Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gene Ther ; 24(1): 21-30, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27779616

RESUMEN

The anti-tumour effects associated with oncolytic virus therapy are mediated significantly through immune-mediated mechanisms, which depend both on the type of virus and the route of delivery. Here, we show that intra-tumoral oncolysis by Reovirus induced the priming of a CD8+, Th1-type anti-tumour response. By contrast, systemically delivered Vesicular Stomatitis Virus expressing a cDNA library of melanoma antigens (VSV-ASMEL) promoted a potent anti-tumour CD4+ Th17 response. Therefore, we hypothesised that combining the Reovirus-induced CD8+ T cell response, with the VSV-ASMEL CD4+ Th17 helper response, would produce enhanced anti-tumour activity. Consistent with this, priming with intra-tumoral Reovirus, followed by an intra-venous VSV-ASMEL Th17 boost, significantly improved survival of mice bearing established subcutaneous B16 melanoma tumours. We also show that combination of either therapy alone with anti-PD-1 immune checkpoint blockade augmented both the Th1 response induced by systemically delivered Reovirus in combination with GM-CSF, and also the Th17 response induced by VSV-ASMEL. Significantly, anti-PD-1 also uncovered an anti-tumour Th1 response following VSV-ASMEL treatment that was not seen in the absence of checkpoint blockade. Finally, the combination of all three treatments (priming with systemically delivered Reovirus, followed by double boosting with systemic VSV-ASMEL and anti-PD-1) significantly enhanced survival, with long-term cures, compared to any individual, or double, combination therapies, associated with strong Th1 and Th17 responses to tumour antigens. Our data show that it is possible to generate fully systemic, highly effective anti-tumour immunovirotherapy by combining oncolytic viruses, along with immune checkpoint blockade, to induce complementary mechanisms of anti-tumour immune responses.


Asunto(s)
Puntos de Control del Ciclo Celular , Inmunoterapia/métodos , Melanoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular Tumoral , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Antígenos Específicos del Melanoma/genética , Antígenos Específicos del Melanoma/inmunología , Ratones , Virus Oncolíticos/genética , Reoviridae/genética , Reoviridae/inmunología , Células TH1/citología , Células TH1/inmunología , Células TH1/virología , Células Th17/citología , Células Th17/inmunología , Células Th17/virología , Vesiculovirus/genética , Vesiculovirus/inmunología
2.
Gene Ther ; 23(4): 357-68, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26814609

RESUMEN

Oncolytic strains of vaccinia virus are currently in clinical development with clear evidence of safety and promising signs of efficacy. Addition of therapeutic genes to the viral genome may increase the therapeutic efficacy of vaccinia. We evaluated the therapeutic potential of vaccinia virus expressing the sodium iodide symporter (NIS) in prostate cancer models, combining oncolysis, external beam radiotherapy and NIS-mediated radioiodide therapy. The NIS-expressing vaccinia virus (VV-NIS), GLV-1h153, was tested in in vitro analyzes of viral cell killing, combination with radiotherapy, NIS expression, cellular radioiodide uptake and apoptotic cell death in PC3, DU145, LNCaP and WPMY-1 human prostate cell lines. In vivo experiments were carried out in PC3 xenografts in CD1 nude mice to assess NIS expression and tumor radioiodide uptake. In addition, the therapeutic benefit of radioiodide treatment in combination with viral oncolysis and external beam radiotherapy was measured. In vitro viral cell killing of prostate cancers was dose- and time-dependent and was through apoptotic mechanisms. Importantly, combined virus therapy and iodizing radiation did not adversely affect oncolysis. NIS gene expression in infected cells was functional and mediated uptake of radioiodide both in vitro and in vivo. Therapy experiments with both xenograft and immunocompetent Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mouse models showed that the addition of radioiodide to VV-NIS-infected tumors was more effective than each single-agent therapy, restricting tumor growth and increasing survival. In conclusion, VV-NIS is effective in prostate cancer models. This treatment modality would be an attractive complement to existing clinical radiotherapy practice.


Asunto(s)
Terapia Genética/métodos , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Simportadores/genética , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/virología , Distribución Aleatoria , Simportadores/metabolismo , Transfección , Virus Vaccinia/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Cancer ; 134(5): 1091-101, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23982804

RESUMEN

Reovirus is an oncolytic virus (OV), which acts by both direct tumor cell killing and priming of antitumor immunity. A major obstacle for effective oncolytic virotherapy is effective delivery of OV to tumor cells. Ovarian cancer is often confined to the peritoneal cavity and therefore i.p. delivery of reovirus may provide the ideal locoregional delivery, avoiding systemic dissemination. However, ovarian cancer is associated with an accumulation of ascitic fluid, which may interfere with oncolytic viral therapy. Here, we investigated the effect of ascites on reovirus-induced oncolysis against primary ovarian cancer cells and ovarian cancer cell lines. In the absence of ascites, reovirus was cytotoxic against ovarian cancer cells; however, cytotoxicity was abrogated in the presence of ascitic fluid. Neutralizing antibodies (NAb) were identified as the cause of this inhibition. Loading OV onto cell carriers may facilitate virus delivery in the presence of NAb and immune cells which have their own antitumor effector activity are particularly appealing. Immature dendritic cells (iDC), Lymphokine-activated killer (LAK) cells and LAKDC cocultures were tested as potential carriers for reovirus for tumor cell killing and immune cell priming. Reovirus-loaded LAKDC, and to a lesser degree iDC, were able to: (i) protect from NAb and hand-off reovirus for tumor cell killing; (ii) induce a proinflammatory cytokine milieu (IFNÉ£, IL-12, IFNα and TNFα) and (iii) generate an innate and specific antitumor adaptive immune response. Hence, LAKDC pulsed with reovirus represent a novel, clinically practical treatment for ovarian cancer to maximise both direct and innate/adaptive immune-mediated tumor cell killing.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Ascitis/inmunología , Células Dendríticas/inmunología , Células Asesinas Activadas por Linfocinas/inmunología , Viroterapia Oncolítica , Neoplasias Ováricas/terapia , Reoviridae/inmunología , Apoptosis , Citocinas/biosíntesis , Femenino , Humanos , Neoplasias Ováricas/inmunología , Células Tumorales Cultivadas
4.
Gene Ther ; 20(1): 7-15, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22170342

RESUMEN

Oncolytic viruses (OV) are promising treatments for cancer, with several currently undergoing testing in randomised clinical trials. Measles virus (MV) has not yet been tested in models of human melanoma. This study demonstrates the efficacy of MV against human melanoma. It is increasingly recognised that an essential component of therapy with OV is the recruitment of host antitumour immune responses, both innate and adaptive. MV-mediated melanoma cell death is an inflammatory process, causing the release of inflammatory cytokines including type-1 interferons and the potent danger signal HMGB1. Here, using human in vitro models, we demonstrate that MV enhances innate antitumour activity, and that MV-mediated melanoma cell death is capable of stimulating a melanoma-specific adaptive immune response.


Asunto(s)
Virus del Sarampión/inmunología , Melanoma/inmunología , Virus Oncolíticos/inmunología , Muerte Celular/inmunología , Línea Celular Tumoral , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Virus del Sarampión/patogenicidad , Melanoma/patología , Melanoma/virología , Virus Oncolíticos/patogenicidad , Regulación hacia Arriba
5.
Gene Ther ; 20(5): 521-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22895509

RESUMEN

Oncolytic reovirus is currently under active investigation in a range of tumour types. Early phase studies have shown that this agent has modest monotherapy efficacy and its future development is likely to focus on combination regimens with cytotoxic chemotherapy. Indeed, phase I/II clinical trials have confirmed that reovirus can be safely combined with cytotoxic drugs, including a platin-taxane doublet regimen, which is currently being tested in a phase III clinical trial in patients with relapsed/metastatic head and neck cancer. Therefore, we have tested this triple (reovirus, cisplatin, paclitaxel) combination therapy in a panel of four head and neck cancer cell lines. Using the combination index (CI) method, the triple therapy demonstrated synergistic cytotoxicity in vitro in both malignant and non-malignant cell lines. In head and neck cancer cell lines, this was associated with enhanced caspase 3 and 7 cleavage, but no increase in viral replication. In vitro analyses confirmed colocalisation of markers of reovirus infection and caspase 3. Triple therapy was significantly more effective than reovirus or cisplatin-paclitaxel in athymic nude mice. These data suggest that the combination of reovirus plus platin-taxane doublet chemotherapy has significant activity in head and neck cancer and underpin the current phase III study in this indication.


Asunto(s)
Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Antineoplásicos , Línea Celular Tumoral , Cisplatino/administración & dosificación , Ensayos Clínicos Fase I como Asunto , Terapia Combinada , Neoplasias de Cabeza y Cuello/virología , Humanos , Ratones , Orthoreovirus/genética , Paclitaxel/administración & dosificación
6.
Gene Ther ; 19(5): 543-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21918546

RESUMEN

Tumor selective, replication competent viruses are being tested for cancer gene therapy. This approach introduces a new therapeutic paradigm due to potential replication of the therapeutic agent and induction of a tumor-specific immune response. However, the experimental outcomes are quite variable, even when studies utilize highly inbred strains of mice and the same cell line and virus. Recognizing that virotherapy is an exercise in population dynamics, we utilize mathematical modeling to understand the variable outcomes observed when B16ova malignant melanoma tumors are treated with vesicular stomatitis virus in syngeneic, fully immunocompetent mice. We show how variability in the initial tumor size and the actual amount of virus delivered to the tumor have critical roles on the outcome of therapy. Virotherapy works best when tumors are small, and a robust innate immune response can lead to superior tumor control. Strategies that reduce tumor burden without suppressing the immune response and methods that maximize the amount of virus delivered to the tumor should optimize tumor control in this model system.


Asunto(s)
Matemática , Melanoma Experimental/terapia , Viroterapia Oncolítica/métodos , Virus de la Estomatitis Vesicular Indiana , Animales , Línea Celular Tumoral , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Replicación Viral
7.
Gene Ther ; 19(11): 1048-57, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22113313

RESUMEN

The limitations of the current oncolytic adenoviruses for cancer therapy include insufficient potency and poor distribution of the virus throughout the tumor mass. To address these problems, we generated an oncolytic adenovirus expressing the hyperfusogenic form of the gibbon-ape leukemia virus (GALV) envelope glycoprotein under the control of the adenovirus major late promoter. The oncolytic properties of the new fusogenic adenovirus, ICOVIR16, were analyzed both in vitro and in vivo, and compared with that of its non-fusogenic counterpart, ICOVIR15. Our results indicate that GALV expression by ICOVIR16 induced extensive syncytia formation and enhanced tumor cell killing in a variety of tumor cell types. When injected intratumorally or intravenously into mice with large pre-established melanoma or pancreatic tumors, ICOVIR16 rapidly reduced tumor burden, and in some cases, resulted in complete eradication of the tumors. Importantly, GALV expression induced tumor cell fusion in vivo and enhanced the spreading of the virus throughout the tumor. Taken together, these results indicate that GALV expression can improve the antitumoral potency of an oncolytic adenovirus and suggest that ICOVIR16 is a promising candidate for clinical evaluation in patients with cancer.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Células Gigantes , Virus de la Leucemia del Gibón/genética , Virus Oncolíticos , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Cricetinae , Femenino , Regulación Viral de la Expresión Génica , Orden Génico , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/metabolismo , Células Gigantes/virología , Humanos , Inyecciones , Masculino , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Nat Med ; 4(5): 581-7, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9585232

RESUMEN

In situ killing of tumor cells using suicide gene transfer to generate death by a non-apoptotic pathway was associated with high immunogenicity and induction of heat shock protein (hsp) expression. In contrast, a syngeneic colorectal tumor line, CMT93, killed predominantly by apoptosis, showed low levels of hsp expression and less immunogenicity. When apoptosis was inhibited in CMT93 cells by overexpression of bcl-2, hsp was also induced. Furthermore, when cDNA encoding hsp70 was stably transfected into B16 and CMT93 cells, its expression significantly enhanced the immunogenicity of both tumors. Increased levels of hsp, induced by non-apoptotic cell killing, may provide an immunostimulatory signal in vivo which helps break tolerance to tumor antigens. These findings have important implications for the development of novel anti-cancer therapies aimed at promoting patients' immune responses to their own tumors.


Asunto(s)
Antígenos de Neoplasias/inmunología , Muerte Celular , Neoplasias Colorrectales/inmunología , Proteínas HSP70 de Choque Térmico/biosíntesis , Melanoma Experimental/inmunología , Animales , Antivirales/farmacología , Apoptosis , Ganciclovir/farmacología , Ratones , Neoplasias Experimentales/inmunología , Proteínas Proto-Oncogénicas c-bcl-2 , Simplexvirus/enzimología , Timidina Quinasa
9.
Gene Ther ; 17(8): 949-60, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20445580

RESUMEN

Viral therapy of cancer includes strategies such as viral transduction of tumour cells with 'suicide genes', using viral infection to trigger immune-mediated tumour cell death and using oncolytic viruses for their direct anti-tumour action. However, problems still remain in terms of adequate viral delivery to tumours. A role is also emerging for single-organ isolation and perfusion. Having begun with the advent of isolated limb perfusion for extremity malignancy, experimental systems have been developed for the perfusion of other organs, particularly the liver, kidneys and lungs. These are beginning to be adopted into clinical treatment pathways. The combination of these two modalities is potentially significant. Locoregional perfusion increases the exposure of tumour cells to viral agents. In addition, the avoidance of systemic elimination through the immune and reticulo-endothelial systems should provide a mechanism for increased transduction/infection of target cells. The translation of laboratory research to clinical practice would occur within the context of perfusion programmes, which are already established in the clinic. Many of these programmes include the use of vasoactive cytokines such as tumour necrosis factor-alpha, which may have an effect on viral uptake. Evidence of activation of specific anti-tumour immunological responses by intratumoural and other existing methods of viral administration raises the intriguing possibility of a locoregional therapy, with the ability to affect distant sites of disease. In this review, we examined the state of the literature in this area and summarized current findings before indicating likely areas of continuing interest.


Asunto(s)
Terapia Genética/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Animales , Humanos , Neoplasias/genética , Neoplasias/virología , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Ratas
10.
Gene Ther ; 17(2): 158-70, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20016540

RESUMEN

A fully intact immune system would be expected to hinder the efficacy of oncolytic virotherapy by inhibiting viral replication. Simultaneously, however, it may also enhance antitumor therapy through initiation of proinflammatory, antiviral cytokine responses at the tumor site. The aim of this study was to investigate the role of a fully intact immune system on the antitumor efficacy of an oncolytic virus. In this respect, injection of oncolytic vesicular stomatitis virus (VSV) into subcutaneous B16ova melanomas in C57Bl/6 mice leads to tumor regression, but it is not associated with viral replicative burst in the tumor. In contrast, intratumoral delivery of VSV induces an acute proinflammatory reaction, which quickly resolves concomitantly with virus clearance. Consistent with the hypothesis that therapy may not be dependent on the ability of VSV to undergo progressive rounds of replication, a single-cycle VSV is equally effective as a fully replication-competent VSV, whereas inactivated viruses do not generate therapy. Even though therapy is dependent on host CD8+ and natural killer cells, these effects are not associated with interferon-gamma-dependent responses against either the virus or tumor. There is, however, a strong correlation between viral gene expression, induction of proinflammatory reaction in the tumor and in vivo therapy. Overall, our results suggest that acute innate antiviral immune response, which rapidly clears VSV from B16ova tumors, is associated with the therapy observed in this model. Therefore, the antiviral immune response to an oncolytic virus mediates an intricate balance between safety, restriction of oncolysis and, potentially, significant immune-mediated antitumor therapy.


Asunto(s)
Melanoma Experimental/genética , Melanoma Experimental/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Vesiculovirus/genética , Animales , Anticuerpos Antivirales/biosíntesis , Genes Virales , Inmunidad Innata , Inflamación/virología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Replicación Viral
11.
Gene Ther ; 17(8): 1000-10, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20410926

RESUMEN

We have assessed the ability of bispecific fusion proteins to improve adenovirus-mediated transfer of therapeutic and marker transgenes. We constructed an expression vector that can be easily modified to synthesize a variety of fusion proteins for retargeting adenoviral gene therapy vectors to cell surface markers, which are differentially expressed between normal and cancer cells. Adenoviral transduction can be improved in a number of tumour cell lines which overexpress EGFR (epidermal growth factor receptor) or uPAR (urokinase-type plasminogen activator receptor), but which have only low levels of endogenous hCAR (human coxsackie B and adenovirus receptor) expression. Up to 40-fold improvement in beta-galactosidase transgene expression was seen using an EGFR retargeting protein, and up to 16-fold using a second fusion protein targeting uPAR. In vitro, our uPAR retargeting fusion protein improved the sensitivity to adenoviral herpes simplex virus thymidine kinase/ganciclovir by an order of magnitude, whereas in vivo, our EGFR retargeting protein is able to significantly delay tumour growth in rodent animal models in a dose-dependent manner. The 'cassette' design of our fusion protein constructs offers a flexible method for the straightforward synthesis of multiple adenoviral retargeting proteins, directed against a variety of tumour-associated antigens, for use in clinical trials.


Asunto(s)
Adenoviridae/genética , Receptores ErbB/genética , Terapia Genética/métodos , Neoplasias/terapia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Antivirales/farmacología , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Resistencia a Antineoplásicos/genética , Receptores ErbB/metabolismo , Ganciclovir/farmacología , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Proteínas de la Membrana/genética , Ingeniería de Proteínas , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Proteínas Recombinantes de Fusión/análisis , Transducción Genética
12.
Gene Ther ; 16(5): 689-99, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19282847

RESUMEN

Reovirus is a naturally occurring oncolytic virus currently in early clinical trials. However, the rapid induction of neutralizing antibodies represents a major obstacle to successful systemic delivery. This study addresses, for the first time, the ability of cellular carriers in the form of T cells and dendritic cells (DC) to protect reovirus from systemic neutralization. In addition, the ability of these cellular carriers to manipulate the subsequent balance of anti-viral versus anti-tumour immune response is explored. Reovirus, either neat or loaded onto DC or T cells, was delivered intravenously into reovirus-naive or reovirus-immune C57Bl/6 mice bearing lymph node B16tk melanoma metastases. Three and 10 days after treatment, reovirus delivery, carrier cell trafficking, metastatic clearance and priming of anti-tumour/anti-viral immunity were assessed. In naive mice, reovirus delivered either neat or through cell carriage was detectable in the tumour-draining lymph nodes 3 days after treatment, though complete clearance of metastases was only obtained when the virus was delivered on T cells or mature DC (mDC); neat reovirus or loaded immature DC (iDC) gave only partial early tumour clearance. Furthermore, only T cells carrying reovirus generated anti-tumour immune responses and long-term tumour clearance; reovirus-loaded DC, in contrast, generated only an anti-viral immune response. In reovirus-immune mice, however, the results were different. Neat reovirus was completely ineffective as a therapy, whereas mDC--though not iDC--as well as T cells, effectively delivered reovirus to melanoma in vivo for therapy and anti-tumour immune priming. Moreover, mDC were more effective than T cells over a range of viral loads. These data show that systemically administered neat reovirus is not optimal for therapy, and that DC may be an appropriate vehicle for carriage of significant levels of reovirus to tumours. The pre-existing immune status against the virus is critical in determining the balance between anti-viral and anti-tumour immunity elicited when reovirus is delivered by cell carriage, and the viral dose and mode of delivery, as well as the immune status of patients, may profoundly affect the success of any clinical anti-tumour viral therapy. These findings are therefore of direct translational relevance for the future design of clinical trials.


Asunto(s)
Células Dendríticas/trasplante , Melanoma Experimental/secundario , Melanoma Experimental/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Linfocitos T/trasplante , Inmunidad Adaptativa , Animales , Muerte Celular , Citotoxicidad Inmunológica , Ganglios Linfáticos/virología , Metástasis Linfática , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Reoviridae/inmunología , Reoviridae/aislamiento & purificación , Resultado del Tratamiento , Células Tumorales Cultivadas , Carga Viral
13.
Gene Ther ; 16(1): 78-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18784747

RESUMEN

Transfer of healthy autologous tissue as a microvascular free flap facilitates reconstruction during ablative cancer surgery. In addition to filling surgical defects, free flaps might concentrate viral vectors at the tumour bed and mediate local therapeutic effects. We evaluated the magnitude, topography and duration of luciferase gene expression after plasmid and adenoviral delivery in rat superficial inferior epigastric (SIE) flaps. For plasmid delivery, luciferase expression was significantly increased by all transduction routes (topical, intraflap injection, intravascular) (P<0.01) at day 1, but not at day 7. The spread of luciferase expression was significantly different between the 4 groups at 1 day (P=0.026) and was greatest for flaps transduced by intravascular injection. For adenoviral transduction, total radiance was significantly different between the transduced groups at 1, 14 and 28 days (P<0.05 for all comparisons). The highest levels of radiance were seen in the intravascular group. There was a statistically significant difference in the spread of light emission between the 3 groups at 1 (P=0.009) and 14 (P=0.013) days, but this was no longer evident at 28 days. Intravascular adenoviral delivery yields high-level, diffuse and durable gene expression in rat SIE flaps and is suitable for examination in therapeutic models.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Plásmidos/farmacología , Colgajos Quirúrgicos , Animales , Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Inyecciones , Operón Lac , Luciferasas/análisis , Luciferasas/genética , Masculino , Modelos Animales , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Transducción Genética/métodos
14.
G Ital Dermatol Venereol ; 144(1): 1-26, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19218908

RESUMEN

Metastatic malignant melanoma is an incurable malignancy with extremely poor prognosis. Patients bearing this diagnosis face a median survival time of approximately 9 months with a probability of surviving 5 years after initial presentation at less than 5%. This is contrasted by the curative nature of surgical resection of early melanoma detected in the skin. To date, no systemic therapy has consistently and predictably impacted the overall survival of patients with metastatic melanoma. However, in recent years, a resurgence of innovative diagnostic and therapeutic developments have broadened our understanding of the natural history of melanoma and identified rational therapeutic targets/strategies that seem poised to significantly change the clinical outcomes in these patients. Herein we review the state-of-the-art in metastatic melanoma diagnostics and therapeutics with particular emphasis on multi-disciplinary clinical management.


Asunto(s)
Melanoma/secundario , Melanoma/terapia , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante , Diagnóstico Diferencial , Medicina Basada en la Evidencia , Fluorodesoxiglucosa F18 , Humanos , Inmunoterapia , Imagen por Resonancia Magnética , Melanoma/diagnóstico , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Melanoma/radioterapia , Melanoma/cirugía , Tomografía de Emisión de Positrones , Pronóstico , Radioterapia Adyuvante , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/radioterapia , Neoplasias Cutáneas/cirugía , Análisis de Supervivencia , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
15.
Gene Ther ; 15(17): 1240-5, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18509378

RESUMEN

Fusogenic membrane glycoproteins (FMGs) may enhance the cytotoxicity of conditionally replicative adenoviruses. However, expression at early stages of infection impairs virus replication. We have inserted the hyperfusogenic form of the gibbon ape leukemia virus (GALV) envelope glycoprotein as a new splice unit of the major late promoter (MLP) to generate a replication-competent adenovirus expressing this protein. At high multiplicity of infection (MOI), this virus replicated efficiently forming clumps of fused cells and showing a faster release. In contrast, at low MOI, infected cells formed syncytia where only one nucleus contained virus DNA, decreasing total virus production but increasing cytotoxicity.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Células Gigantes/fisiología , Virus de la Leucemia del Gibón/fisiología , Viroterapia Oncolítica/métodos , Proteínas Virales de Fusión/genética , Línea Celular Tumoral , Expresión Génica , Ingeniería Genética , Humanos , Virus de la Leucemia del Gibón/genética , Regiones Promotoras Genéticas , Transgenes , Replicación Viral
16.
Gene Ther ; 15(6): 424-33, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18079753

RESUMEN

Escherichia coli nitroreductase (NTR) converts the prodrug CB1954 (5-(aziridin-1-yl)-2,4-dinitrobenzamide) into a bifunctional alkylating agent that causes DNA crosslinks. In this study, the ability of NTR to enhance the combined effects of CB1954 and radiation has been tested in vitro and in vivo. Stably transduced ovarian cancer cells (SKOV3-NTR) that are sensitive to CB1954 (IC(50)=0.35 muM) demonstrated enhanced cytotoxicity when treated with CB1954 and single-fraction irradiation. The NTR-CB1954 system mediated a bystander effect in combination with radiation on transfer of conditioned medium from SKOV3-NTR, but not SKOV3, cells to SW480 target cells. The ability of CB1954 to enhance radiation-induced cytotoxicity in SKOV3-NTR (but not SKOV3) cells was also demonstrated by fluorescence-activated cell sorting (FACS) with dual staining for propidium iodide/fluorescein diacetate, 4',6-diamidino-2-phenylindole dichloride staining of apoptotic cells and measurement of double-stranded DNA breaks by FACS and confocal microscopy for gammaH2AX foci. Adenoviral delivery of NTR, under constitutive cytomegalovirus or tissue-specific CTP1 promoters, increased the in vitro cytotoxicity of CB1954 plus radiation in MTT and clonogenic assays. Finally, adenoviral delivery of NTR plus CB1954 enhanced the effect of fractionated radiotherapy (12 Gy in four fractions) in SW480 xenograft tumours in nude mice.


Asunto(s)
Aziridinas/administración & dosificación , Escherichia coli/enzimología , Terapia Genética/métodos , Nitrorreductasas/genética , Neoplasias Ováricas/radioterapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Apoptosis , Línea Celular Tumoral , Terapia Combinada , Citomegalovirus/genética , Fragmentación del ADN , Femenino , Citometría de Flujo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Transducción Genética/métodos
17.
Gene Ther ; 15(8): 604-16, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18305577

RESUMEN

Although adoptive T-cell therapy has shown clinical success, efficacy is limited by low levels of T-cell trafficking to, and survival in, the immunosuppressive environment of an established tumor. Oncolytic virotherapy has recently emerged as a promising approach to induce both direct tumor cell killing and local proinflammatory environments within tumors. However, inefficient systemic delivery of oncolytic viruses remains a barrier to use of these agents against metastatic disease that is not directly accessible to the end of a needle. Here we show that the ability of antigen-specific T cells to circulate freely, and to localize to tumors, can be exploited to achieve the systemic delivery of replication-competent, oncolytic vesicular stomatitis virus (VSV). Thus, VSV loaded onto OT-I T cells, specific for the SIINFEKL epitope of the ovalbumin antigen, was efficiently delivered to established B16ova tumors in the lungs of fully immune-competent C57Bl/6 mice leading to significant increases in therapy compared to the use of virus, or T cells, alone. Although OT-I T-cell-mediated delivery of VSV led to viral replication within tumors and direct viral oncolysis, therapy was also dependent upon an intact host immune system. Moreover, VSV loading onto the T cells increased both T-cell activation in vitro and T-cell trafficking in vivo. The combination of adoptive T-cell transfer of antigen-specific T cells, along with oncolytic virotherapy, can, therefore, increase the therapeutic utility of both approaches through multiple mechanisms and should be of direct translational value.


Asunto(s)
Traslado Adoptivo/métodos , Antígenos de Neoplasias/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Vesiculovirus/genética , Animales , Movimiento Celular , Terapia Combinada , Terapia Genética/métodos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/virología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Metástasis de la Neoplasia/terapia , Trasplante de Neoplasias , Replicación Viral
18.
Gene Ther ; 15(18): 1257-70, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18401435

RESUMEN

Reovirus is a promising unmodified double-stranded RNA (dsRNA) anti-cancer oncolytic virus, which is thought to specifically target cells with activated Ras. Although reovirus has been tested in a wide range of preclinical models and has entered early clinical trials, it has not previously been tested for the treatment of human melanoma. Here, we show that reovirus effectively kills and replicates in both human melanoma cell lines and freshly resected tumour; intratumoural injection also causes regression of melanoma in a xenograft in vivo model. Reovirus-induced melanoma death is blocked by caspase inhibition and is dependent on constituents of the Ras/RalGEF/p38 pathway. Reovirus melanoma killing is more potent than, and distinct from, chemotherapy or radiotherapy-induced cell death; a range of inflammatory cytokines and chemokines are released by infected tumour cells, while IL-10 secretion is abrogated. Furthermore, the inflammatory response generated by reovirus-infected tumour cells causes bystander toxicity against reovirus-resistant tumour cells and activates human myeloid dendritic cells (DC) in vitro. Hence, reovirus is suitable for clinical testing in melanoma, and may provide a useful danger signal to reverse the immunologically suppressive environment characteristic of this tumour.


Asunto(s)
Melanoma/terapia , Viroterapia Oncolítica/métodos , Reoviridae/fisiología , Neoplasias Cutáneas/terapia , Animales , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Cromonas/farmacología , Citocinas/inmunología , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/inmunología , Citometría de Flujo , Humanos , Imidazoles/farmacología , Melanoma/inmunología , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piridinas/farmacología , Transducción de Señal/fisiología , Neoplasias Cutáneas/inmunología , Células Tumorales Cultivadas , Replicación Viral , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas ras/metabolismo
19.
Gene Ther ; 15(12): 911-20, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18323793

RESUMEN

There is an emerging realization from animal models that the immune response may have both detrimental and beneficial therapeutic effects during cancer virotherapy. However, there is a dearth of clinical data on the immune response to viral agents in patients. During a recently completed phase I trial of intravenous reovirus type 3 Dearing (RT3D), heavily pretreated patients with advanced cancers received RT3D at doses escalating from 1 x 10(8) tissue culture infectious dose-50 (TCID(50)) on day 1 to 3 x 10(10) TCID(50) on 5 consecutive days of a 4 weekly cycle. A detailed analysis of the immune effects was conducted by collecting serial clinical samples for analysis of neutralizing anti-reoviral antibodies (NARA), peripheral blood mononuclear cells (PBMC) and cytokines. Significant increases in NARA were seen with peak endpoint titres >1/10 000 in all but one patient. The median fold increase was 250, with a range of 9-6437. PBMC subset analysis showed marked heterogeneity. At baseline, CD3+CD4+ T cells were reduced in most patients, but after RT3D therapy their numbers increased in 47.6% of patients. In contrast, most patients had high baseline CD3+CD8+ T-cell levels, with 33% showing incremental increases after therapy. In some patients, there was increased cytotoxic T-cell activation post-therapy, as shown by increased CD8+perforin/granzyme+ T-cell numbers. Most patients had high numbers of circulating CD3-CD56+ NK cells before therapy and in 28.6% this increased with treatment. Regulatory (CD3+CD4+CD25+) T cells were largely unaffected by the therapy. Combined Th1 and Th2 cytokine expression increased in 38% of patients. These data confirm that even heavily pretreated patients are capable of mounting dynamic immune responses during treatment with RT3D, although these responses are not clearly related to the administered virus dose. These data will provide the basis for future studies aiming to modulate the immune response during virotherapy.


Asunto(s)
Terapia Genética/métodos , Orthoreovirus Mamífero 3/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Citocinas/sangre , Femenino , Humanos , Inmunidad Innata , Inyecciones Intravenosas , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Subgrupos de Linfocitos T/inmunología , Resultado del Tratamiento
20.
Curr Biol ; 5(1): 10-3, 1995 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-7535170

RESUMEN

Angiostatin, a recently discovered anti-angiogenic factor, offers the hope of long-term control of metastatic cancers following surgery.


Asunto(s)
Metástasis de la Neoplasia/prevención & control , Neovascularización Patológica/prevención & control , Fragmentos de Péptidos/uso terapéutico , Plasminógeno/uso terapéutico , Angiostatinas , Animales , Humanos , Metástasis de la Neoplasia/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA