首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
2.
目的构建表达超抗原金黄色葡萄球菌肠毒素A(SEA)基因的条件增殖型溶瘤腺病毒pPE3-SEA。方法将已构建好的携带SEA基因的非增殖溶瘤腺病毒载体质粒pDC318-SEA经SpeⅠ和SalⅠ双酶切后,将酶切下的超抗原SEA基因片段连接到同样经SpeⅠ和SalⅠ双酶切后的增殖型溶瘤腺病毒穿梭质粒pENTR12中,将鉴定正确的携带SEA基因增殖溶瘤腺病毒载体命名为pENTR12-SEA。增殖溶瘤腺病毒载体pENTR12-SEA与病毒骨架质粒pPE3-ccdB重组,通过Lipofectamine2000共转染HEK293细胞,经同源重组产生重组腺病毒pPE3-SEA。结果应用PCR验证、双酶切分析、序列测定表明,成功构建了携带SEA基因增殖型溶瘤腺病毒载体pENTR12-SEA,通过同源重组成功获得携带超抗原SEA基因片段的pPE3-SEA增殖型溶瘤腺病毒,且病毒滴度为2.5×1010pfu/ml。结论成功构建了表达超抗原SEA基因的条件增殖型溶瘤腺病毒pPE3-SEA,为以后进一步研究该病毒对肿瘤靶向治疗的作用提供了实验基础。  相似文献   

3.
4.
Abstract Prostate cancer (PCa) is the second most commonly diagnosed and sixth leading cause of cancer death in American men and one for which no curative therapy exists after metastasis. To meet this need for novel therapies, our laboratory has previously generated conditionally replicating adenovirus (CRAd) vectors expressing the sodium iodide symporter (hNIS). This virus transduced PCa cells and induced functional NIS expression, allowing for noninvasive tumor imaging and combination therapy with radioiodide, referred to as radiovirotherapy. We have now generated two new modified vectors to further improve efficacy. Ad5/3PB-ADP-hNIS and Ad5/3PB-hNIS include a hybrid Ad5/3 fiber knob to improve transduction efficiency, and express NIS from the endogenous major late promoter to restrict NIS expression to target cells. Additionally, Ad5/3PB-ADP-hNIS includes the adenovirus death protein (ADP), which hastens the release of viral particles after assembly. These two vectors specifically induce radioisotope uptake, cytopathic effect, and viral replication in androgen receptor-expressing PCa cell lines with Ad5/3PB-ADP-hNIS showing earlier (131)I uptake and cytolysis at low multiplicity of infection. SPECT-CT imaging of xenograft tumors infected with Ad5/3PB-hNIS showed steady uptake, whereas infection with Ad5/3PB-ADP-hNIS led to increasing uptake, indicating viral spread. Radiovirotherapy of xenograft LNCaP tumors with Ad5/3PB-ADP-hNIS showed the most significant survival extension versus control tumors (p=0.001), but the benefit of radiovirotherapy was not statistically significant compared with virotherapy alone in this model. These results show the potential of Ad5/3PB-ADP-hNIS as a vector for treatment of prostate cancer.  相似文献   

5.
6.
Conditionally replicating adenoviruses (CRAds) selectively replicate in and thereby kill cancer cells. The CRAd AdDelta24 with pRb-binding-deficient E1A kills cancer cells efficiently. Arming CRAds with genes encoding prodrug-converting enzymes could allow for enhanced anticancer efficacy by the combined effects of oncolytic replication and local prodrug activation. Here, we investigated combination treatment of human colon cancer cell lines with AdDelta24-type CRAds and gene-directed enzyme prodrug therapy (GDEPT) using two different enzyme/prodrug systems, that is, thymidine kinase/ganciclovir (TK/GCV) and carboxylesterase (CE)/CPT-11. On all three cell lines tested, GDEPT with TK/GCV made CRAd treatment less efficacious. In contrast, expression of a secreted form of CE (sCE2) combined with CPT-11 treatment markedly enhanced the efficacy of AdDelta24 virotherapy. Based on this observation, we constructed an AdDelta24 variant expressing sCE2. In the absence of CPT-11, this new CRAd Ad5-Delta24.E3-sCE2 was similarly effective as its parent in killing human colon cancer cells. Low concentrations of CPT-11 inhibited Ad5-Delta24.E3-sCE2 propagation. Nevertheless, CPT-11 specifically augmented the cytotoxicity of Ad5-Delta24.E3-sCE2 against all three-colon cancer cell lines. Hence, the positive contribution of sCE2/CPT-11 GDEPT to colon cancer cytotoxicity outweighed its negative influence on CRAd propagation. Therefore, CRAd-sCE2/CPT-11 combination therapy appears useful for more effective treatment of colon cancer.  相似文献   

7.
Conditionally replicating adenoviruses (CRADs) take advantage of tumor-specific characteristics for preferential replication and subsequent oncolysis of cancer cells. The antitumor effect is determined by the capability to infect tumor cells. Here, we used RGDCRADcox-2R, which features the cyclooxygenase-2 promoter for replication control and an integrin binding RGD-4C motif for enhanced infectivity of ovarian cancer cells. RGDCRADcox-2R replicated in and killed human ovarian cancer cells effectively, while the replication in nonmalignant cells was low. Importantly, the therapeutic efficacy, as evaluated in an orthotopic model of peritoneally disseminated ovarian cancer, was significantly improved and toxicity was lower than with a wild-type virus. Thus, this CRAD could be tested for treatment of ovarian cancer in humans.  相似文献   

8.
We have constructed a new capsid-modified adenovirus (Ad) vector that specifically replicates in tumor cells and expresses TNF-related apoptosis-inducing ligand (TRAIL). The Ad capsid contains short-shafted fibers derived from Ad serotype 35, which allow for efficient infection of malignant tumor cells, and largely avoids innate toxicity after intravenous application. Replication-dependent homologous recombination in Ad genomes was used to achieve tumor-specific expression of Ad E1a (to mediate viral replication) and TRAIL (to mediate apoptosis and enhance release of progeny virus from infected cells). We demonstrated that our oncolytic vector (Ad5/35.IR-E1A/TRAIL) induced apoptosis in human tumor cell lines derived from colorectal, lung, prostate, and liver cancer. Both in vitro and in vivo tumor models showed efficient intratumoral spread of this vector. In a model for metastatic colon cancer, tail vein infusion of Ad5/35.IR-E1A/TRAIL resulted in elimination of preestablished liver metastases. Intravenous injection of this vector caused a transient elevation of serum glutamic pyruvic transaminase in tumor-bearing mice, which we attributed to factors released from apoptotic tumor cells. Liver histology analyzed at day 14 after virus injection did not show signs of hepatocellular damage. This new oncolytic vector represents a potentially efficient means for gene therapy of metastatic cancer.  相似文献   

9.
In clinical trials with cancer patients, the safety of conditionally replicating adenoviruses (CRAds) has been good. However, marginal data are available on the persistence or antitumor efficacy of these agents. The oncolytic potency of CRAds is determined by their capacity for entering target cells. Consequently, we constructed a retargeted CRAd featuring a secreted marker protein, soluble human carcinoembryogenic antigen (hCEA), which can be measured in growth medium or plasma. We found that virus replication closely correlated with hCEA secretion both in vitro and in vivo. Further, antitumor efficacy and the persistence of the virus could be deduced from plasma hCEA levels. Finally, using in vivo bioluminescence imaging, we were able to detect effective tumor cell killing by the virus, which led to enhanced therapeutic efficacy.  相似文献   

10.
We have generated a thymidine kinase gene-deleted vaccinia virus (VV) (Copenhagen strain) that expressed the fusion suicide gene FCU1 derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. Intratumoral inoculation of this thymidine kinase gene-deleted VV encoding FCU1 (VV-FCU1) in the presence of systemically administered prodrug 5-fluorocytosine (5-FC) produced statistically significant reductions in the growth of subcutaneous human colon cancer in nude mice compared with thymidine kinase gene-deleted VV treatments or with control 5-fluorouracil alone. A limitation of prodrug therapies has often been the requirement for the direct injection of the virus into relatively large, accessible tumors. Here we demonstrate vector targeting of tumors growing subcutaneously following systemic administration of VV-FCU1. More importantly we also demonstrate that the systemic injection of VV-FCU1 in nude mice bearing orthotopic liver metastasis of a human colon cancer, with concomitant administration of 5-FC, leads to substantial tumor growth retardation. In conclusion, the insertion of the fusion FCU1 suicide gene potentiates the oncolytic efficiency of the thymidine kinase gene-deleted VV and represents a potentially efficient means for gene therapy of distant metastasis from colon and other cancers.  相似文献   

11.
Liu X  Qian Q  Xu P  Wolf F  Zhang J  Zhang D  Li C  Huang Q 《Human gene therapy》2011,22(4):427-437
Using conditionally replicating adenoviral vectors (CRAds) is a promising strategy in the treatment of solid tumors. The prospective of this study was to design a novel CRAd for the treatment of gastrointestinal cancer and show its efficacy in vitro, as well as in vivo. To determine if aberrant wnt signaling in tumor cells can be used to selectively drive viral replication, we analyzed six colorectal and hepatocellular cell lines, as well as 13 colorectal tumors and 17 gastric tumors, for β-catenin mutation status or aberrant wnt signaling, both of which were found frequently. Based on these findings, a novel CRAd (Ad5F11.wnt-E1A-hIL24) containing an E1A expression cassette driven by an artificial wnt promoter and delivering an apoptosis-inducing gene, interleukin-24 (IL24), was engineered. To enhance infection efficiency, the virus was pseudotyped by replacing adenovirus serotype 5 (Ad5) with Ad11 fiber. Ad5F11.wnt-E1A-hIL24 virus exhibited high selectivity toward cells with aberrant wnt signaling both in vitro and in mouse xenograft tumors. Transduction efficiency was significantly improved compared with that of nonpseudotyped control viruses. The proliferation of tumor cell lines, as well as tumor growth, in mouse xenografts could be profoundly inhibited by viral infection with Ad5F11.wnt-E1A-hIL24. The therapeutic effect was associated with increased apoptosis through caspase-3 activation. In addition, Ad5F11b vector exhibited a more favorable biodistribution, blood clearance, and transgene expression compared with conventional Ad5 vector after systemic or intratumoral injection in human gastrointestinal cancer xenografts. We think that our approach is a promising strategy in the treatment of gastrointestinal cancer, warranting further clinical investigation.  相似文献   

12.
Gutless adenovirus: last-generation adenovirus for gene therapy   总被引:1,自引:0,他引:1  
Alba R  Bosch A  Chillon M 《Gene therapy》2005,12(Z1):S18-S27
Last-generation adenovirus vectors, also called helper-dependent or gutless adenovirus, are very attractive for gene therapy because the associated in vivo immune response is highly reduced compared to first- and second-generation adenovirus vectors, while maintaining high transduction efficiency and tropism. Nowadays, gutless adenovirus is administered in different organs, such as the liver, muscle or the central nervous system achieving high-level and long-term transgene expression in rodents and primates. However, as devoid of all viral coding regions, gutless vectors require viral proteins supplied in trans by a helper virus. To remove contamination by a helper virus from the final preparation, different systems based on the excision of the helper-packaging signal have been generated. Among them, Cre-loxP system is mostly used, although contamination levels still are 0.1-1% too high to be used in clinical trials. Recently developed strategies to avoid/reduce helper contamination were reviewed.  相似文献   

13.
We describe here a novel strategy to enhance the in vivo efficacy of replicating adenovirus therapy, using coinjection of plasmid DNA encoding a fusogenic viral glycoprotein. The combination of fusogenic membrane glycoprotein (FMG)-induced tumor cell fusion and infection with replicating adenovirus effectively treats even large established tumors at doses of plasmid DNA and virus that alone are ineffective. Adenoviral infection appears to increase the transduction of the tumor cells to a modest degree thereby boosting the FMG-mediated component of the therapy. Simultaneously, syncytial formation enhances the therapeutic effects of viral infection by increasing spread of adenoviral particles through the tumor cell population and by increasing titer of virus released from the tumor cells. This effect is due probably to release of intracellular viral particles upon tumor cell death and also to increased levels of E1A protein within syncytia, whose increased metabolic rate is associated with enhanced levels of protein expression. Cotransduction of tumor cells with replicating adenovirus and FMG-expressing vectors could either be combined within single replicating vectors or could be used in strategies using separate administration of two components, both at lower doses than required for either therapy alone.  相似文献   

14.
Conditionally replicating adenoviruses (CRAds) represent a novel approach for the treatment of cancers resistant to conventional therapies. The efficacy of CRAds might be further improved by using chemotherapeutic agents in a multimodal antitumor approach. We have evaluated the use of Ad5/3-Delta24, a serotype 3 receptor targeted Rb/p16 pathway selective CRAd, in combination with gemcitabine against human ovarian adenocarcinoma. The combination of these agents showed synergistic cell killing in vitro compared to single treatments. However, the effect was dependent on dose and sequencing of the agents. Our results also indicate that gemcitabine reduces the initial rate of Ad5/3-Delta24 replication without affecting the total amount of virus produced. Possible reasons for synergy between Ad5/3-Delta24 and gemcitabine include the chemosensitizing activity of E1A and/or altered replication kinetics. In an orthotopic murine model of peritoneally disseminated ovarian cancer, the combination increased the survival of mice over either agent alone, and almost 60% of treated mice were cured. Sequencing of the agents was critical for toxicity versus efficacy. Mice remained free from intraperitoneal disease, but some succumbed to treatment-related hepatic or bone marrow toxicity. This suggests that improved efficacy may uncover treatment-related toxicity, which needs to be monitored closely in clinical trials.  相似文献   

15.
Advances in understanding the interaction of animal viruses with their cognate receptors has led to improvements in the development of cell-specific, targeted viral vectors. Research strategies to generate safe, non-inflammatory viral vectors that are capable of delivering a therapeutic gene to a specific population of cells are currently underway in many laboratories. One approach in the utilization of this cell targeting activity is to ablate the natural interaction of the virus with its native receptor, although this is not an absolute requirement. The initial development of 'viral targeting strategies' was based on the view that by modifying the viral protein/receptor interaction, it would be possible to redirect virus vectors to new host cells. As the understanding of virus/cell interactions increased it was observed, however, that many viruses can use different entry mechanisms for cell attachment and penetration. Adenovirus vectors have been used extensively for the delivery of genes to cells. The entry mechanism for adenoviruses into cells has recently been studied and is relatively well understood, however, there are many aspects of cell receptor/virus interactions, which have still to be elucidated. The single high-affinity receptor on mammalian cells for adenovirus type 5 is recognized as the coxsackie and adenovirus receptor. However, in the absence of coxsackie and adenovirus receptor other receptors are used. A thorough understanding of the biology of adenoviruses is essential in the further development of their use as vectors for cell targeting. One strategy is to modify the viral capsid, either through coating the surface using bispecific antibodies, or by chemically crosslinking the targeting ligand onto the virion surface. Another approach is to genetically modify the virus by incorporating the targeting ligand into the viral 'spike' (fiber) protein. This involves manipulating the adenovirus genome and generating a new targeting ligand on the surface of the fiber protein using recombinant DNA technology. The penton base protein has also received attention as a means of directing adenoviruses via insertion of novel targeting ligands.  相似文献   

16.
Cancer gene therapy using a survivin mutant adenovirus   总被引:141,自引:0,他引:141       下载免费PDF全文
We have constructed a replication-deficient adenovirus encoding a nonphosphorylatable Thr(34)-->Ala mutant of the apoptosis inhibitor survivin (pAd-T34A) to target tumor cell viability in vitro and in vivo. Infection with pAd-T34A caused spontaneous apoptosis in cell lines of breast, cervical, prostate, lung, and colorectal cancer. In contrast, pAd-T34A did not affect cell viability of proliferating normal human cells, including fibroblasts, endothelium, or smooth muscle cells. Infection of tumor cells with pAd-T34A resulted in cytochrome c release from mitochondria, cleavage of approximately 46-kDa upstream caspase-9, processing of caspase-3 to the active subunits of approximately 17 and 19 kDa, and increased caspase-3 catalytic activity. When compared with chemotherapeutic regimens, pAd-T34A was as effective as taxol and considerably more effective than adriamycin in induction of tumor cell apoptosis and enhanced taxol-induced cell death. In three xenograft breast cancer models in immunodeficient mice, pAd-T34A suppressed de novo tumor formation, inhibited by approximately 40% the growth of established tumors, and reduced intraperitoneal tumor dissemination. Tumors injected with pAd-T34A exhibited loss of proliferating cells and massive apoptosis by in situ internucleosomal DNA fragmentation. These data suggest that adenoviral targeting of the survivin pathway may provide a novel approach for selective cancer gene therapy.  相似文献   

17.
Adenovirus-based vectors can efficiently transfer therapeutic genes into cells through an entry process that is initiated be binding to specific receptors on the cell surface. The receptors for the most commonly used Ad vectors include both the Coxsackie and adenovirus receptor (CAR) and omega-integrins. Therapeutic applications of AD vectors could be expanded if the specificity of gene transfer could be modulated to enhance expression of a therapeutic gene in transfer tissues and avoid non-target tissues. Ad vectors have been successfully retargeted to novel receptors using several approaches. The merits and challenges of specific approaches are discussed. In vivo evaluation of these retargeted Ad vectors has given promising results but has also highlighted additional challenges for achieving efficient targeted gene delivery. Additional modifications beyond those affecting interaction with the native receptors, CAR and integrins may be required both to avoid the clearance mechanisms that effectively remove circulating vector following systemic administration and to avoid gene transfer in non-target tissues such as the liver. Developing Ad vector that address these issues and can be targeted to novel receptors would enable gene delivery at the site of disease in applications that are currently not feasible.  相似文献   

18.
Although replication-competent viruses have been developed to treat cancers, their cytotoxic effects are insufficient, as infection is inhibited by the generation of neutralizing antibodies. To address this limitation, we developed a carrier cell system to deliver a replication-competent adenovirus. Carrier cells infected with replication-competent adenovirus were incubated with target cancer cells in a high titer of anti-adenovirus antibody. Carrier cells were injected into syngeneic subcutaneous tumors after immunization with adenovirus. Carrier cell-derived cell fragments containing viral particles were engulfed by proliferative target cancer cells. This engulfment-mediated transfer of adenovirus was not inhibited by the anti-adenovirus antibody and enabled repetitive infection. After the induction of anti-adenoviral cytotoxic T-lymphocyte (CTL) responses by immunization with adenovirus, administration of carrier cells infected with a replication-competent adenovirus induced complete tumor regression. Adenovirus-GM-CSF augmented the anti-tumor effect of carrier cells by increasing anti-adenoviral and anti-tumoral CTL responses and decreased the number of injections of carrier cells required to induce complete tumor regression. This novel carrier cell-mediated viral transfection system might prove useful in a variety of cancer therapies.  相似文献   

19.
Biology of adenovirus and its use as a vector for gene therapy   总被引:1,自引:0,他引:1  
  相似文献   

20.
Replication-incompetent adenoviruses (Ad) carrying the herpes simplex thymidine kinase (HSVtk) gene have been used in a number of human cancer gene therapy trials, however transduction has generally been limited to a small minority of tumor cells. To solve this problem, replication-competent adenoviral vectors carrying transgenes such as HSVtk have been developed. However, contradictory evidence exists regarding the efficacy of these new vectors. Accordingly, we constructed and tested a replication-competent E3-deleted adenoviral vector containing the HSVtk suicide gene driven by the endogenous E3 promoter (Ad.wt.tk). This virus showed high level production of the HSVtk transgene and was more efficacious than a non-replicating virus in vitro, after injection into flank tumors, and against established intraperitoneal tumors. However, addition of ganciclovir (GCV) therapy to cells or tumor-bearing animals treated with the replicating vector containing the HSVtk suicide gene did not result in increased cell killing. Our results indicate that addition of HSVtk to a replicating Ad virus will not likely be useful in augmenting antitumor effects.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号