Network


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

Hotspot


Dive into the research topics where Ivana Yen is active.

Publication


Featured researches published by Ivana Yen.


Nature | 2010

RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth

Georgia Hatzivassiliou; Kyung Song; Ivana Yen; Barbara J. Brandhuber; Daniel J. Anderson; Ryan Alvarado; Mary J. C. Ludlam; David Stokoe; Susan L. Gloor; Guy Vigers; Tony Morales; Ignacio Aliagas; Bonnie Liu; Steve Sideris; Klaus P. Hoeflich; Bijay S. Jaiswal; Somasekar Seshagiri; Hartmut Koeppen; Marcia Belvin; Lori S. Friedman; Shiva Malek

Activating mutations in KRAS and BRAF are found in more than 30% of all human tumours and 40% of melanoma, respectively, thus targeting this pathway could have broad therapeutic effects. Small molecule ATP-competitive RAF kinase inhibitors have potent antitumour effects on mutant BRAF(V600E) tumours but, in contrast to mitogen-activated protein kinase kinase (MEK) inhibitors, are not potent against RAS mutant tumour models, despite RAF functioning as a key effector downstream of RAS and upstream of MEK. Here we show that ATP-competitive RAF inhibitors have two opposing mechanisms of action depending on the cellular context. In BRAF(V600E) tumours, RAF inhibitors effectively block the mitogen-activated protein kinase (MAPK) signalling pathway and decrease tumour growth. Notably, in KRAS mutant and RAS/RAF wild-type tumours, RAF inhibitors activate the RAF–MEK–ERK pathway in a RAS-dependent manner, thus enhancing tumour growth in some xenograft models. Inhibitor binding activates wild-type RAF isoforms by inducing dimerization, membrane localization and interaction with RAS–GTP. These events occur independently of kinase inhibition and are, instead, linked to direct conformational effects of inhibitors on the RAF kinase domain. On the basis of these findings, we demonstrate that ATP-competitive kinase inhibitors can have opposing functions as inhibitors or activators of signalling pathways, depending on the cellular context. Furthermore, this work provides new insights into the therapeutic use of ATP-competitive RAF inhibitors.


Cancer Cell | 2014

Structure of the BRAF-MEK Complex Reveals a Kinase Activity Independent Role for BRAF in MAPK Signaling.

Jacob R. Haling; Jawahar Sudhamsu; Ivana Yen; Steve Sideris; Wendy Sandoval; Wilson Phung; Brandon J. Bravo; Anthony M. Giannetti; Ariana Peck; Alexandre Masselot; Tony Morales; Darin Smith; Barbara J. Brandhuber; Sarah G. Hymowitz; Shiva Malek

Numerous oncogenic mutations occur within the BRAF kinase domain (BRAF(KD)). Here we show that stable BRAF-MEK1 complexes are enriched in BRAF(WT) and KRAS mutant (MT) cells but not in BRAF(MT) cells. The crystal structure of the BRAF(KD) in a complex with MEK1 reveals a face-to-face dimer sensitive to MEK1 phosphorylation but insensitive to BRAF dimerization. Structure-guided studies reveal that oncogenic BRAF mutations function by bypassing the requirement for BRAF dimerization for activity or weakening the interaction with MEK1. Finally, we show that conformation-specific BRAF inhibitors can sequester a dormant BRAF-MEK1 complex resulting in pathway inhibition. Taken together, these findings reveal a regulatory role for BRAF in the MAPK pathway independent of its kinase activity but dependent on interaction with MEK.


Bioorganic & Medicinal Chemistry Letters | 2013

Identification of substituted 2-thio-6-oxo-1,6-dihydropyrimidines as inhibitors of human lactate dehydrogenase.

Peter S. Dragovich; Benjamin P. Fauber; Laura Corson; Charles Z. Ding; Charles Eigenbrot; HongXiu Ge; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; Yichin Liu; Shiva Malek; Borlan Pan; David Peterson; Keith Pitts; Hans E. Purkey; Steve Sideris; Mark Ultsch; Erica VanderPorten; Binqing Wei; Qing Xu; Ivana Yen; Qin Yue; Huihui Zhang; Xuying Zhang

A novel 2-thio-6-oxo-1,6-dihydropyrimidine-containing inhibitor of human lactate dehydrogenase (LDH) was identified by high-throughput screening (IC50=8.1 μM). Biochemical, surface plasmon resonance, and saturation transfer difference NMR experiments indicated that the compound specifically associated with human LDHA in a manner that required simultaneous binding of the NADH co-factor. Structural variation of the screening hit resulted in significant improvements in LDHA biochemical inhibition activity (best IC50=0.48 μM). A crystal structure of an optimized compound bound to human LDHA was obtained and explained many of the observed structure-activity relationships.


Bioorganic & Medicinal Chemistry Letters | 2013

Identification of 2-amino-5-aryl-pyrazines as inhibitors of human lactate dehydrogenase.

Benjamin P. Fauber; Peter S. Dragovich; Jinhua Chen; Laura Corson; Charles Z. Ding; Charles Eigenbrot; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; Yichin Liu; Shiva Malek; David Peterson; Keith Pitts; Steven Sideris; Mark Ultsch; Erica VanderPorten; J Wang; Binqing Wei; Ivana Yen; Qin Yue

A 2-amino-5-aryl-pyrazine was identified as an inhibitor of human lactate dehydrogenase A (LDHA) via a biochemical screening campaign. Biochemical and biophysical experiments demonstrated that the compound specifically interacted with human LDHA. Structural variation of the screening hit resulted in improvements in LDHA biochemical inhibition and pharmacokinetic properties. A crystal structure of an improved compound bound to human LDHA was also obtained and it explained many of the observed structure-activity relationships.


Nature Chemical Biology | 2016

Metabolic plasticity underpins innate and acquired resistance to LDHA inhibition

Aaron Boudreau; Hans E. Purkey; Anna Hitz; Kirk Robarge; David Peterson; Sharada Labadie; Mandy Kwong; Rebecca Hong; Min Gao; Christopher Del Nagro; Raju V. Pusapati; Shuguang Ma; Laurent Salphati; Jodie Pang; Aihe Zhou; Tommy Lai; Yingjie Li; Zhongguo Chen; Binqing Wei; Ivana Yen; Steve Sideris; Mark L. McCleland; Ron Firestein; Laura Corson; Alex Vanderbilt; Simon Williams; Anneleen Daemen; Marcia Belvin; Charles Eigenbrot; Peter K. Jackson

Metabolic reprogramming in tumors represents a potential therapeutic target. Herein we used shRNA depletion and a novel lactate dehydrogenase (LDHA) inhibitor, GNE-140, to probe the role of LDHA in tumor growth in vitro and in vivo. In MIA PaCa-2 human pancreatic cells, LDHA inhibition rapidly affected global metabolism, although cell death only occurred after 2 d of continuous LDHA inhibition. Pancreatic cell lines that utilize oxidative phosphorylation (OXPHOS) rather than glycolysis were inherently resistant to GNE-140, but could be resensitized to GNE-140 with the OXPHOS inhibitor phenformin. Acquired resistance to GNE-140 was driven by activation of the AMPK-mTOR-S6K signaling pathway, which led to increased OXPHOS, and inhibitors targeting this pathway could prevent resistance. Thus, combining an LDHA inhibitor with compounds targeting the mitochondrial or AMPK-S6K signaling axis may not only broaden the clinical utility of LDHA inhibitors beyond glycolytically dependent tumors but also reduce the emergence of resistance to LDHA inhibition.


Molecular Cancer Therapeutics | 2013

Combination Drug Scheduling Defines a “Window of Opportunity” for Chemopotentiation of Gemcitabine by an Orally Bioavailable, Selective ChK1 Inhibitor, GNE-900

Elizabeth Blackwood; Jennifer Epler; Ivana Yen; Michael Flagella; Thomas O'Brien; Marie Evangelista; Stephen Schmidt; Yang Xiao; Jonathan Choi; Kaska Kowanetz; Judi Ramiscal; Kenton Wong; Diana Jakubiak; Sharon Yee; Gary Cain; Lewis J. Gazzard; Karen Williams; Jason S. Halladay; Peter K. Jackson; Shiva Malek

Checkpoint kinase 1 (ChK1) is a serine/threonine kinase that functions as a central mediator of the intra-S and G2–M cell-cycle checkpoints. Following DNA damage or replication stress, ChK1-mediated phosphorylation of downstream effectors delays cell-cycle progression so that the damaged genome can be repaired. As a therapeutic strategy, inhibition of ChK1 should potentiate the antitumor effect of chemotherapeutic agents by inactivating the postreplication checkpoint, causing premature entry into mitosis with damaged DNA resulting in mitotic catastrophe. Here, we describe the characterization of GNE-900, an ATP-competitive, selective, and orally bioavailable ChK1 inhibitor. In combination with chemotherapeutic agents, GNE-900 sustains ATR/ATM signaling, enhances DNA damage, and induces apoptotic cell death. The kinetics of checkpoint abrogation seems to be more rapid in p53-mutant cells, resulting in premature mitotic entry and/or accelerated cell death. Importantly, we show that GNE-900 has little single-agent activity in the absence of chemotherapy and does not grossly potentiate the cytotoxicity of gemcitabine in normal bone marrow cells. In vivo scheduling studies show that optimal administration of the ChK1 inhibitor requires a defined lag between gemcitabine and GNE-900 administration. On the refined combination treatment schedule, gemcitabines antitumor activity against chemotolerant xenografts is significantly enhanced and dose-dependent exacerbation of DNA damage correlates with extent of tumor growth inhibition. In summary, we show that in vivo potentiation of gemcitabine activity is mechanism based, with optimal efficacy observed when S-phase arrest and release is followed by checkpoint abrogation with a ChK1 inhibitor. Mol Cancer Ther; 12(10); 1968–80. ©2013 AACR.


Journal of Medicinal Chemistry | 2014

Discovery of Selective and Noncovalent Diaminopyrimidine-Based Inhibitors of Epidermal Growth Factor Receptor Containing the T790M Resistance Mutation.

Emily J. Hanan; Charles Eigenbrot; Marian C. Bryan; Daniel J. Burdick; Bryan K. Chan; Yuan Chen; Jennafer Dotson; Robert Heald; Philip Stephen Jackson; Hank La; Michael Lainchbury; Shiva Malek; Hans E. Purkey; Gabriele Schaefer; Stephen Schmidt; Eileen Mary Seward; Steve Sideris; Christine Tam; Shumei Wang; Siew Kuen Yeap; Ivana Yen; JianPing Yin; Christine Yu; Inna Zilberleyb; Timothy P. Heffron

Activating mutations within the epidermal growth factor receptor (EGFR) kinase domain, commonly L858R or deletions within exon 19, increase EGFR-driven cell proliferation and survival and are correlated with impressive responses to the EGFR inhibitors erlotinib and gefitinib in nonsmall cell lung cancer patients. Approximately 60% of acquired resistance to these agents is driven by a single secondary mutation within the EGFR kinase domain, specifically substitution of the gatekeeper residue threonine-790 with methionine (T790M). Due to dose-limiting toxicities associated with inhibition of wild-type EGFR (wtEGFR), we sought inhibitors of T790M-containing EGFR mutants with selectivity over wtEGFR. We describe the evolution of HTS hits derived from Jak2/Tyk2 inhibitors into selective EGFR inhibitors. X-ray crystal structures revealed two distinct binding modes and enabled the design of a selective series of novel diaminopyrimidine-based inhibitors with good potency against T790M-containing mutants of EGFR, high selectivity over wtEGFR, broad kinase selectivity, and desirable physicochemical properties.


Bioorganic & Medicinal Chemistry Letters | 2014

Identification of substituted 3-hydroxy-2-mercaptocyclohex-2-enones as potent inhibitors of human lactate dehydrogenase.

Peter S. Dragovich; Benjamin P. Fauber; Jason Boggs; Jinhua Chen; Laura Corson; Charles Z. Ding; Charles Eigenbrot; HongXiu Ge; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; C Li; Yichin Liu; Shuguang Ma; Shiva Malek; David Peterson; Keith Pitts; Hans E. Purkey; Kirk Robarge; Laurent Salphati; Steven Sideris; Mark Ultsch; Erica VanderPorten; J Wang; Binqing Wei; Qing Xu; Ivana Yen; Qin Yue; Huihui Zhang; Xuying Zhang

A novel class of 3-hydroxy-2-mercaptocyclohex-2-enone-containing inhibitors of human lactate dehydrogenase (LDH) was identified through a high-throughput screening approach. Biochemical and surface plasmon resonance experiments performed with a screening hit (LDHA IC50=1.7 μM) indicated that the compound specifically associated with human LDHA in a manner that required simultaneous binding of the NADH co-factor. Structural variation of this screening hit resulted in significant improvements in LDHA biochemical inhibition activity (best IC50=0.18 μM). Two crystal structures of optimized compounds bound to human LDHA were obtained and explained many of the observed structure-activity relationships. In addition, an optimized inhibitor exhibited good pharmacokinetic properties after oral administration to rats (F=45%).


Journal of Medicinal Chemistry | 2015

Noncovalent Mutant Selective Epidermal Growth Factor Receptor Inhibitors: A Lead Optimization Case Study.

Robert Heald; Krista K. Bowman; Marian C. Bryan; Daniel J. Burdick; Bryan K. Chan; Emily Chan; Yuan Chen; Saundra Clausen; Belen Dominguez-Fernandez; Charles Eigenbrot; Richard L. Elliott; Emily J. Hanan; Philip Stephen Jackson; Hank La; Michael Lainchbury; Shiva Malek; Sam Mann; Mark Merchant; Kyle Mortara; Hans E. Purkey; Gabriele Schaefer; Stephen Schmidt; Eileen Mary Seward; Steve Sideris; Lily Shao; Shumei Wang; Kuen Yeap; Ivana Yen; Christine Yu; Timothy P. Heffron

Because of their increased activity against activating mutants, first-generation epidermal growth factor receptor (EGFR) kinase inhibitors have had remarkable success in treating non-small-cell lung cancer (NSCLC) patients, but acquired resistance, through a secondary mutation of the gatekeeper residue, means that clinical responses only last for 8-14 months. Addressing this unmet medical need requires agents that can target both of the most common double mutants: T790M/L858R (TMLR) and T790M/del(746-750) (TMdel). Herein we describe how a noncovalent double mutant selective lead compound was optimized using a strategy focused on the structure-guided increase in potency without added lipophilicity or reduction of three-dimensional character. Following successive rounds of design and synthesis it was discovered that cis-fluoro substitution on 4-hydroxy- and 4-methoxypiperidinyl groups provided synergistic, substantial, and specific potency gain through direct interaction with the enzyme and/or effects on the proximal ligand oxygen atom. Further development of the fluorohydroxypiperidine series resulted in the identification of a pair of diastereomers that showed 50-fold enzyme and cell based selectivity for T790M mutants over wild-type EGFR (wtEGFR) in vitro and pathway knock-down in an in vivo xenograft model.


Bioorganic & Medicinal Chemistry Letters | 2014

Discovery of the 1,7-diazacarbazole class of inhibitors of checkpoint kinase 1.

Lewis J. Gazzard; Brent A. Appleton; Kerry Chapman; Huifen Chen; Kevin Clark; Joy Drobnick; Simon Goodacre; Jason S. Halladay; Joseph P. Lyssikatos; Stephen Schmidt; Steve Sideris; Christian Wiesmann; Karen Williams; Ping Wu; Ivana Yen; Shiva Malek

Checkpoint kinase 1 (ChK1) is activated in response to DNA damage, acting to temporarily block cell cycle progression and allow for DNA repair. It is envisaged that inhibition of ChK1 will sensitize tumor cells to treatment with DNA-damaging therapies, and may enhance the therapeutic window. High throughput screening identified carboxylate-containing diarylpyrazines as a prominent hit series, but with limited biochemical potency and no cellular activity. Through a series of SAR investigations and X-ray crystallographic analysis the critical role of polar contacts with conserved waters in the kinase back pocket was established. Structure-based design, guided by in silico modeling, transformed the series to better satisfy these contacts and the novel 1,7-diazacarbazole class of inhibitors was discovered. Here we present the genesis of this novel series and the identification of GNE-783, a potent, selective and orally bioavailable inhibitor of ChK1.

Collaboration


Dive into the Ivana Yen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans E. Purkey

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge