Network


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

Hotspot


Dive into the research topics where Marie-Claude Bourgeois-Daigneault is active.

Publication


Featured researches published by Marie-Claude Bourgeois-Daigneault.


Science Translational Medicine | 2018

Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy

Marie-Claude Bourgeois-Daigneault; Dominic Roy; Amelia Sadie Aitken; Nader El Sayes; Nikolas Tim Martin; Oliver Varette; Theresa Falls; Lauren Elizabeth St-Germain; Adrian Pelin; Brian D. Lichty; David F. Stojdl; Guy Ungerechts; Jean-Simon Diallo; John C. Bell

Maraba virus treatment before surgery for triple-negative breast cancer promotes antitumor immunity. Viruses team up with cancer immunotherapy Immune checkpoint inhibitors have shown great promise for cancer therapy, but they do not treat all cancers, and neither breast nor brain tumors are usually treatable with these drugs. However, Bourgeois-Daigneault et al. discovered a way to address this for breast cancer, and Samson et al. discovered a way to address this for brain tumors. In both cases, the authors found that oncolytic virus treatment given early, before surgical resection, alters the antitumor immune response and potentiates the effects of subsequent treatment with immune checkpoint inhibitors. Although these studies differ in the details of their methods and the immune effects induced by the oncolytic viruses, they indicate the potential of such viruses for enhancing the potential of checkpoint therapy and expanding it to new types of cancer. Triple-negative breast cancer (TNBC) is an aggressive disease for which treatment options are limited and associated with severe toxicities. Immunotherapeutic approaches like immune checkpoint inhibitors (ICIs) are a potential strategy, but clinical trials have demonstrated limited success in this patient cohort. Clinical studies using ICIs have revealed that patients with preexisting anticancer immunity are the most responsive. Given that oncolytic viruses (OVs) induce antitumor immunity, we investigated their use as an ICI-sensitizing approach. Using a therapeutic model that mimics the course of treatment for women with newly diagnosed TNBC, we demonstrate that early OV treatment coupled with surgical resection provides long-term benefits. OV therapy sensitizes otherwise refractory TNBC to immune checkpoint blockade, preventing relapse in most of the treated animals. We suggest that OV therapy in combination with immune checkpoint blockade warrants testing as a neoadjuvant treatment option in the window of opportunity between TNBC diagnosis and surgical resection.


Cancer Cell | 2015

VEGF-Mediated Induction of PRD1-BF1/Blimp1 Expression Sensitizes Tumor Vasculature to Oncolytic Virus Infection.

Rozanne Arulanandam; Cory Batenchuk; Fernando A. Angarita; Kathryn Ottolino-Perry; Sophie Cousineau; Amelia Mottashed; Emma Burgess; Theresa Falls; Naomi De Silva; Jovian Tsang; Grant A. Howe; Marie-Claude Bourgeois-Daigneault; David P. Conrad; Manijeh Daneshmand; Caroline J. Breitbach; David Kirn; Leda Raptis; Subash Sad; Harold Atkins; Michael S. Huh; Jean-Simon Diallo; Brian D. Lichty; Carolina S. Ilkow; Fabrice Le Boeuf; Christina L. Addison; J. Andrea McCart; John C. Bell

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Molecular Therapy - Oncolytics | 2016

Oncolytic vesicular stomatitis virus expressing interferon-γ has enhanced therapeutic activity

Marie-Claude Bourgeois-Daigneault; Dominic Roy; Theresa Falls; Kwame Twumasi-Boateng; Lauren Elizabeth St-Germain; Monique Marguerie; Vanessa Garcia; Mohammed Selman; Victoria A Jennings; Jessica L. Pettigrew; Sally Amos; Jean-Simon Diallo; Brad H. Nelson; John C. Bell

Oncolytic viruses are known to stimulate the antitumor immune response by specifically replicating in tumor cells. This is believed to be an important aspect of the durable responses observed in some patients and the field is rapidly moving toward immunotherapy. As a further means to engage the immune system, we engineered a virus, vesicular stomatitis virus (VSV), to encode the proinflammatory cytokine interferon-γ. We used the 4T1 mammary adenocarcinoma as well as other murine tumor models to characterize immune responses in tumor-bearing animals generated by treatment with our viruses. The interferon-γ-encoding virus demonstrated greater activation of dendritic cells and drove a more profound secretion of proinflammatory cytokines compared to the parental virus. From a therapeutic point of view, the interferon-γ virus slowed tumor growth, minimized lung tumors, and prolonged survival in several murine tumor models. The improved efficacy was lost in immunocompromized animals; hence the mechanism appears to be T-cell-mediated. Taken together, these results demonstrate the ability of oncolytic viruses to act as immune stimulators to drive antitumor immunity as well as their potential for targeted gene therapy.


Journal of Controlled Release | 2015

Programmable insect cell carriers for systemic delivery of integrated cancer biotherapy

Dominic Roy; A.T. Power; Marie-Claude Bourgeois-Daigneault; Theresa Falls; L. Ferreira; A. Stern; C. Tanese de Souza; J.A. McCart; David F. Stojdl; Brian D. Lichty; Harold Atkins; Rebecca C. Auer; John C. Bell; F. Le Boeuf

Due to cancers genetic complexity, significant advances in the treatment of metastatic disease will require sophisticated, multi-pronged therapeutic approaches. Here we demonstrate the utility of a Drosophila melanogaster cell platform for the production and in vivo delivery of multi-gene biotherapeutic systems. We show that cultured Drosophila S2 cell carriers can stably propagate oncolytic viral therapeutics that are highly cytotoxic for mammalian cancer cells without adverse effects on insect cell viability or gene expression. Drosophila cell carriers administered systemically to immunocompetent animals trafficked to tumors to deliver multiple biotherapeutics with little apparent off-target tissue homing or toxicity, resulting in a therapeutic effect. Cells of this Dipteran invertebrate provide a genetically tractable platform supporting the integration of complex, multi-gene biotherapies while avoiding many of the barriers to systemic administration of mammalian cell carriers. These transporters have immense therapeutic potential as they can be modified to express large banks of biotherapeutics with complementary activities that enhance anti-tumor activity.


Ilar Journal | 2016

Murine Tumor Models for Oncolytic Rhabdo-Virotherapy

Theresa Falls; Dominic Roy; John C. Bell; Marie-Claude Bourgeois-Daigneault

The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.


Biomedicines | 2017

Taking a Stab at Cancer; Oncolytic Virus-Mediated Anti-Cancer Vaccination Strategies

Amelia Sadie Aitken; Dominic Roy; Marie-Claude Bourgeois-Daigneault

Vaccines have classically been used for disease prevention. Modern clinical vaccines are continuously being developed for both traditional use as well as for new applications. Typically thought of in terms of infectious disease control, vaccination approaches can alternatively be adapted as a cancer therapy. Vaccines targeting cancer antigens can be used to induce anti-tumour immunity and have demonstrated therapeutic efficacy both pre-clinically and clinically. Various approaches now exist and further establish the tremendous potential and adaptability of anti-cancer vaccination. Classical strategies include ex vivo-loaded immune cells, RNA- or DNA-based vaccines and tumour cell lysates. Recent oncolytic virus development has resulted in a surge of novel viruses engineered to induce powerful tumour-specific immune responses. In addition to their use as cancer vaccines, oncolytic viruses have the added benefit of being directly cytolytic to cancer cells and thus promote antigen recognition within a highly immune-stimulating tumour microenvironment. While oncolytic viruses are perfectly equipped for efficient immunization, this complicates their use upon previous exposure. Indeed, the host’s anti-viral counter-attacks often impair multiple-dosing regimens. In this review we will focus on the use of oncolytic viruses for anti-tumour vaccination. We will explore different strategies as well as ways to circumvent some of their limitations.


Cancer Research | 2017

Amplification of oncolytic vaccinia virus widespread tumor cell killing by sunitinib through multiple mechanisms

Minah Kim; Maximilian Nitschké; Barbara Sennino; Patrizia Murer; Brian J. Schriver; Alexander Bell; Aishwarya Subramanian; Corry E. McDonald; Jiahu Wang; Howard Cha; Marie-Claude Bourgeois-Daigneault; David Kirn; John C. Bell; Naomi De Silva; Caroline J. Breitbach; Donald M. McDonald

Oncolytic viruses pose many questions in their use in cancer therapy. In this study, we assessed the potential of mpJX-594 (mouse-prototype JX-594), a replication-competent vaccinia virus administered by intravenous injection, to target the tumor vasculature, produce immune activation and tumor cell killing more widespread than the infection, and suppress invasion and metastasis. These actions were examined in RIP-Tag2 transgenic mice with pancreatic neuroendocrine tumors that developed spontaneously and progressed as in humans. mpJX-594 initially infected tumor vascular endothelial cells, leading to vascular pruning and prolonged leakage in tumors but not in normal organs; parallel effects were observed in U87 gliomas. Viral infection spread to tumor cells, where tumor cell killing was much more widespread than the infection. Widespread tumor cell killing at 5 days was prevented by depletion of CD8+ T lymphocytes and did not require GM-CSF, as mpJX-594 variants that expressed human, mouse, or no GM-CSF produced equivalent amounts of killing. The antivascular, antitumor, and antimetastatic effects of mpJX-594 were amplified by concurrent or sequential administration of sunitinib, a multitargeted receptor tyrosine kinase inhibitor. These effects were not mimicked by selective inhibition of VEGFR2 despite equivalent vascular pruning, but were accompanied by suppression of regulatory T cells and greater influx of activated CD8+ T cells. Together, our results showed that mpJX-594 targets tumor blood vessels, spreads secondarily to tumor cells, and produces widespread CD8+ T-cell-dependent tumor cell killing in primary tumors and metastases, and that these effects can be amplified by coadministration of sunitinib.Significance: These findings reveal multiple unrecognized features of the antitumor properties of oncolytic vaccinia viruses, all of which can be amplified by the multitargeted kinase inhibitor sunitinib. Cancer Res; 78(4); 922-37. ©2017 AACR.


Journal for ImmunoTherapy of Cancer | 2018

Enhanced susceptibility of cancer cells to oncolytic rhabdo-virotherapy by expression of Nodamura virus protein B2 as a suppressor of RNA interference

Donald Bastin; Amelia Sadie Aitken; Adrian Pelin; Larissa A. Pikor; Mathieu Jf Crupi; Michael S. Huh; Marie-Claude Bourgeois-Daigneault; John C. Bell; Carolina S. Ilkow

Antiviral responses are barriers that must be overcome for efficacy of oncolytic virotherapy. In mammalian cells, antiviral responses involve the interferon pathway, a protein-signaling cascade that alerts the immune system and limits virus propagation. Tumour-specific defects in interferon signaling enhance viral infection and responses to oncolytic virotherapy, but many human cancers are still refractory to oncolytic viruses. Given that invertebrates, fungi and plants rely on RNA interference pathways for antiviral protection, we investigated the potential involvement of this alternative antiviral mechanism in cancer cells. Here, we detected viral genome-derived small RNAs, indicative of RNAi-mediated antiviral responses, in human cancer cells. As viruses may encode suppressors of the RNA interference pathways, we engineered an oncolytic vesicular stomatitis virus variant to encode the Nodamura virus protein B2, a known inhibitor of RNAi-mediated immune responses. B2-expressing oncolytic virus showed enhanced viral replication and cytotoxicity, impaired viral genome cleavage and altered microRNA processing in cancer cells. Our data establish the improved therapeutic potential of our novel virus which targets the RNAi-mediated antiviral defense of cancer cells.


Journal of Clinical Investigation | 2018

Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade

Joy Hsu; Jonathan J. Hodgins; Malvika Marathe; Chris J. Nicolai; Marie-Claude Bourgeois-Daigneault; Troy N. Trevino; Camillia S. Azimi; Amit K. Scheer; Haley E. Randolph; Thornton W. Thompson; Lily Zhang; Alexandre Iannello; Nikhita Mathur; Karen Jardine; Georgia A. Kirn; John C. Bell; Michael W. McBurney; David H. Raulet; Michele Ardolino

Checkpoint blockade immunotherapy targeting the PD-1/PD-L1 inhibitory axis has produced remarkable results in the treatment of several types of cancer. Whereas cytotoxic T cells are known to provide important antitumor effects during checkpoint blockade, certain cancers with low MHC expression are responsive to therapy, suggesting that other immune cell types may also play a role. Here, we employed several mouse models of cancer to investigate the effect of PD-1/PD-L1 blockade on NK cells, a population of cytotoxic innate lymphocytes that also mediate antitumor immunity. We discovered that PD-1 and PD-L1 blockade elicited a strong NK cell response that was indispensable for the full therapeutic effect of immunotherapy. PD-1 was expressed on NK cells within transplantable, spontaneous, and genetically induced mouse tumor models, and PD-L1 expression in cancer cells resulted in reduced NK cell responses and generation of more aggressive tumors in vivo. PD-1 expression was more abundant on NK cells with an activated and more responsive phenotype and did not mark NK cells with an exhausted phenotype. These results demonstrate the importance of the PD-1/PD-L1 axis in inhibiting NK cell responses in vivo and reveal that NK cells, in addition to T cells, mediate the effect of PD-1/PD-L1 blockade immunotherapy.


Cancer immunology research | 2016

Abstract A068: Determinants of efficacy in cancer immunovirotherapy

Rūta Veinalde; Marie-Claude Bourgeois-Daigneault; Christof von Kalle; Dirk Jaeger; Guy Ungerechts; Christine E. Engeland

Replicating oncolytic viruses (OVs) are emerging as a promising therapeutic approach for different tumor entities. Positive data from numerous clinical studies are accumulating and as a first-in-class oncolytic herpes virus encoding GM-CSF, Talimogene Laherparepvec (T-VEC), has been approved for treatment of unresectable melanoma by the U.S. Food and Drug Administration at the end of 2015. Although it has been recognized that combinations of OVs with different immunomodulators can significantly improve therapeutic outcome, understanding of immune effector mechanisms determining success of a particular approach remains limited, restricting the development of rational combination strategies. In this study we aimed to determine the most beneficial immunomodulation strategies for combination with oncolytic measles virus (MeV). Furthermore, we performed a comprehensive analysis of anti-tumor immune effector mechanisms associated with response to pinpoint mechanisms crucial for therapeutic efficacy. We developed MeV Schwarz vaccine strain vectors (MeVac) encoding different immunomodulators to target the main phases in establishment of an anti-tumor immune response: GM-CSF - to enhance maturation of antigen-presenting cells, an IL-12 fusion protein (FmIL-12) and IP-10 - to enhance immune effector cell responses, antibodies against CTLA-4 and PD-L1 and a soluble form of CD80 - to counteract immunosuppression in the tumor microenvironment. Therapeutic efficacy of the novel vectors was evaluated in the fully immunocompetent murine colon adenocarcinoma model MC38cea. MeVac encoding anti-PD-L1 and FmIL-12, respectively, were identified as the most effective therapeutics. Notably, MeVac encoding FmIL-12 showed a superior therapeutic efficacy, achieving 90% complete remissions of established tumors. Animals treated with the immunomodulatory MeVac vectors rejected secondary tumor engraftments, indicating establishment of protective anti-tumor immunity. Higher IFN-γ production upon restimulation of splenocytes with tumor cells in vitro was observed for animals treated with MeVac FmIL-12 in comparison to treatment with MeVac anti-PD-L1 and a control vector encoding constant region of antibody (MeVac IgG1-Fc), indicating establishment of a more potent memory response. Intratumoral cytokine profiling with cytokine bead arrays revealed upregulation of the effector cytokines IFN-γ and TNF-α after treatment with MeVac FmIL-12. A massive decrease in intratumoral natural killer (NK) cell counts and an increase of activation marker CD69 expression on NK cells as well as an increase in T cell amount was observed in flow cytometry analysis one day after treatment with MeVac FmIL-12. This indicates early direct activation of these lymphocyte subsets through direct IL-12 signaling. An increase in intratumoral T cells and a decrease in NK cells and activated cytotoxic T cells (CD8+CD69+) was observed four days after the last treatment with both MeVac anti-PD-L1 and MeVac IgG1-Fc, with slightly more pronounced effects in the MeVac anti-PD-L1 group. This suggests that MeVac vectors per se activate T and NK cell responses, which can further be supported with PD-L1 blockade. This study establishes MeVac encoding FmIL-12 as a potent immunomodulatory oncolytic vector and provides insights into its mechanisms of action, thereby creating a basis for further rational vector modifications to translate immunomodulatory MeV into clinical application. Citation Format: Rūta Veinalde, Christian Grossardt, Marie-Claude Bourgeois-Daigneault, Christof von Kalle, Dirk Jaeger, Guy Ungerechts, Christine E. Engeland. Determinants of efficacy in cancer immunovirotherapy [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A068.

Collaboration


Dive into the Marie-Claude Bourgeois-Daigneault's collaboration.

Top Co-Authors

Avatar

John C. Bell

Ottawa Hospital Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Theresa Falls

Ottawa Hospital Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jean-Simon Diallo

Ottawa Hospital Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carolina S. Ilkow

Ottawa Hospital Research Institute

View shared research outputs
Top Co-Authors

Avatar

David F. Stojdl

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar

Lauren Elizabeth St-Germain

Ottawa Hospital Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge