Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Lab Invest ; 93(3): 268-78, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23358109

RESUMEN

Metastatic involvement of the skeleton is a frequent consequence of advanced prostate cancer. These skeletal metastases cause a number of debilitating complications and are refractory to current treatments. New therapeutic options are being explored, including conditionally replicating adenoviruses (CRAds). CRAds are engineered to selectively replicate in and destroy tumor cells and can be 'armed' with exogenous transgenes for enhanced potency. We hypothesized that a CRAd armed with osteoprotegerin (OPG), an inhibitor of osteoclastogenesis, would inhibit the progression of prostate cancer bone metastases by directly lysing tumor cells and by reducing osteoclast activity. Although prostate cancer bone metastases are predominantly osteoblastic in nature, increased osteoclast activity is critical for the growth of these lesions. Ad5-Δ24-sOPG-Fc-RGD is a CRAd that carries a fusion of the ligand-binding domains of OPG and the Fc region of human IgG1 in place of the viral E3B genes. To circumvent low tumor cell expression of the native adenoviral receptor, an arginine-glycine-aspartic acid (RGD) peptide insertion within the viral fiber knob allows infection of cells expressing α(v) integrins. A 24-base pair deletion (Δ24) within viral E1A limits replication to cells with aberrant retinoblastoma cell cycle regulator/tumor suppressor expression. We have confirmed that Ad5-Δ24-sOPG-Fc-RGD replicates within and destroys prostate cancer cells and, in both murine and human coculture models, that infection of prostate cancer cells inhibits osteoclastogenesis in vitro. In a murine model, progression of advanced prostate cancer bone metastases was inhibited by treatment with Ad5-Δ24-sOPG-Fc-RGD but not by an unarmed control CRAd.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias Óseas/terapia , Viroterapia Oncolítica/métodos , Osteoprotegerina/farmacología , Neoplasias de la Próstata/patología , Adenoviridae/genética , Análisis de Varianza , Animales , Línea Celular Tumoral , Humanos , Inmunoglobulina G/genética , Luciferasas , Masculino , Ratones , Oligopéptidos/genética , Oligopéptidos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Microtomografía por Rayos X
2.
Mol Ther ; 20(12): 2222-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22948673

RESUMEN

Targeting the tumor stroma in addition to the malignant cell compartment is of paramount importance to achieve complete tumor regression. In this work, we modified a previously designed tumor stroma-targeted conditionally replicative adenovirus (CRAd) based on the SPARC promoter by introducing a mutated E1A unable to bind pRB and pseudotyped with a chimeric Ad5/3 fiber (Ad F512v1), and assessed its replication/lytic capacity in ovary cancer in vitro and in vivo. AdF512v1 was able to replicate in fresh samples obtained from patients: (i) with primary human ovary cancer; (ii) that underwent neoadjuvant treatment; (iii) with metastatic disease. In addition, we show that four intraperitoneal (i.p.) injections of 5 × 10(10) v.p. eliminated 50% of xenografted human ovary tumors disseminated in nude mice. Moreover, AdF512v1 replication in tumor models was enhanced 15-40-fold when the tumor contained a mix of malignant and SPARC-expressing stromal cells (fibroblasts and endothelial cells). Contrary to the wild-type virus, AdF512v1 was unable to replicate in normal human ovary samples while the wild-type virus can replicate. This study provides evidence on the lytic capacity of this CRAd and highlights the importance of targeting the stromal tissue in addition to the malignant cell compartment to achieve tumor regression.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Neoplasias Ováricas/genética , Células del Estroma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Semin Dial ; 24(6): 688-93, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21781173

RESUMEN

The underlying inflammatory component of chronic kidney disease may predispose blood vessels to intimal hyperplasia (IH), which is the primary cause of dialysis access failure. We hypothesize that vascular pathology and markers of IH formation are antecedent to arteriovenous (AV) fistula creation. Blood, cephalic, and basilic vein segments were collected from predialysis chronic kidney disease (CKD) patients with no previous AV access and patients with end-stage renal disease (ESRD). Immunohistochemistry was performed with antibodies against mast cell chymase, transforming growth factor-beta (TGF-ß) and interleukin-6 (IL-6), which cause IH. Plasma chymase was measured by ELISA. IH was present in 91% of CKD and 75% of ESRD vein segments. Chymase was abundant in vessels with IH, with the greatest expression in intima and medial layers, and virtually absent in the controls. Chymase colocalized with TGF-ß1 and IL-6. Plasma chymase concentration was elevated up to 33-fold in patients with CKD versus controls and was associated with increased chymase in vessels with IH. We show that chymase expression in vessels with IH corresponds with plasma chymase concentrations. As chymase inhibition attenuates IH in animal models, and we find chymase is highly expressed in IH lesions of patients with CKD and ESRD, we speculate that chymase inhibition could have therapeutic value in humans.


Asunto(s)
Quimasas/biosíntesis , Quimasas/sangre , Fallo Renal Crónico/sangre , Fallo Renal Crónico/metabolismo , Mastocitos/enzimología , Neointima/metabolismo , Venas/metabolismo , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
4.
J Thromb Haemost ; 19(7): 1653-1665, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33773040

RESUMEN

BACKGROUND: Elucidating the molecular pathogenesis underlying East Texas bleeding disorder (ET) led to the discovery of alternatively spliced F5 transcripts harboring large deletions within exon 13. These alternatively spliced transcripts produce a shortened form of coagulation factor V (FV) in which a large portion of its B-domain is deleted. These FV isoforms bind tissue factor pathway inhibitor alpha (TFPIα) with high affinity, prolonging its circulatory half-life and enhancing its anticoagulant effects. While two missense pathogenic variants highlighted this alternative splicing event, similar internally deleted FV proteins are found in healthy controls. OBJECTIVE: We identified a novel heterozygous 832 base pair deletion within F5 exon 13, termed F5-Atlanta (F5-ATL), in a patient with severe bleeding. Our objective is to investigate the effect of this deletion on F5 and FV expression. METHODS & RESULTS: Assessment of patient plasma revealed markedly elevated levels of total and free TFPI and a FV isoform similar in size to the FV-short described in ET. Sequencing analyses of cDNA revealed the presence of a transcript alternatively spliced using the ET splice sites, thereby removing the F5-ATL deletion. This alternative splicing pattern was recapitulated by heterologous expression in mammalian cells. CONCLUSIONS: These findings support a mechanistic model consisting of cis-acting regulatory sequences encoded within F5 exon 13 that control alternative splicing at the ET splice sites and thereby regulate circulating FV-short and TFPIα levels.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Factor V , Empalme Alternativo , Animales , Trastornos de la Coagulación Sanguínea/genética , Exones , Factor V/genética , Humanos , Mutación , Empalme del ARN
5.
J Spinal Disord Tech ; 23(1): 63-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20084034

RESUMEN

STUDY DESIGN: Ex vivo gene transfer for spinal fusion. OBJECTIVE: This study aimed to evaluate ex vivo transfer of the nuclear-localized Hoxc-8-interacting domain of Smad1 (termed Smad1C) to rabbit bone marrow stromal cells (BMSCs) by a tropism-modified human adenovirus serotype 5 (Ad5) vector as a novel therapeutic approach for spinal fusion. SUMMARY OF BACKGROUND DATA: Novel approaches are needed to improve the success of bone union after spinal fusion. One such approach is the ex vivo transfer of a gene encoding an osteoinductive factor to BMSCs which are subsequently reimplanted into the host. We have previously shown that heterologous expression of the Hoxc-8-interacting domain of Smad1 in the nuclei of osteoblast precursor cells is able to stimulate the expression of genes related to osteoblast differentiation and induce osteogenesis in vivo. Gene delivery vehicles based on human Ad5 are well suited for gene transfer for spinal fusion because they can mediate high-level, short-term gene expression. However, Ad5-based vectors with native tropism poorly transduce BMSCs, necessitating the use of vectors with modified tropism to achieve efficient gene transfer. METHODS: The gene encoding Smad1C was transferred to rabbit BMSCs by an Ad5 vector with native tropism or a vector retargeted to alphav integrins, which are abundantly expressed on rabbit BMSCs. Transduced BMSCs were maintained in osteoblastic differentiation medium for 30 days. Alkaline phosphatase activity was determined and cells stained for calcium deposition. As positive controls for osteogenesis, we used Ad5 vectors expressing bone morphogenetic protein 2. As negative controls, BMSCs were mock-transduced or transduced with an Ad5 vector expressing beta-galactosidase. In an immunocompetent rabbit model of spinal fusion, transduced BMSCs were coated onto absorbable gelatin sponge and implanted between decorticated transverse processes L6 and L7 of 8-week-old female New Zealand white rabbits. Animals were killed 4 weeks after implantation of the sponges, the fusion masses harvested and the area of new bone quantified using image analysis software. RESULTS: The Smad1C-expressing tropism-modified Ad5 vector mediated a significantly higher level of alkaline phosphatase activity and calcium deposition in transduced rabbit BMSCs than all other vectors. The rabbit BMSCs transduced ex vivo with the Smad1C-expressing tropism-modified Ad5 vector mediated a greater amount of new bone formation than BMSCs transduced with any other vector. CONCLUSIONS: Delivery of the Smad1C gene construct to BMSCs by an alphav integrin-targeted Ad5 vector shows promise for spinal fusion and other applications requiring the formation of new bone in vivo.


Asunto(s)
Adenoviridae/genética , Trasplante de Médula Ósea/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteína Smad1/genética , Fusión Vertebral/métodos , Células del Estroma/trasplante , Animales , Regeneración Ósea/genética , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Integrina alfaV/genética , Estructura Terciaria de Proteína/genética , Conejos , Células del Estroma/citología , Células del Estroma/metabolismo , Resultado del Tratamiento
6.
Mol Imaging ; 8(5): 264-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19796604

RESUMEN

We sought to develop a cancer-targeted, infectivity-enhanced oncolytic adenovirus that embodies a capsid-labeling fusion for noninvasive dual-modality imaging of ovarian cancer virotherapy. A functional fusion protein composed of fluorescent and nuclear imaging tags was genetically incorporated into the capsid of an infectivity-enhanced conditionally replicative adenovirus. Incorporation of herpes simplex virus thymidine kinase (HSV-tk) and monomeric red fluorescent protein 1 (mRFP1) into the viral capsid and its genomic stability were verified by molecular analyses. Replication and oncolysis were evaluated in ovarian cancer cells. Fusion functionality was confirmed by in vitro gamma camera and fluorescent microscopy imaging. Comparison of tk-mRFP virus to single-modality controls revealed similar replication efficiency and oncolytic potency. Molecular fusion did not abolish enzymatic activity of HSV-tk as the virus effectively phosphorylated thymidine both ex vivo and in vitro. In vitro fluorescence imaging demonstrated a strong correlation between the intensity of fluorescent signal and cytopathic effect in infected ovarian cancer cells, suggesting that fluorescence can be used to monitor viral replication. We have in vitro validated a new infectivity-enhanced oncolytic adenovirus with a dual-imaging modality-labeled capsid, optimized for ovarian cancer virotherapy. The new agent could provide incremental gains toward climbing the barriers for achieving conditionally replicated adenovirus efficacy in human trials.


Asunto(s)
Adenoviridae/metabolismo , Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Adenoviridae/genética , Cápside/metabolismo , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/fisiología , Microscopía Fluorescente , Simplexvirus/enzimología , Timidina Quinasa/genética , Timidina Quinasa/fisiología , Proteína Fluorescente Roja
7.
Stem Cells ; 26(3): 831-41, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18192232

RESUMEN

Gene therapy represents a promising treatment alternative for patients with malignant gliomas. Nevertheless, in the setting of these highly infiltrative tumors, transgene delivery remains a challenge. Indeed, viral vehicles tested in clinical trials often target only those tumor cells that are adjacent to the injection site. In this study, we examined the feasibility of using human mesenchymal stem cells (hMSC) to deliver a replication-competent oncolytic adenovirus (CRAd) in a model of intracranial malignant glioma. To do so, CRAds with a chimeric 5/3 fiber or RGD backbone with or without CXCR4 promoter driving E1A were examined with respect to replication and toxicity in hMSC, human astrocytes, and the human glioma cell line U87MG by quantitative polymerase chain reaction and membrane integrity assay. CRAd delivery by virus-loaded hMSC was then evaluated in vitro and in an in vivo model of mice bearing intracranial U87MG xenografts. Our results show that hMSC are effectively infected by CRAds that use the CXCR4 promoter. CRAd-CXCR4-RGD had the highest replication, followed by CRAd-CXCR4-5/3, in hMSC, with comparable levels of toxicity. In U87MG tumor cells, CRAd-CXCR4-5/3 showed the highest replication and toxicity. Virus-loaded hMSC effectively migrated in vitro and released CRAds that infected U87MG glioma cells. When injected away from the tumor site in vivo, hMSC migrated to the tumor and delivered 46-fold more viral copies than injection of CRAd-CXCR4-5/3 alone. Taken together, these results indicate that hMSC migrate and deliver CRAd to distant glioma cells. This delivery strategy should be explored further, as it could improve the outcome of oncolytic virotherapy for glioma.


Asunto(s)
Adenoviridae/fisiología , Neoplasias Encefálicas/terapia , Glioma/terapia , Células Madre Mesenquimatosas/citología , Viroterapia Oncolítica , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Regulación de la Expresión Génica , Glioma/patología , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Regiones Promotoras Genéticas/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción Genética , Replicación Viral
8.
Exp Dermatol ; 18(4): 362-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18803656

RESUMEN

The purpose of this study was to determine the effect of transplanted human mesenchymal stem cells (hMSCs) on wound healing. In this model, full-thickness cutaneous wounds were created by incision in the skin of adult New Zealand white rabbits and treated by transplanted hMSCs into the wounds. Wound healing was evaluated by histological analysis and tensiometry over time. A total of 15 New Zealand white rabbits with 10 wounds per animal were examined in this study. Animals were treated with hMSCs and euthanised at 3, 7, 14, 21 and 80 days after manipulation. The hMSCs were labelled with a fluorescent dye (CM-DiI), suspended in phosphate-buffered saline and used to treat full-thickness incisional wounds in rabbit skin. Tensiometry and histology were used to characterise the wound-healing rate of the incisional wounds. These results showed that transplanted hMSCs significantly inhibited scar formation and increased the tensile strength of the wounds. Importantly, MSCs from genetically unrelated donors did not appear to induce an immunologic response. In conclusion, human mesenchymal stem cell therapy is a viable approach to significantly affect the course of normal cutaneous wound healing and significantly increase the tensile strength.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Cicatriz/prevención & control , Humanos , Modelos Animales , Conejos , Piel/patología , Resistencia a la Tracción/fisiología , Factores de Tiempo , Trasplante Heterólogo
9.
J Med Virol ; 80(9): 1595-603, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18649343

RESUMEN

Malignant glioma, in particular glioblastoma multiforme (GBM), represents one of the most devastating cancers currently known and existing treatment regimens do little to change patient prognosis. Conditionally replicating adenoviral vectors (CRAds) represent attractive experimental anti-cancer agents with potential for clinical application. However, early protein products of the wild type adenovirus backbone--such as E1A--limit CRAds' replicative specificity. In this study, we evaluated the oncolytic potency and specificity of CRAds in which p300/CPB and/or pRb binding capacities of E1A were ablated to reduce non-specific replicative cytolysis. In vitro cytopathic assays, quantitative PCR analysis, Western blot, and flow cytometry studies demonstrate the superior anti-glioma efficacy of a double-mutated CRAd, Ad2/24CMV, which harbors mutations that reduce E1A binding to p300/CPB and pRb. When compared to its single-mutated and wild type counterparts, Ad2/24CMV demonstrated attenuated replication and cytotoxicity in representative normal human brain while displaying enhanced replicative cytotoxicity in malignant glioma. These results have implications for the development of double-mutated CRAd vectors for enhanced GBM therapy.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Glioma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Línea Celular , Humanos , Mutación , Replicación Viral
10.
Virol J ; 5: 98, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18718011

RESUMEN

Despite the many potential advantages of Ad vectors for vaccine application, the full utility of current Ad vaccines may be limited by the host anti-vector immune response. Direct incorporation of antigens into the adenovirus capsid offers a new and exciting approach for vaccination strategies; this strategy exploits the inherent antigenicity of the Ad vector. Critical to exploiting Ad in this new context is the placement of antigenic epitopes within the major Ad capsid protein, hexon. In our current study we illustrate that we have the capability to place a range of antigenic epitopes within Ad5 capsid protein hexon hypervariable regions (HVRs) 2 or 5, thus producing viable Ad virions. Our data define the maximal incorporation size at HVR2 or HVR5 as it relates to identical antigenic epitopes. In addition, this data suggests that Ad5 HVR5 is more permissive to a range of insertions. Most importantly, repeated administration of our hexon-modified viruses resulted in a secondary anti-antigen response, whereas minimal secondary effect was present after administration of Ad5 control. Our study describes antigen placement and optimization within the context of the capsid incorporation approach of Ad vaccine employment, thereby broadening this new methodology.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Antígenos/inmunología , Proteínas de la Cápside/genética , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Vacunas/administración & dosificación , Animales , Antígenos/genética , Proteínas de la Cápside/inmunología , Línea Celular , Epítopos/genética , Epítopos/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas/genética , Vacunas/inmunología
11.
J Mol Med (Berl) ; 85(5): 481-96, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17219096

RESUMEN

Fibromodulin, a member of the small leucine-rich proteoglycan family, has been recently suggested as a biologically significant mediator of fetal scarless repair. To assess the role of fibromodulin in the tissue remodeling, we constructed an adenoviral vector expressing human fibromodulin cDNA. We evaluated the effect of adenovirus-mediated overexpression of fibromodulin in vitro on transforming growth factors and metalloproteinases in fibroblasts and in vivo on full-thickness incisional wounds in a rabbit model. In vitro, we found that Ad-Fibromodulin induced a decrease of expression of TGF-beta(1) and TGF-beta(2) precursor proteins, but an increase in expression of TGF-beta(3) precursor protein and TGF-beta type II receptor. In addition, fibromodulin overexpression resulted in decreased MMP-1 and MMP-3 protein secretion but increased MMP-2, TIMP-1, and TIMP-2 secretion, whereas MMP-9 and MMP-13 were not influenced by fibromodulin overexpression. In vivo evaluation by histopathology and tensile strength demonstrated that Ad-Fibromodulin administration could ameliorate wound healing in incisional wounds. In conclusion, although the mechanism of scar formation in adult wounds remains incompletely understood, we found that fibromodulin overexpression improves wound healing in vivo, suggesting that fibromodulin may be a key mediator in reduced scarring.


Asunto(s)
Adenoviridae/genética , Cicatriz/prevención & control , Dermis/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Fibroblastos/metabolismo , Terapia Genética/métodos , Vectores Genéticos , Proteoglicanos/biosíntesis , Cicatrización de Heridas , Animales , Células Cultivadas , Cicatriz/genética , Cicatriz/metabolismo , Cicatriz/patología , Cicatriz/fisiopatología , Procedimientos Quirúrgicos Dermatologicos , Dermis/citología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Fibromodulina , Humanos , Metaloproteinasas de la Matriz Secretadas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteoglicanos/genética , Conejos , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Resistencia a la Tracción , Factores de Tiempo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Transfección , Factores de Crecimiento Transformadores/metabolismo , Cicatrización de Heridas/genética
12.
Hum Gene Ther ; 18(7): 589-602, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17630837

RESUMEN

The poor prognosis of patients with malignant gliomas necessitates the development of novel therapies. Virotherapy, using genetically engineered adenovectors that selectively replicate in and kill neoplastic cells, represents one such strategy. In this study, we examined several oncolytic vectors with modified transcriptional and transductional control of viral replication. First, we incorporated the survivin promoter (S) to drive E1A gene expression. We then modified the adenovirus serotype 5 (Ad5) fiber protein via genetic knob switching or incorporation of peptide ligands to target the following glioma-associated receptors: the Ad3 attachment protein, or CD46, alpha(v) beta(3)/alpha(v)beta(5) integrins, or heparan sulfate proteoglycans. The three conditionally replicative adenoviruses, CRAd-S-5/3, CRAd-S-RGD, and CRAd-S-pk7, were then examined in vitro with respect to transduction efficiency and tissue specificity. The most promising virus was then tested in vivo for evidence of tumor growth inhibition. CRAd-S-pk7 provided the highest level of viral replication and tumor oncolysis in glioma cell lines. At the same time, we observed minimal viral replication and toxicity in normal human brain. Injection of CRAd-S-pk7 inhibited xenograft tumor growth by more than 300% (p < 0.001). Sixty-seven percent of treated mice with intracranial tumors were long-term survivors (>110 days; p < 0.005). Analysis of tumor tissue indicated increased adenoviral infectivity, decreased mitotic activity, and enhanced tumor apoptosis. These findings demonstrate the effectiveness of CRAd-S-pk7 and provide the rationale for further development of this novel oncolytic virus for glioma gene therapy.


Asunto(s)
Adenoviridae , Neoplasias Encefálicas/terapia , Proteínas de la Cápside/genética , Terapia Genética , Glioma/terapia , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Vectores Genéticos , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis , Inyecciones Intralesiones , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas , Survivin , Resultado del Tratamiento , Replicación Viral
13.
Int J Oncol ; 31(5): 1177-85, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17912445

RESUMEN

Targeting gene expression to cancer cells remains a challenge for the development of gene and viral therapy for gliomas. Recent studies have highlighted transcriptional targeting as one of the possible solutions to overcome this limitation. In this context, melanoma associated antigens (MAAs) are usually over-expressed in brain tumors in comparison to normal brain tissue. For this reason, we investigated the use of the tyrosinase promoter as a transcriptional element to target oncolytic therapy for gliomas. Tyrosinase mRNA expression was evaluated by qRT-PCR in normal human brain tissue as well as in human glioma specimens. We found that this gene was significantly over-expressed in glioma cell lines and in primary glioma samples. Tyrosinase expression correlated with the grade of the tumor (p-value range: 0.05-0.001). Furthermore, transfection of several cell cultures with human and mouse tyrosinase promoters driving a luciferase reporter gene confirmed the activity of this promoter in mouse and human cells. To evaluate whether tyrosinase-activated conditionally replicative adenoviruses (CRAds) could induce toxicity in glioma cells, two vectors (Ad h/m and Ad24TYR) were tested in a mouse glioma model. C57BL/6 mice underwent intracranial injection of tumor cell line GL261. Survival was used to evaluate efficacy of the tested vectors. Mice receiving 1 x 10(9) MOI of Ad h/m and Ad24TYR following intracranial tumor implants had a median survival of 46+/-3 days (p<0.05); in contrast, those treated with medium had a median survival of 31+/-2 days. These results suggest that injection of tyrosinase CRAds leads to prolongation of survival in mice with experimental brain tumors. The tyrosinase promoter stands as a proof of principle of the potential use of MAA over-expression patterns for targeting novel anti-glioma therapies.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos/genética , Glioma/terapia , Monofenol Monooxigenasa/genética , Regiones Promotoras Genéticas , Animales , Glioma/enzimología , Humanos , Ratones , Ratones Endogámicos C57BL , Replicación Viral
14.
Int J Oncol ; 31(5): 1213-22, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17912450

RESUMEN

Conventional cancer treatments are not adequate for the majority of most patients stricken with squamous cell carcinomas of the head and neck (SCCHN). Conditionally replicating adenoviruses (CRAds) represent a promising new modality for treating of neoplastic diseases, including SCCHN. Specifically, CRAd agents infect tumor cells and selectively replicate within them, thus causing their death while sparing surrounding normal cells in the host. Oncolysis results from the replicative life cycle of the virus, which lyses infected tumor cells and releases viral progeny for propagation of infection and resultant lysis of neighboring cancer cells, sparing normal host cells. However, to date there have been two main limitations to successful clinical application of these CRAd agents: poor infectivity and poor tumor specificity. Here we report the construction of a CRAd agent, CRAd-CXCR4.F5/3, in which the adenovirus E1 gene is driven by a tumor-specific CXCR4 promoter, and the viral infectivity is enhanced by a fiber modification, F5/3, containing an Ad3 knob chimeric fiber protein. As expected, this agent improved both of the viral infectivity and tumor specificity as evaluated in established SCCHN tumor cell lines and in primary tumor tissues from multiple patients. As an added benefit, the activity of the CXCR4 promoter was low in human liver as described previously. Based on these data, the CRAd-CXCR4.F5/3 is a promising novel CRAd agent for SCCHN targeting with low host toxicity.


Asunto(s)
Adenoviridae/fisiología , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Viroterapia Oncolítica/métodos , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Citomegalovirus/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Regiones Promotoras Genéticas , Receptores CXCR4/genética , Replicación Viral
15.
Lung Cancer ; 55(2): 145-56, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17113184

RESUMEN

Conventional treatments are not adequate for the majority of lung cancer patients. Conditionally replicating adenoviruses (CRAds) represent a promising new modality for the treatment of neoplastic diseases, including non-small cell lung cancer. Specifically, following cellular infection, the virus replicates selectively in the infected tumor cells and kills the cells by cytolysis. Next, the progeny virions infect a new population of surrounding target cells, replicate again and eradicate the infected tumor cells while leaving normal cells unaffected. However, to date, there have been two main limitations to successful clinical application of these CRAd agents; i.e. poor infectivity and poor tumor specificity. Here we report the construction of a CRAd agent, CRAd-CXCR4.RGD, in which the adenovirus E1 gene is driven by a tumor-specific CXCR4 promoter and the viral infectivity is enhanced by a capsid modification, RGD4C. This agent CRAd-CXCR4.RGD, as expected, improved both of the viral infectivity and tumor specificity as evaluated in an established lung tumor cell line and in primary tumor tissue from multiple patients. As an added benefit, the activity of the CXCR4 promoter was low in human liver as compared to three other promoters regularly used for targeting tumors. In addition, this agent has the potential of targeting multiple other tumor cell types. From these data, the CRAd-CXCR4.RGD appears to be a promising novel CRAd agent for lung cancer targeting with low host toxicity.


Asunto(s)
Adenoviridae/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica/métodos , Regiones Promotoras Genéticas , Receptores CXCR4/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Vectores Genéticos , Humanos , Hígado/metabolismo , Neoplasias Pulmonares/genética , Reacción en Cadena de la Polimerasa , Células Tumorales Cultivadas , Replicación Viral
16.
J Neurosurg ; 107(3): 617-27, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17886563

RESUMEN

OBJECT: Gene therapy protocols for malignant gliomas utilize adenoviral vectors that rely almost exclusively on the adenovirus serotype 5 (Ad5) backbone. The authors have previously shown that chimeric vectors that bind to the Ad3 receptor, or CD46, increase the transduction efficiency of malignant brain tumors. In light of the debate regarding the efficacy of CD46 compared with CD80/CD86 in binding Ad3 virions, the authors now examine the expression and transduction efficiency of Ad5/3 chimeras that bind via CD80/CD86. METHODS: The authors first analyzed CD80/CD86 expression in glioma cell lines. They then used three replication-defective vectors containing a luciferase reporter gene: Ad5/3 (containing the tail and shaft domain of Ad5 and the knob domain of Ad3); Ad3/5 (containing the tail of Ad5, shaft of Ad3, and knob of Ad5); and Ad3/3 (containing the tail of Ad5, shaft of Ad3, and knob of Ad3). These vectors were analyzed both in vitro and in vivo against malignant glioma cells. To examine further the effect of Ad5/3 fiber modification, the authors created an oncolytic vector, conditionally replicative Ad5/3 (CRAd5/3). RESULTS: The Ad5/3 vector showed a 10- to 100-fold enhanced transduction efficiency of malignant glioma compared with replication-defective wild-type adenovirus (reAd5) (p < 0.05). Moreover the use of Ad5/3 reduced transgene expression by more than 90% in normal human brain cells compared with reAd5. Finally, the use of CRAd5/3 inhibited tumor cell proliferation by 43% more than replication-competent wild-type virus in vitro (p < 0.05). CONCLUSIONS: The results of this study demonstrate that the Ad5/3 vector offers superior transduction efficiency and low toxicity in the setting of brain tumors, and therefore represents a potential new approach to gene therapy for malignant gliomas.


Asunto(s)
Adenoviridae , Antígeno B7-1/fisiología , Antígeno B7-2/fisiología , Neoplasias Encefálicas/metabolismo , Vectores Genéticos , Glioma/metabolismo , Transducción Genética/métodos , Animales , Técnicas de Cultivo de Célula , Quimera , Humanos , Ratones , Ratones Endogámicos BALB C , Células Tumorales Cultivadas , Acoplamiento Viral
17.
Cancer Biol Ther ; 5(12): 1647-53, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17106248

RESUMEN

Clinical studies of replicating adenoviruses for the treatment of cancer have demonstrated their safety but have yielded disappointing results, indicating the need for new strategies to improve their efficacy. We hypothesized that the efficacy of a replicating adenovirus could be improved by expression of tissue inhibitor of metalloproteinases-2 (TIMP-2), a 21-kDa unglycosylated secretory protein. TIMP-2 specifically inhibits the active forms of a number of matrix metalloproteinases (MMPs) that play a role in the degradation of basement membranes and the extracellular matrix and are therefore involved in the control of the growth, invasion and metastasis of tumor cells, as well as angiogenesis. In addition, TIMP-2 can abrogate tumor growth and angiogenesis by a variety of mechanisms independent of MMP inhibition. In this study, we demonstrate that expression of TIMP-2 enhanced the antitumor efficacy of a replicating adenovirus in vivo, by reducing both tumor growth and angiogenesis.


Asunto(s)
Adenovirus Humanos/fisiología , Neoplasias de la Mama/virología , Neoplasias/prevención & control , Inhibidor Tisular de Metaloproteinasa-2/genética , Replicación Viral/fisiología , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/genética , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citomegalovirus/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/prevención & control , Regiones Promotoras Genéticas , Trasplante Heterólogo
18.
Int J Oncol ; 29(5): 1319-29, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17016667

RESUMEN

Cholangiocarcinoma is a highly malignant neoplasm with no effective treatment. Conditionally replicative adenoviruses (CRAds) represent a promising new modality for the treatment of cancer in general. A key contribution in this regard was the introduction of tumor-selective viral replication for amplification of the initial inoculum in the neoplastic cell population. Under ideal conditions following cellular infection, the viruses replicate selectively in the infected tumor cells and kill the cells by cytolysis, leaving normal cells unaffected. However, to date there have been two limitations to the clinical application of these CRAd agents, i.e. poor viral infectivity and tumor specificity. Here we report the construction of three new CRAd agents, CRAd-S.RGD, CRAd-S.F5/3 and CRAd-S.pk7, in which the tumor specificity is regulated by a tumor-specific promoter, the survivin promoter, and the viral infectivity is enhanced by incorporating a capsid modification (RGD, F5/3 or pk7) in the adenovirus fiber region. These CRAd agents effectively target cholangiocarcinoma cells, induce strong cytoxicity in these cells in vitro and inhibit tumor growth in a murine xenograft model in vivo. In addition, the survivin promoter has extremely low activity both in the non-transformed cell line, HMEC, and in human liver tissue. Our results suggest that the survivin-based CRAds are promising agents for targeting cholangiocarcinoma with low host toxicity. Such results should provide important insights into the identification of novel therapeutic strategies for cholangiocarcinoma.


Asunto(s)
Adenoviridae/genética , Neoplasias de los Conductos Biliares/terapia , Colangiocarcinoma/terapia , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Viroterapia Oncolítica , Replicación Viral/genética , Animales , Neoplasias de los Conductos Biliares/genética , Cápside/metabolismo , Células Cultivadas , Colangiocarcinoma/genética , Citomegalovirus/genética , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas/genética , Survivin , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Neurosurg ; 104(4): 583-92, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16619663

RESUMEN

OBJECT: Malignant brain tumors have been proved to be resistant to standard treatments and therefore require new therapeutic strategies. Survivin, a recently described member of the inhibitor of apoptosis protein family, is overexpressed in several human brain tumors, primarily gliomas, but is downregulated in normal tissues. The authors hypothesized that the expression of tumor-specific survivin could be exploited for treatment of gliomas by targeting the tumors with gene therapy vectors. METHODS: Following confirmation of survivin expression in glioma cell lines, an adenoviral vector containing the survivin promoter and the reporter gene luciferase was tested in established and primary glioma cells, normal astrocytic cells, and normal human brain tissues. High levels of reporter gene expression were observed in established tumor and primary tumor cell lines and low levels of expression in astrocytes and normal human brain tissue. To test oncolytic potency, the authors constructed survivin promoter-based conditionally replicative adenoviruses (CRAds), composed of survivin promoter-regulated E1 gene expression and an RGD-4C capsid modification. These CRAds could efficiently replicate within and kill a variety of established glioma tumor cells, but were inactive in a normal human liver organ culture. Finally, survivin promoter-based CRAds significantly inhibited the growth of glioma xenografts in vivo. CONCLUSIONS: Together these data indicate that the survivin promoter is a promising tumor-specific promoter for transcriptional targeting of adenovirus-based vectors and CRAds for malignant gliomas. The strategy of using survivin-CRAds may thus translate into an experimental therapeutic approach that can be used in human clinical trials.


Asunto(s)
Neoplasias Encefálicas/genética , Marcación de Gen , Terapia Genética/métodos , Glioma/genética , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Adenoviridae , Animales , Apoptosis/genética , Línea Celular Tumoral , Femenino , Vectores Genéticos , Humanos , Proteínas Inhibidoras de la Apoptosis , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Survivin , Trasplante Heterólogo , Replicación Viral/genética
20.
Cancer Res ; 62(16): 4663-70, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12183423

RESUMEN

Malignant melanoma is characterized by growing incidence, early metastasis, and a lack of effective treatment for advanced disease, suggesting a pressing need for novel therapeutic approaches. Conditionally replicative adenoviruses (CRAds) constitute a new and promising strategy for cancer treatment that has been rapidly translated into clinical trials. We engineered novel melanoma-targeted CRAds, AdTyrdelta24 and AdTyrdelta2delta24, by replacing the adenoviral E1A promoter with a cassette containing a polyA sequence and a human tyrosinase enhancer/promoter construct (hTyr2E/P). The small size of this cassette allows retention of the E3 region within these CRAds, which was shown to enhance viral spread and oncolysis. In addition, we introduced mutations (delta24 and delta2delta24) into the viral E1A gene, which attenuate adenoviral replication in quiescent cells. The cell cycle pathways mediating this attenuation are defective in melanoma cells. By analysis of E1A expression, we prove fidelity of hTyr2E/P in the adenoviral genome and in the context of viral replication when an upstream polyA was included. We further show efficient cytotoxicity of AdTyrdelta24 and AdTyrdelta2delta24 in melanoma cell lines and a 100-1000-fold attenuation in cell lines derived from various nonmelanocytic tissues. Virus replication and progeny production of these viruses were similarly selective, resulting in 200-800-fold higher virus yields in melanoma cells versus control cells, thus establishing viral cytolysis and spread as the cause of the observed cell killing. Cytotoxicity of AdTyrdelta24 for normal fibroblasts and keratinocytes was strongly attenuated, and this virus caused selective killing of melanoma cells but not surrounding keratinocytes in a coculture system. Progeny production and cytotoxicity of AdTyrdelta24 in melanoma cells were similar to matching viruses containing the stronger cytomegalovirus enhancer/promoter instead of hTyr2E/P. Furthermore, AdTyrdelta24 showed a cytopathic effect similar to the wild-type E1A containing AdTyrwt and only minimally reduced compared with wild-type adenovirus. We conclude that the generated CRAds AdTyrdelta24 and AdTyrdelta2delta24 constitute novel targeted agents for gene therapy and viral oncolysis of metastatic melanoma.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/fisiología , Melanoma/terapia , Monofenol Monooxigenasa/genética , Proteínas E1A de Adenovirus/metabolismo , Adenovirus Humanos/genética , Técnicas de Cocultivo , Efecto Citopatogénico Viral , Humanos , Queratinocitos/citología , Queratinocitos/virología , Melanoma/virología , Regiones Promotoras Genéticas , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA