Network


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

Hotspot


Dive into the research topics where Nathalie Rioux is active.

Publication


Featured researches published by Nathalie Rioux.


Nature Chemical Biology | 2015

A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models.

Elayne Chan-Penebre; Kristy G Kuplast; Christina R. Majer; P. Ann Boriack-Sjodin; Tim J. Wigle; L. Danielle Johnston; Nathalie Rioux; Michael John Munchhof; Lei Jin; Suzanne L. Jacques; Kip A West; Trupti Lingaraj; Kimberly Stickland; Scott Ribich; Alejandra Raimondi; Margaret Porter Scott; Nigel J. Waters; Roy M. Pollock; Jesse J. Smith; Olena Barbash; Melissa B. Pappalardi; Thau Ho; Kelvin Nurse; Khyati P Oza; Kathleen T Gallagher; Ryan G. Kruger; Mikel P. Moyer; Robert A. Copeland; Richard Chesworth; Kenneth W. Duncan

Protein arginine methyltransferase-5 (PRMT5) is reported to have a role in diverse cellular processes, including tumorigenesis, and its overexpression is observed in cell lines and primary patient samples derived from lymphomas, particularly mantle cell lymphoma (MCL). Here we describe the identification and characterization of a potent and selective inhibitor of PRMT5 with antiproliferative effects in both in vitro and in vivo models of MCL. EPZ015666 (GSK3235025) is an orally available inhibitor of PRMT5 enzymatic activity in biochemical assays with a half-maximal inhibitory concentration (IC50) of 22 nM and broad selectivity against a panel of other histone methyltransferases. Treatment of MCL cell lines with EPZ015666 led to inhibition of SmD3 methylation and cell death, with IC50 values in the nanomolar range. Oral dosing with EPZ015666 demonstrated dose-dependent antitumor activity in multiple MCL xenograft models. EPZ015666 represents a validated chemical probe for further study of PRMT5 biology and arginine methylation in cancer and other diseases.


ACS Medicinal Chemistry Letters | 2015

EPZ011989, A Potent, Orally-Available EZH2 Inhibitor with Robust in Vivo Activity

John E. Campbell; Kevin Wayne Kuntz; Sarah K. Knutson; Natalie Warholic; Heike Keilhack; Tim J. Wigle; Alejandra Raimondi; Christine R. Klaus; Nathalie Rioux; Akira Yokoi; Satoshi Kawano; Yukinori Minoshima; Hyeong-wook Choi; Margaret Porter Scott; Nigel J. Waters; Jesse J. Smith; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland

Inhibitors of the protein methyltransferase Enhancer of Zeste Homolog 2 (EZH2) may have significant therapeutic potential for the treatment of B cell lymphomas and other cancer indications. The ability of the scientific community to explore fully the spectrum of EZH2-associated pathobiology has been hampered by the lack of in vivo-active tool compounds for this enzyme. Here we report the discovery and characterization of EPZ011989, a potent, selective, orally bioavailable inhibitor of EZH2 with useful pharmacokinetic properties. EPZ011989 demonstrates significant tumor growth inhibition in a mouse xenograft model of human B cell lymphoma. Hence, this compound represents a powerful tool for the expanded exploration of EZH2 activity in biology.


ACS Medicinal Chemistry Letters | 2015

Aryl Pyrazoles as Potent Inhibitors of Arginine Methyltransferases: Identification of the First PRMT6 Tool Compound.

Lorna Helen Mitchell; A.E Drew; S.A Ribich; Nathalie Rioux; K.K Swinger; Suzanne L. Jacques; T Lingaraj; P.A Boriack-Sjodin; Nigel J. Waters; Tim J. Wigle; O Moradei; Lei Jin; Thomas V. Riera; M Porter-Scott; Mikel P. Moyer; Jesse J. Smith; Richard Chesworth; Robert A. Copeland

A novel aryl pyrazole series of arginine methyltransferase inhibitors has been identified. Synthesis of analogues within this series yielded the first potent, selective, small molecule PRMT6 inhibitor tool compound, EPZ020411. PRMT6 overexpression has been reported in several cancer types suggesting that inhibition of PRMT6 activity may have therapeutic utility. Identification of EPZ020411 provides the field with the first small molecule tool compound for target validation studies. EPZ020411 shows good bioavailability following subcutaneous dosing in rats making it a suitable tool for in vivo studies.


