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










Base de dados
Intervalo de ano de publicação
1.
Clin Transl Med ; 6(1): 11, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28251571

RESUMO

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.

2.
Mol Ther ; 20(12): 2222-33, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22948673

RESUMO

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.


Assuntos
Proteínas E1A de Adenovirus/genética , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neoplasias Ovarianas/genética , Células Estromais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Imaging ; 8(5): 264-77, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19796604

RESUMO

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.


Assuntos
Adenoviridae/metabolismo , Adenoviridae/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Adenoviridae/genética , Capsídeo/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/fisiologia , Microscopia de Fluorescência , Simplexvirus/enzimologia , Timidina Quinase/genética , Timidina Quinase/fisiologia , Proteína Vermelha Fluorescente
4.
Int J Oncol ; 34(3): 729-42, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19212678

RESUMO

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.


Assuntos
Adenoviridae/fisiologia , Proteínas Reguladoras de Apoptose/biossíntese , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/terapia , Proteínas de Membrana/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Adenoviridae/genética , Proteínas Reguladoras de Apoptose/genética , Autofagia/fisiologia , Proteína Beclina-1 , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virologia , Caspase 3/genética , Caspase 3/metabolismo , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Glioma/genética , Glioma/metabolismo , Glioma/virologia , Células HeLa , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose , Proteínas de Membrana/genética , Análise em Microsséries , Proteínas Associadas aos Microtúbulos/biossíntese , Proteína Oncogênica v-akt/biossíntese , Vírus Oncolíticos/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Survivina , Proteína de Morte Celular Associada a bcl/biossíntese
5.
Am J Obstet Gynecol ; 199(2): 117.e1-6, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18674655

RESUMO

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.


Assuntos
Adenoviridae/genética , Endometriose/terapia , Marcação de Genes/métodos , Terapia Genética/métodos , Proteínas do Capsídeo , Células Cultivadas , Endométrio/citologia , Feminino , Vetores Genéticos , Glucuronidase/metabolismo , Humanos , Fígado/enzimologia , Luciferases/metabolismo , Ativação Transcricional/fisiologia , Transdução Genética/métodos , Transfecção
6.
Cancer Res ; 68(14): 5778-84, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632631

RESUMO

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.


Assuntos
Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Terapia Viral Oncolítica/métodos , Células-Tronco/metabolismo , Antígeno AC133 , Animais , Antígenos CD/biossíntese , Glicoproteínas/biossíntese , Humanos , Proteínas Inibidoras de Apoptose , Camundongos , Camundongos Nus , Transplante de Neoplasias , Peptídeos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Proteínas Repressoras , Survivina
7.
Int J Oncol ; 32(6): 1179-88, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497979

RESUMO

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.


Assuntos
Adenoviridae/genética , Proliferação de Células , Terapia Viral Oncolítica , Neoplasias Ovarianas/terapia , Replicação Viral , Animais , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Transdução Genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Oncol ; 31(5): 1213-22, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17912450

RESUMO

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.


Assuntos
Adenoviridae/fisiologia , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Terapia Viral Oncolítica/métodos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/virologia , Linhagem Celular Tumoral , Citomegalovirus/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Regiões Promotoras Genéticas , Receptores CXCR4/genética , Replicação Viral
9.
J Neurosurg ; 107(3): 617-27, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17886563

RESUMO

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.


Assuntos
Adenoviridae , Antígeno B7-1/fisiologia , Antígeno B7-2/fisiologia , Neoplasias Encefálicas/metabolismo , Vetores Genéticos , Glioma/metabolismo , Transdução Genética/métodos , Animais , Técnicas de Cultura de Células , Quimera , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas , Ligação Viral
10.
Hum Gene Ther ; 18(7): 589-602, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17630837

RESUMO

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.


Assuntos
Adenoviridae , Neoplasias Encefálicas/terapia , Proteínas do Capsídeo/genética , Terapia Genética , Glioma/terapia , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Feminino , Vetores Genéticos , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose , Injeções Intralesionais , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Survivina , Resultado do Tratamento , Replicação Viral
11.
Cancer Biol Ther ; 6(5): 679-85, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17404502

RESUMO

OBJECTIVE: Transcriptional targeting is a key strategy to enhance therapeutic efficacy of gene therapy applications. In the context of oncolytic virotherapy, transcriptional promoter elements are used from genes that are over expressed in a variety of malignant cancers. In the present study, we examined the feasibility of transcriptional targeting to glioma cells by comparing the activity of survivin, midkine, and CXCR4 tumor-specific promoters. METHODS: To evaluate the expression level of several glioma related genes, we performed quantitative RT-PCR analyses on samples obtained from cell lines and patients. To determine specific level of gene expression mediated by selective promoter elements, we measured luciferase expression in glioma samples transduced with replication deficient adenoviral vectors. Finally, we incorporated the optimal promoters into a conditionally replicative adenoviral vector, CRAd-5/3, and examined the cytopathic effect in vitro. RESULTS: The survivin promoter demonstrated the highest level of mRNA expression in primary tumor samples and cell lines. Transcriptional targeting was confirmed by infection of glioma cells with an adenovirus expression vector containing a surviving-driven luciferase reporter gene. Of the tested promoters, minimal level of survivin activity was detected in normal human liver and brain. A novel vector, CRAd-survivin5/3, with E1a under the control of the survivin promoter, exhibited enhanced cytopathic effect in vitro. CONCLUSIONS: Our data demonstrate that the survivin promoter element is very active in glioma samples and has low activity in normal human brain and liver. A novel oncolytic virus, CRAd-survivin-5/3, was effective against a panel of glioma cell lines in vitro. Our results suggest that employing the survivin promoter element in the context of CRAd-5/3 may present a new opportunity for the development of glioma specific oncolytic vectors.


Assuntos
Citocinas/genética , Terapia Genética/métodos , Glioma/terapia , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , Receptores CXCR4/genética , Transcrição Gênica/genética , Adenoviridae/genética , Apoptose/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Citocinas/biossíntese , Marcação de Genes , Vetores Genéticos , Glioma/genética , Glioma/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Fígado/metabolismo , Luciferases/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Midkina , Proteínas de Neoplasias/biossíntese , RNA Mensageiro/metabolismo , Receptores CXCR4/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina
12.
J Gene Med ; 9(3): 151-60, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17351980

RESUMO

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.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos , Glioma/terapia , Transdução Genética/métodos , Animais , Neoplasias Encefálicas/química , Proteínas do Capsídeo/análise , Linhagem Celular Tumoral , Glioma/química , Humanos , Fígado/química , Luciferases/análise , Luciferases/genética , Camundongos , Camundongos SCID , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/metabolismo , Transgenes , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Breast Cancer Res Treat ; 105(2): 157-67, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17221158

RESUMO

PURPOSE: Alternative and complementary therapeutic strategies need to be developed for metastatic breast cancer. Virotherapy is a novel therapeutic approach for the treatment of cancer in which the replicating virus itself is the anticancer agent. However, the success of virotherapy has been limited due to inefficient virus delivery to the tumor site. The present study addresses the utility of human mesenchymal stem cells (hMSCs) as intermediate carriers for conditionally replicating adenoviruses (CRAds) to target metastatic breast cancer in vivo. EXPERIMENTAL DESIGN: HMSC were transduced with CRAds. We used a SCID mouse xenograft model to examine the effects of systemically injected CRAd loaded hMSC or CRAd alone on the growth of MDA-MB-231 derived pulmonary metastases (experimental metastases model) in vivo and on overall survival. RESULTS: Intravenous injection of CRAd loaded hMSCs into mice with established MDA-MB-231 pulmonary metastatic disease homed to the tumor site and led to extended mouse survival compared to mice treated with CRAd alone. CONCLUSION: Injected hMSCs transduced with CRAds suppressed the growth of pulmonary metastases, presumably through viral amplification in the hMSCs. Thus, hMSCs may be an effective platform for the targeted delivery of CRAds to distant cancer sites such as metastatic breast cancer.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Terapia Genética , Neoplasias Pulmonares/terapia , Células-Tronco Mesenquimais , Receptores CXCR4/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Vetores Genéticos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Regiões Promotoras Genéticas , Receptores CXCR4/metabolismo , Taxa de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Lung Cancer ; 55(2): 145-56, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17113184

RESUMO

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.


Assuntos
Adenoviridae/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica/métodos , Regiões Promotoras Genéticas , Receptores CXCR4/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Vetores Genéticos , Humanos , Fígado/metabolismo , Neoplasias Pulmonares/genética , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas , Replicação Viral
15.
Gynecol Oncol ; 105(1): 113-21, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17173958

RESUMO

OBJECTIVES: Current virotherapy strategies for ovarian cancer have been hampered by limitations in target cell infectivity and nonspecific tissue replication. In an effort to circumvent these limitations, we evaluated various CRAds modified to incorporate novel capsid targeting motifs (RGD and chimeric Ad5/3) with a novel tissue-specific promoter (CXCR4). METHODS: Two novel CRAds (Ad5-CXCR4-F5/3 and Ad5-CXCR4-RGD) were constructed via homologous recombination and verified by PCR and DNA sequencing. The infectivity and viral replication rates of these two CRAds were analyzed via quantitative real-time PCR (QRT-PCR) in cell line experiments using three ovarian cancer cell lines (SKOV3.ip1, Hey, and OV4) and compared to that achieved with a clinical grade CRAd (delta24-RGD) to be evaluated in a Phase I trial. Cytocidal effects were determined by crystal violet staining in these same cell lines infected with different concentrations of viral particles per cell (0, 0.1, 1, 10, 100, and 500). Additionally, viral replication was evaluated by QRT-PCR in primary ovarian cancer tissue slices from multiple patients with ovarian cancer as well as in primary human normal liver tissue slices in order to establish CRAd selectivity. All experiments incorporated appropriate controls and repeated in triplicate. RESULTS: Compared to RGD-capsid CRAds (delta24-RGD and CXCR4-RGD), the F5/3-capsid CRAd (CXCR4-F5/3) demonstrated significant improvements in infection rates (p=0.025, 0.006, and 0.006) in all ovarian cancer cell lines tested (SKOV3.ip1, Hey, and OV4, respectively). In addition to improved transduction of virus into the cells, the TSP CXCR4-based CRAds demonstrated improved viral replication. Specifically, CXCR4-F5/3 further enhanced viral replication 89-fold (p=0.009, 0.010, 0.003) in the same cancer cell lines. Furthermore, CXCR4-F5/3 showed a 4-log improvement in oncolytic potential over delta24-RGD. In the ex vivo primary ovarian tissue slices, CXCR4-F5/3 showed a 58-fold improvement in viral replication (p=0.005) compared to the clinical grade delta24-RGD. Both CXCR4-F5/3 and CXCR4-RGD demonstrated significant reduction of viral replication in normal liver slices (p=0.001). CONCLUSIONS: These data suggest that a dual targeted approach is feasible for the combined enhancement of infectivity and replication in ovarian cancer with a specificity that was attenuated in normal liver tissues. In fact, CXCR4-F5/3 outperformed our best CRAd agent to date nearly 60-fold in our most stringent ex vivo model of primary ovarian cancer tissue slices and suggests that this novel agent could be useful for the treatment of ovarian cancer.


Assuntos
Adenovírus Humanos/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/virologia , Replicação Viral/fisiologia , Adenovírus Humanos/genética , Adenovírus Humanos/patogenicidade , Capsídeo/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Fígado/virologia , Oligopeptídeos/genética , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Replicação Viral/genética
16.
Int J Cancer ; 120(4): 935-41, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17131341

RESUMO

Conditionally replicative adenoviruses (CRAds) represent novel therapeutic agents that have been recently applied in the context of breast cancer therapy. However, deficiencies in the ability of the adenovirus to infect target tumor cells and to specifically replicate within the tumor target represent key deficiencies preventing the realization of the full potential of this therapeutic approach. Minimal expression of the adenovirus serotype 5 (Ad5) receptor CAR (coxsackie and adenovirus receptor) on breast cancer cells represents a major limitation for Ad5-based virotherapy. Genetic fiber chimerism is a method to alter the tropism of Ad5-based CRAds to achieve CAR-independent infectivity of tumor cells. Here, we describe the use of a CRAd with cancer specific transcriptional control of the essential Ad5 E1A gene using the human CXCR4 gene promoter. We further modified the fiber protein of this agent by switching the knob domain with that of the adenovirus serotype 3. The oncolytic activity of this 5/3 fiber-modified CRAd was studied in breast cancer cell lines, primary breast cancer and human liver tissue slices from patients, and in a xenograft breast cancer mouse model. This infectivity enhanced CRAd agent showed improved replication and killing in breast cancer cells in vitro and in vivo with a remarkable specificity profile that was strongly attenuated in nonbreast cancer cells, as well as in normal human breast and liver tissues. In conclusion, utilization of a CRAd that combined infectivity enhancement strategies and transcriptional targeting improved the CRAd-based antineoplastic effects for breast cancer therapy.


Assuntos
Adenoviridae/fisiologia , Neoplasias da Mama/terapia , Terapia Viral Oncolítica , Regiões Promotoras Genéticas/genética , Receptores CXCR4/genética , Replicação Viral , Proteínas E1A de Adenovirus/genética , Animais , Mama/metabolismo , Mama/patologia , Mama/virologia , Neoplasias da Mama/metabolismo , Linhagem Celular , Proliferação de Células , Derme/metabolismo , Derme/patologia , Derme/virologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/virologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
17.
Am J Pathol ; 169(5): 1833-42, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17071604

RESUMO

Stimulation of Fas-mediated apoptosis has been promoted as a potential therapy for many cancers, including cholangiocarcinoma. We have previously reported that Fas-resistant, but not Fas-sensitive, cholangiocarcinoma cells are tumorigenic in nude mice. The present studies sought to identify molecular targets that promote Fas-mediated apoptosis in cholangiocarcinoma. We found that Fas-resistant cholangiocarcinoma cells exhibited increased constitutive phosphorylation of AKT compared with Fas-sensitive cells. Increased phosphorylation of AKT was also demonstrated in human cholangiocarcinoma tumors and was evident in a mouse xenograft cholangiocarcinoma model. Furthermore, we found that 3,3'-diindolylmethane (DIM), a vegetable autolysis product, promoted Fas-mediated apoptosis of cholangiocarcinoma cells. DIM inhibited phosphorylation of AKT and activation of FLICE-like-inhibitory-protein (FLIP). Inhibition of phosphatidylinositol 3-kinase/AKT decreased FLIP activation and promoted Fas-mediated apoptosis. By contrast, adenovirus-mediated constitutively activated AKT protected cholangiocarcinoma cells from Fas-mediated apoptosis. Decreased activation of extracellular signal-regulated kinase and nuclear factor-kappaB and increased activation of caspase-3, -8, and -9 were associated with inhibition of AKT and FLIP. These results support AKT and FLIP as potential molecular targets and DIM as a potent compound for cholangiocarcinoma intervention.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/antagonistas & inibidores , Colangiocarcinoma/patologia , Indóis/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Receptor fas/metabolismo , Adenoviridae/metabolismo , Animais , Anticarcinógenos/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Caspases/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Genes Dominantes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Pharmacol ; 70(5): 1488-93, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16896072

RESUMO

Delivery of multiple exogenous genes into target cells is important for a broad range of gene therapy applications, including combined therapeutic gene expression and noninvasive imaging. Previous studies ( Mol Ther 4: 223-231, 2001 ) have described the adenoviral vector RGDTKSSTR with a double-expression cassette that encodes herpes simplex virus thymidine kinase (HSVtk) for molecular chemotherapy and human somatostatin receptor subtype-2 (hSSTR2) for indirect imaging. In this vector, both genes are inserted in place of the E1 region of the adenoviral genome and expressed independently from two cytomegalovirus (CMV) promoters. During production of clinical-grade RGDTKSSTR, we found that the CMV promoters and simian virus 40 (SV40) poly(A) regions located in both expression cassettes provoked homologous recombination and deletion of one of the cassettes. To resolve this problem, we designed a strategy for substituting the duplicate promoters and poly(A) regions. We placed the hSSTR2 gene in the new Ad5.SSTR/TK.RGD vector under the control of a CMV promoter with a bovine growth hormone poly(A) region, whereas the SV40 promoter, enhancer, and poly(A) signal controlled HSVtk expression. This use of different regulatory sequences allowed independent expression of both transgenes from a single adenoviral vector and circumvented the recombination problem. Reconstruction of the vector with a double-expression cassette enables its use in human clinical trials.


Assuntos
Adenoviridae/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Recombinação Genética/genética , Adenoviridae/fisiologia , Sequência de Bases , Células Cultivadas , Genoma Viral/genética , Humanos , Peptídeos/química , Reprodutibilidade dos Testes , Deleção de Sequência , Homologia de Sequência , Transgenes
19.
Gynecol Oncol ; 103(3): 985-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16857247

RESUMO

OBJECTIVE: Icodextrin, a novel glucose polymer solution utilized for peritoneal dialysis, has been demonstrated to have prolonged intraperitoneal (IP) instillation volumes in comparison to standard PBS solutions. In an animal model of ovarian cancer, we explored whether a survival advantage exists utilizing icodextrin rather than PBS as a delivery solution for an infectivity enhanced virotherapy approach. METHODS: Initial experiments evaluated whether icodextrin would adversely affect replication of a clinical grade infectivity enhanced conditionally replicative adenovirus (Delta24-RGD). Virus was added to prepared blinded solutions of PBS or icodextrin (20%) and then evaluated in vitro in various human ovarian cancer cell lines (SKOV3.ip1, PA-1, and Hey) and in vivo in a SKOV3.ip1 human ovarian cancer IP murine model. Viral replication was measured by detecting adenovirus E4 gene levels utilizing QRT-PCR. Survival was subsequently evaluated in a separate SKOV3.ip1 ovarian cancer IP murine model. Cohorts of mice were treated in blinded fashion with PBS alone, icodextrin alone, PBS+Delta24-RGD, or icodextrin+Delta24-RGD. Survival data were plotted on Kaplan-Meier curve and statistical calculations performed using the log-rank test. RESULTS: There was no adverse affect of icodextrin on vector replication in the ovarian cancer cell lines nor murine model tumor samples evaluated. Median survival in the IP treated animal cohorts was 23 days for the PBS group, 40 days for the icodextrin group, 65 days for the PBS+Delta24-RGD group, and 105 days for icodextrin+Delta24-RGD (p=0.023). Of note, 5 of the 10 mice in the icodextrin+Delta24-RGD group were alive at the end of the study period, all without evidence of tumor (120 days). CONCLUSIONS: These experiments suggest that the use of dialysates such as icodextrin may further enhance the therapeutic effects of novel IP virotherapy and other gene therapy strategies for ovarian cancer. Phase I studies utilizing icodextrin-based virotherapy for ovarian cancer are currently in development.


Assuntos
Soluções para Diálise/uso terapêutico , Terapia Genética , Glucanos/uso terapêutico , Glucose/uso terapêutico , Neoplasias Ovarianas/terapia , Animais , Soluções para Diálise/administração & dosagem , Modelos Animais de Doenças , Feminino , Glucanos/administração & dosagem , Glucose/administração & dosagem , Icodextrina , Infusões Parenterais , Camundongos , Neoplasias Ovarianas/patologia
20.
J Neurosurg ; 104(4): 583-92, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16619663

RESUMO

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.


Assuntos
Neoplasias Encefálicas/genética , Marcação de Genes , Terapia Genética/métodos , Glioma/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica/genética , Adenoviridae , Animais , Apoptose/genética , Linhagem Celular Tumoral , Feminino , Vetores Genéticos , Humanos , Proteínas Inibidoras de Apoptose , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Survivina , Transplante Heterólogo , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...