Network


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

Hotspot


Dive into the research topics where Anne Roulston is active.

Publication


Featured researches published by Anne Roulston.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis

Mai Nguyen; Richard C. Marcellus; Anne Roulston; Mark A. Watson; Lucile Serfass; S. R. Murthy Madiraju; Daniel Goulet; Jean Viallet; Laurent Belec; Xavier Billot; Stephane Acoca; Enrico O. Purisima; Adrian Wiegmans; Leonie A. Cluse; Ricky W. Johnstone; Pierre Beauparlant; Gordon C. Shore

Elevated expression of members of the BCL-2 pro-survival family of proteins can confer resistance to apoptosis in cancer cells. Small molecule obatoclax (GX15-070), which is predicted to occupy a hydrophobic pocket within the BH3 binding groove of BCL-2, antagonizes these members and induces apoptosis, dependent on BAX and BAK. Reconstitution in yeast confirmed that obatoclax acts on the pathway and overcomes BCL-2-, BCL-XL-, BCL-w-, and MCL-1-mediated resistance to BAX or BAK. The compound potently interfered with the direct interaction between MCL-1 and BAK in intact mitochondrial outer membrane and inhibited the association between MCL-1 and BAK in intact cells. MCL-1 has been shown to confer resistance to the BCL-2/BCL-XL/BCL-w-selective antagonist ABT-737 and to the proteasome inhibitor bortezomib. In both cases, this resistance was overcome by obatoclax. These findings support a rational clinical development opportunity for the compound in cancer indications or treatments where MCL-1 contributes to resistance to cell killing.


Molecular and Cellular Biology | 2009

The Small Molecule GMX1778 Is a Potent Inhibitor of NAD+ Biosynthesis: Strategy for Enhanced Therapy in Nicotinic Acid Phosphoribosyltransferase 1-Deficient Tumors

Mark A. Watson; Anne Roulston; Laurent Bélec; Xavier Billot; Richard C. Marcellus; Dominique Bédard; Cynthia Bernier; Stéphane Branchaud; Helen S. L. Chan; Kenza Dairi; Daniel Goulet; Michel-Olivier Gratton; Henady Isakau; Anne Jang; Abdelkrim Khadir; Elizabeth Koch; Manon Lavoie; Michael Lawless; Mai Nguyen; Denis Paquette; Émilie Turcotte; Alvin Berger; Matthew W. Mitchell; Gordon C. Shore; Pierre Beauparlant

ABSTRACT GMX1777 is a prodrug of the small molecule GMX1778, currently in phase I clinical trials for the treatment of cancer. We describe findings indicating that GMX1778 is a potent and specific inhibitor of the NAD+ biosynthesis enzyme nicotinamide phosphoribosyltransferase (NAMPT). Cancer cells have a very high rate of NAD+ turnover, which makes NAD+ modulation an attractive target for anticancer therapy. Selective inhibition by GMX1778 of NAMPT blocks the production of NAD+ and results in tumor cell death. Furthermore, GMX1778 is phosphoribosylated by NAMPT, which increases its cellular retention. The cytotoxicity of GMX1778 can be bypassed with exogenous nicotinic acid (NA), which permits NAD+ repletion via NA phosphoribosyltransferase 1 (NAPRT1). The cytotoxicity of GMX1778 in cells with NAPRT1 deficiency, however, cannot be rescued by NA. Analyses of NAPRT1 mRNA and protein levels in cell lines and primary tumor tissue indicate that high frequencies of glioblastomas, neuroblastomas, and sarcomas are deficient in NAPRT1 and not susceptible to rescue with NA. As a result, the therapeutic index of GMX1777 can be widended in the treatment animals bearing NAPRT1-deficient tumors by coadministration with NA. This provides the rationale for a novel therapeutic approach for the use of GMX1777 in the treatment of human cancers.


Anti-Cancer Drugs | 2009

Preclinical development of the nicotinamide phosphoribosyl transferase inhibitor prodrug Gmx1777

Pierre Beauparlant; Dominique Bédard; Cynthia Bernier; Helen S. L. Chan; Daniel Goulet; Michel-Olivier Gratton; Manon Lavoie; Anne Roulston; Émilie Turcotte; Mark A. Watson

GMX1778 was recently shown to function as a potent inhibitor of nicotinamide phosphoribosyl transferase. To translate the discovery of GMX1778 mechanism of action into optimal clinical use of its intravenously administered prodrug, GMX1777, the efficacy of GMX1777 was evaluated in xenograft models and the pharmacokinetic profile of GMX1778 and its effect on nicotinamide adenine dinucleotide cellular levels was measured by liquid chromatography/mass spectrometry. Consistent with the requirement for a prolonged exposure for cytotoxicity in vitro, a dose of 75 mg/kg of GMX1777 administered as two bolus intravenous injections in 1 day were not effective at reducing the growth of multiple myeloma (IM-9) tumors, whereas the same dose of GMX1777 administered over a 24 h intravenous infusion caused tumor regression in the IM-9 model, a small-cell lung cancer (SHP-77) model, and a colon carcinoma (HCT-116) model. A 72 h continuous intravenous infusion of GMX1777 was also effective in the IM-9 model, but was associated with a smaller therapeutic index. GMX1777 at a dose of 75 mg/kg administered over a 24 h intravenous infusion produced GMX1778 steady-state plasma levels of approximately 1 μg/ml and caused nicotinamide adenine dinucleotide levels to decrease significantly in tumors. Consistent with the GMX1778 mechanism of action, nicotinic acid protected mice treated with a lethal dose of GMX1777. These data support the design of an open-label, dose-escalation trial, in which patients with refractory solid tumors and lymphomas receive 24 h infusions of GMX1777 as a single agent in 3-week cycles. Furthermore, these results indicate that nicotinic acid is a potent antidote to treat GMX1777 overdose.


EMBO Reports | 2013

Programming cancer cells for high expression levels of Mcl1

Franziska Ertel; Mai Nguyen; Anne Roulston; Gordon C. Shore

The Bcl2 pro‐survival protein family has long been recognized for its important contributions to cancer. At elevated levels relative to pro‐apoptotic effector members, the survival proteins prevent cancer cells from initiating apoptosis in the face of many intrinsic tumour‐suppressing pathways and extrinsic therapeutic treatments aimed at controlling tumorigenesis. Recent studies, including genome‐wide analyses, have begun to focus attention on a particularly enigmatic member of the family—myeloid cell leukaemia 1 (Mcl1). For reasons that are not clear, Mcl1 in cancer cells is turned over rapidly, eliminated primarily through the ubiquitin–proteasome pathway. Moreover, the mechanistic aspects of this constitutive membrane‐associated protein have not been fully elucidated. As the pro‐cancer activity of Mcl1 requires elevated expression levels of the protein, the cancer genome adapts to ensure either high levels of synthesis or evasion of degradation, or both. Here, we focus on the complex strategies at play and their therapeutic implications.


Cancer Research | 2014

Synergy between the NAMPT Inhibitor GMX1777(8) and Pemetrexed in Non–Small Cell Lung Cancer Cells Is Mediated by PARP Activation and Enhanced NAD Consumption

Manuel Chan; Michel Gravel; Alexandre Bramoullé; Gaëlle Bridon; Daina Avizonis; Gordon C. Shore; Anne Roulston

GMX1778 and its prodrug GMX1777 represent a new class of cancer drugs that targets nicotinamide phosphoribosyltransferase (NAMPT) as a new strategy to interfere with biosynthesis of the key enzymatic cofactor NAD, which is critical for a number of cell functions, including DNA repair. Using a genome-wide synthetic lethal siRNA screen, we identified the folate pathway-related genes, deoxyuridine triphosphatase and dihydrofolate reductase, the silencing of which sensitized non-small cell lung carcinoma (NSCLC) cells to the cytotoxic effects of GMX. Pemetrexed is an inhibitor of dihydrofolate reductase currently used to treat patients with nonsquamous NSCLC. We found that combining pemetrexed with GMX1777 produced a synergistic therapeutic benefit in A549 and H1299 NSCLC cells in vitro and in a mouse A549 xenograft model of lung cancer. Pemetrexed is known to activate PARPs, thereby accelerating NAD consumption. Genetic or pharmacologic blockade of PARP activity inhibited this effect, impairing cell death by pemetrexed either alone or in combination with GMX1777. Conversely, inhibiting the base excision repair pathway accentuated NAD decline in response to GMX and the cytotoxicity of both agents either alone or in combination. These findings provide a mechanistic rationale for combining GMX1777 with pemetrexed as an effective new therapeutic strategy to treat nonsquamous NSCLC.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Translation initiation factor eIF4F modifies the dexamethasone response in multiple myeloma

Francis Robert; William Roman; Alexandre Bramoullé; Christof Fellmann; Anne Roulston; Chaim Shustik; John A. Porco; Gordon C. Shore; Michael Sebag; Jerry Pelletier

Significance Multiple myeloma (MM) is a cancer that develops in the bone marrow and remains incurable to this day. It is a cancer type that shows hallmarks of deregulated protein synthesis control. To uncover new vulnerabilities in this disease, we performed a focused RNAi screen to identify components of the translation apparatus that, when depleted, would sensitize tumor cells to dexamethasone (DEX), a component of frontline therapy in this cancer. We found that suppression of eukaryotic initiation factor 4F, a heterotrimeric complex required for cap-dependent translation initiation, is a modifier of the DEX response in MM. Our efforts uncover a previously unidentified vulnerability in MM that should be explored clinically. Enhanced protein synthesis capacity is associated with increased tumor cell survival, proliferation, and resistance to chemotherapy. Cancers like multiple myeloma (MM), which display elevated activity in key translation regulatory nodes, such as the PI3K/mammalian target of rapamycin and MYC-eukaryotic initiation factor (eIF) 4E pathways, are predicted to be particularly sensitive to therapeutic strategies that target this process. To identify novel vulnerabilities in MM, we undertook a focused RNAi screen in which components of the translation apparatus were targeted. Our screen was designed to identify synthetic lethal relationships between translation factors or regulators and dexamethasone (DEX), a corticosteroid used as frontline therapy in this disease. We find that suppression of all three subunits of the eIF4F cap-binding complex synergizes with DEX in MM to induce cell death. Using a suite of small molecules that target various activities of eIF4F, we observed that cell survival and DEX resistance are attenuated upon eIF4F inhibition in MM cell lines and primary human samples. Levels of MYC and myeloid cell leukemia 1, two known eIF4F-responsive transcripts and key survival factors in MM, were reduced upon eIF4F inhibition, and their independent suppression also synergized with DEX. Inhibition of eIF4F in MM exerts pleotropic effects unraveling a unique therapeutic opportunity.


Science Signaling | 2013

BIM, PUMA, and the Achilles’ Heel of Oncogene Addiction

Anne Roulston; William J. Muller; Gordon C. Shore

Drugs that target oncogenic receptor tyrosine kinases activate parallel signaling pathways that trigger cell death. Cancer cells undergo extensive genetic and epigenetic rewiring to support the malignant phenotype, and yet cell survival and proliferation often remain dependent on one or a limited number of driver mutations. This is the concept of oncogene addiction, the elucidation of which has led to substantial progress in therapeutic interventions. However, because resistance mechanisms often emerge, explicating the pathways that connect therapeutic oncogene inactivation to the cell death machinery is critical to exploiting additional synthetic lethal opportunities.


Molecular and Cellular Oncology | 2016

New strategies to maximize therapeutic opportunities for NAMPT inhibitors in oncology

Anne Roulston; Gordon C. Shore

ABSTRACT Nicotinamide phosphoribosyltransferase (NAMPT) is crucial for nicotinamide adenine dinucleotide (NAD+) biosynthesis in mammalian cells. NAMPT inhibitors represent multifunctional anticancer agents that act on NAD+ metabolism to shut down glycolysis, nucleotide biosynthesis, and ATP generation and act indirectly as PARP and sirtuin inhibitors. The selectivity of NAMPT inhibitors preys on the increased metabolic requirements to replenish NAD+ in cancer cells. Although initial clinical studies with NAMPT inhibitors did not achieve single-agent therapeutic levels before dose-limiting toxicities were reached, a new understanding of alternative rescue pathways and a biomarker that can be used to select patients provides new opportunities to widen the therapeutic window and achieve efficacious doses in the clinic. Recent work has also illustrated the potential for drug combination strategies to further enhance the therapeutic opportunities. This review summarizes recent discoveries in NAD+/NAMPT inhibitor biology in the context of exploiting this new knowledge to optimize the clinical outcomes for this promising new class of agents.


Science Translational Medicine | 2016

The synthetic diazonamide DZ-2384 has distinct effects on microtubule curvature and dynamics without neurotoxicity.

Michal Wieczorek; Joseph Tcherkezian; Cynthia Bernier; Andrea E. Prota; Sami Chaaban; Yannève Rolland; Claude Godbout; Mark A. Hancock; Joseph C. Arezzo; Ozhan Ocal; Cecilia Rocha; Natacha Olieric; Anita Hall; Hui Ding; Alexandre Bramoullé; Matthew G. Annis; George Zogopoulos; Patrick G. Harran; Thomas M. Wilkie; Rolf A. Brekken; Peter M. Siegel; Michel O. Steinmetz; Gordon C. Shore; Gary J. Brouhard; Anne Roulston

A compound that binds to tubulin in an unusual way has superior antitumor efficacy and safety and has a distinctive impact on microtubule curvature and dynamics. Throwing a curve ball to cancer Drugs such as vinca alkaloids, which target tubulin and interfere with microtubule function in mitosis, are commonly used for the treatment of cancer. Unfortunately, they also damage microtubules in normal undividing cells including neurons, resulting in toxicity. Wieczorek et al. identified a drug called DZ-2384, which may offer a safer alternative to the vincas. The authors found that although DZ-2384 is very effective at targeting cancer cells by inhibiting mitosis, it preserves the microtubule network in non-dividing cells and in primary neurons at effective doses and is much safer in mouse models. By analyzing the structure of tubulin with different compounds, the authors determined that DZ-2384 binds at the vinca site but induces a distinctive change in the curvature of growing tubulin protofilaments, which may explain its unusual effects on microtubule dynamics and decreased toxicity. Microtubule-targeting agents (MTAs) are widely used anticancer agents, but toxicities such as neuropathy limit their clinical use. MTAs bind to and alter the stability of microtubules, causing cell death in mitosis. We describe DZ-2384, a preclinical compound that exhibits potent antitumor activity in models of multiple cancer types. It has an unusually high safety margin and lacks neurotoxicity in rats at effective plasma concentrations. DZ-2384 binds the vinca domain of tubulin in a distinct way, imparting structurally and functionally different effects on microtubule dynamics compared to other vinca-binding compounds. X-ray crystallography and electron microscopy studies demonstrate that DZ-2384 causes straightening of curved protofilaments, an effect proposed to favor polymerization of tubulin. Both DZ-2384 and the vinca alkaloid vinorelbine inhibit microtubule growth rate; however, DZ-2384 increases the rescue frequency and preserves the microtubule network in nonmitotic cells and in primary neurons. This differential modulation of tubulin results in a potent MTA therapeutic with enhanced safety.


BMC Cancer | 2015

Obatoclax is a direct and potent antagonist of membrane-restricted Mcl-1 and is synthetic lethal with treatment that induces Bim

Mai Nguyen; Regina Cencic; Franziska Ertel; Cynthia Bernier; Jerry Pelletier; Anne Roulston; John R. Silvius; Gordon C. Shore

BackgroundObatoclax is a clinical stage drug candidate that has been proposed to target and inhibit prosurvival members of the Bcl-2 family, and thereby contribute to cancer cell lethality. The insolubility of this compound, however, has precluded the use of many classical drug-target interaction assays for its study. Thus, a direct demonstration of the proposed mechanism of action, and preferences for individual Bcl-2 family members, remain to be established.MethodsEmploying modified proteins and lipids, we recapitulated the constitutive association and topology of mitochondrial outer membrane Mcl-1 and Bak in synthetic large unilamellar liposomes, and measured bakdependent bilayer permeability. Additionally, cellular and tumor models, dependent on Mcl-1 for survival, were employed.ResultsWe show that regulation of bilayer permeabilization by the tBid – Mcl-1 - Bak axis closely resemblesthe tBid - Bcl-XL - Bax model. Obatoclax rapidly and completely partitioned into liposomal lipid but also rapidly exchanged between liposome particles. In this system, obatoclax was found to be a direct and potent antagonist of liposome-bound Mcl-1 but not of liposome-bound Bcl-XL, and did not directly influence Bak. A 2.5 molar excess of obatoclax relative to Mcl-1 overcame Mcl-1-mediated inhibition of tBid-Bak activation. Similar results were found for induction of Bak oligomers by Bim. Obatoclax exhibited potent lethality in a cellmodel dependent on Mcl-1 for viability but not in cells dependent on Bcl-XL. Molecular modeling predicts that the 3-methoxy moiety of obatoclax penetrates into the P2 pocket of the BH3 binding site of Mcl-1. A desmethoxy derivative of obatoclax failed to inhibit Mcl-1 in proteoliposomes and did not kill cells whose survival depends on Mcl-1. Systemic treatment of mice bearing Tsc2+/- Em-myc lymphomas (whose cells depend on Mcl-1 for survival) with obatoclax conferred a survival advantage compared to vehicle alone (median 31 days vs 22 days, respectively; p=0.003). In an Akt-lymphoma mouse model, the anti-tumor effects of obatoclax synergized with doxorubicin. Finally, treatment of the multiple myeloma KMS11 cell model (dependent on Mcl-1 for survival) with dexamethasone induced Bim and Bim-dependent lethality. As predicted for an Mcl-1 antagonist, obatoclax and dexamethasone were synergistic in this model.ConclusionsTaken together, these findings indicate that obatoclax is a potent antagonist of membranerestricted Mcl-1. Obatoclax represents an attractive chemical series to generate second generation Mcl-1 inhibitors.

Collaboration


Dive into the Anne Roulston'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark A. Watson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge