首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 78 毫秒
1.
唐海军  刘玉琴 《解剖学报》2017,48(1):115-120
嵌合抗原受体修饰T细胞(CAR-T)是目前恶性肿瘤免疫治疗的一种新方法。CAR-T细胞对肿瘤的杀伤不依赖主要组织相容性复合体(MHC),并可克服肿瘤局部免疫抑制微环境和突破宿主免疫耐受状态,因此,CAR-T细胞在治疗肿瘤方面具有独特的优势。CAR-T细胞的构建和选择合适的靶分子是CAR-T细胞免疫治疗的两个关键的问题,我们在文中将围绕这两个问题做一综述。  相似文献   

2.
从1982年人类第一次诱导出淋巴因子激活的杀伤细胞(lymphokine active killer cell, LAK),到2010年嵌合抗原受体T细胞(chimeric antigen receptor T-cell, CAR-T)免疫疗法第一次由Rosenberg博士应用于慢性淋巴细胞白血病治疗并获得成功,再到2...  相似文献   

3.
嵌合抗原受体T(CAR-T)细胞是一种通过基因编辑技术表达多功能合成受体的T细胞.CAR-T细胞在治疗肿瘤尤其是B淋巴母细胞白血病等血液系统恶性肿瘤中取得了巨大的成功,虽然在实体肿瘤的治疗中存在治疗效果有限、易复发等问题,CAR-T细胞治疗也已成为肿瘤治疗的前沿热点.随着CAR-T细胞技术的不断发展,这种细胞特异性疗法...  相似文献   

4.
嵌合抗原受体T细胞(chimeric antigen receptor T cell, CAR-T)疗法是近年来新兴的细胞免疫疗法,它通过基因工程技术使T细胞表达特异性嵌合抗原受体(chimeric antigen receptor, CAR),以非MHC依赖形式识别并杀伤表达相应抗原的靶细胞。CAR-T疗法在血液系统恶性肿瘤的治疗中取得了令人振奋的结果,但在实体瘤治疗领域仍然存在很多困难。溶瘤病毒可直接裂解实体瘤细胞,逆转肿瘤局部的免疫抑制微环境,与CAR-T疗法联用可以增强抗实体瘤效果。此外,CAR-T疗法也显示出一定的抗病毒作用,主要表现在抗人类免疫缺陷病毒(human immunodeficiency virus, HIV)、HBV等方面。文章从CAR-T与病毒的关系入手,主要就CAR-T疗法抗病毒的研究现状及溶瘤病毒与CAR-T疗法联合抗实体瘤机制等方面展开综述,以期为拓宽CAR-T抗病毒治疗领域以及CAR-T的联合治疗方案制定提供新思路。  相似文献   

5.
目的 构建程序死亡受体1(PD-1)敲除及磷脂酰肌醇蛋白多糖-3(GPC3)修饰的嵌合抗原受体T细胞(GPC3-PD1gRNA-CART cells),研究其对肝癌细胞株HepG2的体外杀伤作用,以及其对肝癌动物模型的体内抗肿瘤作用.方法 制备GPC3-PD1gRNA-CART细胞,用流式细胞仪检测PD-1、GPC3 ...  相似文献   

6.
许卓  唐铁军  汤磊  郭薇 《现代免疫学》2022,(3):243-247+272
嵌合抗原受体T细胞(chimeric antigen receptor T cell, CAR-T)免疫疗法作为新型的免疫疗法发展迅速,显示出良好的疗效,并已被批准用于治疗血液系统恶性肿瘤。然而CAR-T免疫治疗在实体瘤中的疗效却未有较大突破。以CAR-T免疫疗法为基础的联合治疗近年来已成为研究的新热点,并为CAR-T免疫疗法治疗血液系统肿瘤和实体瘤提供了广阔的应用前景。CAR-T免疫疗法与免疫检查点、溶瘤病毒等免疫疗法的联合应用,抑制了肿瘤细胞的免疫逃逸,可从不同角度提高机体的抗肿瘤免疫力,显示出了协同增效的作用。CAR-T免疫疗法联合放疗、化疗等传统疗法可以重塑肿瘤免疫微环境,增强抗肿瘤作用。建立以CAR-T为基础的治疗模式是肿瘤免疫治疗的一个新方向,也是对肿瘤治疗新模式的探索。该文概述CAR-T免疫疗法与其他疗法联合治疗的进展,并介绍它们目前的应用状况。  相似文献   

7.
从1989年Gross与Eshhar提出嵌合型抗原受体(CAR)T细胞的理念,到2014年FDA授予CTL019“突破性治疗”荣誉,CAR-T细胞治疗肿瘤已走过25年.从早期的单一CD3ζ结构域到后来的串联CD28、CD137等,随着人类对肿瘤免疫研究的深入,以及临床试验研究开展,CAR T细胞治疗得到不断优化.虽然CD19为靶点的淋巴瘤治疗获得成功,但实体肿瘤治疗仍处于临床试验摸索阶段.基于促进CAR T细胞向肿瘤局部迁移浸润以及克服抗肿瘤微环境的免疫抑制作用等理念及技术优化,CAR T治疗实体肿瘤的研究越来越深入.未来CAR T治疗的适应症将不断扩大.  相似文献   

8.
肿瘤抗原特异性T细胞受体(TCR)基因转导技术作为肿瘤过继性免疫治疗的新手段之一,细胞和动物实验都证明它有很好的抗肿瘤作用,但临床试验结果却不尽人意。影响临床疗效主要原因是肿瘤抗原特异性T细胞受体不能有效地表达于T淋巴细胞表面,因而探索提高抗原特异性TCR高效表达的方法可能是解决这种问题的必要途径。  相似文献   

9.
肿瘤抗原特异性T细胞受体(TCR)基因转导技术作为肿瘤过继性免疫治疗的新手段之一,细胞和动物实验都证明它有很好的抗肿瘤作用,但临床试验结果却不尽人意.影响临床疗效主要原因是肿瘤抗原特异性T细胞受体不能有效地表达于T淋巴细胞表面,因而探索提高抗原特异性TCR高效表达的方法可能是解决这种问题的必要途径.  相似文献   

10.
肿瘤抗原特异性T细胞受体(TCR)基因转导技术作为肿瘤过继性免疫治疗的新手段之一,细胞和动物实验都证明它有很好的抗肿瘤作用,但临床试验结果却不尽人意.影响临床疗效主要原因是肿瘤抗原特异性T细胞受体不能有效地表达于T淋巴细胞表面,因而探索提高抗原特异性TCR高效表达的方法可能是解决这种问题的必要途径.  相似文献   

11.
While the clinical progress of chimeric antigen receptor T cell (CAR-T) immunotherapy has garnered attention to the field, our understanding of the biology of these chimeric molecules is still emerging. Our aim within this review is to bring to light the mechanistic understanding of these multi-modular receptors and how these individual components confer particular properties to CAR-Ts. In addition, we will discuss extrinsic factors that can be manipulated to influence CAR-T performance such as choice of cellular population, culturing conditions and additional modifications that enhance their activity particularly in solid tumors. Finally, we will also consider the emerging toxicity associated with CAR-Ts. By breaking apart the CAR and examining the role of each piece, we can build a better functioning cellular vehicle for optimized treatment of cancer patients.  相似文献   

12.
Chimeric antigen receptors (CARs) are fusion molecules that may be genetically delivered ex-vivo to T-cells and other immune cell populations, thereby conferring specificity for native target antigens found on the surface of tumour and other target cell types. Antigen recognition by CARs is neither restricted by nor dependent upon human leukocyte antigen antigen expression, favouring widespread use of this technology across transplantation barriers. Signalling is delivered by a designer endodomain that provides a tailored and target-dependent activation signal to polyclonal circulating T-cells. Recent clinical data emphasise the enormous promise of this emerging immunotherapeutic strategy for B-cell malignancy, notably acute lymphoblastic leukaemia. In that context, CARs are generally targeted against the ubiquitous B-cell antigen, CD19. However, CAR T-cell immunotherapy is limited by potential for severe on-target toxicity, notably due to cytokine release syndrome. Furthermore, efficacy in the context of solid tumours remains unproven, owing in part to lack of availability of safe tumour-specific targets, inadequate CAR T-cell homing and hostility of the tumour microenvironment to immune effector deployment. Manufacture and commercial development of this strategy also impose new challenges not encountered with more traditional drug products. Finally, there is increasing interest in the application of this technology to the treatment of non-malignant disease states, such as autoimmunity, chronic infection and in the suppression of allograft rejection. Here, we consider the background and direction of travel of this emerging and highly promising treatment for malignant and other disease types.  相似文献   

13.
《Immunobiology》2020,225(1):151850
Glypican-3 (GPC3) is an attractive target for chimeric antigen receptor (CAR)-T cell therapy, as it is overexpressed in most hepatocellular carcinoma (HCC) tissues but shows restricted expression in healthy adult tissues. Herein, we generated GPC3-specific CAR-T cells for HCC therapy by electroporation with plasmid DNA encoding the piggyBac (PB) transposon and the hyperactive piggyBac transposase simultaneously instead of by commonly-used viral vectors. Our results demonstrated that GPC3CAR gene was efficiently integrated into the genome of T cells utilizing the PB transposon system. Upon stimulation with GPC3 antigen, GPC3CAR-T cells could be effectively activated, proliferate strongly and secrete high levels of cytokines. It also was demonstrated that GPC3CAR-T cells displayed potent cytotoxicity against GPC3-positive HCC cell lines in vitro by using real-time cell analyser (RTCA) system and the JuLI™ Stage Cell History Recorder. More importantly, in a Huh-7 xenograft mouse model, GPC3CAR-T cells significantly reduced the tumour burden companied with the secretion of high levels of IFN-γ. Moreover, T cells in mice treated with GPC3CAR-T cells could infiltrate into tumour tissues and persist as effector memory T cells (TEM). Overall, our study suggests that the use of PB system-based GPC3CAR-T cell therapy could be a promising clinical strategy for patients with HCC.  相似文献   

14.
c-Met is a hepatocyte growth factor receptor overexpressed in many tumors such as hepatocellular carcinoma(HCC).Therefore,c-Met may serve as a promising target for HCC immunotherapy.Modifying T cells to express c-Met-specific chimeric antigen receptor(CAR)is an attractive strategy in treating c-Met-positive HCC.This study aimed to systematically evaluate the inhibitory effects of 2nd-and 3rd-generation c-Met CAR-T cells on hepatocellular carcinoma(HCC)cells.Here,2nd-and 3rd-generation c-Met CARs containing an anti-c-Met singlechain variable fragment(scFv)as well as the CD28 signaling domain and CD3ζ(c-Met-28-3ζ),the CD137 signaling domain and CD3ζ(c-Met-137-3ζ),or the CD28 and CD137 signaling domains and CD3ζ(c-Met-28-137-3ζ)were constructed,and their abilities to target c-Met-positive HCC cells were evaluated in vitro and in vivo.All c-Met CARs were stably expressed on T cell membrane,and c-Met CAR-T cells aggregated around c-Met-positive HCC cells and specifically killed them in vitro.c-Met-28-137-3ζCAR-T cells secreted more interferon-gamma(IFN-γ)and interleukin 2(IL-2)than c-Met-28-3ζCAR-T cells and c-Met-137-3ζCAR-T cells.Compared with c-Met low-expressed cells,c-Met CAR-T cells secreted more cytokines when co-cultured with c-Met high-expressed cells.Moreover,c-Met-28-137-3ζCAR-T cells eradicated HCC more effectively in xenograft tumor models compared with the control groups.This study suggests that 3rd-generation c-Met CAR-T cells are more effective in inhibiting c-Met-positive HCC cells than 2nd-generation c-Met CAR-T cells,thereby providing a promising therapeutic intervention for c-Met-positive HCC.  相似文献   

15.
嵌合抗原受体(chimericantigenreceptor,CAR)修饰T细胞是目前肿瘤治疗中新的靶向疗法,通过单链抗体(singlechainfragmentvariable,scFv)-共刺激分子-T细胞信号转导区构成的嵌合模式修饰T细胞,赋予T细胞非MHC依赖性靶向杀伤肿瘤细胞的能力,在动物模型及临床试验中取得了良好的效果.糖蛋白类肿瘤标志物由于其良好的靶向性成为了CAR修饰T细胞新的靶点,针对糖蛋白类肿瘤标志物的靶向研究显示出良好的临床前景,但是也考虑到脱靶效应,转染方式等问题对该治疗在临床运用的限制.相信随着研究的逐渐深入,基于CAR修饰T细胞的糖蛋白类肿瘤标志物靶向治疗会取得更大的突破.  相似文献   

16.
Chimeric antigen receptor (CAR)-engineered T-cell (CAR-T) therapy has demonstrated impressive therapeutic efficacy against hematological malignancies, but multiple challenges have hindered its application, particularly for the eradication of solid tumors. Innate killer cells (IKCs), particularly NK cells, NKT cells, and γδ T cells, employ specific antigen-independent innate tumor recognition and cytotoxic mechanisms that simultaneously display high antitumor efficacy and prevent tumor escape caused by antigen loss or modulation. IKCs are associated with a low risk of developing GVHD, thus offering new opportunities for allogeneic “off-the-shelf” cellular therapeutic products. The unique innate features, wide tumor recognition range, and potent antitumor functions of IKCs make them potentially excellent candidates for cancer immunotherapy, particularly serving as platforms for CAR development. In this review, we first provide a brief summary of the challenges hampering CAR-T-cell therapy applications and then discuss the latest CAR-NK-cell research, covering the advantages, applications, and clinical translation of CAR- and NK-cell receptor (NKR)-engineered IKCs. Advances in synthetic biology and the development of novel genetic engineering techniques, such as gene-editing and cellular reprogramming, will enable the further optimization of IKC-based anticancer therapies.  相似文献   

17.
Abstract

Background: Immunotherapy utilizing T cells genetically modified to express chimeric antigen receptors (CARs) is rapidly emerging as a promising novel treatment for hematological and nonhematological malignancies. In order to target the TKI-insensitive leukemia stem cells (LSCs) in chronic myeloid leukemia (CML) by CAR T cells, we chose CD26 as a cell surface tumor-associated antigen due to preferentially expression on LSCs. Additionally, CD26 has also been suggested to be a multipurpose therapeutic target for other cancer. Therefore, developing the CD26-targeting CAR T cells may be a promising therapy for not only LSCs but also other CD26+ cancer cells.

Methods: We designed the second-generation CD26-targeting CAR utilizing 4-1BB (CD137) as costimulatory domain, and transduced T cells with CD26-CAR containing lentiviral. Then we evaluated the transduction efficiency and expansion ability, and demonstrated the existence of self-antigen-driven fratricide by cytokine assay and cytotoxicity assay.

Results: Anti-CD26-4-1BB-CAR T cells exhibited poor viability, multiple cytokine secretion, down-regulation of CD26 and direct cytotoxicity against themselves, indicating self-antigen-driven fratricide.

Conclusion: Eradicating CML-LSCs via anti-CD26-4-1BB-CAR T cells is not applicable, and optimized design or alternative target is needed.  相似文献   

18.
Tumors originate from a number of genetic events that deregulate homeostatic mechanisms controlling normal cell behavior. The immune system, devoted to patrol the organism against pathogenic events, can identify transformed cells, and in several cases cause their elimination. It is however clear that several mechanisms encompassing both central and peripheral tolerance limit antitumor immunity, often resulting into progressive diseases. Adoptive T‐cell therapy with either allogeneic or autologous T cells can transfer therapeutic immunity. To date, genetic engineering of T cells appears to be a powerful tool for shaping tumor immunity. In this review, we discuss the most recent achievements in the areas of suicide gene therapy, and TCR‐modified T cells and chimeric antigen receptor gene‐modified T cells. We provide an overview of current strategies aimed at improving the safety and efficacy of these approaches, with an outlook on prospective developments.  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