Anti-tumor immunity

抗肿瘤免疫
  • 文章类型: Journal Article
    目的:使用PD-L1阻断的免疫治疗仅对一小部分癌症患者有效,抵抗是常见的。这强调了了解癌症免疫逃避和抵抗机制的重要性。
    方法:基因组规模的CRISPR-Cas9筛选将Bap1鉴定为PD-L1表达的调节因子。为了测量肿瘤大小和存活率,将肿瘤细胞皮下注射到同基因WT小鼠和免疫受损小鼠中。使用流式细胞术检查Bap1缺失肿瘤的表型和转录特征,RNA-seq,以及CUT和Tag-seq分析。
    结果:我们发现癌细胞中组蛋白去泛素酶Bap1的丢失激活了cDC1-CD8+T细胞依赖性抗肿瘤免疫。Bap1的缺失导致与抗肿瘤免疫应答相关的基因增加和与免疫逃避相关的基因减少。因此,肿瘤微环境发炎,更多的cDC1细胞和效应CD8+T细胞,但中性粒细胞和调节性T细胞较少。我们还发现,Bap1缺失肿瘤的消除取决于肿瘤MHCI分子和Fas介导的CD8T细胞毒性。我们对TCGA数据的分析进一步支持了这些发现,显示在各种人类癌症中BAP1表达和活化DC的mRNA签名与T细胞的细胞毒性之间的反向相关性。
    结论:组蛋白去泛素酶Bap1可用作肿瘤分层的生物标志物,并作为癌症免疫治疗的潜在治疗靶点。
    OBJECTIVE: Immunotherapy using PD-L1 blockade is effective in only a small group of cancer patients, and resistance is common. This emphasizes the importance of understanding the mechanisms of cancer immune evasion and resistance.
    METHODS: A genome-scale CRISPR-Cas9 screen identified Bap1 as a regulator of PD-L1 expression. To measure tumor size and survival, tumor cells were subcutaneously injected into both syngeneic WT mice and immunocompromised mice. The phenotypic and transcriptional characteristics of Bap1-deleted tumors were examined using flow cytometry, RNA-seq, and CUT&Tag-seq analysis.
    RESULTS: We found that loss of histone deubiquitinase Bap1 in cancer cells activates a cDC1-CD8+ T cell-dependent anti-tumor immunity. The absence of Bap1 leads to an increase in genes associated with anti-tumor immune response and a decrease in genes related to immune evasion. As a result, the tumor microenvironment becomes inflamed, with more cDC1 cells and effector CD8+ T cells, but fewer neutrophils and regulatory T cells. We also found that the elimination of Bap1-deleted tumors depends on the tumor MHCI molecule and Fas-mediated CD8+ T cell cytotoxicity. Our analysis of TCGA data further supports these findings, showing a reverse correlation between BAP1 expression and mRNA signatures of activated DCs and T-cell cytotoxicity in various human cancers.
    CONCLUSIONS: The histone deubiquitinase Bap1 could be used as a biomarker for tumor stratification and as a potential therapeutic target for cancer immunotherapies.
    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
    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
    肿瘤浸润淋巴细胞(TIL)功能减退有助于晚期癌症的进展,并且是免疫疗法的常见靶标。新的证据表明,在强直刺激期间,代谢不足会导致T细胞功能减退,但是在这种情况下启动代谢重编程的信号在很大程度上是未知的。这里,我们发现类似Meteorin(METRNL),肿瘤微环境(TME)中免疫细胞分泌的代谢活性细胞因子,诱导CD8+T细胞的生物能量衰竭。METRNL在重复刺激期间由CD8+T细胞分泌,并且经由自分泌和旁分泌信号传导两者起作用。机械上,METRNL增加E2F-过氧化物酶体增殖物激活受体δ(PPARδ)活性,导致线粒体去极化和减少的氧化磷酸化,这引发了补偿性生物能量向糖酵解的转变。Metrnl消融或下调改善了CD8+T细胞的代谢适应性,并增强了几种肿瘤模型中的肿瘤控制,证明了靶向METRNL-E2F-PPARδ途径以支持CD8TIL的生物能量适应性的翻译潜力。
    Tumor-infiltrating lymphocyte (TIL) hypofunction contributes to the progression of advanced cancers and is a frequent target of immunotherapy. Emerging evidence indicates that metabolic insufficiency drives T cell hypofunction during tonic stimulation, but the signals that initiate metabolic reprogramming in this context are largely unknown. Here, we found that Meteorin-like (METRNL), a metabolically active cytokine secreted by immune cells in the tumor microenvironment (TME), induced bioenergetic failure of CD8+ T cells. METRNL was secreted by CD8+ T cells during repeated stimulation and acted via both autocrine and paracrine signaling. Mechanistically, METRNL increased E2F-peroxisome proliferator-activated receptor delta (PPARδ) activity, causing mitochondrial depolarization and decreased oxidative phosphorylation, which triggered a compensatory bioenergetic shift to glycolysis. Metrnl ablation or downregulation improved the metabolic fitness of CD8+ T cells and enhanced tumor control in several tumor models, demonstrating the translational potential of targeting the METRNL-E2F-PPARδ pathway to support bioenergetic fitness of CD8+ TILs.
