Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Sci Rep ; 11(1): 15384, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321569

RESUMEN

Determination of treatment response to immunotherapy in glioblastoma multiforme (GBM) is a process which can take months. Detection of CD8+ T cell recruitment to the tumor with a noninvasive imaging modality such as positron emission tomography (PET) may allow for tumor characterization and early evaluation of therapeutic response to immunotherapy. In this study, we utilized 89Zr-labeled anti-CD8 cys-diabody-PET to provide proof-of-concept to detect CD8+ T cell immune response to oncolytic herpes simplex virus (oHSV) M002 immunotherapy in a syngeneic GBM model. Immunocompetent mice (n = 16) were implanted intracranially with GSC005 GBM tumors, and treated with intratumoral injection of oHSV M002 or saline control. An additional non-tumor bearing cohort (n = 4) receiving oHSV M002 treatment was also evaluated. Mice were injected with 89Zr-labeled anti-CD8 cys-diabody seven days post oHSV administration and imaged with a preclinical PET scanner. Standardized uptake value (SUV) was quantified. Ex vivo tissue analyses included autoradiography and immunohistochemistry. PET imaging showed significantly higher SUV in tumors which had been treated with M002 compared to those without M002 treatment (p = 0.0207) and the non-tumor bearing M002 treated group (p = 0.0021). Accumulation in target areas, especially the spleen, was significantly reduced by blocking with the non-labeled diabody (p < 0.001). Radioactive probe accumulation in brains was consistent with CD8+ cell trafficking patterns after oHSV treatment. This PET imaging strategy could aid in distinguishing responders from non-responders during immunotherapy of GBM.


Asunto(s)
Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Glioma/terapia , Viroterapia Oncolítica/métodos , Animales , Antígenos CD8/antagonistas & inhibidores , Antígenos CD8/aislamiento & purificación , Linfocitos T CD8-positivos/virología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioma/diagnóstico por imagen , Glioma/inmunología , Glioma/virología , Humanos , Ratones , Radioisótopos/farmacología , Simplexvirus/genética , Tomografía Computarizada por Rayos X , Circonio/farmacología
3.
Transl Oncol ; 9(5): 419-430, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27751346

RESUMEN

Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, is the most common sarcoma of childhood. Despite multidrug chemotherapy regimens, surgical intervention, and radiation treatment, outcomes remain poor, especially in advanced disease, and novel therapies are needed for the treatment of these aggressive malignancies. Genetically engineered oncolytic viruses, such as herpes simplex virus-1 (HSV), are currently being explored as treatments for pediatric tumors. M002, an oncolytic HSV, has both copies of the γ134.5 gene deleted, enabling replication in tumor cells but thwarting infection of normal, postmitotic cells. We hypothesized that M002 would infect human RMS tumor cells and lead to decreased tumor cell survival in vitro and impede tumor growth in vivo. In the current study, we demonstrated that M002 could infect, replicate in, and decrease cell survival in both embryonal (ERMS) and alveolar rhabdomyosarcoma (ARMS) cells. Additionally, M002 reduced xenograft tumor growth and increased animal survival in both ARMS and ERMS. Most importantly, we showed for the first time that repeated dosing of oncolytic virus coupled with low-dose radiation provided improved tumor response in RMS. These findings provide support for the clinical investigation of oncolytic HSV in pediatric RMS.

4.
Hum Gene Ther Clin Dev ; 25(1): 16-27, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24649838

RESUMEN

Herpes simplex virus type 1 (HSV-1) mutants lacking the γ(1)34.5 neurovirulence loci are promising agents for treating malignant glioma. Arming oncolytic HSV-1 to express immunostimulatory genes may potentiate therapeutic efficacy. We have previously demonstrated improved preclinical efficacy, biodistribution, and safety of M002, a γ(1)34.5-deleted HSV-1 engineered to express murine IL-12. Herein, we describe the safety and biodistribution of M032, a γ(1)34.5-deleted HSV-1 virus that expresses human IL-12 after intracerebral administration to nonhuman primates, Aotus nancymae. Cohorts were administered vehicle, 10(6), or 10(8) pfu of M032 on day 1 and subjected to detailed clinical observations performed serially over a 92-day trial. Animals were sacrificed on days 3, 31, and 91 for detailed histopathologic assessments of all organs and to isolate and quantify virus in all organs. With the possible exception of one animal euthanized on day 16, neither adverse clinical signs nor sex- or dose-related differences were attributed to M032. Elevated white blood cell and neutrophil counts were observed in virus-injected groups on day 3, but no other significant changes were noted in clinical chemistry or coagulation parameters. Minimal to mild inflammation and fibrosis detected, primarily in meningeal tissues, in M032-injected animals on days 3 and 31 had mostly resolved by day 91. The highest viral DNA levels were detected at the injection site and motor cortex on day 3 but decreased in central nervous system tissues over time. These data demonstrate the requisite safety of intracerebral M032 administration for consideration as a therapeutic for treating malignant brain tumors.


Asunto(s)
Glioma/terapia , Herpesvirus Humano 1/genética , Infusiones Intraventriculares , Interleucina-12/genética , Viroterapia Oncolítica/métodos , Animales , Aotidae , Neoplasias Encefálicas/terapia , Vías de Administración de Medicamentos , Femenino , Interleucina-12/biosíntesis , Masculino , Replicación Viral
5.
Mol Ther ; 22(5): 1048-55, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24572293

RESUMEN

G207, a mutant herpes simplex virus (HSV) type 1, is safe when inoculated into recurrent malignant glioma. We conducted a phase 1 trial of G207 to demonstrate the safety of stereotactic intratumoral administration when given 24 hours prior to a single 5 Gy radiation dose in patients with recurrent malignant glioma. Nine patients with progressive, recurrent malignant glioma despite standard therapy were included. Patients received one dose of G207 stereotactically inoculated into the multiple sites of the enhancing tumor margin and were then treated focally with 5 Gy radiation. Treatment was well tolerated, and no patient developed HSV encephalitis. The median interval between initial diagnosis and G207 inoculation was 18 months (mean: 23 months; range: 11-51 months). Six of the nine patients had stable disease or partial response for at least one time point. Three instances of marked radiographic response to treatment occurred. The median survival time from G207 inoculation until death was 7.5 months (95% confidence interval: 3.0-12.7). In conclusion, this study showed the safety and the potential for clinical response of single-dose oncolytic HSV therapy augmented with radiation in the treatment of malignant glioma patients. Additional studies with oncolytic HSV such as G207 in the treatment of human glioma are recommended.


Asunto(s)
Terapia Genética , Glioma/genética , Glioma/radioterapia , Herpesvirus Humano 1/genética , Adulto , Femenino , Glioma/diagnóstico por imagen , Glioma/virología , Herpesvirus Humano 1/patogenicidad , Humanos , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus Oncolíticos/patogenicidad , Radiografía , Replicación Viral/genética
6.
PLoS One ; 9(1): e86843, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24497984

RESUMEN

Recently, investigators showed that mice with syngeneic murine gliomas that were treated with a neuroattenuated oncolytic herpes simplex virus-1 (oHSV), M002, had a significant increase in survival. M002 has deletions in both copies of the γ134.5 gene, enabling replication in tumor cells but precluding infection of normal cells. Previous studies have shown antitumor effects of other oHSV against a number of adult tumors including hepatocellular carcinoma and renal cell carcinoma. The purpose of the current study was to investigate the oncolytic potential of M002 against difficult to treat pediatric liver and kidney tumors. We showed that the oHSV, M002, infected, replicated, and decreased cell survival in hepatoblastoma, malignant rhabdoid kidney tumor, and renal sarcoma cell lines. In addition, we showed that in murine xenografts, treatment with M002 significantly increased survival and decreased tumor growth. Finally, these studies showed that the primary entry protein for oHSV, CD111 (nectin-1) was present in human hepatoblastoma and malignant rhabdoid kidney tumor specimens. We concluded that M002 effectively targeted these rare aggressive tumor types and that M002 may have potential for use in children with unresponsive or relapsed pediatric solid tumors.


Asunto(s)
Neoplasias Renales/terapia , Neoplasias Hepáticas/terapia , Virus Oncolíticos/genética , Tumor Rabdoide/terapia , Sarcoma de Ewing/terapia , Simplexvirus/genética , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos , Neoplasias Renales/patología , Neoplasias Hepáticas/patología , Ratones Desnudos , Nectinas , Trasplante de Neoplasias , Viroterapia Oncolítica , Tumor Rabdoide/patología , Sarcoma de Ewing/patología , Carga Tumoral , Replicación Viral
7.
PLoS One ; 8(10): e77753, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130898

RESUMEN

Despite intensive research efforts and therapeutic advances over the last few decades, the pediatric neural crest tumor, neuroblastoma, continues to be responsible for over 15% of pediatric cancer deaths. Novel therapeutic options are needed for this tumor. Recently, investigators have shown that mice with syngeneic murine gliomas treated with an engineered, neuroattenuated oncolytic herpes simplex virus-1 (oHSV), M002, had a significant increase in survival. M002 has deletions in both copies of the γ 1 34.5 gene, enabling replication in tumor cells but precluding infection of normal neural cells. We hypothesized that M002 would also be effective in the neural crest tumor, neuroblastoma. We showed that M002 infected, replicated, and decreased survival in neuroblastoma cell lines. In addition, we showed that in murine xenografts, treatment with M002 significantly decreased tumor growth, and that this effect was augmented with the addition of ionizing radiation. Importantly, survival could be increased by subsequent doses of radiation without re-dosing of the virus. Finally, these studies showed that the primary entry protein for oHSV, CD111 was expressed by numerous neuroblastoma cell lines and was also present in human neuroblastoma specimens. We concluded that M002 effectively targeted neuroblastoma and that this oHSV may have potential for use in children with unresponsive or relapsed neuroblastoma.


Asunto(s)
Neoplasias Encefálicas/terapia , Ingeniería Genética/métodos , Neuroblastoma/terapia , Viroterapia Oncolítica/métodos , Simplexvirus/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Antígenos CD11/análisis , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Humanos , Ratones , Ratones Desnudos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuroblastoma/virología , Fosforilación , Factor de Transcripción STAT1/metabolismo , Simplexvirus/fisiología , Células Vero , Replicación Viral , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Virol ; 86(9): 5304-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379082

RESUMEN

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Asunto(s)
Vectores Genéticos/genética , Interleucina-12/genética , Simplexvirus/genética , Aciclovir/farmacología , Animales , Antivirales/farmacología , Aotidae , Encéfalo/patología , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Glioma/genética , Glioma/mortalidad , Glioma/terapia , Humanos , Interleucina-12/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones SCID , Simplexvirus/efectos de los fármacos , Análisis de Supervivencia , Replicación Viral/efectos de los fármacos , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cell Biol ; 31(16): 3196-207, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21690298

RESUMEN

Inherently unstable mRNAs contain AU-rich elements (AREs) in the 3' untranslated regions. Expression of ARE-containing type I interferon transcripts is robustly induced upon viral infection and rapidly shut off thereafter. Their transient accumulation is partly mediated through posttranscriptional regulation. Here we show that mouse embryonic fibroblasts derived from knockout mice deficient in KH-type splicing regulatory protein (KSRP), an RNA-binding protein required for ARE-mediated mRNA decay, produce higher levels of Ifna and Ifnb mRNAs in response to viral infection as a result of decreased mRNA decay. Functional analysis showed that KSRP is required for the decay of Ifna4 and Ifnb mRNAs by interaction with AREs. The increased IFN expression renders Ksrp(-)(/)(-) cells refractory to herpes simplex virus type 1 and vesicular stomatitis virus infection. These findings support a role of a posttranscriptional mechanism in the control of type I IFN expression and highlight the function of KSRP in innate immunity by negatively regulating IFN production.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/genética , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Interferencia de ARN/inmunología , Proteínas de Unión al ARN/inmunología , Transactivadores/inmunología , Virosis/inmunología , Animales , Células Cultivadas , Herpes Simple/inmunología , Ratones , Ratones Noqueados , Estabilidad del ARN/inmunología , Infecciones por Rhabdoviridae/inmunología
10.
J Neurooncol ; 95(2): 199-209, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19521665

RESUMEN

Oncolytic herpes simplex viruses (HSV) hold promise for therapy of glioblastoma multiforme (GBM) resistant to traditional therapies. We examined the ability of genetically engineered HSV to infect and kill cells that express CD133, a putative marker of glioma progenitor cells (GPC), to determine if GPC have an inherent therapeutic resistance to HSV. Expression of CD133 and CD111 (nectin-1), the major entry molecule for HSV, was variable in six human glioma xenografts, at initial disaggregation and after tissue culture. Importantly, both CD133+ and CD133- populations of glioma cells expressed CD111 in similar relative proportions in five xenografts, and CD133+ and CD133- glioma cell subpopulations were equally sensitive to killing in vitro by graded dilutions of wild-type HSV-1(F) or several different gamma(1)34.5-deleted viruses. GPC did not display an inherent resistance to HSV. While CD111 expression was an important factor for determining sensitivity of glioma cells to HSV oncolysis, it was not the only factor. Our findings support the notion that HSV will not be able to effectively enter, infect, and kill cells in tumors that have low CD111 expression (<20%). However, virotherapy with HSV may be very effective against CD111+ GPC resistant to traditional therapies.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Encefálicas/terapia , Moléculas de Adhesión Celular/metabolismo , Glioblastoma/terapia , Glicoproteínas/metabolismo , Herpesvirus Humano 1/fisiología , Viroterapia Oncolítica , Péptidos/metabolismo , Antígeno AC133 , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Efecto Citopatogénico Viral , Ingeniería Genética/métodos , Vectores Genéticos , Glioblastoma/metabolismo , Glioblastoma/virología , Humanos , Ratones , Ratones Desnudos , Nectinas , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...