B7 Antigens

B7 抗原
  • 文章类型: Journal Article
    脊索瘤是一种罕见的骨肿瘤,手术后经常复发,目前的治疗方法预后较差。这项研究旨在通过识别临床样品中的靶蛋白以及肿瘤微环境因素来增强疗效,从而确定脊索瘤潜在的新型免疫治疗靶标。通过单细胞RNA测序分析了14个脊索瘤样本,B7-H3和IL-7被确定为潜在的靶标和增效剂,分别。合成B7-H3靶向的嵌合抗原受体T(CAR-T)细胞和表达IL-7的B7-H3CAR-T细胞,并在体外评估其抗肿瘤活性。包括原发性脊索瘤类器官模型。B7-H3CAR-T/IL-7疗法显示出增强的细胞毒性和延长的抗肿瘤细胞作用持续时间。此外,IL-7调节培养的CAR-T细胞的有利亚群,T细胞表面免疫检查点表达减少,和增强的T细胞功能。将IL-7分子掺入B7-H3CAR结构增强了CAR-T细胞功能并改善了CAR-T细胞功效,从而为脊索瘤的治疗提供了一种新的双重治疗策略。
    Chordoma is a rare bone tumor that frequently recurs after surgery, and the prognosis is poor with current treatments. This study aimed to identify potential novel immunotherapeutic targets for chordomas by identifying target proteins in clinical samples as well as tumor microenvironmental factors to enhance efficacy. Fourteen chordoma samples were analyzed by single-cell RNA sequencing, and B7-H3 and IL-7 were identified as potential targets and potentiators, respectively. B7-H3-targeted chimeric antigen receptor T (CAR-T) cells and B7-H3 CAR-T cells expressing IL-7 were synthesized and their anti-tumor activity evaluated in vitro, including in primary chordoma organoid models. The B7-H3 CAR-T/IL-7 therapy showed enhanced cytotoxicity and prolonged duration of action against tumor cells. Additionally, IL-7 modulated favorable subpopulations of cultured CAR-T cells, diminished immune checkpoint expression on T-cell surfaces, and enhanced T-cell functionality. The incorporation of IL-7 molecules into the B7-H3 CAR structure augmented CAR-T-cell function and improved CAR-T-cell efficacy, thus providing a novel dual therapeutic strategy for chordoma 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
    近年来,在新兴免疫疗法的发展中,B7-H3也被称为CD276,并已成为针对神经胶质瘤和其他肿瘤的新型嵌合抗原受体(CAR)-T靶标。引起了广泛的关注。然而,B7-H3具有三种亚型(2、3和4Ig),在肿瘤尤其是神经胶质瘤中具有争议的表达和难以捉摸的功能。目前的研究主要集中在不同B7-H3亚型产生的调控因子和相关机制。首先,我们已经确定2Ig在恶性程度较高的神经胶质瘤中占主导地位,4Ig广泛表达,而3Ig在所有胶质瘤中均显示阴性表达。接下来,我们进一步发现RNA结合蛋白膜联蛋白A2(ANXA2)是B7-H3亚型维持所必需的,但未能确定选择4Ig或2Ig。RNA甲基转移酶NOP2/SunRNA甲基转移酶2(NSUN2)和5-甲基胞嘧啶读取器Y-box结合蛋白1(YBX1)促进2Ig的产生。我们的发现揭示了一系列因素(ANXA2/NSUN2/YBX1),可以确定神经胶质瘤中B7-H3不同亚型的替代产生。我们的结果旨在帮助同行更清楚地了解肿瘤患者中B7H3的表达和调节机制。并为B7H3作为免疫治疗靶点的设计提供更好的策略。
    In recent years, in the development of emerging immunotherapy, B7-H3 is also termed as CD276 and has become a novel chimeric antigen receptor (CAR)-T target against glioma and other tumours, and aroused extensive attention. However, B7-H3 has three isoforms (2, 3 and 4Ig) with the controversial expression and elusive function in tumour especially glioma. The current study mainly focuses on the regulatory factors and related mechanisms of generation of different B7-H3 isoforms. First, we have determined that 2Ig is dominant in glioma with high malignancy, and 4Ig is widely expressed, whereas 3Ig shows negative expression in all glioma. Next, we have further found that RNA binding protein annexin A2 (ANXA2) is essential for B7-H3 isoform maintenance, but fail to determine the choice of 4Ig or 2Ig. RNA methyltransferase NOP2/Sun RNA methyltransferase 2 (NSUN2) and 5-methylcytosine reader Y-box binding protein 1 (YBX1) facilitate the production of 2Ig. Our findings have uncovered a series of factors (ANXA2/NSUN2/YBX1) that can determine the alternative generation of different isoforms of B7-H3 in glioma. Our result aims to help peers gain a clearer understanding of the expression and regulatory mechanisms of B7H3 in tumour patients, and to provide better strategies for designing B7H3 as a target in 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
    嵌合抗原受体(CAR)T细胞靶向成纤维细胞生长因子受体4(FGFR4),横纹肌肉瘤(RMS)中高表达的表面酪氨酸受体,已经处于临床发展阶段,但是肿瘤异质性和次优激活可能会阻碍它们的效力。在这里,我们报告了FGFR4CAR的共刺激和靶向特性的优化策略。我们用CD28的那些替换CD8铰链和跨膜结构域和4-1BB共刺激结构域。除了侵袭性肿瘤细胞系外,所得CAR在几种RMS异种移植模型中显示出增强的抗肿瘤活性。RMS559。通过寻找RMS核心调节转录因子MYOD1的直接靶标,我们确定了另一种表面蛋白,CD276,作为潜在靶标。双顺反子CAR(BiCisCAR)靶向FGFR4和CD276,含有两个不同的共刺激结构域,与优化的FGFR4特异性CAR和具有相同4-1BB共刺激结构域的其他BiCisCAR相比,具有优异的延长的持久性和增强的抗肿瘤活性。因此,我们的研究为针对RMS中的FGFR4和CD276的CART细胞疗法奠定了原理证明。
    Chimeric antigen receptor (CAR) T-cells targeting Fibroblast Growth Factor Receptor 4 (FGFR4), a highly expressed surface tyrosine receptor in rhabdomyosarcoma (RMS), are already in the clinical phase of development, but tumour heterogeneity and suboptimal activation might hamper their potency. Here we report an optimization strategy of the co-stimulatory and targeting properties of a FGFR4 CAR. We replace the CD8 hinge and transmembrane domain and the 4-1BB co-stimulatory domain with those of CD28. The resulting CARs display enhanced anti-tumor activity in several RMS xenograft models except for an aggressive tumour cell line, RMS559. By searching for a direct target of the RMS core-regulatory transcription factor MYOD1, we identify another surface protein, CD276, as a potential target. Bicistronic CARs (BiCisCAR) targeting both FGFR4 and CD276, containing two distinct co-stimulatory domains, have superior prolonged persistent and invigorated anti-tumor activities compared to the optimized FGFR4-specific CAR and the other BiCisCAR with the same 4-1BB co-stimulatory domain. Our study thus lays down the proof-of-principle for a CAR T-cell therapy targeting both FGFR4 and CD276 in RMS.
    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:一些癌症已经发生了儿科肿瘤学的进展;然而,肉瘤的新疗法还不够。使用嵌合抗原受体(CAR)T细胞的细胞免疫疗法在白血病中显示出巨大的益处,淋巴瘤和多发性骨髓瘤,但在小儿实体瘤如横纹肌肉瘤(RMS)和骨肉瘤(OS)中的成功率要低得多。平衡“达到目标”的问题,肿瘤外毒性“,研究者鉴定出B7-H3是一种广泛表达的肿瘤抗原,在正常组织中表达受限.我们假设通过趋化因子受体的快速归巢和通过B7-H3的CAR接合将增强实体瘤中的CART细胞功效。
    方法:我们产生了B7-H3CART细胞,这些细胞也表达白细胞介素-8(IL-8)受体,CXCR2。细胞因子生产,流式细胞术,海马测定和RNA测序用于比较B7-H3CXCR2(BC2)CART细胞与B7-H3CART细胞。我们开发了IL-8过表达的人RMS小鼠模型来测试体内归巢和细胞毒性。
    结果:我们证明了IL-8通过RMS和OS表达,并且在辐射后表达显着增加。IL-8受体的过表达,B7-H3CART细胞上的CXCR2增强了归巢到表达IL-8的肿瘤中,增强T细胞代谢并导致显著的肿瘤消退。
    结论:这些发现值得进一步研究BC2CART细胞作为RMS患者的治疗方法。OS和其他B7-H3-表达,产生IL-8的实体瘤。
    BACKGROUND: Advances in pediatric oncology have occurred for some cancers; however, new therapies for sarcoma have been inadequate. Cellular immunotherapy using chimeric antigen receptor (CAR) T cells has shown dramatic benefits in leukemia, lymphoma, and multiple myeloma but has been far less successful in pediatric solid tumors such as rhabdomyosarcoma (RMS) and osteosarcoma (OS). Balancing issues of \"on-target, off-tumor toxicity\", investigators have identified B7-H3 as a broadly expressed tumor antigen with otherwise restricted expression on normal tissues. We hypothesized that rapid homing via a chemokine receptor and CAR engagement through B7-H3 would enhance CAR T cell efficacy in solid tumors.
    METHODS: We generated B7-H3 CAR T cells that also express the Interleukin-8 (IL-8) receptor, CXCR2. Cytokine production, flow cytometry, Seahorse assays and RNA sequencing were used to compare the B7-H3 CXCR2 (BC2) CAR T cells with B7-H3 CAR T cells. We developed an IL-8 overexpressing human RMS mouse model to test homing and cytotoxicity in vivo.
    RESULTS: We demonstrate that IL-8 is expressed by RMS and OS and expression significantly increases after radiation. Overexpression of an IL-8 receptor, CXCR2, on B7-H3 CAR T cells enhances homing into IL-8 expressing tumors, augments T cell metabolism and leads to significant tumor regression.
    CONCLUSIONS: These findings warrant further investigation into the use of BC2 CAR T cells as a treatment for patients with RMS, OS and other B7-H3-expressing, IL-8 producing solid tumors.
    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
    自然杀伤(NK)细胞传统上通过流式细胞术使用标记物(CD16/CD56/CD3)的组合进行鉴定,因为特定的NK细胞标记仍然缺失。在这里,我们调查了与CD16/CD56/CD3相比,CD314,CD335和NKp80的实用性,以更可靠地识别人类血液中的NK细胞,用于诊断目的。
    从健康供体(HD)和经常与经典NK细胞标志物丢失/下调相关的疾病的患者收集的总共156个外周血(PB)样本按照EuroFlow方案进行了免疫表型分析,旨在比较CD314,CD335和NKp80的总血液NK细胞的染色谱,以及不同标记组合对其准确鉴定的性能。
    NKp80表现出优异的性能(与CD314和CD335)用于鉴定HD血液中的NK细胞。此外,NKp80改进了传统的基于CD16/CD56/CD3的策略,以识别HD和反应过程中的PBNK细胞,特别是当与CD16结合用于进一步准确的NK细胞亚群时。尽管NKp80+CD16改善了克隆/肿瘤NK细胞的鉴定,特别是在CD56-病例中(53%),在25%的患者中观察到NKp80的异常下调,其中CD56可用作互补NK细胞标志物。由于NKp80也在T细胞上表达,我们注意到NKp80+细胞毒性T细胞在更晚期成熟阶段的数量增加,主要是成年人。
    在这里,我们提出了一种新的稳健方法来鉴定PBNK细胞,基于NKp80+CD16的组合。然而,在NK细胞的慢性淋巴增殖性疾病中,建议添加CD56来鉴定克隆NK细胞,由于其频繁的异常NKp80表型。
    UNASSIGNED: Natural killer (NK) cells are traditionally identified by flow cytometry using a combination of markers (CD16/CD56/CD3), because a specific NK-cell marker is still missing. Here we investigated the utility of CD314, CD335 and NKp80, compared to CD16/CD56/CD3, for more robust identification of NK-cells in human blood, for diagnostic purposes.
    UNASSIGNED: A total of 156 peripheral blood (PB) samples collected from healthy donors (HD) and patients with diseases frequently associated with loss/downregulation of classical NK-cell markers were immunophenotyped following EuroFlow protocols, aimed at comparing the staining profile of total blood NK-cells for CD314, CD335 and NKp80, and the performance of distinct marker combinations for their accurate identification.
    UNASSIGNED: NKp80 showed a superior performance (vs. CD314 and CD335) for the identification of NK-cells in HD blood. Besides, NKp80 improved the conventional CD16/CD56/CD3-based strategy to identify PB NK-cells in HD and reactive processes, particularly when combined with CD16 for further accurate NK-cell-subsetting. Although NKp80+CD16 improved the identification of clonal/tumor NK-cells, particularly among CD56- cases (53%), aberrant downregulation of NKp80 was observed in 25% of patients, in whom CD56 was useful as a complementary NK-cell marker. As NKp80 is also expressed on T-cells, we noted increased numbers of NKp80+ cytotoxic T-cells at the more advanced maturation stages, mostly in adults.
    UNASSIGNED: Here we propose a new robust approach for the identification of PB NK-cells, based on the combination of NKp80 plus CD16. However, in chronic lymphoproliferative disorders of NK-cells, addition of CD56 is recommended to identify clonal NK-cells, due to their frequent aberrant NKp80- phenotype.
    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
    尽管免疫疗法取得了成功,克服癌症中的免疫抗性仍然具有挑战性。我们确定了肿瘤相关巨噬细胞(TAMs)的独特生态位,共表达T细胞免疫球蛋白和含粘蛋白结构域3(TIM3)和T细胞活化的V结构域免疫球蛋白抑制剂(VISTA),占主导地位的人类和小鼠肿瘤对大多数目前使用的免疫疗法具有抗性。TIM3+VISTA+TAM由具有低(新)抗原性和T细胞耗尽特征的富含IL-4的肿瘤维持。TIM3+VISTA+TAM显示抗炎和原瘤表型,同时无法感觉到I型干扰素(IFN)。这是由死于免疫原性细胞死亡(ICD)的癌细胞建立的。死亡的癌细胞不仅触发了自分泌I型IFN,而且还暴露了HMGB1/VISTA,使TAM3/VISTA参与抑制旁分泌IFN反应。因此,TIM3/VISTA阻断与紫杉醇协同作用,诱导ICD的化疗,通过肿瘤坏死因子相关的凋亡诱导配体(TRAIL)信号将TIM3+VISTA+TAMs复极化为可杀死癌细胞的促炎TAMs。我们建议靶向TIM3+VISTA+TAM来克服免疫抗性肿瘤。
    Despite the success of immunotherapy, overcoming immunoresistance in cancer remains challenging. We identified a unique niche of tumor-associated macrophages (TAMs), coexpressing T cell immunoglobulin and mucin domain-containing 3 (TIM3) and V-domain immunoglobulin suppressor of T cell activation (VISTA), that dominated human and mouse tumors resistant to most of the currently used immunotherapies. TIM3+VISTA+ TAMs were sustained by IL-4-enriching tumors with low (neo)antigenic and T cell-depleted features. TIM3+VISTA+ TAMs showed an anti-inflammatory and protumorigenic phenotype coupled with inability to sense type I interferon (IFN). This was established with cancer cells succumbing to immunogenic cell death (ICD). Dying cancer cells not only triggered autocrine type I IFNs but also exposed HMGB1/VISTA that engaged TIM3/VISTA on TAMs to suppress paracrine IFN-responses. Accordingly, TIM3/VISTA blockade synergized with paclitaxel, an ICD-inducing chemotherapy, to repolarize TIM3+VISTA+ TAMs to proinflammatory TAMs that killed cancer cells via tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling. We propose targeting TIM3+VISTA+ TAMs to overcome immunoresistant tumors.
    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
    尽管针对PD1/PD-L1免疫检查点的抑制剂显示出相当好的结果,相当比例的头颈部鳞状细胞癌(HNSCC)患者对治疗无反应.除了使用不同的治疗策略,另一种可能性是针对在这些无反应肿瘤中运行的其他免疫检查点。为了获得在HNSCC肿瘤细胞上表达哪些检查点配体的概述,如果这些配体受到HGF/MET信号的影响,我们使用mRNA测序和基于抗体的技术在6种HNSCC肿瘤细胞系中鉴定检查点配体.此外,我们将我们的结果与mRNA测序数据进行了比较.从我们调查的检查点配体来看,VISTA在RNA水平上表达最高,并且也是最普遍表达的。PD-L2和B7-H3的表达相对较低,并且在所有细胞系中不存在相同程度。然而,B7-H4,仅在底特律562细胞系中检测到。关于HGF对配体水平的影响,PD-L2表达随着HGF刺激而增强,而其他检查点配体水平随着刺激而降低。在HGF刺激下,底特律562细胞系中的B7-H4水平急剧下降。这是令人感兴趣的,因为在文献中都报道了检查点配体和生长因子与上皮-间质转化有关。
    Although inhibitors targeting the PD1/PD-L1 immune checkpoint are showing comparably good outcomes, a significant percentage of head and neck squamous cell carcinoma (HNSCC) patients do not respond to treatment. Apart from using different treatment strategies, another possibility would be to target other immune checkpoints operating in these non-responding tumors. To obtain an overview of which checkpoint ligands are expressed on HNSCC tumor cells and if these ligands are affected by HGF/MET signaling, we used mRNA sequencing and antibody-based techniques for identifying checkpoint ligands in six HNSCC tumor cell lines. Furthermore, we compared our results to mRNA sequencing data. From the checkpoint ligands we investigated, VISTA was expressed the highest at the RNA level and was also the most ubiquitously expressed. PD-L2 and B7-H3 were expressed comparably lower and were not present in all cell lines to the same extent. B7-H4, however, was only detectable in the Detroit 562 cell line. Concerning the effect of HGF on the ligand levels, PD-L2 expression was enhanced with HGF stimulation, whereas other checkpoint ligand levels decreased with stimulation. B7-H4 levels in the Detroit 562 cell line drastically decreased with HGF stimulation. This is of interest because both the checkpoint ligand and the growth factor are reported to be connected to epithelial-mesenchymal transition in the literature.
    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:胰腺癌是最致命的恶性肿瘤之一,缺乏治疗选择使其更加致命。嵌合抗原受体T细胞(CAR-T)免疫疗法彻底改变了癌症治疗,并在治疗血液恶性肿瘤方面取得了重大突破,然而,它在治疗实体癌方面的成功仍然有限,主要是由于缺乏肿瘤特异性抗原。另一方面,延长的传统制造过程带来了挑战,服用2至6周,影响患者预后。CD276最近已成为抗实体癌治疗的潜在治疗靶标。这里,我们研究了CD276CAR-T和快速制造的CAR-T对胰腺癌的疗效.
    方法:在本研究中,CD276CAR-T是通过CAR结构制备的,携带376.96scFv序列,CD8铰链和跨膜结构域,4-1BB和CD3ζ胞内结构域。此外,CD276快速制造的CAR-T(名为CD276DashCAR-T)是通过缩短体外培养时间以减少CAR-T制造时间而创新开发的。我们评估了CD276CAR-T的抗肿瘤功效,并通过检测免疫表型进一步比较了DashCAR-T和常规CAR-T在体外和体内的功能评估。杀伤能力,CAR-T的扩增能力和肿瘤根除作用
    结果:我们发现CD276在多种实体癌细胞系中强烈表达,CD276CAR-T可以有效杀死这些实体癌细胞。此外,DashCAR-T在48-72小时内成功制造,随后进行了功能验证。体外,与常规CAR-T相比,CD276DashCAR-T具有较低分化的表型和强大的增殖能力。体内异种移植小鼠模型,CD276DashCAR-T显示出增强的抗胰腺癌功效和T细胞扩增。此外,除了高剂量组,小鼠体重保持稳定,老鼠的状态正常。
    结论:在这项研究中,我们证明了CD276CAR-T在体外和体内对胰腺癌细胞表现出强大的活性。更重要的是,我们证明了制造的可行性,以减少的时间产生的CD276DashCAR-T具有可接受的安全性和优异的抗肿瘤功效。上述研究结果表明,CD276DashCAR-T免疫疗法可能是一种新颖且有前途的胰腺癌治疗策略。
    BACKGROUND: Pancreatic cancer is one of the most lethal malignancies and the lack of treatment options makes it more deadly. Chimeric Antigen Receptor T-cell (CAR-T) immunotherapy has revolutionized cancer treatment and made great breakthroughs in treating hematological malignancies, however its success in treating solid cancers remains limited mainly due to the lack of tumor-specific antigens. On the other hand, the prolonged traditional manufacturing process poses challenges, taking 2 to 6 weeks and impacting patient outcomes. CD276 has recently emerged as a potential therapeutic target for anti-solid cancer therapy. Here, we investigated the efficacy of CD276 CAR-T and rapidly-manufactured CAR-T against pancreatic cancer.
    METHODS: In the present study, CD276 CAR-T was prepared by CAR structure carrying 376.96 scFv sequence, CD8 hinge and transmembrane domain, 4-1BB and CD3ζ intracellular domains. Additionally, CD276 rapidly-manufactured CAR-T (named CD276 Dash CAR-T) was innovatively developed by shortening the duration of ex vitro culture to reduce CAR-T manufacturing time. We evaluated the anti-tumor efficacy of CD276 CAR-T and further compared the functional assessment of Dash CAR-T and conventional CAR-T in vitro and in vivo by detecting the immunophenotypes, killing ability, expansion capacity and tumor-eradicating effect of CAR-T.
    RESULTS: We found that CD276 was strongly expressed in multiple solid cancer cell lines and that CD276 CAR-T could efficiently kill these solid cancer cells. Moreover, Dash CAR-T was successfully manufactured within 48-72 h and the functional validation was carried out subsequently. In vitro, CD276 Dash CAR-T possessed a less-differentiated phenotype and robust proliferative ability compared to conventional CAR-T. In vivo xenograft mouse model, CD276 Dash CAR-T showed enhanced anti-pancreatic cancer efficacy and T cell expansion. Besides, except for the high-dose group, the body weight of mice was maintained stable, and the state of mice was normal.
    CONCLUSIONS: In this study, we proved CD276 CAR-T exhibited powerful activity against pancreatic cancer cells in vitro and in vivo. More importantly, we demonstrated the manufacturing feasibility, acceptable safety and superior anti-tumor efficacy of CD276 Dash CAR-T generated with reduced time. The results of the above studies indicated that CD276 Dash CAR-T immunotherapy might be a novel and promising strategy for pancreatic cancer 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
    背景:尽管在新靶标和嵌合抗原受体(CAR)-T的构建方面不断改进,CAR-T治疗后复发仍是一项重大挑战.肿瘤微环境(TME)与CAR-T疗法的疗效密切相关。T细胞激活的V域Ig抑制因子(VISTA),在调节TME方面发挥着多方面和有争议的作用,不仅在抗原呈递细胞上充当配体,而且在T细胞上充当受体。然而,VISTA控制内源性T细胞激活的特征和潜在机制,这是重塑TME的关键,仍未完全阐明。
    方法:免疫活性B急性淋巴细胞白血病(B-ALL),淋巴瘤和黑色素瘤鼠模型被用来研究在CD19和hCAIXCAR-T细胞治疗后TME内内源性T细胞的特征,分别。此外,我们研究了由干扰素(IFN)-γ信号控制的VISTA在调节B-ALL小鼠内源性T细胞活化和功能中的作用.
    结果:我们证明了CD19CAR-T或hCAIXCAR-T细胞疗法的给药在B-ALL的TME内引起内源性T细胞增强的免疫应答,淋巴瘤和黑色素瘤小鼠,从而证实了CAR-T细胞的功效。然而,在缺乏IFN-γ信号传导的TME中,VISTA水平仍然升高,导致内源性T细胞的细胞毒性减弱,并降低B-ALL受体的存活率。用CD19CAR-T细胞治疗的小鼠在长期缓解期间表现出内源性记忆T细胞的比例增加。具有预防B-ALL再攻击的肿瘤反应能力。与野生型(WT)CAR-T处理的小鼠相比,对WT和IFN-γ-/-受体施用IFN-γ-/-CAR-T导致内源性CD4+和CD8+效应子数量减少,同时表现出原始样CD4+T和记忆CD8+T细胞的数量增加。VISTA表达在静息或记忆CD4+T细胞中持续升高,与程序性细胞死亡蛋白-1(PD-1)表达T亚群不同的定位。阻断VISTA信号增强了树突状细胞诱导的增殖和同系T细胞产生的细胞因子。
    结论:我们的发现证实内源性T细胞活化和功能受VISTA调节,这与CAR-T的治疗效率相关,并为CAR-T治疗中的复发病例提供了有希望的治疗策略。
    BACKGROUND: Despite continuous improvements in the new target and construction of chimeric antigen receptor (CAR)-T, relapse remains a significant challenge following CAR-T therapy. Tumor microenvironment (TME) strongly correlates with the efficacy of CAR-T therapy. V-domain Ig suppressor of T-cell activation (VISTA), which exerts a multifaceted and controversial role in regulating the TME, acts not only as a ligand on antigen-presenting cells but also functions as a receptor on T cells. However, the characteristics and underlying mechanisms governing endogenous T-cell activation by VISTA, which are pivotal for reshaping the TME, remain incompletely elucidated.
    METHODS: The immunocompetent B acute lymphoblastic leukemia (B-ALL), lymphoma, and melanoma murine models were employed to investigate the characteristics of endogenous T cells within the TME following CD19 and hCAIX CAR-T cell therapy, respectively. Furthermore, we examined the role of VISTA controlled by interferon (IFN)-γ signaling in regulating endogenous T-cell activation and functionality in B-ALL mice.
    RESULTS: We demonstrated that the administration of CD19 CAR-T or hCAIX CAR-T cell therapy elicited augmented immune responses of endogenous T cells within the TME of B-ALL, lymphoma, and melanoma mice, thereby substantiating the efficacy of CAR-T cell efficacy. However, in the TME lacking IFN-γ signaling, VISTA levels remained elevated, resulting in attenuated cytotoxicity of endogenous T cells and reduced B-ALL recipient survival. Mice treated with CD19 CAR-T cells exhibited increased proportions of endogenous memory T cells during prolonged remission, which possessed the tumor-responsive capabilities to protect against B-ALL re-challenge. Compared with wild-type (WT) CAR-T treated mice, the administration of IFN-γ-/- CAR-T to both WT and IFN-γ-/- recipients resulted in a reduction in the numbers of endogenous CD4+ and CD8+ effectors, while exhibiting increased populations of naïve-like CD4+ T and memory CD8+ T cells. VISTA expression consistently remained elevated in resting or memory CD4+ T cells, with distinct localization from programmed cell death protein-1 (PD-1) expressing T subsets. Blocking the VISTA signal enhanced dendritic cell-induced proliferation and cytokine production by syngeneic T cells.
    CONCLUSIONS: Our findings confirm that endogenous T-cell activation and functionality are regulated by VISTA, which is associated with the therapeutic efficiency of CAR-T and provides a promising therapeutic strategy for relapse cases in CAR-T 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
    本研究旨在通过C端蛋白质组学策略,使用探索性和验证性队列,确认尿蛋白片段与胰腺导管腺癌(PDAC)的存在有关。通过胰蛋白酶肽的iTRAQ标记检查尿片段,并评估C末端片段的浓度。仅尿CD276片段显示>1.5的倍数变化(FC),健康(H)和PDAC参与者在两个探索性(H,n=42;PDAC,n=39)和验证队列(H,n=36;可切除的PDAC,n=28)。CD276片段诊断可切除PDAC的敏感性和特异性分别为75%和89%,分别,在验证队列中。与手术前相比,术后尿液中CD276片段的水平较低(n=18,P<0.01)。综合C端蛋白质组学鉴定尿CD276片段水平的增加是PDAC患者的特征。尿CD276片段是用于检测可切除的PDAC的潜在生物标志物。
    This study aimed to confirm urinary protein fragments in relation to the presence of pancreatic ductal adenocarcinoma (PDAC) via a C-terminal proteomics strategy using exploratory and validation cohorts. Urinary fragments were examined by iTRAQ-labelling of tryptic peptides and concentrations of C-terminal fragments were evaluated. Only the urinary CD276 fragment showed a fold change (FC) of > 1.5 with a significant difference of P < 0.01 between healthy (H) and PDAC participants in both the exploratory (H, n = 42; PDAC, n = 39) and validation cohorts (H, n = 36; resectable PDAC, n = 28). The sensitivity and specificity of the CD276 fragment for diagnosing resectable PDAC were 75% and 89%, respectively, in the validation cohort. Postoperative urinary levels of the CD276 fragment were low as compared to those before surgery (n = 18, P < 0.01). Comprehensive C-terminus proteomics identified an increase in the urinary CD276 fragment level as a feature of patients with PDAC. The urinary CD276 fragment is a potential biomarker for detecting resectable PDAC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号