Lymphocyte Activation Gene 3 Protein

淋巴细胞活化基因 3 蛋白
  • 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
    目的:免疫检查点抑制剂彻底改变了肾细胞癌(RCC)的治疗方法,但是许多患者对治疗没有反应,大多数患者随着时间的推移会出现耐药性疾病。因此,对替代免疫调节剂的需求日益增加。共抑制分子T细胞免疫球蛋白和ITIM结构域(TIGIT)可能在对批准的免疫检查点抑制剂的抗性中发挥作用,并且正在作为潜在的治疗靶标进行研究。这项研究的目的是量化RCC中肿瘤浸润T细胞的TIGIT阳性。
    方法:我们采用了包含原发性肾癌肿瘤标本的组织微阵列,邻近的正常肾组织,和RCC转移使用定量免疫荧光分析定量肿瘤浸润性CD3+T细胞内的TIGIT。我们还将这些结果与其他四种肿瘤类型(黑色素瘤,非小细胞肺,子宫颈,和头颈癌)。
    结果:我们没有观察到原发性RCC肿瘤和患者匹配的转移样本之间TIGIT阳性的显著差异。我们发现RCC的TIGIT阳性程度与肺癌相当,但低于黑色素瘤,子宫颈,头颈癌.将TIGIT阳性与先前发表的相关分析进行比较,我们小组的患者匹配的空间蛋白质组数据显示TIGIT与检查点蛋白PD-1和LAG3之间呈负相关.
    结论:我们的研究结果支持对可能用TIGIT靶向抗体治疗的患者,对原发性或转移性肾癌标本中T细胞的TIGIT表达进行仔细评估,因为TIGIT阳性的增加可能与治疗应答的可能性更大相关。
    OBJECTIVE: Immune checkpoint inhibitors have revolutionized the treatment of renal cell carcinoma (RCC), but many patients do not respond to therapy and the majority develop resistant disease over time. Thus, there is increasing need for alternative immunomodulating agents. The co-inhibitory molecule T-cell immunoglobulin and ITIM domain (TIGIT) may play a role in resistance to approved immune checkpoint inhibitors and is being investigated as a potential therapeutic target. The purpose of this study was to quantify TIGIT positivity in tumor-infiltrating T cells in RCC.
    METHODS: We employed tissue microarrays containing specimens from primary RCC tumors, adjacent normal renal tissue, and RCC metastases to quantify TIGIT within tumor-infiltrating CD3+ T cells using quantitative immunofluorescent analysis. We also compared these results to TIGIT+ CD3+ levels in four other tumor types (melanoma, non-small cell lung, cervical, and head and neck cancers).
    RESULTS: We did not observe significant differences in TIGIT positivity between primary RCC tumors and patient-matched metastatic samples. We found that the degree of TIGIT positivity in RCC is comparable to that in lung cancer but lower than that in melanoma, cervical, and head and neck cancers. Correlation analysis comparing TIGIT positivity to previously published, patient-matched spatial proteomic data by our group revealed a negative association between TIGIT and the checkpoint proteins PD-1 and LAG3.
    CONCLUSIONS: Our findings support careful evaluation of TIGIT expression on T cells in primary or metastatic RCC specimens for patients who may be treated with TIGIT-targeting antibodies, as increased TIGIT positivity might be associated with a greater likelihood of response to therapy.
    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:淋巴细胞活化基因3(LAG-3)被认为是下一代免疫检查点和免疫疗法的有希望的预后生物标志物。与程序性细胞死亡蛋白1/程序性死亡配体1和细胞毒性T淋巴细胞抗原4抑制剂一样,正电子发射断层扫描(PET)成像策略有助于制定LAG-3相关治疗的临床决策.在这项研究中,我们开发并验证了68Ga标记的环肽示踪剂,用于LAG-3表达的PET成像。
    方法:用68GaCl3修饰和放射性标记一系列LAG-3靶向环肽,并评估其亲和力和特异性,生物分布,药代动力学,和体外和体内辐射剂量学。此外,构建hu-PBL-SCID(PBL)小鼠模型以验证[68Ga]Ga-CC09-1用于使用纵向PET成像绘制LAG-3+淋巴细胞浸润物的能力。最后,[68Ga]Ga-CC09-1被转化为首次人体研究以评估其安全性,LAG-3表达的生物分布和成像潜力。
    结果:使用一系列靶向LAG-3的环肽作为先导化合物来设计和开发68Ga标记的PET示踪剂。体外结合测定显示[68Ga]Ga-CC09-1在中国仓鼠卵巢-人LAG-3细胞和外周血单核细胞中具有更高的亲和力和特异性。体内PET成像显示[68Ga]Ga-CC09-1具有更好的成像能力,在注射后60分钟时,肿瘤摄取更高,为每克1.35±0.33%的注射剂量,肿瘤与肌肉的比率为17.18±3.20。此外,[68Ga]Ga-CC09-1可以检测脾脏中的LAG-3+淋巴细胞浸润,PBL小鼠的肺和唾液腺。在黑色素瘤和非小细胞肺癌患者中,在[68Ga]Ga-CC09-1PET中观察到具有适度肿瘤摄取的原发性病变,与[18F]FDGPET相比。更重要的是,[68Ga]Ga-CC09-1描绘了大肿瘤内LAG-3表达的异质性。
    结论:这些发现巩固了[68Ga]Ga-CC09-1是定量肿瘤微环境中LAG-3表达的有前途的PET示踪剂,表明其在抗LAG-3治疗中作为患者分层和治疗反应监测的伴随诊断的潜力。
    BACKGROUND: Lymphocyte activation gene 3 (LAG-3) has been considered as the next generation of immune checkpoint and a promising prognostic biomarker of immunotherapy. As with programmed cell death protein-1/programmed death-ligand 1 and cytotoxic T-lymphocyte antigen-4 inhibitors, positron emission tomography (PET) imaging strategies could benefit the development of clinical decision-making of LAG-3-related therapy. In this study, we developed and validated 68Ga-labeled cyclic peptides tracers for PET imaging of LAG-3 expression in bench-to-bedside studies.
    METHODS: A series of LAG-3-targeted cyclic peptides were modified and radiolabeled with 68GaCl3 and evaluated their affinity and specificity, biodistribution, pharmacokinetics, and radiation dosimetry in vitro and in vivo. Furthermore, hu-PBL-SCID (PBL) mice models were constructed to validate the capacity of [68Ga]Ga-CC09-1 for mapping of LAG-3+ lymphocytes infiltrates using longitudinal PET imaging. Lastly, [68Ga]Ga-CC09-1 was translated into the first-in-human studies to assess its safety, biodistribution and potential for imaging of LAG-3 expression.
    RESULTS: A series of cyclic peptides targeting LAG-3 were employed as lead compounds to design and develop 68Ga-labeled PET tracers. In vitro binding assays showed higher affinity and specificity of [68Ga]Ga-CC09-1 in Chinese hamster ovary-human LAG-3 cells and peripheral blood mononuclear cells. In vivo PET imaging demonstrated better imaging capacity of [68Ga]Ga-CC09-1 with a higher tumor uptake of 1.35±0.33 per cent injected dose per gram and tumor-to-muscle ratio of 17.18±3.20 at 60 min post-injection. Furthermore, [68Ga]Ga-CC09-1 could detect the LAG-3+ lymphocyte infiltrates in spleen, lung and salivary gland of PBL mice. In patients with melanoma and non-small cell lung cancer, primary lesions with modest tumor uptake were observed in [68Ga]Ga-CC09-1 PET, as compared with that of [18F]FDG PET. More importantly, [68Ga]Ga-CC09-1 delineated the heterogeneity of LAG-3 expression within large tumors.
    CONCLUSIONS: These findings consolidated that [68Ga]Ga-CC09-1 is a promising PET tracer for quantifying the LAG-3 expression in tumor microenvironment, indicating its potential as a companion diagnostic for patients stratification and therapeutic response monitoring in anti-LAG-3 therapy.
    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
    人乳头瘤病毒(HPV)是头颈部鳞状细胞癌(HNSCC)的病因。探讨HPV抗原在抗肿瘤免疫中的作用,我们通过在SCC肿瘤细胞系中表达HPV16E6和E7建立了小鼠模型。我们获得了两个可移植到C57BL/6受体中的HPV抗原表达克隆(C-225和C-100)。我们发现C-225在C57BL/6小鼠中引起完全根除(根除),而C-100逐渐增长(增长)。我们使用流式细胞术检查了免疫肿瘤微环境(TME),发现根除或生长的肿瘤表现出差异的免疫特征,这可能会影响抗肿瘤免疫的结果。令人惊讶的是,CD8和CD4肿瘤浸润淋巴细胞(TIL)的百分比在生长(C-100)中比根除(C-225)肿瘤高得多.然而,与根除肿瘤相比,TIL更有效地上调PD-1和LAG-3,并在生长肿瘤中表现出受损的效应子功能.C-225TME富含骨髓细胞,特别是多形核(PMN)髓源性抑制细胞(MDSC),而M-MDSC和肿瘤相关巨噬细胞(TAMs)的百分比在C-100TME中高得多,特别是M2-TAMs(CD206+)。C-225的完全根除取决于CD8T细胞,并在继发性肿瘤攻击时引起抗肿瘤记忆反应。我们采用DNA测序来鉴定继发性肿瘤攻击前后外周血淋巴细胞T细胞受体的差异。最后,C-225和C-100肿瘤系具有不同的体细胞突变。总的来说,通过建立两个SCC肿瘤细胞系,我们发现了不同的免疫TME,这可能是抗肿瘤免疫不同结果的基础。均表达HPV16E6和E7抗原。我们的实验模型可以提供一个平台,用于确定肿瘤内在与宿主内在的差异,以协调HNSCC中的免疫抑制TME,并鉴定使肿瘤细胞易受免疫攻击的新靶标。
    Human papilloma virus (HPV) is an etiological factor of head and neck squamous cell carcinoma (HNSCC). To investigate the role of HPV antigen in anti-tumor immunity, we established mouse models by expressing HPV16 E6 and E7 in a SCC tumor cell line. We obtained two HPV antigen-expressing clones (C-225 and C-100) transplantable into C57BL/6 recipients. We found that C-225 elicited complete eradication in C57BL/6 mice (eradicated), whereas C-100 grew progressively (growing). We examined immune tumor microenvironment (TME) using flow cytometry and found that eradicated or growing tumors exhibited differential immune profiles that may influence the outcome of anti-tumor immunity. Surprisingly, the percentage of CD8 and CD4 tumor-infiltrating lymphocytes (TILs) was much higher in growing (C-100) than eradicated (C-225) tumor. However, the TILs upregulated PD-1 and LAG-3 more potently and exhibited impaired effector functions in growing tumor compared to their counterparts in eradicated tumor. C-225 TME is highly enriched with myeloid cells, especially polymorphonuclear (PMN) myeloid-derived suppressor cells (MDSC), whereas the percentage of M-MDSC and tumor-associated macrophages (TAMs) was much higher in C-100 TME, especially M2-TAMs (CD206+). The complete eradication of C-225 depended on CD8 T cells and elicited anti-tumor memory responses upon secondary tumor challenge. We employed DNA sequencing to identify differences in the T cell receptor of peripheral blood lymphocytes pre- and post-secondary tumor challenge. Lastly, C-225 and C-100 tumor lines harbored different somatic mutations. Overall, we uncovered differential immune TME that may underlie the divergent outcomes of anti-tumor immunity by establishing two SCC tumor lines, both of which express HPV16 E6 and E7 antigens. Our experimental models may provide a platform for pinpointing tumor-intrinsic versus host-intrinsic differences in orchestrating an immunosuppressive TME in HNSCCs and for identifying new targets that render tumor cells vulnerable to immune attack.
    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:抗程序性死亡-1(PD-1)/抗PD-配体-1(PD-L1)途径抑制是晚期尿路上皮癌(UC)的标准方案;然而,其有限的疗效反映在报告的中等反应率上.本研究探讨了下一代共抑制受体(IRs;淋巴细胞激活基因3(LAG-3),T细胞免疫球蛋白和粘蛋白结构域3(TIM-3),和具有Ig和ITIM结构域的T细胞免疫受体(TIGIT))及其配体(LGs)对PD-(L)1阻断治疗的反应以及UC患者的肿瘤结局。
    方法:我们调查了接受PD-(L)1治疗的转移性UC病例(队列1:n=348,队列2:n=89,队列4:n=29)或涉及手术的晚期UC病例(队列3:n=293,队列5:n=90)。我们使用队列1、2和3评估了关于IR和LGs的mRNA表达谱和相应的临床信息。此外,我们使用多重免疫组织化学(mIHC)对来自队列4和5的福尔马林固定石蜡包埋样品阐明了这些靶向标志物的空间特征.生存,差异表达基因,和基因集富集分析进行。对于mIHC,我们还进行了定量分析,以将免疫和肿瘤细胞密度与患者生存相关联.
    结果:与TIM-3和TIGIT的表达相比,LAG-3的表达与PD-(L)1阻断的反应性强相关。在高LAG-3水平的肿瘤中,纤维蛋白原样蛋白1(FGL1)的表达增加对PD-(L)1阻断反应和总生存期有显著负效应.此外,高FGL1水平与CD4+调节性T细胞基因特征升高以及CD39和神经纤毛蛋白-1上调相关,两者均提示CD8+T细胞耗尽.mIHC分析显示,与CD8+LAG-3+细胞-肿瘤FGL1+细胞慢的患者相比,CD8+LAG-3+细胞-肿瘤FGL1+细胞慢的患者与生存率呈显著负相关。
    结论:LAG-3表达和高FGL1共表达是与免疫抑制背景相关的不良肿瘤结局的重要预测因素。这些发现是产生假设的,需要进一步的机制和临床研究旨在评估UC中的LAG-3/FGL1阻断。
    BACKGROUND: Anti-programmed death-1 (PD-1)/anti-PD-ligand-1 (PD-L1) pathway inhibition is a standard regimen for advanced urothelial carcinoma (UC); however, its limited efficacy has been reflected in reported medium response rates. This study explored the role of next-generation coinhibitory receptors (IRs; lymphocyte activation gene 3 (LAG-3), T-cell immunoglobulin and mucin domain 3 (TIM-3), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT)) and their ligands (LGs) in the response to PD-(L)1 blockade therapy and the oncological outcomes in patients with UC.
    METHODS: We investigated metastatic UC cases who underwent PD-(L)1 therapy (cohort 1: n=348, cohort 2: n=89, and cohort 4: n=29) or advanced UC cases involving surgery (cohort 3: n=293 and cohort 5: n=90). We assessed the mRNA expression profiles and corresponding clinical information regarding IRs and LGs using cohorts 1, 2, and 3. Additionally, we elucidated the spatial features of these targeted markers using multiplex immunohistochemistry (mIHC) on formalin-fixed paraffin-embedded samples from cohorts 4 and 5. Survival, differential expressed gene, and Gene Set Enrichment analyses were performed. For mIHC, quantitative analyses were also performed to correlate immune and tumor cell densities with patient survival.
    RESULTS: LAG-3 expression was strongly associated with the responsiveness of PD-(L)1 blockade compared with the expression of TIM-3 and TIGIT. In tumors with high LAG-3 levels, the increased expression of fibrinogen-like protein 1 (FGL1) had a significantly negative effect on the response to PD-(L)1 blockade and overall survival. Moreover, high FGL1 levels were associated with elevated CD4+ regulatory T-cell gene signatures and the upregulation of CD39 and neuropilin-1, with both indicating CD8+ T-cell exhaustion. mIHC analyses revealed that patients with stromal CD8+LAG-3+cellshigh-tumor FGL1+cellshigh exhibited a significant negative correlation with survival rates compared with those with stromal CD8+LAG-3+cellshigh-tumor FGL1+cellslow.
    CONCLUSIONS: LAG-3 expression and high FGL1 coexpression are important predictive factors of adverse oncological outcomes related to the presence of immunosuppressive contextures. These findings are hypothesis-generating, warranting further mechanistic and clinical studies aimed to evaluate LAG-3/FGL1 blockade in UC.
    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:淋巴细胞活化基因3(LAG-3)在活化的免疫细胞上表达,并已成为免疫检查点阻断的有希望的靶标。然而,关于LAG-3在肿瘤中的表达与患者预后之间的相关性,表明需要进一步研究LAG-3表达水平在肿瘤治疗中的意义。在这项研究中,68Ga-NOTA-XH05是一种靶向LAG-3的新型基于肽的正电子发射断层扫描(PET)示踪剂,用于非侵入性检测CpG寡核苷酸(CpG)治疗后黑色素瘤中LAG-3的表达,并探讨LAG-3表达与治疗效果之间的关系。
    方法:示踪剂68Ga-NOTA-XH05在制备和纯化后通过高效液相色谱法鉴定。进行细胞摄取和阻断实验以验证示踪剂在体外的特异性。流式细胞术检测B16-F10皮下肿瘤中LAG-3的表达,并分析其与示踪剂摄取的相关性以评估示踪剂的特异性。在对单侧或双侧B16-F10皮下肿瘤模型进行CpG治疗后进行PET成像和生物分布研究,以评估68Ga-NOTA-XH05在监测免疫疗法疗效和CpG的横观效应方面的能力。
    结果:纯化后,68Ga-NOTA-XH05表现出高的放射化学纯度和特异性。流式细胞术分析显示肿瘤中的LAG-3表达与68Ga-NOTA-XH05的摄取之间呈正相关。在用CpG处理的带有B16-F10的小鼠中,与对照组相比,使用68Ga-NOTA-XH05的PET成像显示出更高的肿瘤与血液比率(TBR)。此外,从CpG处理的小鼠获得的TBR值允许应答者和非应答者之间的区分。在双侧皮下肿瘤模型中,仅右侧肿瘤用瘤内注射CpG治疗,左侧肿瘤的TBR值明显高于对照组,表明68Ga-NOTA-XH05能有效监测CpG局部注射的全身效应。
    结论:我们的发现强调了68Ga-NOTA-XH05在评估肿瘤内LAG-3表达水平和评估免疫治疗反应方面的检测能力。从而表明有希望的临床转化前景。
    BACKGROUND: Lymphocyte activation gene 3 (LAG-3) is expressed on activated immune cells and has emerged as a promising target for immune checkpoints blockade. However, conflicting findings have been reported regarding the association between LAG-3 expression in tumors and patient prognosis, indicating the need for further investigation into the significance of LAG-3 expression levels in tumor therapies. In this study, 68Ga-NOTA-XH05, a novel peptide-based positron emission tomography (PET) tracer targeting LAG-3, was constructed to non-invasively detect LAG-3 expression in melanoma after CpG oligonucleotide (CpG) treatment and explore the relationship between LAG-3 expression and therapeutic effect.
    METHODS: The tracer 68Ga-NOTA-XH05 was identified by high-performance liquid chromatography after being prepared and purified. Cell uptake and blocking essays were performed to verify the specificity of the tracer in vitro. The expression of LAG-3 in B16-F10 subcutaneous tumors was monitored by flow cytometry, and its correlation with the tracer uptake was analyzed to evaluate the tracer specificity. PET imaging and biodistribution studies were conducted after CpG treatment of unilateral or bilateral B16-F10 subcutaneous tumor models to assess the ability of 68Ga-NOTA-XH05 in monitoring immunotherapy efficacy and the abscopal effect of CpG.
    RESULTS: Following purification, 68Ga-NOTA-XH05 exhibited high radiochemical purity and specificity. Flow cytometry analysis revealed a positive correlation between LAG-3 expression in tumors and the uptake of 68Ga-NOTA-XH05. In B16-F10 bearing mice treated with CpG, PET imaging using 68Ga-NOTA-XH05 demonstrated a higher tumor to blood ratio (TBR) compared with the control group. Furthermore, TBR values obtained from CpG-treated mice allowed for differentiation between responders and non-responders. In a bilateral subcutaneous tumor model where only right-sided tumors were treated with intratumoral injection of CpG, TBR values of left-sided tumors were significantly higher than those in the control group, indicating that 68Ga-NOTA-XH05 could effectively monitor the systemic effect of local CpG injection.
    CONCLUSIONS: Our findings highlight the detection capability of 68Ga-NOTA-XH05 in assessing LAG-3 expression levels within tumors and evaluating response to immunotherapy, thereby suggesting promising clinical translational prospects.
    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
    许多癌症患者无法从PD-L1/PD-1阻断免疫疗法中受益。T细胞中的PD-1和LAG-3共同上调是通过在T细胞中建立高度功能失调状态的抗性的主要机制之一。为了确定与人类癌症和T细胞中PD-1/LAG-3功能障碍相关的共同特征,获得了所有TCGA癌症免疫浸润物的多组表达谱。发现了调节免疫的PD-1/LAG-3功能失调特征,新陈代谢,遗传,和表观遗传途径,但特别是对TCR信号体的强化负调节。这些结果在具有组成型活性PD-1、LAG-3途径及其组合的T细胞系中得到验证。对PD-1/LAG-3T细胞的蛋白质组的差异分析显示参与E3泛素化途径的泛素连接酶的特异性富集。PD-1/LAG-3共阻断抑制CBL-B表达,虽然在临床开发中使用双特异性药物也抑制了C-CBL表达,这逆转了对PD-L1/PD-1阻断耐药的肺癌患者的T细胞功能障碍。CBL-B特异性小分子抑制剂与抗PD-1/抗LAG-3免疫疗法的组合在免疫疗法难以治疗的肺癌模型中显示出显著的治疗效果。克服PD-1/LAG-3介导的耐药性。
    Many cancer patients do not benefit from PD-L1/PD-1 blockade immunotherapies. PD-1 and LAG-3 co-upregulation in T-cells is one of the major mechanisms of resistance by establishing a highly dysfunctional state in T-cells. To identify shared features associated to PD-1/LAG-3 dysfunctionality in human cancers and T-cells, multiomic expression profiles were obtained for all TCGA cancers immune infiltrates. A PD-1/LAG-3 dysfunctional signature was found which regulated immune, metabolic, genetic, and epigenetic pathways, but especially a reinforced negative regulation of the TCR signalosome. These results were validated in T-cell lines with constitutively active PD-1, LAG-3 pathways and their combination. A differential analysis of the proteome of PD-1/LAG-3 T-cells showed a specific enrichment in ubiquitin ligases participating in E3 ubiquitination pathways. PD-1/LAG-3 co-blockade inhibited CBL-B expression, while the use of a bispecific drug in clinical development also repressed C-CBL expression, which reverted T-cell dysfunctionality in lung cancer patients resistant to PD-L1/PD-1 blockade. The combination of CBL-B-specific small molecule inhibitors with anti-PD-1/anti-LAG-3 immunotherapies demonstrated notable therapeutic efficacy in models of lung cancer refractory to immunotherapies, overcoming PD-1/LAG-3 mediated resistance.
    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
    低反应率,治疗复发,耐药性仍然是免疫检查点阻断(ICB)治疗癌症的关键挑战。在这里,我们报道了特异性肿瘤抑制因子(TS)的缺失会诱导炎症反应并促进免疫抑制性肿瘤微环境。重要的是,这些TS的低表达与免疫检查点抑制介质的高表达相关。在这里,我们确定,通过使用基于体内CRISPR/Cas9的功能丧失筛选,NF1、TSC1和TGF-βRII作为TS调节免疫组成。这三个TS中的每一个的缺失导致染色质可及性的改变并增强IL6-JAK3-STAT3/6炎症途径。这导致免疫抑制的景观,以LAG3+CD8和CD4T细胞数量增加为特征。ICB靶向LAG3和PD-L1同时抑制NF1-,临床前三阴性乳腺癌(TNBC)小鼠模型的转移进展。TSC1-或TGF-βRII缺陷型肿瘤。因此,我们的研究揭示了TS通过免疫区室的非细胞自主调节在调节转移中的作用,并为针对NF1-,TSC1或TGF-βRII灭活的癌症。
    Low response rate, treatment relapse, and resistance remain key challenges for cancer treatment with immune checkpoint blockade (ICB). Here we report that loss of specific tumor suppressors (TS) induces an inflammatory response and promotes an immune suppressive tumor microenvironment. Importantly, low expression of these TSs is associated with a higher expression of immune checkpoint inhibitory mediators. Here we identify, by using in vivo CRISPR/Cas9 based loss-of-function screening, that NF1, TSC1, and TGF-β RII as TSs regulating immune composition. Loss of each of these three TSs leads to alterations in chromatin accessibility and enhances IL6-JAK3-STAT3/6 inflammatory pathways. This results in an immune suppressive landscape, characterized by increased numbers of LAG3+ CD8 and CD4 T cells. ICB targeting LAG3 and PD-L1 simultaneously inhibits metastatic progression in preclinical triple negative breast cancer (TNBC) mouse models of NF1-, TSC1- or TGF-β RII- deficient tumors. Our study thus reveals a role of TSs in regulating metastasis via non-cell-autonomous modulation of the immune compartment and provides proof-of-principle for ICB targeting LAG3 for patients with NF1-, TSC1- or TGF-β RII-inactivated cancers.
    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
    自身免疫性甲状腺疾病(AITD)是一种常见的自身免疫性疾病。在110,945例和1,084,290例对照的GWAS荟萃分析中,225个基因座处的290个序列变体与AITD相关。在这些变体中,115以前未报告。多组学分析产生了MHC区外的235个候选基因,该发现强调了参与T细胞调节的基因的重要性。一种罕见的5'-UTR变体(rs781745126-T,MAF=冰岛的0.13%)在LAG3中具有最大的作用(OR=3.42,P=2.2×10-16),并为规范蛋白翻译起始位点上游的开放阅读框产生新的起始密码子。rs781745126-T减少了活化淋巴细胞亚群上抑制性免疫检查点LAG-3共受体的mRNA和表面表达,并在杂合子中降低了血浆中LAG-3水平。rs781745126-T的所有三个纯合携带者都具有AITD,其中一人还患有另外两种T细胞介导的疾病,那就是白癜风和1型糖尿病。rs781745126-T名义上与白癜风相关(OR=5.1,P=6.5×10-3),但与1型糖尿病无关。因此,rs781745126-T的作用类似于抑制LAG-3的药物,LAG-3可释放免疫反应,并可导致甲状腺功能障碍和白癜风作为不良事件.这说明了多组学方法如何揭示潜在的药物靶标和安全性问题。
    Autoimmune thyroid disease (AITD) is a common autoimmune disease. In a GWAS meta-analysis of 110,945 cases and 1,084,290 controls, 290 sequence variants at 225 loci are associated with AITD. Of these variants, 115 are previously unreported. Multiomics analysis yields 235 candidate genes outside the MHC-region and the findings highlight the importance of genes involved in T-cell regulation. A rare 5\'-UTR variant (rs781745126-T, MAF = 0.13% in Iceland) in LAG3 has the largest effect (OR = 3.42, P = 2.2 × 10-16) and generates a novel start codon for an open reading frame upstream of the canonical protein translation initiation site. rs781745126-T reduces mRNA and surface expression of the inhibitory immune checkpoint LAG-3 co-receptor on activated lymphocyte subsets and halves LAG-3 levels in plasma among heterozygotes. All three homozygous carriers of rs781745126-T have AITD, of whom one also has two other T-cell mediated diseases, that is vitiligo and type 1 diabetes. rs781745126-T associates nominally with vitiligo (OR = 5.1, P = 6.5 × 10-3) but not with type 1 diabetes. Thus, the effect of rs781745126-T is akin to drugs that inhibit LAG-3, which unleash immune responses and can have thyroid dysfunction and vitiligo as adverse events. This illustrates how a multiomics approach can reveal potential drug targets and safety concerns.
    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
    结核病(TB)仍然是全球健康挑战,长期联合药物治疗的副作用和日益严重的耐药性问题阻碍了其治疗。因此,开发新的治疗策略至关重要。这项研究的重点是免疫检查点分子(IC)的作用和CD8+T细胞的功能在寻找新的潜在目标的结核病。
    我们对来自TB数据库GSE83456的92个TB样本和61个健康个体(HI)样本进行了差异表达基因分析和CD8+T细胞功能基因分析,其中包含34,603个基因的数据。使用GSE54992数据集来验证发现。此外,我们对感染结核分枝杆菌的灵长类动物和接种卡介苗的灵长类动物的单细胞数据进行了聚类分析.
    发现LAG-3基因的过表达是肺结核病(PTB)和肺外结核病(EPTB)的潜在重要特征。进一步的相关性分析显示,LAG-3基因与GZMB,穿孔素,IL-2和IL-12。在TB感染期间和BCG疫苗接种后,在T细胞和巨噬细胞中观察到LAG-3表达的显着时间和空间变化。
    LAG-3在TB样品中过表达。靶向LAG-3可能代表结核病的潜在治疗靶标。
    UNASSIGNED: Tuberculosis (TB) persists as a global health challenge, with its treatment hampered by the side effects of long-term combination drug therapies and the growing issue of drug resistance. Therefore, the development of novel therapeutic strategies is critical. This study focuses on the role of immune checkpoint molecules (ICs) and functions of CD8+ T cells in the search for new potential targets against TB.
    UNASSIGNED: We conducted differential expression genes analysis and CD8+ T cell functional gene analysis on 92 TB samples and 61 healthy individual (HI) samples from TB database GSE83456, which contains data on 34,603 genes. The GSE54992 dataset was used to validated the findings. Additionally, a cluster analysis on single-cell data from primates infected with mycobacterium tuberculosis and those vaccinated with BCG was performed.
    UNASSIGNED: The overexpression of LAG-3 gene was found as a potentially important characteristic of both pulmonary TB (PTB) and extrapulmonary TB (EPTB). Further correlation analysis showed that LAG-3 gene was correlated with GZMB, perforin, IL-2 and IL-12. A significant temporal and spatial variation in LAG-3 expression was observed in T cells and macrophages during TB infection and after BCG vaccination.
    UNASSIGNED: LAG-3 was overexpressed in TB samples. Targeting LAG-3 may represent a potential therapeutic target for tuberculosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号