Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Mol Ther ; 24(8): 1492-501, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27203445

RESUMEN

We have conducted a phase 1 study of intravenous vvDD, a Western Reserve strain oncolytic vaccinia virus, on 11 patients with standard treatment-refractory advanced colorectal or other solid cancers. The primary endpoints were maximum tolerated dose and associated toxicity while secondary endpoints were pharmacokinetics, pharmacodynamics, immune responses, and antitumor activity. No dose-limiting toxicities and treatment related severe adverse events were observed. The most common adverse events were grades 1/2 flu-like symptoms. Virus genomes were detectable in the blood 15-30 minutes after virus administration in a dose-dependent manner. There was evidence of a prolonged virus replication in tumor tissues in two patients, but no evidence of virus replication in non-tumor tissues, except a healed injury site and an oral thrush. Over 100-fold of anti-viral antibodies were induced in patients' sera. A strong induction of inflammatory and Th1, but not Th2 cytokines, suggested a potent Th1-mediated immunity against the virus and possibly the cancer. One patient showed a mixed response on PET-CT with resolution of some liver metastases, and another patient with cutaneous melanoma demonstrated clinical regression of some lesions. Given the confirmed safety, further trials evaluating intravenous vvDD in combination with therapeutic transgenes, immune checkpoint blockade or complement inhibitors, are warranted.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Poxviridae/genética , Adulto , Anciano , Anticuerpos Antivirales/inmunología , Terapia Combinada , Citocinas/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/farmacocinética , Humanos , Mediadores de Inflamación , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias/diagnóstico , Neoplasias/mortalidad , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Poxviridae/inmunología , Retratamiento , Resultado del Tratamiento
3.
Mol Ther ; 23(9): 1532-40, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26073886

RESUMEN

Fifteen patients with treatment-refractory colorectal cancer were enrolled on a phase 1b study of Pexa-Vec (pexastimogene devacirepvec; JX-594), an oncolytic and immunotherapeutic vaccinia designed to selectively replicate in cancer cells. Pexa-Vec was administered intravenously every 14 days, at dose levels of 1 × 10(6), 1 × 10(7), or 3 × 10(7) plaque-forming units (pfu)/kg. The primary endpoint was to determine the maximum tolerated dose. Secondary endpoints were pharmacokinetics and pharmacodynamics as well as antitumor activity. Patients were heavily pretreated (mean 4.5 lines of therapy). All patients received at least two Pexa-Vec doses (median = 4; range = 2-4). No dose-limiting toxicities were reported, and the maximum tolerated dose was not reached. The most common adverse events were grade 1/2 flu-like symptoms, generally lasting <24 hours. During the first and last cycles, genome pharmacokinetics were unchanged. Infectious pfu could be detected in plasma up to 2 hours after cycle 1 and up to 30 minutes after cycle 4 (when antivaccinia antibody titers are known to have peaked). Ten patients (67%) had radiographically stable disease. Given the acceptable safety profile of multiple intravenous Pexa-Vec infusions in patients with treatment-refractory colorectal cancer, further trials evaluating efficacy of intravenous Pexa-Vec, as monotherapy or in combination with chemotherapeutic agents, is warranted in this patient population.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inmunoterapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Vaccinia/genética , Administración Intravenosa , Adulto , Anciano , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Terapia Combinada , Citocinas/sangre , Esquema de Medicación , Femenino , Vectores Genéticos/administración & dosificación , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Enfermedades de la Piel/etiología , Enfermedades de la Piel/patología , Resultado del Tratamiento
4.
Mol Ther ; 23(1): 202-14, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25292189

RESUMEN

Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.


Asunto(s)
Neoplasias de la Mama/terapia , Neoplasias del Colon/terapia , Melanoma/terapia , Neoplasias Pancreáticas/terapia , Neoplasias Cutáneas/terapia , Virus Vaccinia/inmunología , Replicación Viral/genética , Anciano , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Relación Dosis-Respuesta Inmunológica , Femenino , Eliminación de Gen , Humanos , Inyecciones Intralesiones , Masculino , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Virus Vaccinia/genética , Virus Vaccinia/crecimiento & desarrollo
5.
Mol Ther ; 23(3): 602-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25531693

RESUMEN

Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Vacunas contra el Cáncer/inmunología , Rayos gamma , Inmunoterapia/métodos , Viroterapia Oncolítica/métodos , Virus Vaccinia/inmunología , Adolescente , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/administración & dosificación , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Niño , Preescolar , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Inyecciones Intralesiones , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Masculino , Estadificación de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/patología , Neuroblastoma/terapia , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Sarcoma de Ewing/terapia , Vacunación , Virus Vaccinia/genética , Adulto Joven
6.
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
7.
Nature ; 452(7185): 323-8, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18272964

RESUMEN

Transcriptional activation of cytokines, such as type-I interferons (interferon (IFN)-alpha and IFN-beta), constitutes the first line of antiviral defence. Here we show that translational control is critical for induction of type-I IFN production. In mouse embryonic fibroblasts lacking the translational repressors 4E-BP1 and 4E-BP2, the threshold for eliciting type-I IFN production is lowered. Consequently, replication of encephalomyocarditis virus, vesicular stomatitis virus, influenza virus and Sindbis virus is markedly suppressed. Furthermore, mice with both 4E- and 4E-BP2 genes (also known as Eif4ebp1 and Eif4ebp2, respectively) knocked out are resistant to vesicular stomatitis virus infection, and this correlates with an enhanced type-I IFN production in plasmacytoid dendritic cells and the expression of IFN-regulated genes in the lungs. The enhanced type-I IFN response in 4E-BP1-/- 4E-BP2-/- double knockout mouse embryonic fibroblasts is caused by upregulation of interferon regulatory factor 7 (Irf7) messenger RNA translation. These findings highlight the role of 4E-BPs as negative regulators of type-I IFN production, via translational repression of Irf7 mRNA.


Asunto(s)
Inmunidad Innata/inmunología , Factor 7 Regulador del Interferón/biosíntesis , Biosíntesis de Proteínas , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Células Dendríticas/inmunología , Embrión de Mamíferos/citología , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Fibroblastos/virología , Eliminación de Gen , Inmunidad Innata/genética , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología , Fenómenos Fisiológicos de los Virus , Replicación Viral
8.
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
9.
Mol Ther ; 19(10): 1913-22, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21772252

RESUMEN

JX-594 is a targeted and granulocyte macrophage-colony stimulating factor (GM-CSF)-expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In order to study the mechanisms-of-action (MOA) of JX-594 in humans, a mechanistic proof-of-concept clinical trial was performed at a low dose equivalent to ≤10% of the maximum-tolerated dose (MTD) in other clinical trials. Ten patients with previously treated stage IV melanoma were enrolled. Tumors were injected weekly for up to nine total treatments. Blood samples and tumor biopsies were analyzed for evidence of transgene activity, virus replication, and immune stimulation. The ß-galactosidase (ß-gal) transgene was expressed in all patients as evidenced by antibody induction. Six patients had significant induction of GM-CSF-responsive white blood cell (WBC) subsets such as neutrophils (25-300% increase). JX-594 replication and subsequent shedding into blood was detectable in five patients after cycles 1-9. Tumor biopsies demonstrated JX-594 replication, perivascular lymphocytic infiltration, and diffuse tumor necrosis. Mild flu-like symptoms were the most common adverse events. In sum, JX-594 replication, oncolysis, and expression of both transgenes were demonstrated; replication was still evident after multiple cycles. These findings have implications for further clinical development of JX-594 and other transgene-armed oncolytic viruses.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/terapia , Viroterapia Oncolítica , Poxviridae/genética , Adulto , Anciano , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Poxviridae/fisiología , Transgenes
10.
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
11.
Mol Ther ; 19(6): 1170-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21427706

RESUMEN

JX-594 is a targeted and granulocyte-macrophage colony stimulating factor (GM-CSF) expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In a phase 1 trial, JX-594 injection into hepatocellular carcinoma (HCC) was well-tolerated and associated with viral replication, decreased tumor perfusion, and tumor necrosis. We hypothesized that JX-594 and sorafenib, a small molecule inhibitor of B-raf and vascular endothelial growth factor receptor (VEGFR) approved for HCC, would have clinical benefit in combination given their demonstrated efficacy in HCC patients and their complementary mechanisms-of-action. HCC cell lines were uniformly sensitive to JX-594. Anti-raf kinase effects of concurrent sorafenib inhibited JX-594 replication in vitro, whereas sequential therapy was superior to either agent alone in murine tumor models. We therefore explored pilot safety and efficacy of JX-594 followed by sorafenib in three HCC patients. In all three patients, sequential treatment was (i) well-tolerated, (ii) associated with significantly decreased tumor perfusion, and (iii) associated with objective tumor responses (Choi criteria; up to 100% necrosis). HCC historical control patients on sorafenib alone at the same institutions had no objective tumor responses (0 of 15). Treatment of HCC with JX-594 followed by sorafenib has antitumoral activity, and JX-594 may sensitize tumors to subsequent therapy with VEGF/VEGFR inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencenosulfonatos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/terapia , Piridinas/uso terapéutico , Virus Vaccinia/fisiología , Animales , Línea Celular Tumoral , Femenino , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/terapia , Melanoma/tratamiento farmacológico , Melanoma/terapia , Ratones , Ratones SCID , Niacinamida/análogos & derivados , Viroterapia Oncolítica/métodos , Compuestos de Fenilurea , Sorafenib , Virus Vaccinia/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Front Immunol ; 13: 1029269, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36405739

RESUMEN

Colorectal cancer is the third most diagnosed cancer and the second leading cause of cancer mortality worldwide, highlighting an urgent need for new therapeutic options and combination strategies for patients. The orchestration of potent T cell responses against human cancers is necessary for effective antitumour immunity. However, regression of a limited number of cancers has been induced by immune checkpoint inhibitors, T cell engagers (TCEs) and/or oncolytic viruses. Although one TCE has been FDA-approved for the treatment of hematological malignancies, many challenges exist for the treatment of solid cancers. Here, we show that TCEs targeting CEACAM5 and CD3 stimulate robust activation of CD4 and CD8-positive T cells in in vitro co-culture models with colorectal cancer cells, but in vivo efficacy is hindered by a lack of TCE retention in the tumour microenvironment and short TCE half-life, as demonstrated by HiBiT bioluminescent TCE-tagging technology. To overcome these limitations, we engineered Bispecific Engager Viruses, or BEVirs, a novel tumour-targeted vaccinia virus platform for intra-tumour delivery of these immunomodulatory molecules. We characterized virus-mediated TCE-secretion, TCE specificity and functionality from infected colorectal cancer cells and patient tumour samples, as well as TCE cytotoxicity in spheroid models, in the presence and absence of T cells. Importantly, we show regression of colorectal tumours in both syngeneic and xenograft mouse models. Our data suggest that a different profile of cytokines may contribute to the pro-inflammatory and immune effects driven by T cells in the tumour microenvironment to provide long-lasting immunity and abscopal effects. We establish combination regimens with immune checkpoint inhibitors for aggressive colorectal peritoneal metastases. We also observe a significant reduction in lung metastases of colorectal tumours through intravenous delivery of our oncolytic virus driven T-cell based combination immunotherapy to target colorectal tumours and FAP-positive stromal cells or CTLA4-positive Treg cells in the tumour microenvironment. In summary, we devised a novel combination strategy for the treatment of colorectal cancers using oncolytic vaccinia virus to enhance immune-payload delivery and boost T cell responses within tumours.


Asunto(s)
Neoplasias Colorrectales , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Ratones , Animales , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Virus Vaccinia , Modelos Animales de Enfermedad , Neoplasias Colorrectales/terapia , Microambiente Tumoral
13.
Methods Mol Biol ; 2058: 285-293, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31486046

RESUMEN

Oncolytic viral immunotherapy based on the MG1 Maraba platform has undergone extensive preclinical evaluation, resulting in the advancement of two programs into clinical trials. MG1 Maraba encoding tumor antigens (tumor associated antigens or viral antigens) are used to boost antitumor immunity, while MG1 Maraba infects tumors, causes oncolysis and transforms the tumor microenvironment. An overview of MG1 Maraba clinical development is outlined here, along with general considerations relating to the design of clinical trials for complex biologic products such as oncolytic viral immunotherapies. These include choice of patient population, optimized treatment regimen, and endpoints which provide early signals of activity and inform the late-stage development path of these agents with novel mechanisms of action.


Asunto(s)
Vectores Genéticos/genética , Virus Oncolíticos/genética , Rhabdoviridae/genética , Investigación Biomédica Traslacional , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Terapia Genética/métodos , Humanos , Inmunoterapia/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Proyectos de Investigación , Rhabdoviridae/inmunología
14.
Clin Cancer Res ; 14(15): 4891-7, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676763

RESUMEN

PURPOSE: Nasopharyngeal carcinoma (NPC) is a malignancy of the head and neck region that is associated with EBV latency. Curative treatments for NPC achieve modest survival rates, underscoring a need to develop novel therapies. We evaluated the therapeutic potential of a mutant vesicular stomatitis virus (VSVDelta51) as single treatment modality or in combination with ionizing radiation (RT) in NPC. EXPERIMENTAL DESIGN: MTS assay was used to assess cell viability in vitro; apoptosis was measured using propidium iodide staining and caspase activation. In vivo experiments were conducted using tumor-bearing nude mice with or without local RT (4 Gy). Apoptosis was assessed in excised tumor sections with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining. RESULTS: Our data showed that NPC cells are exquisitely sensitive to VSVDelta51 oncolysis, which correlated with the presence of EBV. Efficacy of VSVDelta51 against NPC cells was further augmented when combined with RT. A single systemic injection of VSVDelta51 achieved 50% survival in treated mice, which increased to 83% when combined with local tumor RT. In addition to induction of apoptosis, an antiangiogenic effect of VSVDelta51 was observed in vivo, suggesting a novel tumoricidal mechanism for VSVDelta51. This virus also prevented growth of NPC sphere-forming cells in vitro, showing potential utility in targeting NPC-initiating cells. CONCLUSIONS: Our data represent the first report showing that EBV-positive NPC cells are exquisitely sensitive to VSVDelta51 oncolysis and documenting the successful utilization of this combinatorial regimen as a novel curative therapeutic strategy for NPC.


Asunto(s)
Carcinoma/terapia , Mutación , Neoplasias Nasofaríngeas/terapia , Vesiculovirus/metabolismo , Animales , Apoptosis , Carcinoma/radioterapia , Línea Celular Tumoral , Supervivencia Celular , Terapia Combinada/métodos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Nasofaríngeas/radioterapia , Trasplante de Neoplasias , Resultado del Tratamiento , Vesiculovirus/genética
15.
Oncoimmunology ; 8(1): e1512329, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30546947

RESUMEN

Multiple immunotherapeutics have been approved for cancer patients, however advanced solid tumors are frequently refractory to treatment. We evaluated the safety and immunogenicity of a vaccination approach with multimodal oncolytic potential in non-human primates (NHP) (Macaca fascicularis). Primates received a replication-deficient adenoviral prime, boosted by the oncolytic Maraba MG1 rhabdovirus. Both vectors expressed the human MAGE-A3. No severe adverse events were observed. Boosting with MG1-MAGEA3 induced an expansion of hMAGE-A3-specific CD4+ and CD8+ T-cells with the latter peaking at remarkable levels and persisting for several months. T-cells reacting against epitopes fully conserved between simian and human MAGE-A3 were identified. Humoral immunity was demonstrated by the detection of circulating MAGE-A3 antibodies. These preclinical data establish the capacity for the Ad:MG1 vaccination to engage multiple effector immune cell populations without causing significant toxicity in outbred NHPs. Clinical investigations utilizing this program for the treatment of MAGE-A3-positive solid malignancies are underway (NCT02285816, NCT02879760).

16.
Curr Opin Mol Ther ; 10(1): 32-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18228179

RESUMEN

The field of oncolytic virus (OV) therapy is an innovative and evolving science, taking advantage of the ability of select viruses to preferentially infect and kill human tumor cells. However, the contribution of the tumor microenvironment, and especially the induced innate immune responses to both the tumor and the virus, has been demonstrated to be a major player in the success of OV therapies. Innate immunity and inflammation in particular can have opposing effects; these can augment OV therapy by enhancing tumor destruction, yet can also recognize and clear the invading virus to significantly hinder viral dissemination through the tumor tissues. This review considers how inflammation and innate immunity impinge on current OV candidates to either facilitate or hinder virotherapy. Novel approaches that modulate or harness the innate immune system to specifically enhance OV-mediated tumor destruction are also discussed.


Asunto(s)
Inmunidad Innata/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Animales , Humanos
17.
Cancer Res ; 78(4): 922-937, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29259007

RESUMEN

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


Asunto(s)
Antineoplásicos/uso terapéutico , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Sunitinib/uso terapéutico , Animales , Antineoplásicos/farmacología , Humanos , Ratones , Ratones Transgénicos , Sunitinib/farmacología , Virus Vaccinia/inmunología
18.
Oncolytic Virother ; 7: 117-128, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538968

RESUMEN

Oncolytic activity of the MG1 strain of the Maraba vesiculovirus has proven efficacy in numerous preclinical cancer models, and relied not only on a direct cytotoxicity but also on the induction of both innate and adaptive antitumor immunity. To further expand tumor-specific T-cell effector and long-lasting memory compartments, we introduced the MG1 virus in a prime-boost cancer vaccine strategy. To this aim, a replication-incompetent adenoviral [Ad] vector together with the oncolytic MG1 have each been armed with a transgene expressing a same tumor antigen. Immune priming with the Ad vaccine subsequently boosted with the MG1 vaccine mounted tumor-specific responses of remarkable magnitude, which significantly prolonged survival in various murine cancer models. Based on these promising results, we validated the safety profile of the Ad:MG1 oncolytic vaccination strategy in nonhuman primates and initiated clinical investigations in cancer patients. Two clinical trials are currently under way (NCT02285816; NCT02879760). The present review will recapitulate the discoveries that led to the development of MG1 oncolytic vaccines from bench to bedside.

19.
EBioMedicine ; 9: 31-36, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27407036

RESUMEN

Oncolytic viruses (OV) are replicating viral therapeutics for the treatment of cancer and have been in laboratory development for about twenty years. Recently, the FDA approved Imlygic, a herpes virus based therapeutic for the treatment of melanoma and thus OVs have entered a new era where they are a weapon in the armament of the oncologist. OVs are unique therapeutics with multiple mechanisms of therapeutic activity. The exact path for their development and eventual uptake by pharmaceutical companies is somewhat clouded by an uncertain identity. Are they vaccines, tumour lysing therapeutics, inducers of innate immunity, gene therapy vectors, anti-vascular agents or all of the above? Should they be developed as stand-alone loco-regional therapeutics, systemically delivered tumour hunters or immune modulators best tested as combination therapeutics? We summarize data here supporting the idea, depending upon the virus, that OVs can be any or all of these things. Pursuing a "one-size fits all" approach is counter-productive to their clinical development and instead as a field we should build on the strengths of individual virus platforms.


Asunto(s)
Vectores Genéticos/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Inmunomodulación , Inmunoterapia/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Resultado del Tratamiento , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
Curr Opin Virol ; 13: 55-60, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25989094

RESUMEN

Developing a live anti-cancer agent derived in most cases from human pathogens presents a unique set of challenges to clinical development versus those anticipated with standard chemotherapeutics and small molecules. The selection of therapeutic targets for oncolytic virus (OV) clinical development, as is true with the development of any agent for cancer therapy, requires careful consideration beyond preclinical and early clinical data, especially when multiple indications may initially appear equally promising. Further, the added complexity of the potential for infectious complications following OV therapy must be anticipated in order to efficiently and safely conduct clinical studies. As more OV enter the clinic, these issues will become increasingly important to successful OV drug development.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Animales , Humanos , Neoplasias/virología , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA