Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Biochem Biophys Res Commun ; 526(3): 641-646, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32248971

RESUMO

Oncolytic viruses (OVs) are a class of biotherapeutics that are currently being explored for the treatment of cancer. While showing promise in several pre-clinical and clinical studies, systemic delivery of these anti-cancer agents is hampered by inefficient tumor targeting and a host immune system that is highly evolved to detect and neutralize pathogens. To shield the virus from immune recognition and destruction, the use of cells as delivery vehicles has been explored for the systemic delivery of OVs. Though several types of cell carriers are able to protect OVs during intravenous delivery, many still lack the ability to specifically home to or accumulate within the tumor microenvironment. Overall, OV-based therapeutics could benefit from tumor targeting strategies to maximize tumor-specific delivery and minimize infection of off-target tissues. In the current study, we examine magnetic targeting as a strategy to improve OV infection of tumor cells in vitro. We found that magnetic targeting of magnetically-labeled VSV particles or VSV-infected cell carriers resulted in increased infection and killing of tumor cells. Furthermore, this enhanced infection of target tumor cells was observed even in the presence of virus-specific neutralizing antibodies. Overall, our findings suggest that magnetic targeting strategies can improve the infection of tumor cells and may be a viable strategy to improve the tumor-targeted delivery of oncolytic VSV-based therapeutics.


Assuntos
Sistemas de Liberação de Medicamentos , Magnetismo , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Drosophila/citologia , Sistemas de Liberação de Medicamentos/métodos , Fenômenos Magnéticos , Magnetismo/métodos , Camundongos , Neoplasias/imunologia , Vírus Oncolíticos/imunologia
2.
Breast Cancer Res ; 18(1): 83, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27503504

RESUMO

BACKGROUND: Breast cancer is the most common malignant disease amongst Western women. The lack of treatment options for patients with chemotherapy-resistant or recurrent cancers is pushing the field toward the rapid development of novel therapies. The use of oncolytic viruses is a promising approach for the treatment of disseminated diseases like breast cancer, with the first candidate recently approved by the Food and Drug Administration for use in patients. In this report, we demonstrate the compatibility of oncolytic virotherapy and chemotherapy using various murine breast cancer models. This one-two punch has been explored in the past by several groups with different viruses and drugs and was shown to be a successful approach. Our strategy is to combine Paclitaxel, one of the most common drugs used to treat patients with breast cancer, and the oncolytic Rhabdovirus Maraba-MG1, a clinical trial candidate in a study currently recruiting patients with late-stage metastatic cancer. METHODS: We used the EMT6, 4 T1 and E0771 murine breast cancer models to evaluate in vitro and in vivo the effects of co-treatment with MG1 and Paclitaxel. Treatment-induced cytotoxicity was assessed and plaque assays, flow cytometry, microscopy and immunocytochemistry analysis were performed to quantify virus production and transgene expression. Orthotopically implanted tumors were measured during and after treatment to evaluate efficacy and Kaplan-Meier survival curves were generated. RESULTS: Our data demonstrate not only the compatibility of the treatments, but also their synergistic cytopathic activity. With Paclitaxel, EMT6 and 4 T1 tumors demonstrated increased virus production both in vitro and in vivo. Our results also show that Paclitaxel does not impair the safety profile of the virus treatment. Importantly, when combined, MG1 and the drug controlled tumor growth and prolonged survival. CONCLUSIONS: The combination of MG1 and Paclitaxel improved efficacy in all of the breast cancer models we tested and thus is a promising alternative approach for the treatment of patients with refractory breast cancer. Our strategy has potential for rapid translation to the clinic, given the current clinical status of both agents.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Paclitaxel/uso terapêutico , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Modelos Animais de Doenças , Feminino , Humanos , Interferon beta/farmacologia , Camundongos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Paclitaxel/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Sci Rep ; 9(1): 1865, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755678

RESUMO

The use of oncolytic viruses (OVs) for cancer treatment is emerging as a successful strategy that combines the direct, targeted killing of the cancer with the induction of a long-lasting anti-tumor immune response. Using multiple aggressive murine models of triple-negative breast cancer, we have recently demonstrated that the early administration of oncolytic Maraba virus (MRB) prior to surgical resection of the primary tumor is sufficient to minimize the metastatic burden, protect against tumor rechallenge, cure a fraction of the mice and sensitize refractory tumors to immune checkpoint blockade without the need for further treatment. Here, we apply our surgical model to other OVs: Vesicular stomatitis virus (VSV), Adenovirus (Ad), Reovirus (Reo) and Herpes simplex virus (HSV) and show that all of the tested OVs could positively change the outcome of the treated animals. The growth of the primary and secondary tumors was differently affected by the various OVs and most of the viruses conferred survival benefits in this neoadjuvant setting despite the absence of direct treatment following rechallenge. This study establishes that OV-therapy confers long-term protection when administered in the pre-operative window of opportunity.


Assuntos
Neoplasias Mamárias Experimentais/prevenção & controle , Terapia Neoadjuvante/métodos , Terapia Viral Oncolítica/métodos , Adenoviridae , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Vírus Oncolíticos , Período Pré-Operatório , Reoviridae , Simplexvirus , Células Vero , Vesiculovirus
4.
Sci Transl Med ; 10(422)2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298865

RESUMO

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.


Assuntos
Terapia Viral Oncolítica/métodos , Neoplasias de Mama Triplo Negativas/terapia , Feminino , Humanos , Terapia Neoadjuvante/métodos , Vírus Oncolíticos/fisiologia
5.
Biomedicines ; 5(1)2017 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-28536346

RESUMO

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.

6.
ILAR J ; 57(1): 73-85, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27034397

RESUMO

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.


Assuntos
Terapia Viral Oncolítica/métodos , Animais , Camundongos , Neoplasias/terapia , Neoplasias/virologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral/fisiologia
7.
Mol Ther Oncolytics ; 3: 16001, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27119116

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA