Cytotoxicity, Immunologic

细胞毒性,免疫学
  • 文章类型: Journal Article
    自然杀伤(NK)细胞在先天免疫中起着至关重要的作用,特别是在对抗感染和肿瘤方面。然而,在血液癌症中,NK细胞通常表现出受损的功能。因此,激活其内体Toll样受体(TLRs)作为恢复其抗肿瘤活性的潜在策略非常重要。我们刺激来自急性淋巴细胞白血病患儿外周血单核细胞的NK细胞,并用特定的TLR配体刺激NK细胞(PolyI:C,咪喹莫特,R848和ODN2006),我们评估了IFN-γ的变化,CD107a,NKG2D,NKp44表达式,颗粒酶B分泌,细胞因子/趋化因子释放,和细胞毒活性。结果表明,PolyI:C和咪喹莫特增强了免疫调节和细胞毒性NK细胞的激活,增加IFN-γ,CD107a,NKG2D,和NKp44表达。R848激活免疫调节NK细胞,而ODN2006提高了CD107a,NKp44,NKG2D,和细胞毒性NK细胞中的IFN-γ分泌。R848还增加了七种细胞因子/趋化因子的分泌。重要的是,R848和ODN2006显著提高了对白血病细胞的细胞毒性。总的来说,TLR刺激增强NK细胞活化,提示TLR8(R848)和TLR9(ODN2006)配体是抗肿瘤免疫疗法的有希望的候选者。
    Natural killer (NK) cells play a crucial role in innate immunity, particularly in combating infections and tumors. However, in hematological cancers, NK cells often exhibit impaired functions. Therefore, it is very important to activate its endosomal Toll-like receptors (TLRs) as a potential strategy to restore its antitumor activity. We stimulated NK cells from the peripheral blood mononuclear cells from children with acute lymphoblastic leukemia and NK cells isolated, and the NK cells were stimulated with specific TLR ligands (Poly I:C, Imiquimod, R848, and ODN2006) and we evaluated changes in IFN-γ, CD107a, NKG2D, NKp44 expression, Granzyme B secretion, cytokine/chemokine release, and cytotoxic activity. Results revealed that Poly I:C and Imiquimod enhanced the activation of both immunoregulatory and cytotoxic NK cells, increasing IFN-γ, CD107a, NKG2D, and NKp44 expression. R848 activated immunoregulatory NK cells, while ODN2006 boosted CD107a, NKp44, NKG2D, and IFN-γ secretion in cytotoxic NK cells. R848 also increased the secretion of seven cytokines/chemokines. Importantly, R848 and ODN 2006 significantly improved cytotoxicity against leukemic cells. Overall, TLR stimulation enhances NK cell activation, suggesting TLR8 (R848) and TLR9 (ODN 2006) ligands as promising candidates for antitumor 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
    免疫疗法已显示出作为癌症治疗中的有效策略的重要前景。然而,在多形性胶质母细胞瘤(GBM)中,成人中最常见的原发性脑肿瘤,这些疗法的疗效低于最初预期.因此,迫切需要提高免疫治疗效果的策略。AURKA已被确定为GBM治疗的潜在药物靶标。对AURKA抑制后的GBM细胞转录组的分析揭示了对免疫系统的潜在影响。我们的研究表明,AURKA在体外和体内影响了各种GBM模型系统中的PD-L1水平。遗传上破坏AURKA功能导致已建立和患者来源的异种移植物GBM培养物中PD-L1水平降低和MHC-I表达增加。这个过程涉及转录和非转录途径,部分涉及GSK3β。干扰AURKA还通过减少PD-L1表达来增强NK细胞介导的GBM消除,正如在救援实验中所证明的那样。此外,使用用患者来源的细胞模拟GBM的小鼠模型,证明了Alisertib降低了活生物体中PD-L1的表达.与媒介物治疗相比,涉及抗PD-1治疗和Alisertib的联合治疗显著延长了总生存期。这些发现表明靶向AURKA可能对调节GBM细胞内的免疫环境具有治疗意义。
    Immunotherapies have shown significant promise as an impactful strategy in cancer treatment. However, in glioblastoma multiforme (GBM), the most prevalent primary brain tumor in adults, these therapies have demonstrated lower efficacy than initially anticipated. Consequently, there is an urgent need for strategies to enhance the effectiveness of immune treatments. AURKA has been identified as a potential drug target for GBM treatment. An analysis of the GBM cell transcriptome following AURKA inhibition revealed a potential influence on the immune system. Our research revealed that AURKA influenced PD-L1 levels in various GBM model systems in vitro and in vivo. Disrupting AURKA function genetically led to reduced PD-L1 levels and increased MHC-I expression in both established and patient-derived xenograft GBM cultures. This process involved both transcriptional and non-transcriptional pathways, partly implicating GSK3β. Interfering with AURKA also enhanced NK-cell-mediated elimination of GBM by reducing PD-L1 expression, as evidenced in rescue experiments. Furthermore, using a mouse model that mimics GBM with patient-derived cells demonstrated that Alisertib decreased PD-L1 expression in living organisms. Combination therapy involving anti-PD-1 treatment and Alisertib significantly prolonged overall survival compared to vehicle treatment. These findings suggest that targeting AURKA could have therapeutic implications for modulating the immune environment within GBM cells.
    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)细胞由于其自发裂解癌细胞的能力而在癌症治疗中具有希望。临床使用,大量的纯净,功能性NK细胞是必需的。将基于粘附的隔离与专门的介质相结合表明隔离方法的不可靠性,但证明了NKMACS®培养基的优越性,特别是在次优条件下。既不是人类汇集的血清,胎牛血清(FCS),人血小板裂解物,化学定义的血清替代也不能替代人AB血清。评估白细胞介素(IL-)2、IL-15、IL-21和组合的CD2/NKp46刺激。IL-21和CD2/NKp46刺激增加细胞毒性,但降低NK细胞增殖。单独的IL-15刺激实现了最高的增殖,但更实惠的IL-2表现类似。RosetteSep™人NK细胞富集试剂盒对分离有效,但是培养物中外周血单核细胞(PBMC)的存在增强了NK细胞的增殖,尽管CD16、NKp46、NKG2D的表达水平相似,和ICAM-1。与此相符,在具有人AB血清和IL-2的NKMACS®培养基中培养的纯化NK细胞表现出对原代成胶质细胞瘤干细胞的高细胞毒性。
    Natural killer (NK) cells hold promise in cancer treatment due to their ability to spontaneously lyse cancer cells. For clinical use, high quantities of pure, functional NK cells are necessary. Combining adherence-based isolation with specialized media showed the unreliability of the isolation method, but demonstrated the superiority of the NK MACS® medium, particularly in suboptimal conditions. Neither human pooled serum, fetal calf serum (FCS), human platelet lysate, nor chemically defined serum replacement could substitute human AB serum. Interleukin (IL-)2, IL-15, IL-21, and combined CD2/NKp46 stimulation were assessed. IL-21 and CD2/NKp46 stimulation increased cytotoxicity, but reduced NK cell proliferation. IL-15 stimulation alone achieved the highest proliferation, but the more affordable IL-2 performed similarly. The RosetteSep™ human NK cell enrichment kit was effective for isolation, but the presence of peripheral blood mononuclear cells (PBMCs) in the culture enhanced NK cell proliferation, despite similar expression levels of CD16, NKp46, NKG2D, and ICAM-1. In line with this, purified NK cells cultured in NK MACS® medium with human AB serum and IL-2 demonstrated high cytotoxicity against primary glioblastoma stem cells.
    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
    多发性骨髓瘤(MM)是一种浆细胞疾病,具有优先的骨髓(BM)嗜性。组织特异性趋化因子受体的强制表达已被证明可以成功地将过继转移的CARNK细胞引导到实体癌的恶性环境中。也包括BM居民AML和MM。为了重定向到BM相关的趋化因子CXCL12,我们用野生型CXCR4或功能获得突变体CXCR4R334X的异位表达对BCMACAR-NK-92以及原代NK细胞进行了装甲。我们的数据显示,配备有CXCR4的BCMCAR-NK-92和原代NK细胞在体外获得了向CXCL12迁移的改善的能力。除了协调趋化性的经典作用之外,CXCR4已被证明参与T细胞共刺激,这促使我们检查CXCR4共转导的BCMA-CARNK细胞的功能。异位CXCR4表达增强BCMACAR-NK细胞的细胞毒性能力,如通过体外消除表达BCMA的靶细胞系和原代MM细胞的能力以及通过加速的溶细胞颗粒释放所证明的。我们表明CXCR4共修饰延长了BCMACAR表面沉积,在汽车参与之后,增强了ZAP-70的招募,和加速远端信号转导动力学。BCMACAR对抗原的敏感性通过增强的ZAP-70募集到免疫突触而增强,揭示了在CXCR4过表达时CAR被触发的倾向增加。出乎意料的是,在不存在CXCL12配体刺激的情况下发生通过CXCR4的共刺激。总的来说,我们的研究结果暗示,CAR-NK细胞与组织相关趋化因子受体的共修饰对过继性NK细胞治疗的影响超出了在肿瘤部位内的转运和滞留改善.
    Multiple myeloma (MM) is a plasma cell disease with a preferential bone marrow (BM) tropism. Enforced expression of tissue-specific chemokine receptors has been shown to successfully guide adoptively-transferred CAR NK cells towards the malignant milieu in solid cancers, but also to BM-resident AML and MM. For redirection towards BM-associated chemokine CXCL12, we armored BCMA CAR-NK-92 as well as primary NK cells with ectopic expression of either wildtype CXCR4 or a gain-of-function mutant CXCR4R334X. Our data showed that BCMA CAR-NK-92 and -primary NK cells equipped with CXCR4 gained an improved ability to migrate towards CXCL12 in vitro. Beyond its classical role coordinating chemotaxis, CXCR4 has been shown to participate in T cell co-stimulation, which prompted us to examine the functionality of CXCR4-cotransduced BCMA-CAR NK cells. Ectopic CXCR4 expression enhanced the cytotoxic capacity of BCMA CAR-NK cells, as evidenced by the ability to eliminate BCMA-expressing target cell lines and primary MM cells in vitro and through accelerated cytolytic granule release. We show that CXCR4 co-modification prolonged BCMA CAR surface deposition, augmented ZAP-70 recruitment following CAR-engagement, and accelerated distal signal transduction kinetics. BCMA CAR sensitivity towards antigen was enhanced by virtue of an enhanced ZAP-70 recruitment to the immunological synapse, revealing an increased propensity of CARs to become triggered upon CXCR4 overexpression. Unexpectedly, co-stimulation via CXCR4 occurred in the absence of CXCL12 ligand-stimulation. Collectively, our findings imply that co-modification of CAR-NK cells with tissue-relevant chemokine receptors affect adoptive NK cell therapy beyond improved trafficking and retention within tumor sites.
    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
    对于先进的治疗药物,需要开发和验证效力测定,根据国际准则,表征产品并获得可靠和一致的数据。我们的目的是验证用于评估自体抗CD19嵌合抗原受体(CAR)T效力的杀伤测定。我们使用CD4+和CD8+淋巴细胞或抗CD19CAR-T细胞作为效应细胞,使用REH(CD19+)或MOLM-13(CD19-)细胞系作为靶细胞。在共培养靶细胞和效应细胞(1:1比例)24小时后,样品用7-AAD标记,通过流式细胞术评估抗CD3和抗CD19抗体以及CD19+死细胞的频率。为了验证CAR-T对CD19+靶标的特异性,安排了CAR-T和REH或MOLM-13以不同的效应器-靶标比进行共培养.此外,测试未转导的CD4+和CD8+淋巴细胞与来自同一供体的CAR-T进行比较,以证明测定特异性.线性和准确性进行了评估,并为这两个参数编制了既定的验收标准(线性为r2≥0.97,准确性为平均相对误差≤10%)。此外,在共培养23到25小时之间进行时,该方法被认为是稳健的,和内部测定,获得了测定间和日间精确度。最后,为了验证分析师之间的精确度,该检验由3个不同的操作者执行,两种情况下类内相关系数均>0.4.总之,我们认为该CAR-T效价测定在产品开发和质量控制的所有步骤中都是有效的.
    For advanced therapy medicinal products, the development and validation of potency assays are required, in accordance with international guidelines, to characterise the product and obtain reliable and consistent data. Our purpose was to validate the killing assay for the evaluation of autologous anti-CD19 chimeric antigen receptor (CAR) T potency. We used CD4 + and CD8 + lymphocytes or anti-CD19 CAR-T cells as effector cells and REH (CD19 +) or MOLM-13 (CD19 -) cell lines as target cells. After co-culturing target and effector cells (1:1 ratio) for 24 h, samples were labelled with 7-AAD, anti-CD3 and anti-CD19 antibodies and the frequency of CD19 + dead cells was evaluated by flow cytometry. In order to verify the CAR-T specificity for the CD19 + target, the co-culture between CAR-T and REH or MOLM-13 at different effector-to-target ratios was scheduled. Moreover, not transduced CD4 + and CD8 + lymphocytes were tested in comparison with CAR-T from the same donor to demonstrate the assay specificity. Linearity and accuracy were evaluated, and established acceptance criteria were compiled for both parameters (r2 ≥ 0.97 for linearity and average relative error ≤ 10% for accuracy). Furthermore, the method was considered robust when performed between 23 and 25 h of co-culture, and the intra-assay, inter-assay and inter-day precision was obtained. Finally, in order to verify the inter-analyst precision, the test was executed by three different operators and the intra-class correlation coefficient was > 0.4 in both cases. In conclusion, we consider this CAR-T potency assay as validated and usable in all steps of product development and quality control.
    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
    免疫细胞具有强烈的身体生活方式,其特征是结构可塑性和力量发挥。为了调查特定的免疫功能是否需要刻板的机械输出,我们使用超分辨率牵引力显微镜比较了细胞毒性T细胞形成的免疫突触与其他T细胞亚群和巨噬细胞形成的接触。T细胞突触是全局压缩的,这与与巨噬细胞吞噬相关的拉动和挤压有着根本的不同。每种细胞类型的力施加模式的光谱分解将细胞毒性与抗压强度联系起来,局部突出,和复杂的归纳,不对称地形。这些特征通过细胞骨架调节因子的遗传破坏被验证为细胞毒性驱动因子,突触分泌的实时成像,和界面变形的计算机模拟分析。突触结构和力的施加对目标刚度和尺寸敏感,这表明杀死的机械增强作用是生物物理适应性的。我们得出结论,细胞毒性和,言下之意,其他效应器响应由传出力的专门模式支持。
    Immune cells have intensely physical lifestyles characterized by structural plasticity and force exertion. To investigate whether specific immune functions require stereotyped mechanical outputs, we used super-resolution traction force microscopy to compare the immune synapses formed by cytotoxic T cells with contacts formed by other T cell subsets and by macrophages. T cell synapses were globally compressive, which was fundamentally different from the pulling and pinching associated with macrophage phagocytosis. Spectral decomposition of force exertion patterns from each cell type linked cytotoxicity to compressive strength, local protrusiveness, and the induction of complex, asymmetric topography. These features were validated as cytotoxic drivers by genetic disruption of cytoskeletal regulators, live imaging of synaptic secretion, and in silico analysis of interfacial distortion. Synapse architecture and force exertion were sensitive to target stiffness and size, suggesting that the mechanical potentiation of killing is biophysically adaptive. We conclude that cellular cytotoxicity and, by implication, other effector responses are supported by specialized patterns of efferent force.
    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
    存在两种已知的自然杀伤(NK)细胞识别和杀死患病靶标的机制:(i)直接杀伤和(ii)抗体依赖性细胞介导的细胞毒性(ADCC)。我们研究了用于增强人类NK细胞杀伤功能的间接NK细胞活化策略。我们通过利用人类外周血单核细胞(PBMC)池内的toll样受体9(TLR9)激动作用导致导致NK细胞活化的强大干扰素信号级联这一事实来做到这一点。TLR9激动剂刺激后,富集NK细胞并将其掺入测定中以评估其杀死肿瘤细胞系靶标的能力。值得注意的是,观察到TLR9激动作用的不同影响-直接杀伤作用增强,而ADCC没有增加.为了确保观察到的差异效应不归因于人类捐赠者之间的差异,我们使用我们的自然杀伤-同时ADCC和直接杀伤试验(NK-SADKA)控制人与人之间的差异来概括观察结果。接下来,我们观察到治疗诱导的NK细胞表面CD16减少-已知由NK细胞活化后脱落。鉴于CD16在ADCC中的重要作用,这种脱落可以解释所观察到的TLR9激动作用对NK细胞介导的杀伤能力的不同影响。
    There are two known mechanisms by which natural killer (NK) cells recognize and kill diseased targets: (i) direct killing and (ii) antibody-dependent cell-mediated cytotoxicity (ADCC). We investigated an indirect NK cell activation strategy for the enhancement of human NK cell killing function. We did this by leveraging the fact that toll-like receptor 9 (TLR9) agonism within pools of human peripheral blood mononuclear cells (PBMCs) results in a robust interferon signaling cascade that leads to NK cell activation. After TLR9 agonist stimulation, NK cells were enriched and incorporated into assays to assess their ability to kill tumor cell line targets. Notably, differential impacts of TLR9 agonism were observed-direct killing was enhanced while ADCC was not increased. To ensure that the observed differential effects were not attributable to differences between human donors, we recapitulated the observation using our Natural Killer-Simultaneous ADCC and Direct Killing Assay (NK-SADKA) that controls for human-to-human differences. Next, we observed a treatment-induced decrease in NK cell surface CD16-known to be shed by NK cells post-activation. Given the essential role of CD16 in ADCC, such shedding could account for the observed differential impact of TLR9 agonism on NK cell-mediated killing capacity.
    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
    增强肿瘤中的免疫细胞功能仍然是癌症免疫治疗的主要挑战。自然杀伤细胞(NK)是主要的先天效应细胞,对肿瘤具有广泛的细胞毒性。因此,NK细胞是癌症免疫治疗的理想候选者。包括胶质母细胞瘤(GBM)。缺氧是实体瘤的共同特征,肿瘤细胞和正常细胞通过上调转录因子缺氧诱导因子(HIF)-1α适应肿瘤微环境,这可能不利于抗肿瘤效应免疫细胞的功能,包括NK细胞。我们使用成簇的规则间隔短回文重复序列(CRISPR)相关蛋白9(Cas9)在人原代NK细胞中敲除HIF-1α。然后,在常氧和低氧条件下进行细胞表征。电穿孔两种HIF-1α靶向指导RNA-Cas9蛋白复合物抑制扩增NK细胞中的HIF-1α表达。HIF-1α敲除人NK细胞,包括处于缺氧状态的人群,增强了同种异体GBM细胞的生长抑制作用,并诱导了GBM细胞衍生球体的凋亡。RNA测序显示,HIF-1α敲除NK细胞的细胞毒性可能与穿孔素和TNF表达增加有关。结果表明HIF-1α敲除人NK细胞,包括人口,在模拟GBM缺氧条件的环境中增强的细胞毒性。CRISPR-Cas9介导的HIF-1α基因敲除NK细胞,包括人口,可能是GBM患者的一种有前途的免疫治疗替代方案。
    Enhancing immune cell functions in tumors remains a major challenge in cancer immunotherapy. Natural killer cells (NK) are major innate effector cells with broad cytotoxicity against tumors. Accordingly, NK cells are ideal candidates for cancer immunotherapy, including glioblastoma (GBM). Hypoxia is a common feature of solid tumors, and tumor cells and normal cells adapt to the tumor microenvironment by upregulating the transcription factor hypoxia-inducible factor (HIF)-1α, which can be detrimental to anti-tumor effector immune cell function, including that of NK cells. We knocked out HIF-1α in human primary NK cells using clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (Cas9). Then, cellular characterizations were conducted in normoxic and hypoxic conditions. Electroporating two HIF-1α-targeting guide RNA-Cas9 protein complexes inhibited HIF-1α expression in expanded NK cells. HIF-1α knockout human NK cells, including populations in hypoxic conditions, enhanced the growth inhibition of allogeneic GBM cells and induced apoptosis in GBM-cell-derived spheroids. RNA-sequencing revealed that the cytotoxicity of HIF-1α knockout NK cells could be related to increased perforin and TNF expression. The results demonstrated that HIF-1α knockout human NK cells, including populations, enhanced cytotoxicity in an environment mimicking the hypoxic conditions of GBM. CRISPR-Cas9-mediated HIF-1α knockout NK cells, including populations, could be a promising immunotherapeutic alternative in patients with GBM.
