首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 125 毫秒
1.
血友病A是X染色体隐性遗传出血性疾病。其发病原因是患者血液中先天缺乏凝血因子FⅧ。用于血友病A基因治疗研究的载体有病毒载体和非病毒载体,目前研究较多的是病毒载体,主要有逆转录病毒载体和慢病毒载体,腺病毒载体及腺相关病毒载体等。非病毒载体主要有质粒、脂质体、转座子等。文章拟对血友病A基因治疗各载体的特点和研究进展作一综述。  相似文献   

2.
血友病A是由于编码凝血因子Ⅷ (简称FⅧ )的基因先天性异常而导致的机体出血紊乱性遗传病 ,基因治疗是彻底治愈血友病A的一种最为理想的治疗措施 .就FⅧ基因及其蛋白质分子的结构与性质、基因治疗载体构建、靶细胞、动物模型及其临床应用作一简要综述 .  相似文献   

3.
研究基因治疗的公司正像这门新兴科学的进步一样在迅速发展。而且,这种势头还不只限于这些公司,一些大制药公司也正以合作和合资方式参与这一事业。最新的加入者是Baxter Healthcare公司(Deerfield,IL),它的基因治疗单位已在93年8月正式成立。 新单位隶属于该公司生物技术集团,致力于对癌、血友病和糖尿病的基因治疗,这类病的治疗技术都有上10亿美元的市场。公司希望在明年晚些时候对癌和血友病A的治疗进行临床试验。美国有2万个A型血友病患者,日本和欧洲约有5万。由于遗传缺陷,这些人不能产生凝血蛋白因子Ⅷ而导致严重失血。该  相似文献   

4.
基因治疗所用的逆转录病毒载体系统徐铿,陈诗书(上海第二医科大学生物化学教研组,200025)关键词基因治疗,逆转录病毒载体,包装细胞八十年代初发展的以逆转录病毒为基础的基因转移系统对今天基因治疗的实现起了关键性的作用。目前近50个基因治疗计划绝大部分...  相似文献   

5.
重组逆转录病毒介导基因治疗(RMGT)的安全性问题许旻,陈诗书(上海第二医科大学人类基因治疗研究中心,上海200025)关键词逆转录病毒,基因治疗,安全性重组逆转录病毒载体系统是基因治疗中最常用的工具。它具有以下优点:①复制缺陷型病毒颗粒比较安全;②...  相似文献   

6.
逆转录病毒是基因治疗研究中最为主要的基因转移载体,目前批准的基因治疗试验方案中绝大多数采用逆转录病毒作为载体。逆转录病毒具有稳定、安全、高效的优点,但仍存在病毒滴度低的问题。目前多从质粒结构和病毒包装两方面来提高病毒滴度。 将HyTK基因替换逆转录病毒载体GlNa  相似文献   

7.
血友病B是一种凝血Ⅸ因子缺陷引起的X连锁隐性遗传的出血性疾病 .近年来 ,国内外学者在基因治疗方面取得的大量研究成果给血友病B患者带来了根治疾病的希望 .就血友病B的基因治疗研究进展及所存在的问题作一综述 .  相似文献   

8.
基因治疗的目的是将导入的外源基因能够得到时序上、水平上的正确表达,其产物发挥治疗作用。目的基因表达的调控是影响基因治疗效果的一个重要因素。本文系统介绍了基因靶技术在基因治疗中的地位,提高逆转录病毒载体滴度的方法,逆转录病毒载体与目的基因序列的影响,反式激活提高目的基因应用以及对基因治疗中目的基因表达调控的展望。  相似文献   

9.
逆转录病毒载体用于基因治疗的安全性   总被引:2,自引:0,他引:2  
有复制能力的逆转录病毒感染,插入突变,无关序列进入靶细胞是基因治疗潜在的危险,但造成实际危害的可能性极小,完善逆转录病毒载体系统的设计,提高检测有复制能力的病毒的灵敏度将进一步保证基因治疗的安全性。  相似文献   

10.
血友病基因治疗经过30年的持续发展,已取得长足的进步。血友病患者FVIII或FIX水平达到正常甚至治愈已成为可能。虽然在多个国家血友病基因治疗已取得显著成果,但仍然有很大的改进空间。目前临床试验中AAV是血友病基因治疗的主要载体,未来的研究将集中在完善病毒衣壳、转基因和启动子的设计上,以追求更高的转导效率、更低的免疫反应和可预测性的治疗结果。目前的研究表明,与在动物模式中近乎100%的高转导效率相比,在人类肝细胞中实现高转导效率及凝血因子的高表达仍有不足之处,需要避免蛋白质过载引起的细胞应激风险。虽然血友病基因治疗面临一定的挑战,但是随着技术的不断发展成熟,相信未来会开发出真正治愈血友病的个体化治疗方案。  相似文献   

11.
Hemophilia A is a common X chromosome-linked genetic bleeding disorder caused by abnormalities in the coagulation factor VIII gene (F8). Hemophilia A patients suffer from a bleeding diathesis, such as life-threatening bleeding in the brain and harmful bleeding in joints and muscles. Because it could potentially be cured by gene therapy, subhuman animal models have been sought. Current mouse hemophilia A models generated by gene targeting of the F8 have difficulties to extrapolate human disease due to differences in the coagulation and immune systems between mice and humans. Here, we generated a porcine model of hemophilia A by nuclear transfer cloning from F8-targeted fibroblasts. The hemophilia A pigs showed a severe bleeding tendency upon birth, similar to human severe hemophiliacs, but in contrast to hemophilia A mice which rarely bleed under standard breed conditions. Infusion of human factor VIII was effective in stopping bleeding and reducing the bleeding frequency of a hemophilia A piglet but was blocked by the inhibitor against human factor VIII. These data suggest that the hemophilia A pig is a severe hemophilia A animal model for studying not only hemophilia A gene therapy but also the next generation recombinant coagulation factors, such as recombinant factor VIII variants with a slower clearance rate.  相似文献   

12.
Factor VIII (FVIII) is an essential component in blood coagulation, a deficiency of which causes the serious bleeding disorder hemophilia A. Recently, with the development of purification level and recombinant techniques, protein replacement treatment to hemophiliacs is relatively safe and can prolong their life expectancy. However, because of the possibility of unknown contaminants in plasma-derived FVIII and recombinant FVIII, and high cost for hemophiliacs to use these products, gene therapy for hemophilia A is an attractive alternative to protein replacement therapy. Thus far, the adeno-associated virus (AAV) is a promising vector for gene therapy. Further improvement of the virus for clinical application depends on better understanding of the molecular structure and fate of the vector genome. It is likely that hemophilia will be the first genetic disease to be cured by somatic cell gene therapy.  相似文献   

13.
14.
15.
Hemophilia is an inherited bleeding disorder caused by a deficiency of functional clotting factors VIII or IX in the blood plasma. The drawbacks of the classical protein substitution therapy fueled interest in alternative treatments by gene therapy. Hemophilia has been recognized as an ideal target disease for gene therapy because a relatively modest increase in clotting factor levels can result in a significant therapeutic benefit. Consequently, introducing a functional FVIII or FIX gene copy into the appropriate target cells could ultimately provide a cure for hemophilic patients. Several cell types have been explored for hemophilia gene therapy, including hepatocytes, muscle, endothelial and hematopoietic cells. Both nonviral and viral vectors have been considered for the development of hemophilia gene therapy, including transposons, γ‐retroviral, lentiviral, adenoviral and adeno‐associated viral vectors. Several of these strategies have resulted in stable correction of the bleeding diathesis in hemophilia A and B murine as well as canine models, paving the way towards clinical trials. Although clotting factor expression has been detected in hemophilic patients treated by gene therapy, the challenge now lies in obtaining prolonged therapeutic FVIII or FIX levels in these patients. This review highlights the benefits and potential risks of the different gene therapy strategies for hemophilia that have been developed. Copyright © 2010 John Wiley & Sons, Ltd.  相似文献   

16.
Developing rDNA products for treatment of hemophilia A.   总被引:2,自引:0,他引:2  
Current therapy for hemophilia A requires frequent infusion of plasma-derived human factor VIII with the associated drawbacks of potential viral contamination, high cost and limited plasma availability. Factor VIII replacement therapy has been improved through increased knowledge of molecular mechanisms regulating blood coagulation, derived largely from the isolation of the factor VIII gene and its expression in mammalian cells. Homogeneous pure preparations of factor VIII--the largest, most complex protein pharmaceutical produced to date through recombinant DNA technology--can now be produced for successful treatment of hemophilia A.  相似文献   

17.
The major complication in the treatment of hemophilia A is the development of neutralizing antibodies (inhibitors) against factor VIII (FVIII). The current method for eradicating inhibitors, termed immune tolerance induction (ITI), is costly and protracted. Clinical protocols that prevent rather than treat inhibitors are not yet established. Liver-directed gene therapy hopes to achieve long-term correction of the disease while also inducing immune tolerance. We sought to investigate the use of adeno-associated viral (serotype 8) gene transfer to induce tolerance to human B domain deleted FVIII in hemophilia A mice. We administered an AAV8 vector with either human B domain deleted FVIII or a codon-optimized transgene, both under a liver-specific promoter to two strains of hemophilia A mice. Protein therapy or gene therapy was given either alone or in conjunction with anti-CD20 antibody-mediated B cell depletion. Gene therapy with a low-expressing vector resulted in sustained near-therapeutic expression. However, supplementary protein therapy revealed that gene transfer had sensitized mice to hFVIII in a high-responder strain but not in mice of a low-responding strain. This heightened response was ameliorated when gene therapy was delivered with anti-murine CD20 treatment. Transient B cell depletion prevented inhibitor formation in protein therapy, but failed to achieve a sustained hypo-responsiveness. Importantly, use of a codon-optimized hFVIII transgene resulted in sustained therapeutic expression and tolerance without a need for B cell depletion. Therefore, anti-CD20 may be beneficial in preventing vector-induced immune priming to FVIII, but higher levels of liver-restricted expression are preferred for tolerance.  相似文献   

18.
Severe hemophilia A (HA) is an inherited bleeding disorder characterized by <1% of residual factor VIII (FVIII) clotting activity. The disease affects several mammals including dogs, and, like humans, is associated with high morbidity and mortality. In gene therapy using adeno-associated viral (AAV) vectors, the canine model has been one of the best predictors of the therapeutic dose tested in clinical trials for hemophilia B (factor IX deficiency) and other genetic diseases, such as congenital blindness. Here we report our experience with liver gene therapy with AAV-FVIII in two outbred, privately owned dogs with severe HA that resulted in sustained expression of 1–2% of normal FVIII levels and prevented 90% of expected bleeding episodes. A Thr62Met mutation in the F8 gene was identified in one dog. These data recapitulate the improvement of the disease phenotype in research animals, and in humans, with AAV liver gene therapy for hemophilia B. Our experience is a novel example of the benefits of a relevant preclinical canine model to facilitate both translational studies in humans and improved welfare of privately owned dogs.  相似文献   

19.
ABSTRACT: Over the past forty years the availability of coagulation factor replacement therapy has greatly contributed to the improved care of people with hemophilia. Following the blood-borne viral infections in the late 1970s and early 1980, caused by coagulation factor concentrates manufactured using non-virally inactivated pooled plasma, the need for safer treatment became crucial to the hemophilia community. The introduction of virus inactivated plasma-derived coagulation factors and then of recombinant products has revolutionized the care of these people. These therapeutic weapons have improved their quality of life and that of their families and permitted home treatment, i.e., factor replacement therapy at regular intervals in order to prevent both bleeding and the resultant joint damage (i.e. primary prophylaxis). Accordingly, a near normal lifestyle and life-expectancy have been achieved. The main current problem in hemophilia is the onset of alloantibodies inactivating the infused coagulation factor, even though immune tolerance regimens based on long-term daily injections of large dosages of coagulation factors are able to eradicate inhibitors in approximately two-thirds of affected patients. In addition availability of products that bypass the intrinsic coagulation defects have dramatically improved the management of this complication. The major challenges of current treatment regimens, such the short half life of hemophilia therapeutics with need for frequent intravenous injections, encourage the current efforts to produce coagulation factors with more prolonged bioavailability. Finally, intensive research is devoted to gene transfer therapy, the only way to ultimately obtain cure in hemophilia.  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