Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Brain ; 143(12): 3629-3652, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33253355

RESUMEN

Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Tolerancia Inmunológica , Mediadores de Inflamación/metabolismo , Animales , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Progresión de la Enfermedad , Femenino , Genes MHC Clase II/genética , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Glioma/inmunología , Glioma/metabolismo , Glioma/patología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Parabiosis , Convulsiones/inducido químicamente , Bazo/inmunología , Bazo/patología , Theilovirus , Timo/patología
2.
Mol Ther ; 28(12): 2540-2552, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-32877695

RESUMEN

Enhancing the immunogenicity of tumor-associated antigens would represent a major advance for anti-tumor vaccination strategies. Here, we investigated structure-directed antigen destabilization as a strategy to improve the degradation, immunogenic epitope presentation, and T cell activation against a vesicular stomatitis virus (VSV)-encoded tumor antigen. We used the crystal structure of the model antigen ovalbumin to identify charge-disrupting amino acid mutations that were predicted to decrease the stability of the protein. One mutation, OVA-C12R, significantly reduced the half-life of the protein and was preferentially degraded in a 26-S proteasomal-dependent manner. The destabilized ovalbumin protein exhibited enhanced presentation of the major histocompatibility complex (MHC) class I immunogenic epitope, SIINFEKL, on the surface of B16F10 cells or murine bone marrow-derived dendritic cells (BMDCs) in vitro. Enhanced presentation correlated with better recognition by cognate CD8 OT-I T cells as measured by activation, proliferation, and effector cytokine production. Finally, VSV encoding the degradation-prone antigen was better able to prime an antigen ovalbumin-specific CD8 T cell response in vivo without altering the anti-viral CD8 T cell response. Our studies highlight that not only is the choice of antigen in cancer vaccines of importance, but that emphasis should be placed on modifying antigen quality to ensure optimal priming of anti-tumor responses.


Asunto(s)
Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Inmunidad , Activación de Linfocitos , Ovalbúmina/genética , Vesiculovirus/genética , Animales , Presentación de Antígeno , Antígenos de Neoplasias/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Epítopos/inmunología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Viroterapia Oncolítica/métodos , Ovalbúmina/química , Estabilidad Proteica
3.
Mol Ther ; 25(4): 962-975, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237836

RESUMEN

Systemic viroimmunotherapy activates endogenous innate and adaptive immune responses against both viral and tumor antigens. We have shown that therapy with vesicular stomatitis virus (VSV) engineered to express a tumor-associated antigen activates antigen-specific adoptively transferred T cells (adoptive cell therapy, ACT) in vivo to generate effective therapy. The overall goal of this study was to phenotypically characterize the immune response to VSV+ACT therapy and use the information gained to rationally improve combination therapy. We observed rapid expansion of blood CD8+ effector cells acutely following VSV therapy with markedly high expression of the immune checkpoint molecules PD-1 and TIM-3. Using these data, we tested a treatment schedule incorporating mAb immune checkpoint inhibitors with VSV+ACT treatment. Unlike clinical scenarios, we delivered therapy at early time points following tumor establishment and treatment. Our goal was to potentiate the immune response generated by VSV therapy to achieve durable control of metastatic disease. Despite the high frequency of endogenous PD-1+ TIM-3+ CD8+ T cells following virus administration, antibody blockade did not improve survival. These findings provide highly significant information about response kinetics to viroimmunotherapy and juxtapose the clinical use of checkpoint inhibitors against chronically dysfunctional T cells and the acute T cell response to oncolytic viruses.


Asunto(s)
Traslado Adoptivo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vectores Genéticos/genética , Inmunoterapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Virus de la Estomatitis Vesicular Indiana/genética , Animales , Modelos Animales de Enfermedad , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Memoria Inmunológica , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Mortalidad , Metástasis de la Neoplasia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Resultado del Tratamiento
4.
Nature ; 477(7362): 99-102, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21886163

RESUMEN

The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Poxviridae/fisiología , Adulto , Anciano , Anciano de 80 o más Años , ADN Viral/sangre , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Neoplasias/patología , Neoplasias/cirugía , Neoplasias/virología , Organismos Modificados Genéticamente/fisiología , Transgenes/genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
5.
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
6.
Methods Mol Biol ; 2748: 41-53, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38070106

RESUMEN

The development of chimeric antigen receptor (CAR) T cells has been a revolutionary technology for the treatment of relapsed and refractory leukemias and lymphomas. The synthetic CAR molecule redirects T cell function toward tumor surface-expressed antigens through a single-chain variable fragment (scFv) fused to CD3z and intracellular costimulatory domains. Here, we describe a protocol for the generation of CAR T cells using primary mouse T cells and a gammaretroviral vector encoding a CAR transgene. This protocol outlines several transduction and expansion methods based on the use of two transduction enhancers, RetroNectin® and Vectofusin®-1, and cell culture systems such as conventional plates or G-Rex® devices.


Asunto(s)
Linfoma , Linfocitos T , Animales , Ratones , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Retroviridae/genética
7.
Trends Pharmacol Sci ; 45(5): 406-418, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38614815

RESUMEN

T cells modified to express intelligently designed chimeric antigen receptors (CARs) are exceptionally powerful therapeutic agents for relapsed and refractory blood cancers and have the potential to revolutionize therapy for many other diseases. To circumvent the complexity and cost associated with broad-scale implementation of ex vivo manufactured adoptive cell therapy products, alternative strategies to generate CAR T cells in vivo by direct infusion of nanoparticle-formulated nucleic acids or engineered viral vectors under development have received a great deal of attention in the past few years. Here, we outline the ex vivo manufacturing process as a motivating framework for direct in vivo strategies and discuss emerging data from preclinical models to highlight the potency of the in vivo approach, the applicability for new disease indications, and the remaining challenges associated with clinical readiness, including delivery specificity, long term efficacy, and safety.


Asunto(s)
Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Linfocitos T , Humanos , Animales , Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos , Ingeniería Celular/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Neoplasias/terapia , Neoplasias/inmunología
8.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937436

RESUMEN

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Asunto(s)
Antígenos de Neoplasias , Antígeno B7-H1 , Linfocitos T CD8-positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Viroterapia Oncolítica , Virus Oncolíticos , Microambiente Tumoral , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Animales , Viroterapia Oncolítica/métodos , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/inmunología , Microambiente Tumoral/inmunología , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Humanos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Interferón beta/metabolismo , Interferón beta/inmunología , Ratones Endogámicos C57BL , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos T/inmunología , Femenino , Vesiculovirus/inmunología , Vesiculovirus/genética
9.
Mol Ther ; 20(4): 749-58, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22186794

RESUMEN

Oncolytic viruses are generally designed to be cancer selective on the basis of a single genetic mutation. JX-594 is a thymidine kinase (TK) gene-inactivated oncolytic vaccinia virus expressing granulocyte-macrophage colony-stimulating factor (GM-CSF) and lac-Z transgenes that is designed to destroy cancer cells through replication-dependent cell lysis and stimulation of antitumoral immunity. JX-594 has demonstrated a favorable safety profile and reproducible tumor necrosis in a variety of solid cancer types in clinical trials. However, the mechanism(s) responsible for its cancer-selectivity have not yet been well described. We analyzed the replication of JX-594 in three model systems: primary normal and cancer cells, surgical explants, and murine tumor models. JX-594 replication, transgene expression, and cytopathic effects were highly cancer-selective, and broad spectrum activity was demonstrated. JX-594 cancer-selectivity was multi-mechanistic; replication was activated by epidermal growth factor receptor (EGFR)/Ras pathway signaling, cellular TK levels, and cancer cell resistance to type-I interferons (IFNs). These findings confirm a large therapeutic index for JX-594 that is driven by common genetic abnormalities in human solid tumors. This appears to be the first description of multiple selectivity mechanisms, both inherent and engineered, for an oncolytic virus. These findings have implications for oncolytic viruses in general, and suggest that their cancer targeting is a complex and multifactorial process.


Asunto(s)
Neoplasias/metabolismo , Virus Oncolíticos/fisiología , Poxviridae/fisiología , Transducción de Señal/fisiología , Replicación Viral/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Leucocitos Mononucleares , Ratones , Ratones Desnudos , Neoplasias/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Poxviridae/genética , Transducción de Señal/genética , Replicación Viral/genética
10.
Res Sq ; 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38045348

RESUMEN

Although immune checkpoint inhibition (ICI) has produced profound survival benefits in a broad variety of tumors, a proportion of patients do not respond. Treatment failure is in part due to immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, we developed a vesicular stomatitis virus expressing interferon-ß (VSV-IFNß) as a viro-immunotherapy against HCC. Since HCC standard of care atezolizumab/bevacizumab incorporates ICI, we tested the hypothesis that pro-inflammatory VSV-IFNß would recruit, prime, and activate anti-tumor T cells, whose activity anti-PD-L1 ICI would potentiate. However, in a partially anti-PD-L1-responsive model of HCC, addition of VSV-IFNß abolished anti-PD-L1 therapy. Cytometry by Time of Flight showed that VSV-IFNß expanded dominant anti-viral effector CD8 T cells with concomitant, relative disappearance of anti-tumor T cell populations which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, the potent anti-viral response became amalgamated with an anti-tumor T cell response generating highly significant cures compared to anti-PD-L1 ICI alone. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, by chimerizing anti-viral and anti-tumor T cell responses through encoding tumor antigens within the virus, oncolytic virotherapy can be purposed for very effective immune driven tumor clearance and can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.

11.
Cancer Immunol Res ; 11(12): 1571-1577, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37906619

RESUMEN

The Arthur and Sandra Irving Cancer Immunology Symposium has been created as a platform for established cancer immunologists to mentor trainees and young investigators as they launch their research career in the field. By sharing their different paths to success, the senior faculty mentors provide an invaluable resource to support the development of the next generation of leaders in the cancer immunology community. This Commentary describes some of the key topics that were discussed during the 2022 symposium: scientific and career trajectory, leadership, mentoring, collaborations, and publishing. For each of these topics, established investigators discussed the elements that facilitate success in these areas as well as mistakes that can hinder progress. Herein, we outline the critical points raised in these discussions for establishing a successful independent research career. These points are highly relevant for the broader scientific community.


Asunto(s)
Tutoría , Neoplasias , Médicos , Humanos , Mentores , Investigadores , Neoplasias/terapia
12.
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
13.
Sci Transl Med ; 14(640): eabn2231, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35417192

RESUMEN

Oncolytic viruses (OVs) encoding a variety of transgenes have been evaluated as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-modified T cells in the solid tumor microenvironment (TME). Here, using systemically delivered OVs and CAR T cells in immunocompetent mouse models, we have defined a mechanism by which OVs can potentiate CAR T cell efficacy against solid tumor models of melanoma and glioma. We show that stimulation of the native T cell receptor (TCR) with viral or virally encoded epitopes gives rise to enhanced proliferation, CAR-directed antitumor function, and distinct memory phenotypes. In vivo expansion of dual-specific (DS) CAR T cells was leveraged by in vitro preloading with oncolytic vesicular stomatitis virus (VSV) or reovirus, allowing for a further in vivo expansion and reactivation of T cells by homologous boosting. This treatment led to prolonged survival of mice with subcutaneous melanoma and intracranial glioma tumors. Human CD19 CAR T cells could also be expanded in vitro with TCR reactivity against viral or virally encoded antigens and was associated with greater CAR-directed cytokine production. Our data highlight the utility of combining OV and CAR T cell therapy and show that stimulation of the native TCR can be exploited to enhance CAR T cell activity and efficacy in mice.


Asunto(s)
Glioma , Melanoma , Viroterapia Oncolítica , Virus Oncolíticos , Receptores Quiméricos de Antígenos , Animales , Glioma/terapia , Inmunoterapia Adoptiva , Melanoma/terapia , Ratones , Virus Oncolíticos/fisiología , Receptores de Antígenos de Linfocitos T , Linfocitos T , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Ther ; 18(5): 896-902, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20160706

RESUMEN

A number of oncolytic virus (OV) candidates currently in clinical trials are human viruses that have been engineered to be safer for patient administration by limiting normal cell targeting and replication. The newest OVs include viruses that cause no disease in humans, yet still have natural tumor tropism. Raccoonpox virus (RCNV) is a member of the Orthopoxvirus genus of Poxviridae and closely related to vaccinia virus, yet has no known pathogenicity in any mammalian species. A screen of cells from the NCI-60 cancer cell panel using growth curves demonstrated greater than a log increase in replication of RCNV in nearly 74% of the cell lines tested, similar to other tested OV poxviruses. In normal cell lines, pretreatment with interferon (IFN)-alpha/beta resulted in significant inhibition of RCNV replication. In both xenograft and syngeneic models of solid tumors, injection of RCNV resulted in significantly slower tumor progression and increased survival of mice. RCNV treatment also prolonged survival in treatment-resistant models of brain tumors and decreased tumor burden by systemic administration in models of lung metastasis.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Poxviridae/fisiología , Animales , Línea Celular Tumoral , Femenino , Humanos , Interferón-alfa/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Virus Oncolíticos/genética , Poxviridae/genética , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancers (Basel) ; 13(5)2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807553

RESUMEN

Oncolytic viruses (OVs) and adoptive T cell therapy (ACT) each possess direct tumour cytolytic capabilities, and their combination potentially seems like a match made in heaven to complement the strengths and weakness of each modality. While providing strong innate immune stimulation that can mobilize adaptive responses, the magnitude of anti-tumour T cell priming induced by OVs is often modest. Chimeric antigen receptor (CAR) modified T cells bypass conventional T cell education through introduction of a synthetic receptor; however, realization of their full therapeutic properties can be stunted by the heavily immune-suppressive nature of the tumour microenvironment (TME). Oncolytic viruses have thus been seen as a natural ally to overcome immunosuppressive mechanisms in the TME which limit CAR T cell infiltration and functionality. Engineering has further endowed viruses with the ability to express transgenes in situ to relieve T cell tumour-intrinsic resistance mechanisms and decorate the tumour with antigen to overcome antigen heterogeneity or loss. Despite this helpful remodeling of the tumour microenvironment, it has simultaneously become clear that not all virus induced effects are favourable for CAR T, begging the question whether viruses act as valets ushering CAR T into their active site, or vandals which cause chaos leading to both tumour and T cell death. Herein, we summarize recent studies combining these two therapeutic modalities and seek to place them within the broader context of viral T cell immunology which will help to overcome the current limitations of effective CAR T therapy to make the most of combinatorial strategies.

16.
Nat Commun ; 12(1): 1930, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772027

RESUMEN

In our clinical trials of oncolytic vesicular stomatitis virus expressing interferon beta (VSV-IFNß), several patients achieved initial responses followed by aggressive relapse. We show here that VSV-IFNß-escape tumors predictably express a point-mutated CSDE1P5S form of the RNA-binding Cold Shock Domain-containing E1 protein, which promotes escape as an inhibitor of VSV replication by disrupting viral transcription. Given time, VSV-IFNß evolves a compensatory mutation in the P/M Inter-Genic Region which rescues replication in CSDE1P5S cells. These data show that CSDE1 is a major cellular co-factor for VSV replication. However, CSDE1P5S also generates a neo-epitope recognized by non-tolerized T cells. We exploit this predictable neo-antigenesis to drive, and trap, tumors into an escape phenotype, which can be ambushed by vaccination against CSDE1P5S, preventing tumor escape. Combining frontline therapy with escape-targeting immunotherapy will be applicable across multiple therapies which drive tumor mutation/evolution and simultaneously generate novel, targetable immunopeptidomes associated with acquired treatment resistance.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Interferón beta/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Proteínas de Unión al ARN/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Replicación Viral/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Inmunoterapia/métodos , Interferón beta/metabolismo , Ratones Endogámicos C57BL , Mutación , Virus Oncolíticos/metabolismo , Virus Oncolíticos/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Virus de la Estomatitis Vesicular Indiana/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología
17.
Mol Ther Oncolytics ; 16: 63-74, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-31930167

RESUMEN

Genetically modified vesicular stomatitis virus (VSV) is an attractive agent for cancer treatment due to rapid intratumoral replication and observed clinical responses. Although VSV selectively kills malignant cells and can boost antitumor immunity, limited induction of intratumoral immune infiltration remains a barrier to efficacy in some cancer models. Here we engineered the oncolytic VSV platform to encode the T cell chemokine CXCL9, which is known to mediate the recruitment of activated CD8+ cytotoxic T cells and CD4+ T helper cells, and demonstrates conserved protein function between mice and humans. Chemotactic activity of the virally encoded chemokine was confirmed in vitro. Intratumoral concentration of CXCL9 was shown to increase after VSV therapy in three different cancer models, but to a much greater degree after VSV-CXCL9 therapy as compared with VSV control viruses. Despite a steep chemokine gradient from the tumor to the bloodstream, tumor trafficking of adoptively transferred and endogenous T cells was not measurably increased following VSV-CXCL9 therapy. Our results indicate that oncolytic VSV infection promotes release of CXCL9 in the tumor microenvironment, but further boosting of the functional chemokine gradient through virus engineering has little incremental impact on intratumoral immune cell infiltration in mouse and human tumor models.

18.
Neuro Oncol ; 22(12): 1757-1770, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-32459315

RESUMEN

BACKGROUND: Diffuse midline glioma, formerly DIPG (diffuse intrinsic pontine glioma), is the deadliest pediatric brainstem tumor with median survival of less than one year. Here, we investigated (i) whether direct delivery of adenovirus-expressing cluster of differentiation (CD)40 ligand (Ad-CD40L) to brainstem tumors would induce immune-mediated tumor clearance and (ii) if so, whether therapy would be associated with a manageable toxicity due to immune-mediated inflammation in the brainstem. METHODS: Syngeneic gliomas in the brainstems of immunocompetent mice were treated with Ad-CD40L and survival, toxicity, and immune profiles determined. A clinically translatable vector, whose replication would be tightly restricted to tumor cells, rAd-Δ24-CD40L, was tested in human patient-derived diffuse midline gliomas and immunocompetent models. RESULTS: Expression of Ad-CD40L restricted to brainstem gliomas by pre-infection induced complete rejection, associated with immune cell infiltration, of which CD4+ T cells were critical for therapy. Direct intratumoral injection of Ad-CD40L into established brainstem tumors improved survival and induced some complete cures but with some acute toxicity. RNA-sequencing analysis showed that Ad-CD40L therapy induced neuroinflammatory immune responses associated with interleukin (IL)-6, IL-1ß, and tumor necrosis factor α. Therefore, to generate a vector whose replication, and transgene expression, would be tightly restricted to tumor cells, we constructed rAd-Δ24-CD40L, the backbone of which has already entered clinical trials for diffuse midline gliomas. Direct intratumoral injection of rAd-Δ24-CD40L, with systemic blockade of IL-6 and IL-1ß, generated significant numbers of cures with readily manageable toxicity. CONCLUSIONS: Virus-mediated delivery of CD40L has the potential to be effective in treating diffuse midline gliomas without obligatory neuroinflammation-associated toxicity.


Asunto(s)
Neoplasias del Tronco Encefálico , Glioma , Adenoviridae , Animales , Neoplasias del Tronco Encefálico/terapia , Linfocitos T CD4-Positivos , Ligando de CD40 , Glioma/terapia , Humanos , Ratones
19.
Nat Commun ; 11(1): 790, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034147

RESUMEN

APOBEC3B, an anti-viral cytidine deaminase which induces DNA mutations, has been implicated as a mediator of cancer evolution and therapeutic resistance. Mutational plasticity also drives generation of neoepitopes, which prime anti-tumor T cells. Here, we show that overexpression of APOBEC3B in tumors increases resistance to chemotherapy, but simultaneously heightens sensitivity to immune checkpoint blockade in a murine model of melanoma. However, in the vaccine setting, APOBEC3B-mediated mutations reproducibly generate heteroclitic neoepitopes in vaccine cells which activate de novo T cell responses. These cross react against parental, unmodified tumors and lead to a high rate of cures in both subcutaneous and intra-cranial tumor models. Heteroclitic Epitope Activated Therapy (HEAT) dispenses with the need to identify patient specific neoepitopes and tumor reactive T cells ex vivo. Thus, actively driving a high mutational load in tumor cell vaccines increases their immunogenicity to drive anti-tumor therapy in combination with immune checkpoint blockade.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Citidina Desaminasa/inmunología , Inmunoterapia/métodos , Antígenos de Histocompatibilidad Menor/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Resistencia a Antineoplásicos , Epítopos/inmunología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Melanoma/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Mutación , Escape del Tumor/efectos de los fármacos
20.
Nat Commun ; 11(1): 3187, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581235

RESUMEN

The application of adoptive T cell therapies, including those using chimeric antigen receptor (CAR)-modified T cells, to solid tumors requires combinatorial strategies to overcome immune suppression associated with the tumor microenvironment. Here we test whether the inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment may help to recruit and potentiate the functionality of CAR T cells. Contrary to our hypothesis, VSVmIFNß infection is associated with attrition of murine EGFRvIII CAR T cells in a B16EGFRvIII model, despite inducing a robust proinflammatory shift in the chemokine profile. Mechanistically, type I interferon (IFN) expressed following infection promotes apoptosis, activation, and inhibitory receptor expression, and interferon-insensitive CAR T cells enable combinatorial therapy with VSVmIFNß. Our study uncovers an unexpected mechanism of therapeutic interference, and prompts further investigation into the interaction between CAR T cells and oncolytic viruses to optimize combination therapy.


Asunto(s)
Inmunoterapia Adoptiva , Interferón beta/metabolismo , Virus Oncolíticos/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Quimiocinas/metabolismo , Terapia Combinada , Femenino , Interferón beta/genética , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Viroterapia Oncolítica , Virus Oncolíticos/genética , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Bazo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA