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 187 毫秒
1.
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   

2.
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   

3.
嵌合抗原受体(chimeric antigen receptor,CAR)是运用重组DNA技术制备的基因工程抗体,由单链抗体、协同刺激分子及T细胞信号转导分子等部分融合而成。全外显子测序技术是传统的c DNA文库表达血清学方法之外的筛选肿瘤抗原的新方法。近年来,嵌合抗原受体-T细胞(chimeric antigen receptor-T cells,CAR-T细胞)在治疗包括实体瘤在内的一系列恶性肿瘤中取得了较大的成就。临床试验表明,CAR-T细胞在产生强大抗肿瘤效应的同时,也具有不容忽视的毒副反应。该文将讨论嵌合抗原受体-T细胞治疗恶性肿瘤基本原理、关键技术和面临的挑战。  相似文献   

4.
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   

5.
T 细胞介导的肿瘤免疫至少需要 T 细胞受体和共刺激分子“双信号”参与的学说目前得到了广泛支持。共抑制或共刺激分子提供的 共信号决定了 T 细胞受体信号介导的免疫应答的最终效应。近年来,靶向共抑制分子如 CTLA-4 和 PD-1 开发的抗体药物在临床应用中获 得了巨大成功,使得肿瘤免疫治疗成为最令人瞩目的研究领域,并被美国《科学》杂志评为 2013 年度十大科学突破之首。肿瘤免疫治疗有 望成为与手术、放化疗和靶向治疗并驾齐驱的抗肿瘤主流治疗方案。针对共抑制分子 CTLA-4、PD-1、PD-L1 和共刺激分子 CD137,综述 其发挥免疫调节作用的分子机制及其相关靶向药物在肿瘤治疗方面的最新进展和应用。  相似文献   

6.
急性髓系白血病(AML)是一种预后较差的血液系统恶性肿瘤,迫切需要新的治疗手段。随着嵌合抗原受体(CAR)T细胞(CAR-T)技术的发展并在B细胞肿瘤中取得显著疗效,人们把目光投入更多的实体瘤与血液肿瘤靶点中。CD123分子是CAR-T治疗AML的潜在靶点,抗CD123 CAR-T具有靶向清除白血病干细胞(LSCs)及原始细胞的能力。本研究总结了目前在靶向CD123 CAR-T治疗急性髓系白血病目前的研究进展。  相似文献   

7.
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   

8.
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   

9.
肿瘤严重威胁着人类健康,当前肿瘤传统的治疗方法有手术治疗、化疗、放疗和靶向药物治疗等。近年来,肿瘤免疫治疗,尤其是嵌合抗原受体(chimeric antigen receptor,CAR) T细胞免疫疗法在基础研究与临床应用中蓬勃发展,并在治疗血液系统恶性肿瘤方面取得了巨大成功。然而,大量研究显示,细胞免疫治疗后可出现不同程度的毒副反应,且部分患者缓解后再次复发。因此,了解细胞治疗面临的挑战与局限性,寻找解决的办法,对继续发挥细胞免疫疗法的潜能具有重要意义。本文就免疫细胞的CAR结构、病毒载体的选择、细胞治疗面临的挑战及前景进行综述。  相似文献   

10.
嵌合抗原受体修饰的T细胞(CAR-T)治疗是过继免疫治疗的一种,通常CAR-T细胞是由患者或供者的T细胞经CAR基因转化并扩增而来,最后再回输到患者体内.CAR-T在治疗B细胞恶性肿瘤中展现出了振奋人心的效果,目前也被用于治疗其他血液肿瘤的研究中.本文就CAR的基本结构、CAR结构的演变、在急性白血病治疗中的进展以及目前CAR-T设计新方案4个方面进行叙述.  相似文献   

11.
Novel immunotherapeutic agents targeting tumor-site microenvironment are revolutionizing cancer therapy. Chimeric antigen receptor (CAR)-engineered T cells are widely studied for cancer immunotherapy. CD19-specific CAR-T cells, tisagenlecleucel, have been recently approved for clinical application. Ongoing clinical trials are testing CAR designs directed at novel targets involved in hematological and solid malignancies. In addition to trials of single-target CAR-T cells, simultaneous and sequential CAR-T cells are being studied for clinical applications. Multi-target CAR-engineered T cells are also entering clinical trials. T cell receptor-engineered CAR-T and universal CAR-T cells represent new frontiers in CAR-T cell development. In this study, we analyzed the characteristics of CAR constructs and registered clinical trials of CAR-T cells in China and provided a quick glimpse of the landscape of CAR-T studies in China.  相似文献   

