ICB, immune checkpoint blockade

ICB,免疫检查点阻断
  • 文章类型: Journal Article
    UASSIGNED:免疫检查点受体淋巴细胞激活基因3(LAG3)是免疫检查点阻断(ICB)的新靶标,但LAG3对动脉粥样硬化的影响尚不清楚。
    未经证实:本研究的目的是使用小鼠动脉粥样硬化的高胆固醇血症模型评估LAG3在斑块炎症中的作用。
    未经批准:为了研究LAG3在动脉粥样硬化中的作用,我们研究了造血细胞中缺乏LAG3的骨髓嵌合体以及全球Lag3-/-基因敲除小鼠。在高胆固醇血症低密度脂蛋白受体敲除(Ldlr-/-)小鼠中测试了抗LAG3单克隆抗体单一疗法和与抗程序性细胞死亡蛋白1(PD-1)的联合疗法的作用,并通过组织学和流式细胞术进行了评估。
    未经证实:LAG3缺乏或阻断抗LAG3单克隆抗体治疗导致产生干扰素γ的辅助性T细胞和效应/记忆T细胞水平升高,通过增加的调节性T细胞水平平衡。斑块大小既不受LAG3缺乏也不受LAG3阻断的影响,尽管LAG3缺失导致斑块中T细胞的密度增加了2倍。抗PD-1和抗LAG3的联合治疗对T细胞活化和细胞因子产生具有累加作用,并促进T细胞的斑块浸润。
    未经证实:LAG3功能的丧失促进了T细胞在斑块中的活化和积累,同时不影响斑块负荷。我们的报告支持进一步的临床研究,调查使用抗LAG3ICB治疗的患者的心血管风险。
    UNASSIGNED: The immune checkpoint receptor lymphocyte-activation gene 3 (LAG3) is a new target for immune checkpoint blockade (ICB), but the effects of LAG3 on atherosclerosis are not known.
    UNASSIGNED: The aim of the study was to evaluate the role of LAG3 on plaque inflammation using murine hypercholesterolemic models of atherosclerosis.
    UNASSIGNED: To study the role of LAG3 in atherosclerosis, we investigated both bone marrow chimeras lacking LAG3 in hematopoietic cells as well as global Lag3 -/- knockout mice. Effects of anti-LAG3 monoclonal antibody monotherapy and combination therapy with anti-programmed cell death protein 1 (PD-1) were tested in hypercholesterolemic low-density lipoprotein receptor knockout (Ldlr -/- ) mice and evaluated by histology and flow cytometry.
    UNASSIGNED: LAG3-deficiency or treatment with blocking anti-LAG3 monoclonal antibodies led to increased levels of both interferon gamma-producing T helper 1 cells and effector/memory T cells, balanced by increased levels of regulatory T cells. Plaque size was affected by neither LAG3 deficiency nor LAG3 blockade, although density of T cells in plaques was 2-fold increased by loss of LAG3. Combination therapy of anti-PD-1 and anti-LAG3 had an additive effect on T cell activation and cytokine production and promoted plaque infiltration of T cells.
    UNASSIGNED: Loss of LAG3 function promoted T cell activation and accumulation in plaques while not affecting plaque burden. Our report supports further clinical studies investigating cardiovascular risk in patients treated with anti-LAG3 ICB.
    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
    带卷曲螺旋的ADP-核糖基化因子(Arf)-GTP酶激活蛋白(GAP),据报道,锚蛋白重复序列和PH结构域1(ACAP1)可作为网格蛋白外套复合物的衔接子,在胞吞再循环和细胞迁移中起作用。ACAP1在肺腺癌(LUAD)中的潜在作用尚未完全确定。我们进行了全面的分析,包括基因表达,生存分析,遗传改变,功能富集,和免疫特性。ACAP1在肿瘤组织中显著下调,并与LUAD患者的临床病理特征有关。预后分析表明,低ACAP1表达与LUAD患者不满意的总生存期(OS)和疾病特异性生存期(DSS)相关。此外,ACAP1可以根据Cox比例风险模型和列线图模型确定为预后生物标志物。我们还证实ACAP1在两个LUAD细胞系中下调,与正常肺细胞相比。ACAP1的过表达导致细胞增殖的严重衰减,迁移,入侵,促进细胞凋亡。此外,功能富集分析证实ACAP1与T细胞活化和免疫应答高度相关.然后,我们进一步进行了免疫景观分析,包括单细胞RNA测序,免疫细胞浸润,和免疫检查点。ACAP1的表达与TME中免疫细胞的浸润水平和免疫检查点分子的表达呈正相关。本研究首先综合分析了分子表达,临床意义,以及LUAD中ACAP1的免疫景观特征,提示ACAP1可预测患者的预后,并可作为预测LUAD患者免疫治疗反应的潜在生物标志物.
    ADP-ribosylation factor (Arf)-GTPase-activating protein (GAP) with coiled-coil, ankyrin repeat and PH domains 1 (ACAP1) has been reported to serve as an adaptor for clathrin coat complex playing a role in endocytic recycling and cellular migration. The potential role of ACAP1 in lung adenocarcinoma (LUAD) has not been yet completely defined. We performed the comprehensive analyses, including gene expression, survival analysis, genetic alteration, function enrichment, and immune characteristics. ACAP1 was remarkably downregulated in tumor tissues, and linked with the clinicopathologic features in LUAD patients. Prognostic analysis demonstrated that low ACAP1 expression was correlated with unsatisfactory overall survival (OS) and disease specific survival (DSS) in LUAD patients. Moreover, ACAP1 could be determined as a prognostic biomarker according to Cox proportional hazard model and nomogram model. We also confirmed that ACAP1 was downregulated in two LUAD cell lines, comparing to normal lung cell. Overexpression of ACAP1 caused a profound attenuation in cell proliferation, migration, invasion, and promoted cell apoptosis. Additionally, functional enrichment analyses confirmed that ACAP1 was highly correlated with T cell activation and immune response. Then, we further conducted immune landscape analyses, including single cell RNA sequencing, immune cells infiltration, and immune checkpoints. ACAP1 expression was positively associated with the infiltrating level of immune cells in TME and the expression of immune checkpoint molecules. This study first comprehensively analyzed molecular expression, clinical implication, and immune landscape features of ACAP1 in LUAD, suggesting that ACAP1 was predictive of prognosis and could serve as a potential biomarker predicting immunotherapy response for LUAD patients.
    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
    中心体和纺锤体极相关蛋白(CSP1)是一种中心体和微管结合蛋白,在细胞周期依赖性细胞骨架组织和纤毛形成中起作用。以前的研究表明,CSP1在肿瘤发生中起作用;然而,尚未进行泛癌症分析.本研究系统地调查了CSP1的表达及其与诊断相关的潜在临床结果。预后,和治疗。CSP1广泛存在于组织和细胞中,其异常表达可作为癌症的诊断生物标志物。CSP1失调是由涉及遗传改变的多维机制驱动的,DNA甲基化,和miRNA。CSP1在特定位点的磷酸化可能在肿瘤发生中起作用。此外,CSP1与多种癌症的临床特征和结果相关。以预后不良的脑低级别胶质瘤(LGG)为例,功能富集分析表明CSP1可能在铁凋亡和肿瘤微环境(TME)中发挥作用,包括调节上皮-间质转化,基质反应,和免疫反应。进一步的分析证实,CSP1在LGG和其他癌症中失调铁凋亡,使得基于铁凋亡的药物有可能用于治疗这些癌症。重要的是,CSP1相关肿瘤在不同的TME中浸润,使免疫检查点阻断治疗对这些癌症患者有益。我们的研究首次证明CSP1是与铁凋亡和TME相关的潜在诊断和预后生物标志物。为特定癌症的药物治疗和免疫治疗提供了新的靶点。
    Centrosome and spindle pole-associated protein (CSPP1) is a centrosome and microtubule-binding protein that plays a role in cell cycle-dependent cytoskeleton organization and cilia formation. Previous studies have suggested that CSPP1 plays a role in tumorigenesis; however, no pan-cancer analysis has been performed. This study systematically investigates the expression of CSPP1 and its potential clinical outcomes associated with diagnosis, prognosis, and therapy. CSPP1 is widely present in tissues and cells and its aberrant expression serves as a diagnostic biomarker for cancer. CSPP1 dysregulation is driven by multi-dimensional mechanisms involving genetic alterations, DNA methylation, and miRNAs. Phosphorylation of CSPP1 at specific sites may play a role in tumorigenesis. In addition, CSPP1 correlates with clinical features and outcomes in multiple cancers. Take brain low-grade gliomas (LGG) with a poor prognosis as an example, functional enrichment analysis implies that CSPP1 may play a role in ferroptosis and tumor microenvironment (TME), including regulating epithelial-mesenchymal transition, stromal response, and immune response. Further analysis confirms that CSPP1 dysregulates ferroptosis in LGG and other cancers, making it possible for ferroptosis-based drugs to be used in the treatment of these cancers. Importantly, CSPP1-associated tumors are infiltrated in different TMEs, rendering immune checkpoint blockade therapy beneficial for these cancer patients. Our study is the first to demonstrate that CSPP1 is a potential diagnostic and prognostic biomarker associated with ferroptosis and TME, providing a new target for drug therapy and immunotherapy in specific 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
    靶向PD-1/PD-L1的免疫检查点阻断(ICB)在治疗癌症方面显示出显著的希望。然而,低反应率和经常观察到的严重副作用限制了其广泛的益处.部分原因是对PD-L1的生物学调控了解较少。这里,我们系统全面地总结了PD-L1从核染色质重组到细胞外呈递的调控。在PD-L1和PD-L2高表达的癌细胞中,在CD274和CD273周围发现了一个新的TAD(拓扑关联域)(chr9:5,400,000-5,600,000),其中包括报道的超级增强子以驱动PD-L1和PD-L2的同步转录。重新成形的TAD允许转录因子如STAT3和IRF1募集到PD-L1基因座以指导PD-L1的表达。转录后,PD-L1通过长3UTR受到miRNA和RNA结合蛋白的严格调控。在翻译层面,PD-L1蛋白及其膜呈递受到翻译后修饰如糖基化和泛素化的严格调节。此外,PD-L1可以通过外泌体分泌以系统地抑制免疫应答。因此,全面剖析PD-L1/PD-L2的调节,彻底检测PD-L1/PD-L2及其调节网络将为ICB和基于ICB的组合治疗带来更多见解。
    The immune checkpoint blockade (ICB) targeting on PD-1/PD-L1 has shown remarkable promise in treating cancers. However, the low response rate and frequently observed severe side effects limit its broad benefits. It is partially due to less understanding of the biological regulation of PD-L1. Here, we systematically and comprehensively summarized the regulation of PD-L1 from nuclear chromatin reorganization to extracellular presentation. In PD-L1 and PD-L2 highly expressed cancer cells, a new TAD (topologically associating domain) (chr9: 5,400,000-5,600,000) around CD274 and CD273 was discovered, which includes a reported super-enhancer to drive synchronous transcription of PD-L1 and PD-L2. The re-shaped TAD allows transcription factors such as STAT3 and IRF1 recruit to PD-L1 locus in order to guide the expression of PD-L1. After transcription, the PD-L1 is tightly regulated by miRNAs and RNA-binding proteins via the long 3\'UTR. At translational level, PD-L1 protein and its membrane presentation are tightly regulated by post-translational modification such as glycosylation and ubiquitination. In addition, PD-L1 can be secreted via exosome to systematically inhibit immune response. Therefore, fully dissecting the regulation of PD-L1/PD-L2 and thoroughly detecting PD-L1/PD-L2 as well as their regulatory networks will bring more insights in ICB and ICB-based combinational 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
    化疗和免疫疗法的结合通过引发免疫原性细胞死亡(ICD)来激发强大的免疫系统,在抑制肿瘤生长和改善免疫抑制肿瘤微环境(ITM)方面显示出巨大的潜力。然而,低劣的药物生物利用度限制了治疗效果。在这里,我们报道了一种通用的生物响应性阿霉素(DOX)基纳米凝胶,可实现肿瘤特异性药物共递送。设计并选择基于DOX的甘露糖纳米凝胶(DMNG)作为示例,以阐明联合化学免疫疗法的机制。不出所料,DMNG表现出显著的胶束稳定性,选择性药物释放和延长生存时间,受益于增强肿瘤通透性和延长血液循环。我们发现由DMNG递送的DOX可以通过促进ICD来诱导强大的抗肿瘤免疫应答。同时,从DMNGs释放的甘露糖被证明在体外和体内对乳腺癌具有强大的协同治疗作用,通过破坏糖酵解和三羧酸循环中的葡萄糖代谢。总的来说,基于DOX的纳米凝胶对肿瘤微环境的调节有望成为一种有效的候选策略,以克服基于ICD的免疫治疗的当前局限性。为免疫调节纳米药物的开发提供了范例。
    The combination of chemotherapy and immunotherapy motivates a potent immune system by triggering immunogenic cell death (ICD), showing great potential in inhibiting tumor growth and improving the immunosuppressive tumor microenvironment (ITM). However, the therapeutic effectiveness has been restricted by inferior drug bioavailability. Herein, we reported a universal bioresponsive doxorubicin (DOX)-based nanogel to achieve tumor-specific co-delivery of drugs. DOX-based mannose nanogels (DM NGs) was designed and choosed as an example to elucidate the mechanism of combined chemo-immunotherapy. As expected, the DM NGs exhibited prominent micellar stability, selective drug release and prolonged survival time, benefited from the enhanced tumor permeability and prolonged blood circulation. We discovered that the DOX delivered by DM NGs could induce powerful anti-tumor immune response facilitated by promoting ICD. Meanwhile, the released mannose from DM NGs was proved as a powerful and synergetic treatment for breast cancer in vitro and in vivo, via damaging the glucose metabolism in glycolysis and the tricarboxylic acid cycle. Overall, the regulation of tumor microenvironment with DOX-based nanogel is expected to be an effectual candidate strategy to overcome the current limitations of ICD-based immunotherapy, offering a paradigm for the exploitation of immunomodulatory nanomedicines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    人工智能(AI)是一个通用术语,是指使用机器来模仿智能行为,以最少的人为干预来执行复杂的任务。比如机器学习;这项技术正在彻底改变和重塑医学。人工智能在诊断等领域具有完善医疗保健系统的巨大潜力,风险分析,卫生信息管理,生活方式监督,虚拟健康援助。在免疫治疗方面,人工智能已被应用于基于免疫特征的免疫治疗反应的预测,医学成像和组织学分析。这些特征在癌症免疫疗法的管理中也非常有用,因为它们在提高诊断准确性方面的性能不断提高。优化治疗计划,预测护理结果并降低人力资源成本。在这次审查中,我们介绍了AI的详细信息以及将AI用于癌症免疫治疗的当前进展和最新技术。此外,我们讨论挑战,应用该技术进行广泛临床部署的机会和相应的策略。最后,我们总结了AI对癌症免疫治疗的影响,并提供了我们对未来AI潜在应用的看法。
    Artificial intelligence (AI) is a general term that refers to the use of a machine to imitate intelligent behavior for performing complex tasks with minimal human intervention, such as machine learning; this technology is revolutionizing and reshaping medicine. AI has considerable potential to perfect health-care systems in areas such as diagnostics, risk analysis, health information administration, lifestyle supervision, and virtual health assistance. In terms of immunotherapy, AI has been applied to the prediction of immunotherapy responses based on immune signatures, medical imaging and histological analysis. These features could also be highly useful in the management of cancer immunotherapy given their ever-increasing performance in improving diagnostic accuracy, optimizing treatment planning, predicting outcomes of care and reducing human resource costs. In this review, we present the details of AI and the current progression and state of the art in employing AI for cancer immunotherapy. Furthermore, we discuss the challenges, opportunities and corresponding strategies in applying the technology for widespread clinical deployment. Finally, we summarize the impact of AI on cancer immunotherapy and provide our perspectives about underlying applications of AI in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    程序性细胞死亡配体1(PD-L1)/程序性细胞死亡蛋白1(PD-1)级联是免疫检查点阻断(ICB)疗法的有效治疗靶标。通过小分子药物靶向PD-L1/PD-1轴是增强抗肿瘤免疫力的有吸引力的方法。使用基于流式细胞术的测定法,我们确定土贝莫苷-1(TBM-1)是一种有前景的抗肿瘤免疫调节剂,可以负调节PD-L1水平.TBM-1破坏PD-1/PD-L1相互作用并通过降低PD-L1的丰度增强T细胞对癌细胞的细胞毒性。此外,TBM-1通过激活肿瘤浸润性T细胞免疫在患有Lewis肺癌(LLC)和B16黑色素瘤肿瘤异种移植物的小鼠中发挥其抗肿瘤作用。机械上,TBM-1在TFEB依赖性中触发PD-L1溶酶体降解,自噬非依赖性途径。TBM-1选择性结合哺乳动物雷帕霉素靶(mTOR)激酶并抑制mTORC1的激活,导致TFEB的核易位和溶酶体生物发生。此外,TBM-1和抗CTLA-4的组合可有效增强抗肿瘤T细胞免疫,并减少髓源性抑制细胞(MDSC)和调节性T(Treg)细胞的免疫抑制浸润。我们的发现揭示了TBM-1以前未被识别的抗肿瘤机制,并代表了一种替代的ICB治疗策略,以增强癌症免疫疗法的功效。
    Programmed cell death ligand 1 (PD-L1)/programmed cell death protein 1 (PD-1) cascade is an effective therapeutic target for immune checkpoint blockade (ICB) therapy. Targeting PD-L1/PD-1 axis by small-molecule drug is an attractive approach to enhance antitumor immunity. Using flow cytometry-based assay, we identify tubeimoside-1 (TBM-1) as a promising antitumor immune modulator that negatively regulates PD-L1 level. TBM-1 disrupts PD-1/PD-L1 interaction and enhances the cytotoxicity of T cells toward cancer cells through decreasing the abundance of PD-L1. Furthermore, TBM-1 exerts its antitumor effect in mice bearing Lewis lung carcinoma (LLC) and B16 melanoma tumor xenograft via activating tumor-infiltrating T-cell immunity. Mechanistically, TBM-1 triggers PD-L1 lysosomal degradation in a TFEB-dependent, autophagy-independent pathway. TBM-1 selectively binds to the mammalian target of rapamycin (mTOR) kinase and suppresses the activation of mTORC1, leading to the nuclear translocation of TFEB and lysosome biogenesis. Moreover, the combination of TBM-1 and anti-CTLA-4 effectively enhances antitumor T-cell immunity and reduces immunosuppressive infiltration of myeloid-derived suppressor cells (MDSCs) and regulatory T (Treg) cells. Our findings reveal a previously unrecognized antitumor mechanism of TBM-1 and represent an alternative ICB therapeutic strategy to enhance the efficacy of cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症治疗的主要挑战是如何有效消除原发性肿瘤并充分诱导免疫原性细胞死亡(ICD)以激发强大的免疫反应来控制转移。