Anti-tumor immunity

抗肿瘤免疫
  • 文章类型: Journal Article
    整合焦亡和铁凋亡混合细胞死亡诱导以增强免疫激活代表了抗肿瘤治疗的有希望的途径。但是缺乏研究。在这里,我们开发了两种铱(III)-三苯胺光敏剂,IrC和IrF,具有破坏氧化还原平衡并诱导对DNA和Kelch样ECH相关蛋白1(KEAP1)的光驱动级联损伤的能力。在黑色素瘤2(AIM2)相关的细胞质核酸传感途径中缺失的激活,由受损的DNA引发,导致GasderminD(GSDMD)介导的焦亡的诱导。同时,铁稳态,受KEAP1/核因子红系2相关因子2(NRF2)/血红素加氧酶1(HO-1)途径调节,作为一个关键的桥梁,不仅促进gasderminE(GSDME)介导的非规范焦亡的诱导,但也与谷胱甘肽过氧化物酶4(GPX4)消耗协同作用。焦亡和铁凋亡的协同作用产生协同作用,引起免疫原性细胞死亡,在体内刺激强大的免疫反应并有效抑制肿瘤生长。我们的工作介绍了第一个金属为基础的小分子双诱导焦亡和铁凋亡的有效的癌症免疫治疗,并强调了铁稳态作为连接焦凋亡和铁凋亡协同作用的重要枢纽的重要性。
    The integration of pyroptosis and ferroptosis hybrid cell death induction to augment immune activation represents a promising avenue for anti-tumor treatment, but there is a lack of research. Herein, we developed two iridium(III)-triphenylamine photosensitizers, IrC and IrF, with the capacity to disrupt redox balance and induce photo-driven cascade damage to DNA and Kelch-like ECH-associated protein 1 (KEAP1). The activation of the absent in melanoma 2 (AIM2)-related cytoplasmic nucleic acid-sensing pathway, triggered by damaged DNA, leads to the induction of gasdermin D (GSDMD)-mediated pyroptosis. Simultaneously, iron homeostasis, regulated by the KEAP1/nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase 1 (HO-1) pathway, serves as a pivotal bridge, facilitating not only the induction of gasdermin E (GSDME)-mediated non-canonical pyroptosis, but also ferroptosis in synergy with glutathione peroxidase 4 (GPX4) depletion. The collaborative action of pyroptosis and ferroptosis generates a synergistic effect that elicits immunogenic cell death, stimulates a robust immune response and effectively inhibits tumor growth in vivo. Our work introduces the first metal-based small molecule dual-inducers of pyroptosis and ferroptosis for potent cancer immunotherapy, and highlights the significance of iron homeostasis as a vital hub connecting synergistic effects of pyroptosis and ferroptosis.
    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
    越来越多的证据表明,免疫疗法受到敌对肿瘤微环境(TME)的阻碍,其特征是缺乏关键营养素和免疫抑制代谢物的汇集。肿瘤细胞和免疫抑制细胞在必需营养素方面胜过免疫效应细胞。同时,广泛的肿瘤细胞源性毒性代谢产物对抗肿瘤免疫反应产生负面影响,降低免疫疗法的疗效。具有优异靶向性的纳米医学提供了一种通过代谢重编程免疫抑制TME来改善癌症免疫治疗的新方法。在这里,我们回顾了通过纳米医学重新连接肿瘤代谢来增强免疫治疗效果的最新策略。通过纳米医学提请注意TME中免疫细胞和基质细胞的免疫代谢策略。此外,我们讨论了未来发展代谢调节纳米药物以实现精确和有效的癌症免疫治疗的方向。
    Increasing evidence indicates that immunotherapy is hindered by a hostile tumor microenvironment (TME) featured with deprivation of critical nutrients and pooling of immunosuppressive metabolites. Tumor cells and immunosuppressive cells outcompete immune effector cells for essential nutrients. Meanwhile, a wide range of tumor cell-derived toxic metabolites exerts negative impacts on anti-tumor immune response, diminishing the efficacy of immunotherapy. Nanomedicine with excellent targetability offers a novel approach to improving cancer immunotherapy via metabolically reprogramming the immunosuppressive TME. Herein, we review recent strategies of enhancing immunotherapeutic effects through rewiring tumor metabolism via nanomedicine. Attention is drawn on immunometabolic tactics for immune cells and stromal cells in the TME via nanomedicine. Additionally, we discuss future directions of developing metabolism-regulating nanomedicine for precise and efficacious cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:使用PD-L1阻断的免疫治疗仅对一小部分癌症患者有效,抵抗是常见的。这强调了了解癌症免疫逃避和抵抗机制的重要性。
    方法:基因组规模的CRISPR-Cas9筛选将Bap1鉴定为PD-L1表达的调节因子。为了测量肿瘤大小和存活率,将肿瘤细胞皮下注射到同基因WT小鼠和免疫受损小鼠中。使用流式细胞术检查Bap1缺失肿瘤的表型和转录特征,RNA-seq,以及CUT和Tag-seq分析。
    结果:我们发现癌细胞中组蛋白去泛素酶Bap1的丢失激活了cDC1-CD8+T细胞依赖性抗肿瘤免疫。Bap1的缺失导致与抗肿瘤免疫应答相关的基因增加和与免疫逃避相关的基因减少。因此,肿瘤微环境发炎,更多的cDC1细胞和效应CD8+T细胞,但中性粒细胞和调节性T细胞较少。我们还发现,Bap1缺失肿瘤的消除取决于肿瘤MHCI分子和Fas介导的CD8T细胞毒性。我们对TCGA数据的分析进一步支持了这些发现,显示在各种人类癌症中BAP1表达和活化DC的mRNA签名与T细胞的细胞毒性之间的反向相关性。
    结论:组蛋白去泛素酶Bap1可用作肿瘤分层的生物标志物,并作为癌症免疫治疗的潜在治疗靶点。
    OBJECTIVE: Immunotherapy using PD-L1 blockade is effective in only a small group of cancer patients, and resistance is common. This emphasizes the importance of understanding the mechanisms of cancer immune evasion and resistance.
    METHODS: A genome-scale CRISPR-Cas9 screen identified Bap1 as a regulator of PD-L1 expression. To measure tumor size and survival, tumor cells were subcutaneously injected into both syngeneic WT mice and immunocompromised mice. The phenotypic and transcriptional characteristics of Bap1-deleted tumors were examined using flow cytometry, RNA-seq, and CUT&Tag-seq analysis.
    RESULTS: We found that loss of histone deubiquitinase Bap1 in cancer cells activates a cDC1-CD8+ T cell-dependent anti-tumor immunity. The absence of Bap1 leads to an increase in genes associated with anti-tumor immune response and a decrease in genes related to immune evasion. As a result, the tumor microenvironment becomes inflamed, with more cDC1 cells and effector CD8+ T cells, but fewer neutrophils and regulatory T cells. We also found that the elimination of Bap1-deleted tumors depends on the tumor MHCI molecule and Fas-mediated CD8+ T cell cytotoxicity. Our analysis of TCGA data further supports these findings, showing a reverse correlation between BAP1 expression and mRNA signatures of activated DCs and T-cell cytotoxicity in various human cancers.
    CONCLUSIONS: The histone deubiquitinase Bap1 could be used as a biomarker for tumor stratification and as a potential therapeutic target for cancer 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
    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
    脑肿瘤如成胶质细胞瘤对免疫检查点阻断治疗有抗性,主要是由于肿瘤中有限的T细胞浸润。这里,我们显示,小鼠患有颅内肿瘤表现出系统性免疫抑制和T细胞在骨髓隔离,导致脑肿瘤中T细胞浸润减少。在荷瘤小鼠中,升高的血浆皮质酮通过糖皮质激素受体驱动T细胞隔离。由糖皮质激素诱导的T细胞动力学介导的免疫抑制和随后的肿瘤生长促进可以通过肾上腺切除术来消除。糖皮质激素激活抑制剂或糖皮质激素受体拮抗剂的给药,和T细胞特异性糖皮质激素受体缺失的小鼠。T细胞中的CCR8表达在荷瘤小鼠中以糖皮质激素受体依赖性方式增加。此外,趋化因子CCL1和CCL8是CCR8的配体,在荷瘤小鼠的骨髓免疫细胞中高度表达以募集T细胞。这些结果表明,脑肿瘤诱导的糖皮质激素激增和T细胞中的CCR8上调导致骨髓中的T细胞螯合,损害抗肿瘤免疫反应。靶向糖皮质激素受体-CCR8轴可能为颅内肿瘤的治疗提供有希望的免疫治疗方法。
    Brain tumors such as glioblastomas are resistant to immune checkpoint blockade therapy, largely due to limited T cell infiltration in the tumors. Here, we show that mice bearing intracranial tumors exhibit systemic immunosuppression and T cell sequestration in bone marrow, leading to reduced T cell infiltration in brain tumors. Elevated plasma corticosterone drives the T cell sequestration via glucocorticoid receptors in tumor-bearing mice. Immunosuppression mediated by glucocorticoid-induced T cell dynamics and the subsequent tumor growth promotion can be abrogated by adrenalectomy, the administration of glucocorticoid activation inhibitors or glucocorticoid receptor antagonists, and in mice with T cell-specific deletion of glucocorticoid receptor. CCR8 expression in T cells is increased in tumor-bearing mice in a glucocorticoid receptor-dependent manner. Additionally, chemokines CCL1 and CCL8, the ligands for CCR8, are highly expressed in bone marrow immune cells in tumor-bearing mice to recruit T cells. These findings suggested that brain tumor-induced glucocorticoid surge and CCR8 upregulation in T cells lead to T cell sequestration in bone marrow, impairing the anti-tumor immune response. Targeting the glucocorticoid receptor-CCR8 axis may offer a promising immunotherapeutic approach for the treatment of intracranial 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
    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
    由于免疫细胞的增殖不足和坚固的免疫抑制肿瘤微环境,癌症免疫治疗仍然是一个重大挑战。在这里,我们提出了通过乳酸调节来激发细胞凋亡和增强抗肿瘤免疫力的假说。为此,开发了带有细菌膜涂层的铜-人血清白蛋白纳米复合物负载的金纳米笼(BAu-CuNC)。肿瘤细胞中BAu-CuNCs的靶向递送和分解引发了级联反应。在近红外(NIR)激光照射下,铜-人血清白蛋白(Cu-HSA)的释放增强,其通过二硫化物交换反应与肿瘤内谷胱甘肽(GSH)反应以释放Cu2离子并产生角化作用。随后,角化效应通过释放损伤相关分子模式(DAMPs)来触发肿瘤中的免疫原性细胞死亡(ICD),从而通过强大的细胞毒性T细胞(CD8)和辅助性T细胞(CD4)的产生实现抗肿瘤免疫。同时,在近红外辐射下,金纳米笼(AuNCs)促进过量的活性氧(ROS)的产生,在抑制糖酵解中起主要作用,降低乳酸和ATP水平。较低乳酸水平的联合作用,ATP减少和GSH消耗进一步使肿瘤细胞对角质形成敏感。此外,较低的乳酸产生导致免疫抑制性T调节细胞(Tregs)的显着阻断,并增强抗肿瘤免疫力。此外,有效抑制乳腺癌向肺的转移增强了BAu-CuNCs+NIR治疗的抗肿瘤治疗效果。因此,BAu-CuNCs+NIR同时诱导角化,ICD和乳酸生产受阻,导致肿瘤生长的抑制,重塑免疫抑制肿瘤微环境和抑制肺转移。因此,利用铜凋亡-乳酸调节,这种方法为增强肿瘤免疫治疗提供了一种新的策略.
    Cancer immunotherapy remains a significant challenge due to insufficient proliferation of immune cells and the sturdy immunosuppressive tumor microenvironment. Herein, we proposed the hypothesis of cuproptosis-lactate regulation to provoke cuproptosis and enhance anti-tumor immunity. For this purpose, copper-human serum albumin nanocomplex loaded gold nanocages with bacterial membrane coating (BAu-CuNCs) were developed. The targeted delivery and disassembly of BAu-CuNCs in tumor cells initiated a cascade of reactions. Under near infrared (NIR) laser irradiation, the release of copper-human serum albumin (Cu-HSA) was enhanced that reacted with intratumoral glutathione (GSH) via a disulfide exchange reaction to liberate Cu2+ ions and exert cuproptosis. Subsequently, the cuproptosis effect triggered immunogenic cell death (ICD) in tumor by the release of damage associated molecular patterns (DAMPs) to realize anti-tumor immunity via robust production of cytotoxic T cells (CD8+) and helper T cells (CD4+). Meanwhile, under NIR irradiation, gold nanocages (AuNCs) promoted excessive reactive oxygen species (ROS) generation that played a primary role in inhibiting glycolysis, reducing the lactate and ATP level. The combine action of lower lactate level, ATP reduction and GSH depletion further sensitized the tumor cells to cuproptosis. Also, the lower lactate production led to the significant blockage of immunosuppressive T regulatory cells (Tregs) and boosted the anti-tumor immunity. Additionally, the effective inhibition of breast cancer metastasis to the lungs enhanced the anti-tumor therapeutic impact of BAu-CuNCs + NIR treatment. Hence, BAu-CuNCs + NIR concurrently induced cuproptosis, ICD and hindered lactate production, leading to the inhibition of tumor growth, remodeling of the immunosuppressive tumor microenvironment and suppression of lung metastasis. Therefore, leveraging cuproptosis-lactate regulation, this approach presents a novel strategy for enhanced tumor immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    恶性肿瘤的免疫抑制微环境严重阻碍了抗肿瘤治疗的有效性。此外,异常的肿瘤血管与免疫细胞相互作用,形成进一步干扰抗肿瘤免疫并促进肿瘤进展的恶性循环。我们的基础前发现c-di-AMP和RRx-001分别具有优异的抗肿瘤作用,我们进一步探讨了它们是否可以协同组合用于抗肿瘤免疫疗法。我们选择将这两种药物加载到PVA-TSPBA水凝胶支架上,通过原位注射在肿瘤微环境中明确释放药物。研究表明c-di-AMP激活STING通路,增强免疫细胞浸润,逆转肿瘤免疫抑制.同时,RRx-001释放一氧化氮,增加肿瘤细胞的氧化应激损伤并促进细胞凋亡。此外,与单用药物相比,这两种药物的联合使用显示出更强大的促血管正常化和逆转的肿瘤免疫抑制.这项研究证明了抗肿瘤联合治疗的新设计选择以及肿瘤环境响应性水凝胶支架与抗肿瘤免疫疗法结合的潜力。
    The immunosuppressive microenvironment of malignant tumors severely hampers the effectiveness of anti-tumor therapy. Moreover, abnormal tumor vasculature interacts with immune cells, forming a vicious cycle that further interferes with anti-tumor immunity and promotes tumor progression. Our pre-basic found excellent anti-tumor effects of c-di-AMP and RRx-001, respectively, and we further explored whether they could be combined synergistically for anti-tumor immunotherapy. We chose to load these two drugs on PVA-TSPBA hydrogel scaffolds that expressly release drugs within the tumor microenvironment by in situ injection. Studies have shown that c-di-AMP activates the STING pathway, enhances immune cell infiltration, and reverses tumor immunosuppression. Meanwhile, RRx-001 releases nitric oxide, which increases oxidative stress injury in tumor cells and promotes apoptosis. Moreover, the combination of the two presented more powerful pro-vascular normalization and reversed tumor immunosuppression than the drug alone. This study demonstrates a new design option for anti-tumor combination therapy and the potential of tumor environmentally responsive hydrogel scaffolds in combination with anti-tumor immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    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
    免疫检查点抑制剂(ICIs)已经改变了癌症治疗,然而,低应答率和显著异质性等持续挑战需要引起关注.主要组织相容性复合体(MHC)在ICI疗效中的关键作用,其复杂的影响和潜力作为预后标志,值得全面探索。本研究整合了单细胞RNA测序(scRNA-seq),批量RNA-seq,和空间转录组学分析,以揭示受MHC转录特征(MHC。Sig).通过对不同队列的663,760个细胞的scRNA-seq分析开发,并在30种实体癌类型中进行了验证。MHC。sig证明了免疫相关基因和浸润免疫细胞之间的强相关性,强调其作为抗肿瘤免疫通用泛癌标志物的潜力。MHC的筛选。使用CRISPR数据的治疗靶标的sig识别了用于免疫治疗协同作用的潜在基因,并验证了其对跨不同数据集和癌症类型的ICIs响应性的预测功效。最后,细胞通讯模式的分析揭示了C1QC+巨噬细胞和恶性细胞之间的相互作用,提供对潜在治疗剂及其敏感性特征的见解。这种综合分析定位了MHC。sig作为预测免疫治疗结果和指导组合治疗策略的有希望的标志物。
    Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet persistent challenges such as low response rate and significant heterogeneity necessitate attention. The pivotal role of the major histocompatibility complex (MHC) in ICI efficacy, its intricate impacts and potentials as a prognostic marker, warrants comprehensive exploration. This study integrates single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and spatial transcriptomic analyses to unveil pan-cancer immune characteristics governed by the MHC transcriptional feature (MHC.sig). Developed through scRNA-seq analysis of 663,760 cells across diverse cohorts and validated in 30 solid cancer types, the MHC.sig demonstrates a robust correlation between immune-related genes and infiltrating immune cells, highlighting its potential as a universal pan-cancer marker for anti-tumor immunity. Screening the MHC.sig for therapeutic targets using CRISPR data identifies potential genes for immune therapy synergy and validates its predictive efficacy for ICIs responsiveness across diverse datasets and cancer types. Finally, analysis of cellular communication patterns reveals interactions between C1QC+macrophages and malignant cells, providing insights into potential therapeutic agents and their sensitivity characteristics. This comprehensive analysis positions the MHC.sig as a promising marker for predicting immune therapy outcomes and guiding combinatorial therapeutic strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号