首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到17条相似文献,搜索用时 93 毫秒
1.
溶瘤单纯疱疹病毒治疗技术及其进展   总被引:3,自引:0,他引:3  
田聆  薛京伦  贾韦国 《生命科学》2008,20(5):734-741
溶瘤病毒治疗是当前肿瘤治疗技术的研究热点,溶瘤单纯疱疹病毒是目前研究最多的,也是抗肿瘤作用最好的溶瘤病毒之一,本文就主要介绍溶瘤单纯疱疹病毒的优势、遗传操作、发展策略、研究进展及靶向性策略等。  相似文献   

2.
1型单纯疱疹病毒(Herpes simplex virustype1,HSV-1)感染效率高且易于通过基因工程改造,己广泛应用于肿瘤治疗研究和临床实验。溶瘤HSV-1可通过基因工程改造或从HSV-1自发突变株中筛选获得。研究证实溶瘤HSV-1能够有效抑杀肿瘤细胞,可通过多种机制靶向肿瘤细胞,溶瘤HSV-1与放化疗联合使用治疗肿瘤的研究也取得了理想的结果。目前,已有多个溶瘤HSV-1进入临床试验。  相似文献   

3.
1型单纯疱疹病毒(herpes simplex virus type1,HSV-1)具天然的嗜神经性和高效的感染能力,已广泛应用于肿瘤治疗研究和临床实验。目前开发的溶瘤HSV-1包括扩增子(amplicon)、复制缺陷型病毒(replication-defective virus)和条件选择性复制病毒(conditional-replicative virus)三类。前两类HSV-1在细胞中不能复制,溶瘤效应限于局部组织,而复制型溶瘤HSV-1能够在肿瘤细胞中复制,复制周期完成后可裂解肿瘤细胞并释放子代病毒,继而感染临近的肿瘤细胞。目前,一些复制型溶瘤HSV-1已进入临床试验阶段,并在肿瘤治疗方面取得了一些令人鼓舞的结果。  相似文献   

4.
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   

5.
1型单纯疱疹病毒(herpes simplex virus 1, HSV-1)具有基因组大、允许插入多个外源基因、病毒复制周期短、可感染多种肿瘤细胞并易在肿瘤细胞中复制、对人体致病性弱且目前抗疱疹病毒药物(如阿昔洛韦)可有效控制病毒复制等优点,因而以HSV-1为基础开发的溶瘤1型单纯疱疹病毒(oncolytic herpes simplex virus 1, oHSV-1)产品在肿瘤治疗研究中受到广泛关注。虽然目前已经有多个产品获批开展临床试验,但在药学研究中仍存在诸多问题。现就oHSV-1的研究现状及目前该类产品在申报临床阶段药学研究中存在的主要问题展开论述和总结,以促进此类产品的临床开发与应用。  相似文献   

6.
基因工程改造的Ⅰ型单纯疱疹病毒(HSV-1)对肿瘤具有特异的靶向性和显著的杀伤能力,已经成为肿瘤治疗研究的热点。HSV-1类溶瘤病毒在临床上对多种肿瘤表现出良好的治疗效果,也被证实能够刺激机体产生较强的抗肿瘤免疫应答。2015年,Amgen公司的T-VEC被美国FDA批准用于治疗恶性黑色素瘤。溶瘤病毒疗法联合放、化疗等治疗方式能进一步提高肿瘤治疗效果。更重要的是,靶向免疫检查点疗法在肿瘤免疫治疗中有着巨大的应用前景,有望与溶瘤病毒疗法联用,成为未来肿瘤治疗的新方向。  相似文献   

7.
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   

8.
近年来,随着国内外几款溶瘤病毒制剂的相继上市,溶瘤病毒疗法成为肿瘤免疫治疗的焦点。溶瘤病毒可选择性感染并裂解肿瘤细胞,同时释放肿瘤相关抗原激活机体的抗肿瘤免疫反应,达到杀伤肿瘤细胞和抑制肿瘤生长的目的。溶瘤病毒对肿瘤的靶向杀伤作用决定了其安全性和溶瘤效果。为了开发出安全高效的溶瘤病毒,目前主要采用以下策略:利用某些病毒载体对肿瘤细胞的天然靶向性,使溶瘤病毒选择性地在肿瘤细胞内复制并杀伤肿瘤细胞;或者对病毒基因组进行缺失和插入等修饰,通过靶向肿瘤细胞特异性表面受体、胞内信号通路或者肿瘤微环境等提高溶瘤病毒的肿瘤靶向性。其中,肿瘤微环境中的低氧状态、新血管生成以及免疫抑制状态等都可成为溶瘤病毒的靶点。而溶瘤病毒通过表达细胞因子和免疫检查点抑制剂,或者与CAR-T细胞联合作用,靶向调节肿瘤微环境中免疫抑制状态,成为提高溶瘤病毒肿瘤靶向性的常用方法。本文将对以上溶瘤病毒靶向治疗肿瘤策略的研究进展进行综述。  相似文献   

9.
溶瘤病毒利用肿瘤细胞抗病毒信号通路缺损或病毒受体过表达的特点,实现在其中选择性高复制进而杀伤肿瘤细胞,同时刺激机体产生特异性抗肿瘤免疫反应,是目前肿瘤治疗研究领域的热点。水疱性口炎病毒(vesicular stomatitis virus,VSV)能依赖肿瘤细胞干扰素信号通路的缺陷特异性靶向肿瘤细胞,具有复制高效、广泛组织嗜性、人群低致病性、基因组较小且易操纵等优势,是一种具有发展潜力的溶瘤病毒载体。对水疱性口炎病毒的病毒学特征以及目前基于VSV溶瘤病毒关于提高肿瘤选择性、延长半衰期、增强溶瘤效果的研究进展进行综述,为基于VSV溶瘤制剂的开发提供依据,为肿瘤治疗提供新的策略。  相似文献   

10.
溶瘤病毒可以感染和破坏癌组织,是一种治疗癌症的新型生物疗法。溶瘤病毒在癌细胞中选择性复制,进而导致癌细胞裂解,释放肿瘤特异性抗原,诱导抗癌免疫反应,充当原位肿瘤疫苗。对病毒进入、复制、诱导和抑制免疫反应机制的深入了解促进了利用病毒治疗人类疾病技术的发展。过去十年溶瘤病毒领域临床试验的进展证实了其对癌症患者的治疗益处,利用溶瘤病毒作为载体治疗特定类型的癌症是肿瘤免疫治疗市场的新增长点。通过全面分析溶瘤病毒免疫治疗市场的细分领域及其市场动态,从关键技术进展、主要企业竞争格局和产品研发进展角度进行分析,并展望溶瘤病毒的发展前景,旨在为相关企业研发方向选择及地区产业决策提供参考。  相似文献   

11.
Oncolytic virotherapy is an emerging treatment modality that uses replication-competent viruses to destroy cancers. Recent advances include preclinical proof of feasibility for a single-shot virotherapy cure, identification of drugs that accelerate intratumoral virus propagation, strategies to maximize the immunotherapeutic action of oncolytic viruses and clinical confirmation of a critical viremic threshold for vascular delivery and intratumoral virus replication. The primary clinical milestone has been completion of accrual in a phase 3 trial of intratumoral herpes simplex virus therapy using talimogene laherparepvec for metastatic melanoma. Key challenges for the field are to select 'winners' from a burgeoning number of oncolytic platforms and engineered derivatives, to transiently suppress but then unleash the power of the immune system to maximize both virus spread and anticancer immunity, to develop more meaningful preclinical virotherapy models and to manufacture viruses with orders-of-magnitude higher yields than is currently possible.  相似文献   

12.
13.
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   

14.
Viruses used for gene therapy are usually genetically modified to deliver therapeutic transgenes and prevent viral replication. In contrast, replication-competent viruses may be used for cancer therapy because replication of some viruses within cancer cells can result in their destruction (oncolysis). Viral ribonucleotide reductase expression is defective in the HSV1 mutant hrR3. Cellular ribonucleotide reductase, which is scarce in normal liver and abundant in liver metastases, can substitute for its viral counterpart to allow hrR3 replication in infected cells. Two or three log orders more of hrR3 virions are produced from infection of colon carcinoma cells than from infection of normal hepatocytes in viral replication assays. This viral replication is oncolytic. A single intravascular administration of hrR3 into immune-competent mice bearing diffuse liver metastases dramatically reduces tumor burden. hrR3-mediated tumor inhibition is equivalent in immune-competent and immune-incompetent mice, suggesting that viral oncolysis and not the host immune response is the primary mechanism of tumor destruction. HSV1-mediated oncolysis of diffuse liver metastases is effective in mice preimmunized against HSV1. These results indicate that replication-competent HSV1 mutants hold significant promise as cancer therapeutic agents. Yoon, S. S., Nakamura, H., Carroll, N. M., Bode, B. P., Chiocca, E. A., Tanabe, K. K. An oncolytic herpes simplex virus type 1 selectively destroys diffuse liver metastases from colon carcinoma.  相似文献   

15.
Molecular therapy using viruses would benefit greatly from a non-invasive modality for assessing dissemination of viruses. Here we investigated whether positron emission tomography (PET) scanning using [(124)I]-5-iodo-2'-fluoro-1-beta-d-arabinofuranosyl-uracil (FIAU) could image cells infected with herpes simplex viruses (HSV). Using replication-competent HSV-1 oncolytic viruses with thymidine kinase (TK) under control of different promoters, we demonstrate that viral infection, proliferation and promoter characteristics all interact to influence FIAU accumulation and imaging. In vivo, as few as 1 x 107 viral particles injected into a 0.5-cm human colorectal tumor can be detected by [(124)I]FIAU PET imaging. PET signal intensity is significantly greater at 48 hours compared with that at 8 hours after viral injection, demonstrating that PET scanning can detect changes in TK activity resulting from local viral proliferation. We also show the ability of FIAU-PET scanning to detect differences in viral infectivity at 0.5 log increments. Non-invasive imaging might be useful in assessing biologically relevant distribution of virus in therapies using replication-competent HSV.  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.
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   

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