Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Curr Cancer Drug Targets ; 18(2): 109-123, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28176648

RESUMEN

Oncolytic viruses are a promising anti-cancer platform, achieving significant pre-clinical and clinical milestones in recent years. A full arsenal of selective, safe, and effective viruses has been developed with some emerging pre-clinical research focusing on optimizing these therapies in the face of remaining challenges, both in the bloodstream and in the tumour microenvironment. Herein we discuss the recent progress in pre-clinical virotherapy research to address these challenges, with special focus on innovative strategies that seek to complement the current strengths of virotherapy, ensuring an optimal multi-faceted attack on cancer. This review highlights the research areas that we believe provide the most potential to increase the efficacy of this exciting biotherapy platform: cell carriers, tumour vascular destruction, microenvironment modulation, combination therapies, and virus-mediated anti-tumour immune responses.


Asunto(s)
Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Humanos
2.
Mol Ther ; 23(6): 1066-1076, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25807289

RESUMEN

Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Virus Vaccinia/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Línea Celular Tumoral , Chlorocebus aethiops , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Estudios de Factibilidad , Femenino , Células HeLa , Humanos , Inyecciones Intralesiones , Macaca fascicularis/inmunología , Masculino , Neoplasias/sangre , Neoplasias/terapia , Pruebas de Neutralización , Piridonas/inmunología , Piridonas/farmacología , Ratas , Ratas Endogámicas F344 , Vacuna contra Viruela/sangre , Vacuna contra Viruela/inmunología , Vacunación , Células Vero
3.
Clin Cancer Res ; 19(14): 3832-43, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23714728

RESUMEN

PURPOSE: Acute lymphoblastic leukemia (ALL) remains incurable in most adults. It has been difficult to provide effective immunotherapy to improve outcomes for the majority of patients. Rhabdoviruses induce strong antiviral immune responses. We hypothesized that mice administered ex vivo rhabdovirus-infected ALL cells [immunotherapy by leukemia-oncotropic virus (iLOV)] would develop robust antileukemic immune responses capable of controlling ALL. EXPERIMENTAL DESIGN: Viral protein production, replication, and cytopathy were measured in human and murine ALL cells exposed to attenuated rhabdovirus. Survival following injection of graded amounts of ALL cells was compared between cohorts of mice administered γ-irradiated rhabdovirus-infected ALL cells (iLOV) or multiple control vaccines to determine key immunotherapeutic components and characteristics. Host immune requirements were assessed in immunodeficient and bone marrow-transplanted mice or by adoptive splenocyte transfer from immunized donors. Antileukemic immune memory was ascertained by second leukemic challenge in long-term survivors. RESULTS: Human and murine ALL cells were infected and killed by rhabdovirus; this produced a potent antileukemia vaccine. iLOV protected mice from otherwise lethal ALL by developing durable leukemia-specific immune-mediated responses (P < 0.0001), which required an intact CTL compartment. Preexisting antiviral immunity augmented iLOV potency. Splenocytes from iLOV-vaccinated donors protected 60% of naïve recipients from ALL challenge (P = 0.0001). Injecting leukemia cells activated by, or concurrent with, multiple Toll-like receptor agonists could not reproduce the protective effect of iLOV. Similarly, injecting uninfected irradiated viable, apoptotic, or necrotic leukemia cells with/without concurrent rhabdovirus administration was ineffective. CONCLUSION: Rhabdovirus-infected leukemia cells can be used to produce a vaccine that induces robust specific immunity against aggressive leukemia.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Rhabdoviridae/fisiología , Traslado Adoptivo , Animales , Trasplante de Médula Ósea , Vacunas contra el Cáncer , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Desnudos , Trasplante de Neoplasias , Medicina de Precisión , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Vero
4.
Mol Ther ; 21(4): 887-94, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23295947

RESUMEN

Histone deacetylase inhibitors (HDACi) can modulate innate antiviral responses and render tumors more susceptible to oncolytic viruses (OVs); however, their effects on adaptive immunity in this context are largely unknown. Our present study reveals an unexpected property of the HDACi MS-275 that enhances viral vector-induced lymphopenia leading to selective depletion of bystander lymphocytes and regulatory T cells while allowing expansion of antigen-specific secondary responses. Coadministration of vaccine plus drug during the boosting phase focuses the immune response on the tumor by suppressing the primary immune response against the vaccine vector and enhancing the secondary response against the tumor antigen. Furthermore, improvement of T cell functionality was evident suggesting that MS-275 can orchestrate a complex array of effects that synergize immunotherapy and viral oncolysis. Surprisingly, while MS-275 dramatically enhanced efficacy, it suppressed autoimmune pathology, profoundly improving the therapeutic index.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Autoinmunidad/efectos de los fármacos , Línea Celular Tumoral , Femenino , Melanoma/tratamiento farmacológico , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico
5.
Mol Ther ; 20(9): 1791-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22760544

RESUMEN

Treatment of permissive tumors with the oncolytic virus (OV) VSV-Δ51 leads to a robust antitumor T-cell response, which contributes to efficacy; however, many tumors are not permissive to in vivo treatment with VSV-Δ51. In an attempt to channel the immune stimulatory properties of VSV-Δ51 and broaden the scope of tumors that can be treated by an OV, we have developed a potent oncolytic vaccine platform, consisting of tumor cells infected with VSV-Δ51. We demonstrate that prophylactic immunization with this infected cell vaccine (ICV) protected mice from subsequent tumor challenge, and expression of granulocyte-monocyte colony stimulating factor (GM-CSF) by the virus (VSVgm-ICV) increased efficacy. Immunization with VSVgm-ICV in the VSV-resistant B16-F10 model induced maturation of dendritic and natural killer (NK) cell populations. The challenge tumor is rapidly infiltrated by a large number of interferon γ (IFNγ)-producing T and NK cells. Finally, we demonstrate that this approach is robust enough to control the growth of established tumors. This strategy is broadly applicable because of VSV's extremely broad tropism, allowing nearly all cell types to be infected at high multiplicities of infection in vitro, where the virus replication kinetics outpace the cellular IFN response. It is also personalized to the unique tumor antigen(s) displayed by the cancer cell.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/terapia , Vesiculovirus/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inmunización , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Vero , Vesiculovirus/genética , Replicación Viral
6.
Mol Ther ; 20(6): 1148-57, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22273579

RESUMEN

Replicating viruses for the treatment of cancer have a number of advantages over traditional therapeutic modalities. They are highly targeted, self-amplifying, and have the added potential to act as both gene-therapy delivery vehicles and oncolytic agents. Parapoxvirus ovis or Orf virus (ORFV) is the prototypic species of the Parapoxvirus genus, causing a benign disease in its natural ungulate host. ORFV possesses a number of unique properties that make it an ideal viral backbone for the development of a cancer therapeutic: it is safe in humans, has the ability to cause repeat infections even in the presence of antibody, and it induces a potent T(h)-1-dominated immune response. Here, we show that live replicating ORFV induces an antitumor immune response in multiple syngeneic mouse models of cancer that is mediated largely by the potent activation of both cytokine-secreting, and tumoricidal natural killer (NK) cells. We have also highlighted the clinical potential of the virus by demonstration of human cancer cell oncolysis including efficacy in an A549 xenograft model of cancer.


Asunto(s)
Vectores Genéticos/administración & dosificación , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Virus del Orf/inmunología , Animales , Línea Celular Tumoral , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Vectores Genéticos/efectos adversos , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Neoplasias Pulmonares/secundario , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/genética , Virus Oncolíticos/genética , Virus del Orf/genética , Bazo/inmunología , Bazo/metabolismo , Carga Tumoral , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 107(4): 1576-81, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080710

RESUMEN

Oncolytic viruses constitute a promising therapy against malignant gliomas (MGs). However, virus-induced type I IFN greatly limits its clinical application. The kinase mammalian target of rapamycin (mTOR) stimulates type I IFN production via phosphorylation of its effector proteins, 4E-BPs and S6Ks. Here we show that mouse embryonic fibroblasts and mice lacking S6K1 and S6K2 are more susceptible to vesicular stomatitis virus (VSV) infection than their WT counterparts as a result of an impaired type I IFN response. We used this knowledge to employ a pharmacoviral approach to treat MGs. The highly specific inhibitor of mTOR rapamycin, in combination with an IFN-sensitive VSV-mutant strain (VSV(DeltaM51)), dramatically increased the survival of immunocompetent rats bearing MGs. More importantly, VSV(DeltaM51) selectively killed tumor, but not normal cells, in MG-bearing rats treated with rapamycin. These results demonstrate that reducing type I IFNs through inhibition of mTORC1 is an effective strategy to augment the therapeutic activity of VSV(DeltaM51).


Asunto(s)
Glioma/metabolismo , Glioma/terapia , Interferón Tipo I/biosíntesis , Factores de Transcripción/metabolismo , Estomatitis Vesicular/metabolismo , Vesiculovirus/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Glioma/genética , Glioma/virología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos , Trasplante de Neoplasias , Viroterapia Oncolítica , Proteínas , Ratas , Ratas Endogámicas F344 , Proteínas Quinasas S6 Ribosómicas/deficiencia , Proteínas Quinasas S6 Ribosómicas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Estomatitis Vesicular/genética , Estomatitis Vesicular/virología , Vesiculovirus/genética
8.
Mol Ther ; 15(9): 1686-93, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17579581

RESUMEN

Oncolytic viruses (OVs) are selected or designed to eliminate malignancies by direct infection and lysis of cancer cells. In contrast to this concept of direct tumor lysis by viral infection, we observed that a significant portion of the in vivo tumor killing activity of two OVs, vesicular stomatitis virus (VSV) and vaccinia virus is caused by indirect killing of uninfected tumor cells. Shortly after administering the oncolytic virus we observed limited virus infection, coincident with a loss of blood flow to the interior of the tumor that correlated with induction of apoptosis in tumor cells. Transcript profiling of tumors showed that virus infection resulted in a dramatic transcriptional activation of pro-inflammatory genes including the neutrophil chemoattractants CXCL1 and CXCL5. Immunohistochemical examination of infected tumors revealed infiltration by neutrophils correlating with chemokine induction. Depletion of neutrophils in animals prior to VSV administration eliminated uninfected tumor cell apoptosis and permitted more extensive replication and spreading of the virus throughout the tumor. Taken all together, these results indicate that targeted recruitment of neutrophils to infected tumor beds enhances the killing of malignant cells. We propose that activation of inflammatory cells can be used for enhancing the effectiveness of oncolytic virus therapeutics, and that this approach should influence the planning of therapeutic doses.


Asunto(s)
Inflamación/terapia , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Animales , Apoptosis/fisiología , Velocidad del Flujo Sanguíneo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Inflamación/genética , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Neoplasias/genética , Neoplasias/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...