Network


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

Hotspot


Dive into the research topics where Paul A. Barsanti is active.

Publication


Featured researches published by Paul A. Barsanti.


Bioorganic & Medicinal Chemistry Letters | 2010

The discovery of tetrahydro-β-carbolines as inhibitors of the kinesin Eg5

Paul A. Barsanti; Weibo Wang; Zhi-Jie Ni; David Duhl; Nathan Brammeier; Eric J. Martin; Dirksen E. Bussiere; Annette Walter

A series of tetrahydro-beta-carbolines were identified by HTS as inhibitors of the kinesin Eg5. Molecular modeling and medicinal chemistry techniques were employed to explore the SAR for this series with a focus of removing potential metabolic liabilities and improving cellular potency.


Molecular Cancer Research | 2015

A Synthetic Lethal Screen Reveals Enhanced Sensitivity to ATR Inhibitor Treatment in Mantle Cell Lymphoma with ATM Loss-of-Function

Daniel Menezes; Jenny Holt; Yan Tang; Jiajia Feng; Paul A. Barsanti; Yue Pan; Majid Ghoddusi; Wei Zhang; George Thomas; Jocelyn Holash; Emma Lees; Lorena Taricani

Mechanisms to maintain genomic integrity are essential for cells to remain viable. Not surprisingly, disruption of key DNA damage response pathway factors, such as ataxia telangiectasia-mutated (ATM)/ataxia telangiectasia and RAD3-related (ATR) results in loss of genomic integrity. Here, a synthetic lethal siRNA-screening approach not only confirmed ATM but identified additional replication checkpoint proteins, when ablated, enhanced ATR inhibitor (ATRi) response in a high-content γ-H2AX assay. Cancers with inactivating ATM mutations exhibit impaired DNA double-stranded break (DSB) repair and rely on compensatory repair pathways for survival. Therefore, impairing ATR activity may selectively sensitize cancer cells to killing. ATR inhibition in an ATM-deficient context results in phosphorylation of DNA-dependent protein kinase catalytic subunits (DNA-PKcs) and leads to induction of γ-H2AX. Using both in vitro and in vivo models, ATR inhibition enhanced efficacy in ATM loss-of-function mantle cell lymphoma (MCL) compared with ATM wild-type cancer cells. In summary, single-agent ATR inhibitors have therapeutic utility in the treatment of cancers, like MCL, in which ATM function has been lost. Implications: These data suggest that single-agent ATR inhibitors have therapeutic utility and that ATR uses a complex and coordinated set of proteins to maintain genomic stability that could be further exploited. Mol Cancer Res; 13(1); 120–9. ©2014 AACR.


ACS Medicinal Chemistry Letters | 2015

Structure-Based Drug Design of Novel Potent and Selective Tetrahydropyrazolo[1,5-a]pyrazines as ATR Inhibitors.

Paul A. Barsanti; Robert Aversa; Xianming Jin; Yue Pan; Yipin Lu; Robert A. Elling; Rama Jain; Mark Knapp; Jiong Lan; Xiaodong Lin; Patrick Rudewicz; Janet Sim; Lorena Taricani; George Thomas; Linda Xiao; Qin Yue

A saturation strategy focused on improving the selectivity and physicochemical properties of ATR inhibitor HTS hit 1 led to a novel series of highly potent and selective tetrahydropyrazolo[1,5-a]pyrazines. Use of PI3Kα mutants as ATR crystal structure surrogates was instrumental in providing cocrystal structures to guide the medicinal chemistry designs. Detailed DMPK studies involving cyanide and GSH as trapping agents during microsomal incubations, in addition to deuterium-labeled compounds as mechanistic probes uncovered the molecular basis for the observed CYP3A4 TDI in the series.


Journal of Medicinal Chemistry | 2017

Design and Discovery of N-(2-Methyl-5′-morpholino-6′-((tetrahydro-2H-pyran-4-yl)oxy)-[3,3′-bipyridin]-5-yl)-3-(trifluoromethyl)benzamide (RAF709): A Potent, Selective, and Efficacious RAF Inhibitor Targeting RAS Mutant Cancers

Gisele Nishiguchi; Alice Rico; Huw Tanner; Robert Aversa; Benjamin Taft; Sharadha Subramanian; Lina Setti; Matthew Burger; Lifeng Wan; Victoriano Tamez; Aaron Smith; Yan Lou; Paul A. Barsanti; Brent A. Appleton; Mulugeta Mamo; Laura Tandeske; Ina Dix; John E. Tellew; Shenlin Huang; Lesley A. Mathews Griner; Vesselina G. Cooke; Anne Van Abbema; Hanne Merritt; Sylvia Ma; Kalyani Gampa; Fei Feng; Jing Yuan; Yingyun Wang; Jacob R Haling; Sepideh Vaziri

RAS oncogenes have been implicated in >30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.


Journal of Molecular Biology | 2017

Rationally Designed PI3Kα Mutants to Mimic ATR and Their Use to Understand Binding Specificity of ATR Inhibitors

Yipin Lu; Mark Knapp; Kenneth Crawford; Robert Warne; Robert Elling; Kelly Yan; Michael V. Doyle; Gwynn Pardee; Li Zhang; Sylvia Ma; Mulugeta Mamo; Elizabeth Ornelas; Yue Pan; Dirksen E. Bussiere; Johanna M. Jansen; Isabel Zaror; Albert Lai; Paul A. Barsanti; Janet Sim

ATR, a protein kinase in the PIKK family, plays a critical role in the cell DNA-damage response and is an attractive anticancer drug target. Several potent and selective inhibitors of ATR have been reported showing significant antitumor efficacy, with most advanced ones entering clinical trials. However, due to the absence of an experimental ATR structure, the determinants contributing to ATR inhibitors potency and specificity are not well understood. Here we present the mutations in the ATP-binding site of PI3Kα to progressively transform the pocket to mimic that of ATR. The generated PI3Kα mutants exhibit significantly improved affinity for selective ATR inhibitors in multiple chemical classes. Furthermore, we obtained the X-ray structures of the PI3Kα mutants in complex with the ATR inhibitors. The crystal structures together with the analysis on the inhibitor affinity profile elucidate the roles of individual amino acid residues in the binding of ATR inhibitors, offering key insights for the binding mechanism and revealing the structure features important for the specificity of ATR inhibitors. The ability to obtain structural and binding data for these PI3Kα mutants, together with their ATR-like inhibitor binding profiles, makes these chimeric PI3Kα proteins valuable model systems for structure-based inhibitor design.


Archive | 2007

Pyrimidine derivatives used as PI-3-kinase inhibitors

Matthew Burger; Zhi-Jie Ni; Sabina Pecchi; Gordana Atallah; Sarah Bartulis; Kelly Frazier; Aaron Smith; Joelle Verhagen; Yanchen Zhang; Allan S. Wagman; Simon Ng; Keith B. Pfister; Daniel J. Poon; Alicia Louie; Teresa E. Pick; Paul A. Barsanti; Edwin Iwanowicz; Wendy J. Fantl; Thomas Hendrickson; Mark Knapp; Hanne Meritt; Charles Voliva; Marion Wiesmann; Xiahua Xin


Archive | 2006

2-amino-7,8-dihydro-6h-pyrido[4,3-d] pyrimidin-5-ones

Timothy D. Machajewski; Cynthia Shafer; Christopher Mcbride; William R. Antonios-McCrea; Brandon M. Doughan; Barry Haskell Levine; Yi Xia; Maureen Mckenna; X. Michael Wang; Kris Mendenhall; Yasheen Zhou; Baoqing Gong; Dan Gu; John Dolan; John Tulinsky; Kristin Brinner; Zhenhai Gao; Daniel Poon; Paul A. Barsanti; Xiaodong Lin; Abran Costales; Alice Rico; Nathan Brammeier; Teresa E. Pick; Paul A. Renhowe


Archive | 2005

Tetrahydrocarboline compounds as anticancer agents

Weibo Wang; Zhi-Jie Ni; Paul A. Barsanti; Sabina Pecchi; Yi Xia; Nathan Brammeier; Megan C. Phillips; Eliza Jazan; Kelly Wayman; David Dibble; Jie-Kai Cheng


Archive | 2010

Pyridine and pyrazine derivatives as protein kinase modulators

Paul A. Barsanti; Cheng Hu; Jeff Jin; Robert Keyes; Robert Kucejko; Xiaodong Lin; Yue Pan; Keith B. Pfister; Martin Sendzik; James Sutton; Lifeng Wan


Archive | 2011

3-(AMINOARYL)-PYRIDINE COMPOUNDS

William R. Antonios-McCrea; Paul A. Barsanti; Cheng Hu; Xianming Jin; Eric J. Martin; Yue Pan; Keith B. Pfister; Martin Sendzik; James Sutton; Lifeng Wan

Collaboration


Dive into the Paul A. Barsanti's collaboration.

Researchain Logo
Decentralizing Knowledge