Immune Evation

免疫反应
  • 文章类型: Journal Article
    背景:EB病毒(EBV)是一种双链DNA致癌病毒。几种类型的实体瘤,如鼻咽癌,EBV相关性胃癌,和淋巴上皮瘤样肺癌,与EBV感染有关.目前,几种用于EBV相关肿瘤的TCR-T细胞疗法正在临床试验中,但是由于实体瘤的免疫抑制免疫微环境,TCR-T细胞治疗EBV相关实体瘤的临床应用有限.阐明EBV参与肿瘤免疫抑制微环境形成的机制将有助于T细胞或TCR-T细胞突破限制并发挥更强的抗肿瘤潜力。
    方法:流式细胞术用于分析EBV感染和未感染EBV的肿瘤诱导的巨噬细胞分化表型,以及与这些巨噬细胞共培养的T细胞的功能。小鼠异种移植模型用于探讨M2巨噬细胞的作用,TCR-T细胞,和基质金属蛋白9(MMP9)抑制剂对EBV感染的肿瘤生长的影响。
    结果:EBV阳性肿瘤表现出T细胞耗竭特征,尽管存在大量T细胞浸润。EBV感染的肿瘤招募了大量带有CCL5的单核巨噬细胞,并通过分泌CSF1和促进单核巨噬细胞产生自分泌IL10来诱导CD163M2巨噬细胞极化。EBV感染诱导该组CD163+M2巨噬细胞大量分泌MMP9是导致EBV阳性肿瘤T细胞耗竭和TCR-T细胞治疗抵抗的重要因素,MMP9抑制剂的使用改善了与M2巨噬细胞共培养的T细胞的功能。最后,MMP9抑制剂与靶向EBV阳性肿瘤的TCR-T细胞的组合显著抑制小鼠异种移植物的生长。
    结论:MMP9抑制剂可改善EBV诱导的M2巨噬细胞抑制的TCR-T细胞功能。TCR-T细胞疗法联合MMP9抑制剂是EBV阳性实体瘤的有效治疗策略。
    BACKGROUND: Epstein-Barr virus (EBV) is a double-stranded DNA oncogenic virus. Several types of solid tumors, such as nasopharyngeal carcinoma, EBV-associated gastric carcinoma, and lymphoepithelioma-like carcinoma of the lung, have been linked to EBV infection. Currently, several TCR-T-cell therapies for EBV-associated tumors are in clinical trials, but due to the suppressive immune microenvironment of solid tumors, the clinical application of TCR-T-cell therapy for EBV-associated solid tumors is limited. Figuring out the mechanism by which EBV participates in the formation of the tumor immunosuppressive microenvironment will help T cells or TCR-T cells break through the limitation and exert stronger antitumor potential.
    METHODS: Flow cytometry was used for analyzing macrophage differentiation phenotypes induced by EBV-infected and EBV-uninfected tumors, as well as the function of T cells co-cultured with these macrophages. Xenograft model in mice was used to explore the effects of M2 macrophages, TCR-T cells, and matrix metalloprotein 9 (MMP9) inhibitors on the growth of EBV-infected tumors.
    RESULTS: EBV-positive tumors exhibited an exhaustion profile of T cells, despite the presence of a large T-cell infiltration. EBV-infected tumors recruited a large number of mononuclear macrophages with CCL5 and induced CD163+M2 macrophages polarization through the secretion of CSF1 and the promotion of autocrine IL10 production by mononuclear macrophages. Massive secretion of MMP9 by this group of CD163+M2 macrophages induced by EBV infection was an important factor contributing to T-cell exhaustion and TCR-T-cell therapy resistance in EBV-positive tumors, and the use of MMP9 inhibitors improved the function of T cells cocultured with M2 macrophages. Finally, the combination of an MMP9 inhibitor with TCR-T cells targeting EBV-positive tumors significantly inhibited the growth of xenografts in mice.
    CONCLUSIONS: MMP9 inhibitors improve TCR-T cell function suppressed by EBV-induced M2 macrophages. TCR-T-cell therapy combined with MMP9 inhibitors was an effective therapeutic strategy for EBV-positive 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
    背景:虽然程序性细胞死亡蛋白1(PD-1)/程序性细胞死亡配体1(PD-L1)阻断是一种有效的抗肿瘤治疗策略,它仅对有限的癌症患者有效,强调需要确定额外的免疫检查点。据报道,嗜丁基酶1A1(BTN1A1)具有潜在的免疫调节活性,但是它作为免疫检查点的功能仍有待系统评估,而这种活动的潜在机制尚未得到表征。
    方法:在原发肿瘤组织样本中评估BTN1A1的表达,并检查了其抑制T细胞活化和T细胞依赖性肿瘤清除的能力。BTN1A1和PD-L1表达之间的关系进一步表征,随后开发了一种BTN1A1特异性抗体,该抗体施用于荷瘤小鼠,以测试该靶标对免疫检查点抑制的适应性。
    结果:证实BTN1A1在体外和体内抑制T细胞活化。在一系列实体瘤组织样品中检测到稳健的BTN1A1表达,BTN1A1的表达与PD-L1的表达是相互排斥的,因为它抑制了Janus激活的激酶/信号转导子和转录信号激活因子诱导的PD-L1上调。抗体介导的BTN1A1阻断抑制了同基因荷瘤小鼠的肿瘤生长并增强了免疫细胞浸润。
    结论:一起,这些结果证实,BTN1A1的潜力是一个真正的免疫检查点和可行的免疫治疗靶标,用于治疗抗PD-1/PD-L1难治性或耐药性疾病的个体,为改善一系列癌症患者的生存结果开辟了新的途径。
    BACKGROUND: While Programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) blockade is a potent antitumor treatment strategy, it is effective in only limited subsets of patients with cancer, emphasizing the need for the identification of additional immune checkpoints. Butyrophilin 1A1 (BTN1A1) has been reported to exhibit potential immunoregulatory activity, but its ability to function as an immune checkpoint remains to be systematically assessed, and the mechanisms underlying such activity have yet to be characterized.
    METHODS: BTN1A1 expression was evaluated in primary tumor tissue samples, and its ability to suppress T-cell activation and T cell-dependent tumor clearance was examined. The relationship between BTN1A1 and PD-L1 expression was further characterized, followed by the development of a BTN1A1-specific antibody that was administered to tumor-bearing mice to test the amenability of this target to immune checkpoint inhibition.
    RESULTS: BTN1A1 was confirmed to suppress T-cell activation in vitro and in vivo. Robust BTN1A1 expression was detected in a range of solid tumor tissue samples, and BTN1A1 expression was mutually exclusive with that of PD-L1 as a consequence of its inhibition of Janus-activated kinase/signal transducer and activator of transcription signaling-induced PD-L1 upregulation. Antibody-mediated BTN1A1 blockade suppressed tumor growth and enhanced immune cell infiltration in syngeneic tumor-bearing mice.
    CONCLUSIONS: Together, these results confirm that the potential of BTN1A1 is a bona fide immune checkpoint and a viable immunotherapeutic target for the treatment of individuals with anti-PD-1/PD-L1 refractory or resistant disease, opening new avenues to improving survival outcomes for patients with a range of 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
    背景:恩扎鲁胺,下一代抗雄激素药物,已被批准用于治疗转移性去势抵抗性前列腺癌(CRPC)。虽然恩扎鲁他胺已被证明可以改善CRPC患者的进展时间并延长总体生存率,大多数患者最终会对治疗产生抗药性。免疫治疗方法在该患者群体中显示出有限的临床益处;了解抗性机制可以帮助开发针对CRPC的新颖且更有效的治疗方法。肿瘤对各种疗法的耐药机制之一是肿瘤表型可塑性,癌细胞获得间质特征,无论是否丧失经典的上皮特征。这项工作研究了恩杂鲁胺耐药性之间的潜在联系,肿瘤表型可塑性,以及对前列腺癌中免疫介导的裂解的抗性。
    方法:通过将人前列腺癌细胞系长期暴露于培养的药物,建立了对恩杂鲁胺耐药的前列腺癌模型。在体外和体内评估肿瘤细胞的表型特征,以及对免疫效应细胞介导的细胞毒性的敏感性。
    结果:恩杂鲁胺耐药与间充质肿瘤特征的增加有关,雌激素受体表达上调,并显著降低肿瘤对自然杀伤(NK)介导的裂解的易感性,与恩杂鲁胺耐药细胞的肿瘤/NK细胞缀合物形成减少相关的效应。Fulvestrant,一种选择性雌激素受体降解剂,在体外恢复靶/NK细胞缀合物的形成并增加对NK细胞裂解的易感性。在体内,氟维司群对恩杂鲁胺耐药细胞具有抗肿瘤活性,与NK细胞活化相关的效应。
    结论:NK细胞正在成为前列腺癌的一种有希望的治疗方法。通过氟维司群阻断雌激素受体来改变肿瘤的可塑性可能为通过基于NK细胞的方法在耐恩杂鲁胺的CRPC中进行免疫干预提供了机会。
    Enzalutamide, a next-generation antiandrogen agent, is approved for the treatment of metastatic castration-resistant prostate cancer (CRPC). While enzalutamide has been shown to improve time to progression and extend overall survival in men with CRPC, the majority of patients ultimately develop resistance to treatment. Immunotherapy approaches have shown limited clinical benefit in this patient population; understanding resistance mechanisms could help develop novel and more effective treatments for CRPC. One of the mechanisms involved in tumor resistance to various therapeutics is tumor phenotypic plasticity, whereby carcinoma cells acquire mesenchymal features with or without the loss of classical epithelial characteristics. This work investigated a potential link between enzalutamide resistance, tumor phenotypic plasticity, and resistance to immune-mediated lysis in prostate cancer.
    Models of prostate cancer resistant to enzalutamide were established by long-term exposure of human prostate cancer cell lines to the drug in culture. Tumor cells were evaluated for phenotypic features in vitro and in vivo, as well as for sensitivity to immune effector cell-mediated cytotoxicity.
    Resistance to enzalutamide was associated with gain of mesenchymal tumor features, upregulation of estrogen receptor expression, and significantly reduced tumor susceptibility to natural killer (NK)-mediated lysis, an effect that was associated with decreased tumor/NK cell conjugate formation with enzalutamide-resistant cells. Fulvestrant, a selective estrogen receptor degrader, restored the formation of target/NK cell conjugates and increased susceptibility to NK cell lysis in vitro. In vivo, fulvestrant demonstrated antitumor activity against enzalutamide-resistant cells, an effect that was associated with activation of NK cells.
    NK cells are emerging as a promising therapeutic approach in prostate cancer. Modifying tumor plasticity via blockade of estrogen receptor with fulvestrant may offer an opportunity for immune intervention via NK cell-based approaches in enzalutamide-resistant CRPC.
    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
    抗癌免疫疗法,如免疫检查点抑制剂,双特异性抗体,和嵌合抗原受体T细胞,改善了各种恶性肿瘤患者的预后。然而,由于肿瘤微环境的原发性或适应性/获得性免疫抵抗机制,大多数患者最初没有反应或没有表现出持久的反应。这些抑制程序是无数的,表面上相同癌症类型的患者不同,并且可以利用多种细胞类型来增强其稳定性。因此,单一疗法的总体获益仍然有限.尖端技术现在允许广泛的肿瘤分析,可用于定义肿瘤细胞的原发性和/或获得性免疫抗性的内在和外在途径,本文中称为对当前疗法的免疫抗性的特征或特征集。我们建议癌症可以通过免疫抗性原型来表征,由包含已知免疫抵抗机制的五个特征集组成。抗性的原型可以提供新的治疗策略,同时解决多个细胞轴和/或抑制机制。因此,临床医生可能能够优先考虑针对个体患者的靶向治疗组合,以改善总体疗效和结局.
    Anticancer immunotherapies, such as immune checkpoint inhibitors, bispecific antibodies, and chimeric antigen receptor T cells, have improved outcomes for patients with a variety of malignancies. However, most patients either do not initially respond or do not exhibit durable responses due to primary or adaptive/acquired immune resistance mechanisms of the tumor microenvironment. These suppressive programs are myriad, different between patients with ostensibly the same cancer type, and can harness multiple cell types to reinforce their stability. Consequently, the overall benefit of monotherapies remains limited. Cutting-edge technologies now allow for extensive tumor profiling, which can be used to define tumor cell intrinsic and extrinsic pathways of primary and/or acquired immune resistance, herein referred to as features or feature sets of immune resistance to current therapies. We propose that cancers can be characterized by immune resistance archetypes, comprised of five feature sets encompassing known immune resistance mechanisms. Archetypes of resistance may inform new therapeutic strategies that concurrently address multiple cell axes and/or suppressive mechanisms, and clinicians may consequently be able to prioritize targeted therapy combinations for individual patients to improve overall efficacy and outcomes.
    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:I型干扰素(IFN-Is),由造血细胞分泌,驱动实体瘤的免疫监视。然而,在造血系统恶性肿瘤包括B细胞急性淋巴细胞白血病(B-ALL)中,IFN-I驱动的免疫应答的抑制机制尚不清楚.
    方法:使用高维细胞计数,我们描述了高级别原发性人类和小鼠B-ALL中IFN-I产生和IFN-I驱动的免疫应答的缺陷。我们开发了自然杀伤(NK)细胞作为治疗来对抗B-ALL中IFN-I产生的内在抑制。
    结果:我们发现IFN-I信号基因的高表达预示着B-ALL患者的良好临床结局,强调IFN-I途径在这种恶性肿瘤中的重要性。我们表明,人和小鼠B-ALL微环境在旁分泌(浆细胞样树突状细胞)和/或自分泌(B细胞)IFN-I产生和IFN-I驱动的免疫反应中存在内在缺陷。减少的IFN-I产生足以抑制免疫系统并促进易于MYC驱动的B-ALL的小鼠中的白血病发展。在抗白血病免疫亚群中,抑制IFN-I的产生最显着降低IL-15的转录并减少B-ALL微环境中的NK细胞数量和效应物成熟。健康NK细胞的过继转移可显着延长带有ALL的转基因小鼠的存活。对B-ALL易感小鼠施用IFN-Is减少白血病进展并增加循环中总NK和NK细胞效应物的频率。用IFN-Is对原代小鼠B-ALL微环境中的恶性和非恶性免疫细胞的离体治疗完全恢复了近端IFN-I信号传导并部分恢复了IL-15的产生。在B-ALL患者中,IL-15的抑制在MYC过表达的难以治疗的亚型中是最严重的。MYC过表达促进B-ALL对NK细胞介导的杀伤的敏感性。为了对抗MYCHH人B-ALL中IFN-I诱导的IL-15产生的抑制,我们CRISPRa改造了一种分泌IL-15的新型人类NK细胞系。分泌CRISPRaIL-15的人NK细胞比不产生IL-15的NK细胞更有效地在体外杀死高级人B-ALL并在体内阻断白血病进展。
    结论:我们发现,在B-ALL中恢复固有抑制的IFN-I产生是产生IL-15的NK细胞的治疗功效的基础,并且这种NK细胞代表了在高级B-ALL中对MYC进行药物治疗的有吸引力的治疗解决方案。
    Type I interferons (IFN-Is), secreted by hematopoietic cells, drive immune surveillance of solid tumors. However, the mechanisms of suppression of IFN-I-driven immune responses in hematopoietic malignancies including B-cell acute lymphoblastic leukemia (B-ALL) are unknown.
    Using high-dimensional cytometry, we delineate the defects in IFN-I production and IFN-I-driven immune responses in high-grade primary human and mouse B-ALLs. We develop natural killer (NK) cells as therapies to counter the intrinsic suppression of IFN-I production in B-ALL.
    We find that high expression of IFN-I signaling genes predicts favorable clinical outcome in patients with B-ALL, underscoring the importance of the IFN-I pathway in this malignancy. We show that human and mouse B-ALL microenvironments harbor an intrinsic defect in paracrine (plasmacytoid dendritic cell) and/or autocrine (B-cell) IFN-I production and IFN-I-driven immune responses. Reduced IFN-I production is sufficient for suppressing the immune system and promoting leukemia development in mice prone to MYC-driven B-ALL. Among anti-leukemia immune subsets, suppression of IFN-I production most markedly lowers the transcription of IL-15 and reduces NK-cell number and effector maturation in B-ALL microenvironments. Adoptive transfer of healthy NK cells significantly prolongs survival of overt ALL-bearing transgenic mice. Administration of IFN-Is to B-ALL-prone mice reduces leukemia progression and increases the frequencies of total NK and NK-cell effectors in circulation. Ex vivo treatment of malignant and non-malignant immune cells in primary mouse B-ALL microenvironments with IFN-Is fully restores proximal IFN-I signaling and partially restores IL-15 production. In B-ALL patients, the suppression of IL-15 is the most severe in difficult-to-treat subtypes with MYC overexpression. MYC overexpression promotes sensitivity of B-ALL to NK cell-mediated killing. To counter the suppressed IFN-I-induced IL-15 production in MYChigh human B-ALL, we CRISPRa-engineered a novel human NK-cell line that secretes IL-15. CRISPRa IL-15-secreting human NK cells kill high-grade human B-ALL in vitro and block leukemia progression in vivo more effectively than NK cells that do not produce IL-15.
    We find that restoration of the intrinsically suppressed IFN-I production in B-ALL underlies the therapeutic efficacy of IL-15-producing NK cells and that such NK cells represent an attractive therapeutic solution for the problem of drugging MYC in high-grade B-ALL.
    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:新数据表明,程式化死亡配体1(PD-L1)表达增加的恩杂鲁胺治疗的前列腺癌患者可能受益于抗PD-L1治疗。不幸的是,IMbassor250III期临床试验显示,阿特珠单抗(一种PD-L1抑制剂)和恩杂鲁胺联合应用未能延长去势抵抗性前列腺癌(CRPC)患者的总生存期.然而,治疗失败的潜在机制仍然未知.
    方法:将人CRPCC4-2B细胞和鼠Myc-CaP细胞长期暴露于浓度增加的恩杂鲁胺中,对恩杂鲁胺有抗性的细胞称为C4-2BMDVR和Myc-CaPMDVR,分别。使用RNA测序分析确定了耐药前列腺癌细胞的作用机制,RNA干扰,实时PCR,西方印迹,和共同培养技术。Myc-CaP和Myc-CaPMDVR肿瘤在同基因FVB小鼠中建立,和肿瘤浸润的白细胞在恩杂鲁胺治疗后分离。通过流式细胞术测定染色的免疫细胞。并使用FlowJo对数据进行分析。
    结果:免疫相关信号通路(干扰素α/γ反应,炎症反应,和细胞趋化性)在人抗恩杂鲁胺前列腺癌细胞中受到抑制。在耐药细胞和CRPC患者队列中,PD-L1过表达并受到雄激素受体信号传导的负调控。恩扎鲁胺治疗减少了CD8+T细胞数量,但增加了小鼠Myc-CaP肿瘤中的单核细胞髓源性抑制细胞(M-MDSC)群体和PD-L1表达。同样,趋化性和免疫反应调节信号通路被抑制,使用耐恩杂鲁胺的Myc-CaPMDVR细胞,PD-L1表达也增加。值得注意的是,与Myc-CaP亲本肿瘤相比,Myc-CaPMDVR原位肿瘤中的MDSC群体显着增加。骨髓细胞与Myc-CaPMDVR细胞共培养可显着促进MDSC分化并向M2巨噬细胞偏斜转变。
    结论:我们的研究表明,恩杂鲁胺耐药前列腺癌细胞可以直接促进免疫抑制信号传导,并且可能是免疫检查点抑制剂在恩杂鲁胺耐药前列腺癌中的疗效降低的潜在手段。
    Emerging data suggest that patients with enzalutamide-treated prostate cancer with increased programmed death-ligand 1 (PD-L1) expression may benefit from anti-PD-L1 treatment. Unfortunately, the Phase III IMbassador250 clinical trial revealed that the combination of atezolizumab (a PD-L1 inhibitor) and enzalutamide failed to extend overall survival in patients with castration-resistant prostate cancer (CRPC). However, the mechanisms underlying treatment failure remain unknown.
    Human CRPC C4-2B cells and murine Myc-CaP cells were chronically exposed to increasing concentrations of enzalutamide and the cells resistant to enzalutamide were referred to as C4-2B MDVR and Myc-CaP MDVR, respectively. The mechanisms of action in drug-resistant prostate cancer cells were determined using RNA sequencing analyses, RNA interference, real-time PCR, western blotting, and co-culturing technologies. Myc-CaP and Myc-CaP MDVR tumors were established in syngeneic FVB mice, and tumor-infiltrating leukocytes were isolated after enzalutamide treatment. The stained immune cells were determined by flow cytometry, and the data were analyzed using FlowJo.
    Immune-related signaling pathways (interferon alpha/gamma response, inflammatory response, and cell chemotaxis) were suppressed in human enzalutamide-resistant prostate cancer cells. PD-L1 was overexpressed and negatively regulated by androgen receptor signaling in resistant cells and patient with CRPC cohorts. Enzalutamide treatment decreased CD8+ T-cell numbers but increased monocytic myeloid-derived suppressor cell (M-MDSC) populations and PD-L1 expression within murine Myc-CaP tumors. Similarly, chemotaxis and immune response-regulating signaling pathways were suppressed, and PD-L1 expression was also increased using enzalutamide-resistant Myc-CaP MDVR cells. Notably, MDSC populations were significantly increased in Myc-CaP MDVR orthotopic tumors compared with those in Myc-CaP parental tumors. Co-culturing bone marrow cells with Myc-CaP MDVR cells significantly promoted MDSC differentiation and shifted towards M2 macrophage skewing.
    Our study suggests that immunosuppressive signaling can be promoted directly by enzalutamide-resistant prostate cancer cells and may be a potential means by which the efficacy of immune checkpoint inhibitors in enzalutamide-resistant prostate cancer is diminished.
    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:针对程序性细胞死亡1(PD-1)/程序性细胞死亡配体1(PD-L1)的免疫检查点抑制剂(ICIs)疗法显示出有希望的临床益处。然而,相对较低的应答率凸显了开发针对PD-1/PD-L1免疫检查点的替代策略的必要性.我们的研究重点是膜联蛋白A1(ANXA1)衍生肽A11降解PD-L1的作用和机制,以及A11对多种癌症中肿瘤免疫逃避的影响。
    方法:通过结合质谱分析的生物素下拉法鉴定A11与PD-L1的结合。通过筛选人去泛素酶cDNA文库,发现USP7作为PD-L1的去泛素酶。分析了A11与USP7竞争降解PD-L1的作用和机制。研究了A11通过抑制肿瘤免疫逃避增强T细胞介导的肿瘤细胞杀伤活性和抗肿瘤作用的能力。还在小鼠中研究了A11和PD-L1mAb(单克隆抗体)通过抑制肿瘤免疫逃避的协同抗肿瘤作用。采用免疫组织化学方法评价USP7和PD-L1在癌组织中的表达及临床意义。
    结果:A11通过泛素蛋白酶体途径降低乳腺癌中PD-L1蛋白的稳定性和水平,肺癌和黑色素瘤细胞。机械上,A11与PD-L1去泛素酶USP7竞争结合PD-L1,然后通过抑制USP7介导的PD-L1去泛素化降解PD-L1。功能上,A11促进T细胞体外杀伤癌细胞的能力,通过增加肿瘤微环境中CD8+T细胞的数量和激活来抑制小鼠的肿瘤免疫逃避,A11和PD-1mAb在小鼠体内具有协同抗肿瘤作用。此外,USP7和PD-L1的表达水平在乳腺癌中明显更高,非小细胞肺癌组织和皮肤黑色素瘤组织比相应的正常组织和癌组织呈正相关,预测PD-1mAb免疫治疗疗效和患者预后的两种蛋白均优于单个蛋白。
    结论:我们的结果表明,A11与USP7竞争结合并降解癌细胞中的PD-L1,A11具有明显的抗肿瘤作用,并通过抑制肿瘤免疫逃避与PD-1mAb具有协同抗肿瘤活性,A11可作为多种癌症ICIs治疗的替代策略。
    Immune checkpoint inhibitors (ICIs) therapy targeting programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) shows promising clinical benefits. However, the relatively low response rate highlights the need to develop an alternative strategy to target PD-1/PD-L1 immune checkpoint. Our study focuses on the role and mechanism of annexin A1 (ANXA1)-derived peptide A11 degrading PD-L1 and the effect of A11 on tumor immune evasion in multiple cancers.
    Binding of A11 to PD-L1 was identified by biotin pull-down coupled with mass spectrometry analysis. USP7 as PD-L1\'s deubiquitinase was found by screening a human deubiquitinase cDNA library. The role and mechanism of A11 competing with USP7 to degrade PD-L1 were analyzed. The capability to enhance the T cell-mediated tumor cell killing activity and antitumor effect of A11 via suppressing tumor immune evasion were investigated. The synergistic antitumor effect of A11 and PD-L1 mAb (monoclonal antibody) via suppressing tumor immune evasion were also studied in mice. The expression and clinical significance of USP7 and PD-L1 in cancer tissues were evaluated by immunohistochemistry.
    A11 decreases PD-L1 protein stability and levels by ubiquitin proteasome pathway in breast cancer, lung cancer and melanoma cells. Mechanistically, A11 competes with PD-L1\'s deubiquitinase USP7 for binding PD-L1, and then degrades PD-L1 by inhibiting USP7-mediated PD-L1 deubiquitination. Functionally, A11 promotes T cell ability of killing cancer cells in vitro, inhibits tumor immune evasion in mice via increasing the population and activation of CD8+ T cells in tumor microenvironment, and A11 and PD-1 mAb possess synergistic antitumor effect in mice. Moreover, expression levels of both USP7 and PD-L1 are significantly higher in breast cancer, non-small cell lung cancer and skin melanoma tissues than those in their corresponding normal tissues and are positively correlated in cancer tissues, and both proteins for predicting efficacy of PD-1 mAb immunotherapy and patient prognosis are superior to individual protein.
    Our results reveal that A11 competes with USP7 to bind and degrade PD-L1 in cancer cells, A11 exhibits obvious antitumor effects and synergistic antitumor activity with PD-1 mAb via inhibiting tumor immune evasion and A11 can serve as an alternative strategy for ICIs therapy in multiple 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
    异基因造血干细胞移植(allo-HSCT)代表了许多血液肿瘤疾病的唯一治愈性治疗选择。事实上,allo-HSCT被认为是最成功的免疫疗法之一,因为其临床疗效基于供体T细胞控制残留疾病的能力。该过程称为移植物抗白血病(GvL)反应。然而,同种反应性T细胞也可将宿主识别为外来物,并引发全身性潜在危及生命的炎症性疾病,称为移植物抗宿主病(GvHD).更好地了解导致GvHD或疾病复发的潜在机制可以帮助我们提高allo-HSCT的疗效和安全性。近年来,细胞外囊泡(EV)已成为细胞间串扰的关键组成部分。表达免疫检查点分子程序性死亡配体1(PD-L1)的癌症相关EV可以抑制T细胞反应,从而有助于免疫逃逸。同时,已经观察到,作为负反馈网络的一部分,炎症会触发PD-L1表达.这里,我们调查了allo-HSCT后循环EV是否表达PD-L1,并测试了其抑制(自体)T细胞有效靶向AML母细胞的能力的功效.最后,我们评估了EV上PD-L1水平与(T-)细胞重建之间的联系,GvHD,和疾病复发。我们能够检测到在allo-HSCT后6周达到峰值PD-L1表达的PD-L1+EV。急性GvHD的发展与allo-HSCT后PD-L1高EV的出现有关。此外,PD-L1水平与GvHD等级呈正相关,并且(仅)在成功的治疗干预后下降。与PD-L1低对应体相比,PD-L1高EV的T细胞抑制能力更高,并且可以使用PD-L1/PD-1阻断抗体拮抗。T细胞抑制性PD-L1高EV的丰度似乎也影响GvL疗效,因为患者复发的风险较高。最后,PD-L1高队列患者的总生存率降低.一起来看,我们显示,在allo-HSCT后存在表达PD-L1的EV。EV上的PD-L1水平与其抑制T细胞的能力和GvHD的发生相关。后一种观察可能表明负反馈机制来控制炎症(GvHD)活性。这种内在的免疫抑制可以随后促进疾病复发。
    Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents the only curative treatment option for a number of hemato-oncological disorders. In fact, allo-HSCT is considered as one of the most successful immunotherapies as its clinical efficacy is based on the donor T-cells\' capacity to control residual disease. This process is known as the graft-versus-leukemia (GvL) reaction. However, alloreactive T-cells can also recognize the host as foreign and trigger a systemic potentially life-threatening inflammatory disorder termed graft-versus-host disease (GvHD). A better understanding of the underlying mechanisms that lead to GvHD or disease relapse could help us to improve efficacy and safety of allo-HSCT. In recent years, extracellular vesicles (EVs) have emerged as critical components of intercellular crosstalk. Cancer-associated EVs that express the immune checkpoint molecule programmed death-ligand 1 (PD-L1) can suppress T-cell responses and thus contribute to immune escape. At the same time, it has been observed that inflammation triggers PD-L1 expression as part of a negative feedback network.Here, we investigated whether circulating EVs following allo-HSCT express PD-L1 and tested their efficacy to suppress the ability of (autologous) T-cells to effectively target AML blasts. Finally, we assessed the link between PD-L1 levels on EVs to (T-)cell reconstitution, GvHD, and disease relapse.We were able to detect PD-L1+ EVs that reached a peak PD-L1 expression at 6 weeks post allo-HSCT. Development of acute GvHD was linked to the emergence of PD-L1high EVs following allo-HSCT. Moreover, PD-L1 levels correlated positively with GvHD grade and declined (only) on successful therapeutic intervention. T-cell-inhibitory capacity was higher in PD-L1high EVs as compared with their PD-L1low counterparts and could be antagonized using PD-L1/PD-1 blocking antibodies. Abundance of T-cell-suppressive PD-L1high EVs appears to also impact GvL efficacy as patients were at higher risk for relapse. Finally, patients of PD-L1high cohort displayed a reduced overall survival.Taken together, we show that PD-L1-expressing EVs are present following allo-HSCT. PD-L1 levels on EVs correlate with their ability to suppress T-cells and the occurrence of GvHD. The latter observation may indicate a negative feedback mechanism to control inflammatory (GvHD) activity. This intrinsic immunosuppression could subsequently promote disease relapse.
    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:曲妥珠单抗治疗HER2阳性(HER2+)乳腺癌的成功,针对HER2的抗体依赖于免疫反应。我们证明了TNFα诱导粘蛋白4(MUC4)表达,屏蔽HER2分子上的曲妥珠单抗表位,从而降低其治疗效果。这里,我们使用小鼠模型和HER2+乳腺癌患者的样本,通过促进免疫逃避,揭示MUC4参与阻碍曲妥珠单抗效应的作用.
    方法:我们与曲妥珠单抗一起使用了对可溶性TNFα(sTNFα)具有选择性的显性阴性TNFα抑制剂(DN)。使用两种条件MUC4沉默的肿瘤模型进行临床前实验以表征免疫细胞浸润。用曲妥珠单抗治疗的91名患者的队列用于将肿瘤MUC4与肿瘤浸润淋巴细胞相关联。
    结果:在携带从头对曲妥珠单抗耐药的HER2+乳腺肿瘤的小鼠中,用DN中和sTNFα诱导MUC4下调。使用有条件的MUC4沉默的肿瘤模型,曲妥珠单抗的抗肿瘤作用得以恢复,加入TNFα阻断剂并未进一步降低肿瘤负荷.DN给予曲妥珠单抗通过M1样表型巨噬细胞极化和NK细胞脱颗粒改变免疫抑制肿瘤环境。耗竭实验揭示了巨噬细胞和NK细胞之间的串扰是曲妥珠单抗抗肿瘤作用所必需的。此外,接受DN治疗的肿瘤细胞对曲妥珠单抗依赖性细胞吞噬作用更敏感.最后,MUC4在HER2+乳腺癌中的表达与免疫性沙漠肿瘤有关。
    结论:这些发现为MUC4+和HER2+乳腺癌患者应用sTNFα阻断联合曲妥珠单抗或曲妥珠单抗药物偶联物克服曲妥珠单抗耐药提供了理论基础。
    The success of HER2-positive (HER2+) breast cancer treatment with trastuzumab, an antibody that targets HER2, relies on immune response. We demonstrated that TNFα induces mucin 4 (MUC4) expression, which shields the trastuzumab epitope on the HER2 molecule decreasing its therapeutic effect. Here, we used mouse models and samples from HER2+ breast cancer patients to unravel MUC4 participation in hindering trastuzumab effect by fostering immune evasion.
    We used a dominant negative TNFα inhibitor (DN) selective for soluble TNFα (sTNFα) together with trastuzumab. Preclinical experiments were performed using two models of conditionally MUC4-silenced tumors to characterize the immune cell infiltration. A cohort of 91 patients treated with trastuzumab was used to correlate tumor MUC4 with tumor-infiltrating lymphocytes.
    In mice bearing de novo trastuzumab-resistant HER2+ breast tumors, neutralizing sTNFα with DN induced MUC4 downregulation. Using the conditionally MUC4-silenced tumor models, the antitumor effect of trastuzumab was reinstated and the addition of TNFα-blocking agents did not further decrease tumor burden. DN administration with trastuzumab modifies the immunosuppressive tumor milieu through M1-like phenotype macrophage polarization and NK cells degranulation. Depletion experiments revealed a cross-talk between macrophages and NK cells necessary for trastuzumab antitumor effect. In addition, tumor cells treated with DN are more susceptible to trastuzumab-dependent cellular phagocytosis. Finally, MUC4 expression in HER2+ breast cancer is associated with immune desert tumors.
    These findings provide rationale to pursue sTNFα blockade combined with trastuzumab or trastuzumab drug conjugates for MUC4+ and HER2+ breast cancer patients to overcome trastuzumab resistance.
    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:大约一半的弥漫性大B细胞淋巴瘤(DLBCL)被大量调节性T细胞(Tregs)浸润。尽管在接受标准利妥昔单抗加环磷酰胺治疗的患者中,特别是效应物Treg的存在与预后较差相关,阿霉素,长春新碱,和泼尼松(R-CHOP)免疫化疗,这种细胞类型在淋巴瘤发生和发展过程中的作用知之甚少.
    方法:这里,我们使用包含前瞻性收集的DLBCL患者标本的组织微阵列,以及来自公开可用队列的数据,以探索Treg浸润的DLBCL的突变情况。我们进一步利用MYC驱动的淋巴瘤模型,从机械上剖析Tregs对淋巴瘤发病机理的贡献,并开发Treg选择性白介素2(IL-2)饥饿的策略,以改善MYC驱动的淋巴瘤的免疫控制。
    结果:我们发现所有遗传DLBCL亚型,除了以共同发生的MYD88/CD79突变为特征的一个,被Tregs严重渗透.光谱流式细胞术和scRNA测序揭示了Tregs浸润性MYC驱动的淋巴瘤的功能和免疫抑制标志物的稳健表达;我们发现肿瘤内Treg是由于肿瘤接触时幼稚CD4+前体的局部转化而产生的。Foxp3iDTR小鼠中的Treg消融,或通过抗体介导的Treg选择性阻断IL-2信号传导,大大减轻了淋巴瘤的负担。我们确定淋巴瘤B细胞是IL-2的主要来源,并显示Treg耗竭的作用被同时耗竭的Foxp3阴性CD4+T细胞逆转。但不是CD8+T细胞或自然杀伤(NK)细胞。Tregs对ATP水解和腺苷产生的抑制至少部分地表型复制了Treg消耗的影响。Treg消耗进一步与促凋亡CD40活化协同作用以维持持久的反应。
    结论:综合数据表明Tregs是DLBCL的潜在治疗靶点,尤其是与其他免疫疗法结合。
    Roughly half of all diffuse large B-cell lymphomas (DLBCLs) are infiltrated by large numbers of regulatory T-cells (Tregs). Although the presence of \'effector\' Tregs in particular is associated with an inferior prognosis in patients on standard rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) immunochemotherapy, the role of this cell type during lymphoma initiation and progression is poorly understood.
    Here, we use tissue microarrays containing prospectively collected DLBCL patient specimens, as well as data from publicly available cohorts to explore the mutational landscape of Treg-infiltrated DLBCL. We further take advantage of a model of MYC-driven lymphoma to mechanistically dissect the contribution of Tregs to lymphoma pathogenesis and to develop a strategy of Treg-selective interleukin-2 (IL-2) starvation to improve immune control of MYC-driven lymphoma.
    We find that all genetic DLBCL subtypes, except for one characterized by co-occurring MYD88/CD79 mutations, are heavily infiltrated by Tregs. Spectral flow cytometry and scRNA-sequencing reveal the robust expression of functional and immunosuppressive markers on Tregs infiltrating MYC-driven lymphomas; notably, we find that intratumoral Tregs arise due to local conversion from naïve CD4+ precursors on tumor contact. Treg ablation in Foxp3iDTR mice, or by antibody-mediated Treg-selective blockade of IL-2 signaling, strongly reduces the lymphoma burden. We identify lymphoma B-cells as a major source of IL-2, and show that the effects of Treg depletion are reversed by the simultaneous depletion of Foxp3-negative CD4+ T-cells, but not CD8+ T-cells or natural killer (NK) cells. The inhibition of ATP hydrolyzation and adenosine production by Tregs at least partly phenocopies the effects of Treg depletion. Treg depletion further synergizes with pro-apoptotic CD40 activation to sustain durable responses.
    The combined data implicate Tregs as a potential therapeutic target in DLBCL, especially in combination with other immunotherapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号