Interferon Inducers

干扰素诱导剂
  • 文章类型: Journal Article
    背景:高级别浆液性卵巢癌(HGSC)是最致命的妇科恶性肿瘤,其特征是对化疗耐药和高复发率。HGSC肿瘤表现出抑癌基因丢失的高患病率。鉴于BRCA1和PTEN基因的1型干扰素调节功能及其相关的对比T细胞浸润和非浸润肿瘤免疫微环境(TIME)状态,分别,在这项研究中,我们研究了干扰素基因刺激因子(STING)途径激活在通过增强化疗反应改善总体生存率方面的潜力,特别是在PTEN缺乏的肿瘤中。
    方法:在组织微阵列中评估PTEN蛋白的表达,所述组织微阵列使用从110名患有HGSC的患者的队列收集的预处理肿瘤产生。进行多重免疫荧光染色以确定所选淋巴和骨髓细胞的空间分布和密度。使用同系小鼠HGSC细胞系的体内研究,ID8-Trp53-/-;Pten-/-和ID8-Trp53-/-;Brca1-/-,进行了表征时间和对卡铂化疗联合外源性STING激活疗法的反应。
    结果:与完整的PTEN表达相比,缺乏PTEN蛋白的患者肿瘤表现出疾病特异性存活和上皮内CD68+巨噬细胞浸润的显著降低。体内研究表明,Pten缺乏的卵巢癌细胞建立了免疫抑制的时间,其特征是M2样巨噬细胞的比例增加,腹水中的GR1+MDSCs,与Brca1缺陷细胞相比,效应CD8+细胞毒性T细胞功能降低;进一步,注射Pten缺陷型ID8细胞的小鼠肿瘤由于抑制恶性腹水中的巨噬细胞优势而表现出攻击行为。联合化疗,外源性STING激活导致注射Pten缺陷ID8细胞的小鼠的总生存期更长,将Pten缺陷性腹水衍生的腹膜内M2样巨噬细胞重编程为M1样表型,并挽救了CD8细胞毒性T细胞活化。
    结论:本研究揭示了考虑癌细胞内在遗传改变对治疗选择时间的影响的重要性。我们建立了最佳掺入干扰素激活疗法作为PTEN缺陷型HGSC的新型组合策略的基本原理。
    High-grade serous ovarian carcinoma (HGSC) is the most lethal gynecologic malignancy characterized by resistance to chemotherapy and high rates of recurrence. HGSC tumors display a high prevalence of tumor suppressor gene loss. Given the type 1 interferon regulatory function of BRCA1 and PTENgenes and their associated contrasting T-cell infiltrated and non-infiltrated tumor immune microenvironment (TIME) states, respectively, in this study we investigated the potential of stimulator of interferon genes (STING) pathway activation in improving overall survival via enhancing chemotherapy response, specifically in tumors with PTEN deficiency.
    Expression of PTEN protein was evaluated in tissue microarrays generated using pretreatment tumors collected from a cohort of 110 patients with HGSC. Multiplex immunofluorescence staining was performed to determine spatial profiles and density of selected lymphoid and myeloid cells. In vivo studies using the syngeneic murine HGSC cell lines, ID8-Trp53 -/-; Pten -/- and ID8-Trp53 -/-; Brca1 -/-, were conducted to characterize the TIME and response to carboplatin chemotherapy in combination with exogenous STING activation therapy.
    Patient tumors with absence of PTEN protein exhibited a significantly decreased disease specific survival and intraepithelial CD68+ macrophage infiltration as compared with intact PTEN expression. In vivo studies demonstrated that Pten-deficient ovarian cancer cells establish an immunosuppressed TIME characterized by increased proportions of M2-like macrophages, GR1+MDSCs in the ascites, and reduced effector CD8+ cytotoxic T-cell function compared with Brca1-deficient cells; further, tumors from mice injected with Pten-deficient ID8 cells exhibited an aggressive behavior due to suppressive macrophage dominance in the malignant ascites. In combination with chemotherapy, exogenous STING activation resulted in longer overall survival in mice injected with Pten-deficient ID8 cells, reprogrammed intraperitoneal M2-like macrophages derived from Pten-deficient ascites to M1-like phenotype and rescued CD8+ cytotoxic T-cell activation.
    This study reveals the importance of considering the influence of cancer cell intrinsic genetic alterations on the TIME for therapeutic selection. We establish the rationale for the optimal incorporation of interferon activating therapies as a novel combination strategy in PTEN-deficient HGSC.
    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
    The review contains a brief analysis of the 60-year history of the discovery, study and medical application of interferons, a new group of remarkable proteins that have found wide medical application in the therapy of virological, oncological, neurological, ophthalmic and other pathologies. Modern data on the classification of interferons and the mechanisms of their action are given. Particular attention is paid to the clinical use of medications of interferon and its inducers.
    Обзор содержит краткий анализ 60-летней истории открытия, изучения и медицинского применения интерферонов - новой группы замечательных белков, нашедших широкое применение в терапии вирусологических, онкологических, неврологических, офтальмологических и иных форм патологии. Приводятся современные данные о классификации интерферонов и механизмах их действия. Особое внимание уделено клиническому применению лекарственных препаратов интерферона и его индукторов.
    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
    卵巢癌(OC)迫切需要新的治疗方法。女性第五大致命癌症。临床前工作表明,DNA甲基转移酶抑制剂(DNMTis)可以逆转OC中的免疫抑制肿瘤微环境。抑制DNA甲基转移酶激活双链(ds)RNA的转录,包括转座元素。这些dsRNA激活细胞质中的传感器并触发I型干扰素(IFN)信号,招募宿主免疫细胞杀死肿瘤细胞。腺苷脱氨酶1(ADAR1)由IFN信号诱导,并编辑哺乳动物dsRNA,其被读取为测序数据中的A到G变化。这些编辑的dsRNA不能被dsRNA传感器感知,因此ADAR1在负反馈回路中抑制I型IFN应答。我们假设减少ADAR1编辑将增强DNMTi诱导的免疫应答。
    人OC细胞系在体外用DNMTi处理,然后RNA测序以测量RNA编辑。Adar1在ID8Trp53-/-小鼠OC细胞中稳定敲低。用模拟或DNMTi处理测试对照细胞(shGFP)或shAdar1细胞。使用流式细胞术对肿瘤浸润性免疫细胞进行免疫表型分析,并分析细胞培养上清液的分泌趋化因子/细胞因子。将小鼠注射同基因shAdar1ID8Trp53-/-细胞并用四氢尿苷/DNMTi处理,同时每3天给予抗-干扰素α和β受体1、抗-CD8或抗-NK1.1抗体。
    我们显示,在体外DNMTi处理后,ADAR1在人OC细胞系中编辑转座因子。与单独的任一种扰动相比,将ADAR1敲低与DNMTi组合显著增加促炎细胞因子/趋化因子的产生和对IFN-β的敏感性。此外,在OC的免疫活性小鼠模型中,DNMTi治疗和Adar1损失减少肿瘤负荷并延长存活。Adar1损失和DNMTi的组合引发了最强大的抗肿瘤反应,并通过增加CD8T细胞的募集和激活来转化免疫微环境。
    总之,我们表明,DNMTi+ADAR1抑制的生存获益依赖于I型IFN信号传导.因此,表观遗传诱导转座因子转录结合抑制RNA编辑是逆转OC免疫逃避的一种新的治疗策略,一种对目前的免疫疗法没有反应的疾病。
    Novel therapies are urgently needed for ovarian cancer (OC), the fifth deadliest cancer in women. Preclinical work has shown that DNA methyltransferase inhibitors (DNMTis) can reverse the immunosuppressive tumor microenvironment in OC. Inhibiting DNA methyltransferases activate transcription of double-stranded (ds)RNA, including transposable elements. These dsRNAs activate sensors in the cytoplasm and trigger type I interferon (IFN) signaling, recruiting host immune cells to kill the tumor cells. Adenosine deaminase 1 (ADAR1) is induced by IFN signaling and edits mammalian dsRNA with an A-to-I nucleotide change, which is read as an A-to-G change in sequencing data. These edited dsRNAs cannot be sensed by dsRNA sensors, and thus ADAR1 inhibits the type I IFN response in a negative feedback loop. We hypothesized that decreasing ADAR1 editing would enhance the DNMTi-induced immune response.
    Human OC cell lines were treated in vitro with DNMTi and then RNA-sequenced to measure RNA editing. Adar1 was stably knocked down in ID8 Trp53-/- mouse OC cells. Control cells (shGFP) or shAdar1 cells were tested with mock or DNMTi treatment. Tumor-infiltrating immune cells were immunophenotyped using flow cytometry and cell culture supernatants were analyzed for secreted chemokines/cytokines. Mice were injected with syngeneic shAdar1 ID8 Trp53-/- cells and treated with tetrahydrouridine/DNMTi while given anti-interferon alpha and beta receptor 1, anti-CD8, or anti-NK1.1 antibodies every 3 days.
    We show that ADAR1 edits transposable elements in human OC cell lines after DNMTi treatment in vitro. Combining ADAR1 knockdown with DNMTi significantly increases pro-inflammatory cytokine/chemokine production and sensitivity to IFN-β compared with either perturbation alone. Furthermore, DNMTi treatment and Adar1 loss reduces tumor burden and prolongs survival in an immunocompetent mouse model of OC. Combining Adar1 loss and DNMTi elicited the most robust antitumor response and transformed the immune microenvironment with increased recruitment and activation of CD8+ T cells.
    In summary, we showed that the survival benefit from DNMTi plus ADAR1 inhibition is dependent on type I IFN signaling. Thus, epigenetically inducing transposable element transcription combined with inhibition of RNA editing is a novel therapeutic strategy to reverse immune evasion in OC, a disease that does not respond to current immunotherapies.
    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
    树突状细胞(DC)是启动对病原体和肿瘤细胞的免疫防御的专职抗原呈递细胞。人类肿瘤仅含有很少的DC,其主要表现出非活化表型。因此,肿瘤相关DC的激活可以提高癌症免疫疗法的疗效.已知Toll样受体(TLR)激动剂和干扰素促进DC成熟。然而,目前尚不清楚人肿瘤中的DC是否对激活信号有反应,以及哪种刺激诱导人肿瘤DC的最佳激活。
    我们首先筛选了TLR激动剂的组合,STING激动剂和干扰素(IFN)激活人常规DC(cDC)的能力。更详细地研究了两种组合:TL8-506(TLR8激动剂)+IFN-γ和TL8-506+聚(I:C)(TLR3激动剂)。cDC1s和cDC2s来源于脐带血干细胞,用TL8-506+IFN-γ或TL8-506+Poly(I:C)刺激血液或患者肿瘤样品。通过ELISA分析不同的活化标志物,流式细胞术,NanoStringnCounter技术或单细胞RNA测序。进行T细胞活化和迁移测定以评估cDC活化的功能后果。
    我们显示TL8-506与IFN-γ或Poly(I:C)协同诱导不同趋化因子和细胞因子(包括白细胞介素(IL)-12p70)在人脐带血和血液cDC亚群中以组合特异性方式高表达。重要的是,两种组合体外诱导患者肿瘤样品中cDC亚群的激活。对抗癌反应重要的免疫刺激基因包括CD40、IFNB1、IFNL1、IL12A和IL12B的表达在刺激时上调。此外,响应于TL8-506组合,在肿瘤来源的cDC中诱导与CD8+T细胞募集相关的趋化因子。体外活化和迁移测定证实,刺激的cDC诱导T细胞活化和迁移。
    我们的数据表明,脐带血来源和血液来源的cDC是研究人类肿瘤cDC治疗反应的良好替代品。虽然人类肿瘤中的大多数cDC表现出非活化表型,TL8-506组合驱动人肿瘤cDC朝向与刺激时的Th1应答相关的免疫刺激表型。因此,基于TL8-506的组合可能是启动或促进癌症患者抗肿瘤反应的有希望的候选者。
    Dendritic cells (DCs) are professional antigen presenting cells that initiate immune defense to pathogens and tumor cells. Human tumors contain only few DCs that mostly display a non-activated phenotype. Hence, activation of tumor-associated DCs may improve efficacy of cancer immunotherapies. Toll-like receptor (TLR) agonists and interferons are known to promote DC maturation. However, it is unclear if DCs in human tumors respond to activation signals and which stimuli induce the optimal activation of human tumor DCs.
    We first screened combinations of TLR agonists, a STING agonist and interferons (IFNs) for their ability to activate human conventional DCs (cDCs). Two combinations: TL8-506 (a TLR8 agonist)+IFN-γ and TL8-506+Poly(I:C) (a TLR3 agonist) were studied in more detail. cDC1s and cDC2s derived from cord blood stem cells, blood or patient tumor samples were stimulated with either TL8-506+IFN-γ or TL8-506+Poly(I:C). Different activation markers were analyzed by ELISA, flow cytometry, NanoString nCounter Technology or single-cell RNA-sequencing. T cell activation and migration assays were performed to assess functional consequences of cDC activation.
    We show that TL8-506 synergized with IFN-γ or Poly(I:C) to induce high expression of different chemokines and cytokines including interleukin (IL)-12p70 in human cord blood and blood cDC subsets in a combination-specific manner. Importantly, both combinations induced the activation of cDC subsets in patient tumor samples ex vivo. The expression of immunostimulatory genes important for anticancer responses including CD40, IFNB1, IFNL1, IL12A and IL12B were upregulated on stimulation. Furthermore, chemokines associated with CD8+ T cell recruitment were induced in tumor-derived cDCs in response to TL8-506 combinations. In vitro activation and migration assays confirmed that stimulated cDCs induce T cell activation and migration.
    Our data suggest that cord blood-derived and blood-derived cDCs are a good surrogate to study treatment responses in human tumor cDCs. While most cDCs in human tumors display a non-activated phenotype, TL8-506 combinations drive human tumor cDCs towards an immunostimulatory phenotype associated with Th1 responses on stimulation. Hence, TL8-506-based combinations may be promising candidates to initiate or boost antitumor responses in patients with cancer.
    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
    转化生长因子-β(TGFβ)可以限制癌症治疗的疗效。包括放疗(RT),通过诱导免疫抑制的肿瘤环境。TGFβ与受损的T细胞浸润和抗肿瘤免疫的关联是已知的,但TGFβ参与免疫细胞排斥和限制抗肿瘤治疗疗效的机制需要进一步研究.
    我们使用了临床相关的TGFβ受体2(TGFβR2)中和抗体MT1和小分子TGFβR1抑制剂LY3200882,并评估了它们与RT联合对头颈部和肺癌小鼠原位模型的疗效。
    我们证明了TGFβ途径抑制强烈增加了RT的功效。TGFβR2抗体上调受照射的肿瘤内肿瘤相关巨噬细胞中的干扰素β表达,并有利于肿瘤病变周围和核心的T细胞浸润。我们强调,MT1和RT联合观察到的抗肿瘤功效和淋巴细胞浸润增加都依赖于I型干扰素信号传导。
    这些数据为TGFβ在限制RT疗效中的作用提供了新的思路,鉴定涉及抑制巨噬细胞衍生的I型干扰素产生的新机制,并促进在头颈部和肺癌的治疗策略中结合使用TGFβR抑制剂与RT。
    Transforming growth factor-beta (TGFβ) can limit the efficacy of cancer treatments, including radiotherapy (RT), by inducing an immunosuppressive tumor environment. The association of TGFβ with impaired T cell infiltration and antitumor immunity is known, but the mechanisms by which TGFβ participates in immune cell exclusion and limits the efficacy of antitumor therapies warrant further investigations.
    We used the clinically relevant TGFβ receptor 2 (TGFβR2)-neutralizing antibody MT1 and the small molecule TGFβR1 inhibitor LY3200882 and evaluated their efficacy in combination with RT against murine orthotopic models of head and neck and lung cancer.
    We demonstrated that TGFβ pathway inhibition strongly increased the efficacy of RT. TGFβR2 antibody upregulated interferon beta expression in tumor-associated macrophages within the irradiated tumors and favored T cell infiltration at the periphery and within the core of the tumor lesions. We highlighted that both the antitumor efficacy and the increased lymphocyte infiltration observed with the combination of MT1 and RT were dependent on type I interferon signaling.
    These data shed new light on the role of TGFβ in limiting the efficacy of RT, identifying a novel mechanism involving the inhibition of macrophage-derived type I interferon production, and fostering the use of TGFβR inhibition in combination with RT in therapeutic strategies for the management of head and neck and lung cancer.
    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
    已知放射疗法(RT)以免疫原性方式杀死癌细胞的能力。最近的临床前数据表明,靶向α-粒子治疗与RT具有引发免疫刺激效应的能力,在免疫“冷”肿瘤中,作为一种规避免疫检查点抑制剂耐药性的有前途的策略。
    Radiation therapy (RT) is known for its ability to kill cancer cells in an immunogenic manner. Recent preclinical data demonstrate that targeted alpha-particle therapy shares with RT the capacity to elicit immunostimulatory effects, standing out as a promising strategy to circumvent immune checkpoint inhibitor resistance in immunologically \'cold\' tumors.
    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
    银屑病是一种慢性炎症性皮肤病。最近,据报道,溶血磷脂酸(LPA)/LPAR5信号参与巨噬细胞中的NLRP3炎性体激活和角质形成细胞激活以产生炎性细胞因子,有助于银屑病的发病机制。然而,LPA/LPAR信号在银屑病角质形成细胞增殖中的作用和分子机制尚不清楚.在这项研究中,我们研究了LPAR1/3抑制对咪喹莫特(IMQ)诱导的银屑病样小鼠的影响。用LPAR1/3拮抗剂治疗,ki16425减轻了IMQ诱导的银屑病样小鼠模型的皮肤症状,并减少了病变中的角质形成细胞增殖。它还通过增加细胞周期蛋白A2,细胞周期蛋白D1,细胞周期蛋白依赖性激酶(CDK)2和CDK4的表达和减少HaCaT细胞中p27Kip1的表达来降低LPA诱导的细胞增殖和细胞周期进程。在HaCaT细胞中LPAR1敲低降低LPA诱导的增殖,抑制细胞周期蛋白A2和CDK2表达,并恢复p27Kip1表达。LPA增加Rho相关蛋白激酶2(ROCK2)表达和PI3K/AKT激活;ROCK2和PI3K/AKT信号的药理学抑制抑制LPA诱导的细胞周期进程。总之,我们证明LPAR1/3拮抗剂可缓解IMQ诱导的小鼠银屑病样症状,特别是,LPAR1信号传导通过角质形成细胞中的ROCK2/PI3K/AKT途径参与细胞周期进程。
    Psoriasis is a chronic inflammatory skin disease. Recently, lysophosphatidic acid (LPA)/LPAR5 signaling has been reported to be involved in both NLRP3 inflammasome activation in macrophages and keratinocyte activation to produce inflammatory cytokines, contributing to psoriasis pathogenesis. However, the effect and molecular mechanisms of LPA/LPAR signaling in keratinocyte proliferation in psoriasis remain unclear. In this study, we investigated the effects of LPAR1/3 inhibition on imiquimod (IMQ)-induced psoriasis-like mice. Treatment with the LPAR1/3 antagonist, ki16425, alleviated skin symptoms in IMQ-induced psoriasis-like mouse models and decreased keratinocyte proliferation in the lesion. It also decreased LPA-induced cell proliferation and cell cycle progression via increased cyclin A2, cyclin D1, cyclin-dependent kinase (CDK)2, and CDK4 expression and decreased p27Kip1 expression in HaCaT cells. LPAR1 knockdown in HaCaT cells reduced LPA-induced proliferation, suppressed cyclin A2 and CDK2 expression, and restored p27Kip1 expression. LPA increased Rho-associated protein kinase 2 (ROCK2) expression and PI3K/AKT activation; moreover, the pharmacological inhibition of ROCK2 and PI3K/AKT signaling suppressed LPA-induced cell cycle progression. In conclusion, we demonstrated that LPAR1/3 antagonist alleviates IMQ-induced psoriasis-like symptoms in mice, and in particular, LPAR1 signaling is involved in cell cycle progression via ROCK2/PI3K/AKT pathways in keratinocytes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人类流行病学研究表明,在神经发育障碍的病因中,妊娠期间暴露于感染。母体免疫激活(MIA)的动物模型已将母体免疫反应确定为母体感染与异常后代大脑和行为发育之间的关键联系。在这里,我们评估了由MIA处理的水母(n=14)出生的雄性恒河猴(Macacamulatta)的神经发育,这些水母在孕早期末注射了改良形式的病毒模拟物多聚肌胞苷:多聚胞嘧啶酸。对照大坝在相同的妊娠时间点接受盐水注射(n=10)或未经处理(n=4)。MIA治疗的水坝表现出强烈的免疫反应,以疾病行为的短暂增加为索引,温度,和炎性细胞因子。尽管控制或MIA处理的水坝所生的后代在身体生长和早期发育里程碑方面没有差异,MIA治疗的动物在认知发育方面表现出微妙的变化,并且偏离了物种典型的大脑生长轨迹。纵向MRI显示,经过MIA治疗的后代在6个月时前额叶和额叶皮质的灰质体积显着减少,并持续到45个月的最后时间点,而经过MIA治疗的动物在36和45个月时额叶白质体积较小。这些发现提供了MIA暴露的非人灵长类动物出生后早期大脑发育变化的第一个证据,并建立了一个翻译相关的模型系统,以探索从出生到青春期后期与产前免疫挑战相关的风险的神经发育轨迹。重要声明怀孕期间暴露于感染的妇女生孩子的风险增加,孩子后来被诊断患有神经发育障碍。临床前母体免疫激活(MIA)模型已证明母体感染对胎儿脑发育的影响是由母体免疫反应介导的。由于大多数MIA模型都是在啮齿动物中进行的,非人灵长类动物提供了一个独特的系统来评估与人类密切相关的物种中的MIA假说。在这里,我们报告了在非人灵长类MIA模型中进行的第一项纵向研究。MIA暴露的后代表现出认知发育的微妙变化,并伴有额叶灰质和白质的明显减少,进一步支持产前免疫攻击与后代神经发育改变之间的关联。
    Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.
    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
    Viral-based immunotherapy can overcome resistance to immune checkpoint blockade (ICB) and fill the unmet needs of many patients with cancer. Oncolytic viruses (OVs) are defined as engineered or naturally occurring viruses that selectively replicate in and kill cancer cells. OVs also induce antitumor immunity. The purpose of this study was to compare the antitumor effects of live oncolytic vaccinia viruses versus the inactivated versions and elucidate their underlying immunological mechanisms.
    We engineered a replication-competent, oncolytic vaccinia virus (OV-GM) by inserting a murine GM-CSF gene into the thymidine kinase locus of a mutant vaccinia E3L∆83N, which lacks the Z-DNA-binding domain of vaccinia virulence factor E3. We compared the antitumor effects of intratumoral (IT) delivery of live OV-GM versus heat-inactivated OV-GM (heat-iOV-GM) in a murine B16-F10 melanoma bilateral implantation model. We also generated vvDD, a well-studied oncolytic vaccinia virus, and compared the antitumor effects of live vvDD vs heat-inactivated vvDD (heat-ivvDD) in a murine A20 B-cell lymphoma bilateral tumor implantation model.
    Heat-iOV-GM infection of dendritic cells (DCs) and tumor cells in vitro induced type I interferon and proinflammatory cytokines and chemokines, whereas live OV-GM did not. IT live OV-GM was less effective in generating systemic antitumor immunity compared with heat-iOV-GM. Similar to heat-iOV-GM, the antitumor effects of live OV-GM also require Batf3-dependent CD103+ dendritic cells. When combined with systemic delivery of ICB, IT heat-iOV-GM was more effective in eradicating tumors, compared with live OV-GM. IT heat-ivvDD was also more effective in treating murine A20 B-cell lymphoma, compared with live vvDD.
    Tumor lysis induced by the replication of oncolytic vaccinia virus has a limited effect on the generation of systemic antitumor immunity. The activation of Batf3-dependent CD103+ DCs is critical for antitumor effects induced by both live OV-GM and heat-iOV-GM, with the latter being more potent than live OV-GM in inducing innate and adaptive immunity in both locally injected and distant, non-injected tumors. We propose that evaluations of both innate and adaptive immunity, induced by IT oncolytic viral immunotherapy at injected and non-injected tumors, should be included as potential biomarkers for host responses to viral 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
    修饰的安卡拉痘苗病毒(MVA)是基因工程改造的非复制型病毒载体。MVA的瘤内施用诱导环GMP-AMP合酶介导的I型干扰素(IFN)应答和高水平的转基因的产生,所述转基因被工程化到病毒基因组中,例如肿瘤抗原以构建癌症疫苗。尽管I型IFN对于建立CD8介导的抗肿瘤反应至关重要,这种细胞因子家族也可能产生免疫抑制机制。
    进行体外测定以评价辛伐他汀和阿托伐他汀对I型IFN信号传导和抗原呈递的活性。IFNα/β受体1的表面水平,牛血清白蛋白-荧光素5(6)-异硫氰酸盐的内吞作用,信号转导和转录激活因子(STAT)磷酸化,在鼠成纤维细胞系L929中评估IFN刺激的基因和实时PCR。在表达卵清蛋白的B16黑素瘤(OVA)和Lewis肺癌(LLC)-OVA肿瘤模型中,进行体内实验以表征辛伐他汀对MVA诱导的先天免疫应答和对基于MVA的抗肿瘤疫苗的抗肿瘤作用。RNAseq分析,消耗单克隆抗体,和流式细胞术用于评估MVA介导的免疫应答。
    在这项工作中,由于他汀类药物能够降低IFN-α/β受体1的表面表达水平和减少网格蛋白介导的内吞作用,我们将常用的他汀类药物鉴定为有效的IFNα药物抑制剂.辛伐他汀和阿托伐他汀有效地消除了8小时对IFNα的转录组反应,并增加了体内呈递与主要组织相容性复合物(MHC)I类结合的OVA衍生肽的树突状细胞的数量。辛伐他汀的腹膜内或肌内给药减少了肿瘤周围给药MVA介导的炎症反应,并增强了编码肿瘤相关抗原的MVA的抗肿瘤活性。协同抗肿瘤作用关键取决于CD8+细胞,而它们通过CD4+淋巴细胞的消耗而显著改善,调节性T细胞,或NK细胞。MVA-OVA单独或联合辛伐他汀增强的B细胞,CD4+淋巴细胞,CD8+淋巴细胞,和肿瘤引流淋巴结中的肿瘤特异性CD8+。然而,只有治疗组合增加了肿瘤微环境和脾脏中这些淋巴细胞群的数量。
    总之,辛伐他汀对IFNα功能的阻断显着增强了淋巴细胞浸润和MVA的抗肿瘤活性,促使一个可行的药物再利用。
    Modified vaccinia virus Ankara (MVA) are genetically engineered non-replicating viral vectors. Intratumoral administration of MVA induces a cyclic GMP-AMP synthase-mediated type I interferon (IFN) response and the production of high levels of the transgenes engineered into the viral genome such as tumor antigens to construct cancer vaccines. Although type I IFNs are essential for establishing CD8-mediated antitumor responses, this cytokine family may also give rise to immunosuppressive mechanisms.
    In vitro assays were performed to evaluate the activity of simvastatin and atorvastatin on type I IFN signaling and on antigen presentation. Surface levels of IFN α/β receptor 1, endocytosis of bovine serum albumin-fluorescein 5 (6)-isothiocyanate, signal transducer and activator of transcription (STAT) phosphorylation, and real-time PCR of IFN-stimulated genes were assessed in the murine fibroblast cell line L929. In vivo experiments were performed to characterize the effect of simvastatin on the MVA-induced innate immune response and on the antitumor effect of MVA-based antitumor vaccines in B16 melanoma expressing ovalbumin (OVA) and Lewis lung carcinoma (LLC)-OVA tumor models. RNAseq analysis, depleting monoclonal antibodies, and flow cytometry were used to evaluate the MVA-mediated immune response.
    In this work, we identified commonly prescribed statins as potent IFNα pharmacological inhibitors due to their ability to reduce surface expression levels of IFN-α/β receptor 1 and to reduce clathrin-mediated endocytosis. Simvastatin and atorvastatin efficiently abrogated for 8 hours the transcriptomic response to IFNα and enhanced the number of dendritic cells presenting an OVA-derived peptide bound to major histocompatibility complex (MHC) class I. In vivo, intraperitoneal or intramuscular administration of simvastatin reduced the inflammatory response mediated by peritumoral administration of MVA and enhanced the antitumor activity of MVA encoding tumor-associated antigens. The synergistic antitumor effects critically depend on CD8+ cells, whereas they were markedly improved by depletion of CD4+ lymphocytes, T regulatory cells, or NK cells. Either MVA-OVA alone or combined with simvastatin augmented B cells, CD4+ lymphocytes, CD8+ lymphocytes, and tumor-specific CD8+ in the tumor-draining lymph nodes. However, only the treatment combination increased the numbers of these lymphocyte populations in the tumor microenvironment and in the spleen.
    In conclusion, blockade of IFNα functions by simvastatin markedly enhances lymphocyte infiltration and the antitumor activity of MVA, prompting a feasible drug repurposing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号