Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
Int J Oncol ; 34(3): 729-42, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19212678

RESUMEN

Survivin has gained attention as a tumor-specific marker which is upregulated in a variety of neoplasms. Although the survivin protein is implicated in anti-apoptotic tumor pathways, little is known about the function of the survivin promoter. In this study, we constructed a conditionally replicative adenoviral vector (CRAd) that utilizes the survivin promoter and examined the mechanism of CRAd induced cell death in malignant glioma. Our results indicate that CRAd vectors which utilize the survivin promoter effectively replicate in glioma cells and exhibit a high oncolytic effect. The survivin-mediated CRAd appeared to induce apoptosis as measured by Annexin/7-AAD. Caspase-3 and BAX mRNAs were upregulated based on microarray data, however, Western blot analysis of infected cells showed no evidence of elevated caspase-3, BAX, or p53 protein expression. Of note, at each time point infected glioma cells showed no evidence of activated BAD or AKT. The inhibition of AKT signaling led us to examine autophagy in infected cells. Electron micrographs of virally infected glioma cells suggested auto-phagosomal-mediated cell death and selective blocking of beclin with siRNA prevented autophagy. These results indicate that the survivin promoter enhances viral replication and induces autophagy of infected glioma cells via a beclin-dependent mechanism.


Asunto(s)
Adenoviridae/fisiología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/terapia , Proteínas de la Membrana/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/genética , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/fisiología , Beclina-1 , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Caspasa 3/genética , Caspasa 3/metabolismo , Ciclo Celular/fisiología , Línea Celular Tumoral , Glioma/genética , Glioma/metabolismo , Glioma/virología , Células HeLa , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis , Proteínas de la Membrana/genética , Análisis por Micromatrices , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteína Oncogénica v-akt/biosíntesis , Virus Oncolíticos/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Survivin , Proteína Letal Asociada a bcl/biosíntesis
4.
Int J Oncol ; 32(6): 1179-88, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18497979

RESUMEN

Human ovarian cancer is a highly lethal malignant neoplasm in woman with no effective treatment if conventional chemotherapy fails. In this regard, conditionally replicative adenoviruses (CRAds) represent a promising new modality for the treatment of cancer. A key contribution to the development of CRAds was the introduction of tumor-selective viral replication to restrict amplification to the neoplastic cell population. Under ideal conditions following cellular infection, the viruses replicate selectively in the infected tumor cells, killing the cells by cytolysis, leaving normal cells unaffected. However, to date, there have been limitations to the clinical application of these CRAd agents i.e. poor viral infectivity, poor tumor specificity and high toxicity. Here, we report the in vitro and in vivo comparison of four CRAd agents developed for ovarian cancer application, specifically, Ad-Delta24.F5/3, CRAd-C.F5/3, CRAd-M.F5/3 and CRAd-S.F5/3. All CRAd agents contained fiber knob chimeras of adenovirus serotype 3, which enhanced the viral infectivity at the transductional level via a non-Coxsackie-Adenovirus Receptor alternative pathway. In addition, these CRAds embodied distinct mechanisms for the achievement of replication specificity. Tumor cell killing was assessed by using an oncolytic assay and a cell viability assay (MTS) in vitro, while tumor growth was examined in a xenograft model in vivo by using a bioluminescent imaging assay. In addition, the replication rates of the CRAd agents were determined in human liver slices. Both the Ad-Delta24.F5/3 and CRAd-S.F5/3 were demonstrated to have higher tumor killing effects in tumor cells and a lower viral replication rate in human liver. These agents are thus excellent candidates for clinical trials of CRAd agents against human ovarian cancer.


Asunto(s)
Adenoviridae/genética , Proliferación Celular , Viroterapia Oncolítica , Neoplasias Ováricas/terapia , Replicación Viral , Animales , Femenino , Terapia Genética , Vectores Genéticos , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/genética , Transducción Genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Am J Obstet Gynecol ; 199(2): 117.e1-6, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18674655

RESUMEN

OBJECTIVE: The purpose of this study was to screen a panel of targeted adenoviruses as vectors for endometriosis gene therapy. STUDY DESIGN: Endometriotic cells were obtained from subjects with ovarian endometriomas. Liver tissues were taken from donors during hepatic transplantation surgery. Human endometriotic cells and liver tissues were transfected by targeted adenoviruses expressing luciferase reporter gene. Luciferase activity that was mediated by each virus was expressed as a percentage of adenovirus serotype 5 (Ad5-CMV-luc) activity. The 2-tailed Studentt test was used to compare the adenovirus data. RESULTS: In endometriotic cells, the adenovirus-RGD (Ad-RGD-luc), adenovirus under secretory leukocyte protease inhibitor promoter (Ad-SLPI-luc), and adenovirus under heparanase promoter (Ad-heparanase-luc) showed significantly higher activity, compared with the adenovirus serotype 5. In liver tissues, adenovirus-survivin (Ad-survivin-luc) and Ad-heparanase-luc had significantly lower activity, compared with adenovirus serotype 5. CONCLUSION: Ad-heparanase-luc showed "endometriosis on, liver off" phenotype and is a promising vector for endometriosis gene therapy.


Asunto(s)
Adenoviridae/genética , Endometriosis/terapia , Marcación de Gen/métodos , Terapia Genética/métodos , Proteínas de la Cápside , Células Cultivadas , Endometrio/citología , Femenino , Vectores Genéticos , Glucuronidasa/metabolismo , Humanos , Hígado/enzimología , Luciferasas/metabolismo , Activación Transcripcional/fisiología , Transducción Genética/métodos , Transfección
6.
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
7.
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
8.
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
9.
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
10.
Clin Transl Med ; 6(1): 11, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28251571

RESUMEN

BACKGROUND: Radiogenetic therapy is a novel approach in the treatment of cancer, which employs genetic modification to alter the sensitivity of tumor cells to the effect of applied radiation. AIM: To select a potent radiation inducible promoter in the context of brain tumors and to investigate if CArG radio responsive motifs or other elements in the promoter nucleotide sequences can correlate to its response to radiation. METHODS: To select initial candidates for promoter inducible elements, the levels of mRNA expression of six different promoters were assessed using Quantitative RTPCR in D54 MG cells before and after radiation exposure. Recombinant Ad/reporter genes driven by five different promoters; CMV, VEGF, FLT-1, DR5 and survivin were constructed. Glioma cell lines were infected with different multiplicity of infection of the (promoter) Ad or CMV Ad. Cells were then exposed to a range of radiation (0-12 Gy) at single fraction. Fluorescent microscopy, Luc assay and X-gal staining was used to detect the level of expression of related genes. Different glioma cell lines and normal astrocytes were infected with Ad survivin and exposed to radiation. The promoters were analyzed for presence of CArG radio-responsive motifs and CCAAT box consensus using NCBI blast bioinformatics software. RESULTS: Radiotherapy increases the expression of gene expression by 1.25-2.5 fold in different promoters other than survivin after 2 h of radiation. RNA analysis was done and has shown an increase in copy number of tenfold for survivin. Most importantly cells treated with RT and Ad Luc driven by survivin promoter showed a fivefold increase in expression after 2 Gy of radiation in comparison to non-irradiated cells. Presence or absence of CArG motifs did not correlate with promoter response to radiation. Survivin with the best response to radiation had the lowest number of CCAAT box. CONCLUSION: Survivin is a selective potent radiation inducible promoter for glioblastoma viral gene therapy and this response to radiation could be independent of CArG motifs.

11.
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
12.
Breast Cancer Res ; 7(6): R1141-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16457694

RESUMEN

INTRODUCTION: In view of the limited success of available treatment modalities for metastatic breast cancer, alternative and complementary strategies need to be developed. Adenoviral vector mediated strategies for breast cancer gene therapy and virotherapy are a promising novel therapeutic platform for the treatment of breast cancer. However, the promiscuous tropism of adenoviruses (Ads) is a major concern. Employing tissue specific promoters (TSPs) to restrict transgene expression or viral replication is an effective way to increase specificity towards tumor tissues and to reduce adverse effects in non-target tissues such as the liver. In this regard, candidate breast cancer TSPs include promoters of the genes for the epithelial glycoprotein 2 (EGP-2), cyclooxygenase-2 (Cox-2), alpha-chemokine SDF-1 receptor (stromal-cell-derived factor, CXCR4), secretory leukoprotease inhibitor (SLPI) and survivin. METHODS: We employed E1-deleted Ads that express the reporter gene luciferase under the control of the promoters of interest. We evaluated this class of vectors in various established breast cancer cell lines, primary breast cancer cells and finally in the most stringent preclinical available substrate system, constituted by precision cut tissue slices of human breast cancer and liver. RESULTS: Overall, the CXCR4 promoter exhibited the highest luciferase activity in breast cancer cell lines, primary breast cancer cells and breast cancer tissue slices. Importantly, the CXCR4 promoter displayed a very low activity in human primary fibroblasts and human liver tissue slices. Interestingly, gene expression profiles correlated with the promoter activities both in breast cancer cell lines and primary breast cancer cells. CONCLUSION: These data suggest that the CXCR4 promoter has an ideal 'breast cancer-on/liver-off' profile, and could, therefore, be a powerful tool in Ad vector based gene therapy or virotherapy of the carcinoma of the breast.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Adenoviridae/genética , Línea Celular Tumoral , Femenino , Fibroblastos , Marcadores Genéticos , Vectores Genéticos , Humanos , Hígado/citología , Luciferasas/biosíntesis , Regiones Promotoras Genéticas , Receptores CXCR4 , Tropismo
13.
Cancer Biol Ther ; 4(11): 1203-10, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16177567

RESUMEN

Successful adenoviral (Ad) vector-mediated strategies for breast cancer gene therapy and virotherapy have heretofore been hindered by low transduction efficiency. This has recently been understood to result from a relative paucity of expression of the primary adenovirus receptor, coxsackie-adenovirus-receptor (CAR), on primary tumor cells. To further investigate this issue, we evaluated the expression of CAR on breast cancer cell lines as well as primary breast cancer cells. With the exception of one patient sample, CAR expression was notably higher in the tumor cells from patients compared to CAR expression in the tumor cell lines. Furthermore, we explored CAR-independent targeting strategies to breast cancer tissue by exploring a panel of infectivity-enhanced Ad vectors, which contain CAR-independent targeting motifs for their utility in breast cancer gene therapy and virotherapy. These targeting motifs included Ad 3 knob (Ad5/3), canine Ad serotype 2 knob (Ad5CAV-2), RGD (Ad5.RGD), polylysine (Ad5.pK7), or both RGD and polylysine (Ad5.RGD.pK7), and were tested using the breast cancer tissue slice model, which is the most stringent substrate system available. Of all the tested tropism modified Ad vectors, Ad5/3 exhibited the highest transductional efficiency in breast cancer. These preclinical results suggest that Ad5/3 is the most useful modification to achieve higher clinical efficacy of breast cancer gene therapy and virotherapy.


Asunto(s)
Adenoviridae/química , Adenoviridae/genética , Neoplasias de la Mama/terapia , Técnicas de Transferencia de Gen , Vectores Genéticos , Adenoviridae/clasificación , Adulto , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma/patología , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Femenino , Humanos , Persona de Mediana Edad , Modelos Biológicos , Receptores Virales/metabolismo , Serotipificación , Especificidad por Sustrato , Transducción Genética , Células Tumorales Cultivadas
14.
Int J Oncol ; 27(1): 237-46, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15942665

RESUMEN

Conditionally replicating adenoviruses (CRAds) represent a promising new modality for the treatment of cancer. A key contribution in this regard was the introduction of tumor-selective viral replication for amplification of the initial inoculum. Specifically, following cellular infection, the virus replicates selectively in the infected tumor cells and kills the cells by cytolysis. Next, the progeny virions infect surrounding target cells, replicate and eradicate the infected tumor cells, leaving normal cells unaffected. However, to date there have been two limitations to clinical application of these CRAd agents; i.e., both infectivity and tumor specificity are poor. Survivin protein is a novel member of the inhibitor of apoptosis (IAP) protein family, which plays an important role in the survival of cancer cells and progression of malignancies. Previous data have shown the survivin promoter has high activities in multiple cancer cells with a low activity in mouse liver. In this study, we propose an improved CRAd agent to circumvent the obstacles. We constructed a novel CRAd agent, CRAd-Survivin-RGD, which contains both the survivin promoter (either the short version, S-S, or the long version, S-L) to selectively drive E1 gene expression in tumor cells and a capsid modification and RGD4C to specifically enhance the tumor infectivity of CRAd agents. Both CRAd agents (S-S and S-L) showed high replication rates in the breast cancer cell line, MDA-MB-361, and low promoter activity in both normal mouse and human liver, thus signifying the CRAd agents have the phenotype of 'tumor on/liver off'. In cytocidal experiments, the CRAd agents demonstrated a high cytocidal effect on multiple cancer cell lines, including the breast cancer cell line, MDA-MB-231; the glioma cell line, D65, the melanoma cell line, MEL-28; and mesothelioma, Meso2374. The results also showed the tumor growth was dramatically inhibited by intertumoral administration of the CRAd agents in a breast cancer (MDA-MB-361) xenograft animal model. These data clearly demonstrate that CRAd-Survivin-RGD is a potential novel therapeutic agent for treatment in many, but not all, human cancers.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Terapia Genética/métodos , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Oligopéptidos/genética , Regiones Promotoras Genéticas , Proteínas E1 de Adenovirus/genética , Secuencias de Aminoácidos , Animales , Línea Celular Tumoral , Femenino , Vectores Genéticos , Humanos , Técnicas In Vitro , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Modelos Genéticos , Familia de Multigenes , Trasplante de Neoplasias , Fenotipo , Reacción en Cadena de la Polimerasa , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Survivin , Factores de Tiempo
15.
Leuk Lymphoma ; 46(11): 1627-1638, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16334907

RESUMEN

Lymphomas and leukemias, neoplasms of hematopoetic lineage, pose unique challenges that require novel treatment paradigms. The inter-relationship between the immune system and the neoplastic lesion in these diseases dictates that, to evaluate novel therapies, models are needed that mimic human disease in an immunocompetent host. In the present study, we describe a disseminated, syngeneic model of B-cell lymphoma in the Balb/c mouse based upon the A20 cell line. This model mimics aspects of diffuse large B-cell lymphomas in humans, and recapitulates para-spinous tumor growth, bone destruction and nerve root compression, which may complicate disseminated disease. Furthermore, this tumor expresses a key marker of interest, CD40, which is a candidate for tumor-specific vector targeting via current modalities. The present study therefore describes a high-fidelity model of disseminated lymphoma with implications for novel targeted therapeutics. Lymphomas and leukemias, neoplasms of hematopoetic lineage, pose unique challenges that require novel treatment paradigms. The inter-relationship between the immune system and the neoplastic lesion in these diseases dictates that, to evaluate novel therapies, models are needed that mimic human disease in an immunocompetent host. In the present study, we describe a disseminated, syngeneic model of B-cell lymphoma in the Balb/c mouse based upon the A20 cell line. This model mimics aspects of diffuse large B-cell lymphomas in humans, and recapitulates para-spinous tumor growth, bone destruction and nerve root compression, which may complicate disseminated disease. Furthermore, this tumor expresses a key marker of interest, CD40, which is a candidate for tumor-specific vector targeting via current modalities. The present study therefore describes a high-fidelity model of disseminated lymphoma with implications for novel targeted therapeutics.


Asunto(s)
Linfoma de Células B/patología , Linfoma de Células B Grandes Difuso/patología , Neoplasias Experimentales/patología , Animales , Enfermedades Óseas , Antígenos CD40/análisis , Línea Celular Tumoral , Humanos , Huésped Inmunocomprometido , Linfoma de Células B/complicaciones , Linfoma de Células B Grandes Difuso/complicaciones , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/complicaciones , Radiculopatía , Neoplasias de la Columna Vertebral , Trasplante Isogénico
16.
Mol Cancer Ther ; 3(6): 687-91, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15210854

RESUMEN

Metastatic renal cell carcinoma (RCC) is often resistant to standard treatment, thereby requiring new therapeutic strategies. In this regard, tumor cell migration and metastasis have recently been shown to be regulated by chemokines and their respective receptors (e.g., SDF-1alpha/CXCR4). In the context of RCC, up-regulation of CXCR4 expression is closely related to the development of invasive cancer. Thus, we hypothesized that the CXCR4 pathway could be exploited for RCC targeting with gene therapy vectors. In this regard, targeting adenoviral vectors to tumor cells is critically dependent on tumor-specific gene expression. Toward the end of RCC tumor targeting, we evaluated the utility of the CXCR4 promoter in an adenoviral context. First, overexpression of CXCR4 was confirmed in several RCC cell lines. Next, an adenoviral vector was constructed, whereby the human CXCR4 promoter drives the expression of a reporter gene. We tested the activity of the CXCR4 promoter in vitro and in vivo in relevant models. Our data indicate that the human CXCR4 promoter is highly active in RCC cells but not in normal human cells. Finally, biodistribution studies in mice demonstrated dramatic repression of the CXCR4 promoter in the liver but not in the kidney. In conclusion, the unique activity of the CXCR4 promoter in RCC lines and its repression in normal human cells and in the murine liver underscore its potential utility as a novel candidate for transcriptional targeting of RCC.


Asunto(s)
Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Regiones Promotoras Genéticas/genética , Receptores CXCR4/genética , Transcripción Genética/genética , Adenoviridae/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Genes Reporteros/genética , Terapia Genética , Vectores Genéticos/genética , Humanos , Hígado/metabolismo , Ratones
17.
Cancer Gene Ther ; 11(4): 256-62, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15017380

RESUMEN

It has been demonstrated that survivin, a novel member of the inhibitor of apoptosis (IAP) protein family, is expressed in human cancers but is undetectable in normal differentiated tissues. We employed a recombinant adenoviral vector (reAdGL3BSurvivin) in which a tumor-specific survivin promoter and a luciferase reporter gene were inserted into the E1-deleted region of adenovirus vector. Luciferase activity was measured in both multiple tumor cell lines and two primary melanoma cells infected with reAdGL3BSurvivin. Human fibroblast and mammary epithelial cell lines were used as negative controls. A reAdGL3CMV, containing the CMV promoter and luciferase gene, was used as a positive control to normalize the luciferase activity generated by the survivin promoter. Our data revealed that the survivin promoter showed high activity in both established tumor cell lines and the primary melanoma cells. In contrast, the in vivo studies indicated that the activities of survivin promoter were extremely low in the major mouse organs. The survivin promoter appears to be a promising tumor-specific promoter exhibiting a "tumor on" and "liver off" profile, and therefore, it may prove to be a good candidate for transcriptional targeting of cancer gene therapy in a wide variety of tumors.


Asunto(s)
Terapia Genética/métodos , Proteínas Asociadas a Microtúbulos/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas/genética , Transcripción Genética , Adenoviridae/genética , Animales , Línea Celular Tumoral , Ciclooxigenasa 2 , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Transferencia de Gen , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Isoenzimas/genética , Luciferasas/análisis , Luciferasas/genética , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias , Neoplasias/terapia , Prostaglandina-Endoperóxido Sintasas/genética , ARN Mensajero/análisis , Survivin , Transcripción Genética/genética
19.
Cancer Res ; 68(14): 5778-84, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632631

RESUMEN

To improve the efficacy and selectivity of virotherapy for malignant glioma, we designed a strategy to amplify adenoviral replication in conjunction with radiotherapy using a radioinducible promoter. First, we compared the radiation-inducible activity of FLT-1, vascular endothelial growth factor, DR5, Cox2, and survivin. We then examined the capacity of the optimal promoter to modulate transgene expression followed by E1A activity in vitro and in vivo in a glioma stem cell model. In the presence of radiation, survivin mRNA activity increased 10-fold. Luciferase transgene expression was dose dependent and optimal at 2 Gy. A novel oncolytic adenovirus, CRAd-Survivin-pk7, showed significant toxicity and replication against a panel of passaged and primary CD133(+) glioma stem cells. On delivery of radiation, the toxicity associated with CRAd-Survivin-pk7 increased by 20% to 50% (P < 0.05). At the same time, the level of E1A activity increased 3- to 10-fold. In vivo, treatment of U373MG CD133(+) stem cells with CRAd-Survivin-pk7 and radiation significantly inhibited tumor growth (P < 0.05). At the same time, the level of E1A activity was 100-fold increased versus CRAd-Survivin-pk7 alone. Selected genes linked to radioinducible promoters whose expression can be regulated by ionizing radiation may improve the therapeutic ratio of virotherapy. In this study, we have identified a new radioinducible promoter, survivin, which greatly enhances the activity of an oncolytic adenovirus in the presence of low-dose radiotherapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Viroterapia Oncolítica/métodos , Células Madre/metabolismo , Antígeno AC133 , Animales , Antígenos CD/biosíntesis , Glicoproteínas/biosíntesis , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Péptidos , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Proteínas Represoras , Survivin
20.
J Gene Med ; 9(3): 151-60, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17351980

RESUMEN

BACKGROUND: Malignant gliomas remain refractory to Ad5-mediated gene therapy due to deficiency of the coxsackie adenovirus receptor on tumor cells. The purpose of this study was to evaluate whether changes in adenoviral tropism can enhance gene transfer in the context of malignant glioma. METHODS: We have identified several receptors that are over-expressed on tumor cells and created a series of pseudotyped Ad5 vectors that recognize these receptors: Ad5-RGD which binds alpha(v)beta3/alpha(v)beta5 integrins; Ad5/3 which contains adenovirus serotype 3 knob and binds to CD46; Ad5-Sigma which incorporates the reovirus sigma knob and binds to junctional adhesion molecule-1; and Ad5-pk7 which contains the polylysine motif and binds heparan sulfate proteoglycans. We also investigated the Ad5-CAV1 vector, which contains the knob of canine adenovirus type 1, a virus previously shown to infect glioma via an unknown mechanism. In this study, we compared these modified vectors for their ability to promote the expression of luciferase transgene both in vitro and in vivo. RESULTS: Our results indicate that all five modified vectors attained higher mean luciferase activity vs. control. Among them, Ad5-CAV1 and Ad5-pk7 attained the highest transduction efficiency independent of different tumor lines or infection time. Ad5-Sigma and Ad5-pk7 also demonstrated the least nonspecific infection in normal human astrocytes. Most importantly, Ad5-pk7 achieved 1000-fold increased transgene expression in human glioma xenografts in vivo. CONCLUSIONS: These results indicate that modifications of adenoviral tropism can enhance gene transfer in tumors that are poorly susceptible to adenoviral vectors and warrant further development of Ad5-pk7 for glioma gene therapy.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vectores Genéticos , Glioma/terapia , Transducción Genética/métodos , Animales , Neoplasias Encefálicas/química , Proteínas de la Cápside/análisis , Línea Celular Tumoral , Glioma/química , Humanos , Hígado/química , Luciferasas/análisis , Luciferasas/genética , Ratones , Ratones SCID , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/metabolismo , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA