Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 283
Filtrar
1.
Cancer Gene Ther ; 22(4): 222, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25904493

RESUMO

Correction to: Cancer Gene Therapy (2013) 20, 445­452; doi:10.1038/cgt.2013.39; published online 9 August 2013. Laura Timares was unintentionally left off the list of authors. She should have been listed along with her affiliation, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA. Her NIH funding sources, R21AR055252 and P30AR050948 should also have been listed. The authors regret the errors.

2.
Cancer Gene Ther ; 22(4): 172-80, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25721205

RESUMO

Radiation therapy is a critical component of cancer treatment with over half of patients receiving radiation during their treatment. Despite advances in image-guided therapy and dose fractionation, patients receiving radiation therapy are still at risk for side effects due to off-target radiation damage of normal tissues. To reduce normal tissue damage, researchers have sought radioprotectors, which are agents capable of protecting tissue against radiation by preventing radiation damage from occurring or by decreasing cell death in the presence of radiation damage. Although much early research focused on small-molecule radioprotectors, there has been a growing interest in gene therapy for radioprotection. The amenability of gene therapy vectors to targeting, as well as the flexibility of gene therapy to accomplish ablation or augmentation of biologically relevant genes, makes gene therapy an excellent strategy for radioprotection. Future improvements to vector targeting and delivery should greatly enhance radioprotection through gene therapy.


Assuntos
Terapia Genética , Neoplasias/radioterapia , Lesões por Radiação/prevenção & controle , Tolerância a Radiação/genética , Animais , Expressão Gênica , Humanos , Lesões por Radiação/genética , Transgenes
3.
Cancer Gene Ther ; 21(10): 427-33, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25213663

RESUMO

Increased expression of sialyl Lewis(x/a) carbohydrates, ligands for E-selectin, correlates with clinically advanced stages and metastasis of gastric and colon cancers. In contrast, Sd(a) carbohydrate is abundantly detected in the normal gastrointestinal mucosa but dramatically reduced or lost in cancer tissues. A glycosyltransferase, ß1,4N-acetylgalactosaminyltransferase 2 (B4GALNT2) that catalyzes Sd(a) carbohydrate synthesis, is silenced in cancer. In the present study, we aimed at reducing the expression of sialyl Lewis(x/a) of cancer cells in vivo by forced expression of B4GALNT2 and Sd(a), thereby preventing dissemination/metastasis, especially metastasis triggered by surgical maneuvers. We used a fiber-modified adenovirus (Ad) vector that contained a chimeric construct with a serotype 5 shaft and a serotype 3 knob. Using this Ad5/3 vector, we successfully introduced the B4GALNT2 gene into a human gastric cancer cell line KATO III in vitro and confirmed replacement of sialyl Lewis(x) to Sd(a) with a decrease in E-selectin-dependent adhesion. Administration of Ad5/3-B4GALNT2 vectors into the peritoneal cavity of mice after inoculation of KATO III cells with laparotomy significantly reduced the incidence of metastasis. Our results indicate that the transfer of a single gene encoding B4GALNT2 modified carbohydrate chains of cancer cells in vivo and decreased tumor dissemination and metastasis.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Glicosiltransferases/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Animais , Proteínas do Capsídeo/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Selectina E/metabolismo , Ativação Enzimática , Expressão Gênica , Glicosiltransferases/metabolismo , Humanos , Masculino , Camundongos , Metástase Neoplásica , Peritônio/patologia , Neoplasias Gástricas/metabolismo , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Gene Ther ; 21(7): 264-74, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24903014

RESUMO

Pancreatic cancer is a significant clinical problem and novel therapeutic approaches are desperately needed. Recent advances in conditionally replicative adenovirus-based (CRAd) oncolytic virus design allow the application of CRAd vectors as a therapeutic strategy to efficiently target and eradicate chemoresistant pancreatic cancer cells, thereby improving the efficacy of pancreatic cancer treatment. The goal of this study was to construct and validate the efficacy of an infectivity-enhanced, liver-untargeted, tumor-specific CRAd vector. A panel of CRAds has been derived that embodies the C-X-C chemokine receptor type 4 promoter for conditional replication, two-fiber complex mosaicism for targeting expansion and hexon hypervariable region 7 (HVR7) modification for liver untargeting. We evaluated CRAds for cancer virotherapy using a human pancreatic tumor xenograft model. Employment of the fiber mosaic approach improved CRAd replication in pancreatic tumor xenografts. Substitution of the HVR7 of the Ad5 hexon for Ad serotype 3 hexon resulted in decreased liver tropism of systemically administrated CRAd. Obtained data demonstrated that employment of complex mosaicism increased efficacy of the combination of oncolytic virotherapy with chemotherapy in a human pancreatic tumor xenograft model.


Assuntos
Adenoviridae/genética , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/virologia , Receptores CXCR4/genética , Animais , Feminino , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Distribuição Aleatória , Receptores CXCR4/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
5.
Cancer Gene Ther ; 20(8): 445-52, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23928733

RESUMO

Adenoviruses are currently used in a variety of bench and bedside applications. However, their employment in gene delivery to lymphocyte lineages is hampered by the lack of coxsackie virus and adenovirus receptor (CAR) on the cell surface. Exploitation of an alternative receptor on the surface of T lymphocytes can allow for utilization of adenovirus in a variety of T lymphocyte-based diseases and therapies. Here, we describe how resistance to infection can be overcome by the utilization of a bi-specific fusion protein, soluble CAR murine interleukin 2 (sCAR-mIL-2), that retargets adenovirus to the murine IL-2 receptor (IL-2R). Infection of a murine T-cell line, CTLL-2, with a sCAR-mIL-2/Adenovirus conjugate provided a ninefold increase in both green fluorescence protein-positive cells and luciferase expression. In addition, this increase in infection was also seen in isolated primary murine T lymphocytes. In this context, the sCAR-mIL-2 adapter provided a fourfold gene transduction increase in activated primary murine T lymphocytes. Our results show that recombinant sCAR-mIL-2 fusion protein promotes IL-2R-targeted gene transfer to murine T lymphocytes and that alternative targeting can abrogate their native resistance to infection.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Receptores de Interleucina-2/metabolismo , Linfócitos T/fisiologia , Linfócitos T/virologia , Adenoviridae/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-2/biossíntese , Receptores de Interleucina-2/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/metabolismo , Transdução Genética
6.
Gynecol Obstet Invest ; 76(2): 119-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23920223

RESUMO

Postoperative abdominal/pelvic peritoneal adhesions are a major source of morbidity (bowel obstruction, infertility, ectopic gestation as well as chronic pelvic pain) in women. In this study, we screened various transduction and transcription modifications of adenovirus (Ad) to identify those that support maximal Ad-mediated gene delivery to human adhesion fibroblasts, which in turn would enhance the efficacy of this novel treatment/preventative strategy for postoperative adhesions. We transduced primary cultures of human peritoneal adhesion fibroblasts with fiber-modified Ad vectors Ad5-RGD-luc, Ad5-Sigma-luc, Ad5/3-luc and Ad5-CAV2-luc as well as transcriptional targeting viruses Ad5-survivin-luc, Ad5-heparanase-luc, Ad5-mesothelin (MSLN)-CRAd-luc and Ad5-secretory leukoprotease inhibitor (SLPI)-luc, and compared their activity to wild-type Ad5-luc. At 48 h, luciferase activity was measured and normalized to the total protein content in the cells. Among the fiber-modified Ad vectors, Ad5-Sigma-luc and among the transcriptional targeting modified Ad vectors, Ad5-MSLN-CRAd-luc showed significantly increased expression levels of luciferase activity at 5, 10 and 50 plaque forming units/cell in adhesion fibroblast cells compared with wild-type Ad5-luc (p < 0.05). Specific modifications of Ad improve their gene delivery efficiency towards human peritoneal adhesion fibroblasts. Developing a safe localized method to prevent/treat postoperative adhesion formation would have a major impact on women health.


Assuntos
Adenoviridae/genética , Fibroblastos , Terapia Genética , Aderências Teciduais/terapia , Transdução Genética , Células Cultivadas , Fibroblastos/enzimologia , Expressão Gênica , Vetores Genéticos , Humanos , Luciferases/genética , Luciferases/metabolismo , Mesotelina , Aderências Teciduais/prevenção & controle , Transcrição Gênica
7.
Hum Reprod ; 28(9): 2398-406, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23820419

RESUMO

STUDY QUESTION: Is targeted adenovirus vector, Ad-SSTR-RGD-TK (Adenovirus -human somatostatin receptor subtype 2- arginine, glycine and aspartate-thymidine kinase), given in combination with ganciclovir (GCV) against immortalized human leiomyoma cells (HuLM) a potential therapy for uterine fibroids? SUMMARY ANSWER: Ad-SSTR-RGD-TK/GCV, a targeted adenovirus, effectively reduces cell growth in HuLM cells and to a significantly greater extent than in human uterine smooth muscle cells (UtSM). WHAT IS KNOWN ALREADY: Uterine fibroids (leiomyomas), a major cause of morbidity and the most common indication for hysterectomy in premenopausal women, are well-defined tumors, making gene therapy a suitable and potentially effective non-surgical approach for treatment. Transduction of uterine fibroid cells with adenoviral vectors such as Ad-TK/GCV (herpes simplex virus thymidine kinase gene) decreases cell proliferation. STUDY DESIGN, SIZE, DURATION: An in vitro cell culture method was set up to compare and test the efficacy of a modified adenovirus vector with different multiplicities of infection in two human immortalized cell lines for 5 days. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immortalized human leiomyoma cells and human uterine smooth muscle cells were infected with different multiplicities of infection (MOI) (5-100 plaque-forming units (pfu)/cell) of a modified Ad-SSTR-RGD-TK vector and subsequently treated with GCV. For comparison, HuLM and UtSM cells were transfected with Ad-TK/GCV and Ad-LacZ/GCV. Cell proliferation was measured using the CyQuant assay in both cell types. Additionally, western blotting was used to assess the expression of proteins responsible for regulating proliferation and apoptosis in the cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transduction of HuLM cells with Ad-SSTR-RGD-TK/GCV at 5, 10, 50 and 100 pfu/cell decreased cell proliferation by 28, 33, 45, and 84%, respectively (P < 0.05) compared with untransfected cells, whereas cell proliferation in UtSM cells transfected with the same four MOIs of Ad-SSTR-RGD-TK/GCV compared with that of untransfected cells was decreased only by 8, 23, 25, and 28%, respectively (P < 0.01). Western blot analysis showed that, in comparison with the untargeted vector Ad-TK, Ad-SSTR-RGD-TK/GCV more effectively reduced expression of proteins that regulate the cell cycle (Cyclin D1) and proliferation (PCNA, Proliferating Cell Nuclear Antigen), and it induced expression of the apoptotic protein BAX, in HuLM cells. LIMITATIONS, REASONS FOR CAUTION: Results from this study need to be replicated in an appropriate animal model before testing this adenoviral vector in a human trial. WIDER IMPLICATIONS OF THE FINDINGS: Effective targeting of gene therapy to leiomyoma cells enhances its potential as a non-invasive treatment of uterine fibroids.


Assuntos
DNA Recombinante/metabolismo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/metabolismo , Leiomioma/metabolismo , Miométrio/metabolismo , Transdução Genética , Neoplasias Uterinas/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Adenoviridae/patogenicidade , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , DNA Recombinante/efeitos adversos , DNA Recombinante/uso terapêutico , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/uso terapêutico , Humanos , Leiomioma/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Uterinas/terapia
8.
Gene Ther ; 20(7): 733-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23171918

RESUMO

Specific and efficient gene delivery to the lung has been hampered by liver sequestration of adenovirus serotype 5 (Ad5) vectors. The complexity of Ad5 liver tropism has largely been unraveled, permitting improved efficacy of Ad5 gene delivery. However, Kupffer cell (KC) scavenging and elimination of Ad5 still represent major obstacles to lung gene delivery strategies. KC uptake substantially reduces bioavailability of Ad5 for target tissues and compensatory dose escalation leads to acute hepatotoxicity and a potent innate immune response. Here, we report a novel lung-targeting strategy through redirection of Ad5 binding to the concentrated leukocyte pool within the pulmonary microvasculature. We demonstrate that this leukocyte-binding approach retargets Ad5 specifically to lung endothelial cells and prevents KC uptake and hepatocyte transduction, resulting in 165,000-fold enhanced lung targeting, compared with Ad5. In addition, myeloid cell-specific binding is preserved in single-cell lung suspensions and only Ad.MBP-coated myeloid cells achieved efficient endothelial cell transduction ex vivo. These findings demonstrate that KC sequestration of Ad5 can be prevented through more efficient uptake of virions in target tissues and suggest that endothelial transduction is achieved by leukocyte-mediated 'hand-off' of Ad.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Terapia Genética , Células Mieloides/citologia , Tropismo Viral , Células Endoteliais/citologia , Células Endoteliais/virologia , Vetores Genéticos , Hepatócitos/citologia , Hepatócitos/virologia , Humanos , Células de Kupffer/citologia , Células de Kupffer/virologia , Fígado/citologia , Fígado/virologia , Pulmão/citologia , Pulmão/virologia , Células Mieloides/virologia , Transdução Genética
9.
Hum Reprod ; 25(8): 2068-83, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20573677

RESUMO

BACKGROUND: Novel therapeutic approaches for endometriosis based on molecular strategies may prove to be useful. Conditionally replicative adenoviruses (CRAds) are designed to exploit key differences between target and normal cells. The wild-type adenovirus (Adwt) promoter can be replaced by tissue-specific promoters, allowing viral replication only in target cells. Viral infectivity can be enhanced by altering Ad tropism via fiber modification. We investigated whether CRAds can be used to target endometriosis and determined the most efficient transcriptional- and transductional-targeting strategy. METHODS: An in vitro study was carried out using human endometriotic cell lines, 11Z (epithelial) and 22B (stromal), normal human ovarian surface epithelial cell line (NOSE006) and primary human endometriosis cells. A total of 9 promoters and 12 Ad tropism modifications were screened by means of a luciferase reporter assay. From this screening data, three CRAds (CRAd-S-pK7, CRAd-S-RGD, CRAd-S-F5/3sigma1, all incorporating the survivin promoter but with different fiber modifications) were selected to perform experiments using Adwt and a replication-deficient virus as controls. CRAds were constructed using a plasmid recombination system. Viral-binding capacity, rates of entry and DNA replication were evaluated by quantitative real-time PCR of viral genome copy. Cell-killing effects were determined by crystal violet staining and a cell viability assay for different concentrations of viral particles per cell. RESULTS: Comparison of promoters demonstrated that the survivin promoter exhibited the highest induction in both endometriotic cell lines. Among the fiber-modified viruses, the polylysine modification (pK7) showed the best infection enhancement. CRAd-S-pK7 was validated as the optimal CRAd to target endometriosis in terms of binding ability, entry kinetics, DNA replication and cell-killing effect. CRAd-S-pK7 also exhibited a high level of DNA replication in primary endometriosis cells. CONCLUSIONS: CRAd-S-pK7 has the best infection and cell-killing effect in the context of endometriosis. It could prove to be a useful novel method to target refractory cases of endometriosis.


Assuntos
Adenoviridae/genética , Endometriose/terapia , Replicação Viral , Linhagem Celular , Endometriose/genética , Feminino , Humanos , Regiões Promotoras Genéticas , Ligação Viral , Replicação Viral/genética
10.
Cancer Gene Ther ; 17(7): 447-56, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20150932

RESUMO

Gene therapy is being examined as a potential strategy for treating prostate cancer. Serotype 5 adenovirus (Ad.5) is routinely used as a vector for transgene delivery. However, the infectivity of Ad.5 is dependent on Coxsackie-adenovirus receptors (CARs); many tumor types show a reduction in this receptor in vivo, thereby limiting therapeutic gene transduction. Serotype chimerism is one approach to circumvent CAR deficiency; this strategy is used to generate an Ad.5/3-recombinant Ad that infects cancer cells through Ad.3 receptors in a CAR-independent manner. In this report, the enhanced transgene delivery and efficacy of Ad.5/3-recombinant virus was evaluated using an effective wide-spectrum anticancer therapeutic melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24). Our data show that in low CAR human prostate cancer cells (PC-3), a recombinant Ad.5/3 virus delivering mda-7/IL-24 (Ad.5/3-mda-7) is more efficacious than an Ad.5 virus encoding mda-7/IL-24 (Ad.5-mda-7) in infecting tumor cells, expressing MDA-7/IL-24 protein, inducing cancer-specific apoptosis, inhibiting in vivo tumor growth and exerting an antitumor 'bystander' effect in a nude mouse xenograft model. Considering the fact that Ad.5-mda-7 has shown significant objective responses in a phase I clinical trial for solid tumors, Ad.5/3-mda-7 is predicted to exert enhanced therapeutic benefit in patients with prostate cancer.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Interleucinas/genética , Neoplasias da Próstata/terapia , Receptores Virais/metabolismo , Animais , Linhagem Celular Tumoral , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Humanos , Imuno-Histoquímica , Interleucinas/biossíntese , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Transgenes , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Biol Ther ; 7(5): 786-93, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18756624

RESUMO

Malignant glioma continues to be a major target for gene therapy and virotherapy due to its aggressive growth and the current lack of effective treatment. However, these approaches have been hampered by inefficient infection of glioma cells by viral vectors,particularly vectors derived from serotype 5 adenoviruses (Ad5). This results from limited cell surface expression of the primary adenovirus receptor, coxsackie-adenovirus-receptor (CAR), on tumor cells. To circumvent this problem, Ad fiber pseudotyping,the genetic replacement of either the entire fiber or fiber knob domain with its structural counterpart from another human Ad serotype that recognizes a cellular receptor other than CAR, has been shown to enhance Ad infectivity in a variety of tumor types,including human glioma. Here, we have extended the paradigm of genetic pseudotyping to include fiber domains from non-human or"xenotype" Ads for infectivity enhancement of human glioma cell populations. In this study, we evaluated the gene transfer efficiency of a panel of Ad vectors which express one of five different "xenotype"fiber knob domains, including those derived from murine,ovine, porcine and canine species, in both human glioma cell lines as well as primary glioma tumor cells from patients. Adenovirus vectors displaying either canine Ad or porcine Ad fiber elements had the highest gene transfer to both glioma cell lines and primary tumor cells. The correlation between the viral infectivity of modified adenovirus vectors and expression of human CAR and CD46(an adenovirus type B receptor) on the surfaces of tumor cells was also analyzed. Taken together, human adenovirus vectors modified with "xenotype" fiber elements could be excellent candidates to target human glioma.


Assuntos
Adenoviridae/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Animais , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Citomegalovirus/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/metabolismo , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Vírus/metabolismo
12.
Gene Ther ; 15(10): 716-29, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18369326

RESUMO

Use of cells as therapeutic carriers has increased in the past few years and has developed as a distinct concept and delivery method. Cell-based vehicles are particularly attractive for delivery of biotherapeutic agents that are difficult to synthesize, have reduced half-lives, limited tissue penetrance or are rapidly inactivated upon direct in vivo introduction. Initial studies using cell-based approaches served to identify some of the key factors for the success of this type of therapeutic delivery. These factors include the efficiency of cell loading with a therapeutic payload, the means of cell loading and the nature of therapeutics that cells can carry. However, one important aspect of cell-based delivery yet to be fully investigated is the process of actual delivery of the cell payload in vivo. In this regard, the potential ability of cell carriers to provide site-specific or targeted delivery of therapeutics deserves special attention. The present review focuses on a variety of targeting approaches that may be utilized to improve cell-based therapeutic delivery strategies. The different aspects of targeting that can be applied to cell vehicles will be discussed, including physical methods for directing cell distribution, intrinsic cell-mediated homing mechanisms and the feasibility of engineering cells with novel targeting mechanisms. Development of cell targeting strategies will further advance cell vehicle applications, broaden the applicability of this delivery approach and potentiate therapeutic outcomes.


Assuntos
Transplante de Células/métodos , Terapia Genética/métodos , Neoplasias/terapia , Movimento Celular , Proliferação de Células , Humanos , Neoplasias/patologia
13.
Cancer Gene Ther ; 15(5): 293-302, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18323853

RESUMO

The prognosis and response to conventional therapies of malignant melanoma inversely correlate with disease progression. With increasing thickness, melanomas acquire metastatic potential and become inherently resistant to radiotherapy and chemotherapy. These harsh realities mandate the design of improved therapeutic modalities, especially those targeting metastases. To develop an approach to effectively treat this aggressive disease, we constructed a conditionally replication-competent adenovirus in which expression of the adenoviral E1A gene, necessary for replication, is driven by the cancer-specific promoter of progression-elevated gene-3 (PEG-3) and which simultaneously expresses mda-7/IL-24 in the E3 region of the adenovirus (Ad.PEG-E1A-mda-7), a cancer terminator virus (CTV). This CTV produces large quantities of MDA-7/IL-24 protein as a function of adenovirus replication uniquely in cancer cells. Infection of Ad.PEG-E1A-mda-7 (CTV) in normal human immortal melanocytes and human melanoma cells demonstrates cancer cell-selective adenoviral replication, mda-7/IL-24 expression, growth inhibition and apoptosis induction. Injecting Ad.PEG-E1A-mda-7 CTV into xenografts derived from MeWo human metastatic melanoma cells in athymic nude mice completely eliminated not only primary treated tumors but also distant non-treated tumors (established in the opposite flank), thereby implementing a cure. These provocative findings advocate potential therapeutic applications of this novel virus for treating patients with advanced melanomas with metastases.


Assuntos
Adenoviridae/genética , Adenoviridae/isolamento & purificação , Melanoma/genética , Melanoma/virologia , Regiões Terminadoras Genéticas/genética , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/genética , Animais , Progressão da Doença , Terapia Genética/métodos , Humanos , Melanoma/patologia , Camundongos , Camundongos Nus , Metástase Neoplásica/genética , Metástase Neoplásica/prevenção & controle , Transplante de Neoplasias , Transplante Heterólogo , Replicação Viral
14.
Hum Reprod ; 23(3): 514-24, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18184643

RESUMO

BACKGROUND: To circumvent the paucity of the primary adenovirus (Ad5) receptor and the non-specific Ad5 tropism in the context of uterine leiomyoma cells, Ad5 modification strategies would be beneficial. METHODS: We screened several modified adenoviruses to identify the most efficient and selective virus toward human leiomyoma cells to be used as candidate for delivering therapeutic genes. We propagated: wild-type Ad5-luc, fiber-modified viruses: ad5 RGD-luc, Ad5-Sigma-luc, Ad5/3-luc and Ad5-CAV2-luc, as well as transcriptional targeted viruses: ad5 survivin-luc, Ad5-heparanase-luc, Ad5-MSLN-CRAD-luc and Ad5-SLPI-luc, on 293 cells and purified them by double CsCL density centrifugation. Then we transfected primary cultures of human leiomyoma cells derived from fibroids of four different patients, telomerase-immortalized human leiomyoma cell line (huLM), telomerase-immortalized normal human myometrial cell line (HM9) and immortalized normal human liver cells (THLE3) with the viruses at 5, 10 and 50 plaque-forming units (PFU)/cell. After 48 h, luciferase activities were measured and normalized to the total cellular protein content. RESULTS: Ad5-RGD-luc and Ad5-CAV2-luc, Ad5-SLPI-luc and Ad5-MSLN-CRAD-luc at 5, 10 and 50 pfu/cell showed significantly higher expression levels of luciferase activity in both primary and immortalized human leiomyoma cells when compared with Ad5-Luc. Additionally, these modified viruses demonstrated selectivity toward leiomyoma cells, compared with myometrial cells and exhibited lower liver cell transduction, compared with Ad5-luc, at the same dose levels. CONCLUSIONS: Ad5-CAV2-luc, Ad5-RGD-luc, Ad5-SLPI-luc and Ad5-MSLN-CRAD-luc are promising delivery vehicles in the context of leiomyoma gene therapy.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Leiomioma/terapia , Leiomioma/virologia , Feminino , Humanos , Fígado/citologia , Mesotelina , Miométrio/citologia , Miométrio/virologia , Receptores Virais/genética
15.
Cancer Gene Ther ; 15(2): 94-100, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18157147

RESUMO

Irinotecan (CPT-11) is a key drug for the treatment of various cancers. CPT-11 can be considered to be a prodrug, since it needs to be activated into the toxic drug SN-38 by the enzyme carboxylesterase. However, CPT-11 may induce severe diarrhea and bone marrow suppression as adverse effects, thus leading to treatment interruption. The tumor-specific activation of CPT-11 is a possible strategy to avoid the severe toxicities by reducing the serum concentration of CPT-11. In this study, we constructed human liver carboxylesterase-2 fused with anticarcinoembryonic antigen (CEA) scFv as a targeting molecule. The recombinant enzyme anchors onto the tumor cell surface CEA, and thus metabolize CPT-11 extracellularly. In addition a secreted tumor-targeted form of carboxylesterase should help prevent the leakage of the enzyme from the site of the tumor into the circulation. This fusion protein showed CPT-11 activation to SN-38 and specific binding to CEA-expressing cells. In combination with CPT-11, the recombinant carboxylesterase protein exerted antiproliferative effects on human cancer cells. This recombinant enzyme is, therefore, a promising new tool in enzyme prodrug therapy for the treatment of carcinoma with CPT-11.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Carboxilesterase/genética , Antígeno Carcinoembrionário/genética , Região Variável de Imunoglobulina/genética , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/toxicidade , Camptotecina/farmacocinética , Camptotecina/farmacologia , Camptotecina/toxicidade , Carboxilesterase/antagonistas & inibidores , Carboxilesterase/metabolismo , Antígeno Carcinoembrionário/imunologia , Antígeno Carcinoembrionário/metabolismo , Relação Dose-Resposta Imunológica , Marcação de Genes , Células HeLa , Humanos , Região Variável de Imunoglobulina/fisiologia , Irinotecano , Proteínas Recombinantes de Fusão/genética
16.
Gene Ther ; 15(4): 298-308, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18046426

RESUMO

CD40 ligation has been shown to promote antigen-presenting functions of dendritic cells, which express CD40 receptor. Here we reported significantly altered biodistribution and immune responses with the use of CD40-targeted adenovirus. Compared with unmodified adenovirus 5, the CD40-targeted adenovirus following intravenous administration (i.v.) resulted in increased transgene expressions in the lung and thymus, which normally do not take up significant amounts of adenovirus. Intradermal injection saw modified adenovirus being mainly processed in local draining lymph nodes and skin. Following intranasal administration (i.n.), neither unmodified nor targeted viruses were found to be in the liver or spleen, which predominantly took up the virus following i.v. administration. However, inadvertent infection of the brain was found with unmodified adenoviruses, with the second highest gene expression among 14 tissues examined. Importantly, such undesirable effects were largely ablated with the use of targeted vector. Moreover, the targeted adenovirus elicited more sustained antigen-specific cellular immune responses (up to 17-fold) at later time points (30 days post boosting), but also significantly hampered humoral responses irrespective of administration routes. Additional data suggest the skewed immune responses induced by the targeted adenoviruses were not due to the identity of the transgene but more likely a combination of overall transgene load and CD40 stimulation.


Assuntos
Adenoviridae/genética , Antígenos CD40/genética , Células Dendríticas/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Distribuição Tecidual , Transgenes
17.
Gene Ther ; 14(20): 1448-60, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17700705

RESUMO

Targeting of gene transfer at the level of cell entry is one of the most attractive challenges in vector development. However, attempts to redirect adenovirus vectors to alternative receptors by engineering the capsid-coding region have shown limited success because proper targeting ligand-receptor systems on the cells of interest are generally unknown. Systematic approaches to generate adenovirus vectors targeting any given cell type need to be developed to achieve this goal. Here, we constructed an adenovirus library that was generated by a Cre-lox-mediated in vitro recombination between an adenoviral fiber-modified plasmid library and genomic DNA to display random peptides on a fiber knob. As proof of concept, we screened the adenovirus display library on a glioma cell line and observed selection of several particular peptide sequences. The targeted vector carrying the most frequently isolated peptide significantly enhanced gene transduction in the glioma cell line but not in many other cell lines. Because the insertion of a pre-selected peptide into a fiber knob often fails to generate an adenovirus vector, the selection of targeting peptides is highly useful in the context of the adenoviral capsid. This vector-screening system can facilitate the development of a targeted adenovirus vector for a variety of applications in medicine.


Assuntos
Adenoviridae/genética , Proteínas do Capsídeo/genética , DNA Viral/genética , Vetores Genéticos/genética , Biblioteca de Peptídeos , Animais , Reatores Biológicos , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Engenharia Genética , Glioma/genética , Humanos , Camundongos , Camundongos Nus , Plasmídeos , Recombinação Genética , Transdução Genética/métodos , Transfecção/métodos
18.
Cancer Gene Ther ; 14(4): 421-30, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17235353

RESUMO

Oncolytic viruses represent a novel cancer treatment strategy. Despite their promising preclinical data, however, corresponding clinical trials have disappointed. To aid preclinical analyses, we hypothesized that three-dimensional tumor cell clusters or spheroids might provide an assay system superior to conventional monolayer cell cultures. Spheroids show viral infection, replication and oncolytic patterns distinct from conventional monolayer assays. Therefore, viral tumor penetration and oncolysis measurements may be improved with such three-dimensional models. Also, preclinical analyses of oncolytic viruses frequently measure mitochondrial activity, but more accurate measures of oncolysis might involve quantitation of intracellular protein release. Therefore, we measured luciferase released from luciferase-expressing spheroids and found unique patterns that maintained consistency with various viruses and doses. The relative variations between viruses and doses may represent temporal differences in oncolysis dynamics. Analysis of five recombinant replicative adenoviruses with promise for clinical application showed that Ad5/3-Delta24 produced the most luciferase release 1 week after infection and achieved the earliest and highest peak luciferase release level. Ad5/3-Delta24 also effected the earliest subtotal spheroid cell death. These findings closely parallel monolayer oncolysis assays with these agents. Therefore, the luciferase-expressing tumor spheroid assay represents a promising three-dimensional model for preclinical analysis of replicative oncolytic agents.


Assuntos
Adenoviridae/fisiologia , Bioensaio , Luciferases/análise , Vírus Oncolíticos/fisiologia , Replicação Viral , Adenoviridae/genética , Humanos , Luciferases/genética , Vírus Oncolíticos/genética , Esferoides Celulares/virologia , Células Tumorais Cultivadas
19.
Gene Ther ; 14(8): 627-37, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17251987

RESUMO

Adenovirus (Ad)-based vectors are useful gene delivery vehicles for a variety of applications. Despite their attractive properties, many in vivo applications require modulation of the viral tropism. Targeting approaches applied to adenoviral vectors included genetic modification of the viral capsid, controlled expression of the transgene and combinatorial approaches that combine two or more targeting elements in single vectors. Most of these studies confirmed successful retargeting in cell cultures, however, in vivo gains of targeted adenoviral vectors have not been widely demonstrated. We have developed a combinatorial retargeting approach utilizing metabolically biotinylated Ad, where the biotin acceptor peptide was incorporated in one of the fibers in a dual fiber viral particle resulting in metabolically biotinylated fiber-mosaic Ad (mBfMAd). We have utilized this vector in complex with epidermal growth factor (EGF)-Streptavidin to retarget fiber-mosaic virus to EGF receptor (EGFR) expressing cells in vitro and confirmed an increased infectivity of the retargeting complex. Most importantly, the utility of this strategy was demonstrated in vivo in two distinct animal models. In both models tested, retargeted mBfMAd demonstrated an increased ratio of gene expression in target tissues compared to the liver expression profile. Thus, metabolically biotinylated fiber-mosaic virus in combination with appropriate adapters can be successfully exploited for adenoviral retargeting strategies.


Assuntos
Adenoviridae/genética , Receptores ErbB/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Neoplasias/terapia , Transdução Genética/métodos , Adenoviridae/metabolismo , Animais , Biotinilação , Linhagem Celular Tumoral , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Fator de Crescimento Epidérmico/genética , Feminino , Marcação de Genes/métodos , Vetores Genéticos/genética , Humanos , Luciferases/genética , Luciferases/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Neoplasias/metabolismo , Receptores Virais/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Estreptavidina/genética , Transgenes
20.
Cancer Gene Ther ; 14(1): 105-16, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17024232

RESUMO

Treatment of advanced lung cancer is one of the major challenges in current medicine because of the high morbidity and mortality of the disease. Advanced stage lung cancer is refractory to conventional therapies and has an extremely poor prognosis. Thus, new therapeutic approaches are needed. Lung tumor formation depends on angiogenesis in which the vascular endothelial growth factor (VEGF) produced by cancer cells plays a pivotal role. Neutralizing VEGF with a soluble VEGF receptor suppresses tumor growth; however, the anticancer effect with this therapy is weakened after the intratumoral vascular network is completed. In this study, we turned the expression of VEGF by tumors to therapeutic advantage using a conditionally replication-competent adenovirus (CRAd) in which the expression of E1 is controlled by the human VEGF promoter. This virus achieved good levels of viral replication in lung cancer cells and induced a substantial anticancer effect in vitro and in vivo. As a further enhancement, the cancer cell killing effect was improved with tropism modification of the virus to express the knob domain of Ad3, which improved infectivity for cancer cells. These VEGF promoter-based CRAds also showed a significant cell killing effect for various types of cancer lines other than lung cancer. Conversely, the VEGF promoter has low activity in normal tissues, and the CRAd caused no damage to normal bronchial epithelial cells. Since tumor-associated angiogenesis via VEGF signalling is common in many types of cancers, these CRAds may be applicable to a wide range of tumors. We concluded that VEGF promoter-based CRAds have the potential to be an effective strategy for cancer treatment.


Assuntos
Adenoviridae/genética , Terapia Genética , Neoplasias/terapia , Regiões Promotoras Genéticas , Fator A de Crescimento do Endotélio Vascular/genética , Replicação Viral , Adenoviridae/fisiologia , Sequência de Bases , Primers do DNA , Humanos , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA