Network


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

Hotspot


Dive into the research topics where Melissa Chenard is active.

Publication


Featured researches published by Melissa Chenard.


Bioorganic & Medicinal Chemistry Letters | 2008

Exploration of the internal cavity of histone deacetylase (HDAC) with selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Prasun K. Chakravarty; Melissa Chenard; Joshua Close; Jonathan C. Cruz; William K. Dahlberg; Judith C. Fleming; Christopher Hamblett; Julie E. Hamill; Paul Harrington; Andreas Harsch; Richard Heidebrecht; Bethany Hughes; Joon Jung; Candia M. Kenific; Astrid M. Kral; Peter T. Meinke; Richard E. Middleton; Nicole Ozerova; David L. Sloman; Matthew G. Stanton; Alexander A. Szewczak; Sriram Tyagarajan; David J. Witter; J. Paul Secrist; Thomas A. Miller

We report herein the initial exploration of novel selective HDAC1/HDAC2 inhibitors (SHI-1:2). Optimized SHI-1:2 structures exhibit enhanced intrinsic activity against HDAC1 and HDAC2, and are greater than 100-fold selective versus other HDACs, including HDAC3. Based on the SAR of these agents and our current understanding of the HDAC active site, we postulate that the SHI-1:2 extend the existing HDAC inhibitor pharmacophore to include an internal binding domain.


BMC Medical Genomics | 2010

A gene expression signature of RAS pathway dependence predicts response to PI3K and RAS pathway inhibitors and expands the population of RAS pathway activated tumors

Andrey Loboda; Michael Nebozhyn; Rich Klinghoffer; Jason Frazier; Michael Chastain; William T. Arthur; Brian Roberts; Theresa Zhang; Melissa Chenard; Brian B. Haines; Jannik N. Andersen; Kumiko Nagashima; Cloud Paweletz; Bethany Lynch; Igor Feldman; Hongyue Dai; Pearl S. Huang; James Watters

BackgroundHyperactivation of the Ras signaling pathway is a driver of many cancers, and RAS pathway activation can predict response to targeted therapies. Therefore, optimal methods for measuring Ras pathway activation are critical. The main focus of our work was to develop a gene expression signature that is predictive of RAS pathway dependence.MethodsWe used the coherent expression of RAS pathway-related genes across multiple datasets to derive a RAS pathway gene expression signature and generate RAS pathway activation scores in pre-clinical cancer models and human tumors. We then related this signature to KRAS mutation status and drug response data in pre-clinical and clinical datasets.ResultsThe RAS signature score is predictive of KRAS mutation status in lung tumors and cell lines with high (> 90%) sensitivity but relatively low (50%) specificity due to samples that have apparent RAS pathway activation in the absence of a KRAS mutation. In lung and breast cancer cell line panels, the RAS pathway signature score correlates with pMEK and pERK expression, and predicts resistance to AKT inhibition and sensitivity to MEK inhibition within both KRAS mutant and KRAS wild-type groups. The RAS pathway signature is upregulated in breast cancer cell lines that have acquired resistance to AKT inhibition, and is downregulated by inhibition of MEK. In lung cancer cell lines knockdown of KRAS using siRNA demonstrates that the RAS pathway signature is a better measure of dependence on RAS compared to KRAS mutation status. In human tumors, the RAS pathway signature is elevated in ER negative breast tumors and lung adenocarcinomas, and predicts resistance to cetuximab in metastatic colorectal cancer.ConclusionsThese data demonstrate that the RAS pathway signature is superior to KRAS mutation status for the prediction of dependence on RAS signaling, can predict response to PI3K and RAS pathway inhibitors, and is likely to have the most clinical utility in lung and breast tumors.


Cancer Research | 2010

MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor

Bo-Sheng Pan; Grace K.Y. Chan; Melissa Chenard; An Chi; Lenora Davis; Sujal V. Deshmukh; Jackson B. Gibbs; Susana Gil; Gaozhen Hang; Harold Hatch; James P. Jewell; Ilona Kariv; Jason D. Katz; Kaiko Kunii; Wei Lu; Bart Lutterbach; Cloud P. Paweletz; Xianlu Qu; John F. Reilly; Alexander A. Szewczak; Qinwen Zeng; Nancy E. Kohl; Christopher J. Dinsmore

The receptor tyrosine kinase c-Met is an attractive target for therapeutic blockade in cancer. Here, we describe MK-2461, a novel ATP-competitive multitargeted inhibitor of activated c-Met. MK-2461 inhibited in vitro phosphorylation of a peptide substrate recognized by wild-type or oncogenic c-Met kinases (N1100Y, Y1230C, Y1230H, Y1235D, and M1250T) with IC(50) values of 0.4 to 2.5 nmol/L. In contrast, MK-2461 was several hundredfold less potent as an inhibitor of c-Met autophosphorylation at the kinase activation loop. In tumor cells, MK-2461 effectively suppressed constitutive or ligand-induced phosphorylation of the juxtamembrane domain and COOH-terminal docking site of c-Met, and its downstream signaling to the phosphoinositide 3-kinase-AKT and Ras-extracellular signal-regulated kinase pathways, without inhibiting autophosphorylation of the c-Met activation loop. BIAcore studies indicated 6-fold tighter binding to c-Met when it was phosphorylated, suggesting that MK-2461 binds preferentially to activated c-Met. MK-2461 displayed significant inhibitory activities against fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor, and other receptor tyrosine kinases. In cell culture, MK-2461 inhibited hepatocyte growth factor/c-Met-dependent mitogenesis, migration, cell scatter, and tubulogenesis. Seven of 10 MK-2461-sensitive tumor cell lines identified from a large panel harbored genomic amplification of MET or FGFR2. In a murine xenograft model of c-Met-dependent gastric cancer, a well-tolerated oral regimen of MK-2461 administered at 100 mg/kg twice daily effectively suppressed c-Met signaling and tumor growth. Similarly, MK-2461 inhibited the growth of tumors formed by s.c. injection of mouse NIH-3T3 cells expressing oncogenic c-Met mutants. Taken together, our findings support further preclinical development of MK-2461 for cancer therapy.


Cancer Cell | 2016

Long-Term ERK Inhibition in KRAS-Mutant Pancreatic Cancer Is Associated with MYC Degradation and Senescence-like Growth Suppression

Tikvah K. Hayes; Nicole F. Neel; Chaoxin Hu; Prson Gautam; Melissa Chenard; Brian Long; Meraj Aziz; Michelle Kassner; Kirsten L. Bryant; Mariaelena Pierobon; Raoud Marayati; Swapnil Kher; Samuel D. George; Mai Xu; Andrea Wang-Gillam; Ahmed A. Samatar; Anirban Maitra; Krister Wennerberg; Emanuel F. Petricoin; Hongwei H. Yin; Barry D. Nelkin; Adrienne D. Cox; Jen Jen Yeh; Channing J. Der

Induction of compensatory mechanisms and ERK reactivation has limited the effectiveness of Raf and MEK inhibitors in RAS-mutant cancers. We determined that direct pharmacologic inhibition of ERK suppressed the growth of a subset of KRAS-mutant pancreatic cancer cell lines and that concurrent phosphatidylinositol 3-kinase (PI3K) inhibition caused synergistic cell death. Additional combinations that enhanced ERK inhibitor action were also identified. Unexpectedly, long-term treatment of sensitive cell lines caused senescence, mediated in part by MYC degradation and p16 reactivation. Enhanced basal PI3K-AKT-mTOR signaling was associated with de novo resistance to ERK inhibitor, as were other protein kinases identified by kinome-wide siRNA screening and a genetic gain-of-function screen. Our findings reveal distinct consequences of inhibiting this kinase cascade at the level of ERK.


Bioorganic & Medicinal Chemistry Letters | 2008

SAR profiles of spirocyclic nicotinamide derived selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Christopher Hamblett; Dawn M. Mampreian; Joon Jung; Andreas Harsch; Alexander A. Szewczak; William K. Dahlberg; Richard E. Middleton; Bethany Hughes; Judith C. Fleming; Hongmei Wang; Astrid M. Kral; Nicole Ozerova; Jonathan C. Cruz; Brian B. Haines; Melissa Chenard; Candia M. Kenific; J. Paul Secrist; Thomas A. Miller

A potent family of spirocyclic nicotinyl aminobenzamide selective HDAC1/HDAC2 inhibitors (SHI-1:2) is profiled. The incorporation of a biaryl zinc-binding motif into a nicotinyl scaffold resulted in enhanced potency and selectivity versus HDAC3, but also imparted hERG activity. It was discovered that increasing polar surface area about the spirocycle attenuates this liability. Compound 12 induced a 4-fold increase in acetylated histone H2B in an HCT-116 xenograft model study with acute exposure, and inhibited tumor growth in a 21-day efficacy study with qd dosing.


Bioorganic & Medicinal Chemistry Letters | 2008

Phenylglycine and phenylalanine derivatives as potent and selective HDAC1 inhibitors (SHI-1)

Kevin J. Wilson; David J. Witter; Jonathan Grimm; Phieng Siliphaivanh; Karin M. Otte; Astrid M. Kral; Judith C. Fleming; Andreas Harsch; Julie E. Hamill; Jonathan C. Cruz; Melissa Chenard; Alexander A. Szewczak; Richard E. Middleton; Bethany Hughes; William K. Dahlberg; J. Paul Secrist; Thomas A. Miller

An HTS screening campaign identified a series of low molecular weight phenols that showed excellent selectivity (>100-fold) for HDAC1/HDAC2 over other Class I and Class II HDACs. Evolution and optimization of this HTS hit series provided HDAC1-selective (SHI-1) compounds with excellent anti-proliferative activity and improved physical properties. Dose-dependent efficacy in a mouse HCT116 xenograft model was demonstrated with a phenylglycine SHI-1 analog.


Journal of Medicinal Chemistry | 2013

Discovery of 1-[3-(1-Methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5]cyclohepta[1,2-b]pyridin-7-yl]-N-(pyridin-2-ylmethyl)methanesulfonamide (MK-8033): A Specific c-Met/Ron Dual Kinase Inhibitor with Preferential Affinity for the Activated State of c-Met

Alan B. Northrup; Matthew H. Katcher; Michael D. Altman; Melissa Chenard; Matthew H. Daniels; Sujal V. Deshmukh; Danielle Falcone; David J. Guerin; Harold Hatch; Chaomin Li; Wei Lu; Bart Lutterbach; Timothy J. Allison; Sangita B. Patel; John F. Reilly; Michael H. Reutershan; Keith Rickert; Craig Rosenstein; Stephen M. Soisson; Alexander A. Szewczak; Deborah Walker; Kevin J. Wilson; Jonathan R. Young; Bo Sheng Pan; Christopher J. Dinsmore

This report documents the first example of a specific inhibitor of protein kinases with preferential binding to the activated kinase conformation: 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one 11r (MK-8033), a dual c-Met/Ron inhibitor under investigation as a treatment for cancer. The design of 11r was based on the desire to reduce time-dependent inhibition of CYP3A4 (TDI) by members of this structural class. A novel two-step protocol for the synthesis of benzylic sulfonamides was developed to access 11r and analogues. We provide a rationale for the observed selectivity based on X-ray crystallographic evidence and discuss selectivity trends with additional examples. Importantly, 11r provides full inhibition of tumor growth in a c-Met amplified (GTL-16) subcutaneous tumor xenograft model and may have an advantage over inactive form kinase inhibitors due to equal potency against a panel of oncogenic activating mutations of c-Met in contrast to c-Met inhibitors without preferential binding to the active kinase conformation.


Bioorganic & Medicinal Chemistry Letters | 2009

Parallel medicinal chemistry approaches to selective HDAC1/HDAC2 inhibitor (SHI-1:2) optimization

Solomon Kattar; Laura Surdi; Anna A. Zabierek; Joey L. Methot; Richard E. Middleton; Bethany Hughes; Alexander A. Szewczak; William K. Dahlberg; Astrid M. Kral; Nicole Ozerova; Judith C. Fleming; Hongmei Wang; Paul Secrist; Andreas Harsch; Julie E. Hamill; Jonathan C. Cruz; Candia M. Kenific; Melissa Chenard; Thomas A. Miller; Scott C. Berk; Paul Tempest

The successful application of both solid and solution phase library synthesis, combined with tight integration into the medicinal chemistry effort, resulted in the efficient optimization of a novel structural series of selective HDAC1/HDAC2 inhibitors by the MRL-Boston Parallel Medicinal Chemistry group. An initial lead from a small parallel library was found to be potent and selective in biochemical assays. Advanced compounds were the culmination of iterative library design and possess excellent biochemical and cellular potency, as well as acceptable PK and efficacy in animal models.


Biochemistry | 2014

Divergent Kinetics Differentiate the Mechanism of Action of Two HDAC Inhibitors

Astrid M. Kral; Nicole Ozerova; Joshua Close; Joon Jung; Melissa Chenard; Judith C. Fleming; Brian B. Haines; Paul Harrington; John Maclean; Thomas A. Miller; Paul Secrist; Hongmei Wang; Richard Heidebrecht

Histone deacetylases (HDACs) play diverse roles in many diseases including cancer, sarcopenia, and Alzheimers. Different isoforms of HDACs appear to play disparate roles in the cell and are associated with specific diseases; as such, a substantial effort has been made to develop isoform-selective HDAC inhibitors. Our group focused on developing HDAC1/HDAC2-specific inhibitors as a cancer therapeutic. In the course of characterizing the mechanism of inhibition of a novel HDAC1/2-selective inhibitor, it was determined that it did not exhibit classical Michaelis-Menten kinetic behavior; this result is in contrast to the seminal HDAC inhibitor SAHA. Enzymatic assays, along with a newly developed binding assay, were used to determine the rates of binding and the affinities of both the HDAC1/2-selective inhibitor and SAHA. The mechanism of action studies identified a potential conformational change required for optimal binding by the selective inhibitor. A model of this putative conformational change is proposed.


Bioorganic & Medicinal Chemistry Letters | 2009

Exploring the pharmacokinetic properties of phosphorus-containing selective HDAC 1 and 2 inhibitors (SHI-1:2).

Richard Heidebrecht; Melissa Chenard; Joshua Close; William K. Dahlberg; Judith C. Fleming; Jonathan Grimm; Julie E. Hamill; Andreas Harsch; Brian B. Haines; Bethany Hughes; Astrid M. Kral; Richard E. Middleton; Chandrasekhar Mushti; Nicole Ozerova; Alexander A. Szewczak; Hongmei Wang; Kevin J. Wilson; David J. Witter; J. Paul Secrist; Thomas A. Miller

We report the preparation and structure-activity relationships of phosphorus-containing histone deacetylase inhibitors. A strong trend between decreasing phosphorus functional group size and superior mouse pharmacokinetic properties was identified. In addition, optimized candidates showed tumor growth inhibition in xenograft studies.

Researchain Logo
Decentralizing Knowledge