TIGIT

TIGIT
  • 文章类型: Journal Article
    T细胞免疫球蛋白和基于免疫受体酪氨酸的抑制基序结构域(TIGIT),一个新发现的检查站,其特征在于其在CD4+T细胞上的表达升高,CD8+T细胞,自然杀伤(NK)细胞,调节性T细胞(Tregs),和肿瘤浸润淋巴细胞(TIL)。迄今为止的研究表明,TIGIT与许多癌症中NK细胞和T细胞的耗竭有关。CD155是人类TIGIT的特异性配体,由于其与TIGIT的关键相互作用,成为免疫治疗的关键靶标。此外,许多研究表明,TIGIT与其他免疫检查点抑制剂(ICIs)和/或传统治疗的组合在结直肠癌(CRC)中引起有效的抗肿瘤反应。这篇评论概述了该结构,函数,以及与跨多种免疫系统细胞类型的TIGIT相关的信号通路。此外,关注TIGIT在CRC进展中的作用,这项研究回顾了探索CRC中基于TIGIT的免疫治疗的各种研究。
    The T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), a newly discovered checkpoint, is characterized by its elevated expression on CD4 + T cells, CD8 + T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). Research to date has been shown that TIGIT has been linked to exhaustion of NK cell both and T cells in numerous cancers. CD155, being the specific ligand of TIGIT in humans, emerges as a key target for immunotherapy owing to its crucial interaction with TIGIT. Furthermore, numerous studies have demonstrated that the combination of TIGIT with other immune checkpoint inhibitors (ICIs) and/or traditional treatments elicits a potent antitumor response in colorectal cancer (CRC). This review provides an overview of the structure, function, and signaling pathways associated with TIGIT across multiple immune system cell types. Additionally, focusing on the role of TIGIT in the progression of CRC, this study reviewed various studies exploring TIGIT-based immunotherapy in CRC.
    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
    近年来,免疫疗法已成为癌症治疗的重要组成部分。然而,它的功效在各种癌症类型中仍然有限,突出未满足的需求。脊髓灰质炎病毒受体相关2(PVRL2)和脊髓灰质炎病毒受体(PVR)是Nectin和Nectin样分子家族的成员,以其作为细胞-细胞粘附分子的作用而闻名。随着免疫治疗的发展,它们作为免疫检查点因子参与肿瘤免疫机制的研究受到了广泛关注.PVRL2和PVR主要在肿瘤细胞和抗原呈递细胞上表达,绑定到PVRIG和TIGIT,分别,主要在T和NK细胞上发现,从而抑制抗肿瘤免疫。值得注意的是,妇科癌症如卵巢癌和子宫内膜癌表现出高表达水平的PVRL2和PVR,在各种其他实体和血液肿瘤中观察到类似的趋势。靶向这些免疫检查点途径提供了一个有前途的治疗途径,可能与现有的治疗方法相结合。然而,涉及这些结合的免疫调节机制,被称为DNAM-1轴,是复杂的,强调了解它对开发新疗法的重要性。本文全面综述了以PVRL2和PVR为中心的免疫调节机制。阐明它们对各种癌症类型的影响。
    In recent years, immunotherapy has emerged as a crucial component of cancer treatment. However, its efficacy remains limited across various cancer types, highlighting unmet needs. Poliovirus receptor-related 2 (PVRL2) and Poliovirus receptor (PVR) are members of the Nectin and Nectin-like Molecules family, known for their role as cell-cell adhesion molecules. With the development of immunotherapy, their involvement in tumor immune mechanisms as immune checkpoint factors has garnered significant attention. PVRL2 and PVR are predominantly expressed on tumor cells and antigen-presenting cells, binding to PVRIG and TIGIT, respectively, which are primarily found on T and NK cells, thereby suppressing antitumor immunity. Notably, gynecological cancers such as ovarian and endometrial cancers exhibit high expression levels of PVRL2 and PVR, with similar trends observed in various other solid and hematologic tumors. Targeting these immune checkpoint pathways offers a promising therapeutic avenue, potentially in combination with existing treatments. However, the immunomodulatory mechanism involving these bindings, known as the DNAM-1 axis, is complex, underscoring the importance of understanding it for developing novel therapies. This article comprehensively reviews the immunomodulatory mechanisms centered on PVRL2 and PVR, elucidating their implications for various cancer types.
    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
    小细胞肺癌(SCLC)占所有肺癌诊断的约10%至15%,并且由于其高死亡率而代表了紧迫的全球公共卫生挑战。常规治疗SCLC的疗效欠佳,以有限的抗肿瘤作用和频繁复发为特征。在这种情况下,新兴的研究转向免疫疗法与化疗相结合,这是一个快速发展的领域,在改善SCLC患者的临床结局方面显示出希望。通过最初开发用于非小细胞肺癌(NSCLC),这些疗法拓展了SCLC的新治疗途径.目前,一系列新兴热点治疗方法已显示出显著的治疗效果.基于化疗和免疫治疗的融合,以及新的免疫治疗剂的开发,SCLC的治疗已经看到了希望的未来。通过伴随使用化疗,在增强肿瘤免疫微环境方面取得了进展。免疫疗法,和酪氨酸激酶抑制剂(TKI),正如新兴的临床试验数据所证明的那样。此外,涉及免疫疗法的三方方法,靶向治疗,化疗对未来的临床应用来说似乎是吉祥的。克服对免疫治疗后方案的耐药性仍然是一个紧迫的探索领域。最后,双特异性抗体,过继细胞转移(ACT),溶瘤病毒,单一疗法,包括Delta样配体3(DLL3)和具有Ig和ITIM结构域的T细胞免疫受体(TIGIT),以及精准医学,可能是在SCLC中实现治愈结果的前瞻性途径。这篇综述旨在综合现有文献,并强调SCLC治疗的未来方向,承认该领域的持续挑战。此外,新的治疗药物和技术的不断发展使得SCLC治疗的未来越来越乐观.
    Small cell lung cancer (SCLC) constitutes approximately 10% to 15% of all lung cancer diagnoses and represents a pressing global public health challenge due to its high mortality rates. The efficacy of conventional treatments for SCLC is suboptimal, characterized by limited anti-tumoral effects and frequent relapses. In this context, emerging research has pivoted towards immunotherapy combined with chemotherapy, a rapidly advancing field that has shown promise in ameliorating the clinical outcomes of SCLC patients. Through originally developed for non-small cell lung cancer (NSCLC), these therapies have extended new treatment avenues for SCLC. Currently, a nexus of emerging hot-spot treatments has demonstrated significant therapeutic efficacy. Based on the amalgamation of chemotherapy and immunotherapy, and the development of new immunotherapy agents, the treatment of SCLC has seen the hoping future. Progress has been achieved in enhancing the tumor immune microenvironment through the concomitant use of chemotherapy, immunotherapy, and tyrosine kinase inhibitors (TKI), as evinced by emerging clinical trial data. Moreover, a tripartite approach involving immunotherapy, targeted therapy, and chemotherapy appears auspicious for future clinical applications. Overcoming resistance to post-immunotherapy regimens remains an urgent area of exploration. Finally, bispecific antibodies, adoptive cell transfer (ACT), oncolytic virus, monotherapy, including Delta-like ligand 3 (DLL3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT), as well as precision medicine, may present a prospective route towards achieving curative outcomes in SCLC. This review aims to synthesize extant literature and highlight future directions in SCLC treatment, acknowledging the persistent challenges in the field. Furthermore, the continual development of novel therapeutic agents and technologies renders the future of SCLC treatment increasingly optimistic.
    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
    骨髓来源的抑制细胞(MDSC)是免疫细胞的异质家族,包括粒细胞(CD14neg/CD15/HLA-DRneg)和单核细胞亚型(CD14/CD15neg/HLA-DRneg)。在本研究中,我们发现表达粒细胞标志物CD15的单核细胞群体在结直肠癌(CRC)患者的外周血(PB)和肿瘤组织中均显著增加.进一步的表型分析证实了该单核细胞亚群的粒细胞样特征,这与这些患者(pts)的PB中粒细胞-单核细胞前体(GMPs)的增加有关。机械上,这种粒细胞样单核细胞群通过诱导TIGIT和参与NKp30抑制NK细胞活性。因此,在CRC患者的PB和肿瘤组织中发现了功能受损的TIGIT+NK细胞的频率增加.总的来说,我们通过显示这种新型MDSC的增加,为CRC中发生的肿瘤免疫逃逸提供了新的机制解释,在PB和CRC组织中,能够显著损害NK细胞的效应子功能,从而代表了癌症免疫疗法的潜在治疗靶标。
    Myeloid-derived suppressor cells (MDSCs) are a heterogeneous family of immune cells including granulocytic (CD14neg/CD15+/HLA-DRneg) and monocytic subtypes (CD14+/CD15neg/HLA-DRneg). In the present study, we found a population of monocytes expressing the granulocyte marker CD15 that significantly increased in both peripheral blood (PB) and tumoral tissues of patients with colorectal cancer (CRC). Further phenotypical analysis confirmed the granulocytic-like features of this monocyte subpopulation that is associated with an increase in granulocyte-monocyte precursors (GMPs) in the PB of these patients (pts). Mechanistically, this granulocyte-like monocyte population suppressed NK cell activity by inducing TIGIT and engaging NKp30. Accordingly, an increased frequency of TIGIT+ NK cells with impaired functions was found in both the PB and tumoral tissue of CRC pts. Collectively, we provided new mechanistic explanations for tumor immune escape occurring in CRC by showing the increase in this new kind of MDSC, in both PB and CRC tissue, which is able to significantly impair the effector functions of NK cells, thereby representing a potential therapeutic target for cancer immunotherapy.
    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
    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: 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
    本研究旨在探讨银屑病和特应性皮炎的共同致病机制,两种T细胞介导的自身免疫性疾病。利用单细胞转录组测序数据,我们发现Treg细胞主要在银屑病和特应性皮炎中表达TIGIT,并鉴定了高度表达SGK1的巨噬细胞亚群。这些细胞可以通过NECTIN2-TIGIT信号通路与T细胞相互作用,抑制T细胞分化为促炎表型,从而揭示了两种疾病中共同的免疫调节机制。此外,我们发现,在两种疾病模型中,SGK1的抑制都会加剧炎症反应.这些发现不仅为银屑病和特应性皮炎的常见治疗策略提供了新的视角,而且突出了在未来治疗中考虑这些分子相互作用的重要性。通过进一步的qPCR验证这些观察结果,免疫荧光,动物研究已经确定了治疗银屑病和特应性皮炎的潜在新靶点。
    This study aims to explore the common pathogenic mechanisms of psoriasis and atopic dermatitis, two T-cell-mediated autoimmune diseases. Utilizing single-cell transcriptomic sequencing data, we revealed that Treg cells primarily express TIGIT in both psoriasis and atopic dermatitis, and identified a subset of macrophages that highly express SGK1. These cells can interact with T cells via the NECTIN2-TIGIT signaling pathway, inhibiting the differentiation of T cells into a pro-inflammatory phenotype, thereby uncovering a common immunoregulatory mechanism in both diseases. Furthermore, we discovered that inhibition of SGK1 exacerbates the inflammatory response in disease models of both conditions. These findings not only provide a new perspective for a common therapeutic strategy for psoriasis and atopic dermatitis but also highlight the importance of considering these molecular interactions in future treatments. Validation of these observations through further qPCR, immunofluorescence, and animal studies has identified potential new targets for the treatment of psoriasis and atopic dermatitis.
    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:鼻窦粘膜黑色素瘤(SNMM)是一种罕见但侵袭性肿瘤,预后不良。共抑制受体T细胞免疫球蛋白和含粘结构域-3(TIM-3),淋巴细胞活化基因-3(LAG-3)、T细胞免疫球蛋白和基于免疫受体酪氨酸的抑制基序结构域(TIGIT)是抗癌免疫治疗的有希望的新靶点.这些免疫检查点分子(ICM)的表达谱和潜在的预后影响尚未在SNMM中表征。
    方法:TIGIT的免疫组织化学染色,对来自27例原发性SNMM患者的肿瘤组织样品进行LAG-3和TIM-3。ICM表达式和人口统计参数之间的关联,AJCC肿瘤分期,总生存率,对无复发生存率进行回顾性分析。
    结果:SNMM患者在原发肿瘤中TIGIT+和TIM-3+肿瘤浸润淋巴细胞(TIL)的数量较低,存活时间明显长于TIGIT+和TIM-3+TILs程度较高的患者。TIM-3或TIGIT淋巴细胞的高浸润与较高的T4期和5年生存率降低有关。
    结论:我们确定高密度的TIM-3+和TIGIT+TILs是SNMM中强的阴性预后生物标志物。这表明TIM-3和TIGIT有助于SNMM中的免疫抑制,并为基于这种下一代免疫检查点抑制剂的新型治疗策略提供了理论基础。有必要对更多病例进行前瞻性研究,以证实我们的发现及其对免疫治疗的意义。
    BACKGROUND: Sinonasal mucosal melanoma (SNMM) is a rare but aggressive tumor with a poor prognosis. The co-inhibitory receptors T cell immunoglobulin and mucinodomain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3) and T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) are promising new targets in anti-cancer immunotherapy. The expression profiles of these immune checkpoint molecules (ICMs) and potential prognostic implications have not been characterized in SNMM yet.
    METHODS: Immunohistochemical staining for TIGIT, LAG-3 and TIM-3 was performed on tumor tissue samples from 27 patients with primary SNMM. Associations between ICM expression and demographic parameters, AJCC tumor stage, overall survival, and recurrence-free survival were retrospectively analyzed.
    RESULTS: SNMM patients with low numbers of TIGIT+ and TIM-3+ tumor infiltrating lymphocytes (TILs) in the primary tumor survived significantly longer than patients with a high degree of TIGIT+ and TIM-3+ TILs. High infiltration with TIM-3+ or TIGIT+ lymphocytes was associated with the higher T4 stage and decreased 5-year survival.
    CONCLUSIONS: We identified high densities of TIM-3+ and TIGIT+ TILs as strong negative prognostic biomarkers in SNMM. This suggests that TIM-3 and TIGIT contribute to immunosuppression in SNMM and provides a rationale for novel treatment strategies based on this next generation of immune checkpoint inhibitors. Prospective studies with larger case numbers are warranted to confirm our findings and their implications for 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
    免疫检查点(IC)阻断和肿瘤特异性T细胞(ACT)的过继转移是治疗转移性黑色素瘤的两种主要策略。它们的组合可以增强抑制性肿瘤微环境中的T细胞激活,但与全身注射IC阻滞剂相关的自身免疫不良反应在此策略中持续存在.IC表达缺陷的肿瘤反应性T细胞的ACT将克服这个问题。为此,PD-1和TIGIT似乎是相关的候选人,因为它们在高度肿瘤反应性淋巴细胞上的共表达限制了它们在肿瘤微环境中的治疗功效,NT.我们的研究比较了PDCD1或TIGIT基因缺失对黑色素瘤特异性T淋巴细胞的抗肿瘤特性和T细胞适应性的影响。转录组学分析显示PD-1KOT细胞中细胞周期相关基因的下调,与生物学观察一致,而增殖途径保留在TIGITKOT细胞中。功能分析表明,PD-1KO和TIGITKOT细胞在体外和使用免疫缺陷小鼠的临床前黑色素瘤模型中表现出比其野生型对应物更好的抗肿瘤反应性。有趣的是,看来TIGITKOT细胞在体内更有效地抑制肿瘤细胞增殖,在肿瘤内的存留时间比PD-1KOT细胞长,与TIGIT缺失对T细胞适应性没有影响一致。一起来看,这些结果表明TIGIT缺失,超过PD-1删除,黑色素瘤特异性T细胞是未来免疫治疗策略的一个令人信服的选择.
    Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of PDCD1 or TIGIT genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1KO T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGITKO T-cells. Functional analyses showed that PD-1KO and TIGITKO T-cells displayed superior antitumor reactivity than their wild-type counterpart in vitro and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGITKO T-cells were more effective at inhibiting tumor cell proliferation in vivo, and persist longer within tumors than PD-1KO T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic 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
    TIGIT是一种替代的检查点受体(CR),其抑制作用可促进NK细胞的移植物抗白血病作用。鉴于在急性髓系白血病(AML)患者中循环的NK细胞具有显着的免疫能力,我们询问活化NK细胞的过继转移是否会受益于额外的TIGIT阻断.因此,我们表征了细胞因子诱导的记忆样(CIML)-NK细胞和NK细胞系的抑制性CRs表达。此外,我们分析了AML患者(CCLE和BeatAML2.0队列)中CR配体的转录,并使用体外细胞毒性试验评估了CR阻断的功效,CD69、CD107a和IFN-γ表达。替代但非经典的CR在健康供体NK细胞上大量表达,甚至进一步上调了CIML-NK细胞。根据我们的发现,CD155,一种重要的TIGIT配体,在AML上可靠地表达,我们显示NK-92对CD155+-AML母细胞的杀伤作用得到改善,但有趣的是,在TIGIT阻断的存在下,ML-NK细胞的杀伤作用并非如此。此外,我们的计算机模拟数据(n=671)显示预后不良的AML患者表现出CD86lowCD112/CD155high表型,而预后较好的患者则表现为CD86高CD112/CD155低表型。总的来说,我们的数据表明,AML母细胞中复杂的CR配体表达谱可能是AML患者中观察到的内在NK细胞耗竭的一种解释,过继性NK-92转移联合TIGIT阻断可以克服这种耗竭.
    TIGIT is an alternative checkpoint receptor (CR) whose inhibition promotes Graft-versus-Leukemia effects of NK cells. Given the significant immune-permissiveness of NK cells circulating in acute myeloid leukemia (AML) patients, we asked whether adoptive transfer of activated NK cells would benefit from additional TIGIT-blockade. Hence, we characterized cytokine-induced memory-like (CIML)-NK cells and NK cell lines for the expression of inhibitory CRs. In addition, we analyzed the transcription of CR ligands in AML patients (CCLE and Beat AML 2.0 cohort) in silico and evaluated the efficacy of CR blockade using in vitro cytotoxicity assays, CD69, CD107a and IFN-γ expression. Alternative but not classical CRs were abundantly expressed on healthy donor NK cells and even further upregulated on CIML-NK cells. In line with our finding that CD155, one important TIGIT-ligand, is reliably expressed on AMLs, we show improved killing of CD155+-AML blasts by NK-92 but interestingly not CIML-NK cells in the presence of TIGIT-blockade. Additionally, our in silico data (n = 671) show that poor prognosis AML patients rather displayed a CD86low CD112/CD155high phenotype, whereas patients with a better outcome rather exhibited a CD86high CD112/CD155low phenotype. Collectively, our data evidence that the complex CR ligand expression profile on AML blasts may be one explanation for the intrinsic NK cell exhaustion observed in AML patients which might be overcome with adoptive NK-92 transfer in combination with TIGIT-blockade.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号