Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(5): 896-915.e19, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35180381

RESUMEN

The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Inmunidad Mucosa , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Citocinas/sangre , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Neutralización , Nucleocápside/genética , Nucleocápside/inmunología , Nucleocápside/metabolismo , Pan troglodytes , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
Nat Immunol ; 17(5): 514-522, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043414

RESUMEN

Cytosolic DNA-mediated activation of the transcription factor IRF3 is a key event in host antiviral responses. Here we found that infection with DNA viruses induced interaction of the metabolic checkpoint kinase mTOR downstream effector and kinase S6K1 and the signaling adaptor STING in a manner dependent on the DNA sensor cGAS. We further demonstrated that the kinase domain, but not the kinase function, of S6K1 was required for the S6K1-STING interaction and that the TBK1 critically promoted this process. The formation of a tripartite S6K1-STING-TBK1 complex was necessary for the activation of IRF3, and disruption of this signaling axis impaired the early-phase expression of IRF3 target genes and the induction of T cell responses and mucosal antiviral immunity. Thus, our results have uncovered a fundamental regulatory mechanism for the activation of IRF3 in the cytosolic DNA pathway.


Asunto(s)
ADN/inmunología , Factor 3 Regulador del Interferón/inmunología , Proteínas de la Membrana/inmunología , Proteínas Quinasas S6 Ribosómicas 90-kDa/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Citosol/inmunología , Citosol/metabolismo , Citosol/virología , ADN/genética , ADN/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células HEK293 , Herpes Simple/inmunología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Humanos , Inmunización/métodos , Immunoblotting , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Nucleotidiltransferasas/metabolismo , Ovalbúmina/genética , Ovalbúmina/inmunología , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
3.
J Immunol ; 196(11): 4587-95, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183620

RESUMEN

Effector T cells (TEFF) are a barrier to booster vaccination because they can rapidly kill Ag-bearing APCs before memory T cells are engaged. We report in this study that i.v. delivery of rhabdoviral vectors leads to direct infection of follicular B cells in the spleen, where the earliest evidence of secondary T cell responses was observed. This allows booster immunizations to rapidly expand CD8(+) central memory T cells (TCM) during the acute phase of the primary response that is dominated by TEFF Interestingly, although the ablation of B cells before boosting with rhabdoviral vectors diminishes the expansion of memory T cells, B cells do not present Ags directly. Instead, depletion of CD11c(+) dendritic cells abrogates secondary T cell expansion, suggesting that virus-infected follicular B cells may function as an Ag source for local DCs to subsequently capture and present the Ag. Because TCM are located within B cell follicles in the spleen whereas TEFF cannot traffic through follicular regions, Ag production and presentation by follicular APCs represent a unique mechanism to secure engagement of TCM during an ongoing effector response. Our data offer insights into novel strategies for rapid expansion of CD8(+) T cells using prime-boost vaccines by targeting privileged sites for Ag presentation.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas Foliculares/inmunología , Bazo/citología , Bazo/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas Virales/inmunología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Virol ; 88(1): 54-65, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24109243

RESUMEN

The human coronavirus OC43 is a major contributor to the common cold worldwide, though due to its low mortality rate, little research has focused on this human pathogen. The nucleocapsid is an essential structural protein with conserved functions across the coronavirus family. While a multitude of studies have examined nucleocapsid function, none have described the effects of OC43 nucleocapsid on the transcription factor NF-κB. We report that the nucleocapsid protein of OC43 causes potentiation of NF-κB activation. This prolonged activation is the direct result of the ability of the nucleocapsid to bind RNA, specifically microRNA 9 (miR-9), which is a negative regulator of NF-κB. This previously undescribed interaction between virus and host is a potential mechanism of immune evasion in RNA viruses.


Asunto(s)
Coronavirus Humano OC43/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Proteínas de la Nucleocápside/metabolismo , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Inmunoprecipitación , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Blood ; 121(13): 2432-9, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23325836

RESUMEN

Rapid boosting of memory CD8(+) T cells (TM) is essential in cancer immunotherapy and the control of certain infectious diseases. However, effector T cells (TE) are a barrier to booster vaccination because they can rapidly kill antigen-bearing antigen-presenting cells (APCs) before TM are engaged. We demonstrate that viral-vectored vaccines delivered by B cells elicit robust TM expansion in the presence of TE, enabling booster immunizations to bypass TE-mediated negative feedback regulation. Our data indicate that viral vector-loaded B cells home to the follicular regions in secondary lymphoid organs, which are anatomically separated from TE and in close proximity to TM. The B cells, however, do not serve as APCs in this area. Rather, classic CD11c(+) dendritic cells serve to stimulate the secondary CD8(+) T-cell response. Our data reveal that B cells represent a novel and readily accessible delivery system that can effectively engage secondary CD8(+) T-cell activation for prime-boost strategies.


Asunto(s)
Adenoviridae , Linfocitos B/trasplante , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/administración & dosificación , Memoria Inmunológica , Inmunoterapia Adoptiva/métodos , Aceleración , Adenoviridae/genética , Animales , Linfocitos B/metabolismo , Células Cultivadas , Técnicas de Transferencia de Gen , Inmunización Secundaria/métodos , Memoria Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vacunación/métodos
6.
Mol Ther ; 22(1): 123-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24343053

RESUMEN

Within the oncolytic virus field, the extent of virus replication that is essential for immune stimulation to control tumor growth remains unresolved. Using infected cell protein 0 (ICP0)-defective oncolytic Herpes simplex virus type 1 (HSV-1) and HSV-2 viruses (dICP0 and dNLS) that show differences in their in vitro replication and cytotoxicity, we investigated the inherent features of oncolytic HSV viruses that are required for potent antitumor activity. In vitro, the HSV-2 vectors showed rapid cytotoxicity despite lower viral burst sizes compared to HSV-1 vectors. In vivo, although both of the dICP0 vectors initially replicated to a similar level, HSV-1 dICP0 was rapidly cleared from the tumors. In spite of this rapid clearance, HSV-1 dICP0 treatment conferred significant survival benefit. HSV-1 dICP0-treated tumors showed significantly higher levels of danger-associated molecular patterns that correlated with higher numbers of antigen-presenting cells within the tumor and increased antigen-specific CD8+ T-cell levels in the peripheral blood. This study suggests that, at least in the context of oncolytic HSV, the initial stages of immunogenic virus replication leading to activation of antitumor immunity are more important than persistence of a replicating virus within the tumor. This knowledge provides important insight for the design of therapeutically successful oncolytic viruses.


Asunto(s)
Vectores Genéticos/genética , Neoplasias/genética , Neoplasias/inmunología , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Simplexvirus/genética , Simplexvirus/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Apoptosis/genética , Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Proteína HMGB1/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Mutación , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Viroterapia Oncolítica , Receptor ErbB-2/inmunología , Carga Tumoral/genética , Carga Tumoral/inmunología , Replicación Viral
7.
Mol Ther ; 22(2): 420-429, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24322333

RESUMEN

The rhabdovirus Maraba has recently been characterized as a potent oncolytic virus. In the present study, we engineered an attenuated Maraba strain, defined as MG1, to express a melanoma-associated tumor antigen. Its ability to mount an antitumor immunity was evaluated in tumor-free and melanoma tumor-bearing mice. Alone, the MG1 vaccine appeared insufficient to prime detectable adaptive immunity against the tumor antigen. However, when used as a boosting vector in a heterologous prime-boost regimen, MG1 vaccine rapidly generated strong antigen-specific T-cell immune responses. Once applied for treating syngeneic murine melanoma tumors, our oncolytic prime-boost vaccination protocol involving Maraba MG1 dramatically extended median survival and allowed complete remission in more than 20% of the animals treated. This work describes Maraba virus MG1 as a potent vaccine vector for cancer immunotherapy displaying both oncolytic activity and a remarkable ability to boost adaptive antitumor immunity.


Asunto(s)
Vectores Genéticos/genética , Virus Oncolíticos/genética , Rhabdoviridae/genética , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Efecto Citopatogénico Viral , Femenino , Expresión Génica , Vectores Genéticos/inmunología , Inmunización Secundaria/métodos , Oxidorreductasas Intramoleculares/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Melanoma Experimental , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Virus Oncolíticos/inmunología , Rhabdoviridae/inmunología , Resultado del Tratamiento , Vesiculovirus/genética , Vesiculovirus/inmunología , Tropismo Viral
8.
Mol Ther ; 22(7): 1320-1332, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24695102

RESUMEN

This study characterizes the ability of novel oncolytic rhabdoviruses (Maraba MG1) to boost natural killer (NK) cell activity. Our results demonstrate that MG1 activates NK cells via direct infection and maturation of conventional dendritic cells. Using NK depletion and conventional dendritic cells ablation studies in vivo, we established that both are required for MG1 efficacy. We further explored the efficacy of attenuated MG1 (nonreplicating MG1-UV(2min) and single-cycle replicating MG1-Gless) and demonstrated that these viruses activate conventional dendritic cells, although to a lesser extent than live MG1. This translates to equivalent abilities to remove tumor metastases only at the highest viral doses of attenuated MG1. In tandem, we characterized the antitumor ability of NK cells following preoperative administration of live and attenuated MG1. Our results demonstrates that a similar level of NK activation and reduction in postoperative tumor metastases was achieved with equivalent high viral doses concluding that viral replication is important, but not necessary for NK activation. Biochemical characterization of a panel of UV-inactivated MG1 (2-120 minutes) revealed that intact viral particle and target cell recognition are essential for NK cell-mediated antitumor responses. These findings provide mechanistic insight and preclinical rationale for safe perioperative virotherapy to effectively reduce metastatic disease following cancer surgery.


Asunto(s)
Células Dendríticas/citología , Células Asesinas Naturales/citología , Melanoma/terapia , Rhabdoviridae/fisiología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos
9.
J Immunol ; 188(9): 4149-57, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22447977

RESUMEN

IL-15 plays many important roles within the immune system. IL-15 signals in lymphocytes via trans presentation, where accessory cells such as macrophages and dendritic cells present IL-15 bound to IL-15Rα in trans to NK cells and CD8(+) memory T cells expressing IL-15/IL-2Rß and common γ chain (γ(c)). Previously, we showed that the prophylactic delivery of IL-15 to Rag2(-/-)γ(c)(-/-) mice (mature T, B, and NK cell negative) afforded protection against a lethal HSV-2 challenge and metastasis of B16/F10 melanoma cells. In this study, we demonstrated that in vivo delivery of an adenoviral construct optimized for the secretion of human IL-15 to Rag2(-/-)γ(c)(-/-) mice resulted in significant increases in spleen size and cell number, leading us to hypothesize that IL-15 signals differently in myeloid immune cells compared with lymphocytes, for which IL-15/IL-2Rß and γ(c) expression are essential. Furthermore, treatment with IL-15 induced RANTES production by Rag2(-/-)γ(c)(-/-) bone marrow cells, but the presence of γ(c) did not increase bone marrow cell sensitivity to IL-15. This IL-15-mediated RANTES production by Rag2(-/-)γ(c)(-/-) bone marrow cells occurred independently of the IL-15/IL-2Rß and Jak/STAT pathways and instead required IL-15Rα signaling as well as activation of JNK and NF-κB. Importantly, we also showed that the trans presentation of IL-15 by IL-15Rα boosts IL-15-mediated IFN-γ production by NK cells but reduces IL-15-mediated RANTES production by Rag2(-/-)γ(c)(-/-) myeloid bone marrow cells. Our data clearly show that IL-15 signaling in NK cells is different from that of myeloid immune cells. Additional insights into IL-15 biology may lead to novel therapies aimed at bolstering targeted immune responses against cancer and infectious disease.


Asunto(s)
Quimiocina CCL5/inmunología , Subunidad alfa del Receptor de Interleucina-15/inmunología , Interleucina-15/inmunología , MAP Quinasa Quinasa 4/inmunología , Macrófagos/inmunología , FN-kappa B/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Memoria Inmunológica/fisiología , Infecciones/genética , Infecciones/inmunología , Infecciones/metabolismo , Infecciones/patología , Interleucina-15/genética , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/inmunología , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Factores de Transcripción STAT/metabolismo
10.
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
11.
Sci Rep ; 14(1): 18522, 2024 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-39122821

RESUMEN

One major limitation of effective vaccine delivery is its dependency on a robust cold chain infrastructure. While Vesicular stomatitis virus (VSV) has been demonstrated to be an effective viral vaccine vector for diseases including Ebola, its -70 °C storage requirement is a significant limitation for accessing disadvantaged locations and populations. Previous work has shown thermal stabilization of viral vaccines with a combination of pullulan and trehalose (PT) dried films. To improve the thermal stability of VSV, we optimized PT formulation concentrations and components, as well as drying methodology with enhanced vacuum drying. When formulated in PT films, VSV can be stored for 32 weeks at 4 °C with less than 2 log PFU loss, at 25 °C with 2.5 log PFU loss, and at 37 °C with 3.1 log PFU loss. These results demonstrate a significant advancement in VSV thermal stabilization, decreasing the cold chain requirements for VSV vectored vaccines.


Asunto(s)
Glucanos , Trehalosa , Trehalosa/química , Glucanos/química , Vacio , Vectores Genéticos , Desecación/métodos , Vacunas Virales/química , Vesiculovirus/genética , Animales , Temperatura
12.
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
13.
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
14.
J Virol ; 85(23): 12160-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917977

RESUMEN

Oncolytic virotherapy is a promising biological approach to cancer treatment that contributes to tumor eradication via immune- and non-immune-mediated mechanisms. One of the remaining challenges for these experimental therapies is the necessity to develop a durable adaptive immune response against the tumor. Vesicular stomatitis virus (VSV) is a prototypical oncolytic virus (OV) that exemplifies the multiple mechanisms of oncolysis, including direct cell lysis, cellular hypoxia resulting from the shutdown of tumor vasculature, and inflammatory cytokine release. Despite these properties, the generation of sustained antitumor immunity is observed only when VSV is engineered to express a tumor antigen directly. In the present study, we sought to increase the number of tumor-associated dendritic cells (DC) in vivo and tumor antigen presentation by combining VSV treatment with recombinant Fms-like tyrosine kinase 3 ligand (rFlt3L), a growth factor promoting the differentiation and proliferation of DC. The combination of VSV oncolysis and rFLt3L improved animal survival in two different tumor models, i.e., VSV-resistant B16 melanoma and VSV-sensitive E.G7 T lymphoma; however, increased survival was independent of the adaptive CD8 T cell response. Tumor-associated DC were actively infected by VSV in vivo, which reduced their viability and prevented their migration to the draining lymph nodes to prime a tumor-specific CD8 T cell response. These results demonstrate that VSV interferes with tumor DC functions and blocks tumor antigen presentation.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , Linfoma de Células T/prevención & control , Melanoma Experimental/prevención & control , Viroterapia Oncolítica , Virus de la Estomatitis Vesicular Indiana/fisiología , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Movimiento Celular , Proliferación Celular , Terapia Combinada , Células Dendríticas/metabolismo , Células Dendríticas/virología , Femenino , Citometría de Flujo , Terapia Genética , Linfoma de Células T/inmunología , Linfoma de Células T/virología , Melanoma Experimental/inmunología , Melanoma Experimental/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Tirosina Quinasa 3 Similar a fms/genética
15.
J Immunol ; 184(8): 4269-75, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20237288

RESUMEN

Although vaccines targeting tissue differentiation Ags represent a promising strategy for cancer immunotherapy, the risk of triggering autoimmune damage to normal tissues remains to be determined. Immunizing against a melanoma-associated Ag, dopachrome tautomerase (DCT), which normal melanocytes and glial cells also express, allowed concurrent analysis of autoimmune consequences in multiple tissues. We show that vaccination with recombinant adenovirus expressing DCT elicited a strong CTL response in C57BL/6 mice, leading to protection against intracranial challenge with B16-F10 melanoma cells. Both histological analysis and behavioral testing indicated that there was no evidence of neuropathology in vaccinated animals and long-term survivors. Although vitiligo or demyelination could be induced by additional stimuli (i.e., surgery or inflammation) in DCT-vaccinated mice, it did not extend beyond the inflammatory area, suggesting that there is self-regulatory negative feedback in normal tissues. These results demonstrate that it is possible to vaccinate against a tumor embedded in a vital organ that shares the target Ag.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Rechazo de Injerto/inmunología , Oxidorreductasas Intramoleculares/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/genética , Neoplasias Encefálicas/patología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Femenino , Marcación de Gen/métodos , Rechazo de Injerto/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/prevención & control , Inyecciones Intraventriculares , Oxidorreductasas Intramoleculares/administración & dosificación , Oxidorreductasas Intramoleculares/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Vitíligo/inmunología , Vitíligo/patología , Vitíligo/prevención & control
16.
Cancer Cell ; 4(4): 263-75, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14585354

RESUMEN

Ideally, an oncolytic virus will replicate preferentially in malignant cells, have the ability to treat disseminated metastases, and ultimately be cleared by the patient. Here we present evidence that the attenuated vesicular stomatitis strains, AV1 and AV2, embody all of these traits. We uncover the mechanism by which these mutants are selectively attenuated in interferon-responsive cells while remaining highly lytic in 80% of human tumor cell lines tested. AV1 and AV2 were tested in a xenograft model of human ovarian cancer and in an immune competent mouse model of metastatic colon cancer. While highly attenuated for growth in normal mice, both AV1 and AV2 effected complete and durable cures in the majority of treated animals when delivered systemically.


Asunto(s)
Inmunidad Innata/fisiología , Interferón beta/metabolismo , Virus de la Estomatitis Vesicular Indiana/metabolismo , Transporte Activo de Núcleo Celular , Animales , Neoplasias del Colon/terapia , Neoplasias del Colon/virología , Femenino , Humanos , Inmunidad Innata/inmunología , Interferón beta/inmunología , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/virología , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , Neoplasias Experimentales/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/terapia , Neoplasias Ováricas/virología , Transducción de Señal , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas de la Matriz Viral/metabolismo , Replicación Viral/genética , Replicación Viral/fisiología
17.
Mol Ther ; 19(2): 335-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21119618

RESUMEN

Oncolytic virotherapy, the selective killing of tumor cells by oncolytic viruses (OVs), has emerged as a promising avenue of anticancer research. We have previously shown that KM100, a Herpes simplex virus type-1 (HSV) deficient for infected cell protein 0 (ICP0), possesses substantial oncolytic properties in vitro and has antitumor efficacy in vivo, in part by inducing antitumor immunity. Here, we illustrate through T-cell immunodepletion studies in nontolerized tumor-associated antigen models of breast cancer that KM100 treatment promotes antiviral and antitumor CD8(+) cytotoxic T-cell responses necessary for complete tumor regression. In tolerized tumor-associated antigen models of breast cancer, antiviral CD8(+) cytotoxic T-cell responses against infected tumor cells correlated with the induction of significant tumoristasis in the absence of tumor-associated antigen-specific CD8(+) cytotoxic T-cells. To enhance oncolysis, we tested a more cytopathic ICP0-null HSV and a vesicular stomatitis virus M protein mutant and found that despite improved in vitro replication, oncolysis in vivo did not improve. These studies illustrate that the in vitro cytolytic properties of OVs are poor prognostic indicators of in vivo antitumor activity, and underscore the importance of adaptive antiviral CD8(+) cytotoxic T-cells in effective cancer virotherapy.


Asunto(s)
Inmunidad Adaptativa/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Viroterapia Oncolítica/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Simplexvirus/genética , Simplexvirus/fisiología
18.
Mol Ther ; 19(5): 886-94, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21364541

RESUMEN

Oncolytic viruses (OVs) have been engineered or selected for cancer cell-specific infection however, we have found that following intravenous administration of vesicular stomatitis virus (VSV), tumor cell killing rapidly extends far beyond the initial sites of infection. We show here for the first time that VSV directly infects and destroys tumor vasculature in vivo but leaves normal vasculature intact. Three-dimensional (3D) reconstruction of infected tumors revealed that the majority of the tumor mass lacks significant blood flow in contrast to uninfected tumors, which exhibit relatively uniform perfusion. VSV replication in tumor neovasculature and spread within the tumor mass, initiates an inflammatory reaction including a neutrophil-dependent initiation of microclots within tumor blood vessels. Within 6 hours of intravenous administration of VSV and continuing for at least 24 hours, we observed the initiation of blood clots within the tumor vasculature whereas normal vasculature remained clot free. Blocking blood clot formation with thrombin inhibitors prevented tumor vascular collapse. Our results demonstrate that the therapeutic activity of an OV can go far beyond simple infection and lysis of malignant cells.


Asunto(s)
Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus de la Estomatitis Vesicular Indiana , Adenocarcinoma/genética , Animales , Coagulación Sanguínea , Línea Celular Tumoral , Proliferación Celular , Ratones , Ratones Endogámicos BALB C , Neutrófilos , Trombina/antagonistas & inhibidores
19.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35246474

RESUMEN

BACKGROUND: Pancreatic cancer is one of the leading causes of cancer death, with a 5-year -year survival rate of less than 10%. This results from late detection, high rates of metastasis, and resistance to standard chemotherapies. Furthermore, chemotherapy and radiation are associated with significant morbidity, underscoring the need for novel therapies. Recent clinical studies have shown that immunotherapies can provide durable outcomes in cancer patients, but successes in pancreatic cancer have been limited. It is likely that novel and combined therapies will be needed to achieve clinical benefits. METHODS: Using experimental mouse models of pancreatic ductal adenocarcinoma, we examined natural killer T (NKT) cell activation therapy in combination with a recombinant oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express the cytokine IL-15 (VSV-IL-15). Panc02 pancreatic ductal carcinoma cells were implanted subcutaneously or orthotopically into syngeneic C57BL/6 mice. Mice were then treated with VSV expressing green fluorescent protein (VSV-GFP) or VSV-IL-15 and/or NKT cell activation therapy via delivery of α-GalCer-loaded DCs. We further assessed whether the addition of PD-1 blockade could increase the therapeutic benefit of our combination treatment. Three days after NKT cell activation, some groups of mice were treated with anti-PD-1 antibodies weekly for 3 weeks. RESULTS: VSV-GFP and VSV-IL-15 mediated equal killing of human and mouse pancreatic cancer lines in vitro. In vivo, VSV-IL-15 combined with NKT cell activation therapy to enhance tumor regression and increase survival time over individual treatments, and was also superior to NKT cell therapy combined with VSV-GFP. Enhanced tumor control was associated with increased immune cell infiltration and anti-tumor effector functions (cytotoxicity and cytokine production). While ineffective as a monotherapy, the addition of blocking PD-1 antibodies to the combined protocol sustained immune cell activation and effector functions, resulting in prolonged tumor regression and complete tumor clearance in 20% of mice. Mice who cleared the initial tumor challenge exhibited reduced tumor growth uponon rechallenge, consistent with the formation of immune memory. CONCLUSION: TThese results demonstrate that NKT cell immunotherapy combined with oncolytic VSV-IL-15 virotherapy and PD-1 blockade enhances tumor control and presents a promising treatment strategy for targeting pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Células T Asesinas Naturales , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Pancreáticas , Animales , Carcinoma Ductal Pancreático/terapia , Línea Celular Tumoral , Citocinas/metabolismo , Humanos , Inmunoterapia , Interleucina-15/genética , Interleucina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Virus de la Estomatitis Vesicular Indiana/metabolismo , Vesiculovirus , Neoplasias Pancreáticas
20.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34990408

RESUMEN

BackgroundAdenovirus-vectored (Ad-vectored) vaccines are typically administered via i.m. injection to humans and are incapable of inducing respiratory mucosal immunity. However, aerosol delivery of Ad-vectored vaccines remains poorly characterized, and its ability to induce mucosal immunity in humans is unknown. This phase Ib trial evaluated the safety and immunogenicity of human serotype-5 Ad-vectored tuberculosis (TB) vaccine (AdHu5Ag85A) delivered to humans via inhaled aerosol or i.m. injection.MethodsThirty-one healthy, previously BCG-vaccinated adults were enrolled. AdHu5Ag85A was administered by single-dose aerosol using Aeroneb Solo Nebulizer or by i.m. injection. The study consisted of the low-dose (LD) aerosol, high-dose (HD) aerosol, and i.m. groups. The adverse events were assessed at various times after vaccination. Immunogenicity data were collected from the peripheral blood and bronchoalveolar lavage samples at baseline, as well as at select time points after vaccination.ResultsThe nebulized aerosol droplets were < 5.39 µm in size. Both LD and HD of AdHu5Ag85A administered by aerosol inhalation and i.m. injection were safe and well tolerated. Both aerosol doses, particularly LD, but not i.m., vaccination markedly induced airway tissue-resident memory CD4+ and CD8+ T cells of polyfunctionality. While as expected, i.m. vaccination induced Ag85A-specific T cell responses in the blood, the LD aerosol vaccination also elicited such T cells in the blood. Furthermore, the LD aerosol vaccination induced persisting transcriptional changes in alveolar macrophages.ConclusionInhaled aerosol delivery of Ad-vectored vaccine is a safe and superior way to elicit respiratory mucosal immunity. This study warrants further development of aerosol vaccine strategies against respiratory pathogens, including TB and COVID-19.Trial registrationClinicalTrial.gov, NCT02337270.FundingThe Canadian Institutes for Health Research (CIHR) and the Natural Sciences and Engineering Research Council of Canada funded this work.


Asunto(s)
Aerosoles/farmacología , COVID-19/prevención & control , SARS-CoV-2/efectos de los fármacos , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Administración por Inhalación , Adolescente , Adulto , Aerosoles/administración & dosificación , Anticuerpos Neutralizantes/sangre , Vacuna BCG/inmunología , COVID-19/inmunología , Femenino , Humanos , Inmunidad Mucosa/efectos de los fármacos , Inmunidad Mucosa/inmunología , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Tuberculosis/inmunología , Vacunación/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA