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 156 毫秒
1.
嵌合抗原受体T(CAR-T)细胞疗法是一种利用合成受体特异性靶向抗原的过继性细胞疗法(ACT),目前在血液肿瘤的治疗中有极大的临床应用价值。虽然美国食品药品监督管理局(FDA)已经批准两款CAR-T药物上市,但CAR-T疗法在治疗过程中仍然存在一些副作用,如细胞因子释放综合征(CRS)、神经毒性、B细胞功能缺失等。同时,CAR-T疗法在实体瘤治疗中的效果甚微,主要原因是缺乏特异性靶点以及肿瘤微环境对CAR-T细胞功能的抑制等。文中将从CAR的结构设计、临床应用、合成生物学对新型CAR的优化来阐述应用CAR-T细胞疗法治疗肿瘤所面临的挑战及广阔前景。  相似文献   

2.
自然杀伤(NK)细胞是一种重要的肿瘤杀伤性免疫细胞,在细胞免疫治疗中有着良好的应用前景。嵌合抗原受体(CAR)是一种人工修饰的融合蛋白,它可以通过胞外区特异性识别肿瘤并激活胞内区信号域。CAR-T细胞在临床中取得了巨大的成功,而NK细胞比T细胞在CAR其相对于T细胞的优势,并就NK细胞的来源、CAR-NK在临床中的应用及存在的挑战做简要综述。  相似文献   

3.
嵌合抗原受体(chimeric antigen receptors,CAR)细胞疗法已广泛用于白血病、淋巴瘤的治疗, CD19和CD22靶向CAR-T已在复发、难治性急性B淋巴细胞白血病(RR-B-ALL)等血液系统疾病的治疗上取得了显著疗效,而在T细胞肿瘤治疗上进展缓慢。介绍了目前国内外利用CAR细胞技术与CRISPR / Cas9基因编码技术,设计了T-ALL相关的CAR细胞免疫疗法并进行了CAR细胞免疫疗法在T-ALL治疗上的初步探索。  相似文献   

4.
癌症是威胁人类健康最主要的疾病之一,目前临床上主要采用的手术/放化疗联合治疗的疗效有限,因此如何进一步提高肿瘤临床疗效仍是巨大的挑战。免疫细胞过继是肿瘤治疗技术中迅速发展的一种生物治疗技术,通过输入自身或同种"抗肿瘤免疫效应细胞"达到直接杀伤肿瘤或增强机体自身细胞免疫功能的作用。由于γδT细胞对肿瘤细胞具有强有力的直接细胞毒性,并可与其他免疫细胞协同作用发挥其抗肿瘤活性,因而成为了细胞免疫治疗中新的研究热点。本文主要叙述了γδT细胞与肿瘤相关的基础研究及临床试验的最新进展。  相似文献   

5.
过继性免疫治疗(adoptive cellular immunotherapy,ACI)是将供体的淋巴细胞转移给受体,增强其细胞免疫功能。这种治疗是目前较为有效的恶性肿瘤的治疗方法之一,已在实体瘤和血液肿瘤的临床治疗中取得较好疗效。近年来发展的嵌合抗原受体(chimeric antigen receptor,CAR)T细胞是利用基因技术改造得来的,它表达肿瘤特异性,并显示出靶向性、杀伤活性和持久性,这为过继性细胞免疫治疗注入了新的解决方案。CAR发展到现在共经历过三代改进,前人也就每代CAR进行了大量临床试验,得出对临床治疗具有现实意义的实验结果。本文就CAR技术对B细胞淋巴瘤的作用影响作一综述。  相似文献   

6.
嵌合抗原受体T细胞免疫疗法(chimeric antigen receptor-T, CAR-T),是通过体外激活和扩增肿瘤特异或非特异性杀伤细胞达到抗肿瘤效果,在肿瘤免疫治疗方面具有良好的应用前景。本研究构建靶向EGFRⅢ(epidermal growth factor receptor variant III)的嵌合抗原受体(CAR)的重组慢病毒表达载体,利用慢病毒感染并筛选能够稳定表达该嵌合抗原受体的Jurkat细胞系。通过EGFRvⅢ分子刺激、与U87MG细胞共培养的方式检测细胞系的活化状况。结果显示,成功构建了pCDH-EGFRvⅢscFv-CAR-copGFP-T2A-puro慢病毒表达重组质粒,并筛选出可稳定表达EGFRⅢ-CAR的Jurkat 细胞系。CCK-8 法检测显示,EGFRvⅢ分子刺激12 h的Jurkat-CAR细胞增殖率约是对照组的1.36倍(P<0.05);ELISA法检测显示,与U87MG细胞共孵育后,细胞上清中IL-2的浓度约是单独培养分泌在上清中IL-2的1.625倍(P < 0.01)。以上结果表明,稳定表达CAR的jurkat细胞,可以靶向性识别EGFRvⅢ分子及EGFRvⅢ阳性的靶细胞,并引起IL-2细胞因子释放,为后续临床细胞免疫治疗提供了理论基础。  相似文献   

7.
多年来,癌症治疗一直是困扰人类的难题之一,常规的治疗手段都各有局限。随着基因转移和细胞培养技术的更新换代,细胞免疫治疗逐渐走入公众视线。嵌合抗原受体(chimeric antigen receptor,CAR)修饰的T淋巴细胞(CAR-T)治疗是几年来肿瘤过继免疫治疗的新方法,能特异性识别靶抗原,并杀伤肿瘤细胞。我们将CAR-T细胞在肿瘤治疗中的应用及构建CAR-T细胞的策略做简要综述。  相似文献   

8.
过继性细胞免疫治疗(adoptive cellular immunotherapy,ACI)是目前较为有效的恶性肿瘤的治疗方法之一。随着技术的日趋成熟,已在多种实体瘤和血液肿瘤的t临床治疗中取得较好疗效。其中,嵌合抗原受体(chimeric antigen receptor,CAR)T细胞技术是近年来发展非常迅速的一种细胞治疗技术。通过基因改造技术,效应T细胞的靶向性、杀伤活性和持久性均较常规应用的免疫细胞高,并可克服肿瘤局部免疫抑制微环境和打破宿主免疫耐受状态。目前,CAR的信号域已从第一代的单一信号分子发展为包含CD28、4—1BB等共刺激分子的多信号结构域(第二、三代),临床应用广泛。但是,该技术也存在脱靶效应、插入突变等临床应用风险。该文将就CAR—T细胞技术在恶性肿瘤免疫治疗中的应用及可能存在的问题作一综述。  相似文献   

9.
嵌合抗原受体T细胞免疫疗法(chimeric antigen receptor T-cell immunotherapy,CAR-T)是近年来迅速发展的肿瘤过继免疫治疗方法,其胞外段抗体以非主要组织相容性复合物(major histocompatibility complex,MHC)方式与相应的肿瘤相关抗原结合识别后,使T细胞活化而发挥抗肿瘤效应。CAR-T在血液疾病治疗中取得较好的效果,现就CAR的结构、CAR-T治疗的靶点、出现的不良反应及采取的相应策略等作一概述。  相似文献   

10.
嵌合抗原受体T细胞免疫疗法(CAR-T疗法)是一种治疗肿瘤的新免疫疗法,通过向患者自身T细胞中导入已被修饰的CAR基因,使T细胞表达结合肿瘤表面抗原的特异性受体来实现对肿瘤的精准治疗.目前已发展到第四代.该免疫疗法在血液瘤和实体瘤治疗中都有一定疗效,同时也存在一些待解决难题.本文就近年来CAR-T在血液瘤和实体瘤中的研究治疗进展及存在的问题进行综述.  相似文献   

11.
Glioblastoma multiforme (GBM) is the most common primary malignant cancer of brain, which is extremely aggressive and carries a dreadful prognosis. Current treatment protocol runs around radiotherapy, surgical resection, and temozolomide with median overall survival of around 12–15 months. Due to its heterogeneity and mutational load, immunotherapy with chimeric antigen receptor (CAR) T cell therapy can be a promising treatment option for recurrent glioblastoma. Initial phase 1 studies have shown that this therapy is safe without dose-limiting side effects and it also has a better clinical outcome. Therefore, CAR T cell therapy can be a great future tool in our armamentarium to treat advanced GBM. In this article, we have explained the structure, mechanism of action, and rationale of CAR T cell therapy in GBM; we also discussed various antigenic targets and clinical outcome of initial studies of this novel therapy.  相似文献   

12.
The adoptive transfer of chimeric antigen receptor (CAR)-expressing T cells is a relatively new but promising approach in the field of cancer immunotherapy. This therapeutic strategy is based on the genetic reprogramming of T cells with an artificial immune receptor that redirects them against targets on malignant cells and enables their destruction by exerting T cell effector functions. There has been an explosion of interest in the use of CAR T cells as an immunotherapy for cancer. In the pre-clinical setting, there has been a considerable focus upon optimizing the structural and signaling potency of the CAR while advances in bio-processing technology now mean that the clinical testing of these gene-modified T cells has become a reality. This review will summarize the concept of CAR-based immunotherapy and recent clinical trial activity and will further discuss some of the likely future challenges facing CAR-modified T cell therapies.  相似文献   

13.
The host immune system plays an instrumental role in the surveillance and elimination of tumors by recognizing and destroying cancer cells. In recent decades, studies have mainly focused on adoptive immunotherapy using engineered T cells for the treatment of malignant diseases. Through gene engraftment of the patient’s own T cells with chimeric antigen receptor (CAR), they can recognize tumor specific antigens effectively and eradicate selectively targeted cells in an MHC-independent fashion. To date, CAR-T cell therapy has shown great clinical utility in patients with B-cell leukemias. Owing to different CAR designs and tumor complex microenvironments, genetically redirected T cells may generate diverse biological properties and thereby impact their long-term clinical performance and outcome. Meanwhile some unexpected toxicities that result from CAR-T cell application have been examined and limited the curative effects. Diverse important parameters are closely related with adoptively transferred cell behaviors, including CAR-T cells homing, CAR constitutive signaling, T cell differentiation and exhaustion. Thus, understanding CARs molecular design to improve infused cell efficacy and safety is crucial to clinicians and patients who are considering this novel cancer therapeutics. In this review, the developments in CAR-T cell therapy and the limitations and perspectives in optimizing this technology towards clinical application are discussed.  相似文献   

14.
胃癌是目前世界上发病率及致死率较高的恶性肿瘤之一,在东亚地区尤其显著。针对胃癌的治疗手段仍是传统的手术联合化疗、放疗为主,尽管靶向药物治疗提供了新的选择,但其对晚期胃癌的疗效仍然有限。胃癌的免疫治疗作为独特的治疗手段,在近十多年发展较为活跃,特别是过继性免疫治疗手段不断有创新。过继性免疫治疗主要依赖回输具有抗肿瘤活性的细胞,目前回输的细胞由具有非特异性抗肿瘤作用向具有特异性抗肿瘤作用演变,特别是嵌合性抗原T细胞治疗的出现,为进展期胃癌患者提供了有一种潜在的选择。本文对胃癌过继性免疫治疗中采用的不同免疫活性细胞的作用机制、临床应用等进行总结,并针对其不足提出利用基因工程技术增强治疗靶向性、降低免疫逃逸的研究方向。  相似文献   

15.
Chimeric antigen receptor (CAR) T-cell therapy is an immunotherapy approach that has played a tremendous role in the battle against cancer for years. Since the CAR T lymphocytes are unrestricted-major histocompatibility complex T lymphocytes, they could identify more targets than natural T cells, resulting in practical and widespread functions. The good prospects of CAR T-cell therapy in oncology can be additionally applied to treat other diseases such as autoimmune and infectious diseases. CAR-T cell-derived immunotherapy for autoimmune disorders can be allocated to CAR-Tregs and chimeric autoantibody receptor T cells. Other generations of CARs target human immunodeficiency virus (HIV) proteins. In this review, we summarize CAR-T cell therapies in autoimmune disorders and HIV infection.  相似文献   

16.
Recent reports on the impressive efficacy of chimeric antigen receptor (CAR)-modified T cells against hematologic malignancies have inspired oncologists to extend these efforts for the treatment of solid tumors. Clinical trials of CAR-T-based cancer immunotherapy for solid tumors showed that the efficacies are not as remarkable as in the case of hematologic malignancies. There are several challenges that researchers must face when treating solid cancers with CAR-T cells, these include choosing an ideal target, promoting efficient trafficking and infiltration, overcoming the immunosuppressive microenvironment, and avoiding associated toxicity. In this review, we discuss the obstacles imposed by solid tumors on CAR-T cell-based immunotherapy and strategies adopted to improve the therapeutic potential of this approach. Continued investigations are necessary to improve therapeutic outcomes and decrease the adverse effects of CAR-T cell therapy in patients with solid malignancies in the future.  相似文献   

17.
肿瘤细胞免疫疗法近年来的发展颇为瞩目,嵌合抗原受体T(CAR-T)的临床研究显示其对血液系统肿瘤具有良好的治疗效果。自然杀伤细胞(NK)是人体固有免疫的一类重要细胞,其不同于T细胞的非特异性识靶及杀伤机制吸引科学家将工程CAR-T技术沿袭并用于嵌合抗原受体NK(CAR-NK)改造。目前,无论在体外细胞模型还是小鼠动物模型中,CAR-?NK均显示出良好的肿瘤杀伤效果。最新的临床研究显示,CAR-NK细胞对血液系统肿瘤有良好的治疗效果,但治疗实体瘤效果尚待验证。与CAR-T细胞疗法一样,CAR-NK也有问题亟需解决,但是NK细胞作为效应细胞,其自身优点预示CAR-NK细胞在实体瘤治疗方面拥有良好的发展前景。  相似文献   

18.
In the past 50 years, disease burden has steadily shifted from infectious disease to cancer. Standard chemotherapy has long been the mainstay of cancer medical management, and despite vast efforts towards more targeted and personalized drug therapy, many cancers remain refractory to treatment, with high rates of relapse and poor prognosis. Recent dramatic immunotherapy clinical trials have demonstrated that engineering T-cells with chimeric antigen receptors (CARs) to target CD19 can lead to complete remission in relapsed or refractory B-cell malignancies, generating a great deal of enthusiasm in the field. Here we provide a comprehensive overview of the history of adoptive T-cell therapy, including CARs, in solid tumors as well as hematologic malignancies. CAR therapy has the potential to fundamentally transform cancer treatment with specific and even personalized targeting of tissue- and tumor-specific antigens. However, before CARs become standard first-line treatment modalities, critical issues regarding efficacy, combinatorial regimens, and mechanisms of treatment failure and toxicity will need to be addressed.  相似文献   

19.
Natural killer (NK) cells are an important subset of lymphocytes which play a critical role in host immunity against cancers. With MHC-independent recognition, short lifespan and potent cytotoxicity, NK cells make a promising candidate for chimeric antigen receptor (CAR)-engineered cancer immunotherapy. Due to innate biological properties of NK cells, CAR-NK may outperform CAR-T therapy in terms of less side effects and more universal access, which may become a great reformation in CAR-based cancer immunotherapy. The CARs used in peripheral blood (PB) NK cells as well as NK cell line like NK-92 are the most important outfits defining antigenic specificity. The constructs of CARs used in NK cells from different sources vary, which all undergo generational optimization. The anti-tumor effects of CAR-NK have been validated in numerous preclinical trials for cancers, including hematologic malignancies and many solid tumors, which provide evidence for potential clinical application of CAR-NK. Additionally, this review concludes the challenges faced in the application of CAR-NK. Although CAR-NK is considered as one of the most possible “off-the-shelf” products, the improvement for the efficiency of expansion and transduction as well as the solution for underlying safety issues is still needed. Possible coping strategies for challenges and upgrades in techniques are also highlighted for future development in CAR-NK cancer immunotherapy.  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