T-Cell Exhaustion

T 细胞耗尽
  • 文章类型: Journal Article
    克服免疫介导的对PD-1阻断的抗性仍然是主要的临床挑战。在联合使用nivolumab(抗PD-1)和relatlimab(抗LAG-3)治疗的黑色素瘤患者中,已证明疗效增强。这是同类产品中第一个获得FDA批准的。然而,这两种抑制性受体如何协同作用以阻碍抗肿瘤免疫仍然未知。这里,我们显示,CD8+T细胞缺乏PD-1和LAG-3,与缺乏任一受体的CD8+T细胞相反,在黑色素瘤小鼠模型中介导增强的肿瘤清除和长期存活。PD-1-和LAG-3缺陷型CD8+T细胞在转录上不同,具有广泛的TCR克隆性和效应物样和干扰素反应基因的富集,导致增强的IFN-γ释放指示功能性。LAG-3和PD-1联合驱动T细胞耗尽,在调节TOX表达中起主导作用。机械上,自分泌,PD-1-和LAG-3-缺陷的CD8+T细胞需要细胞固有的IFN-γ信号传导来增强抗肿瘤免疫力,深入了解LAG-3和PD-1的组合靶向如何增强疗效。
    Overcoming immune-mediated resistance to PD-1 blockade remains a major clinical challenge. Enhanced efficacy has been demonstrated in melanoma patients with combined nivolumab (anti-PD-1) and relatlimab (anti-LAG-3) treatment, the first in its class to be FDA approved. However, how these two inhibitory receptors synergize to hinder anti-tumor immunity remains unknown. Here, we show that CD8+ T cells deficient in both PD-1 and LAG-3, in contrast to CD8+ T cells lacking either receptor, mediate enhanced tumor clearance and long-term survival in mouse models of melanoma. PD-1- and LAG-3-deficient CD8+ T cells were transcriptionally distinct, with broad TCR clonality and enrichment of effector-like and interferon-responsive genes, resulting in enhanced IFN-γ release indicative of functionality. LAG-3 and PD-1 combined to drive T cell exhaustion, playing a dominant role in modulating TOX expression. Mechanistically, autocrine, cell-intrinsic IFN-γ signaling was required for PD-1- and LAG-3-deficient CD8+ T cells to enhance anti-tumor immunity, providing insight into how combinatorial targeting of LAG-3 and PD-1 enhances efficacy.
    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
    肝细胞癌(HCC)是一种常见的恶性肿瘤,其复杂的免疫逃避机制给治疗带来了挑战。S100A10基因在各种癌症中的作用引起了极大的关注。本研究旨在阐明S100A10通过cPLA2和5-LOX轴对CD8+T细胞衰竭的影响,从而阐明其在HCC免疫逃避中的作用。通过分析GEO和TCGA数据库中与HCC相关的数据,我们鉴定了与脂质代谢相关的差异表达基因,并建立了预后风险模型.随后,通过RNA-seq和PPI分析,我们确定了重要的脂质代谢基因和下游因子S100A10,ACOT7和SMS,与CD8+T细胞浸润显著相关。鉴于最显著的表达差异,我们选择S100A10进行进一步研究.进行了体外和体内实验,包括CD8+T细胞与MHCC97-L细胞的共培养实验,Co-IP实验,并在HCC小鼠模型中进行验证。S100A10在HCC组织中显着过表达,并可能通过cPLA2和5-LOX轴调节CD8T细胞耗竭和脂质代谢重编程。沉默S100A10可以抑制CD8+T细胞衰竭,进一步抑制HCC的免疫逃避。S100A10可以激活cPLA2和5-LOX轴,启动脂质代谢重编程和上调LTB4水平,从而促进HCC组织中CD8+T细胞耗尽,促进HCC细胞的免疫逃避,最终影响肝癌细胞的生长和迁移。这项研究强调了S100A10通过cPLA2和5-LOX轴在HCC免疫逃避中的关键作用,为HCC的诊断和治疗提供了新的理论基础和潜在的靶点。
    Hepatocellular carcinoma (HCC) is a common malignant tumor with a complex immune evasion mechanism posing a challenge to treatment. The role of the S100A10 gene in various cancers has garnered significant attention. This study aims to elucidate the impact of S100A10 on CD8+ T cell exhaustion via the cPLA2 and 5-LOX axis, thereby elucidating its role in immune evasion in HCC. By analyzing the HCC-related data from the GEO and TCGA databases, we identified differentially expressed genes associated with lipid metabolism and developed a prognostic risk model. Subsequently, through RNA-seq and PPI analyses, we determined vital lipid metabolism genes and downstream factors S100A10, ACOT7, and SMS, which were significantly correlated with CD8+ T cell infiltration. Given the most significant expression differences, we selected S100A10 for further investigation. Both in vitro and in vivo experiments were conducted, including co-culture experiments of CD8+ T cells with MHCC97-L cells, Co-IP experiments, and validation in an HCC mouse model. S100A10 was significantly overexpressed in HCC tissues and potentially regulates CD8+ T cell exhaustion and lipid metabolism reprogramming through the cPLA2 and 5-LOX axis. Silencing S100A10 could inhibit CD8+ T cell exhaustion, further suppressing immune evasion in HCC. S100A10 may activate the cPLA2 and 5-LOX axis, initiating lipid metabolism reprogramming and upregulating LTB4 levels, thus promoting CD8+ T cell exhaustion in HCC tissues, facilitating immune evasion by HCC cells, ultimately impacting the growth and migration of HCC cells. This research highlights the critical role of S100A10 via the cPLA2 and 5-LOX axis in immune evasion in HCC, providing new theoretical foundations and potential targets for diagnosing and treating HCC.
    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
    由于抑制性肿瘤微环境和T细胞耗竭,胰腺导管癌(PDAC)的免疫治疗仍然令人失望。其中干扰素刺激基因的作用在很大程度上是未知的。这里,我们专注于一个典型的干扰素刺激基因,GBP4,并研究其在胰腺癌中的潜在诊断和治疗价值。对本地样本和公共数据库的表达分析表明,GBP4是PDAC微环境中最主要的GBP家族成员之一。GBP4的表达水平与患者生存率呈负相关。然后我们鉴定了PDAC中GBP4调控区的DNA低甲基化,并通过在选定的DNA基因座上使用dCas9-SunTag-DNMAT3A-sgRNA靶向甲基化系统进行靶向甲基化来验证其对GBP4表达的调节作用。之后,我们研究了GBP4的下游功能,趋化试验表明GBP4过表达显著改善了CD8+T细胞的浸润,而且还诱导了免疫检查点基因的上调和T细胞耗竭。最后,使用初级类器官的体外T细胞杀伤试验表明,具有高水平GBP4表达的PDAC样品显示出抗PD-1治疗的显著更高的敏感性。一起来看,我们的研究揭示了GBP4在胰腺癌中的表达模式和表观遗传调控机制,阐明了GBP4对T细胞耗竭和抗肿瘤免疫学的影响.
    Immunotherapy for pancreatic ductal carcinoma (PDAC) remains disappointing due to the repressive tumor microenvironment and T cell exhaustion, in which the roles of interferon-stimulated genes were largely unknown. Here, we focused on a typical interferon-stimulated gene, GBP4, and investigated its potential diagnostic and therapeutic value in pancreatic cancer. Expression analysis on both local samples and public databases indicated that GBP4 was one of the most dominant GBP family members present in the PDAC microenvironment, and the expression level of GBP4 was negatively associated with patient survival. We then identified DNA hypo-methylation in regulatory regions of GBP4 in PDAC, and validated its regulatory role on GBP4 expression via performing targeted methylation using dCas9-SunTag-DNMAT3A-sgRNA-targeted methylation system on selected DNA locus. After that, we investigated the downstream functions of GBP4, and chemotaxis assays indicated that GBP4 overexpression significantly improved the infiltration of CD8+T cells, but also induced upregulation of immune checkpoint genes and T cell exhaustion. Lastly, in vitro T cell killing assays using primary organoids suggested that the PDAC samples with high level of GBP4 expression displayed significantly higher sensitivity to anti-PD-1 treatment. Taken together, our studies revealed the expression patterns and epigenetic regulatory mechanisms of GBP4 in pancreatic cancer and clarified the effects of GBP4 on T cell exhaustion and antitumor immunology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    作为抗肿瘤免疫的主要组成部分,T细胞在肿瘤微环境(TME)中易于耗尽和功能障碍。彻底了解TME中的T细胞耗竭(TEX)对于有效解决临床环境中的TEX和促进免疫检查点阻断疗法的功效至关重要。在真核生物中,许多细胞表面蛋白通过糖基化磷脂酰肌醇(GPI)锚与质膜相连,在促进膜蛋白的正确易位中起着至关重要的作用。然而,现有证据不足以支持GPI锚的任何额外功能参与.这里,我们调查了乳腺癌(BC)患者TME中GPI锚生物合成的特征,特别是它与TEX的相关性。GPI锚生物合成应被视为BC的预后危险因素。具有高GPI锚生物合成的患者显示更严重的TEX。耗竭的CD8T细胞中GPI锚生物合成水平高于正常的CD8T细胞,在恶性上皮细胞和正常乳腺上皮细胞之间未观察到。此外,我们还发现GPI-锚生物合成相关基因可用于诊断BC患者的TEX状态和预测预后。TEX诊断模型和预后模型均显示良好的AUC值.最后,我们在细胞和临床样本中证实了我们的发现.敲除PIGU基因表达显著降低MDA-MB-231和MCF-7细胞系的增殖率。来自临床样品的免疫荧光结果显示CD8T细胞在具有GPAA1和PIGU高表达的组织中的聚集减少。
    As the primary component of anti-tumor immunity, T cells are prone to exhaustion and dysfunction in the tumor microenvironment (TME). A thorough understanding of T cell exhaustion (TEX) in the TME is crucial for effectively addressing TEX in clinical settings and promoting the efficacy of immune checkpoint blockade therapies. In eukaryotes, numerous cell surface proteins are tethered to the plasma membrane via Glycosylphosphatidylinositol (GPI) anchors, which play a crucial role in facilitating the proper translocation of membrane proteins. However, the available evidence is insufficient to support any additional functional involvement of GPI anchors. Here, we investigate the signature of GPI-anchor biosynthesis in the TME of breast cancer (BC)patients, particularly its correlation with TEX. GPI-anchor biosynthesis should be considered as a prognostic risk factor for BC. Patients with high GPI-anchor biosynthesis showed more severe TEX. And the levels of GPI-anchor biosynthesis in exhausted CD8 T cells was higher than normal CD8 T cells, which was not observed between malignant epithelial cells and normal mammary epithelial cells. In addition, we also found that GPI -anchor biosynthesis related genes can be used to diagnose TEX status and predict prognosis in BC patients, both the TEX diagnostic model and the prognostic model showed good AUC values. Finally, we confirmed our findings in cells and clinical samples. Knockdown of PIGU gene expression significantly reduced the proliferation rate of MDA-MB-231 and MCF-7 cell lines. Immunofluorescence results from clinical samples showed reduced aggregation of CD8 T cells in tissues with high expression of GPAA1 and PIGU.
    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
    在肿瘤微环境(TME)中,由于肿瘤抗原的持续刺激,CD8+T细胞显示出阶段耗尽。评估CD8+T细胞状态并逆转耗竭是评估肿瘤患者预后和治疗效果的关键。这项研究的目的是建立一个预后标志,可以有效地预测肝细胞癌(HCC)患者的预后和免疫治疗反应。我们使用单变量Cox分析从癌症基因组图谱数据集获得与CD8+T细胞耗尽相关的转录因子。然后,转录因子碱性亮氨酸拉链ATF样转录因子的预后特征,Eomesodermin,利用LASSOCox回归构建了调节T细胞衰竭的T-box蛋白21。采用逆转录-定量聚合酶链反应法检测23对肝癌和癌旁组织中3种转录因子mRNA的相对表达水平,并在癌症基因组图谱数据集中进行内部验证,在国际癌症基因组联盟数据集中进行外部验证。Cox回归分析显示风险评分是独立的预后变量。高危组的总生存期明显低于低危组。低危组有较高的免疫评分,矩阵得分,和估计分数,低危组中大多数免疫检查点基因的表达水平显着增加。因此,风险评分较低的患者从免疫治疗中获益更多.3种转录因子的组合可以评价TME中CD8+T细胞的耗尽状态,为评估HCC患者的TME和免疫治疗疗效奠定了基础。
    In the tumor microenvironment (TME), CD8+ T cells showed stage exhaustion due to the continuous stimulation of tumor antigens. To evaluate the status of CD8+ T cells and reverse the exhaustion is the key to evaluate the prognosis and therapeutic effect of tumor patients. The aim of this study was to establish a prognostic signature that could effectively predict prognosis and response to immunotherapy in patients with hepatocellular carcinoma (HCC). We used univariate Cox analysis to obtain transcription factors associated with CD8+ T cell exhaustion from The Cancer Genome Atlas dataset. Then, the prognostic signature for transcription factors basic leucine zipper ATF-like transcription factor, Eomesodermin, and T-box protein 21 regulating T cell exhaustion was constructed using LASSO Cox regression. The relative expression levels of the mRNA of the 3 transcription factors were detected by reverse transcription-quantitative polymerase chain reaction in 23 pairs of HCC and paracancer tissues, and verified internally in The Cancer Genome Atlas dataset and externally in the International Cancer Genome Consortium dataset. Cox regression analysis showed that risk score was an independent prognostic variable. The overall survival of the high-risk group was significantly lower than that of the low-risk group. The low-risk group had higher immune scores, matrix scores, and ESTIMATE scores, and significantly increased expression levels of most immune checkpoint genes in the low-risk group. Therefore, patients with lower risk scores benefit more from immunotherapy. The combination of the 3 transcription factors can evaluate the exhaustion state of CD8+ T cells in the TME, laying a foundation for evaluating the TME and immunotherapy efficacy in patients with HCC.
    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
    化学免疫疗法通过激活细胞毒性淋巴细胞(CTL)免疫应答而具有特异性抗肿瘤作用的优势。然而,多重屏障限制了结局,部分原因是免疫抑制性肿瘤微环境(iTME)中肿瘤细胞介导的CTL耗竭.这里,我们合理地设计了一种简单而通用的Ca2+纳米发生器来调节iTME,以增强2-脱氧葡萄糖(2-DG)介导的化学免疫疗法.简而言之,2-DG化疗后,涂有EL4细胞膜的CaO2纳米颗粒(表示为CaNP@ECM)可以通过EL4细胞膜上的LFA-1与肿瘤组织中发炎的内皮细胞上的ICAM-1之间的粘附优先在肿瘤组织中积累,并显示出一系列对CTL的益处:i)增加CTL的葡萄糖利用率,同时通过Ca2过载介导的肿瘤细胞糖酵解抑制减少乳酸分泌,以及缓解缺氧;ii)通过EL4细胞膜上的PD-1和TGF-β1R通过TGF-β1清除和PD-L1阻断来逆转CTL耗竭;iii)通过Ca2过载诱导的肿瘤细胞的免疫死亡(ICD)来增强肿瘤免疫治疗。我们证明了多模态Ca2纳米发生器在体外和体内挽救了T细胞的耗竭并抑制了肿瘤的生长。更重要的是,该研究还通过Ca2+超负荷促进了基于葡萄糖代谢抑制的肿瘤免疫治疗的发展.
    Chemo-immunotherapy holds the advantage of specific antitumor effects by activating cytotoxic lymphocyte cells (CTLs) immune response. However, multiple barriers have limited the outcomes partly due to tumor-cell-mediated exhaustion of CTLs in the immunosuppressive tumor microenvironment (iTME). Here, we rationally designed a simple-yet-versatile Ca2+ nanogenerator to modulate iTME for enhancing 2-deoxyglucose (2-DG) mediated chemo-immunotherapy. Briefly, after 2-DG chemotherapy, CaO2 nanoparticles coated with EL4 cell membrane (denoted as CaNP@ECM) could preferentially accumulate in tumor tissue via adhesion between LFA-1 on EL4 cell membrane and ICAM-1 on inflamed endothelial cell in tumor tissues and display a series of benefits for CTLs: i) Increasing glucose availability of CTLs while reducing lactic acid secretion through Ca2+ overloading mediated inhibition of tumor cell glycolysis, as well as relieving hypoxia; ii) Reversing CTLs exhaustion via TGF-β1 scavenging and PD-L1 blockade through PD-1 and TGF-β1R on EL4 cell membrane; iii) Boosting tumor immunotherapy via immunologic death (ICD) of tumor cells induced by Ca2+ overloading. We demonstrate that the multi-modal Ca2+ nanogenerator rescues T cells from exhaustion and inhibits tumor growth both in vitro and in vivo. More importantly, the study also facilitate the development of glucose metabolism inhibition-based tumor immunotherapy via Ca2+ overloading.
    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细胞长期受到抗原刺激,能量供应不足,导致它们的效应器功能下降,记忆能力,和增殖能力,最终导致T细胞耗尽和无法在肿瘤微环境中执行正常的免疫功能。因此,探索如何将这些耗尽的T细胞恢复到具有效应子功能的状态具有重要意义。耗尽的T细胞表现出一系列分子改变,例如抑制受体的表达增强,转录因子谱的变化,以及表观遗传上的修饰,新陈代谢,和转录景观。这篇综述全面概述了逆转T细胞衰竭的各种策略,包括免疫检查点封锁,并探讨了联合多种方法逆转T细胞耗竭的潜在协同作用。它为实现更持久和有效的T细胞衰竭逆转提供了新的见解和方法。
    T cells suffer from long-term antigen stimulation and insufficient energy supply, leading to a decline in their effector functions, memory capabilities, and proliferative capacity, ultimately resulting in T cell exhaustion and an inability to perform normal immune functions in the tumor microenvironment. Therefore, exploring how to restore these exhausted T cells to a state with effector functions is of great significance. Exhausted T cells exhibit a spectrum of molecular alterations, such as heightened expression of inhibitory receptors, shifts in transcription factor profiles, and modifications across epigenetic, metabolic, and transcriptional landscapes. This review provides a comprehensive overview of various strategies to reverse T cell exhaustion, including immune checkpoint blockade, and explores the potential synergistic effects of combining multiple approaches to reverse T cell exhaustion. It offers new insights and methods for achieving more durable and effective reversal of T cell exhaustion.
    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细胞和双特异性抗体(BsAb)是针对表达特异性抗原的细胞重定向免疫系统的主要平台。彻底改变血液恶性肿瘤的治疗方法,包括多发性骨髓瘤(MM)。在MM中,耐药复发是主要的治疗限制因素,也是导致该疾病仍被认为无法治愈的主要原因。T细胞参与疗法有望改善MM的治疗。然而,T细胞适应性可能会阻碍这些治疗的有效性。T细胞衰竭是效应子功能逐渐下降的条件,细胞因子分泌减少,和慢性抗原刺激引起的抑制性受体表达增加。
    这篇综述探讨了在T细胞重定向BsAb和CAR-T治疗的背景下,MM中T细胞耗竭的发现。
    T细胞的适应性已成为T细胞重定向疗法发展的重要因素。T细胞耗竭与这些疗法相关的方式可能会影响CAR和BsAbs技术的进一步发展。以及临床使用的策略。因此,这篇综述旨在探讨目前对MM中T细胞耗竭的认识及其与这些治疗的关系。
    UNASSIGNED: Chimeric Antigen Receptor (CAR) T-cells and Bispecific Antibodies (BsAb) are the leading platforms for redirecting the immune system against cells expressing the specific antigen, revolutionizing the treatment of hematological malignancies, including multiple myeloma (MM). In MM, drug-resistant relapses are the main therapy-limiting factor and the leading cause of why the disease is still considered incurable. T-cell-engaging therapies hold promise in improving the treatment of MM. However, the effectiveness of these treatments may be hindered by T-cell fitness. T-cell exhaustion is a condition of a gradual decline in effector function, reduced cytokine secretion, and increased expression of inhibitory receptors due to chronic antigen stimulation.
    UNASSIGNED: This review examines findings about T-cell exhaustion in MM in the context of T-cell redirecting BsAbs and CAR-T treatment.
    UNASSIGNED: The fitness of T-cells has become an important factor in the development of T-cell redirecting therapies. The way T-cell exhaustion relates to these therapies could affect the further development of CAR and BsAbs technologies, as well as the strategies used for clinical use. Therefore, this review aims to explore the current understanding of T-cell exhaustion in MM and its relationship to these therapies.
    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
    肿瘤来源的细胞外囊泡(EV)是细胞间通讯和信号传导的最重要方式之一。癌症干细胞(CSC)分泌EV以调节免疫检查点分子并逃避免疫监视。被称为细胞毒性T淋巴细胞(CTL)的活化CD8+T细胞是最强大的抗癌适应性细胞。它们的活性在肿瘤微环境(TME)内遇到细胞和信号时受到损害,导致反应不足,称为精疲力竭。CSC衍生的外泌体在CD8+T细胞上表达程序性死亡配体-1(PD-L1)并上调程序性死亡-1(PD-1)以促进其耗尽。肿瘤衍生的外泌体上的PD-L1表达似乎由含有转化生长因子(TGF)-β的CSC衍生的外泌体诱导。生腱蛋白-C是CSC外泌体的另一种成分,其作用于T细胞中的哺乳动物雷帕霉素靶标(mTOR)信号传导。糖酵解是由CSC衍生的外泌体对缺氧诱导因子-1α(HIF-1α)的诱导作用促进的代谢事件。CSC与CD8+T细胞的相互作用更加复杂,因为CSC衍生的外泌体含有Notch1以刺激非肿瘤细胞的干性,Notch1对PD-1的诱导作用促进CD8+T细胞衰竭。CSC外泌体靶向尚未被广泛研究。该领域的进展将打开新的治疗窗口,并塑造癌症免疫治疗的未来。
    Tumor-derived extracellular vesicles (EVs) are one of the most important ways of intercellular communication and signaling. Cancer stem cells (CSCs) secrete EVs to modulate immune checkpoint molecules and evade immune surveillance. Activated CD8+ T cells known as cytotoxic T lymphocytes (CTLs) are the most powerful anti-cancer adaptive cells. Their activity is compromised upon encountering cells and signaling within the tumor microenvironment (TME), resulting in hyporesponsiveness called exhaustion. CSC-derived exosomes express programmed death ligand-1 (PD-L1) and upregulate programmed death-1 (PD-1) on CD8+ T cells to promote their exhaustion. PD-L1 expression on tumor-derived exosomes appears to be induced by CSC-derived exosomes containing transforming growth factor (TGF)-β. Tenascin-C is another constituent of CSC exosomes that acts on mammalian target of rapamycin (mTOR) signaling in T cells. Glycolysis is a metabolic event promoted by the inducing effect of CSC-derived exosomes on hypoxia-inducible factor-1α (HIF-1α). CSC interaction with CD8+ T cells is even more complex as the CSC-derived exosomes contain Notch1 to stimulate stemness in non-tumor cells, and the inducible effect of Notch1 on PD-1 promotes CD8+ T cell exhaustion. CSC exosome targeting has not been extensively studied yet. Advances in the field will open up new therapeutic windows and shape the future of cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号