Tumor antigen

肿瘤抗原
  • 文章类型: Journal Article
    免疫疗法通过利用免疫系统的先天能力来对抗恶性肿瘤,彻底改变了癌症治疗。尽管肿瘤抗原有望刺激抗肿瘤免疫反应,它们的临床应用受到引起强大而持久的免疫反应的限制的阻碍,肿瘤异质性和免疫逃避机制加剧。对宿主同源微生物抗原的免疫原性特性的最新见解引起了人们对它们增强抗肿瘤免疫力同时最小化脱靶效应的潜力的兴趣。本文综述了微生物抗原肽在肿瘤免疫治疗中的治疗潜力。首先概述了肿瘤抗原及其在临床翻译中的挑战。我们进一步探索微生物与肿瘤发展之间的复杂关系,阐明分子模拟的概念及其对肿瘤相关抗原免疫识别的意义。最后,我们讨论了鉴定和表征微生物抗原肽的方法,强调其免疫原性和治疗应用前景。
    Immunotherapy has revolutionized cancer treatment by leveraging the immune system\'s innate capabilities to combat malignancies. Despite the promise of tumor antigens in stimulating anti-tumor immune responses, their clinical utility is hampered by limitations in eliciting robust and durable immune reactions, exacerbated by tumor heterogeneity and immune evasion mechanisms. Recent insights into the immunogenic properties of host homologous microbial antigens have sparked interest in their potential for augmenting anti-tumor immunity while minimizing off-target effects. This review explores the therapeutic potential of microbial antigen peptides in tumor immunotherapy, beginning with an overview of tumor antigens and their challenges in clinical translation. We further explore the intricate relationship between microorganisms and tumor development, elucidating the concept of molecular mimicry and its implications for immune recognition of tumor-associated antigens. Finally, we discuss methodologies for identifying and characterizing microbial antigen peptides, highlighting their immunogenicity and prospects for therapeutic application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    白细胞介素-10(IL-10)是一种高度多效性的细胞因子,其通过抗炎和/或免疫刺激功能调节免疫稳态。此外,众所周知,IL-10在肿瘤免疫学和免疫疗法中发挥着不同的作用。本研究调查了头颈部鳞状细胞癌(HNSCC)患者中循环肿瘤抗原特异性IL-10产生T细胞的存在,以及确定可能影响产生IL-10的T细胞的免疫动力学的因素。体外,用肿瘤抗原p53和MAGE-A4刺激的外周血单个核细胞(PBMC)使用IFN-γ/IL-10双色酶联免疫吸附斑点试验评估干扰素(IFN)-γ/IL-10的产生.使用流式细胞术分析PBMC中表达免疫检查点分子的T细胞的比例。在18例HNSCC患者中,2(11.1%)和9(50.0%)表现出p53特异性IFN-γ和IL-10的产生,分别。同时,14例患者中有4例(28.6%)和7例(50.0%)检测到MAGE-A4特异性IFN-γ和IL-10的产生。在p53特异性反应中,在患者中观察到产生IL-10的T细胞明显多于产生IFN-γ的T细胞(P=0.0275)。在CD4+和CD8+T细胞中,有p53特异性IL-10产生的患者中表达淋巴细胞活化基因-3(Lag-3)的T细胞比例显著低于无p53特异性IL-10产生的患者.在某些患者中,Lag-3阻断增强了肿瘤抗原特异性IL-10。一起来看,本研究成功证明HNSCC患者外周血中存在产生肿瘤抗原特异性IL-10的T细胞,Lag-3+T细胞可能在调节产生IL-10的T细胞中起重要作用。这些发现为IL-10和Lag-3在介导抗肿瘤免疫反应中的作用提供了新的见解。
    Interleukin-10 (IL-10) is a highly pleiotropic cytokine that regulates immunological homeostasis through anti-inflammatory and/or immunostimulatory functions. Moreover, IL-10 is well known to exert diverse roles in tumor immunology and immunotherapy. The present study investigated the presence of circulating tumor antigen-specific IL-10-producing T cells in patients with head and neck squamous cell carcinoma (HNSCC), and determined factors that may influence the immunodynamics of IL-10-producing T cells. In vitro, peripheral blood mononuclear cells (PBMCs) stimulated with the tumor antigens p53 and MAGE-A4 were evaluated for interferon (IFN)-γ/IL-10 production using the IFN-γ/IL-10 double-color enzyme-linked immunosorbent spot assay. The proportion of T cells expressing immune checkpoint molecules in PBMCs was analyzed using flow cytometry. Of the 18 patients with HNSCC, 2 (11.1%) and 9 (50.0%) exhibited p53-specific IFN-γ and IL-10 production, respectively. Meanwhile, MAGE-A4-specific IFN-γ and IL-10 production was detected in 4 (28.6%) and 7 (50.0%) of 14 patients. In the p53-specific responses, IL-10-producing T cells were observed in significantly more patients than IFN-γ producing T cells (P=0.0275). In both CD4+ and CD8+ T cells, the proportion of T cells expressing lymphocyte activation gene-3 (Lag-3) was significantly lower in patients with p53-specific IL-10 production than in those without. In certain patients, Lag-3 blockade enhanced tumor antigen-specific IL-10. Taken together, the present study successfully demonstrated that tumor antigen-specific IL-10-producing T cells exist in the peripheral blood of patients with HNSCC and that Lag-3+ T cells may serve an important role in modulating IL-10-producing T cells. These findings provide novel insights into the roles of IL-10 and Lag-3 in mediating antitumor immune responses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胶质瘤,中枢神经系统(CNS)最常见的原发性恶性肿瘤,缺乏有效的治疗方法,>60%的病例是胶质母细胞瘤(GBM),最具侵略性的形式。尽管免疫疗法取得了进展,GBM仍然具有很高的抗性。靶向肿瘤抗原的方法加快了免疫疗法的发展,包括个性化的肿瘤特异性疫苗,患者特异性目标选择,树突状细胞(DC)疫苗,以及嵌合抗原受体(CAR)和T细胞受体(TCR)T细胞。最近的研究表明,在治疗GBM和低度胶质瘤(LGG)方面取得了有希望的结果,培养未来免疫疗法的希望。这篇综述讨论了针对神经胶质瘤的肿瘤疫苗,免疫学研究中的临床前模型,以及CD4+T细胞在疫苗诱导的抗肿瘤免疫中的作用。我们还总结了临床方法,挑战,以及未来创造更有效疫苗的研究。
    Glioma, the most common primary malignant tumor in the central nervous system (CNS), lacks effective treatments, and >60% of cases are glioblastoma (GBM), the most aggressive form. Despite advances in immunotherapy, GBM remains highly resistant. Approaches that target tumor antigens expedite the development of immunotherapies, including personalized tumor-specific vaccines, patient-specific target selection, dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) and T cell receptor (TCR) T cells. Recent studies show promising results in treating GBM and lower-grade glioma (LGG), fostering hope for future immunotherapy. This review discusses tumor vaccines against glioma, preclinical models in immunological research, and the role of CD4+ T cells in vaccine-induced antitumor immunity. We also summarize clinical approaches, challenges, and future research for creating more effective vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    传统疗法(TTS)的利用,比如化疗,基于活性氧的治疗,和热疗,在肿瘤细胞中诱导免疫原性细胞死亡(ICD)已成为激活抗肿瘤免疫反应的有希望的策略。然而,大多数TTS在诱导肿瘤ICD效应方面的功效有限,阻碍了它们与免疫治疗联合应用.为了应对这一挑战,已经提出了各种智能策略来增强这些TTS的免疫激活效果,然后与免疫治疗实现协同抗肿瘤功效。这些策略主要集中在增强肿瘤ICD效应或促进TTS期间的抗原(由ICD肿瘤细胞释放)呈递过程。在这篇综述中对它们进行了系统的总结。最后,并讨论了TTS在肿瘤免疫调节应用中存在的瓶颈和前景。
    The utilization of traditional therapies (TTS), such as chemotherapy, reactive oxygen species-based therapy, and thermotherapy, to induce immunogenic cell death (ICD) in tumor cells has emerged as a promising strategy for the activation of the antitumor immune response. However, the limited effectiveness of most TTS in inducing the ICD effect of tumors hinders their applications in combination with immunotherapy. To address this challenge, various intelligent strategies have been proposed to strengthen the immune activation effect of these TTS, and then achieve synergistic antitumor efficacy with immunotherapy. These strategies primarily focus on augmenting the tumor ICD effect or facilitating the antigen (released by the ICD tumor cells) presentation process during TTS, and they are systematically summarized in this review. Finally, the existing bottlenecks and prospects of TTS in the application of tumor immune regulation are also discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    由于高发病率和死亡率,结肠腺癌(COAD)是一个严重的公共卫生问题。本研究旨在确定可能的肿瘤抗原和COAD的坏死亚型,以开发mRNA疫苗并选择合适的患者进行精确治疗。
    COAD的基因表达谱和临床信息来自癌症基因组图谱和基因表达综合,分别。我们使用cBioPortal全面研究了坏死相关基因(NRGs)的变化,并使用基因表达谱交互分析2筛选与COAD患者预后相关的中心NRGs。进行共识聚类分析以鉴定坏死亚型。使用加权基因共表达网络分析(WGCNA)来鉴定NRG的共表达模块。使用基于图形学习的降维方法评估了COAD的坏死景观。最后,对两种坏死亚型进行了药物敏感性分析.
    两种肿瘤抗原,根据BLC-2相关X蛋白(BAX)和白介素1β(IL1B)与患者预后和抗原呈递细胞浸润的关系,对其进行鉴定。在COAD患者中区分了两种坏死亚型(N1和N2),它们的特征在于它们的差异生存状态和免疫检查点蛋白和免疫遗传学细胞死亡调节剂的分子表达水平。此外,COAD的坏死景观表明,个体患者具有明显的异质性。使用WGCNA鉴定共表达模块,发现中心NRG参与各种免疫过程。药物敏感性分析表明,N1和N2亚型之间的药物敏感性存在显着差异。细胞实验表明,BAX和IL1B的过表达均促进COAD细胞的坏死,并增强CD8T细胞的细胞毒性。
    BAX和IL1B是开发抗COADmRNA疫苗的潜在抗原,特别适用于N2亚型患者。因此,这项研究将指导开发更有效的免疫治疗方法和确定合适的患者。
    UNASSIGNED: Colon adenocarcinoma (COAD) is a serious public health issue due to high incidence and mortality rate. This study aimed to identify possible tumor antigens and necroptosis subtypes of COAD for the development of mRNA vaccines and the selection of appropriate patients for precision therapy.
    UNASSIGNED: Gene expression profiles and clinical information for COAD were obtained from The Cancer Genome Atlas and Gene Expression Omnibus, respectively. We comprehensively studied the alterations in necroptosis-related genes (NRGs) using cBioPortal, and screened the hub NRGs associated with the prognosis of patients with COAD using Gene Expression Profiling Interactive Analysis 2. Consensuses clustering analysis was performed to identify necroptosis subtypes. Weighted gene co-expression network analysis (WGCNA) was used to identify the co-expression modules of the NRGs. The necroptosis landscape of COAD was assessed using graph learning-based dimensionality reduction. Finally, a drug sensitivity analysis of the two necroptosis subtypes was performed.
    UNASSIGNED: Two tumor antigens, BLC-2-associated X protein (BAX) and interleukin 1 beta (IL1B) were identified based on their associations with prognosis of patients and antigen presenting cell infiltration. Two necroptosis subtypes (N1 and N2) were distinguished in patients with COAD, and they were characterized by their differential survival status and molecular expression levels of immune checkpoint proteins and immunogenetic cell death modulators. Furthermore, the necroptosis landscape of COAD indicated that individual patients had obvious heterogeneity. Co-expression modules were identified using WGCNA, and the hub NRGs were found to be involved in various immune processes. Drug sensitivity analysis indicated that there were significant differences in drug sensitivity between the N1 and N2 subtypes. Cell experiments suggested that both overexpression of BAX and IL1B promoted necroptosis of COAD cells and enhanced the cytotoxicity of CD8+ T cells.
    UNASSIGNED: BAX and IL1B are potential antigens for the development of anti-COAD mRNA vaccines, specifically for patients with the N2 subtype. Consequently, this study will guide the development of more effective immunotherapeutic approaches and the identification of appropriate patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肿瘤疫苗的开发已成为骨肉瘤(OS)免疫治疗的热门话题;然而,需要鉴定更多具有更强免疫原性的肿瘤抗原。
    我们从在线数据库下载了六组基因表达谱数据。分析了过表达的基因,相交,并用于根据其表达矩阵计算TARGETOS数据集中的免疫浸润丰度。根据潜在的肿瘤抗原基因是否与抗原呈递细胞(APC)表现出高度相关性来鉴定它们。根据ImmPort网站的表达情况,共检索到1330个免疫相关基因(IRGs),并使用共识聚类方法获得OS样本的免疫亚型。预后,免疫微环境,比较了免疫亚型对药物的敏感性。
    总共,680个基因在至少两个数据集中过表达,其中TREM2、TNFRSF12A、和THY1与不同APC呈正相关。基于TARGET-OS中1330个IRG的表达式矩阵,两种免疫亚型,确定了IS1和IS2。IS1亚型的预后优于IS2,IS1亚型患者的免疫检查点(ICP)相关基因表达较高,IS1亚型患者的免疫细胞浸润和对16种药物的敏感性普遍较高。
    我们鉴定了三个APC相关基因,这些基因可以被认为是OS疫苗的潜在新型肿瘤抗原的编码。确定了OS患者中的两种免疫亚型以使用mRNA疫苗实施个性化治疗。
    UNASSIGNED: The development of tumor vaccines has become a hot topic in immunotherapy for osteosarcoma (OS); however, more tumor antigens with stronger immunogenicity need to be identified.
    UNASSIGNED: We downloaded six sets of gene expression profile data from online databases. The overexpressed genes were analyzed, intersected, and used to calculate the immune infiltration abundance in the TARGET OS dataset based on their expression matrix. Potential tumor antigen genes were identified based on whether they exhibited a high correlation with the antigen-presenting cells (APCs). A total of 1330 immune-related genes (IRGs) from the ImmPort website were retrieved based on their expression, and the Consensus Cluster method was used to obtain immune subtypes of the OS samples. Prognosis, immune microenvironment, and sensitivity to drugs were compared among the immune subtypes.
    UNASSIGNED: In total, 680 genes were overexpressed in at least two datasets, of which TREM2, TNFRSF12A, and THY1 were positively correlated with different APCs. Based on the expression matrix of 1330 IRGs in TARGET-OS, two immune subtypes, IS1 and IS2, were identified. The prognosis of the IS1 subtype was better than that of IS2, the expression of immune checkpoint (ICP)-related genes was higher in patients with the IS1 subtype, and immune cell infiltration and sensitivity to 16 drugs were generally higher in IS1 subtype patients.
    UNASSIGNED: We identified three APC-correlated genes that can be considered to code for potential novel tumor antigens for OS vaccines. Two immune subtypes in patients with OS were identified to implement personalized treatments using mRNA vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肿瘤特异性CD8+T细胞经常是功能失调的并且不能阻止肿瘤生长。我们研究了是否可以招募肿瘤特异性CD4+T细胞来克服肿瘤内的CD8+T细胞功能障碍。我们发现CD8+和CD4+T细胞的空间定位和相互作用,但不是他们的数字,在过继性T细胞治疗以及免疫检查点阻断(ICB)的背景下,决定抗肿瘤反应:在效应期,CD4+T细胞必须与CD8+T细胞在相同的树突状细胞上参与,形成三细胞型簇(三联组)以许可CD8T细胞的细胞毒性和癌细胞消除。当瘤内三联体形成被破坏时,尽管肿瘤特异性CD8+和CD4+T细胞数量相等,肿瘤仍在进展。在ICB治疗的胸膜间皮瘤患者中,三合会与临床反应有关。因此,在效应物阶段和肿瘤消除期间,CD4+T细胞和三联体是CD8+T细胞毒性所必需的。
    Tumor-specific CD8+ T cells are frequently dysfunctional and unable to halt tumor growth. We investigated whether tumor-specific CD4+ T cells can be enlisted to overcome CD8+ T cell dysfunction within tumors. We find that the spatial positioning and interactions of CD8+ and CD4+ T cells, but not their numbers, dictate anti-tumor responses in the context of adoptive T cell therapy as well as immune checkpoint blockade (ICB): CD4+ T cells must engage with CD8+ T cells on the same dendritic cell during the effector phase, forming a three-cell-type cluster (triad) to license CD8+ T cell cytotoxicity and cancer cell elimination. When intratumoral triad formation is disrupted, tumors progress despite equal numbers of tumor-specific CD8+ and CD4+ T cells. In patients with pleural mesothelioma treated with ICB, triads are associated with clinical responses. Thus, CD4+ T cells and triads are required for CD8+ T cell cytotoxicity during the effector phase and tumor elimination.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    嵌合抗原受体(CAR)T细胞疗法是一种过继性免疫疗法,自传统癌症治疗如化疗以来,为增强癌症治疗提供了有希望的途径。手术,事实证明,放射治疗不足以完全根除肿瘤,尽管取得了相对积极的成果。已经观察到CAR-T细胞疗法在治疗大多数血液恶性肿瘤方面显示出有希望的结果,但对于其他癌症类型也具有广泛的范围。CAR是T细胞上的额外受体,通过有效激活免疫系统来帮助增加和加速肿瘤破坏。它由三个领域组成,胞外域,跨膜,和内域。胞外域是抗原识别和结合所必需的,而共刺激信号由内结构域转导。迄今为止,美国食品和药物管理局(FDA)已经批准了6种CAR-T细胞疗法。然而,尽管取得了显著的成功,CAR-T治疗与许多不良事件相关,并具有一定的局限性。本章主要介绍CAR域的结构和功能,各代的汽车,以及CAR-T细胞发育的过程,不利影响,以及CAR-T疗法的挑战。CAR-T细胞疗法也适用于其他疾病,包括系统性红斑狼疮,多发性硬化症,和心肌纤维化,等。
    Chimeric Antigen Receptor (CAR) T cell therapy is a type of adoptive immunotherapy that offers a promising avenue for enhancing cancer treatment since traditional cancer treatments like chemotherapy, surgery, and radiation therapy have proven insufficient in completely eradicating tumors, despite the relatively positive outcomes. It has been observed that CAR-T cell therapy has shown promising results in treating the majority of hematological malignancies but also have a wide scope for other cancer types. CAR is an extra receptor on the T-cell that helps to increase and accelerate tumor destruction by efficiently activating the immune system. It is made up of three domains, the ectodomain, transmembrane, and the endodomain. The ectodomain is essential for antigen recognition and binding, whereas the co-stimulatory signal is transduced by the endodomain. To date, the Food and Drug Administration (FDA) has granted approval for six CAR-T cell therapies. However, despite its remarkable success, CAR-T therapy is associated with numerous adverse events and has certain limitations. This chapter focuses on the structure and function of the CAR domain, various generations of CAR, and the process of CAR-T cell development, adverse effects, and challenges in CAR-T therapy. CAR-T cell therapy also has scopes in other disease conditions which include systemic lupus erythematosus, multiple sclerosis, and myocardial fibrosis, etc.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    免疫检查点抑制剂(ICIs)治疗在一些Lynch综合征相关结肠癌患者中显示出疗效。但是有些病人仍然没有从中受益。在这项研究中,我们采用了肿瘤疫苗和ICIs的组合策略,以最大限度地发挥免疫治疗的益处.这里,我们通过裂解MC38Mlh1KD细胞获得了含肿瘤抗原的细胞裂解物(TCL),并通过薄膜水化制备了具有典型球形形态的脂质体纳米颗粒(Lipo-PEG).通过酰胺化反应将抗PD-L1偶联至脂质体表面。正如观察到的,在安全浓度范围内,抗PD-L1/TCL@Lipo-PEG对小鼠肠上皮细胞(MODE-K)无明显毒性,也不引起小鼠红细胞溶血.此外,抗PD-L1/TCL@Lipo-PEG通过抗PD-L1抗体降低了结肠癌细胞(MC38Mlh1KD)的免疫逃逸,恢复了CD8+T细胞的杀伤功能,并将更多的肿瘤抗原靶向骨髓来源的树突状细胞(BMDCs),它也表达PD-L1,以刺激BMDC抗原呈递。在同基因移植的Lynch综合征相关结肠癌小鼠中,抗PD-L1和TCL的组合比单一免疫疗法提供了更好的癌症抑制,抗PD-L1/TCL@Lipo-PEG治疗的抑癌效果甚至优于游离药物。同时抗PD-L1/TCL@Lipo-PEG增强了免疫抑制肿瘤微环境。活体荧光成像和H&E染色显示纳米药物主要保留在肿瘤部位,对其他主要器官无明显毒副作用。本研究制备的抗PD-L1/TCL@Lipo-PEG对缓解Lynch综合征相关性结肠癌的进展具有较高的疗效和良好的生物安全性。它有望成为Lynch综合征相关结肠癌的治疗候选药物。
    Treatment with immune checkpoint inhibitors (ICIs) has shown efficacy in some patients with Lynch syndrome-associated colon cancer, but some patients still do not benefit from it. In this study, we adopted a combination strategy of tumor vaccines and ICIs to maximize the benefits of immunotherapy. Here, we obtained tumor-antigen-containing cell lysate (TCL) by lysing MC38Mlh1 KD cells and prepared liposome nanoparticles (Lipo-PEG) with a typical spherical morphology by thin-film hydration. Anti-PD-L1 was coupled to the liposome surface by the amidation reaction. As observed, anti-PD-L1/TCL@Lipo-PEG was not significantly toxic to mouse intestinal epithelial cells (MODE-K) in the safe concentration range and did not cause hemolysis of mouse red blood cells. In addition, anti-PD-L1/TCL@Lipo-PEG reduced immune escape from colon cancer cells (MC38Mlh1 KD) by the anti-PD-L1 antibody, restored the killing function of CD8+ T cells, and targeted more tumor antigens to bone marrow-derived dendritic cells (BMDCs), which also expressed PD-L1, to stimulate BMDC antigen presentation. In syngeneic transplanted Lynch syndrome-associated colon cancer mice, the combination of anti-PD-L1 and TCL provided better cancer suppression than monoimmunotherapy, and the cancer suppression effect of anti-PD-L1/TCL@Lipo-PEG treatment was even better than that of the free drug. Meanwhile anti-PD-L1/TCL@Lipo-PEG enhanced the immunosuppressive tumor microenvironment. In vivo fluorescence imaging and H&E staining showed that the nanomedicine was mainly retained in the tumor site and had no significant toxic side effects on other major organs. The anti-PD-L1/TCL@Lipo-PEG prepared in this study has high efficacy and good biosafety in alleviating the progression of Lynch syndrome-associated colon cancer, and it is expected to be a therapeutic candidate for Lynch syndrome-associated colon cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    T细胞景观中的过继性免疫疗法在癌症治疗中表现出功效。在过去的几十年里,转基因T细胞,特别是嵌合抗原受体T细胞,在血液恶性肿瘤的治疗方面取得了显著进展。此外,用于治疗实体瘤的多种抗原的广泛探索已经引起了对表达工程化T细胞受体(TCR)的T细胞的潜力的临床兴趣。TCR-T细胞具有识别细胞内抗原家族的能力,并在对靶表位的亲和力和信号转导方面保持TCR的固有特性。最近的研究提供了他们的能力和治疗多个难治性实体瘤的目标的关键见解,但也暴露了一些持久疗效的挑战。在这次审查中,我们描述了可用的肿瘤抗原的筛选和鉴定,以及TCR-T细胞治疗TCR的获取和优化。此外,我们总结了TCR-T细胞从实验室到临床应用的完整流程。最后,我们提出了通过联合疗法或TCR-T衍生产品提高癌症治疗效果的未来前景。总之,这篇综述描述了我们目前对TCR-T细胞治疗实体瘤的理解,并为扩大其临床应用和提高治疗效果提供了新的视角。
    Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from  laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号