most sensitive model. This study defines PAK4-related pathways, provides additional support for PAK4 as a therapeutic target with a unique combination of functions (apoptotic, cytoskeletal, cell-cycle), and identifies a potent, orally available small-molecule PAK inhibitor with significant promise for the treatment of human cancers.


Journal of Medicinal Chemistry | 2011

Systematic Structure Modifications of Imidazo[1,2-a]pyrimidine to Reduce Metabolism Mediated by Aldehyde Oxidase (AO)

Angelica Linton; Ping Kang; Martha Ornelas; Susan Kephart; Qiyue Hu; Mason Alan Pairish; Ying Jiang; Chuangxing Guo

N-{trans-3-[(5-Cyano-6-methylpyridin-2-yl)oxy]-2,2,4,4-tetramethylcyclobutyl}imidazo[1,2-a]pyrimidine-3-carboxamide (1) was recently identified as a full antagonist of the androgen receptor, demonstrating excellent in vivo tumor growth inhibition in castration-resistant prostate cancer (CRPC). However, the imidazo[1,2-a]pyrimidine moiety is rapidly metabolized by aldehyde oxidase (AO). The present paper describes a number of medicinal chemistry strategies taken to avoid the AO-mediated oxidation of this particular system. Guided by an AO protein structure-based model, our investigation revealed the most probable site of AO oxidation and the observation that altering the heterocycle or blocking the reactive site are two of the more effective strategies for reducing AO metabolism. These strategies may be useful for other drug discovery programs.


Journal of Medicinal Chemistry | 2012

Discovery of Pyrroloaminopyrazoles as Novel Pak Inhibitors.

Chuangxing Guo; Indrawan McAlpine; Junhu Zhang; Daniel D. Knighton; Susan Kephart; M. Catherine Johnson; Haitao Li; Djamal Bouzida; Anle Yang; Liming Dong; Joseph Timothy Marakovits; Jayashree Girish Tikhe; Paul G. Richardson; Lisa C. Guo; Robert Steven Kania; Martin Paul Edwards; Eugenia Kraynov; James G. Christensen; Joseph Piraino; Joseph H. Lee; Eleanor Dagostino; Christine Del-Carmen; Ya-Li Deng; Tod Smeal; Brion W. Murray

The P21-activated kinases (PAK) are emerging antitumor therapeutic targets. In this paper, we describe the discovery of potent PAK inhibitors guided by structure-based drug design. In addition, the efflux of the pyrrolopyrazole series was effectively reduced by applying multiple medicinal chemistry strategies, leading to a series of PAK inhibitors that are orally active in inhibiting tumor growth in vivo.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of 2-((1H-benzo[d]imidazol-1-yl)methyl)-4H-pyrido[1,2-a]pyrimidin-4-ones as novel PKM2 activators.

Chuangxing Guo; Angelica Linton; Mehran Jalaie; Susan Kephart; Martha Ornelas; Mason Alan Pairish; Samantha Greasley; Paul G. Richardson; Karen Maegley; Michael J. Hickey; John Li; Xin Wu; Xiaodong Ji; Zhi Xie

The M2 isoform of pyruvate kinase is an emerging target for antitumor therapy. In this letter, we describe the discovery of 2-((1H-benzo[d]imidazol-1-yl)methyl)-4H-pyrido[1,2-a]pyrimidin-4-ones as potent and selective PKM2 activators which were found to have a novel binding mode. The original lead identified from high throughput screening was optimized into an efficient series via computer-aided structure-based drug design. Both a representative compound from this series and an activator described in the literature were used as molecular tools to probe the biological effects of PKM2 activation on cancer cells. Our results suggested that PKM2 activation alone is not sufficient to alter cancer cell metabolism.


Journal of Medicinal Chemistry | 2016

Design and Synthesis of Pyridone-Containing 3,4-Dihydroisoquinoline-1(2H)-ones as a Novel Class of Enhancer of Zeste Homolog 2 (EZH2) Inhibitors

Pei-Pei Kung; Eugene Rui; Simon Bergqvist; Patrick Bingham; John Frederick Braganza; Michael Raymond Collins; Mei Cui; Wade Diehl; Dac M. Dinh; Connie Fan; Valeria R. Fantin; Hovhannes J. Gukasyan; Wenyue Hu; Buwen Huang; Susan Kephart; Cody Krivacic; Robert Arnold Kumpf; Gary Li; Karen Maegley; Indrawan McAlpine; Lisa Nguyen; Sacha Ninkovic; Martha Ornelas; Michael Ryskin; Stephanie Scales; Scott C. Sutton; John Howard Tatlock; Dominique Verhelle; Fen Wang; Peter A. Wells

A new enhancer of zeste homolog 2 (EZH2) inhibitor series comprising a substituted phenyl ring joined to a dimethylpyridone moiety via an amide linkage has been designed. A preferential amide torsion that improved the binding properties of the compounds was identified for this series via computational analysis. Cyclization of the amide linker resulted in a six-membered lactam analogue, compound 18. This transformation significantly improved the ligand efficiency/potency of the cyclized compound relative to its acyclic analogue. Additional optimization of the lactam-containing EZH2 inhibitors focused on lipophilic efficiency (LipE) improvement, which provided compound 31. Compound 31 displayed improved LipE and on-target potency in both biochemical and cellular readouts relative to compound 18. Inhibitor 31 also displayed robust in vivo antitumor growth activity and dose-dependent de-repression of EZH2 target genes.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of 3-aryloxy-lactam analogs as potent androgen receptor full antagonists for treating castration resistant prostate cancer.

Chuangxing Guo; Susan Kephart; Martha Ornelas; Javier Gonzalez; Angelica Linton; Mason Alan Pairish; Asako Nagata; Samantha Greasley; Jeff Elleraas; Natilie Hosea; Jon Engebretsen; Andrea Fanjul

High throughput cell-based screening led to the identification of 3-aryloxy lactams as potent androgen receptor (AR) antagonists. Refinement of these leads to improve the ADME profile and remove residual agonism led to the discovery of 12, a potent full antagonist with greater oral bioavailability. Improvements in the ADME profile were realized by designing more ligand-efficient molecules with reduced molecular weights and lower lipophilicities.


Bioorganic & Medicinal Chemistry Letters | 2012

Structural modifications of a 3-methoxy-2-aminopyridine compound to reduce potential for mutagenicity and time-dependent drug–drug interaction

Cynthia Louise Palmer; Mason Alan Pairish; Susan Kephart; Djamal Bouzida; Jingrong Cui; Judith Gail Deal; Liming Dong; Danlin Gu; Angelica Linton; Indrawan McAlpine; Shinji Yamazaki; Evan Smith; Annette John-Baptiste; Shubha Bagrodia; Robert Steven Kania; Chuangxing Guo

(S)-1-((4-(3-(6-Amino-5-methoxypyridin-3-yl)-1-isopropyl-1H-pyrazol-4-yl)pyrimidin-2-yl)amino)propan-2-ol, 1, was recently identified as a potent inhibitor of the oncogenic kinase bRAF. Compounds containing 3-methoxy-2-aminopyridine, as in 1, comprised a promising lead series because of their high ligand efficiency and excellent ADME profile. However, following metabolic oxidation, compounds in this series also demonstrated two significant safety risks: mutagenic potential and time-dependent drug-drug interaction (TDI). Metabolite identification studies revealed formation of a reactive metabolite. We hypothesized that minimizing or blocking the formation of such a metabolite would mitigate the safety liabilities. Our investigation demonstrated that structural modifications which either reduced the electron density of the 3-methoxy-2-aminopyridine ring or blocked the reactive site following metabolic oxidation were successful in reducing TDI and AMES mutagenicity.


Molecular Cancer Therapeutics | 2009

Abstract PR-2: Discovery of p21‐activated kinase inhibitor PF‐03758309

Chuangxing Guo; Junhu Zhang; Indrawan McAlpine; Catherine Johnson; Joseph Marakovits; Liming Dong; Susan Kephart; Anle Yang; Jayashree Tikhe; Haitao Li; Lisa Guo; Djamal Bouzida; Ya-Li Deng; Dan Knighton; Joseph Piraino; Joseph Lee; Tod Smeal; Jamie Christensen; Eugenia Kraynov; Cho‐Ming Loi; Hasam Younis; Eleanor Dagostino; Brion W. Murray

The p21‐activated kinase (PAK) family members are key effectors of Rho family GTPases, which act as regulatory switches that control such cellular processes as motility, proliferation, and cell survival. Some members of this family (such as Cdc42) have been shown to be required for Ras driven tumorigenesis. PAK4 is a key effector for Cdc42 and mediates downstream signals that control cell motility, proliferation and cell survival. The PAK family consists of PAK1,2,3 (‘group 1’) and PAK4,5,6 (‘group 2’). Both PAK4 and PAK1 have been shown to be required for Ras driven transformation. PAK4 has been shown to be oncogenic and able to drive anchorage independent growth when activated. PAK4 expression and activity is broadly up‐regulated in solid tumors such as colon, ovarian and pancreatic cancers. Pfizer9s PAK4 inhibitor program started with HTS of kinase focus library compounds. Multiple series were identified from the screening effort. Initial optimization mainly focused on kinase selectivity, ADME properties and ability to modulate target in vivo, leading to selection of pyrrolopyrazoles as the lead series. By hybridizing the pyrrolopyrazole core with aminopyrimidine series, we discovered potent PAK4 inhibitors with low to sub nM cellular activity. For the new hybrid series, the challenge was to attain good oral bioavailability. The observed poor absorption was likely due to high efflux nature of the template. Medicinal chemistry strategies, such as reducing molecular charge, lowering polar surface area and improving ligand efficiency, were applied to reduce efflux. As a result, two sub‐series achieved excellent in vivo tumor growth inhibition when dosed orally. PF‐03758309 has demonstrated excellent profile, leading to its selection as a clinical development candidate. PF‐3758309 is a potent ATP‐competitive inhibitor of PAK4 kinase domain (Kd = 4.5 nM). In engineered cell assays, PF‐3758309 inhibited PAK4 dependent phosphorylation of its substrate GEF‐H1 (IC50 = 1 nM). It potently inhibits the anchorage independent growth of HCT116 cells (IC50 = 0.24 nM). PF‐3758309 exhibited broad anti‐proliferative activity across a panel of 67 cell lines (CRC/pancreatic/NSCLC): 66% IC50 70%TGI at 15–20 mg/kg PO) by PF‐3758309: HCT116, A549, MDAMB231, M24met, and Colo205. Broad kinase screening has demonstrated that this is a selective pan‐PAK inhibitor with potential additional activities (e.g. AMPK). The pharmacodynamic and antitumor effects of PF‐3758309 support its evaluation as an anticancer agent. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):PR-2.


Archive | 2001

Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation

Siegfried Heinz Reich; Ted Michael Bleckman; Susan Kephart; William H. Romines; Michael B. Wallace


Archive | 2004

3,5 disubstituted indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation

Susan Kephart; Indrawan McAlpine; Siegfried Heinz Agouron Pharmaceuticals Reich

Collaboration


Dive into the Susan Kephart's collaboration.

Researchain Logo
Decentralizing Knowledge