    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T细胞在肿瘤免疫中起着核心作用,试图利用它们的细胞毒性能力作为一种治疗方法,效果有限,部分是由于抑制微环境限制了它们的迁移和激活。相比之下,骨髓细胞大量浸润肿瘤,并且很好地适应了这些苛刻的条件。虽然它们具备杀伤细胞的能力,它们通常在迁移到肿瘤后采用免疫抑制表型。因此,如何修改他们针对癌症的激活程序,以及在骨髓细胞中应该激活哪些信号级联以引起其细胞毒性仍不清楚。这里,我们发现在小鼠骨髓细胞中IgM诱导的信号激活导致溶解颗粒的分泌和大量肿瘤细胞死亡。这些发现为设计新型免疫疗法开辟了场所,通过为单核细胞配备靶向肿瘤抗原的嵌合受体,信号通过IgM受体。尽管如此,我们发现,由于ER应激反应的诱导,骨髓细胞不表达用于识别肿瘤抗原的抗体衍生部分.为了克服这个限制,我们设计了基于IgG高亲和力FcγRI的嵌合受体。表达这些受体的巨噬细胞与肿瘤结合IgG一起孵育诱导大量肿瘤细胞杀伤和分泌活性氧和颗粒酶B。这项工作突出了在基因重编程髓样细胞信号中涉及的挑战,并为赋予髓样细胞抗原特异性细胞毒性提供了框架.
    Despite the central role of T cells in tumor immunity, attempts to harness their cytotoxic capacity as a therapy have met limited efficacy, partially as a result of the suppressive microenvironment which limits their migration and activation. In contrast, myeloid cells massively infiltrate tumors and are well adapted to survive these harsh conditions. While they are equipped with cell-killing abilities, they often adopt an immunosuppressive phenotype upon migration to tumors. Therefore, the questions of how to modify their activation programming against cancer, and what signaling cascades should be activated in myeloid cells to elicit their cytotoxicity have remained unclear. Here, we found that activation of IgM-induced signaling in murine myeloid cells results in secretion of lytic granules and massive tumor cell death. These findings open venues for designing novel immunotherapy by equipping monocytes with chimeric receptors that target tumor antigens and consequently, signal through IgM receptor. Nonetheless, we found that myeloid cells do not express the antibody-derived portion used to recognize the tumor antigen due to the induction of an ER stress response. To overcome this limitation, we designed chimeric receptors that are based on the high-affinity FcγRI for IgG. Incubation of macrophages expressing these receptors along with tumor-binding IgG induced massive tumor cell killing and secretion of reactive oxygen species and Granzyme B. Overall, this work highlights the challenges involved in genetically reprogramming the signaling in myeloid cells and provides a framework for endowing myeloid cells with antigen-specific cytotoxicity.
    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:过继性癌症免疫治疗,使用工程化的T细胞,表达嵌合抗原受体或自体肿瘤浸润淋巴细胞,近年来,不同类型癌症的主要治疗方法。然而,尽管过继性癌症免疫疗法具有转化潜力,这个领域仍然面临重大挑战,表现为效应CD8+T细胞的细胞毒性能力在其扩增后明显下降。为了应对这些挑战,我们开发了一种离体“合成免疫生态位”(SIN),由固定的CCL21和ICAM1组成,它们协同诱导抗原特异性CD8+T细胞的有效扩增,甚至增强它们的细胞毒性。
    方法:为了探索基于CCL21+ICAM1的SIN调节抗原激活和CD3/CD28激活的效应CD8+T细胞的增殖和细胞毒性之间的相互作用的分子机制,我们通过流式细胞术对特异性分化标志物进行了综合分析,以及基因表达谱分析。
    结果:在第3天,对于树突细胞(DC)/卵清蛋白(OVA)活化的和抗CD3/CD28活化的细胞,由SIN诱导的转录组效应在很大程度上相似。细胞增殖增加,细胞表现出高杀伤能力。在第四天和以后,增殖/细胞毒性表型完全变成“激活特异性”;DC/OVA激活的细胞失去其细胞毒活性,which,反过来,被SIN治疗救出。在更长的孵化时间里,细胞毒活性进一步下降,在第7天,不能被罪拯救。用抗CD3/CD28珠活化后的SIN刺激诱导增殖表型的主要增加,同时瞬时抑制其细胞毒性2-3天,并在第7天完全恢复其杀伤活性。确定了SIN效应的潜在分子调控途径,基于转录组学和多光谱成像分析。
    结论:这些数据表明细胞增殖和细胞毒性呈负相关,它们之间的相互作用受到初始激活模式的不同调节。SIN刺激大大增强了细胞的扩增,在两种激活模式下,在刺激后的特定时间点显示出高存活率和细胞毒性,这表明它可以有效地加强过继性癌症免疫疗法。
    BACKGROUND: Adoptive cancer immunotherapy, using engineered T-cells, expressing chimeric antigen receptor or autologous tumor infiltrating lymphocytes became, in recent years, a major therapeutic approach for diverse types of cancer. However, despite the transformative potential of adoptive cancer immunotherapy, this field still faces major challenges, manifested by the apparent decline of the cytotoxic capacity of effector CD8+ T cells upon their expansion. To address these challenges, we have developed an ex vivo \"synthetic immune niche\" (SIN), composed of immobilized CCL21 and ICAM1, which synergistically induce an efficient expansion of antigen-specific CD8+ T cells while retaining, and even enhancing their cytotoxic potency.
    METHODS: To explore the molecular mechanisms through which a CCL21+ICAM1-based SIN modulates the interplay between the proliferation and cytotoxic potency of antigen-activated and CD3/CD28-activated effector CD8+ T cells, we performed integrated analysis of specific differentiation markers via flow cytometry, together with gene expression profiling.
    RESULTS: On day 3, the transcriptomic effect induced by the SIN was largely similar for both dendritic cell (DC)/ovalbumin (OVA)-activated and anti-CD3/CD28-activated cells. Cell proliferation increased and the cells exhibited high killing capacity. On day 4 and on, the proliferation/cytotoxicity phenotypes became radically \"activation-specific\"; The DC/OVA-activated cells lost their cytotoxic activity, which, in turn, was rescued by the SIN treatment. On longer incubation, the cytotoxic activity further declined, and on day7, could not be rescued by the SIN. SIN stimulation following activation with anti-CD3/CD28 beads induced a major increase in the proliferative phenotype while transiently suppressing their cytotoxicity for 2-3 days and fully regaining their killing activity on day 7. Potential molecular regulatory pathways of the SIN effects were identified, based on transcriptomic and multispectral imaging profiling.
    CONCLUSIONS: These data indicate that cell proliferation and cytotoxicity are negatively correlated, and the interplay between them is differentially regulated by the mode of initial activation. The SIN stimulation greatly enhances the cell expansion, following both activation modes, while displaying high survival and cytotoxic potency at specific time points following stimulation, suggesting that it could effectively reinforce adoptive cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号