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 109 毫秒
1.
溶瘤病毒(Oncolytic virus,OV)是可以靶向感染并杀伤肿瘤细胞的一类病毒,其中溶瘤I型单纯疱疹病毒(Oncolytic herpes simplex virus type 1,OHSV-1)是目前研究最多的溶瘤病毒之一,可通过多种策略进行构建,已有多种OHSV-1进入临床试验,大量结果显示其具有较好的安全性和有效性。本文主要介绍OHSV-1的分子生物学特性与优势、主要的开发及靶向性策略、各类OHSV-1的研究进展以及目前存在的问题等。  相似文献   

2.
溶瘤单纯疱疹病毒(oncolytic HSV)是一种重要的具有临床应用前景的病毒,但是该病毒的感染缺乏肿瘤细胞靶向性,因而在相当程度上限制了其进一步的临床应用。首先把特异结合肿瘤细胞表面叶酸受体的叶酸(FA)与聚乙二醇(PEG)进行化学交联,然后用PEG化的叶酸对溶瘤单纯疱疹病毒G207进行共价表面化学修饰,并检测修饰后的病毒FA-PEG-HSV和PEG-HSV的物理和生物学活性。结果显示,FA-PEG-HSV和PEG-HSV的稳定性较未修饰的HSV有所提高,但病毒活力则分别下降为未修饰HSV的22.5%和27.5%,而FA-PEG-HSV对叶酸受体过表达的肿瘤细胞KB的感染效率则比叶酸受体低表达A549细胞提高了300%。以上结果表明,FA-PEG-HSV是一种成功的叶酸受体靶向性溶瘤病毒复合体。  相似文献   

3.
本研究旨在检测BALB/c小鼠肿瘤注射部位溶瘤Ⅱ型单纯疱疹病毒(oncolytic herpes simplex virus type 2, oHSV2)的持续时间和复制水平。同时检测血清中人粒细胞-巨噬细胞集落刺激因子(human granulocyte macrophage colony-stimulating factor, hGM-CSF)的表达量和HSV-2抗体水平。小鼠经瘤内注射高、低两个剂量oHSV2-Fluc[萤火虫荧光素酶(firefly luciferase, Fluc)],检测其荧光表达变化及持续时间。采用实时荧光定量聚合酶链式反应(quantitative real-time polymerase chain reaction, qPCR)检测肿瘤组织中oHSV2基因拷贝数。酶联免疫吸附法(enzyme linked immunosorbent assay, ELISA)检测血清中hGM-CSF表达量以及HSV-2抗体水平。结果表明高剂量组肿瘤体积显著小于对照组(P<0.01)。瘤内注射oHSV2-Fluc表明病毒携带的Fluc可以在肿瘤中持续表达,于第11天仍能检测到荧光,直至第18天低于检测线。通过提取肿瘤组织RNA进行qPCR,表明第9天高、低剂量组肿瘤组织均存在oHSV2的mRNA。对血清进行ELISA检测,相较于对照组,高、低剂量组hGM-CSF及HSV2抗体水平显著提高(P<0.05)。这些发现表明,oHSV2能够在瘤内正常复制,同时所携带的外源因子可持续表达,对肿瘤产生杀伤作用。瘤内注射oHSV2后,小鼠血清中存在较高水平的hGM-CSF和HSV-2抗体。  相似文献   

4.
肿瘤已成为危及全人类生命的重大疾病,虽然常规治疗手段如手术及放疗/化疗等不断发展,但对某些复发、难治性恶性肿瘤仍然束手无策,亟需安全、有效可行的治疗手段。在肿瘤基因治疗领域,能在肿瘤细胞内大量自我复制并选择性杀伤肿瘤细胞的溶瘤病毒(Oncolytic virus)逐渐崭露头角,目前溶瘤病毒抗瘤治疗备受关注。其中溶瘤麻疹病毒疫苗株(MV-Edm)因其可靠的安全性和优良的溶瘤效果已进入几项临床试验,为推动其临床转化奠定了基础,期望为肿瘤患者带来治疗上的突破。本文就目前溶瘤麻疹病毒的临床研究与转化进展的相关研究成果进行综述。  相似文献   

5.
溶瘤病毒(oncolytic viruses, OVs)是一种具有发展潜力的肿瘤免疫治疗方法,是天然或经基因改造后对肿瘤具有靶向性的DNA病毒和RNA病毒。溶瘤病毒具有肿瘤靶向性、作为载体传递多种转基因表达、诱导免疫性细胞死亡和促进抗肿瘤免疫反应等优点,而且具有可耐受的安全性。该文将从溶瘤病毒的发展历程、分类、作用机制、改造策略、生物标志物和临床应用的研究现状和现存问题展开综述。  相似文献   

6.
采用溶瘤腺病毒策略进行肝细胞癌基因治疗的思考   总被引:1,自引:0,他引:1  
研制有效的盱细胞癌生物治疗制剂,作为现有治疗手段的有益补充或替代,具有重大的社会和经济效益。采用腺病毒溶瘤策略开展肝细胞癌的基因治疗研究是一个新的方向。章概述了此方向研究所面临的问题,并提出了一些设想。  相似文献   

7.
钟江 《微生物与感染》2004,27(6):1-4,10
研究发现有些病毒具有特异性感染并杀伤肿瘤细胞的能力,这类病毒被称为“溶瘤病毒”。天然的溶瘤病毒有细小病毒、呼肠孤病毒、新城疫病毒、水疱性口炎病毒、麻疹病毒等。有些病毒缺失一些病毒基因的突变体,具有溶瘤病毒的特性,其中包括腺病毒、疱疹病毒和甲型流感病毒。通过病毒基因工程可以进一步提高溶瘤病毒的安全性和杀伤肿瘤细胞的效果。一系列体外、动物模型和临床的试验证实,不少溶瘤病毒具有良好的安全性和抑制肿瘤的能力。  相似文献   

8.
段海潇  程奕宁  汪洋  胡翰  刘滨磊 《生命科学》2023,(11):1462-1474
嵌合抗原受体修饰的T细胞(chimeric antigen receptor T cells, CAR-T)疗法属于肿瘤免疫治疗的范畴。该疗法在血液系统恶性肿瘤治疗中表现优异,但在实体瘤治疗中存在诸多挑战。近年来,溶瘤病毒(oncolytic viruses, OVs)在针对黑色素瘤、脑胶质瘤等实体瘤适应证的临床试验中展现出良好的疗效。溶瘤病毒一方面选择性地在肿瘤细胞中复制杀伤肿瘤细胞,另一方面通过激活机体自身的免疫系统发挥抗肿瘤作用。因此,溶瘤病毒与免疫检查点抑制剂、肿瘤浸润淋巴细胞疗法(tumor infiltrating lymphocytes, TIL)等免疫疗法的联合应用也在广泛开展。目前研究表明,溶瘤病毒不仅能够增加CAR-T细胞的抗肿瘤活性,而且通过传递肿瘤相关抗原或特异性抗原来增强CAR-T细胞对肿瘤的杀伤作用,同时溶瘤病毒还可以帮助CAR-T细胞克服免疫抑制性的肿瘤微环境。两种疗法的联合应用在临床前研究中展现出了良好的疗效和安全性,能够解决CAR-T在实体瘤治疗领域的瓶颈,具有广阔的临床转化前景。本文就溶瘤病毒与CAR-T细胞联合治疗实体瘤的药效及相关机制研究展开论述...  相似文献   

9.
溶瘤病毒疗法属于免疫治疗的手段之一。其可通过病毒特异性地感染裂解肿瘤细胞和激活肿瘤免疫两种途径来达到杀伤肿瘤的目的;同传统疗法比,具有安全、高效、副作用小等优点。流感病毒自1900年代首次发现其可能作为“有益”的病毒缓解白血病病情以来,不断有研究证明流感病毒具有杀伤肿瘤细胞的能力;利用反向遗传操作技术对病毒进行改造,有望将其发展成为一种更加安全、有效的肿瘤治疗生物制剂。本文将对近年来溶瘤流感病毒利用肿瘤分泌的胰蛋白酶促进病毒感染并在RAS基因突变导致干扰素缺陷的肿瘤中复制来提高肿瘤靶向性,编码CTLA-4的单链抗体或HER-2增强流感病毒的抗癌特异性及作为外源基因IL-2、IL-15、GM-CSF和抗PD-1单克隆抗体的载体激活机体免疫几个方向进行综述。  相似文献   

10.
溶瘤病毒(oncolytic virus,OVs)历经百年发展,应用于当前最具潜力的肿瘤免疫疗法。它主要是天然的或基因修饰的DNA病毒和RNA病毒。近年来随着基因工程技术的飞跃发展,经基因改造的溶瘤病毒在肿瘤治疗领域取得很大进展,很多不同类型的病毒(包括单纯疱疹病毒、腺病毒、痘病毒、麻疹病毒和呼肠孤病毒等)正处于临床前研究、临床试验阶段或已批准上市,显示了良好的安全性和临床疗效。普遍认为溶瘤病毒靶向杀伤肿瘤细胞是通过选择性在肿瘤细胞内自我复制,最终裂解肿瘤细胞,同时可激发机体的免疫应答反应,进而增强抗肿瘤免疫效果,靶向杀伤肿瘤细胞而对正常细胞无明显影响。运用基因重组技术将溶瘤病毒与免疫检查点相结合以及肿瘤免疫联合疗法的兴起和不断进步,使溶瘤病毒的应用更加广泛,但仍存在病毒靶向性、安全性、给药途径等瓶颈问题。本文综述了溶瘤病毒的发展史、病毒分类、不同类型溶瘤病毒产品的临床研究进展、溶瘤病毒靶向杀伤肿瘤的免疫学机制及未来发展面临的挑战与展望等。  相似文献   

11.
12.
Advances in gene modification and viral therapy have led to the development of a variety of vectors in several viral families that are capable of replication specifically in tumor cells. Because of the nature of viral delivery, infection, and replication, this technology, oncolytic virotherapy, may prove valuable for treating cancer patients, especially those with inoperable tumors. Current limitations exist, however, for oncolytic virotherapy. They include the body's B and T cell responses, innate inflammatory reactions, host range, safety risks involved in using modified viruses as treatments, and the requirement that most currently available oncolytic viruses require local administration. Another important constraint is that genetically enhanced vectors may or may not adhere to their replication restrictions in long-term applications. Several solutions and strategies already exist, however, to minimize or circumvent many of these limitations, supporting viral oncolytic therapy as a viable option and powerful tool in the fight against cancer.  相似文献   

13.
Herpes simplex virus type 1 (HSV-1) deleted for the immediate-early gene was applied for treatment of hepatoma cells of SKHep 1 and Huh-7. Hepatoma cells were cultured in medium containing HSV1 expressing GFP gene (QOZ/HG) to determine its transfection rate, and both cell lines infected by MOI 1 of QOZ/HG were found to have high expression of GFP without cytotoxicity. Subcutaneous growth of SKHep 1 cell tumor in nude mice was significantly reduced by injection of replicative-deficient herpes virus (TOZ.1) containing Tk-gene with administration of GCV, in comparison with that of noninjected tumor. SCID mice of peritonitis carcinomatosis due to Huh-7 hepatoma cells infected with TOZ.1 could survive longer under administration of GCV than those without TOZ.1. Therefore replicative-deficient HSV1 is a useful vector for treatment of human hepatoma cells, and TOZ.1 with GCV may be applied to suicide gene therapy for hepatoma and peritonitis carcinomatosis of hepatoma cells.  相似文献   

14.
Oncolytic virus therapy using genetically engineered herpes simplex viruses   总被引:4,自引:0,他引:4  
Todo T 《Human cell》2002,15(3):151-159
An increasing number of oncolytic virus vectors has been developed lately for cancer therapy. Herpes simplex virus type 1 (HSV-1) vectors are particularly useful, because they can be genetically engineered to replicate and spread highly selectively in tumor cells and can also express multiple foreign transgenes. These vectors can manifest cytopathic effect in a wide variety of tumor types without damaging normal tissues, provide amplified gene delivery within the tumor, and induce specific antitumor immunity. Multiple recombinant HSV-1 vectors have been tested in patients with brain tumors and other cancers, which showed the feasibility of administering replication-competent HSV-1 vectors safely in human organs including the brain. Different approaches are currently undertaken to improve the efficacy of oncolytic HSV-1 therapy which include development of new generation vectors via further genetic engineering of existing safe vectors, combination with immune gene therapy, and combination with conventional therapies. Oncolytic virus therapy is a promising therapeutic modality that awaits establishing as an important treatment option for cancer patients in the near future.  相似文献   

15.
BACKGROUND: Tumour necrosis factor alpha (TNFalpha) therapy is a promising anti-cancer treatment when combined with radiotherapy due to its potent radio sensitising effects, but systemic toxicity has limited its clinical use. Previously, non-replicative adenovirus vectors have been used to deliver TNFalpha directly to the tumour, including under the control of a radiation sensitive promoter. Here, we have used an ICP34.5 deleted, oncolytic herpes simplex virus (HSV) for delivery to increase expression levels and spread through the tumour, and the use of the US11 true late HSV promoter to limit expression to where the virus replicates, i.e. selectively in tumour tissue. METHODS: TNFalpha expression under the CMV or US11 promoter was compared on cell lines CT26, BHK and Fadu. To further compare the activities of the promoters, expression of human TNFalpha was analysed in the presence and absence of acyclovir--an inhibitor of viral DNA replication and on HSV/ICP34.5- non-permissive cell line 3T6. The in vivo efficacy and toxicity of TNFalpha viruses were compared using A20 double flank tumour model in Balb/C mice and Fadu tumour model in nude mice. RESULTS: The results demonstrated that the US11 promoter significantly reduced and delayed TNFalpha expression as compared to use of the CMV promoter, especially in non-permissive cells or in the presence of acyclovir. Despite the reduced and more selective expression levels, US11 driven TNFalpha showed improved anti-tumour effects compared to CMV driven TNFalpha, and without the toxic side effects. CONCLUSIONS: This approach is therefore beneficial in increasing localised TNFalpha expression as compared to the use of non-replicative approaches, and combines the effects of TNFalpha with oncolytic virus replication which is expected to further enhance the efficacy of radiotherapy in a combined treatment approach.  相似文献   

16.
BACKGROUND: Malignant pleural mesothelioma (MPM) is an aggressive cancer that is refractory to current treatment modalities. Oncolytic herpes simplex viruses (HSV) used for gene therapy are genetically engineered, replication-competent viruses that selectively target tumor cells while sparing normal host tissue. The localized nature, the potential accessibility and the relative lack of distant metastasis make MPM a particularly suitable disease for oncolytic viral therapy. METHODS: The infectivity, selective replication, vector spread and cytotoxic ability of three oncolytic HSV: G207, NV1020 and NV1066, were tested against eleven pathological types of MPM cell lines including those that are resistant to radiation therapy, gemcitabine or cisplatin. The therapeutic efficacy and the effect on survival of NV1066 were confirmed in a murine MPM model. RESULTS: All three oncolytic HSV were highly effective against all the MPM cell lines tested. Even at very low concentrations of MOI 0.01 (MOI: multiplicity of viral infection, ratio of viral particles per cancer cell), HSV were highly effective against MPM cells that are resistant to radiation, gemcitabine and cisplatin. NV1066, an oncolytic HSV that expresses green fluorescent protein (GFP), was able to delineate the extent of the disease in a murine model of MPM due to selective infection and expression of GFP in tumor cells. Furthermore, NV1066 was able to reduce the tumor burden and prolong survival even when treatment was at an advanced stage of the disease. CONCLUSION: These findings support the continued investigation of oncolytic HSV as potential therapy for patients with therapy-resistant MPM.  相似文献   

17.
溶瘤病毒(oncolytic virus,OVs)疗法是治疗肿瘤的一种新方法,它可通过直接杀死肿瘤细胞并引起机体的免疫反应发挥作用.然而天然溶瘤病毒有一定的局限性,因此需将各种不同的病毒作为载体,通过基因修饰的方法增强或减弱病毒毒力并导入新的功能性基因,以提高其溶瘤作用.目前可以作为溶瘤病毒载体的有单纯疱疹病毒、腺病毒、牛痘病毒、水泡性口炎病毒、麻疹病毒、腮腺炎病毒、脊髓灰质炎病毒等.这些病毒载体均可通过不同的基因修饰方法,靶向性感染并杀死不同的肿瘤细胞,获得较好的溶瘤作用.本文综述了几种溶瘤病毒载体的基因修饰方法,以及修饰后的溶瘤效果.  相似文献   

18.
19.
Herpes simplex virus vectors for gene therapy   总被引:2,自引:0,他引:2  
Herpes simplex virus (HSV) has a number of advantages as a vector for delivering specific genes to the nervous system. These include its large size, wide host range, and its ability to establish long-lived asymptomatic infections in neuronal cells in which a specific region of the viral genome continues to be expressed. Unfortunately, the large size of this virus and difficulty in manipulating it has led to its use as a vector lagging behind that of other, smaller viruses such as the retroviruses. In addition, the virus's ability to replicate lytically in the brain, under some circumstances, causing encephalitis, has led to fears about its potential safety for ultimate use in humans. This review will discuss a number of new approaches that are aimed at rendering simpler the insertion of foreign genes into the virus and making it as safe as possible. Ultimately, these advances offer real hope for the use of HSV vectors in gene therapy procedures.  相似文献   

20.
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