Toll-Like Receptor Agonists

Toll 样受体激动剂
  • 文章类型: Journal Article
    Toll样受体(TLR)激动剂由于其有效的免疫刺激特性而被开发为抗癌治疗剂。然而,将TLR激动剂作为单一疗法进行试验的临床试验通常未能证明与标准治疗相比有显著改善.我们假设TLR激动剂免疫疗法的抗癌功效可以通过组合方法得到改善。为了防止毒性增加,常见的全身联合疗法,我们开发了一种以低剂量递送TLR激动剂组合的水凝胶,本地,在癌症切除手术期间。使用WEHI164和双侧M3-9-M肉瘤和CT26结肠癌的肿瘤模型,我们评估了成对组合的聚(I:C),R848和CpG在控制局部和远处肿瘤生长中的作用。我们显示TLR3激动剂聚(I:C)和TLR7/8激动剂R848的组合驱动针对局部和远端肿瘤的抗肿瘤免疫。此外,局部聚(I:C)和R848的组合对全身免疫检查点阻断致敏肿瘤,改善肿瘤控制。机械上,我们证明,poly(I:C)和R848的局部治疗在抗肿瘤反应的早期将炎性单核细胞募集到肿瘤引流淋巴结。最后,我们提供了术中通过可手术应用的生物可降解水凝胶一起递送聚(I:C)和R848的概念证明.
    Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.
    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
    在过去的几十年里,开发有效和安全的免疫激活佐剂技术已成为持续对抗高度突变和免疫逃避病毒如流感的深入研究的核心,严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),和人类免疫缺陷病毒(HIV)。在这里,我们开发了一个高度模块化的基于皂苷的纳米颗粒平台,其中包含Toll样受体激动剂(TLRas),包括TLR1/2a,TLR4a,和TLR7/8a佐剂及其混合物。这些不同的TLRa-皂苷纳米佐剂构建体诱导独特的急性细胞因子和免疫信号谱,导致特定的T辅助反应,这可能取决于预防的目标疾病。在一项鼠类疫苗研究中,佐剂大大提高了效力,耐用性,广度,并中和COVID-19和HIV候选疫苗,表明这些佐剂构建体对一系列不同抗原的潜在广泛应用。总的来说,这项工作证明了一个模块化的TLRa-SNP佐剂平台,该平台可以改善疫苗的设计并影响现代疫苗的开发.
    Over the past few decades, the development of potent and safe immune-activating adjuvant technologies has become the heart of intensive research in the constant fight against highly mutative and immune evasive viruses such as influenza, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and human immunodeficiency virus (HIV). Herein, we developed a highly modular saponin-based nanoparticle platform incorporating Toll-like receptor agonists (TLRas) including TLR1/2a, TLR4a, and TLR7/8a adjuvants and their mixtures. These various TLRa-saponin nanoparticle adjuvant constructs induce unique acute cytokine and immune-signaling profiles, leading to specific T helper responses that could be of interest depending on the target disease for prevention. In a murine vaccine study, the adjuvants greatly improved the potency, durability, breadth, and neutralization of both COVID-19 and HIV vaccine candidates, suggesting the potential broad application of these adjuvant constructs to a range of different antigens. Overall, this work demonstrates a modular TLRa-SNP adjuvant platform that could improve the design of vaccines and affect modern vaccine development.
    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
    Toll样受体(TLR)是刺激针对各种感染的免疫应答的关键组分。最近,TLR激动剂已成为激活抗肿瘤免疫的有希望的方法。L-Pampo,TLR1/2和TLR3激动剂,诱导体液和细胞免疫反应,也导致癌细胞死亡。在这项研究中,我们在免疫细胞,结肠癌和前列腺癌细胞中使用RNA-seq研究了L-pampo诱导的信号,并描述了它们与分子信号通路的相互作用.我们首先使用加权基因共表达网络分析构建了一个具有差异表达基因和网络传播影响基因的模板网络。接下来,我们使用上述方法获得了扰动模块,并通过进行Walktrap从其中提取了核心子模块。最后,我们利用最短路径查找算法重建了主要分子信号的子网络,TOPAS.我们的分析表明,L-pampo激活的TLR信号仅在高度表达TLR的免疫和前列腺癌细胞中通过PI3K-AKT和JAK-STAT传递给活性氧(ROS)的氧化磷酸化(OXPHOS)。由于其OXPHOS和ROS的高基础表达水平,该信号流可以进一步使前列腺癌对L-pampo敏感。我们的计算方法可用于从复杂的基因表达谱推断潜在的分子机制。
    Toll-like receptors (TLRs) are critical components to stimulate immune responses against various infections. Recently, TLR agonists have emerged as a promising way to activate anti-tumor immunity. L-pampo, a TLR1/2 and TLR3 agonist, induces humoral and cellular immune responses and also causes cancer cell death. In this study, we investigated the L-pampo-induced signals and delineated their interactions with molecular signaling pathways using RNA-seq in immune cells and colon and prostate cancer cells. We first constructed a template network with differentially expressed genes and influential genes from network propagation using the weighted gene co-expression network analysis. Next, we obtained perturbed modules using the above method and extracted core submodules from them by conducting Walktrap. Finally, we reconstructed the subnetworks of major molecular signals utilizing a shortest path-finding algorithm, TOPAS. Our analysis suggests that TLR signaling activated by L-pampo is transmitted to oxidative phosphorylation (OXPHOS) with reactive oxygen species (ROS) through PI3K-AKT and JAK-STAT only in immune and prostate cancer cells that highly express TLRs. This signal flow may further sensitize prostate cancer to L-pampo due to its high basal expression level of OXPHOS and ROS. Our computational approaches can be applied for inferring underlying molecular mechanisms from complex gene expression profiles.
    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

       PDF(Pubmed)

  • 文章类型: Journal Article
    产前酒精暴露(PAE)的患病率正在增加,有证据表明PAE与感染风险增加有关。PAE被认为会影响先天免疫系统,通过模式识别受体识别病原体,其中Toll样受体(TLRs)是关键成分。我们假设轻度至中度PAE会损害免疫反应,如通过TLR刺激后细胞因子水平的增强反应所测量的。来自乙醇子集的脐带样本(10个对照和8个PAE),神经发育,纳入婴儿和儿童健康研究-2队列。用一种激动剂(TLR2、TLR3、TLR4或TLR9)刺激外周血单核细胞(PMBCs)。TLR2激动剂刺激在24小时后显著增加PAE组中的促炎性白介素-1-β。在用TLR2激动剂刺激后,促炎性和抗炎细胞因子增加。用TLR3或TLR9激动剂刺激显示总体影响最小,但24小时后与PAE相比,对照组的变化百分比显着增加。这项初步研究的结果支持进一步研究PAE后对TLR2和TLR4反应的影响,以确定促炎和抗炎细胞因子水平的变化是否具有可用于患者管理和/或关注随访的临床意义。
    The prevalence of prenatal alcohol exposure (PAE) is increasing, with evidence suggesting that PAE is linked to an increased risk of infections. PAE is hypothesized to affect the innate immune system, which identifies pathogens through pattern recognition receptors, of which toll-like receptors (TLRs) are key components. We hypothesized that light-to-moderate PAE would impair immune responses, as measured by a heightened response in cytokine levels following TLR stimulation. Umbilical cord samples (10 controls and 8 PAE) from a subset of the Ethanol, Neurodevelopment, Infant and Child Health Study-2 cohort were included. Peripheral blood mononuclear cells (PMBCs) were stimulated with one agonist (TLR2, TLR3, TLR4, or TLR9). TLR2 agonist stimulation significantly increased pro-inflammatory interleukin-1-beta in the PAE group after 24 h. Pro- and anti-inflammatory cytokines were increased following stimulation with the TLR2 agonists. Stimulation with TLR3 or TLR9 agonists displayed minimal impact overall, but there were significant increases in the percent change of the control compared to PAE after 24 h. The results of this pilot investigation support further work into the impact on TLR2 and TLR4 response following PAE to delineate if alterations in levels of pro- and anti-inflammatory cytokines have clinical significance that could be used in patient management and/or attention to follow-up.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:小肠是最容易受到电离辐射(IR)损伤的器官之一。然而,防止IR诱导的肠损伤的方法是有限的。CBLB502,一种来自沙门氏菌鞭毛蛋白的Toll样受体5(TLR5)激动剂,对各种组织和器官发挥辐射防护作用。然而,CBLB502对抗IR诱导的肠损伤的分子机制尚不清楚.因此,本研究旨在阐明IR诱导小鼠肠损伤的潜在机制以及CBLB502对这种情况的保护作用。
    方法:在不同时间点,在IR之前以不同剂量给予小鼠0.2mg/kgCBLB502,然后是存活率,体重,血象,并对小鼠进行组织病理学分析。
    结果:CBLB502减少IR诱导的肠损伤。RNA-seq分析显示不同剂量和持续时间的IR诱导不同的调控模式。CBLB502主要通过逆转IR诱导基因的表达并调节免疫过程和代谢途径来保护IR后的肠道损伤。
    结论:本研究初步阐述了CBLB502对IR诱导肠损伤的调控机制和潜在的分子保护机制,为鉴定介导IR诱导肠损伤保护的功能基因和分子机制提供了依据。
    OBJECTIVE: The small intestine is one of the organs most vulnerable to ionizing radiation (IR) damage. However, methods to protect against IR-induced intestinal injury are limited. CBLB502, a Toll-like receptor 5 (TLR5) agonist from Salmonella flagellin, exerts radioprotective effects on various tissues and organs. However, the molecular mechanisms by which CBLB502 protects against IR-induced intestinal injury remain unclear. Thus, this study aimed to elucidate the mechanisms underlying IR-induced intestinal injury and the protective effects of CBLB502 against this condition in mice.
    METHODS: Mice were administered 0.2 mg/kg CBLB502 before IR at different doses for different time points, and then the survival rate, body weight, hemogram, and histopathology of the mice were analyzed.
    RESULTS: CBLB502 reduced IR-induced intestinal injury. RNA-seq analysis revealed that different doses and durations of IR induced different regulatory patterns. CBLB502 protected against intestinal injury mainly after IR by reversing the expression of IR-induced genes and regulating immune processes and metabolic pathways.
    CONCLUSIONS: This study preliminarily describes the regulatory mechanism of IR-induced intestinal injury and the potential molecular protective mechanism of CBLB502, providing a basis for identifying the functional genes and molecular mechanisms that mediate protection against IR-induced injury.
    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
    利什曼病,由利什曼原虫(L.)物种,这是一种被忽视的感染。治疗性疫苗接种为其治疗提供了有希望的策略。在这项研究中,我们的目标是使用包封在聚乳酸-羟基乙酸共聚物(PLGA)纳米颗粒(NP)中的利什曼抗原(SLA)和Toll样受体(TLR)7/8激动剂(R848)开发治疗性疫苗候选物.此外,将TLR1/2激动剂(Pam3CSK4)加载到NP上。在L.major感染的BALB/c小鼠中评价这些NP的治疗效果。脚垫肿胀,寄生虫负荷,细胞和体液免疫反应,分析了一氧化氮(NO)的产生。结果表明,PLGANP(SLA-R848-Pam3CSK4)治疗性疫苗有效刺激Th1细胞反应,诱导的体液反应,促进NO生产,和限制寄生虫负担和病变大小。我们的发现表明,在PLGANP中使用SLA与TLR1/2和TLR7/8激动剂联合接种作为治疗性疫苗可提供对L的强大保护。主要感染。这些结果代表了针对旧世界皮肤利什曼病的新型微粒治疗性疫苗。
    Leishmaniasis, caused by Leishmania (L.) species, remains a neglected infection. Therapeutic vaccination presents a promising strategy for its treatment. In this study, we aimed to develop a therapeutic vaccine candidate using Leishmaniaantigens (SLA) and Toll-like receptor (TLR) 7/8 agonist (R848) encapsulated into the poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). Moreover, TLR1/2 agonist (Pam3CSK4) was loaded onto the NPs. The therapeutic effects of these NPs were evaluated in L. major-infected BALB/c mice. Footpad swelling, parasite load, cellular and humoral immune responses, and nitric oxide (NO) production were analyzed. The results demonstrated that the PLGA NPs (SLA-R848-Pam3CSK4) therapeutic vaccine effectively stimulated Th1 cell responses, induced humoral responses, promoted NO production, and restricted parasite burden and lesion size.Our findings suggest that vaccination with SLA combined with TLR1/2 and TLR7/8 agonists in PLGA NPs as a therapeutic vaccine confers strong protection againstL. majorinfection. These results represent a novel particulate therapeutic vaccine against Old World cutaneous leishmaniasis.
    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
    CD47,一种已知抑制吞噬作用的细胞表面蛋白,在肿瘤微环境(TME)中起着至关重要的作用,并且是癌症的潜在生物标志物。然而,直接应用αCD47,一种靶向CD47的亲水性大分子抗体,在体内用于癌症检测会对正常细胞产生不良影响,引起全身毒性,并导致对抗癌疗法的抵抗。在这项研究中,我们开发了一种新型的配合物,其中包含负载有吲哚菁绿(ICG)的铝基金属有机骨架(Al-MOF),αCD47和瑞喹莫特(R848),疏水性小分子Toll样受体7/8(TLR7/8)激动剂。用红外808nm激光激活后,纳米复合材料表现出光热效应,触发了负载试剂的释放,诱导ROS产生,并诱导TME的变化。这导致免疫抑制M2巨噬细胞向免疫刺激M1表型极化,促进树突状细胞(DC)成熟,并使成熟的DC促进抗原呈递,T细胞激活,以及在肿瘤免疫中的关键作用。此外,体内成像成功检测到肿瘤细胞上αCD47与CD47的特异性结合。总的来说,由αCD47抗体和Toll样受体激动剂组成的复合物在肿瘤诊断和治疗中显示出良好的疗效。为检测早期肺癌和调节肿瘤微环境以改善治疗结果提供潜在策略。
    CD47, a cell surface protein known for inhibiting phagocytosis, plays a critical role in the tumor microenvironment (TME) and is a potential biomarker for cancer. However, directly applying αCD47, a hydrophilic macromolecular antibody that targets CD47, in vivo for cancer detection can have adverse effects on normal cells, cause systemic toxicities, and lead to resistance against anti-cancer therapies. In this study, we developed a novel complex incorporating aluminum-based metal-organic frameworks (Al-MOF) loaded with indocyanine green (ICG), αCD47, and resiquimod (R848), a hydrophobic small molecule Toll-like receptor 7/8 (TLR7/8) agonist. Upon activation with an infrared 808 nm laser, the nanocomposites exhibited photothermal effects that triggered the release of the loaded reagents, induced ROS production, and induced changes in the TME. This led to the polarization of immune-suppressive M2 macrophages towards an immune-stimulatory M1 phenotype, promoted dendritic cell (DC) maturation, and enabled mature DCs to facilitate antigen presentation, T-cell activation, and critical roles in tumor immunity. Furthermore, in vivo imaging successfully detected the specific binding of αCD47 with CD47 on tumor cells. Overall, the complex composed of αCD47 antibody and toll-like receptor agonist showed promising efficacy in both tumor diagnosis and therapy, providing a potential strategy for detecting early lung cancer and modulating the tumor microenvironment for improved treatment outcomes.
    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
    在这项随机II期临床试验中,我们评估了添加TLR激动剂的有效性,聚-ICLC或瑞喹莫特,对新诊断或复发的WHOIII-IV级恶性神经胶质瘤患者进行自体肿瘤裂解物脉冲树突状细胞(ATL-DC)疫苗接种。主要终点是评估疫苗和佐剂的最有效组合,以增强免疫效力,还有安全。ATL-DC疫苗和TLR激动剂的组合是安全的,并且发现可以增强全身免疫反应。如干扰素基因表达增加和免疫细胞活化变化所示。具体来说,PD-1表达在CD4+T细胞上增加,而CD38和CD39在CD8+T细胞上的表达减少,伴随着单核细胞的增加。Poly-ICLC处理放大了单核细胞和T淋巴细胞中干扰素诱导的基因的诱导。表现出较高干扰素应答基因表达的患者表现出延长的生存期和延迟的疾病进展。这些发现表明,ATL-DC与poly-ICLC的结合可以诱导循环单核细胞和CD8+T细胞的极化干扰素应答,这可能是该患者人群中免疫疗法的重要血液生物标志物。试用注册:ClinicalTrials.gov标识符:NCT01204684。
    In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.
    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:T细胞检查点受体在T细胞被激活时表达,和这些受体的表达或信号的调节可以改变T细胞的功能及其抗肿瘤功效。我们以前发现,用同源抗原激活的T细胞在PD-1的表达中增加,并且在存在多种toll样受体(TLR)激动剂的情况下,特别是TLR3加上TLR9。在当前的报告中,我们试图研究在小鼠肿瘤模型中,将TLR激动剂与免疫检查点阻断联合使用是否可进一步增强疫苗介导的T细胞抗肿瘤免疫力.
    方法:将TLR激动剂(TLR3加TLR9)和免疫检查点抑制剂(靶向PD-1,CTLA-4,LAG-3,TIM-3或VISTA的抗体)组合并与疫苗或疫苗激活的CD8T细胞一起递送至E.G7-OVA或MyC-CaP荷瘤小鼠。评估肿瘤的生长,然后收集并通过流式细胞术分析。
    结果:用SIINFEKL肽疫苗免疫E.G7-OVA荷瘤小鼠,与TLR激动剂和αCTLA-4共同给药,证明了比单独用TLR激动剂或αCTLA-4免疫更大的抗肿瘤功效。相反,当疫苗和TLR激动剂与αPD-1联合使用时,抗肿瘤功效被取消.TLR激动剂抑制调节性T细胞(Tregs)上的PD-1表达并激活该群体。Tregs在荷瘤小鼠中的消耗导致这种联合疗法的更大抗肿瘤功效,即使存在αPD-1。与TLR激动剂和αCTLA-4或αLAG-3的组合疫苗接种比与αTIM-3或αVISTA的组合显示更大的抗肿瘤。
    结论:TLR激动剂和αCTLA-4或αLAG-3的组合可以进一步提高癌症疫苗的功效,当αPD-1与TLR3和TLR9激动剂组合时,由于Treg的激活,使用αPD-1未观察到的效果。这些数据表明TLR激动剂和免疫检查点阻断的最佳组合可以提高人抗癌疫苗的功效。
    BACKGROUND: T cell checkpoint receptors are expressed when T cells are activated, and modulation of the expression or signaling of these receptors can alter the function of T cells and their antitumor efficacy. We previously found that T cells activated with cognate antigen had increases in the expression of PD-1, and this was attenuated in the presence of multiple toll-like receptor (TLR) agonists, notably TLR3 plus TLR9. In the current report, we sought to investigate whether combining TLR agonists with immune checkpoint blockade can further augment vaccine-mediated T cell antitumor immunity in murine tumor models.
    METHODS: TLR agonists (TLR3 plus TLR9) and immune checkpoint inhibitors (antibodies targeting PD-1, CTLA-4, LAG-3, TIM-3 or VISTA) were combined and delivered with vaccines or vaccine-activated CD8+T cells to E.G7-OVA or MyC-CaP tumor-bearing mice. Tumors were assessed for growth and then collected and analyzed by flow cytometry.
    RESULTS: Immunization of E.G7-OVA tumor-bearing mice with SIINFEKL peptide vaccine, coadministered with TLR agonists and αCTLA-4, demonstrated greater antitumor efficacy than immunization with TLR agonists or αCTLA-4 alone. Conversely, the antitumor efficacy was abrogated when vaccine and TLR agonists were combined with αPD-1. TLR agonists suppressed PD-1 expression on regulatory T cells (Tregs) and activated this population. Depletion of Tregs in tumor-bearing mice led to greater antitumor efficacy of this combination therapy, even in the presence of αPD-1. Combining vaccination with TLR agonists and αCTLA-4 or αLAG-3 showed greater antitumor than with combinations with αTIM-3 or αVISTA.
    CONCLUSIONS: The combination of TLR agonists and αCTLA-4 or αLAG-3 can further improve the efficacy of a cancer vaccine, an effect not observed using αPD-1 due to activation of Tregs when αPD-1 was combined with TLR3 and TLR9 agonists. These data suggest that optimal combinations of TLR agonists and immune checkpoint blockade may improve the efficacy of human anticancer vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号