这里,开发了一种自组装的级联生物反应器,以增强肿瘤渗透和饥饿的协同治疗来改善癌症治疗,化学动力学(CDT)和光热疗法。以葡萄糖氧化酶(GOx)为模板合成超小FeS-GOx纳米点,紫杉醇(PTX)通过疏水作用诱导形成自组装FeS-GOx@PTX(FGP)。在肿瘤部位积累后,FGP分解为较小的FeS-GOx,以增强肿瘤的深层渗透。GOx维持高的酶活性以在氧的辅助下催化葡萄糖以产生过氧化氢(H2O2)作为饥饿疗法。涉及再生H2O2的Fenton反应进而产生更多的羟基自由基以增强CDT。跟随808nm的近红外激光,通过联合治疗,FGP在体外和体内显示出显著的肿瘤抑制。随之而来的钙网织蛋白暴露增加了ICD并促进了树突状细胞的成熟。结合抗CTLA4检查点封锁,由于细胞毒性T淋巴细胞的肿瘤内浸润增强,FGP可以绝对消除原发性肿瘤并积极抑制远处肿瘤。我们的工作提出了一种有希望的原发性肿瘤和转移抑制策略。
    Major challenges for cancer treatment are how to effectively eliminate primary tumor and sufficiently induce immunogenic cell death (ICD) to provoke a robust immune response for metastasis control. Here, a self-assembled cascade bioreactor was developed to improve cancer treatment with enhanced tumor penetration and synergistic therapy of starvation, chemodynamic (CDT) and photothermal therapy. Ultrasmall FeS-GOx nanodots were synthesized with glucose oxidase (GOx) as template and induced by paclitaxel (PTX) to form self-assembling FeS-GOx@PTX (FGP) via hydrophobic interaction. After accumulated at tumor sites, FGP disassembles to smaller FeS-GOx for enhanced deep tumor penetration. GOx maintains high enzymatic activity to catalyze glucose with assistant of oxygen to generate hydrogen peroxide (H2O2) as starvation therapy. Fenton reaction involving the regenerated H2O2 in turn produced more hydroxyl radicals for enhanced CDT. Following near-infrared laser at 808 nm, FGPs displayed pronounced tumor inhibition in vitro and in vivo by the combination therapy. The consequent increased exposure to calreticulin amplified ICD and promoted dendritic cells maturation. In combination with anti-CTLA4 checkpoint blockade, FGP can absolutely eliminate primary tumor and avidly inhibit distant tumors due to the enhanced intratumoral infiltration of cytotoxic T lymphocytes. Our work presents a promising strategy for primary tumor and metastasis inhibition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    微小RNA(miRNA)失调通过调节mRNA水平在前列腺癌(PCa)的异质性发展中起关键作用。在这里,我们旨在通过非负矩阵分解对miRNA调节的转录组进行聚类来表征PCa的分子特征。使用来自癌症基因组图谱的478个PCa样本,四种分子亚型(S-I,S-II,S-III,和S-IV)在两个合并的微阵列和RNAseq数据集中的656和252个样本中进行了鉴定和验证,分别。有趣的是,4种亚型在综合分析临床特征后表现出明显的临床和生物学特征,多维配置文件,免疫浸润,和药物敏感性。S-I是基底/干/间充质样的,免疫排除有明显的转化生长因子β,上皮-间质转化和缺氧信号,增加对奥拉帕尼的敏感性,和中间预后。S-II具有腔/代谢活性,对雄激素剥夺治疗有反应,经常进行TMPRSS2-ERG融合,预后良好。S-III的特征是适度的增殖和代谢活性,对基于紫杉烷的化疗的敏感性,和中间预后。S-IV具有高度增生性,具有中等EMT和干性,频繁删除TP53、PTEN和RB,和最差的预后;它也是免疫发炎和敏感的抗PD-L1治疗。总的来说,基于miRNA调节的基因谱,这项研究确定了4种不同的PCa亚型,这些亚型可以改善诊断时的风险分层并提供治疗指导.
    MicroRNA (miRNA) deregulation plays a critical role in the heterogeneous development of prostate cancer (PCa) by tuning mRNA levels. Herein, we aimed to characterize the molecular features of PCa by clustering the miRNA-regulated transcriptome with non-negative matrix factorization. Using 478 PCa samples from The Cancer Genome Atlas, four molecular subtypes (S-I, S-II, S-III, and S-IV) were identified and validated in two merged microarray and RNAseq datasets with 656 and 252 samples, respectively. Interestingly, the four subtypes showed distinct clinical and biological features after comprehensive analyses of clinical features, multiomic profiles, immune infiltration, and drug sensitivity. S-I is basal/stem/mesenchymal-like and immune-excluded with marked transforming growth factor β, epithelial-mesenchymal transition and hypoxia signals, increased sensitivity to olaparib, and intermediate prognosis. S-II is luminal/metabolism-active and responsive to androgen deprivation therapy with frequent TMPRSS2-ERG fusion and a good prognosis. S-III is characterized by moderate proliferative and metabolic activity, sensitivity to taxane-based chemotherapy, and intermediate prognosis. S-IV is highly proliferative with moderate EMT and stemness, frequent deletions of TP53, PTEN and RB, and the poorest prognosis; it is also immune-inflamed and sensitive to anti-PD-L1 therapy. Overall, based on miRNA-regulated gene profiles, this study identified four distinct PCa subtypes that could improve risk stratification at diagnosis and provide therapeutic guidance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:免疫检查点阻断(ICB)已被批准用于肝细胞癌(HCC)的治疗。然而,许多晚期HCC患者对ICB单药治疗无反应。已提出细胞毒性化学疗法来调节肿瘤微环境(TME)并使肿瘤对ICB敏感。因此,我们的目的是在原位HCC模型中研究细胞毒性化疗和ICB的组合。
    方法:使用临床前原位HCC小鼠模型来阐明5-氟尿嘧啶(5-FU)和ICB的功效。小鼠肝内注射RIL-175或Hepa1-6细胞,然后用5-FU和抗程序性细胞死亡配体1(PD-L1)抗体处理。骨髓来源的抑制细胞(MDSC)被耗尽以验证其在减弱对免疫疗法的敏感性中的作用。在小鼠和患者样本中进行基于流式细胞术的免疫谱分析和免疫荧光染色,分别。
    结果:5-FU可以诱导肿瘤内MDSC的积累,以抵消T淋巴细胞和自然杀伤细胞的浸润,从而取消PD-L1阻断的抗肿瘤功效。在临床样本中,经动脉化疗栓塞后,MDSCs积累,CD8+T细胞数量减少。
    结论:5-FU可以引发免疫抑制MDSCs的积累,在HCC中损害对PD-L1阻断的反应。我们的数据表明,特异性化疗和ICB的组合可能会损害抗肿瘤免疫反应,需要在临床前模型中进一步研究,并在临床环境中考虑。
    背景:我们的研究结果表明,一些化疗可能会损害免疫治疗的抗肿瘤功效。需要进一步的研究来揭示不同化疗对肿瘤免疫谱的具体影响。这些数据对于合理设计肝细胞癌患者的联合免疫治疗策略至关重要。
    OBJECTIVE: Immune checkpoint blockade (ICB) has been approved for treatment of hepatocellular carcinoma (HCC). However, many patients with advanced HCC are non-responders to ICB monotherapy. Cytotoxic chemotherapy has been proposed to modulate the tumor microenvironment (TME) and sensitize tumors to ICB. Thus, we aimed to study the combination of cytotoxic chemotherapy and ICB in an orthotopic HCC model.
    METHODS: Preclinical orthotopic HCC mouse models were used to elucidate the efficacy of 5-fluorouracil (5-FU) and ICB. The mice were intrahepatically injected with RIL-175 or Hepa1-6 cells, followed by treatment with 5-FU and anti-programmed cell death ligand 1 (PD-L1) antibody. Myeloid-derived suppressor cells (MDSCs) were depleted to validate their role in attenuating sensitivity to immunotherapy. Flow cytometry-based immune profiling and immunofluorescence staining were performed in mice and patient samples, respectively.
    RESULTS: 5-FU could induce intratumoral MDSC accumulation to counteract the infiltration of T lymphocytes and natural killer cells, thus abrogating the anti-tumor efficacy of PD-L1 blockade. In clinical samples, MDSCs accumulated and CD8+ T cell numbers decreased following transarterial chemoembolization.
    CONCLUSIONS: 5-FU can trigger the accumulation of immunosuppressive MDSCs, impairing the response to PD-L1 blockade in HCC. Our data suggest that the combination of specific chemotherapy and ICB may impair anti-tumor immune responses, warranting further study in preclinical models and consideration in clinical settings.
    BACKGROUND: Our findings suggest that some chemotherapies may impair the anti-tumor efficacy of immunotherapy. Further studies are required to uncover the specific effects of different chemotherapies on the immunological profile of tumors. This data will be critical for the rational design of combination immunotherapy strategies for patients with hepatocellular carcinoma.
    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