Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Cancer Res ; 53(17): 3860-4, 1993 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8395331

RESUMEN

We report here the use of the 5' flanking region of the murine tyrosinase gene to direct expression of the herpes simplex virus thymidine kinase (tk) gene specifically to murine melanoma cells, whilst not permitting expression in a range of other cell types. Expression of the herpes simplex virus tk gene from the tyrosinase promoter in melanoma cells rendered them sensitive to killing by ganciclovir (100% cell death of a tk-expressing B16 clone after 12 days in culture at 1 microgram/ml ganciclovir). We also observed a substantial bystander killing effect when expressing cells were mixed with nontransfected parental B16 cells. When transfected murine melanoma cells expressing tk were injected into syngeneic mice both their tumorigenicity and experimental metastatic potential were abrogated completely when the mice were treated with ganciclovir (27 of 28 mice treated with water developed progressively growing tumors versus 1 of 30 in the ganciclovir-treated group). Direct injection of the tk gene under control of the tyrosinase promoter into established tumors in mice, followed by treatment with ganciclovir, led to significant reductions in resultant tumor size relative to the size of tumor developing in mice treated with water (median tumor weight, 1.65 g versus 2.75 g). Therefore, direct transfer of recombinant genes by injection of DNA can significantly reduce established tumor burden in vivo.


Asunto(s)
ADN/administración & dosificación , Ganciclovir/farmacología , Regulación Enzimológica de la Expresión Génica , Melanoma Experimental/terapia , Monofenol Monooxigenasa/genética , Simplexvirus/enzimología , Animales , Medios de Cultivo , Esquema de Medicación , Resistencia a Medicamentos , Ganciclovir/administración & dosificación , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Inyecciones Intralesiones , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Melanoma Experimental/prevención & control , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Puromicina , Transfección , Células Tumorales Cultivadas
13.
Cancer Res ; 53(5): 962-7, 1993 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8439971

RESUMEN

Gene therapy protocols for cancer usually involve removal of tumor cells, culture in vitro to allow gene transfer, and subsequent reintroduction in vivo. Targeting therapeutic genes to tumor cells in situ requires an accuracy of gene delivery that currently is not possible with the use of existing techniques. To overcome these limitations we have used two promoters, which are preferentially active in melanocytic cells, to direct gene expression specifically to melanoma cells both in vitro and in vivo. Here we describe experiments showing that as little as 769 base pairs of the 5'-flanking regions of the tyrosinase, and 1.4 kilobase pair of the tyrosinase-related protein 1, genes are sufficient to direct expression of the beta-galactosidase gene to both human and murine melanoma cells and melanocytes, while not permitting expression in a range of other cell types in vitro. These promoters showed high levels of activity in 12 of 14 murine and human melanoma cell lines tested but showed only basal levels of activity, similar to that of a promoterless construct, in a range of 12 other cell types. Cell type specificity is maintained when the construct is delivered to cells either by physical means or by inclusion of the cell type-specific expression cassette into a retroviral vector. Direct injection of DNA, encoding the beta-galactosidase gene expressed from either promoter, into established B16 melanomas or Colo 26 tumors in syngeneic mice resulted in extensive transduction of tumor cells in the B16 melanomas (approximately 10% of tumor cells expressing 10 days after DNA injection), whereas no blue-staining cells were seen in the Colo 26 tumors. The reporter gene was expressed in melanoma cells and in some normal melanocytes but not in other surrounding normal tissue. We propose that the combination of a tissue-specific promoter driving a therapeutic gene, with delivery of such a construct directly to sites of tumor growth in vivo, either by direct DNA injection or by retroviral infection, may provide significantly enhanced safety for gene therapy for solid tumors.


Asunto(s)
Terapia Genética/métodos , Melanoma/terapia , Glicoproteínas de Membrana , Oxidorreductasas , Animales , Cricetinae , Expresión Génica , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monofenol Monooxigenasa/genética , Regiones Promotoras Genéticas , Proteínas/genética , Ratas , Retroviridae/genética , Transfección , beta-Galactosidasa/genética
14.
Cancer Res ; 54(23): 6228-34, 1994 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7954471

RESUMEN

Previously we have demonstrated safe and effective transfer of the HSVtk cytotoxic gene to primary murine melanoma tumors by direct injection of plasmid and retroviral vectors in which the HSVtk gene is driven by the tissue-specific tyrosinase promoter. However, for general clinical application such forms of therapy should, ideally, be effective against disseminated metastases. We report here that the number of recently established lung metastases of B16 melanoma in C57BL mice treated with ganciclovir is reduced compared to controls after multiple i.v. administrations of high titer retroviral supernatant encoding the HSVtk gene, but not after administration of liposome-complexed plasmid DNA. Using polymerase chain reaction analysis, integration of the provirus was observed in metastasis-bearing lungs (4 of 6 mice) and in the spleens of some ganciclovir-treated animals (2 of 6 mice) but not in the testes, brain, heart, liver, or kidney. The reduction in the number of experimental metastases in C57BL mice exceeded the anticipated extent of transduction of tumor cells, which is indicative of a marked bystander effect. This magnitude of reduction was not observed in immunodeficient athymic mice, suggesting that the immune system plays some part in the bystander effect. In support of these data, we show that, whereas the parental tumor cells are only poorly immunogenic, an effective antitumor immune response is generated following the killing of neoplastic cells in vivo as a result of treatment with ganciclovir. These effects may be responsible for augmenting the efficacy of retroviral infection. The combination of local cell killing by the HSVtk/ganciclovir system and the induction of antitumor immunity suggests new opportunities for the design of vectors for the gene therapy of cancer.


Asunto(s)
Terapia Genética , Melanoma Experimental/terapia , Simplexvirus/genética , Timidina Quinasa/genética , Animales , Células Cultivadas , Ganciclovir/uso terapéutico , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Monofenol Monooxigenasa/genética , Especificidad de Órganos , Retroviridae/genética , Timidina Quinasa/biosíntesis
15.
Cancer Res ; 60(22): 6396-402, 2000 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11103804

RESUMEN

Viral fusogenic membrane glycoproteins (FMGs) are candidates for gene therapy of solid tumors because they cause cell fusion, leading to formation of lethal multinucleated syncytia. However, the cellular mechanisms mediating cell death after FMG-induced cell fusion remain unclear. The present study was designed to examine the mechanisms by which FMG expression in hepatocellular carcinoma cells lead to cell death. Transfection of Hep3B cells with the Gibbon Ape leukemia virus hyperfusogenic envelope protein (GALV-FMG) resulted in the formation of multinucleated syncytia that reached a maximum 5 days after transfection (100 nuclei/syncytia). The syncytia were viable for a period of 2 days and then rapidly lost viability by day 5. Mitochondrial dysfunction occurred in GALV-FMG-induced syncytia prior to loss of viability with loss of the mitochondrial membrane potential, cellular ATP depletion, and release of mitochondrial cytochrome c-GFP into the cytosol. The pan-caspase inhibitor, Z-VAD-fmk, did not prevent cell death. However, glycolytic generation of ATP with fructose effectively increased cellular ATP and preserved syncytial viability. These data suggest that expression of FMG in hepatoma cells results in the formation of multinucleated syncytia, causing mitochondrial failure with ATP depletion, a bioenergetic form of cell death with necrosis. This form of cell death should be effective in vivo and enhance the bystander effect, suggesting that FMG-based gene therapy deserves further study for the treatment of hepatocellular and other cancers.


Asunto(s)
Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Células Gigantes/patología , Virus de la Leucemia del Gibón/genética , Neoplasias Hepáticas/terapia , Proteínas Virales de Fusión/genética , Adenosina Trifosfato/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Inhibidores de Caspasas , Caspasas/metabolismo , Muerte Celular/fisiología , Fusión Celular/métodos , Metabolismo Energético/fisiología , Fructosa/farmacología , Células Gigantes/metabolismo , Células Gigantes/virología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/fisiología , Necrosis , Fosforilación Oxidativa , Células Tumorales Cultivadas , Proteínas Virales de Fusión/biosíntesis
16.
Cancer Res ; 59(12): 2802-5, 1999 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10383135

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells that are capable of priming systemic antitumor immune responses in animal tumor models. However, many of the model tumor systems tested need definition of the specific tumor antigens involved. To use DCs in situations that are more relevant to the majority of human cancers, where the antigens are unknown, we have tested the adoptive transfer of immature DCs in mouse colorectal and melanoma models of varying immunogenicity but with undefined antigens. When DCs admixed with a syngeneic primary tumor inoculum were seeded s.c., the growth of the primary tumor was unchanged; however, if the primary tumor was then surgically excised and the animal was rechallenged with the same tumor, significant protection (75%) was generated when DCs were present in the original primary inoculum of a moderately immunogenic colorectal model (CMT93tk). This effect was not observed when a nonimmunogenic melanoma (B16) was tested in an identical protocol. Next, DCs were injected directly into 6-9-mm established tumors; again, protection (55%) was achieved against a secondary tumor challenge following excision of the primary, but only in the CMT93tk model of moderate immunogenicity. To increase the clinical relevance of this approach still further, we tested irradiated allogeneic K1735 melanoma cells mixed with syngeneic DCs as a vaccine against subsequent challenge with the poorly immunogenic syngeneic melanoma B16. The allogeneic vaccine alone was ineffective, but when admixed with DCs, a significant number of animals rejected a subsequent B16 challenge, suggesting that DCs are able to prime an immune response against melanoma antigens shared between K1735 and B16. The generation of systemic antitumor immunity by adoptive transfer of DCs has significant clinical potential because it is technically straightforward and does not require the definition of specific tumor antigens.


Asunto(s)
Células Dendríticas/trasplante , Inmunoterapia Adoptiva , Melanoma Experimental/terapia , Animales , Vacunas contra el Cáncer/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Inmunidad , Inmunoterapia , Isoantígenos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Trasplante Autólogo
17.
Cancer Res ; 61(19): 7240-7, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11585761

RESUMEN

The mechanisms by which the immune system distinguishes normal developmental cell death from pathological immunogenic cell killing are central to effective cancer immunotherapy. Using HSVtk suicide gene therapy, we showed that macrophages can distinguish between tumor cells dying through classical apoptosis and tumor cells engineered to die through nonapoptotic mechanisms, resulting in secretion of either immunosuppressive cytokines (interleukin 10 and transforming growth factor beta) or inflammatory cytokines (tumor necrosis factor alpha or interleukin 1beta), respectively. Additionally heat shock protein 70 acts as one component of a bimodal alarm signal that activates macrophages in the presence of stressful, immunogenic tumor cell killing. These differential responses of macrophages can also be used to vaccinate mice against tumor challenge, using adoptive transfer, as well as to cure mice of established tumors.


Asunto(s)
Apoptosis/inmunología , Macrófagos/inmunología , Neoplasias Experimentales/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Muerte Celular/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Citocinas/inmunología , Citocinas/metabolismo , Ganciclovir/farmacología , Terapia Genética , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/farmacología , Inmunoterapia Adoptiva , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Transfección
18.
Cancer Res ; 60(6): 1492-7, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10749110

RESUMEN

We report here the use of viral fusogenic membrane glycoproteins (FMGs) as a new class of therapeutic genes for the control of tumor growth. FMGs kill cells by fusing them into large multinucleated syncytia, which die by sequestration of cell nuclei and subsequent nuclear fusion by a mechanism that is nonapoptotic, as assessed by multiple criteria. Direct and bystander killing of three different FMGs were at least one log more potent than that of herpes simplex virus thymidine kinase or cytosine deaminase suicide genes. Transduction of human tumor xenografts with plasmid DNA prevented tumor outgrowth in vivo, and cytotoxicity could be regulated through transcriptional targeting. Syncytial formation is accompanied by the induction of immunostimulatory heat shock proteins, and tumor-associated FMG expression in immunocompetent animals generated specific antitumor immunity.


Asunto(s)
Fusión Celular/genética , Terapia Genética/métodos , Neoplasias Experimentales/terapia , Proteínas Virales de Fusión/genética , Animales , Apoptosis/genética , División Celular/genética , Fusión Celular/inmunología , Línea Celular , ADN Complementario/genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Plásmidos/genética , Simplexvirus/enzimología , Timidina Quinasa/genética , Transfección , Células Tumorales Cultivadas , Proteínas Virales de Fusión/inmunología
19.
Clin Cancer Res ; 7(2): 243-54, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11234875

RESUMEN

The biodistribution and pharmacokinetics of (111)In-DTPA-labeled pegylated liposomes (IDLPL) were studied in 17 patients with locally advanced cancers. The patients received 65-107 MBq of IDLPL, and nuclear medicine whole body gamma camera imaging was used to study liposome biodistribution. The t(1/2beta) of IDLPL was 76.1 h. Positive tumor images were obtained in 15 of 17 studies (4 of 5 breast, 5 of 5 head and neck, 3 of 4 bronchus, 2 of 2 glioma, and 1 of 1 cervix cancer). The levels of tumor liposome uptake estimated from regions of interest on gamma camera images were approximately 0.5-3.5% of the injected dose at 72 h. The greatest levels of uptake were seen in the patients with head and neck cancers [33.0 +/- 15.8% ID/kg (percentage of injected dose/kg)]. The uptake in the lung tumors was at an intermediate level (18.3 +/- 5.7% ID/kg), and the breast cancers showed relatively low levels of uptake (5.3 +/- 2.6% ID/kg). These liposome uptake values mirrored the estimated tumor volumes of the various tumor types (36.2 +/- 18.0 cm3 for squamous cell cancer of the head and neck, 114.5 +/- 42.0 cm3 for lung tumors, and 234.7 +/- 101.4 cm3 for breast tumors). In addition, significant localization of the liposomes was seen in the tissues of the reticuloendothelial system (liver, spleen, and bone marrow). One patient with extensive mucocutaneous AIDS-related Kaposi sarcoma was also studied according to a modified protocol, and prominent deposition of the radiolabeled liposomes was demonstrated in these lesions. An additional two patients with resectable head and neck cancer received 26 MBq of IDLPL 48 h before undergoing surgical excision of their tumors. Samples of the tumor, adjacent normal mucosa, muscle, fat, skin, and salivary tissue were obtained at operation. The levels of tumor uptake were 8.8 and 15.9% ID/kg, respectively, with tumor uptake exceeding that in normal mucosa by a mean ratio of 2.3:1, in skin by 3.6:1, in salivary gland by 5.6:1, in muscle by 8.3:1, and in fat by 10.8:1. These data strongly support the development of pegylated liposomal agents for the treatment of solid tumors, particularly those of the head and neck.


Asunto(s)
Neoplasias/metabolismo , Ácido Pentético/farmacocinética , Radiofármacos/farmacocinética , Adulto , Anciano , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Femenino , Humanos , Radioisótopos de Indio/farmacocinética , Liposomas , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico por imagen , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Orina/química
20.
Clin Cancer Res ; 6(12): 4939-49, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11156255

RESUMEN

Concomitant chemotherapy and radiotherapy (CCRT) has recently been shown to improve treatment outcome in a range of solid tumors. Pegylated liposomes have the potential to target drugs directly to tumors and may increase the efficacy and reduce the toxicity of CCRT by selectively delivering radiosensitizing agents to tumor, as opposed to normal, tissues. In these studies, we have assessed CCRT using pegylated liposome encapsulated doxorubicin (PLED) and pegylated liposome encapsulated cisplatin (PLEC) against KB head and neck cancer xenograft tumors in nude mice. The addition of low-dose (2 mg/kg) PLED (P < 0.001) and PLEC (P < 0.001) significantly increased the effect of 4.5 Gy, but not 9 Gy, single-fraction radiotherapy (SFRT). Both PLED and PLEC were significantly more effective than their unencapsulated counterparts in increasing the effect of SFRT. In addition, PLED (P < 0.001) and PLEC (P < 0.05) significantly increased the effect of fractionated radiotherapy (9 Gy in 3 fractions) in two different dosing schedules (2 mg/kg single dose or three sequential doses of 0.67 mg/kg). Unencapsulated diethylenetriaminepentaacetic acid and pegylated liposomal diethylenetriaminepentaacetic acid were used as controls to test the effect of the liposome vehicle and showed no interaction with 4.5 Gy or 9 Gy SFRT (P > 0.1). CCRT was well-tolerated, with no evidence of increased local or systemic toxicity, as compared with radiotherapy alone. This study is the first to demonstrate the value of pegylated liposomes as vehicles for the delivery of radiosensitizing drugs in CCRT strategies.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Doxorrubicina/administración & dosificación , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Liposomas/química , Polietilenglicoles/química , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Terapia Combinada , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Doxorrubicina/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Factores de Tiempo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA