Tumor immune microenvironment

肿瘤免疫微环境
  • 文章类型: Journal Article
    在宫颈癌(CC)病理生理学的动态景观中,本研究旨在阐明坏死性凋亡在调节肿瘤增殖中的作用,入侵,和CC中的免疫微环境。在这项研究中,通过一系列生物信息学分析和实验方法评估了坏死对CC的影响。通过分析坏死对肿瘤侵袭的影响来说明坏死对CC的影响,免疫反应,和JAK2-STAT3信号通路。贝伐单抗,一种靶向血管内皮生长因子(VEGF)的单克隆抗体,还评估了其在CC细胞中对坏死的潜在诱导及其与坏死抑制剂的相互作用。此外,这项研究评估了坏死对免疫微环境的影响,特别是在T细胞相关途径和CC中抑癌基因的表达中。发现坏死通过激活JAK2-STAT3途径增强VEGFA表达,促进CC中的肿瘤增殖和侵袭能力。贝伐单抗诱导CC细胞坏死,可能导致对治疗的抵抗。贝伐单抗与坏死性凋亡抑制剂的组合减弱了VEGFA的表达,提出了一种新的治疗策略。此外,坏死激活T细胞相关通路,促进JurkatT细胞的浸润和活化。CD3D-CC中的抑癌基因被鉴定为关键标志物,其表达可通过JurkatT细胞中的JAK2-STAT3途径通过坏死上调。用来自坏死凋亡诱导的Jurkat细胞的上清液处理CC细胞导致肿瘤细胞增殖和侵袭减少。这项研究揭示了坏死之间的复杂相互作用,肿瘤进展,和CC中的免疫反应。研究结果提出了一种细微差别的方法来利用坏死进行治疗干预,强调了将坏死性凋亡抑制剂与现有疗法相结合以改善CC治疗结果的潜力。
    In the dynamic landscape of cervical cancer (CC) pathophysiology, this study aimed to elucidate the role of necroptosis in modulating tumor proliferation, invasion, and the immune microenvironment in CC. In this study, the impact of necroptosis on CC was evaluated through a series of bioinformatical analyses and experimental approaches. The impact of necroptosis on CC was illustrated by analyzing its effects on tumor aggression, immune responses, and the JAK2-STAT3 signaling pathway. Bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor (VEGF), was also evaluated for its potential induction of necroptosis in CC cells and its interaction with necroptosis inhibitors. Additionally, the study assessed the influence of necroptosis on the immune microenvironment, particularly in T-cell-related pathways and the expression of tumor suppressor genes in CC. Necroptosis was found to enhance VEGFA expression through the activation of the JAK2-STAT3 pathway, promoting tumor proliferative and invasive capabilities in CC. Bevacizumab induced necroptosis in CC cells, potentially leading to resistance to therapy. The combination of bevacizumab with necroptosis inhibitors attenuated VEGFA expression, suggesting a novel therapeutic strategy. Additionally, necroptosis activated T-cell-related pathways and promoted the infiltration and activation of Jurkat T cells. CD3D-a tumor suppressor gene in CC-was identified as a critical marker and its expression could be upregulated by necroptosis via the JAK2-STAT3 pathway in Jurkat T cells. Treatment of CC cells with supernatants from necroptosis-induced Jurkat cells resulted in reduced tumor cell proliferation and invasion. This study reveals a complex interaction between necroptosis, tumor progression, and the immune response in CC. The findings propose a nuanced approach to leveraging necroptosis for therapeutic interventions, highlighting the potential of combining necroptosis inhibitors with existing therapies to improve treatment outcomes in CC.
    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:膀胱癌(BLCA)是一种高度异质性的肿瘤。我们旨在从N6-甲基腺苷甲基化(m6A)的角度构建分类器,以识别具有不同预后风险和治疗反应性的患者,以进行精确治疗。方法:基因表达谱数据,突变,临床特征主要来自TCGA-BLCA队列。进行无监督聚类以构建m6A亚型。通过使用ssGSEA探索肿瘤微环境(TME)景观,估计,和MCPcounter算法。使用K-M生存曲线和Cox回归分析来证明m6A亚型在预测预后中的意义。pRrophetic,oncoPredict,和TIDE算法用于评估抗肿瘤治疗的反应性.最后基于随机森林和人工神经网络(ANN)开发了m6a亚型分类器。结果:两种m6A亚型具有明显不同的m6A相关基因表达谱和突变格局。TME分析显示,与亚型A相比,亚型B中的基质和抑制性免疫成分水平更高。m6A亚型是BLCA的临床独立预后预测因子,B亚型预后较差。药物敏感性分析显示,B亚型对顺铂和多西他赛的IC50值和AUC值均较低。疗效评估显示,B亚型的放疗疗效明显较差,免疫治疗反应性较低。我们最终构建了一个ANN分类器,将BLCA患者准确分为两个m6A亚型。结论:我们的研究开发了一个分类器,用于识别具有不同m6A特征的亚型,具有不同m6A亚型的BLCA患者的预后和对抗肿瘤治疗的反应性明显不同。
    Purpose: Bladder cancer (BLCA) is a highly heterogeneous tumor. We aim to construct a classifier from the perspective of N6-methyladenosine methylation (m6A) to identify patients with different prognostic risks and treatment responsiveness for precision therapy. Methods: Data on gene expression profile, mutation, and clinical characteristics were mainly obtained from the TCGA-BLCA cohort. Unsupervised clustering was performed to construct m6A subtypes. The tumor microenvironment (TME) landscapes were explored by using ssGSEA, ESTIMATE, and MCPcounter algorithms. K-M survival curves and Cox regression analysis were used to demonstrate the significance of m6A subtypes in predicting prognosis. pRRophetic, oncoPredict, and TIDE algorithms were used to evaluate responsiveness to antitumor therapy. A classifier of m6a subtypes was finally developed based on random forest and artificial neural network (ANN). Results: The two m6A subtypes have significantly different m6A-related gene expression profiles and mutational landscapes. TME analysis showed a higher level of stromal and Inhibitory immune components in subtype B compared with subtype A. The m6A subtype is a clinically independent prognostic predictor of BLCA, subtype B has a poorer prognosis. Drug sensitivity analysis showed that subtype B has lower IC50 values and AUC values for cisplatin and docetaxel. Efficacy assessment showed significantly poorer radiotherapy efficacy and lower immunotherapy responsiveness in subtype B. We finally constructed an ANN classifier to accurately classify BLCA patients into two m6A subtypes. Conclusion: Our study developed a classifier for identifying subtypes with different m6A characteristics, and BLCA patients with different m6A subtypes have significantly different prognosis and responsiveness to antitumor 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
    CD8+T细胞是抗肿瘤免疫应答中的关键效应细胞。食管鳞状细胞癌(ESCC)中CD8T细胞浸润的潜在机制尚未明确阐明。
    收集新鲜的ESCC组织并根据CD8+T细胞的浸润密度分组。在对这些样品进行转录组测序并结合癌症基因组图谱(TCGA)ESCC数据进行分析后,选择分泌蛋白DEFB1以探索其在CD8+T细胞浸润中的潜在作用.生物信息学分析,然后进行组织学验证和体外实验。
    DEFB1在ESCC中高表达,DEFB1的高表达是总生存期的独立危险因素。由于DEFB1的上调或下调不影响增殖,ESCC细胞的迁移和凋亡,我们推测DEFB1的致癌作用是通过调节微环境特征来实现的。生物信息学分析表明,DEFB1可能在炎症反应和抗肿瘤免疫反应中起重要作用,并与ESCC中未成熟树突状细胞(imDC)的浸润有关。组织学分析进一步证实,有较少的CD8+T细胞浸润,在DEFB1高表达的ESCC样品中,较少的CD83成熟DC(mDC)浸润,而较多的CD1aimDC浸润。用重组DEFB1蛋白处理后,DC的成熟受到显著阻碍,其次是体外2D和3D培养中T细胞的杀伤作用受损。
    肿瘤来源的DEFB1可抑制DC的成熟,削弱CD8+T细胞的功能,考虑ESCC中的免疫耐受。DEFB1在ESCC中的作用值得进一步探讨。
    UNASSIGNED: CD8+ T cells are the key effector cells in the anti-tumor immune response. The mechanism underlying the infiltration of CD8+ T cells in esophageal squamous cell carcinoma (ESCC) has not been clearly elucidated.
    UNASSIGNED: Fresh ESCC tissues were collected and grouped according to the infiltration density of CD8+ T cells. After the transcriptome sequencing on these samples and the combined analyses with The Cancer Genome Atlas (TCGA) ESCC data, a secreted protein DEFB1 was selected to explore its potential role in the infiltration of CD8+ T cells. Bioinformatics analyses, histological verification and in vitro experiments were then performed.
    UNASSIGNED: DEFB1 was highly expressed in ESCC, and the high expression of DEFB1 was an independent risk factor for overall survival. Since the up-regulation or down-regulation of DEFB1 did not affect the proliferation, migration and apoptosis of ESCC cells, we speculated that the oncogenic effect of DEFB1 was achieved by regulating microenvironmental characteristics. Bioinformatics analyses suggested that DEFB1 might play a major role in the inflammatory response and anti-tumor immune response, and correlate to the infiltration of immature dendritic cell (imDC) in ESCC. Histological analyses further confirmed that there were less CD8+ T cells infiltrated, less CD83+ mature DC (mDC) infiltrated and more CD1a+ imDC infiltrated in those ESCC samples with high expression of DEFB1. After the treatment with recombinant DEFB1 protein, the maturation of DC was hindered significantly, followed by the impairment of the killing effects of T cells in both 2D and 3D culture in vitro.
    UNASSIGNED: Tumor-derived DEFB1 can inhibit the maturation of DC and weaken the function of CD8+ T cells, accounting for the immune tolerance in ESCC. The role of DEFB1 in ESCC deserves further exploration.
    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:SEPT9是一种关键的细胞骨架GTP酶,可调节包括有丝分裂和胞质分裂在内的多种生物学过程。虽然先前的研究涉及SEPT9与肿瘤发生和发展有关,但尚未进行全面的泛癌症分析。本研究旨在系统探讨其在癌症筛查中的作用,预后,和治疗,解决这一关键差距。
    方法:包含临床信息的基因和蛋白质表达数据从公共数据库获得,用于泛癌症分析。此外,我们使用来自90例肺鳞癌(LUSC)患者的临床样本,进一步通过实验验证SEPT9的临床意义.此外,分子对接工具用于分析SEPT9蛋白与药物之间的亲和力.
    结果:SEPT9在各种癌症中高表达,其异常表达与遗传变化和表观遗传修饰相关,导致不良临床结果。以LUSC为例,额外的数据集分析和免疫组织化学实验进一步证实了SEPT9基因和蛋白的诊断和预后价值以及临床相关性.功能富集,单细胞表达,和免疫浸润分析显示,SEPT9促进恶性肿瘤进展和调节免疫微环境,使患者受益于免疫疗法。此外,药物敏感性和分子对接分析表明,SEPT9与多种药物的敏感性和耐药性有关,包括Vorinostat和OTS-964。为了利用其在LUSC中的预后和治疗潜力,有丝分裂纺锤体相关预后模型,包括SEPT9、HSF1、ARAP3、KIF20B、FAM83D,TUBB8和几个临床特征,已开发。该模型不仅改善了临床结果预测,而且重塑了免疫微环境,使免疫疗法对LUSC患者更有效。
    结论:这是第一个系统分析SEPT9在癌症中的作用并创新性地将有丝分裂纺锤体相关模型应用于LUSC的研究,充分展示其作为癌症筛查和预后的有价值的生物标志物的潜力,并突出其在促进免疫治疗和化疗中的应用价值,特别是对于LUSC。
    BACKGROUND: SEPT9 is a pivotal cytoskeletal GTPase that regulates diverse biological processes encompassing mitosis and cytokinesis. While previous studies have implicated SEPT9 in tumorigenesis and development; comprehensive pan-cancer analyses have not been performed. This study aims to systematically explore its role in cancer screening, prognosis, and treatment, addressing this critical gap.
    METHODS: Gene and protein expression data containing clinical information were obtained from public databases for pan-cancer analyses. Additionally, clinical samples from 90 patients with lung squamous cell carcinoma (LUSC) were used to further experimentally validate the clinical significance of SEPT9. In addition, the molecular docking tool was used to analyze the affinities between SEPT9 protein and drugs.
    RESULTS: SEPT9 is highly expressed in various cancers, and its aberrant expression correlates with genetic alternations and epigenetic modifications, leading to adverse clinical outcomes. Take LUSC as an example, additional dataset analyses and immunohistochemical experiments further confirm the diagnostic and prognostic values as well as the clinical relevance of the SEPT9 gene and protein. Functional enrichment, single-cell expression, and immune infiltration analyses revealed that SEPT9 promotes malignant tumor progression and modulates the immune microenvironments, enabling patients to benefit from immunotherapy. Moreover, drug sensitivity and molecular docking analyses showed that SEPT9 is associated with the sensitivity and resistance of multiple drugs and has stable binding activity with them, including Vorinostat and OTS-964. To harness its prognostic and therapeutic potential in LUSC, a mitotic spindle-associated prognostic model including SEPT9, HSF1, ARAP3, KIF20B, FAM83D, TUBB8, and several clinical characteristics, was developed. This model not only improves clinical outcome predictions but also reshapes the immune microenvironment, making immunotherapy more effective for LUSC patients.
    CONCLUSIONS: This is the first study to systematically analyze the role of SEPT9 in cancers and innovatively apply the mitotic spindle-associated model to LUSC, fully demonstrating its potential as a valuable biomarker for cancer screening and prognosis, and highlighting its application value in promoting immunotherapy and chemotherapy, particularly for LUSC.
    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:尽管唾液腺癌(SGC)的免疫检查点抑制剂(ICPi)已经在临床试验中进行了研究,肿瘤免疫微环境(TIME)的细节尚不清楚.本研究旨在阐明SGC的时间及其与肿瘤突变负荷(TMB)的关系,并探讨ICPi适用性的原理。
    方法:我们选择了五种病理类型,即腺样囊性癌(ACC);腺癌,未另作说明(ANOS);唾液导管癌(SDC);和低级/高级粘液表皮样癌(MEClow/high)。我们调查了每种病理类型的TIME和TMB。通过多重荧光免疫组织化学评估TIME。通过下一代测序测量TMB。
    结果:ACC和MEChigh在肿瘤和基质中均显示出最低和最高的免疫效应细胞和抑制细胞浸润。ANOS,SDC,MEClow显示肿瘤中免疫效应细胞的适度浸润。相关性分析显示肿瘤中CD3+CD8+T细胞与TMB呈正相关(r=0.647)。肿瘤中CD3+CD8+T细胞与程序性细胞死亡-配体1表达呈正相关(r=0.513),与肿瘤中CD3+CD4+Foxp3+细胞呈弱正相关(r=0.399)。然而,在肿瘤中CD3+CD8+T细胞和CD204+细胞之间未观察到相关性(r=-0.049)。
    结论:ACC的时间是所谓的免疫沙漠类型,这可能解释了先前临床试验中对ICPi反应不佳的机制。另一方面,MEChigh是免疫发炎型,这可能支持ICPi这种病理亚型的基本原理.
    OBJECTIVE: Although immune checkpoint inhibitors (ICPi) for salivary gland cancer (SGC) have been investigated in clinical trials, details of the tumor immune microenvironment (TIME) remain unclear. This research aimed to elucidate the TIME of SGC and its relationship with tumor mutation burden (TMB) and to explore the rationale for the applicability of ICPi.
    METHODS: We selected five pathological types, namely adenoid cystic carcinoma (ACC); adenocarcinoma, not otherwise specified (ANOS); salivary duct carcinoma (SDC); and low/high-grade mucoepidermoid carcinoma (MEClow/high). We investigated the TIME and TMB of each pathological type. TIME was evaluated by multiplexed fluorescent immunohistochemistry. TMB was measured by next-generation sequencing.
    RESULTS: ACC and MEChigh showed the lowest and highest infiltration of immune effector and suppressor cells in both tumor and stroma. ANOS, SDC, and MEClow showed modest infiltration of immune effector cells in tumors. Correlation analysis showed a positive correlation between CD3+CD8+ T cells in tumor and TMB (r = 0.647). CD3+CD8+ T cells in tumors showed a positive correlation with programmed cell death-ligand 1 expression in tumor cells (r = 0.513) and a weak positive correlation with CD3+CD4+Foxp3+ cells in tumors (r = 0.399). However, no correlation was observed between CD3+CD8+ T cells and CD204+ cells in tumors (r = -0.049).
    CONCLUSIONS: The TIME of ACC was the so-called immune desert type, which may explain the mechanisms of the poor response to ICPi in previous clinical trials. On the other hand, MEChigh was the immune-inflamed type, and this may support the rationale of ICPi for this pathological subtype.
    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
    免疫疗法已在各种肿瘤中显示出有希望的抗肿瘤作用,然而,它遇到了来自抑制性肿瘤免疫微环境(TIME)的挑战。浸润性调节性T细胞(Tregs)是免疫抑制时间的重要贡献者,限制肿瘤免疫监视和阻断有效的抗肿瘤免疫反应。尽管系统性Tregs的消耗或抑制增强了抗肿瘤免疫力,自身免疫后遗症降低了对该方法的期望。在这里,我们总结新兴战略,特异性靶向肿瘤浸润(TI)-Tregs,通过重新编程其表型来提高生物体抵抗肿瘤的能力。还讨论了Treg重编程的调节机制以及如何利用这些知识来开发新的和有效的癌症免疫疗法。
    Immunotherapy has shown promising anti-tumor effects across various tumors, yet it encounters challenges from the inhibitory tumor immune microenvironment (TIME). Infiltrating regulatory T cells (Tregs) are important contributors to immunosuppressive TIME, limiting tumor immunosurveillance and blocking effective anti-tumor immune responses. Although depletion or inhibition of systemic Tregs enhances the anti-tumor immunity, autoimmune sequelae have diminished expectations for the approach. Herein, we summarize emerging strategies, specifically targeting tumor-infiltrating (TI)-Tregs, that elevate the capacity of organisms to resist tumors by reprogramming their phenotype. The regulatory mechanisms of Treg reprogramming are also discussed as well as how this knowledge could be utilized to develop novel and effective cancer immunotherapies.
    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:我们的研究致力于开发一种基于超级增强子相关基因(SERGs)的强大预测特征,具有预测生存结果和评估肝细胞癌(HCC)肿瘤免疫微环境(TiME)的双重目标。
    方法:从癌症基因组图谱(TCGA)检索HCCRNA测序数据,365例患者以1:1的比例被随机分配到训练或测试组.HCC的SERGs从超级增强子数据库(SEdb)下载。在训练集的基础上,确定了SERGs签名,其预后价值通过内部和外部验证(GSE14520)组得到证实。我们随后检查了该模型的潜在功能富集和肿瘤免疫浸润程度。此外,我们进行了体外实验来研究CBX2基因的生物学功能。
    结果:建立并验证了包括CBX2,TPX2,EFNA3,DNASE1L3和SOCS2的SE相关预后模型。根据这个风险模型,高风险组患者的预后明显较差,免疫细胞浸润与低危组差异显著。此外,高危人群表现出肿瘤相关病理通路的显著富集.SERGs签名通常可用于筛选可能对免疫疗法有反应的HCC患者,因为风险评分与肿瘤免疫功能障碍和排除(TIDE)评分之间存在正相关。此外,CBX2基因表达的下调被发现抑制HCC细胞活力,迁移,和细胞周期进程,同时促进细胞凋亡。
    结论:我们利用SERGs开发了一种新的HCC预后模型,这表明高风险评分的患者不仅预后较差,而且对免疫检查点抑制剂(ICIs)的治疗反应也可能减弱。该模型旨在针对每个患者的个性化需求定制个性化治疗策略,从而改善HCC患者的整体临床结果。此外,CBX2是HCC治疗干预的有希望的候选者。
    BACKGROUND: Our research endeavored to develop a robust predictive signature grounded in super-enhancer-related genes (SERGs), with the dual objectives of forecasting survival outcomes and evaluating the tumor immune microenvironment (TiME) in hepatocellular carcinoma (HCC).
    METHODS: HCC RNA-sequencing data were retrieved from The Cancer Genome Atlas (TCGA), and 365 patients were randomly assigned to training or testing sets in 1:1 ratio. SERGs of HCC were downloaded from Super-Enhancer Database (SEdb). On the basis of training set, a SERGs signature was identified, and its prognostic value was confirmed by internal and external validation (GSE14520) sets. We subsequently examined the model for potential functional enrichment and the degree of tumor immune infiltration. Additionally, we carried out in vitro experiments to delve into the biological functions of CBX2 gene.
    RESULTS: An SE-related prognostic model including CBX2, TPX2, EFNA3, DNASE1L3 and SOCS2 was established and validated. According to this risk model, patients in the high-risk group had a significantly worse prognosis, and their immune cell infiltration was significantly different from that of low-risk group. Moreover, the high-risk group exhibited a significant enrichment of tumor-associated pathological pathways. The SERGs signature can generally be utilized to screen HCC patients who are likely to respond to immunotherapy, as there is a positive correlation between the risk score and the Tumor Immune Dysfunction and Exclusion (TIDE) score. Furthermore, the downregulation of the CBX2 gene expression was found to inhibit HCC cell viability, migration, and cell cycle progression, while simultaneously promoting apoptosis.
    CONCLUSIONS: We developed a novel HCC prognostic model utilizing SERGs, indicating that patients with high-risk score not only face a poorer prognosis but also may exhibit a diminished therapeutic response to immune checkpoint inhibitors (ICIs). This model is designed to tailor personalized treatment strategies to the individual needs of each patient, thereby improving the overall clinical outcomes for HCC patients. Furthermore, CBX2 is a promising candidate for therapeutic intervention in HCC.
    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
    嵌合抗原受体T(CAR-T)细胞疗法作为过继性细胞疗法(ACT)的一种形式在癌症治疗中显示出巨大的前景。由FDA批准的针对CD19或B细胞成熟抗原(BCMA)的CAR-T细胞疗法证明,尽管实体瘤的预后中等。然而,尽管有这些进步,CAR-T疗法的功效通常会受到T细胞耗尽的影响,一种阻碍CAR-T细胞持久性和效应子功能的现象,导致使用CD19或CD22CAR-T细胞治疗血液系统恶性肿瘤的患者复发率高达75%。克服CAR-T耗尽的策略采用最先进的基因组工程工具和单细胞测序技术。在这次审查中,我们全面了解了T细胞耗竭的最新机制及其对当前优化CAR-T细胞治疗的影响.这些见解,结合最近临床试验中基于CAR-T的基准产品的经验教训,旨在解决T细胞耗尽带来的挑战,可能为开发量身定制的下一代癌症治疗方法奠定基础。
    Chimeric antigen receptor T (CAR-T) cell therapy as a form of adoptive cell therapy (ACT) has shown significant promise in cancer treatment, demonstrated by the FDA-approved CAR-T cell therapies targeting CD19 or B cell maturation antigen (BCMA) for hematological malignancies, albeit with moderate outcomes in solid tumors. However, despite these advancements, the efficacy of CAR-T therapy is often compromised by T cell exhaustion, a phenomenon that impedes the persistence and effector function of CAR-T cells, leading to a relapse rate of up to 75% in patients treated with CD19 or CD22 CAR-T cells for hematological malignancies. Strategies to overcome CAR-T exhaustion employ state-of-the-art genomic engineering tools and single-cell sequencing technologies. In this review, we provide a comprehensive understanding of the latest mechanistic insights into T cell exhaustion and their implications for the current efforts to optimize CAR-T cell therapy. These insights, combined with lessons learned from benchmarking CAR-T based products in recent clinical trials, aim to address the challenges posed by T cell exhaustion, potentially setting the stage for the development of tailored next-generation approaches to cancer treatment.
    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
    抗肿瘤药物的使用代表了癌症治疗的可靠策略。不幸的是,耐药已越来越普遍,并导致肿瘤的转移和局部复发。肿瘤免疫微环境(TME)由免疫细胞组成,细胞因子和免疫调节剂,它们共同影响对治疗的反应。表观遗传变化,包括DNA甲基化和组蛋白修饰,据报道,药物出口的增加也有助于癌症耐药性的发展。在过去的几年里,大多数关于肿瘤的研究仅从机制的角度关注肿瘤的发展和进展;很少有研究检查TME的变化是否也会影响肿瘤的生长和耐药性.最近,新的证据引起了人们对TME在耐药性发展中的作用的更多关注。在本次审查中,讨论了抑制性TME如何适应以免疫细胞合作为特征的耐药性,细胞因子,免疫调节剂,基质细胞和细胞外基质。此外,综述了这些免疫或代谢变化如何改变免疫监视,从而促进肿瘤耐药性。此外,我们揭示了TME中存在的潜在靶标,用于开发新的治疗策略,以改善癌症治疗的个体化治疗.
    The use of antitumor drugs represents a reliable strategy for cancer therapy. Unfortunately, drug resistance has become increasingly common and contributes to tumor metastasis and local recurrence. The tumor immune microenvironment (TME) consists of immune cells, cytokines and immunomodulators, and collectively they influence the response to treatment. Epigenetic changes including DNA methylation and histone modification, as well as increased drug exportation have been reported to contribute to the development of drug resistance in cancers. In the past few years, the majority of studies on tumors have only focused on the development and progression of a tumor from a mechanistic standpoint; few studies have examined whether the changes in the TME can also affect tumor growth and drug resistance. Recently, emerging evidence have raised more concerns regarding the role of TME in the development of drug resistance. In the present review, it was discussed how the suppressive TME adapts to drug resistance characterized by the cooperation of immune cells, cytokines, immunomodulators, stromal cells and extracellular matrix. Furthermore, it was reviewed how these immunological or metabolic changes alter immuno‑surveillance and thus facilitate tumor drug resistance. In addition, potential targets present in the TME for developing novel therapeutic strategies to improve individualized therapy for cancer treatment were revealed.
    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
    身体活动通过多种机制降低癌症相关死亡率,包括肿瘤免疫微环境(TIME)重编程。然而,生理干预是否以及如何促进抗肿瘤免疫仍然难以捉摸。这里,我们报告说,临床相关的自愿运动促进患者和小鼠模型中肌肉来源的细胞外囊泡(EV)相关的miR-29a-3p抑制肿瘤细胞外基质(ECM),从而允许免疫细胞浸润和免疫疗法。机械上,一项无偏倚的筛查确定了EV相关的miR-29a-3p对休闲时间体力活动或自愿运动的反应.含有MiR-29a-3p的EV在肿瘤中积累,并通过靶向COL1A1下调胶原蛋白组成。功能增益和功能丧失实验以及通过飞行时间(CyTOF)的细胞计数证明肌细胞分泌的miR-29a-3p促进抗肿瘤免疫。免疫疗法与自愿运动或miR-29a-3p的组合进一步增强抗肿瘤功效。临床上,miR-29a-3p与减少的ECM相关,T细胞浸润增加,和对免疫疗法的反应。我们的工作揭示了miR-29a-3p对免疫治疗的预测价值,提供了对运动诱导的抗癌免疫的机械见解,并强调了自愿运动在敏化免疫疗法中的潜力。
    Physical activity reduces cancer-associated mortality through multiple mechanisms, including tumor immune microenvironment (TIME) reprogramming. However, whether and how physiological interventions promote anti-tumor immunity remain elusive. Here, we report that clinically relevant voluntary exercise promotes muscle-derived extracellular vesicle (EV)-associated miR-29a-3p for tumor extracellular matrix (ECM) inhibition in patients and mouse models, thereby permitting immune cell infiltration and immunotherapy. Mechanistically, an unbiased screening identifies EV-associated miR-29a-3p in response to leisure-time physical activity or voluntary exercise. MiR-29a-3p-containing EVs accumulate in tumors and downregulate collagen composition by targeting COL1A1. Gain- and loss-of-function experiments and cytometry by time of flight (CyTOF) demonstrate that myocyte-secreted miR-29a-3p promotes anti-tumor immunity. Combining immunotherapy with voluntary exercise or miR-29a-3p further enhances anti-tumor efficacy. Clinically, miR-29a-3p correlates with reduced ECM, increased T cell infiltration, and response to immunotherapy. Our work reveals the predictive value of miR-29a-3p for immunotherapy, provides mechanistic insights into exercise-induced anti-cancer immunity, and highlights the potential of voluntary exercise in sensitizing immunotherapy.
    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