12.
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   

13.
《Biotechnology advances》2019,37(7):107411
T cells engineered to express a chimeric antigen receptor (CAR) have re-shaped the way hematological malignancies are treated. Despite the overwhelming early clinical success, CAR-T therapies are associated with severe side-effects, disease relapse and often exhibit limited efficacy. In this Review article we summarize the most recent biotechnological advances that have been developed to enhance the efficacy and specificity of CAR-T therapies, as well as to address the key challenges associated with them. We place particular emphasis on the most recent clinical data that indicate which CAR-T populations are the most relevant to clinical success, and indicate how the molecular structure of the CAR receptor can affect clinical outcome. Finally, we outline what we believe is the next generation of immunotherapies.  相似文献   

14.
CAR-T cell therapy has already achieved world-renowned clinical effects in the treatment of hematological malignancies. Due to the tumor heterogeneity, immunosuppressive microenvironment, and other factors, CAR-T cell therapy has still not shown obvious clinical efficacy in clinical treatment of solid tumors. However, great progress has been made in the preparation of CAR-T cells in recent years, including T cells redirected for universal cytokine mediated killing, universal CAR -T cells, non-viral vector CAR-T cells, SynNotch technology, SUPRA CAR technology, regulated CAR-T cells, and bi-specific CAR-T cells, etc. Future research and development of CAR-T cell therapy will be focused on these following aspects: the combined application of CAR-T cells with different targets, known as "Cocktail CAR-T cells", is expected to increase efficiency toward solid tumors; based on systemic biology/synthetic biology theories, CAR-T cells are likely to be transformed to robot or intelligent system by introducing sensors, logic gates, and logic circuits. This article mainly comments on research progress and perspectives on CAR-T cell therapy in solid tumor treatment.  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.
Hepatocellular carcinoma (HCC), the most common primary liver cancer has a high mortality in China, and it is usually diagnosed at a late stage, thereby leaving patients with few effective treatment options. Chimeric antigen receptor-T (CAR-T) cell therapy, a novel immunotherapy that has shown promising results in leukemia, lymphoma and multiple myeloma, is also expected to work well in solid tumors, including HCC. However, the ideal therapeutic efficacy has not yet been achieved, in part due to tumor antigen escape caused by antigen heterogeneity. To overcome such challenge, we screened a panel of biomarkers in HCC cell lines and found that GPC3 and B7H3 were highly expressed on HCC with expression heterogeneity. Then we developed a novel bispecific T cell engagers CAR-T (CAR.T-BiTEs) that drives the expression of a CAR specific for GPC3 and BiTEs against CD3 and B7H3, herein referred to as “GPC3-BiTE CAR.” We found that BiTEs promoted the increased activation of untransduced T cells and IFN-γ release. Moreover, BiTEs secreted by GPC3-BiTE CAR-HEK293T cells promoted increased cytotoxicity activity of untransduced T cells against GPC3+/B7H3+ (GPC3 positive/B7H3 positive) and GPC3-/B7H3+(GPC3 negative/B7H3 positive) HCC cell lines. In vitro function assays showed that GPC3-BiTE CAR-T cells exhibited greater cytotoxicity activity against GPC3+/B7H3+ HCC cell lines than GPC3 CAR-T cells (GPC3-targeted CAR-T cells) and B7H3 CAR-T cells (B7H3-targeted CAR-T cells). Furthermore, GPC3-BiTE CAR-T cells exhibited superior cytotoxicity against GPC3 negative HCC cell lines compared with GPC3 CAR T cells. In conclusion, our study showed that GPC3-BiTE CAR T cells exhibited superior antitumor activity than single-target CAR-T cells and can overcome tumor escape induced by antigen heterogeneity, suggesting that this could be a promising therapeutic strategy for HCC.  相似文献   

17.
基于嵌合抗原受体修饰的T细胞(CAR-T)的过继免疫疗法已被证明是治疗恶性肿瘤最有希望的策略之一,但是目前其在实体瘤中的应用依然有限。研究表明磷脂酰肌醇蛋白聚糖3 (GPC3)对肝细胞癌来说是一种有意义的诊断、治疗和预后生物标志物,且已有利用第二代/第三代GPC3靶向的CAR-T细胞治疗肝细胞癌的研究报道。为了进一步提高治疗效果,构建同时表达GPC3 CAR、人源IL-7和CCL19细胞因子的第四代慢病毒载体,转染293T细胞包装慢病毒并感染人T淋巴细胞制备靶向GPC3的第四代CAR-T细胞(GPC3-BBZ-7×19)。利用细胞计数、趋化小室、荧光素酶生物发光法以及流式细胞术等比较其与第二代GPC3 CAR-T细胞(GPC3-BBZ)在增殖、迁移、杀伤以及亚型分布方面的区别,评估GPC3-BBZ-7×19 CAR-T细胞对免疫缺陷小鼠体内GPC3阳性的肝细胞癌腹腔移植瘤生长的作用。结果表明与GPC3-BBZ CAR-T细胞相比,GPC3-BBZ-7×19 CAR-T细胞具备更强的增殖能力、趋化能力以及记忆干细胞(Stem memory T cell,Tscm)组成比(P值均0.05),而在体外特异性杀伤GPC3阳性的肝细胞癌细胞以及细胞因子分泌水平方面无显著差异。此外,GPC3-BBZ-7×19 CAR-T细胞能够明显抑制免疫缺陷小鼠体内的GPC3阳性肝细胞癌移植瘤模型。因此靶向GPC3的第四代CAR-T细胞(分泌IL-7和CCL19)有望比第二代GPC3靶向的CAR-T细胞更持久有效地对抗肿瘤,并产生针对肿瘤的记忆功能,为以后的临床试验提供了临床前研究基础。  相似文献   

18.
BackgroundChimeric antigen receptor (CAR)–T cell therapy opens a new era for cancer treatment. However, in prolonged follow-up, relapse has emerged as one of the major obstacles. Dendritic cell (DC) vaccination is a promising treatment to eradicate tumor cells and prevent relapse. The epidermal growth factor receptor (EGFR) pathway substrate 8 (Eps8) gene is involved in regulating cancer progression and is considered an attractive target for specific cancer immunotherapy. The purpose of this study was to explore a combinatorial therapy using CAR-T cells and a DC vaccine such as Eps8-DCs to increase leukemia treatment efficacy.MethodsWe pulsed DCs with Eps8-derived peptides to generate Eps8-DCs, engineered T cells to express a second-generation CAR specific for CD19, and analyzed the effects of the Eps8-DCs on the in vitro expansion, phenotype and effector functions of the CD19 CAR-T cells.ResultsThe Eps8-DCs significantly reduced the activation-induced cell death and enhanced the proliferative potential of CAR-T cells during in vitro expansion. In addition, the expanded T cells co-cultured with the Eps8-DCs exhibited an increased percentage of central memory T cells (Tcms) and a decreased percentage of effector memory T cells (Tems). The Eps8-DCs enhanced CD19 CAR-T cell immune functions, including cytokine production, CD107a degranulation activity and cytotoxicity.DiscussionThis study demonstrates that Eps8-DCs exert synergistic effect on CD19 targeting CAR-T cells and paves the way for clinical trials using the combination of DC vaccination and engineered T cells in relapsed leukemia.  相似文献   

19.
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   

20.
以嵌合抗原受体修饰T细胞(chimeric antigen receptor modified-T lymphocytes,CAR-T)为基础的过继细胞免疫治疗近年来成为治疗恶性肿瘤的一种新模式。CAR-T细胞赋予T细胞靶向杀伤活性,并可克服肿瘤免疫抑制和打破宿主免疫耐受。尽管目前CAR-T的临床应用还存在诸如脱靶效应、细胞因子风暴、插入突变和移植物抗宿主病样损伤等许多问题,但我们相信随着对CAR-T细胞的深入研究及推广应用将会大幅度提高肿瘤的临床治疗效果。本文就CAR-T细胞应用于肿瘤免疫治疗的前生、今世和将来作一综述。  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