Anti-tumor immunity

抗肿瘤免疫
  • 文章类型: Journal Article
    最近的研究已经将升高的肿瘤非整倍体与抗肿瘤免疫抑制和免疫疗法后的不良存活联系起来。在这里,我们为肿瘤非整倍性作为非小细胞肺癌(NSCLC)患者免疫治疗反应的生物标志物提供了支持性证据.我们确定了非整倍体评分与患者预后之间的剂量反应关系。在两个独立的NSCLC队列(n=659例)中,我们证明了非整倍体升高与非吸烟相关的致癌驱动突变之间的新关联.最后,我们报道了高度非整倍体NSCLC的TERT扩增和免疫抑制表型的富集。一起来看,我们的研究结果强调了肿瘤非整倍体在指导免疫治疗策略方面的潜在关键作用.
    Recent studies have linked elevated tumor aneuploidy to anti-tumor immune suppression and adverse survival following immunotherapy. Herein, we provide supportive evidence for tumor aneuploidy as a biomarker of response to immunotherapy in patients with non-small cell lung cancer (NSCLC). We identify a dose-response relationship between aneuploidy score and patient outcomes. In two independent NSCLC cohorts (n = 659 patients), we demonstrate a novel association between elevated aneuploidy and non-smoking-associated oncogenic driver mutations. Lastly, we report enrichment of TERT amplification and immune-suppressive phenotypes of highly aneuploid NSCLC. Taken together, our findings emphasize a potentially critical role for tumor aneuploidy in guiding immunotherapy treatment strategies.
    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
    克服免疫介导的对PD-1阻断的抗性仍然是主要的临床挑战。在联合使用nivolumab(抗PD-1)和relatlimab(抗LAG-3)治疗的黑色素瘤患者中,已证明疗效增强。这是同类产品中第一个获得FDA批准的。然而,这两种抑制性受体如何协同作用以阻碍抗肿瘤免疫仍然未知。这里,我们显示,CD8+T细胞缺乏PD-1和LAG-3,与缺乏任一受体的CD8+T细胞相反,在黑色素瘤小鼠模型中介导增强的肿瘤清除和长期存活。PD-1-和LAG-3缺陷型CD8+T细胞在转录上不同,具有广泛的TCR克隆性和效应物样和干扰素反应基因的富集,导致增强的IFN-γ释放指示功能性。LAG-3和PD-1联合驱动T细胞耗尽,在调节TOX表达中起主导作用。机械上,自分泌,PD-1-和LAG-3-缺陷的CD8+T细胞需要细胞固有的IFN-γ信号传导来增强抗肿瘤免疫力,深入了解LAG-3和PD-1的组合靶向如何增强疗效。
    Overcoming immune-mediated resistance to PD-1 blockade remains a major clinical challenge. Enhanced efficacy has been demonstrated in melanoma patients with combined nivolumab (anti-PD-1) and relatlimab (anti-LAG-3) treatment, the first in its class to be FDA approved. However, how these two inhibitory receptors synergize to hinder anti-tumor immunity remains unknown. Here, we show that CD8+ T cells deficient in both PD-1 and LAG-3, in contrast to CD8+ T cells lacking either receptor, mediate enhanced tumor clearance and long-term survival in mouse models of melanoma. PD-1- and LAG-3-deficient CD8+ T cells were transcriptionally distinct, with broad TCR clonality and enrichment of effector-like and interferon-responsive genes, resulting in enhanced IFN-γ release indicative of functionality. LAG-3 and PD-1 combined to drive T cell exhaustion, playing a dominant role in modulating TOX expression. Mechanistically, autocrine, cell-intrinsic IFN-γ signaling was required for PD-1- and LAG-3-deficient CD8+ T cells to enhance anti-tumor immunity, providing insight into how combinatorial targeting of LAG-3 and PD-1 enhances efficacy.
    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
    肿瘤浸润淋巴细胞(TIL)功能减退有助于晚期癌症的进展,并且是免疫疗法的常见靶标。新的证据表明,在强直刺激期间,代谢不足会导致T细胞功能减退,但是在这种情况下启动代谢重编程的信号在很大程度上是未知的。这里,我们发现类似Meteorin(METRNL),肿瘤微环境(TME)中免疫细胞分泌的代谢活性细胞因子,诱导CD8+T细胞的生物能量衰竭。METRNL在重复刺激期间由CD8+T细胞分泌,并且经由自分泌和旁分泌信号传导两者起作用。机械上,METRNL增加E2F-过氧化物酶体增殖物激活受体δ(PPARδ)活性,导致线粒体去极化和减少的氧化磷酸化,这引发了补偿性生物能量向糖酵解的转变。Metrnl消融或下调改善了CD8+T细胞的代谢适应性,并增强了几种肿瘤模型中的肿瘤控制,证明了靶向METRNL-E2F-PPARδ途径以支持CD8TIL的生物能量适应性的翻译潜力。
    Tumor-infiltrating lymphocyte (TIL) hypofunction contributes to the progression of advanced cancers and is a frequent target of immunotherapy. Emerging evidence indicates that metabolic insufficiency drives T cell hypofunction during tonic stimulation, but the signals that initiate metabolic reprogramming in this context are largely unknown. Here, we found that Meteorin-like (METRNL), a metabolically active cytokine secreted by immune cells in the tumor microenvironment (TME), induced bioenergetic failure of CD8+ T cells. METRNL was secreted by CD8+ T cells during repeated stimulation and acted via both autocrine and paracrine signaling. Mechanistically, METRNL increased E2F-peroxisome proliferator-activated receptor delta (PPARδ) activity, causing mitochondrial depolarization and decreased oxidative phosphorylation, which triggered a compensatory bioenergetic shift to glycolysis. Metrnl ablation or downregulation improved the metabolic fitness of CD8+ T cells and enhanced tumor control in several tumor models, demonstrating the translational potential of targeting the METRNL-E2F-PPARδ pathway to support bioenergetic fitness of CD8+ TILs.
    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
    凭借其独特的性能和潜在的应用,近年来,用于信使RNA(mRNA)疫苗的基于纳米颗粒的递送平台获得了显著的关注。纳米颗粒具有增强免疫原性的优点,靶向递送,提高稳定性,为药物和疫苗提供新的解决方案。在一些临床研究中,各种纳米颗粒递送平台已逐步应用于广泛的疫苗应用。当前的研究重点是探索各种类型的纳米颗粒作为疫苗递送系统以增强疫苗稳定性和免疫原性。脂质纳米颗粒(LNP)在将抗原有效递送至免疫细胞的临床前和临床研究中显示出有希望的潜力。此外,脂质纳米颗粒和其他用于核酸的纳米颗粒,特别是mRNA递送系统,已经显示出疫苗开发的巨大潜力。在这次审查中,我们提出了各种疫苗平台,重点是纳米颗粒作为mRNA疫苗递送载体。我们描述了几种用于mRNA疫苗的新型纳米颗粒递送平台,例如脂质-,聚合物-,和基于蛋白质的纳米颗粒。此外,我们概述了纳米疫苗在癌症免疫治疗中对不同肿瘤的抗肿瘤免疫。最后,我们概述了这种有前途的基于纳米颗粒的疫苗递送平台技术的未来前景和仍然存在的挑战.
    With its unique properties and potential applications, nanoparticle-based delivery platforms for messenger RNA (mRNA) vaccines have gained significant attention in recent years. Nanoparticles have the advantages of enhancing immunogenicity, targeting delivery, and improving stability, providing a new solution for drug and vaccine delivery. In some clinical studies, a variety of nanoparticle delivery platforms have been gradually applied to a wide range of vaccine applications. Current research priorities are exploring various types of nanoparticles as vaccine delivery systems to enhance vaccine stability and immunogenicity. Lipid nanoparticles (LNPs) have shown promising potential in preclinical and clinical studies on the efficient delivery of antigens to immune cells. Moreover, lipid nanoparticles and other nanoparticles for nucleic acids, especially for mRNA delivery systems, have shown vast potential for vaccine development. In this review, we present various vaccine platforms with an emphasis on nanoparticles as mRNA vaccine delivery vehicles. We describe several novel nanoparticle delivery platforms for mRNA vaccines, such as lipid-, polymer-, and protein-based nanoparticles. In addition, we provide an overview of the anti-tumor immunity of nanovaccines against different tumors in cancer immunotherapy. Finally, we outline future perspectives and remaining challenges for this promising technology of nanoparticle-based delivery platforms for vaccines.
    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:在三阴性乳腺癌(TNBC)治疗中,淋巴细胞浸润不足会显著阻碍免疫检查点抑制剂的疗效.我们以前已经证明海纳宁-1(HN-1),从海南青蛙皮肤中鉴定出的宿主防御肽(HDP),诱导乳腺癌细胞凋亡,并通过未知的机制引导抗肿瘤免疫。
    方法:我们使用体外实验观察HN-1处理的TNBC细胞系中的免疫原性细胞死亡(ICD)指标,小鼠肿瘤模型验证HN-1促进小鼠抗肿瘤免疫应答,并对患者来源的乳腺癌细胞进行体外药敏试验,以验证HN-1的抑制作用。
    结果:HN-1在TNBC中诱导ICD,在此期间释放了损伤相关分子模式(DAMPs),可以进一步增加抗肿瘤免疫反应。白细胞介素2(IL-2)的分泌水平,IL-12和干扰素γ在共培养上清液中增加,和树突细胞(DC)通过与HN-1预处理的TNBC细胞共培养而被激活。因此,HN-1增加了携带4T1和EMT6肿瘤的小鼠模型中抗肿瘤免疫细胞(DC和T淋巴细胞)的浸润。同时,调节性T细胞和骨髓来源的抑制细胞受到抑制。此外,HN-1诱导DNA损伤,胞质溶胶中的双链DNA释放显着增强,表明HN-1可能通过激活STING途径刺激ICD。STING的敲低抑制HN-1诱导的ICD。值得注意的是,在三维培养条件下,HN-1对患者来源的乳腺癌细胞表现出抑制作用。
    结论:总的来说,我们的研究表明,HN-1可作为一种潜在化合物,可增强TNBC患者的免疫治疗效果.
    BACKGROUND: In triple-negative breast cancer (TNBC) therapy, insufficient tumor infiltration by lymphocytes significantly hinders the efficacy of immune checkpoint inhibitors. We have previously demonstrated that Hainanenin-1 (HN-1), a host defense peptide (HDP) identified from Hainan frog skin, induces breast cancer apoptosis and boots anti-tumor immunity via unknown mechanism.
    METHODS: We used in vitro experiments to observe immunogenic cell death (ICD) indicators in HN-1-treated TNBC cell lines, a mouse tumor model to verify HN-1 promotion of mice anti-tumor immune response, and an in vitro drug sensitivity test of patient-derived breast cancer cells to verify the inhibitory effect of HN-1.
    RESULTS: HN-1 induced ICD in TNBC in a process during which damage-associated molecular patterns (DAMPs) were released that could further increase the anti-tumor immune response. The secretion level of interleukin 2 (IL-2), IL-12, and interferon γ in the co-culture supernatant was increased, and dendritic cells (DCs) were activated via a co-culture with HN-1-pretreated TNBC cells. As a result, HN-1 increased the infiltration of anti-tumor immune cells (DCs and T lymphocytes) in the mouse model bearing both 4T1 and EMT6 tumors. Meanwhile, regulatory T cells and myeloid-derived suppressor cells were suppressed. In addition, HN-1 induced DNA damage, and double-strand DNA release in the cytosol was significantly enhanced, indicating that HN-1 might stimulate ICD via activation of STING pathway. The knockdown of STING inhibited HN-1-induced ICD. Of note, HN-1 exhibited inhibitory effects on patient-derived breast cancer cells under three-dimensional culture conditions.
    CONCLUSIONS: Collectively, our study demonstrated that HN-1 could be utilized as a potential compound that might augment immunotherapy effects in patients with TNBC.
    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
    T细胞受体(TCR)发出的信号,共刺激受体,和细胞因子受体各自影响CD8T细胞命运。了解这些信号对稳态和微环境线索的反应可以揭示治疗性指导T细胞功能的新方法。通过小鼠的正向遗传筛查,我们发现LDL受体相关蛋白10(Lrp10)的功能缺失突变导致幼稚和中枢记忆性CD8T细胞在外周淋巴器官中积累.Lrp10编码免疫功能未知的保守细胞表面蛋白。T细胞活化诱导Lrp10表达,它在翻译后抑制IL7受体(IL7R)水平。因此,Lrp10缺失通过IL7R信号增强T细胞稳态扩增。Lrp10缺陷型小鼠也对同基因肿瘤具有内在抗性。这种表型取决于CD8T细胞的致密肿瘤浸润,显示出增强的存储单元特性,减少终端疲惫,和增强对免疫检查点抑制的反应。这里,我们提出Lrp10作为CD8T细胞稳态的一种新的负调节因子和一种控制肿瘤耐药的宿主因子,对免疫治疗有意义.
    Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for 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: Systematic Review
    近年来,由于高死亡率和令人沮丧的五年生存率,治疗非小细胞肺癌(NSCLC)的重要性日益增加。免疫检查点抑制剂(ICI)是一种有前途的方法,由于细胞的抗原性,在NSCLC中具有出色的结果。相反,ICI对免疫系统的过度刺激是一把双刃剑,可导致从轻度到危及生命的各种负面影响。这篇综述探讨了基于纳米粒子的ICI的当前突破及其局限性。PubMed,检查了Scopus和WebofScience的相关出版物。分析中包括38项试验(N=16,781)。量化治疗效果的混合效果分析对ICI治疗效果研究中的亚组做出了显着贡献。模型证实,与常规治疗方案相比,ICI对治疗效果的影响更大,且降低了受访者的死亡率。由于ICI已被证明的有效性和安全性,ICI可能被用作一线治疗。
    Treating non-small-cell lung cancer (NSCLC) has gained increased importance in recent years due to the high mortality rate and dismal five-year survival rate. Immune checkpoint inhibitors (ICI) are a promising approach with exceptional outcomes in NSCLC thanks to the antigenic nature of cells. Conversely, immune system over-stimulation with ICI is a double-edged sword that can lead to various negative effects ranging from mild to life-threatening. This review explores current breakthroughs in nanoparticle-based ICI and their limitations. The PubMed, Scopus and Web of Science were examined for relevant publications. Thirty-eight trials (N = 16,781) were included in the analyses. The mixed effects analyses on quantifying the treatment effect contributed significantly to the subgroups within studies for ICI treatment effect. Models confirmed ICI\'s higher impact on treatment effectivity and the decrease in respondents\' mortality compared to conventional treatment regiments. ICI might be used as first-line therapy due to their proven effectiveness and safety profile.
    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
    化疗和免疫治疗对不可切除的胰腺导管腺癌(PDAC)没有显着结果。多药联合治疗已成为临床试验的共识,以探索如何激发抗肿瘤免疫力,同时克服杀瘤效果差和极大阻碍药物渗透的基质屏障。为了应对这一挑战,提出了一种全面的策略,即充分利用PDAC的铁毒性来有效刺激抗肿瘤免疫,并通过在可注射的水凝胶中持续释放药物来完全改善免疫抑制微环境,以增加药物在肿瘤位置的渗透并避免系统毒性。可注射水凝胶ED-M@CS/MC与装载有专门诱导铁凋亡的erastin和抑制基质形成的FAK抑制剂defactinib的胶束杂交,并实现药物持续释放长达12天。只需单次肿瘤内注射,在异种移植和KrasG12D工程化的原代PDAC小鼠中,erastin和defactinib的联合治疗可产生进一步的抗肿瘤效果,并协同促进CD8+细胞毒性T细胞的浸润和II型巨噬细胞的减少.这些发现可能为PDAC的临床治疗提供新的有希望的策略。
    Chemotherapy and immunotherapy have shown no significant outcome for unresectable pancreatic ductal adenocarcinoma (PDAC). Multi-drug combination therapy has become a consensus in clinical trials to explore how to arouse anti-tumor immunity and meanwhile overcome the poorly tumoricidal effect and the stroma barrier that greatly hinders drug penetration. To address this challenge, a comprehensive strategy is proposed to fully utilize both the ferroptotic vulnerability of PDAC to potently irritate anti-tumor immunity and the desmoplasia-associated focal adhesion kinase (FAK) to wholly improve the immunosuppressive microenvironment via sustained release of drugs in an injectable hydrogel for increasing drug penetration in tumor location and averting systematic toxicity. The injectable hydrogel ED-M@CS/MC is hybridized with micelles loaded with erastin that exclusively induces ferroptosis and a FAK inhibitor defactinib for inhibiting stroma formation, and achieves sustained release of the drugs for up to 12 days. With only a single intratumoral injection, the combination treatment with erastin and defactinib produces further anti-tumor performance both in xenograft and KrasG12D-engineered primary PDAC mice and synergistically promotes the infiltration of CD8+ cytotoxic T cells and the reduction of type II macrophages. The findings may provide a novel promising strategy for the clinical treatment of PDAC.
    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:端粒酶,通过保护受损的端粒和支持DNA损伤修复,有能力提高肿瘤细胞的抗辐射能力。因此,反过来,可能会损害放射治疗(RT)和放射免疫治疗的疗效。我们以前的研究表明,高选择性端粒酶抑制剂,BIBR1532具有增强非小细胞肺癌(NSCLC)的放射敏感性的潜力。在这项研究中,我们进一步探讨了BIBR1532对RT诱导的免疫激活的影响,并阐明了其潜在机制。
    方法:生物信息分析,免疫荧光测定,蛋白质印迹试验,流式细胞术分析阐明BIBR1532联合放疗在NSCLC中的作用.脂质过氧化物的细胞内水平,谷胱甘肽,丙二醛,和Fe2+作为铁凋亡状态的指标。进行了体外和体内研究以检查抗肿瘤作用。
    结果:我们的发现表明,BIBR1532与RT的汇合显着增强了体内和体外环境中cGAS-STING途径的激活,从而促进有效的抗肿瘤免疫反应。这些影响可以归因于两个关键过程。首先,电离辐射,在沉淀DNA双链断裂(DSB)中,促使肿瘤衍生的双链DNA(dsDNA)释放到细胞质中。随后,BIBR1532在RT后通过dsDNA扩增抗原呈递细胞的活化并激发cGAS-STING途径。其次,BIBR1532增强RT后NSCLC的铁凋亡反应,从而促进肿瘤细胞内未受约束的脂质过氧化和活性氧(ROS)水平升高。这最终导致线粒体应激和内源性线粒体DNA(mtDNA)释放到细胞质中,从而促进STING途径的激活和I型干扰素(IFN)相关的适应性免疫应答的诱导。
    结论:本研究强调了BIBR1532作为一种有效和安全的放射增敏剂和放射免疫疗法增效剂的潜力,为NSCLC的治疗提供有力的临床前研究证据。
    BACKGROUND: Telomerase, by safeguarding damaged telomeres and bolstering DNA damage repair, has the capacity to heighten the radioresistance of tumour cells. Thus, in turn, can compromise the efficacy of radiotherapy (RT) and radioimmunotherapy. Our previous studies have revealed that the highly selective telomerase inhibitor, BIBR1532, possesses the potential to enhance the radiosensitivity of Non-small cell lung cancer (NSCLC). In this study, we delve further into the impact of BIBR1532 on the immune activation induced by RT and elucidate the underlying mechanisms.
    METHODS: Biological information analyses, immunofluorescence assays, western blot assays, flow cytometry analysis were conducted to elucidate the functions of the combination of BIBR1532 with radiotherapy in NSCLC. Intracellular levels of lipid peroxides, glutathione, malondialdehyde, and Fe2+ were measured as indicators of ferroptosis status. Both in vitro and in vivo studies were conducted to examine the antitumor effects.
    RESULTS: Our findings indicate that the confluence of BIBR1532 with RT significantly augments the activation of the cGAS-STING pathway in both in vivo and in vitro settings, thereby fostering an effective anti-tumoral immune response. The effects can be ascribed to two key processes. Firstly, ionizing radiation, in precipitating DNA double-strand breaks (DSBs), prompts the release of tumour-derived double-stranded DNA (dsDNA) into the cytoplasm. Subsequently, BIBR1532 amplifies the activation of antigen-presenting cells by dsDNA post-RT and instigates the cGAS-STING pathway. Secondly, BIBR1532 enhances the ferroptosis response in NSCLC following RT, thereby promoting unrestrained lipid peroxidation and elevated levels of reactive oxygen species (ROS) within tumour cells. This ultimately leads to mitochondrial stress and the release of endogenous mitochondrial DNA (mtDNA) into the cytoplasm, thus facilitating the activation of the STING pathway and the induction of a type I interferon (IFN)-linked adaptive immune response.
    CONCLUSIONS: This study underscores the potential of BIBR1532 as an efficacious and safe radiosensitizer and radioimmunotherapy synergist, providing robust preclinical research evidence for the treatment of NSCLC.
    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
    乳酸乳球菌亚种。cremorisC60是乳酸菌(LAB)的益生菌菌株,可在骨髓谱系细胞中诱导各种免疫修饰。这些修饰随后调节T细胞功能,导致局部和全身免疫力增强。这里,我们报道C60通过代谢改变增强巨噬细胞功能来抑制肿瘤生长,从而增加鼠黑素瘤模型中的三磷酸腺苷(ATP)产生。与小鼠中的盐水施用相比,C60的胃内(i.g.)施用显著减小了肿瘤体积。在施用C60的小鼠中,肿瘤内(IT)巨噬细胞的抗肿瘤功能上调,如炎症表型(M1)而不是抗炎/修复(M2)表型增加所证明的,随着抗原呈递能力的增强,导致肿瘤抗原特异性CD8+T细胞增加。通过这种功能修改,我们发现C60建立了糖酵解主导的代谢,而不是脂肪酸氧化(粮农组织),在IT巨噬细胞中,导致细胞内ATP水平增加。为了解决为什么口服补充的C60在远端显示功能的问题,我们发现细菌细胞壁成分的可能性,它可以从肠道分布在全身,可能通过Toll样受体(TLR)信号激活在外周巨噬细胞中诱导刺激信号。因此,C60通过在TLR介导的刺激下促进向糖酵解的主要代谢转变来增强巨噬细胞抗肿瘤免疫力,从而增加了大量的能源生产。
    Lactococcus lactis subsp. cremoris C60 is a probiotic strain of lactic acid bacteria (LAB) which induces various immune modifications in myeloid lineage cells. These modifications subsequently regulate T cell function, resulting in enhanced immunity both locally and systemically. Here, we report that C60 suppresses tumor growth by enhancing macrophage function via metabolic alterations, thereby increasing adenosine triphosphate (ATP) production in a murine melanoma model. Intragastric (i.g.) administration of C60 significantly reduced tumor volume compared to saline administration in mice. The anti-tumor function of intratumor (IT) macrophage was upregulated in mice administered with C60, as evidenced by an increased inflammatory phenotype (M1) rather than an anti-inflammatory/reparative (M2) phenotype, along with enhanced antigen-presenting ability, resulting in increased tumor antigen-specific CD8+ T cells. Through this functional modification, we identified that C60 establishes a glycolysis-dominant metabolism, rather than fatty acid oxidation (FAO), in IT macrophages, leading to increased intracellular ATP levels. To address the question of why orally supplemented C60 exhibits functions in distal places, we found a possibility that bacterial cell wall components, which could be distributed throughout the body from the gut, may induce stimulatory signals in peripheral macrophages via Toll-like receptors (TLRs) signaling activation. Thus, C60 strengthens macrophage anti-tumor immunity by promoting a predominant metabolic shift towards glycolysis upon TLR-mediated stimulation, thereby increasing substantial energy production.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号