Lag-3

LAG - 3
  • 文章类型: Journal Article
    淋巴细胞活化基因3(LAG-3),也称为CD223,是一种新兴的免疫检查点,紧随PD-1和CTLA-4。临床试验中的几种LAG-3靶向抑制剂以及relatlimab(抗LAG-3)和nivolumab(抗PD-1)的组合已被批准用于治疗不可切除或转移性黑色素瘤。尽管LAG-3具有令人鼓舞的临床潜力,但肿瘤中的生理功能和作用机制仍未得到很好的了解。在这次审查中,系统总结了LAG-3的结构、LAG-3的配体、LAG-3的细胞特异性功能和信号传导以及LAG-3抑制剂的研发现状。
    Lymphocyte activation gene 3 (LAG-3), also known as CD223, is an emerging immune checkpoint that follows PD-1 and CTLA-4. Several LAG-3 targeting inhibitors in clinical trials and the combination of relatlimab (anti-LAG-3) and nivolumab (anti-PD-1) have been approved for treating - unresectable or metastatic melanoma. Despite the encouraging clinical potential of LAG-3, the physiological function and mechanism of action in tumors are still not well understood. In this review, we systematically summarized the structure of LAG-3, ligands of LAG-3, cell-specific functions and signaling of LAG-3, and the current status of LAG-3 inhibitors under development.
    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
    目的:结直肠癌是全球最常见的恶性肿瘤。目前许多病理和分子遗传标准被用作该疾病的预测因子。它们包括MMR缺陷或MSI/MSS状态的评估,其中,确定肿瘤的免疫原性。在这方面,根据MMR状态评估不同肿瘤区室的PD-L1,CTLA-4和LAG-3免疫检查点分子值得特别关注.
    方法:使用多重免疫组织化学评估肿瘤核心和浸润边缘的免疫检查点分子的表达。
    结果:数据分析显示PD-L1占优势(p=0.011),CTLA-4(p=0.004),与pMMR样品相比,dMMR癌的浸润边缘和LAG-3(p=0.013)表达。对肿瘤核心和浸润性边缘的TIL群体进行定量分析,可以确定dMMR癌浸润性边缘的CD3和CD8淋巴细胞的优势。对相同肿瘤区室中CD163巨噬细胞群的研究表明,所研究的TAM在dMMR癌的核心和侵袭性边缘占主导地位,而在肿瘤基质中具有PD-L1表型的CD163巨噬细胞占主导地位。
    结论:这项研究揭示了PD-L1,CTLA-4,LAG-3和CD3+的显着优势,dMMR结直肠癌中的CD8+淋巴细胞。进一步研究不同肿瘤区室的免疫景观可能对CRC患者具有较高的预后价值。因为它可能会扩大处方免疫治疗的标准。
    OBJECTIVE: Colorectal cancer is the most common malignancy worldwide. A number of pathological and molecular genetic criteria are currently used as predictors of the disease. They include assessment of MMR deficiency or MSI/MSS status, which among others, determine the immunogenicity of the tumor. In this regard, the evaluation of PD-L1, CTLA-4, and LAG-3 immune checkpoint molecules in different tumor compartments according to MMR status deserves special attention.
    METHODS: Multiplex immunohistochemistry was used to evaluate the expression of immune checkpoint molecules in the tumor core and at the invasive margin.
    RESULTS: Data analysis showed the predominance of PD-L1 (p = 0.011), CTLA-4 (p = 0.004), and LAG-3 (p = 0.013) expression at the invasive margin of dMMR carcinomas compared to pMMR samples. Quantitative analysis of TILs population in the tumor core and at the invasive margin allowed establishment of the predominance of CD3+ and CD8+ lymphocytes at the invasive margin of dMMR carcinomas. Study of the CD163+ macrophages population in the same tumor compartments revealed the predominance of the studied TAMs in the core and at the invasive margin of dMMR carcinomas and the predominance of CD163+ macrophages with PD-L1-phenotype in the tumor stroma.
    CONCLUSIONS: This study revealed a significant predominance of PD-L1, CTLA-4, LAG-3, and CD 3+ ,CD8+ lymphocytes in dMMR colorectal carcinomas. Further research on the immune landscape in different tumor compartments will likely have high prognostic value for CRC patients, as it might expand the criteria for prescribing immunotherapy.
    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
    淋巴细胞活化基因(Lag)-3是癌症的抑制性共受体和免疫检查点抑制剂(ICI)治疗的靶标。分析了Lag-3在T细胞激活后的免疫突触处的动态行为以阐明Lag-3抑制机制。Lag-3形成簇并且在T细胞活化后与T细胞受体微簇(TCR-MC)共定位,类似于PD-1。Lag-3阻断抗体(Abs)抑制Lag-3和TCR-MC之间的共定位而不抑制Lag-3簇形成。Lag-3还抑制MHC-II非依赖性刺激和Lag-3Ab,不阻断MHC-II结合的化合物仍然可以阻断Lag-3的抑制功能,这表明Lag-3Ab通过解离TCR-MC和Lag-3簇的共组装来阻断Lag-3抑制信号。与PD-1和Lag-3Ab增强T细胞反应的组合益处一致,双特异性Lag-3/PD-1拮抗剂有效抑制簇形成和PD-1和Lag-3与TCR-MC的共定位。因此,Lag-3抑制TCR-MC的T细胞活化,Lag-3ICI的目标是解离Lag-3与TCR-MC的共定位。
    Lymphocyte activation gene (Lag)-3 is an inhibitory co-receptor and target of immune checkpoint inhibitor (ICI) therapy for cancer. The dynamic behavior of Lag-3 was analyzed at the immune synapse upon T-cell activation to elucidate the Lag-3 inhibitory mechanism. Lag-3 formed clusters and co-localized with T-cell receptor microcluster (TCR-MC) upon T-cell activation similar to PD-1. Lag-3 blocking antibodies (Abs) inhibited the co-localization between Lag-3 and TCR-MC without inhibiting Lag-3 cluster formation. Lag-3 also inhibited MHC-II-independent stimulation and Lag-3 Ab, which did not block MHC-II binding could still block Lag-3\'s inhibitory function, suggesting that the Lag-3 Ab blocks the Lag-3 inhibitory signal by dissociating the co-assembly of TCR-MC and Lag-3 clusters. Consistent with the combination benefit of PD-1 and Lag-3 Abs to augment T-cell responses, bispecific Lag-3/PD-1 antagonists effectively inhibited both cluster formation and co-localization of PD-1 and Lag-3 with TCR-MC. Therefore, Lag-3 inhibits T-cell activation at TCR-MC, and the target of Lag-3 ICI is to dissociate the co-localization of Lag-3 with TCR-MC.
    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
    背景:与其他种族/族裔群体相比,患有乳腺癌的非裔美国人(AA)的生存率更高。迄今未研究的结果差异是可溶性LAG-3(sLAG-3),从LAG-3免疫检查点受体切割,由于其在肿瘤微环境中的显着免疫抑制功能,LAG-3免疫检查点受体是新兴免疫疗法中失活的拟议靶标。先前的研究发现较低的sLAG-3基线水平与不良结局相关。
    方法:在95例原发性乳腺癌患者的横断面研究中(n=58白种人,n=37AA),我们使用非参数Mann-Whitneyu-Test对独立样本测量了治疗前血液样本中的sLAG-3(ELISApg/ml),and,按种族计算sLAG-3与循环细胞因子的Pearsonr相关系数。
    结果:与高加索患者相比,AA患者的平均sLAG-3水平较低(1377.6vs3690.3,P=.002),三阴性乳腺癌(TNBC)患者与非TNBC恶性肿瘤患者相比(P=0.02)。当TNBC肿瘤患者被排除在分析之外时,AA患者(n=21)和白种人患者(n=40)的sLAG-3水平差异显著(1937.4vs4182.4,P=.06).在白种人患者中,sLAG-3与IL-6、IL-8和IL-10相关(r=.69,P<.001;r=.70,P<.001;r=.46,P=.01;分别)。对于AA患者,sLAG-3仅与IL-6相关(r=.37,P=.03)。
    结论:我们首次报道非裔美国人乳腺癌患者治疗前sLAG-3水平相对较低,独立于TNBC地位,与循环细胞因子的共表达减少。裂解LAG-3的机制和预后作用,特别是在不同的结果中,还有待阐明。
    BACKGROUND: Worse survival persists for African-Americans (AA) with breast cancer compared to other race/ethnic groups despite recent improvements for all. Unstudied in outcomes disparities to date is soluble LAG-3 (sLAG-3), cleaved from the LAG-3 immune checkpoint receptor which is a proposed target for deactivation in emerging immunotherapies due to its prominent immunosuppressive function in the tumoral microenvironment. A prior study has found that lower sLAG-3 baseline level was associated with poor outcomes.
    METHODS: In a cross-sectional study of 95 patients with primary breast cancer (n = 58 Caucasian, n = 37 AA), we measured sLAG-3 (ELISA pg/ml) in pre-treatment blood samples using the non-parametric Mann-Whitney u-Test for independent samples, and, calculated Pearson r correlation coefficients of sLAG-3 with circulating cytokines by race.
    RESULTS: Mean sLAG-3 level was lower in AA compared to Caucasian patients (1377.6 vs 3690.3, P = .002), and in patients with triple-negative breast cancer (TNBC) compared to those with non-TNBC malignancies (P = .02). When patients with TNBC tumors were excluded from analyses, the difference in sLAG-3 level between AA (n = 21) and Caucasian patients (n = 40) substantially remained (1937.4 vs 4182.4, P = .06). Among Caucasian patients, sLAG-3 was correlated with IL-6, IL-8 and IL-10 (r = .69, P < .001; r = .70, P < .001; and, r = .46, P = .01; respectively). For AA patients, sLAG-3 was correlated only with IL-6 (r = .37, P = .03).
    CONCLUSIONS: We present the first report that African-American breast cancer patients might have comparatively low pre-treatment sLAG-3 levels, independent of TNBC status, along with reduced co-expression with circulating cytokines. The mechanistic and prognostic role of cleaved LAG-3, particularly in disparate outcomes, remains to be elucidated.
    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
    系统性红斑狼疮(SLE)代表一种影响多个系统的自身免疫性疾病。在治疗这种情况时,重点主要围绕炎症抑制和免疫抑制。因此,靶向治疗已成为一种流行的方法.目前,在SLE治疗的背景下,寻求高度敏感和特别有效的靶标获得了显著的动力.淋巴细胞活化基因-3(LAG-3)作为与pMHC-II结合的关键抑制性受体,从而有效地抑制自身免疫反应。纤维蛋白原样蛋白1(FGL1)是LAG-3的主要免疫抑制配体,它们的联合作用显示出有效的免疫抑制作用。这种复杂的机制通过用FGL1靶向LAG-3为潜在的SLE治疗铺平了道路。这项工作全面总结了LAG-3在SLE发病机制中的作用,并阐明了利用FGL1作为SLE治疗方法的可行性。它引入了一种新的治疗靶标,并在SLE治疗的临床背景下开辟了治疗考虑的新途径。
    Systemic lupus erythematosus (SLE) represents an autoimmune disorder that affects multiple systems. In the treatment of this condition, the focus primarily revolves around inflammation suppression and immunosuppression. Consequently, targeted therapy has emerged as a prevailing approach. Currently, the quest for highly sensitive and specifically effective targets has gained significant momentum in the context of SLE treatment. Lymphocyte activation gene-3 (LAG-3) stands out as a crucial inhibitory receptor that binds to pMHC-II, thereby effectively dampening autoimmune responses. Fibrinogen-like protein 1 (FGL1) serves as the principal immunosuppressive ligand for LAG-3, and their combined action demonstrates a potent immunosuppressive effect. This intricate mechanism paves the way for potential SLE treatment by targeting LAG-3 with FGL1. This work provides a comprehensive summary of LAG-3\'s role in the pathogenesis of SLE and elucidates the feasibility of leveraging FGL1 as a therapeutic approach for SLE management. It introduces a novel therapeutic target and opens up new avenues of therapeutic consideration in the clinical context of SLE treatment.
    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
    免疫检查点受体的阻断在肿瘤免疫治疗中显示出突出的功效。抗淋巴细胞激活基因-3(LAG-3)的有希望的治疗已经被认为是下一个有效的治疗,但对LAG-3介导的免疫抑制机制的了解仍然有限。这里,利用高分辨率分子成像,我们发现了通过LAG-3抑制CD4T细胞的机制,其中抗原呈递细胞(APC)上的LAG-3结合的主要组织相容性复合物(MHC)II类分子聚集在免疫突触的中心区域,并通过T细胞受体驱动的内化运动性向表达LAG-3的CD4和CD8T细胞转运。因此,APC上MHCII类分子的下调导致其抗原呈递功能的减弱和CD4T细胞活化的损害。从这些数据来看,建议抗LAG-3治疗具有通过LAG-3直接阻断抑制性信号传导的效力,并同时通过LAG-3介导的反式内吞作用降低APC上MHCII类表达,以从T细胞耗尽中恢复。
    Blockade of immune checkpoint receptors has shown outstanding efficacy for tumor immunotherapy. Promising treatment with anti-lymphocyte-activation gene-3 (LAG-3) has already been recognized as the next efficacious treatment, but there is still limited understanding of the mechanism of LAG-3-mediated immune suppression. Here, utilizing high-resolution molecular imaging, we find a mechanism of CD4 T cell suppression via LAG-3, in which LAG-3-bound major histocompatibility complex (MHC) class II molecules on antigen-presenting cells (APCs) gather at the central region of an immunological synapse and are trans-endocytosed by T cell receptor-driven internalization motility toward CD4 and CD8 T cells expressing LAG-3. Downregulation of MHC class II molecules on APCs thus results in the attenuation of their antigen-presentation function and impairment of CD4 T cell activation. From these data, anti-LAG-3 treatment is suggested to have potency to directly block the inhibitory signaling via LAG-3 and simultaneously reduce MHC class II expression on APCs by LAG-3-mediated trans-endocytosis for recovery from 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
    肿瘤免疫治疗已经引起了相当大的关注,正在成为癌症治疗的新标准。常规目标,如VEGF和EGFR,已扩展到其他,包括BRAF和PD-1/PD-L1,它们在最近的癌症治疗中显示出显著的潜力。这篇综述旨在简要概述通过靶向VEGF调节PD-1/PD-L1表达的疗法的影响和机制。EGFR,LAG-3、CTLA-4和BRAF。我们调查了PD-1/PD-L1表达的调节如何影响生长因子信号传导,揭示肿瘤微环境中免疫调节途径和生长因子网络之间的相互作用。通过阐明这些相互作用,我们旨在为癌症免疫治疗提供新的潜在协同治疗策略。
    Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
    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
    尽管对早发型先兆子痫的临床方面进行了大量研究,我们对胎盘发育不充分的免疫学后果的理解仍然不完整.早发型先兆子痫的Th1优势特征显著影响母体免疫耐受,免疫检查点分子在这些机制中的作用尚未完全阐明。我们的研究旨在填补这些关键的知识空白。本研究共纳入34例诊断为早发型先兆子痫的孕妇和34例健康孕妇。将来自静脉血的单核细胞片段分离并冷冻。使用多色流式细胞术鉴定CD8+和CD8-NK细胞亚群并与其免疫检查点分子表达进行比较。通过ELISA测量血清CD226水平。根据我们的措施,在NKdim和NKbright亚群中,CD8-亚群的频率均显著高于CD8+亚群.在所有研究的亚群中,与健康女性相比,在先兆子痫组中检测到显著更低的CD226表面表达。然而,而两组间可溶性CD226分子水平无差异,CD112和CD155表面表达差异显著。我们的研究结果强调了CD8+和CD8-NK亚群在Th1型免疫环境中的重要作用。这加深了我们对早发型先兆子痫的理解,并表明每个亚群都可能有助于这种情况下的补偿机制和免疫平衡的恢复。这是制定有效干预措施的关键一步。
    Despite the numerous studies on the clinical aspects of early-onset preeclampsia, our understanding of the immunological consequences of inadequate placenta development remains incomplete. The Th1-predominance characteristic of early-onset preeclampsia significantly impacts maternal immunotolerance, and the role of immune checkpoint molecules in these mechanisms is yet to be fully elucidated. Our study aims to fill these crucial knowledge gaps. A total of 34 pregnant women diagnosed with early-onset preeclampsia and 34 healthy pregnant women were enrolled in this study. A mononuclear cell fragment from the venous blood was separated and frozen. The CD8+ and CD8- NK cell subpopulations were identified and compared to their immune checkpoint molecule expressions using multicolor flow cytometry. The serum CD226 levels were measured by ELISA. Based on our measures, the frequency of the CD8- subpopulation was significantly higher than that of the CD8+ counterpart in both the NKdim and NKbright subsets. Significantly lower CD226 surface expressions were detected in the preeclamptic group compared to healthy women in all the investigated subpopulations. However, while no difference was observed in the level of the soluble CD226 molecule between the two groups, the CD112 and CD155 surface expressions were significantly different. Our study\'s findings underscore the significant role of the CD8+ and CD8- NK subpopulations in the Th1-dominated immune environment. This deepens our understanding of early-onset preeclampsia and suggests that each subpopulation could contribute to the compensation mechanisms and the restoration of the immunological balance in this condition, a crucial step toward developing effective interventions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase II
    Relatlimab(rela;抗LAG-3)加nivolumab(nivo;抗PD-1)可安全有效地治疗晚期黑色素瘤。我们设计了一项试验(NCT03743766),其中晚期黑色素瘤患者接受rela,尼沃,或rela+nivo询问rela+nivo的免疫机制。这项正在进行的试验的生物标本分析表明,rela+nivo导致CD8+T细胞受体信号传导能力增强,CD8+T细胞分化改变,尽管保留了耗尽曲线,但仍导致细胞毒性升高。细胞毒性和耗竭特征的共表达是由PRDM1、BATF、ETV7和TOX。在rela+nivo后出现的克隆扩增的CD8+T细胞中,效应子功能上调。rela+nivo肿瘤内CD8+T细胞特征与良好的预后相关。这种肿瘤内rela+nivo特征在外周血中被验证为CD38+TIM3+CD8+T细胞的频率升高。总的来说,我们证明,尽管保留了耗尽特征,但细胞毒性可以增强,这将为未来的治疗策略提供信息。
    Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future therapeutic strategies.
    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
    慢性病毒感染和癌症中的耗尽的CD8T(Tex)细胞具有抑制受体(IR)的持续共表达。Tex细胞可以通过阻断IR来恢复活力,例如PD-1,但是通过共同靶向包括PD-1和LAG-3在内的多个IR,可以实现协同恢复和增强的疾病控制。为了剖析这些IR通路被破坏时固有的分子变化,我们研究了慢性感染期间PD-1和/或LAG-3丢失对Tex细胞的影响.这些分析揭示了PD-1和LAG-3在调节Tex细胞增殖和效应子功能中的不同作用。分别。此外,这些研究确定了LAG-3在维持TOX和Tex细胞耐久性方面的重要作用,以及LAG-3依赖性电路,该电路产生了Tex细胞的CD94/NKG2亚群,具有通过识别应激配体Qa-1b介导的增强的细胞毒性,在人类中也有类似的观察。这些分析解开了PD-1和LAG-3的非冗余机制及其在调节Tex细胞中的协同作用。
    Exhausted CD8 T (Tex) cells in chronic viral infection and cancer have sustained co-expression of inhibitory receptors (IRs). Tex cells can be reinvigorated by blocking IRs, such as PD-1, but synergistic reinvigoration and enhanced disease control can be achieved by co-targeting multiple IRs including PD-1 and LAG-3. To dissect the molecular changes intrinsic when these IR pathways are disrupted, we investigated the impact of loss of PD-1 and/or LAG-3 on Tex cells during chronic infection. These analyses revealed distinct roles of PD-1 and LAG-3 in regulating Tex cell proliferation and effector functions, respectively. Moreover, these studies identified an essential role for LAG-3 in sustaining TOX and Tex cell durability as well as a LAG-3-dependent circuit that generated a CD94/NKG2+ subset of Tex cells with enhanced cytotoxicity mediated by recognition of the stress ligand Qa-1b, with similar observations in humans. These analyses disentangle the non-redundant mechanisms of PD-1 and LAG-3 and their synergy in regulating Tex cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号