Antimicrobial Agents and Chemotherapy | 2014

Preclinical Profile of BI 224436, a Novel HIV-1 Non-Catalytic Site Integrase Inhibitor

Craig Fenwick; Ma’an Amad; Murray D. Bailey; Richard C. Bethell; Michael Bös; Pierre R. Bonneau; Michael G. Cordingley; René Coulombe; Jianmin Duan; Paul Edwards; Lee Fader; Anne-Marie Faucher; Michel Garneau; Araz Jakalian; Stephen H. Kawai; Louie Lamorte; Steven R. LaPlante; Laibin Luo; Steve Mason; Marc-André Poupart; Nathalie Rioux; Patricia Schroeder; Bruno Simoneau; Sonia Tremblay; Youla S. Tsantrizos; Myriam Witvrouw; Christiane Yoakim

ABSTRACT BI 224436 is an HIV-1 integrase inhibitor with effective antiviral activity that acts through a mechanism that is distinct from that of integrase strand transfer inhibitors (INSTIs). This 3-quinolineacetic acid derivative series was identified using an enzymatic integrase long terminal repeat (LTR) DNA 3′-processing assay. A combination of medicinal chemistry, parallel synthesis, and structure-guided drug design led to the identification of BI 224436 as a candidate for preclinical profiling. It has antiviral 50% effective concentrations (EC50s) of <15 nM against different HIV-1 laboratory strains and cellular cytotoxicity of >90 μM. BI 224436 also has a low, ∼2.1-fold decrease in antiviral potency in the presence of 50% human serum and, by virtue of a steep dose-response curve slope, exhibits serum-shifted EC95 values ranging between 22 and 75 nM. Passage of virus in the presence of inhibitor selected for either A128T, A128N, or L102F primary resistance substitutions, all mapping to a conserved allosteric pocket on the catalytic core of integrase. BI 224436 also retains full antiviral activity against recombinant viruses encoding INSTI resistance substitutions N155S, Q148H, and E92Q. In drug combination studies performed in cellular antiviral assays, BI 224436 displays an additive effect in combination with most approved antiretrovirals, including INSTIs. BI 224436 has drug-like in vitro absorption, distribution, metabolism, and excretion (ADME) properties, including Caco-2 cell permeability, solubility, and low cytochrome P450 inhibition. It exhibited excellent pharmacokinetic profiles in rat (clearance as a percentage of hepatic flow [CL], 0.7%; bioavailability [F], 54%), monkey (CL, 23%; F, 82%), and dog (CL, 8%; F, 81%). Based on the excellent biological and pharmacokinetic profile, BI 224436 was advanced into phase 1 clinical trials.


ACS Medicinal Chemistry Letters | 2016

Novel Oxindole Sulfonamides and Sulfamides: EPZ031686, the First Orally Bioavailable Small Molecule SMYD3 Inhibitor.

Lorna Helen Mitchell; P. Ann Boriack-Sjodin; Sherri Smith; Michael Thomenius; Nathalie Rioux; Michael John Munchhof; James E. Mills; Christine R. Klaus; Jennifer Totman; Thomas V. Riera; Alejandra Raimondi; Suzanne L. Jacques; Kip West; Megan Alene Cloonan Foley; Nigel J. Waters; Kevin Wayne Kuntz; Tim J. Wigle; Margaret Porter Scott; Robert A. Copeland; Jesse J. Smith; Richard Chesworth

SMYD3 has been implicated in a range of cancers; however, until now no potent selective small molecule inhibitors have been available for target validation studies. A novel oxindole series of SMYD3 inhibitors was identified through screening of the Epizyme proprietary histone methyltransferase-biased library. Potency optimization afforded two tool compounds, sulfonamide EPZ031686 and sulfamide EPZ030456, with cellular potency at a level sufficient to probe the in vitro biology of SMYD3 inhibition. EPZ031686 shows good bioavailability following oral dosing in mice making it a suitable tool for potential in vivo target validation studies.


ACS Medicinal Chemistry Letters | 2016

Structure and Property Guided Design in the Identification of PRMT5 Tool Compound EPZ015666

Kenneth W. Duncan; Nathalie Rioux; P. Ann Boriack-Sjodin; Michael John Munchhof; Lawrence A. Reiter; Christina R. Majer; Lei Jin; L. Danielle Johnston; Elayne Chan-Penebre; Kristy G Kuplast; Margaret Porter Scott; Roy M. Pollock; Nigel J. Waters; Jesse J. Smith; Mikel P. Moyer; Robert A. Copeland; Richard Chesworth

The recent publication of a potent and selective inhibitor of protein methyltransferase 5 (PRMT5) provides the scientific community with in vivo-active tool compound EPZ015666 (GSK3235025) to probe the underlying pharmacology of this key enzyme. Herein, we report the design and optimization strategies employed on an initial hit compound with poor in vitro clearance to yield in vivo tool compound EPZ015666 and an additional potent in vitro tool molecule EPZ015866 (GSK3203591).


Bioorganic & Medicinal Chemistry Letters | 2013

Structure-based design of novel HCV NS5B thumb pocket 2 allosteric inhibitors with submicromolar gt1 replicon potency: Discovery of a quinazolinone chemotype

Pierre L. Beaulieu; René Coulombe; Jianmin Duan; Gulrez Fazal; Cédrickx Godbout; Oliver Hucke; Araz Jakalian; Marc-André Joly; Olivier Lepage; Montse Llinas-Brunet; Julie Naud; Martin Poirier; Nathalie Rioux; Bounkham Thavonekham; George Kukolj; Timothy Stammers

We describe the structure-based design of a novel lead chemotype that binds to thumb pocket 2 of HCV NS5B polymerase and inhibits cell-based gt1 subgenomic reporter replicons at sub-micromolar concentrations (EC50<200nM). This new class of potent thumb pocket 2 inhibitors features a 1H-quinazolin-4-one scaffold derived from hybridization of a previously reported, low affinity thiazolone chemotype with our recently described anthranilic acid series. Guided by X-ray structural information, a key NS5B-ligand interaction involving the carboxylate group of anthranilic acid based inhibitors was replaced by a neutral two-point hydrogen bonding interaction between the quinazolinone scaffold and the protein backbone. The in vitro ADME and in vivo rat PK profile of representative analogs are also presented and provide areas for future optimization of this new class of HCV polymerase inhibitors.


Drug Metabolism and Disposition | 2016

Physiologically Based Pharmacokinetic Modeling in Pediatric Oncology Drug Development

Nathalie Rioux; Nigel J. Waters

Childhood cancer represents more than 100 rare and ultra-rare diseases, with an estimated 12,400 new cases diagnosed each year in the United States. As such, this much smaller patient population has led to pediatric oncology drug development lagging behind that for adult cancers. Developing drugs for pediatric malignancies also brings with it a number of unique trial design considerations, including flexible enrollment approaches, age-appropriate formulation, acceptable sampling schedules, and balancing the need for age-stratified dosing regimens, given the smaller patient populations. The regulatory landscape for pediatric pharmacotherapy has evolved with U.S. Food and Drug Administration (FDA) legislation such as the 2012 FDA Safety and Innovation Act. In parallel, regulatory authorities have recommended the application of physiologically based pharmacokinetic (PBPK) modeling, for example, in the recently issued FDA Strategic Plan for Accelerating the Development of Therapies for Pediatric Rare Diseases. PBPK modeling provides a quantitative and systems-based framework that allows the effects of intrinsic and extrinsic factors on drug exposure to be modeled in a mechanistic fashion. The application of PBPK modeling in drug development for pediatric cancers is relatively nascent, with several retrospective analyses of cytotoxic therapies, and latterly for targeted agents such as obatoclax and imatinib. More recently, we have employed PBPK modeling in a prospective manner to inform the first pediatric trials of pinometostat and tazemetostat in genetically defined populations (mixed lineage leukemia–rearranged and integrase interactor-1–deficient sarcomas, respectively). In this review, we evaluate the application of PBPK modeling in pediatric cancer drug development and discuss the important challenges that lie ahead in this field.


Xenobiotica | 2012

A high throughput in vitro mrp2 assay to predict in vivo biliary excretion

Federico Colombo; Catherine Armstrong; Jianmin Duan; Nathalie Rioux

Prediction of biliary excretion is a challenge for drug discovery scientists due to the lack of in vitro assays. This study explores the possibility of establishing a simple assay to predict in vivo biliary excretion via the mrp2 transport system. In vitro mrp2 activity was determined by measuring the ATP-dependent uptake of 5(6)-carboxy-2′,7′-dichlorofluorescein (CDCF) in canalicular plasma membrane vesicles (cLPM) from rat livers. The CDCF uptake was time- and concentration-dependent (Km of 2.2 ± 0.3 µM and Vmax of 115 ± 26 pmol/mg/min) and strongly inhibited by the mrp2 inhibitors, benzbromarone, MK-571, and cyclosporine A, with IC50 values ≤ 1.1 µM. Low inhibition of CDCF uptake by taurocholate (BSEP inhibitor; 57 µM) and digoxin (P-gp inhibitor; 101 µM) demonstrated assay specificity towards mrp2. A highly significant correlation (r2 = 0.959) between the in vitro IC50 values from the described mrp2 assay and in vivo biliary excretion in rats was observed using 10 literature compounds. This study demonstrated, for the first time, that a high throughput assay could be established with the capability of predicting biliary excretion in the rat using CDCF as a substrate.


Xenobiotica | 2016

Species differences in metabolism of EPZ015666, an oxetane-containing protein arginine methyltransferase-5 (PRMT5) inhibitor

Nathalie Rioux; Kenneth W. Duncan; Ronald J. Lantz; Xiusheng Miao; Elayne Chan-Penebre; Mikel P. Moyer; Michael John Munchhof; Robert A. Copeland; Richard Chesworth; Nigel J. Waters

Abstract 1. Metabolite profiling and identification studies were conducted to understand the cross-species differences in the metabolic clearance of EPZ015666, a first-in-class protein arginine methyltransferase-5 (PRMT5) inhibitor, with anti-proliferative effects in preclinical models of Mantle Cell Lymphoma. EPZ015666 exhibited low clearance in human, mouse and rat liver microsomes, in part by introduction of a 3-substituted oxetane ring on the molecule. In contrast, a higher clearance was observed in dog liver microsomes (DLM) that translated to a higher in vivo clearance in dog compared with rodent. 2. Structure elucidation via high resolution, accurate mass LC-MSn revealed that the prominent metabolites of EPZ015666 were present in hepatocytes from all species, with the highest turnover rate in dogs. M1 and M2 resulted from oxidative oxetane ring scission, whereas M3 resulted from loss of the oxetane ring via an N-dealkylation reaction. 3. The formation of M1 and M2 in DLM was significantly abrogated in the presence of the specific CYP2D inhibitor, quinidine, and to a lesser extent by the CYP3A inhibitor, ketoconazole, corroborating data from human recombinant isozymes. 4. Our data indicate a marked species difference in the metabolism of the PRMT5 inhibitor EPZ015666, with oxetane ring scission the predominant metabolic pathway in dog mediated largely by CYP2D.

Collaboration


Dive into the Nathalie Rioux's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tim J. Wigle

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge