Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Ther Oncolytics ; 24: 299-318, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35118189

RESUMEN

This study determined the influence of intravenous (i.v.) oncolytic vaccinia virus mpJX-594 (mpJX) on antitumor activity of anti-programmed death receptor-1 antibody (aPD1) in functional and metastatic pancreatic neuroendocrine tumors (PanNETs). One i.v. dose of mpJX, engineered for mice with the same plasmid design as clinical virus Pexa-Vec, was administered alone or with repeated dosing of aPD1 (mpJX+aPD1) to two contrasting genetic models of PanNET: one developing benign insulin-secreting tumors (RIP1-Tag2;C57BL/6J mice) and the other developing liver metastases (RIP1-Tag2;AB6F1 mice). Experiments revealed that aPD1 had synergistic actions with mpJX on CD8+ T cell and natural killer (NK) cell influx, apoptosis, and suppression of proliferation in PanNETs. After mpJX+aPD1, the 53-fold increase in apoptosis (5 days) and 85% reduction in proliferation (20 days) exceeded the sum of mpJX and aPD1 given separately. mpJX+aPD1 also stabilized blood insulin and glucose in mice with functional PanNETs, regressed liver metastases in mice with aggressive PanNETs, and prolonged survival of both. The findings revealed that mpJX+aPD1 converted "cold" PanNETs into immunogenic tumors with widespread cytotoxic T cell influx, tumor cell killing, and suppression of proliferation. Reduction of tumor insulin secretion from functional PanNETs prolonged survival, and anti-metastatic actions on aggressive PanNETs reduced the metastatic burden to less than before treatment. The findings support the efficacy of the vaccinia virus with aPD1 for functional and metastatic PanNETs.

2.
Cancer Gene Ther ; 28(7-8): 769-784, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32647136

RESUMEN

We have adapted a zebrafish (Danio rerio) tumor xenograft model for use in the study of oncolytic virotherapy. Following implantation of mammalian cancer cells into the perivitelline space of developing zebrafish embryos, both local and intravenous oncolytic virus treatments produce a tumor-specific infection with measurable antitumor effects. Tumor cells are injected at 48 h post fertilization, with oncolytic virus treatment then being administered 24 h later to allow for an initial period of tumor development and angiogenesis. Confocal fluorescent imaging is used to quantify dynamics within the tumor environment. The natural translucency of zebrafish at the embryo stage, coupled with the availability of strains with fluorescent immune and endothelial cell reporter lines, gives the model broad potential to allow for real time, in vivo investigation of important events within tumors throughout the course of virotherapy. Zebrafish xenografts offer a system with biologic fidelity to processes in human cancer development that influence oncolytic virus efficacy, and to our knowledge this is the first demonstration of the model's use in the context of virotherapy. Compared with other models, our protocol offers a powerful, inexpensive approach to evaluating novel oncolytic viruses and oncolytic virus-based combination therapies, with potential application to investigating the impacts of virotherapy on immune response, tumor vasculature, and metastatic disease.


Asunto(s)
Virus Oncolíticos/metabolismo , Animales , Modelos Animales , Microambiente Tumoral , Pez Cebra
3.
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
4.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26212250

RESUMEN

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Asunto(s)
Neoplasias/virología , Virus Oncolíticos/fisiología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/virología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Interferencia de ARN , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Virus Vaccinia/fisiología
5.
J Pediatr Surg ; 50(6): 983-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25840604

RESUMEN

PURPOSE: We characterize the outcomes of pediatric spontaneous pneumomediastinum in the largest series to date and propose a management pathway. METHODS: All patients at our institution with ICD-9 code 518.1 confirmed to have isolated radiographic findings of spontaneous pneumomediastinum between January 2003 and February 2014 were retrospectively reviewed for admission, intensive care unit (ICU) stay, complications, and outcome. RESULTS: We identified 96 children with 99 episodes, median age 14.1 years (IQR: 8.7-16.4). Primary symptoms were chest pain, cough, and dyspnea. Most were hospitalized (n=91, 91.9%), with 20 (20.2%) admitted to ICU. Median lengths of stay (LOS) were 1 day (IQR: 1-2) for non-ICU admissions and 3 days (IQR: 2-3) for ICU admissions. The surgical service discharged non-ICU patients 0.94 days earlier than medical services (95% CI 0.38-1.50, p=0.0014). Asthma affected neither LOS nor ICU admission rates. Follow-up imaging, when obtained (n=81, 81.8%), did not alter management. Recurrences occurred in three asthmatics, all after one year. Each was rehospitalized and discharged uneventfully. No patient developed pneumomediastinum-related complications (e.g., pneumothorax, pneumopericardium, or mediastinitis). CONCLUSION: Spontaneous pneumomediastinum without associated comorbidities can be managed with expectant outpatient observation without further imaging. Children with asthma should be treated independent of spontaneous pneumomediastinum.


Asunto(s)
Enfisema Mediastínico/terapia , Adolescente , Asma/complicaciones , Dolor en el Pecho/etiología , Niño , Tos/etiología , Disnea/etiología , Femenino , Humanos , Unidades de Cuidados Intensivos , Tiempo de Internación , Masculino , Enfisema Mediastínico/complicaciones , Enfisema Mediastínico/diagnóstico por imagen , Radiografía , Recurrencia , Estudios Retrospectivos , Espera Vigilante
6.
Nat Commun ; 6: 6410, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25817275

RESUMEN

In this study, we show that several microtubule-destabilizing agents used for decades for treatment of cancer and other diseases also sensitize cancer cells to oncolytic rhabdoviruses and improve therapeutic outcomes in resistant murine cancer models. Drug-induced microtubule destabilization leads to superior viral spread in cancer cells by disrupting type I IFN mRNA translation, leading to decreased IFN protein expression and secretion. Furthermore, microtubule-destabilizing agents specifically promote cancer cell death following stimulation by a subset of infection-induced cytokines, thereby increasing viral bystander effects. This study reveals a previously unappreciated role for microtubule structures in the regulation of the innate cellular antiviral response and demonstrates that unexpected combinations of approved chemotherapeutics and biological agents can lead to improved therapeutic outcomes.


Asunto(s)
Efecto Espectador/efectos de los fármacos , Citocinas/efectos de los fármacos , Interferón Tipo I/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Viroterapia Oncolítica , Virus Oncolíticos , ARN Mensajero/efectos de los fármacos , Infecciones por Rhabdoviridae/inmunología , Moduladores de Tubulina/farmacología , Albendazol/farmacología , Animales , Bencimidazoles/farmacología , Efecto Espectador/inmunología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Colchicina/farmacología , Citocinas/inmunología , Células HT29 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Nocodazol/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Rhabdoviridae , Células Vero , Vinblastina/análogos & derivados , Vinblastina/farmacología , Vinorelbina
8.
Cancer Res ; 73(4): 1265-75, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23393196

RESUMEN

Efforts to selectively target and disrupt established tumor vasculature have largely failed to date. We hypothesized that a vaccinia virus engineered to target cells with activation of the ras/MAPK signaling pathway (JX-594) could specifically infect and express transgenes (hGM-CSF, ß-galactosidase) in tumor-associated vascular endothelial cells in humans. Efficient replication and transgene expression in normal human endothelial cells in vitro required either VEGF or FGF-2 stimulation. Intravenous infusion in mice resulted in virus replication in tumor-associated endothelial cells, disruption of tumor blood flow, and hypoxia within 48 hours; massive tumor necrosis ensued within 5 days. Normal vessels were not affected. In patients treated with intravenous JX-594 in a phase I clinical trial, we showed dose-dependent endothelial cell infection and transgene expression in tumor biopsies of diverse histologies. Finally, patients with advanced hepatocellular carcinoma, a hypervascular and VEGF-rich tumor type, were treated with JX-594 on phase II clinical trials. JX-594 treatment caused disruption of tumor perfusion as early as 5 days in both VEGF receptor inhibitor-naïve and -refractory patients. Toxicities to normal blood vessels or to wound healing were not evident clinically or on MRI scans. This platform technology opens up the possibility of multifunctional engineered vaccinia products that selectively target and infect tumor-associated endothelial cells, as well as cancer cells, resulting in transgene expression, vasculature disruption, and tumor destruction in humans systemically.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Neovascularización Patológica/prevención & control , Virus Oncolíticos/fisiología , Virus Vaccinia/fisiología , Animales , Western Blotting , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Células Cultivadas , Ensayos Clínicos Fase I como Asunto , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Células Endoteliales/virología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/virología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/terapia , Neoplasias Experimentales/virología , Neovascularización Patológica/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Conejos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Virus Vaccinia/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Replicación Viral
9.
Diabetes Care ; 36(2): 480-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23349152

RESUMEN

OBJECTIVE: Depression is associated with the onset of type 2 diabetes. A systematic review and meta-analysis of observational studies, controlled trials, and unpublished data was conducted to examine the association between depression and insulin resistance (IR). RESEARCH DESIGN AND METHODS: Medline, EMBASE, and PsycINFO were searched for studies published up to September 2011. Two independent reviewers assessed the eligibility of each report based on predefined inclusion criteria (study design and measure of depression and IR, excluding prevalent cases of diabetes). Individual effect sizes were standardized, and a meta-analysis was performed to calculate a pooled effect size using random effects. Subgroup analyses and meta-regression were conducted to explore any potential source of heterogeneity between studies. RESULTS: Of 967 abstracts reviewed, 21 studies met the inclusion criteria of which 18 studies had appropriate data for the meta-analysis (n = 25,847). The pooled effect size (95% CI) was 0.19 (0.11-0.27) with marked heterogeneity (I(2) = 82.2%) using the random-effects model. Heterogeneity between studies was not explained by age or sex, but could be partly explained by the methods of depression and IR assessments. CONCLUSIONS: A small but significant cross-sectional association was observed between depression and IR, despite heterogeneity between studies. The pathophysiology mechanisms and direction of this association need further study using a purposively designed prospective or intervention study in samples at high risk for diabetes.


Asunto(s)
Depresión/epidemiología , Depresión/fisiopatología , Resistencia a la Insulina/fisiología , Diabetes Mellitus Tipo 2/epidemiología , Humanos , Factores de Riesgo
10.
Emerg Radiol ; 20(1): 39-44, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22898866

RESUMEN

The objective of this study is to compare the dose of CT angiography (CTA) for the diagnosis of pulmonary embolism (PE) performed using a reduced z-axis to conventional CTA for PE, both using adaptive iterative reconstruction technique on a 64-detector row device. The institutional review board approved a waiver of informed consent. A study was performed to consecutive patients having CTA for PE in the emergency department (ED). The patients underwent a reduced z-axis CTA from the top of the aortic arch to the bottom of the heart using the appropriate CT parameters and standard IV contrast injections. All patients had scans performed with 40 % ASIR and had a breast shield placed to limit breast dose. Per ED ordering criteria, the reduced z-axis protocol was appropriate for patients under 50 years old with no significant comorbidity. The control group consisted of patients from the same time period under 50 years of age who received a full z-axis scan. Technical parameters were the same for both groups other than scan length. Dose-length product (DLP) and volume CT dose index (CTDIvol) were the parameters used to evaluate differences in radiation dose to patients. The average effective dose of the full z-axis group was significantly higher (10.9 mSv (SD 4.7, range = 2.8-22)) compared to the reduced z-axis group (5.5 mSv (SD 3.0, range = 1.6-13, p < 0.001). The average effective dose for the reduced z-axis group was 49 % less than that of the full z-axis group. Reducing the z-axis of a CTA for PE significantly reduces effective radiation dose.


Asunto(s)
Angiografía/métodos , Embolia Pulmonar/diagnóstico por imagen , Dosis de Radiación , Tomografía Computarizada por Rayos X/métodos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Interpretación de Imagen Radiográfica Asistida por Computador
11.
J Virol ; 87(4): 2363-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221568

RESUMEN

Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.


Asunto(s)
Glioma/terapia , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/inmunología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
12.
Curr Psychiatry Rep ; 14(1): 8-14, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22094982

RESUMEN

We review the validity of the evidence for an association between depression and the risk of insulin resistance (IR). We describe the potentially plausible biological and behavioral mechanisms that explain how depression increases the risk of IR and consequent overt diabetes. We have identified gaps in the literature to guide future research. Evidence for bidirectional associations between depression and IR is inconsistent. Results showing positive associations between depression and IR are derived from cross-sectional studies, whereas negative findings are typically reported in cohort studies. On the other hand, tentative trial evidence suggests that the effective treatment of depression can improve IR, and that lifestyle programs improve IR and reduce depressive symptoms. These emerging themes could lead to potential new multidisciplinary approaches to preventing diabetes.


Asunto(s)
Trastorno Depresivo/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Resistencia a la Insulina/fisiología , Antidepresivos/uso terapéutico , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/epidemiología , Trastorno Depresivo/psicología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Estudios Epidemiológicos , Femenino , Humanos , Estilo de Vida , Masculino
13.
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
14.
J Vis Exp ; (52)2011 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-21730946

RESUMEN

Oncolytic Viruses (OVs) are novel therapeutics that selectively replicate in and kill tumor cells(1). Several clinical trials evaluating the effectiveness of a variety of oncolytic platforms including HSV, Reovirus, and Vaccinia OVs as treatment for cancer are currently underway(2-5). One key characteristic of oncolytic viruses is that they can be genetically modified to express reporter transgenes which makes it possible to visualize the infection of tissues by microscopy or bio-luminescence imaging(6,7). This offers a unique advantage since it is possible to infect tissues from patients ex vivo prior to therapy in order to ascertain the likelihood of successful oncolytic virotherapy(8). To this end, it is critical to appropriately sample tissue to compensate for tissue heterogeneity and assess tissue viability, particularly prior to infection(9). It is also important to follow viral replication using reporter transgenes if expressed by the oncolytic platform as well as by direct titration of tissues following homogenization in order to discriminate between abortive and productive infection. The object of this protocol is to address these issues and herein describes 1. The sampling and preparation of tumor tissue for cell culture 2. The assessment of tissue viability using the metabolic dye alamar blue 3. Ex vivo infection of cultured tissues with vaccinia virus expressing either GFP or firefly luciferase 4. Detection of transgene expression by fluorescence microscopy or using an In Vivo Imaging System (IVIS) 5. Quantification of virus by plaque assay. This comprehensive method presents several advantages including ease of tissue processing, compensation for tissue heterogeneity, control of tissue viability, and discrimination between abortive infection and bone fide viral replication.


Asunto(s)
Neoplasias/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Luciferasas de Luciérnaga/análisis , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Microscopía Fluorescente/métodos , Neoplasias/terapia , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Transgenes , Células Tumorales Cultivadas , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Virus Vaccinia/fisiología , Ensayo de Placa Viral/métodos
15.
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
16.
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
17.
Cytokine Growth Factor Rev ; 21(2-3): 135-41, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20338801

RESUMEN

Oncolytic viruses (OVs) are selected based on their ability to eliminate malignancies by direct infection and lysis of cancer cells. Originally, OVs were designed to target malignancies by taking advantage of the defects of cancer cells observed in vitro. Subsequent analysis of virus delivery and spread in vivo has demonstrated that the tumour microenvironment can impede the ability of OVs to effectively infect and spread. Despite this limitation, it is becoming increasingly evident that OVs are also able to take advantage of certain features of the tumour microenvironment. Currently, a growing body of the literature is delineating the complex interaction between OVs and the tumour microenvironment that results in an additional therapeutic activity; these viruses are able to target malignancies by rapidly altering the tumour microenvironment into a milieu that potentiates anticancer activity. Herein, we discuss strategies that capitalize on the multifaceted relationship between OVs and host-tumour interactions that enhance the toxicity of OVs to the tumour microenvironment.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Animales , Ensayos Clínicos como Asunto , Humanos , Inmunidad Innata , Neoplasias/inmunología , Neoplasias/patología , Virus Oncolíticos/genética , Resultado del Tratamiento
18.
PLoS One ; 5(12): e14462, 2010 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21283510

RESUMEN

Histone deacetylase inhibitors (HDI) dampen cellular innate immune response by decreasing interferon production and have been shown to increase the growth of vesicular stomatitis virus and HSV. As attenuated tumour-selective oncolytic vaccinia viruses (VV) are already undergoing clinical evaluation, the goal of this study is to determine whether HDI can also enhance the potency of these poxviruses in infection-resistant cancer cell lines. Multiple HDIs were tested and Trichostatin A (TSA) was found to potently enhance the spread and replication of a tumour selective vaccinia virus in several infection-resistant cancer cell lines. TSA significantly decreased the number of lung metastases in a syngeneic B16F10LacZ lung metastasis model yet did not increase the replication of vaccinia in normal tissues. The combination of TSA and VV increased survival of mice harbouring human HCT116 colon tumour xenografts as compared to mice treated with either agent alone. We conclude that TSA can selectively and effectively enhance the replication and spread of oncolytic vaccinia virus in cancer cells.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Neoplasias/metabolismo , Neoplasias/virología , Viroterapia Oncolítica/métodos , Virus Vaccinia/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Humanos , Ácidos Hidroxámicos/farmacología , Sistema Inmunológico , Interferones/metabolismo , Melanoma Experimental , Ratones , Trasplante de Neoplasias , Virus Oncolíticos/metabolismo
19.
J Biol Chem ; 281(22): 15155-63, 2006 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-16595687

RESUMEN

Components of the cellular translation machinery are targets of caspase-mediated cleavage during apoptosis that correlates with the inhibition of protein synthesis, which accompanies apoptosis. Paradoxically, protein synthesis is required for apoptosis to occur in many experimental settings. Previous studies showed that two proteins that regulate apoptosis by controlling caspase activity, XIAP and Apaf-1, are translated by a unique, cap-independent mechanism mediated by an internal ribosome entry site (IRES) that is used preferentially under conditions in which normal cap-dependent translation is repressed. We investigated the regulation of XIAP and Apaf-1 following UVC irradiation. We show that UVC irradiation leads to the inhibition of translation and cell death. Furthermore, IRES-mediated translation of Apaf-1, but not XIAP, is enhanced by UVC irradiation, and this increase in Apaf-1 translation correlated with cell death. The enhanced Apaf-1 IRES-mediated translation is caspase-independent but is negatively modulated by the eIF2alpha kinase protein kinase RNA-like endoplasmic reticulum kinase. These data suggest that progression of UV-induced apoptosis requires IRES-mediated translation of Apaf-1 to ensure continuous levels of Apaf-1 despite an overall suppression of protein synthesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas/genética , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Animales , Factor Apoptótico 1 Activador de Proteasas , Secuencia de Bases , Muerte Celular/genética , Muerte Celular/efectos de la radiación , Línea Celular , Células Cultivadas , Humanos , Ratones , Ratones Noqueados , Biosíntesis de Proteínas/efectos de la radiación , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Rayos Ultravioleta , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...