    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
    肥胖与13种不同癌症的风险增加有关。肥胖中患癌症的风险增加是由肥胖相关的免疫系统变化介导的。肥胖对与肿瘤发生有关的不同类型的炎症有不同的影响。例如,肥胖会促进脂肪组织中的慢性炎症,从而促进外周组织中的肿瘤。相反,肥胖抑制排斥肿瘤的急性炎症。因此,肥胖通过差异调节慢性与急性炎症而促进癌症。鉴于肥胖是慢性的,脂肪组织的初始炎症会导致全身性炎症,可在外周组织中诱导代偿性抗炎反应以抑制慢性炎症。因此,肥胖在外周组织中的总体作用取决于肥胖的持续时间和严重程度。脂肪组织是由除脂肪细胞之外的许多细胞类型组成的复杂组织。Further,在整个身体的不同解剖部位,脂肪组织的细胞组成是不同的。因此,脂肪组织对肥胖的敏感性取决于脂肪库的解剖位置。例如,肥胖引起的内脏炎症比皮下脂肪组织多。基于这些研究,肥胖促进肿瘤发生的机制是多因素和免疫细胞类型特异性的.本文的目的是讨论肥胖通过调节脂肪组织和肿瘤微环境中不同类型的炎症来促进肿瘤发生的细胞机制。
    Obesity is associated with an increased risk for 13 different cancers. The increased risk for cancer in obesity is mediated by obesity-associated changes in the immune system. Obesity has distinct effects on different types of inflammation that are tied to tumorigenesis. For example, obesity promotes chronic inflammation in adipose tissue that is tumor-promoting in peripheral tissues. Conversely, obesity inhibits acute inflammation that rejects tumors. Obesity therefore promotes cancer by differentially regulating chronic versus acute inflammation. Given that obesity is chronic, the initial inflammation in adipose tissue will lead to systemic inflammation that could induce compensatory anti-inflammatory reactions in peripheral tissues to suppress chronic inflammation. The overall effect of obesity in peripheral tissues is therefore dependent on the duration and severity of obesity. Adipose tissue is a complex tissue that is composed of many cell types in addition to adipocytes. Further, adipose tissue cellularity is different at different anatomical sites throughout the body. Consequently, the sensitivity of adipose tissue to obesity is dependent on the anatomical location of the adipose depot. For example, obesity induces more inflammation in visceral than subcutaneous adipose tissue. Based on these studies, the mechanisms by which obesity promotes tumorigenesis are multifactorial and immune cell type-specific. The objective of our paper is to discuss the cellular mechanisms by which obesity promotes tumorigenesis by regulating distinct types of inflammation in adipose tissue and the tumor microenvironment.
    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
    凭借其独特的性能和潜在的应用,近年来,用于信使RNA(mRNA)疫苗的基于纳米颗粒的递送平台获得了显著的关注。纳米颗粒具有增强免疫原性的优点,靶向递送,提高稳定性,为药物和疫苗提供新的解决方案。在一些临床研究中,各种纳米颗粒递送平台已逐步应用于广泛的疫苗应用。当前的研究重点是探索各种类型的纳米颗粒作为疫苗递送系统以增强疫苗稳定性和免疫原性。脂质纳米颗粒(LNP)在将抗原有效递送至免疫细胞的临床前和临床研究中显示出有希望的潜力。此外,脂质纳米颗粒和其他用于核酸的纳米颗粒,特别是mRNA递送系统,已经显示出疫苗开发的巨大潜力。在这次审查中,我们提出了各种疫苗平台,重点是纳米颗粒作为mRNA疫苗递送载体。我们描述了几种用于mRNA疫苗的新型纳米颗粒递送平台,例如脂质-,聚合物-,和基于蛋白质的纳米颗粒。此外,我们概述了纳米疫苗在癌症免疫治疗中对不同肿瘤的抗肿瘤免疫。最后,我们概述了这种有前途的基于纳米颗粒的疫苗递送平台技术的未来前景和仍然存在的挑战.
    With its unique properties and potential applications, nanoparticle-based delivery platforms for messenger RNA (mRNA) vaccines have gained significant attention in recent years. Nanoparticles have the advantages of enhancing immunogenicity, targeting delivery, and improving stability, providing a new solution for drug and vaccine delivery. In some clinical studies, a variety of nanoparticle delivery platforms have been gradually applied to a wide range of vaccine applications. Current research priorities are exploring various types of nanoparticles as vaccine delivery systems to enhance vaccine stability and immunogenicity. Lipid nanoparticles (LNPs) have shown promising potential in preclinical and clinical studies on the efficient delivery of antigens to immune cells. Moreover, lipid nanoparticles and other nanoparticles for nucleic acids, especially for mRNA delivery systems, have shown vast potential for vaccine development. In this review, we present various vaccine platforms with an emphasis on nanoparticles as mRNA vaccine delivery vehicles. We describe several novel nanoparticle delivery platforms for mRNA vaccines, such as lipid-, polymer-, and protein-based nanoparticles. In addition, we provide an overview of the anti-tumor immunity of nanovaccines against different tumors in cancer immunotherapy. Finally, we outline future perspectives and remaining challenges for this promising technology of nanoparticle-based delivery platforms for 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
    在最近的一篇文章中,麦克斯韦等人。报告肿瘤细胞特异性AT丰富的相互作用域1A(ARID1A)的丢失,染色质重塑SWI/SNF复合物的一个组成部分,通过R-loop介导的I型干扰素(IFN)途径的上调触发抗肿瘤免疫。这些最新发现揭示了在具有ARID1A功能丧失突变的患者中观察到的改善对免疫检查点治疗(ICT)反应的分子机制。
    In a recent article, Maxwell et al. report that loss of tumor cell-specific AT-rich interaction domain 1A (ARID1A), a component of the chromatin remodeling SWI/SNF complex, triggers antitumor immunity via R-loop-mediated upregulation of the type-I interferon (IFN) pathway. These recent findings uncover a molecular mechanism underlying improved responses to immune checkpoint therapy (ICT) seen in patients harboring an ARID1A loss-of-function mutation.
    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
    肠道微生物组显着影响免疫应答和免疫检查点抑制剂的功效。我们在13例抗PD-1难治性晚期实体癌患者中进行了一项临床试验(NCT04264975),将抗程序性死亡-1(PD-1)抑制剂与抗PD-1应答者的粪便微生物群移植(FMT)相结合。FMT在13例患者中有6例引起持续的微生物群变化和临床获益,有1个部分反应和5个稳定的疾病,达到7.7%的客观反应率和46.2%的疾病控制率。临床反应与血液和肿瘤中细胞毒性T细胞和免疫细胞因子的增加相关。我们从FMT的应答者中分离出免疫普雷氏菌,通过增强细胞毒性T细胞浸润刺激T细胞活性并抑制小鼠肿瘤生长。此外,我们发现唾液乳杆菌和拟杆菌属可能抑制抗肿瘤免疫。我们的研究结果表明,具有有益微生物群的FMT可以克服晚期实体癌对抗PD-1抑制剂的耐药性,尤其是胃肠道癌症。
    The gut microbiome significantly influences immune responses and the efficacy of immune checkpoint inhibitors. We conducted a clinical trial (NCT04264975) combining an anti-programmed death-1 (PD-1) inhibitor with fecal microbiota transplantation (FMT) from anti-PD-1 responder in 13 patients with anti-PD-1-refractory advanced solid cancers. FMT induced sustained microbiota changes and clinical benefits in 6 of 13 patients, with 1 partial response and 5 stable diseases, achieving an objective response rate of 7.7% and a disease control rate of 46.2%. The clinical response correlates with increased cytotoxic T cells and immune cytokines in blood and tumors. We isolated Prevotella merdae Immunoactis from a responder to FMT, which stimulates T cell activity and suppresses tumor growth in mice by enhancing cytotoxic T cell infiltration. Additionally, we found Lactobacillus salivarius and Bacteroides plebeius may inhibit anti-tumor immunity. Our findings suggest that FMT with beneficial microbiota can overcome resistance to anti-PD-1 inhibitors in advanced solid cancers, especially gastrointestinal cancers.
    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
    脑肿瘤如成胶质细胞瘤对免疫检查点阻断治疗有抗性,主要是由于肿瘤中有限的T细胞浸润。这里,我们显示,小鼠患有颅内肿瘤表现出系统性免疫抑制和T细胞在骨髓隔离,导致脑肿瘤中T细胞浸润减少。在荷瘤小鼠中,升高的血浆皮质酮通过糖皮质激素受体驱动T细胞隔离。由糖皮质激素诱导的T细胞动力学介导的免疫抑制和随后的肿瘤生长促进可以通过肾上腺切除术来消除。糖皮质激素激活抑制剂或糖皮质激素受体拮抗剂的给药,和T细胞特异性糖皮质激素受体缺失的小鼠。T细胞中的CCR8表达在荷瘤小鼠中以糖皮质激素受体依赖性方式增加。此外,趋化因子CCL1和CCL8是CCR8的配体,在荷瘤小鼠的骨髓免疫细胞中高度表达以募集T细胞。这些结果表明,脑肿瘤诱导的糖皮质激素激增和T细胞中的CCR8上调导致骨髓中的T细胞螯合,损害抗肿瘤免疫反应。靶向糖皮质激素受体-CCR8轴可能为颅内肿瘤的治疗提供有希望的免疫治疗方法。
    Brain tumors such as glioblastomas are resistant to immune checkpoint blockade therapy, largely due to limited T cell infiltration in the tumors. Here, we show that mice bearing intracranial tumors exhibit systemic immunosuppression and T cell sequestration in bone marrow, leading to reduced T cell infiltration in brain tumors. Elevated plasma corticosterone drives the T cell sequestration via glucocorticoid receptors in tumor-bearing mice. Immunosuppression mediated by glucocorticoid-induced T cell dynamics and the subsequent tumor growth promotion can be abrogated by adrenalectomy, the administration of glucocorticoid activation inhibitors or glucocorticoid receptor antagonists, and in mice with T cell-specific deletion of glucocorticoid receptor. CCR8 expression in T cells is increased in tumor-bearing mice in a glucocorticoid receptor-dependent manner. Additionally, chemokines CCL1 and CCL8, the ligands for CCR8, are highly expressed in bone marrow immune cells in tumor-bearing mice to recruit T cells. These findings suggested that brain tumor-induced glucocorticoid surge and CCR8 upregulation in T cells lead to T cell sequestration in bone marrow, impairing the anti-tumor immune response. Targeting the glucocorticoid receptor-CCR8 axis may offer a promising immunotherapeutic approach for the treatment of intracranial tumors.
    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
    背景:在三阴性乳腺癌(TNBC)治疗中,淋巴细胞浸润不足会显著阻碍免疫检查点抑制剂的疗效.我们以前已经证明海纳宁-1(HN-1),从海南青蛙皮肤中鉴定出的宿主防御肽(HDP),诱导乳腺癌细胞凋亡,并通过未知的机制引导抗肿瘤免疫。
    方法:我们使用体外实验观察HN-1处理的TNBC细胞系中的免疫原性细胞死亡(ICD)指标,小鼠肿瘤模型验证HN-1促进小鼠抗肿瘤免疫应答,并对患者来源的乳腺癌细胞进行体外药敏试验,以验证HN-1的抑制作用。
    结果:HN-1在TNBC中诱导ICD,在此期间释放了损伤相关分子模式(DAMPs),可以进一步增加抗肿瘤免疫反应。白细胞介素2(IL-2)的分泌水平,IL-12和干扰素γ在共培养上清液中增加,和树突细胞(DC)通过与HN-1预处理的TNBC细胞共培养而被激活。因此,HN-1增加了携带4T1和EMT6肿瘤的小鼠模型中抗肿瘤免疫细胞(DC和T淋巴细胞)的浸润。同时,调节性T细胞和骨髓来源的抑制细胞受到抑制。此外,HN-1诱导DNA损伤,胞质溶胶中的双链DNA释放显着增强,表明HN-1可能通过激活STING途径刺激ICD。STING的敲低抑制HN-1诱导的ICD。值得注意的是,在三维培养条件下,HN-1对患者来源的乳腺癌细胞表现出抑制作用。
    结论:总的来说,我们的研究表明,HN-1可作为一种潜在化合物,可增强TNBC患者的免疫治疗效果.
    BACKGROUND: In triple-negative breast cancer (TNBC) therapy, insufficient tumor infiltration by lymphocytes significantly hinders the efficacy of immune checkpoint inhibitors. We have previously demonstrated that Hainanenin-1 (HN-1), a host defense peptide (HDP) identified from Hainan frog skin, induces breast cancer apoptosis and boots anti-tumor immunity via unknown mechanism.
    METHODS: We used in vitro experiments to observe immunogenic cell death (ICD) indicators in HN-1-treated TNBC cell lines, a mouse tumor model to verify HN-1 promotion of mice anti-tumor immune response, and an in vitro drug sensitivity test of patient-derived breast cancer cells to verify the inhibitory effect of HN-1.
    RESULTS: HN-1 induced ICD in TNBC in a process during which damage-associated molecular patterns (DAMPs) were released that could further increase the anti-tumor immune response. The secretion level of interleukin 2 (IL-2), IL-12, and interferon γ in the co-culture supernatant was increased, and dendritic cells (DCs) were activated via a co-culture with HN-1-pretreated TNBC cells. As a result, HN-1 increased the infiltration of anti-tumor immune cells (DCs and T lymphocytes) in the mouse model bearing both 4T1 and EMT6 tumors. Meanwhile, regulatory T cells and myeloid-derived suppressor cells were suppressed. In addition, HN-1 induced DNA damage, and double-strand DNA release in the cytosol was significantly enhanced, indicating that HN-1 might stimulate ICD via activation of STING pathway. The knockdown of STING inhibited HN-1-induced ICD. Of note, HN-1 exhibited inhibitory effects on patient-derived breast cancer cells under three-dimensional culture conditions.
    CONCLUSIONS: Collectively, our study demonstrated that HN-1 could be utilized as a potential compound that might augment immunotherapy effects in patients with TNBC.
    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
    T细胞受体(TCR)发出的信号,共刺激受体,和细胞因子受体各自影响CD8T细胞命运。了解这些信号对稳态和微环境线索的反应可以揭示治疗性指导T细胞功能的新方法。通过小鼠的正向遗传筛查,我们发现LDL受体相关蛋白10(Lrp10)的功能缺失突变导致幼稚和中枢记忆性CD8T细胞在外周淋巴器官中积累.Lrp10编码免疫功能未知的保守细胞表面蛋白。T细胞活化诱导Lrp10表达,它在翻译后抑制IL7受体(IL7R)水平。因此,Lrp10缺失通过IL7R信号增强T细胞稳态扩增。Lrp10缺陷型小鼠也对同基因肿瘤具有内在抗性。这种表型取决于CD8T细胞的致密肿瘤浸润,显示出增强的存储单元特性,减少终端疲惫,和增强对免疫检查点抑制的反应。这里,我们提出Lrp10作为CD8T细胞稳态的一种新的负调节因子和一种控制肿瘤耐药的宿主因子,对免疫治疗有意义.
    Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号