Network


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

Hotspot


Dive into the research topics where Isabelle Dussault is active.

Publication


Featured researches published by Isabelle Dussault.


Journal of Medicinal Chemistry | 2008

Discovery and Optimization of Triazolopyridazines as Potent and Selective Inhibitors of the c-Met Kinase.

Brian K. Albrecht; Jean-Christophe Harmange; David Bauer; Loren Berry; Christiane Bode; Alessandro Boezio; April Chen; Deborah Choquette; Isabelle Dussault; Cary Fridrich; Satoko Hirai; Doug Hoffman; Jay Larrow; Paula Kaplan-Lefko; Jasmine Lin; Julia Lohman; Alexander M. Long; Jodi Moriguchi; Anne O'connor; Michele Potashman; Monica Reese; Karen Rex; Aaron C. Siegmund; Kavita Shah; Roman Shimanovich; Stephanie K. Springer; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon

Tumorigenesis is a multistep process in which oncogenes play a key role in tumor formation, growth, and maintenance. MET was discovered as an oncogene that is activated by its ligand, hepatocyte growth factor. Deregulated signaling in the c-Met pathway has been observed in multiple tumor types. Herein we report the discovery of potent and selective triazolopyridazine small molecules that inhibit c-Met activity.


Cancer Research | 2011

STK33 Kinase Activity Is Nonessential in KRAS-Dependent Cancer Cells

Carol Babij; Yihong Zhang; Robert J.M. Kurzeja; Anke Munzli; Amro Shehabeldin; Manory Fernando; Kim Quon; Paul Kassner; Astrid Ruefli-Brasse; Vivienne Watson; Flordeliza Fajardo; Angela Jackson; James Zondlo; Yu Sun; Aaron R. Ellison; Cherylene Plewa; Miguel Tisha San; John C. Robinson; John D. McCarter; Ralf Schwandner; Ted Judd; Josette Carnahan; Isabelle Dussault

Despite the prevalence of KRAS mutations in human cancers, there remain no targeted therapies for treatment. The serine-threonine kinase STK33 has been proposed to be required for the survival of mutant KRAS-dependent cell lines, suggesting that small molecule kinase inhibitors of STK33 may be useful to treat KRAS-dependent tumors. In this study, we investigated the role of STK33 in mutant KRAS human cancer cells using RNA interference, dominant mutant overexpression, and small molecule inhibitors. As expected, KRAS downregulation decreased the survival of KRAS-dependent cells. In contrast, STK33 downregulation or dominant mutant overexpression had no effect on KRAS signaling or survival of these cells. Similarly, a synthetic lethal siRNA screen conducted in a broad panel of KRAS wild-type or mutant cells identified KRAS but not STK33 as essential for survival. We also obtained similar negative results using small molecule inhibitors of the STK33 kinase identified by high-throughput screening. Taken together, our findings refute earlier proposals that STK33 inhibition may be a useful therapeutic approach to target human KRAS mutant tumors.


Cancer Research | 2010

Preclinical Evaluation of AMG 900, a Novel Potent and Highly Selective Pan-Aurora Kinase Inhibitor with Activity in Taxane-Resistant Tumor Cell Lines

Marc Payton; Tammy L. Bush; Grace Tin-Yun Chung; Beth Ziegler; Patrick Eden; Patricia McElroy; Sandra L. Ross; Victor J. Cee; Holly L. Deak; Brian L. Hodous; Hanh Nho Nguyen; Philip R. Olivieri; Karina Romero; Laurie B. Schenkel; Annette Bak; Mary K. Stanton; Isabelle Dussault; Vinod F. Patel; Stephanie Geuns-Meyer; Robert Radinsky; Richard Kendall

In mammalian cells, the aurora kinases (aurora-A, -B, and -C) play essential roles in regulating cell division. The expression of aurora-A and -B is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis, making them attractive targets for anticancer therapy. AMG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser(10), a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes.


Journal of Medicinal Chemistry | 2008

Design, synthesis, and biological evaluation of potent c-Met inhibitors.

Noel D'angelo; Steven Bellon; Shon Booker; Yuan Cheng; Angela Coxon; Celia Dominguez; Ingrid M. Fellows; Douglas Hoffman; Randall W. Hungate; Paula Kaplan-Lefko; Matthew R. Lee; Chun Li; Longbin Liu; Elizabeth Rainbeau; Paul J. Reider; Karen Rex; Aaron C. Siegmund; Yaxiong Sun; Andrew Tasker; Ning Xi; Shimin Xu; Yajing Yang; Yihong Zhang; Teresa L. Burgess; Isabelle Dussault; Tae-Seong Kim

c-Met is a receptor tyrosine kinase that plays a key role in several cellular processes but has also been found to be overexpressed and mutated in different human cancers. Consequently, targeting this enzyme has become an area of intense research in drug discovery. Our studies began with the design and synthesis of novel pyrimidone 7, which was found to be a potent c-Met inhibitor. Subsequent SAR studies identified 22 as a more potent analog, whereas an X-ray crystal structure of 7 bound to c-Met revealed an unexpected binding conformation. This latter finding led to the development of a new series that featured compounds that were more potent both in vitro and in vivo than 22 and also exhibited different binding conformations to c-Met. Novel c-Met inhibitors have been designed, developed, and found to be potent in vitro and in vivo.


Journal of Medicinal Chemistry | 2008

Discovery of a Potent, Selective, and Orally Bioavailable c-Met Inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458)

Longbin Liu; Aaron C. Siegmund; Ning Xi; Paula Kaplan-Lefko; Karen Rex; April Chen; Jasmine Lin; Jodi Moriguchi; Loren Berry; Liyue Huang; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon; Matthew R. Lee; Roman Shimanovich; Annette Bak; Celia Dominguez; Mark H. Norman; Jean-Christophe Harmange; Isabelle Dussault; Tae-Seong Kim

Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.


Journal of Medicinal Chemistry | 2012

Structure-based design of novel class II c-Met inhibitors: 1. Identification of pyrazolone-based derivatives.

Mark H. Norman; Longbin Liu; Matthew R. Lee; Ning Xi; Ingrid M. Fellows; Noel D'angelo; Celia Dominguez; Karen Rex; Steven Bellon; T.S Kim; Isabelle Dussault

Deregulation of c-Met receptor tyrosine kinase activity leads to tumorigenesis and metastasis in animal models. More importantly, the identification of activating mutations in c-Met, as well as MET gene amplification in human cancers, points to c-Met as an important target for cancer therapy. We have previously described two classes of c-Met kinase inhibitors (class I and class II) that differ in their binding modes and selectivity profiles. The class II inhibitors tend to have activities on multiple kinases. Knowledge of the binding mode of these molecules in the c-Met protein led to the design and evaluation of several new class II c-Met inhibitors that utilize various 5-membered cyclic carboxamides to conformationally restrain key pharmacophoric groups within the molecule. These investigations resulted in the identification of a potent and novel class of pyrazolone c-Met inhibitors with good in vivo activity.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery and optimization of potent and selective triazolopyridazine series of c-Met inhibitors

Christiane Bode; Alessandro Boezio; Brian K. Albrecht; Steven Bellon; Loren Berry; Martin A. Broome; Deborah Choquette; Isabelle Dussault; Richard T. Lewis; Min-Hwa Jasmine Lin; Karen Rex; Douglas A. Whittington; Yajing Yang; Jean-Christophe Harmange

Deregulation of the receptor tyrosine kinase c-Met has been implicated in several human cancers and is an attractive target for small molecule drug discovery. We previously showed that O-linked triazolopyridazines can be potent inhibitors of c-Met. Herein, we report the discovery of a related series of N-linked triazolopyridazines which demonstrate nanomolar inhibition of c-Met kinase activity and display improved pharmacodynamic profiles. Specifically, the potent time-dependent inhibition of cytochrome P450 associated with the O-linked triazolopyridazines has been eliminated within this novel series of inhibitors. N-linked triazolopyridazine 24 exhibited favorable pharmacokinetics and displayed potent inhibition of HGF-mediated c-Met phosphorylation in a mouse liver PD model. Once-daily oral administration of 24 for 22days showed significant tumor growth inhibition in an NIH-3T3/TPR-Met xenograft mouse efficacy model.


Molecular Cancer Therapeutics | 2016

In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models

Paul E. Hughes; Karen Rex; Sean Caenepeel; Yajing Yang; Yihong Zhang; Martin A. Broome; Hue T. Kha; Teresa L. Burgess; Benny Amore; Paula Kaplan-Lefko; Jodi Moriguchi; Jonathan A. Werner; Michael A. Damore; Daniel M. Baker; Deborah Choquette; Jean-Christophe Harmange; Robert Radinsky; Richard Kendall; Isabelle Dussault; Angela Coxon

The MET receptor tyrosine kinase is involved in cell growth, survival, and invasion. Clinical studies with small molecule MET inhibitors have shown the role of biomarkers in identifying patients most likely to benefit from MET-targeted therapy. AMG 337 is an oral, small molecule, ATP-competitive, highly selective inhibitor of the MET receptor. Herein, we describe AMG 337 preclinical activity and mechanism of action in MET-dependent tumor models. These studies suggest MET is the only therapeutic target for AMG 337. In an unbiased tumor cell line proliferation screen (260 cell lines), a closely related analogue of AMG 337, Compound 5, exhibited activity in 2 of 260 cell lines; both were MET-amplified. Additional studies examining the effects of AMG 337 on the proliferation of a limited panel of cell lines with varying MET copy numbers revealed that high-level focal MET amplification (>12 copies) was required to confer MET oncogene addiction and AMG 337 sensitivity. One MET-amplified cell line, H1573 (>12 copies), was AMG 337 insensitive, possibly because of a downstream G12A KRAS mutation. Mechanism-of-action studies in sensitive MET-amplified cell lines demonstrated that AMG 337 inhibited MET and adaptor protein Gab-1 phosphorylation, subsequently blocking the downstream PI3K and MAPK pathways. AMG 337 exhibited potency in pharmacodynamic assays evaluating MET signaling in tumor xenograft models; >90% inhibition of Gab-1 phosphorylation was observed at 0.75 mg/kg. These findings describe the preclinical activity and mechanism of action of AMG 337 in MET-dependent tumor models and indicate its potential as a novel therapeutic for the treatment of MET-dependent tumors. Mol Cancer Ther; 15(7); 1568–79. ©2016 AACR.


Cancer Research | 2014

Abstract 728: AMG 337, a novel, potent and selective MET kinase inhibitor, has robust growth inhibitory activity in MET-dependent cancer models

Paul E. Hughes; Yajing Yang; Karen Rex; Yihong Zhang; Paula Kaplan-Lefko; Sean Caenepeel; Jodi Moriguchi; Martin A. Broome; Deborah Choquette; Robert Radinsky; Richard Kendall; Angela Coxon; Isabelle Dussault

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Signaling through the MET receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), promotes cell proliferation, survival, and invasion. Activation of MET signaling is a relatively common hallmark of a diverse range of human cancer types, and as a result, inhibition of MET signaling represents an attractive therapeutic opportunity for the treatment of cancer. In this study, we describe the characterization of AMG 337, a potent and highly selective small molecule ATP-competitive MET kinase inhibitor that demonstrates robust activity in MET-dependent cancer models. In enzymatic assays, AMG 337 inhibited MET kinase activity with an IC50 of 10 μM in all other tested cell lines. Further studies in an expanded panel of additional cancer cell lines derived from gastric, NSCLC, and esophageal cancer confirmed that the in-vitro anti-proliferative activity of AMG 337 correlated with amplification of MET. In those cell lines, treatment with AMG 337 inhibited downstream PI3K and MAPK signaling pathways, which translated into growth arrest as evidenced by an accumulation of cells in the G1 phase of the cell cycle, a concomitant reduction in DNA synthesis, and the induction of apoptosis. In vivo, oral administration of AMG 337 resulted in robust dose-dependent anti-tumor efficacy in MET amplified gastric cancer xenograft models, with inhibition of tumor growth consistent with the pharmacodynamic modulation of MET signaling. In conclusion, these findings illustrate the potential clinical utility of AMG 337 as a therapeutic agent for the treatment of tumors with evidence of dysregulated MET signaling, including MET amplification. A phase 1 clinical study is currently evaluating the safety, tolerability and pharmacokinectics of AMG 337 in patients with solid tumors. Citation Format: Paul E. Hughes, Yajing Yang, Karen Rex, Yihong Zhang, Paula J. Kaplan-Lefko, Sean Caenepeel, Jodi Moriguchi, Martin Broome, Deborah Choquette, Robert Radinsky, Richard Kendall, Angela Coxon, Isabelle Dussault. AMG 337, a novel, potent and selective MET kinase inhibitor, has robust growth inhibitory activity in MET-dependent cancer models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 728. doi:10.1158/1538-7445.AM2014-728


Cancer Research | 2011

STK33 Kinase Is Not Essential in KRAS-Dependent Cells–Response

Isabelle Dussault; Josette Carnahan; Carol Babij; Yihong Zhang; Vivienne Watson; Kim Quon; Paul Kassner

Our recently published work ([1][1]) was initiated based on the conclusion in Scholl and colleagues that “cells that are dependent on mutant KRAS exhibit sensitivity to suppression of STK33, irrespective of tissue of origin” and that “STK33 promotes cancer cell viability in a kinase activity

Researchain Logo
Decentralizing Knowledge