tumour microenvironment

肿瘤微环境
  • 文章类型: Journal Article
    用于对抗抗性癌症的基于细胞的免疫疗法中的尖端方法涉及基因工程化嵌合抗原受体T(CAR-T)淋巴细胞。近年来,这些疗法已经证明是有效的,导致它们针对某些类型的癌症的商业化和临床应用。然而,CAR-T疗法面临局限,如免疫抑制肿瘤微环境(TME),可以使CAR-T细胞无效,以及治疗的副作用,包括细胞因子释放综合征(CRS)。细胞外囊泡(EV)是由几乎所有细胞类型释放到细胞外环境中的不同组的膜结合颗粒。它们对于细胞间通讯至关重要,转移诸如蛋白质之类的货物,脂质,各种类型的RNA,和靶细胞的DNA片段,在局部和系统上穿越生物屏障。电动汽车在许多生理过程中发挥作用,来自免疫和非免疫细胞的那些能够通过激活或抑制来调节免疫系统。利用电动汽车的这种能力来增强CAR-T细胞疗法可能代表着克服其当前局限性的重大进展。这篇综述探讨了CAR-T细胞免疫治疗的现状,并探讨了电动汽车在增强其治疗效果方面的潜在作用。
    A cutting-edge approach in cell-based immunotherapy for combating resistant cancer involves genetically engineered chimeric antigen receptor T (CAR-T) lymphocytes. In recent years, these therapies have demonstrated effectiveness, leading to their commercialization and clinical application against certain types of cancer. However, CAR-T therapy faces limitations, such as the immunosuppressive tumour microenvironment (TME) that can render CAR-T cells ineffective, and the adverse side effects of the therapy, including cytokine release syndrome (CRS). Extracellular vesicles (EVs) are a diverse group of membrane-bound particles released into the extracellular environment by virtually all cell types. They are essential for intercellular communication, transferring cargoes such as proteins, lipids, various types of RNAs, and DNA fragments to target cells, traversing biological barriers both locally and systemically. EVs play roles in numerous physiological processes, with those from both immune and non-immune cells capable of modulating the immune system through activation or suppression. Leveraging this capability of EVs to enhance CAR-T cell therapy could represent a significant advancement in overcoming its current limitations. This review examines the current landscape of CAR-T cell immunotherapy and explores the potential role of EVs in augmenting its therapeutic efficacy.
    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
    在全球范围内,癌症是一种困难和毁灭性的疾病。当前化疗的几个问题包括细胞毒性,缺乏选择性,干细胞样细胞生长,和多重耐药性。用于癌症治疗的最合适的纳米材料是那些具有特征的纳米材料,如细胞毒性,限制性特异性,和药物容量和生物利用度;这些材料是纳米级的(1-100nm)。尽管研究不断发展,但纳米药物很少被许可用于治疗用途。这些化合物需要纳米载体靶向药物递送实验来改善它们的翻译。这篇综述描述了文献中报道的新纳米材料,阻碍他们的临床研究,以及它们有益的癌症治疗用途。它还提出了更有效地在癌症治疗中使用纳米材料的方法,并描述了癌症治疗的内在挑战以及可用于特定肿瘤靶向的不同纳米载体和化学物质。此外,它提供了癌症治疗方法的简要概述,重点是那些利用纳米材料的人。虽然纳米技术为癌症检测和治疗的未来进步提供了一个很好的来源,有一个新兴的需要更多的研究,以解决目前的障碍,临床翻译.
    On a global scale, cancer is a difficult and devastating illness. Several problems with current chemotherapies include cytotoxicity, lack of selectivity, stem-like cell growth, and multi-drug resistance. The most appropriate nanomaterials for cancer treatment are those with characteristics, such as cytotoxicity, restricted specificity, and drug capacity and bioavailability; these materials are nanosized (1-100 nm). Nanodrugs are rarely licenced for therapeutic use despite growing research. These compounds need nanocarrier-targeted drug delivery experiments to improve their translation. This review describes new nanomaterials reported in the literature, impediments to their clinical studies, and their beneficial cancer therapeutic use. It also suggests ways to use nanomaterials in cancer therapy more efficiently and describes the intrinsic challenges of cancer treatment and the different nanocarriers and chemicals that can be utilised for specified tumour targeting. Furthermore, it provides a concise overview of cancer theranostics methods, with a focus on those that make use of nanomaterials. Although nanotechnology offers a great source for future advancements in cancer detection and therapy, there is an emerging need for more studies to address the present barriers to clinical translation.
    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
    癌细胞表现出改变的代谢表型,以糖酵解和乳酸产生增加为特征,即使在充足的氧气存在下-这种现象被称为Warburg效应。这种代谢重编程是使癌细胞能够满足其提高的能量和生物合成需求的关键适应。重要的是,肿瘤微环境在形成和维持癌细胞的这种代谢转变中起着关键作用。本文综述了肿瘤微环境与Warburg效应之间的复杂关系。强调在这个利基内的交流如何调节癌细胞代谢并影响肿瘤进展和治疗抗性。我们讨论了靶向Warburg效应作为一种有前途的治疗策略的潜力,目的是破坏癌细胞的代谢优势,并增强我们对肿瘤微环境中这种复杂相互作用的理解。
    Cancer cells display an altered metabolic phenotype, characterised by increased glycolysis and lactate production, even in the presence of sufficient oxygen - a phenomenon known as the Warburg effect. This metabolic reprogramming is a crucial adaptation that enables cancer cells to meet their elevated energy and biosynthetic demands. Importantly, the tumor microenvironment plays a pivotal role in shaping and sustaining this metabolic shift in cancer cells. This review explores the intricate relationship between the tumor microenvironment and the Warburg effect, highlighting how communication within this niche regulates cancer cell metabolism and impacts tumor progression and therapeutic resistance. We discuss the potential of targeting the Warburg effect as a promising therapeutic strategy, with the aim of disrupting the metabolic advantage of cancer cells and enhancing our understanding of this complex interplay within the tumor microenvironment.
    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
    癌症对发达国家和发展中国家都有重大的负面社会和经济影响。因此,了解癌症的发病和进展对于开发可以改善癌症患者的福祉和健康的疗法至关重要。随着时间的推移,研究表明,肿瘤微环境对这一过程有很大的影响。微观和纳米尺度的工程技术可用于研究肿瘤微环境。纳米级和微型工程使用具有小尺寸的新颖技术和设计来重新创建TME。了解癌细胞如何相互作用,可以帮助研究人员开发治疗方法,预测和抵消癌细胞的技术,以逃避检测和对抗抗癌治疗。例如微加工技术,微流体装置,纳米传感器,和用于研究或重建肿瘤微环境的纳米设备。然而,一个复杂的行动,就像生长和癌症进展一样,以及它们在周围环境中的密集关联,这必须进行更详细的研究。
    Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells\' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
    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
    免疫治疗对透明细胞肾细胞癌(ccRCC)的疗效,特别是先进的ccRCC,是有限的,提出了临床挑战。这种限制与免疫调节网络密切相关。了解肿瘤微环境(TME)的异质性对于开发先进的ccRCC疗法至关重要。使用公开可用的ccRCC数据(scRNA-seq,批量RNA-seq,和体细胞突变数据),进行了一项多组学研究,以探讨TME异质性.通过组合的scRNA-seq和整体RNA-seq分析鉴定了三种不同的ccRCC免疫亚型。在TCGA和CheckMate队列中验证了基于免疫抑制肿瘤亚型中独特细胞信号分子的预后模型。MDK作为免疫抑制亚型中的关键调节基因出现,预测不良ccRCC预后和不良的免疫治疗反应。MDK通过MDK-LRP1相互作用促进M2巨噬细胞极化,MDK的抑制抑制了M2极化。这项研究揭示了ccRCCTME的异质性和可靠的预后模型,阐明MDK在免疫抑制TME中的重要作用,为优化ccRCC免疫治疗铺平了道路。
    The efficacy of immunotherapy for clear cell renal cell carcinoma (ccRCC), especially advanced ccRCC, is limited, presenting a clinical challenge. This limitation is closely tied to the immune regulation network. Understanding the heterogeneity of the tumour microenvironment (TME) is crucial for developing advanced ccRCC therapies. Using publicly available ccRCC data (scRNA-seq, bulk RNA-seq, and somatic mutation data), a multiomics study was performed to explore TME heterogeneity. Three distinct ccRCC immune subtypes were identified through combined scRNA-seq and bulk RNA-seq analysis. A prognostic model based on unique cell signalling molecules in immunosuppressive tumour subtype was validated in the TCGA and CheckMate cohorts. MDK emerged as a critical regulatory gene in the immunosuppressive subtype, predicting a poor ccRCC prognosis and a poor immunotherapy response. MDK promotes M2 macrophage polarization via the MDK-LRP1 interaction, and the inhibition of MDK suppressed M2 polarization. This study revealed the heterogeneity of the ccRCC TME and a reliable prognostic model, shedding light on the vital role of MDK in the immunosuppressive TME and paving the way for optimized ccRCC 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
    背景:免疫检查点阻断(ICB)的治疗潜力遍及各种癌症;然而,其治疗肝细胞癌(HCC)的有效性经常受到固有和已发展的耐药性的削弱。
    目的:本研究探讨了将安洛替尼(一种广谱酪氨酸激酶抑制剂)与程序性死亡-1(PD-1)阻断相结合的有效性,并为更有效的治疗HCC策略提供了机制见解。
    方法:使用患者来源的器官型组织球体和原位HCC小鼠模型,我们评估了安洛替尼联合PD-1阻断的有效性.对肿瘤免疫微环境和潜在机制的影响使用飞行时间质量细胞计数评估,RNA测序,和跨细胞系的蛋白质组学,小鼠模型,和HCC患者样本。
    结果:在临床前模型中,安洛替尼与抗PD-1抗体的组合增强了针对HCC的免疫应答。安洛替尼通过VEGFR2/AKT/HIF-1α信号轴显著抑制转铁蛋白受体(TFRC)的表达。CD8+T细胞浸润到肿瘤微环境中与TFRC的低表达相关。安洛替尼还增加了趋化因子CXCL14的水平,这对于吸引CD8+T细胞至关重要。CXCL14成为TFRC的下游效应物,TFRC沉默后表现出表达升高。重要的是,低TFRC表达也与更好的预后相关,增强对联合治疗的敏感性,和肝癌患者抗PD-1治疗的良好反应。
    结论:我们的发现强调了安洛替尼通过靶向TFRC和增强CXCL14介导的CD8+T细胞浸润来增强抗PD-1免疫治疗在HCC中的疗效的潜力。这项研究有助于开发新的肝癌治疗策略,强调精准医学在肿瘤学中的作用。
    结论:在HCC临床前模型中证明了安洛替尼和抗PD-1免疫疗法的协同作用。安洛替尼通过VEGFR2/AKT/HIF-1α途径抑制TFRC表达。CXCL14通过TFRC抑制上调促进CD8+T细胞募集。TFRC成为评估晚期HCC患者预后和预测基于抗PD-1疗法反应的潜在生物标志物。
    BACKGROUND: The therapeutic potential of immune checkpoint blockade (ICB) extends across various cancers; however, its effectiveness in treating hepatocellular carcinoma (HCC) is frequently curtailed by both inherent and developed resistance.
    OBJECTIVE: This research explored the effectiveness of integrating anlotinib (a broad-spectrum tyrosine kinase inhibitor) with programmed death-1 (PD-1) blockade and offers mechanistic insights into more effective strategies for treating HCC.
    METHODS: Using patient-derived organotypic tissue spheroids and orthotopic HCC mouse models, we assessed the effectiveness of anlotinib combined with PD-1 blockade. The impact on the tumour immune microenvironment and underlying mechanisms were assessed using time-of-flight mass cytometry, RNA sequencing, and proteomics across cell lines, mouse models, and HCC patient samples.
    RESULTS: The combination of anlotinib with an anti-PD-1 antibody enhanced the immune response against HCC in preclinical models. Anlotinib remarkably suppressed the expression of transferrin receptor (TFRC) via the VEGFR2/AKT/HIF-1α signaling axis. CD8+ T-cell infiltration into the tumour microenvironment correlated with low expression of TFRC. Anlotinib additionally increased the levels of the chemokine CXCL14, crucial for attracting CD8+ T cells. CXCL14 emerged as a downstream effector of TFRC, exhibiting elevated expression following the silencing of TFRC. Importantly, low TFRC expression was also associated with a better prognosis, enhanced sensitivity to combination therapy, and a favourable response to anti-PD-1 therapy in patients with HCC.
    CONCLUSIONS: Our findings highlight anlotinib\'s potential to augment the efficacy of anti-PD-1 immunotherapy in HCC by targeting TFRC and enhancing CXCL14-mediated CD8+ T-cell infiltration. This study contributes to developing novel therapeutic strategies for HCC, emphasizing the role of precision medicine in oncology.
    CONCLUSIONS: Synergistic effects of anlotinib and anti-PD-1 immunotherapy demonstrated in HCC preclinical models. Anlotinib inhibits TFRC expression via the VEGFR2/AKT/HIF-1α pathway. CXCL14 upregulation via TFRC suppression boosts CD8+ T-cell recruitment. TFRC emerges as a potential biomarker for evaluating prognosis and predicting response to anti-PD-1-based therapies in advanced HCC 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
    背景:由于存在由肿瘤细胞和免疫群体之间的相互作用建立的免疫抑制肿瘤微环境(TME),人类肝细胞癌(HCC)获得了对抗癌剂的抗性。迫切需要针对相互作用的新治疗方法,并且对HCC患者有益。本研究旨在探讨中药复方四物汤的抗肿瘤作用及其潜在机制。
    方法:用高效液相色谱-质谱联用法测定SWD的化学谱。评估SWD在消退的HCC中的体外和体内作用。通过过继转移测定确定了骨髓来源的抑制细胞(MDSC)在介导SWD诱导的HCC抑制中的作用。SWD诱导的HCC细胞和MDSC之间的相互作用的调节也在体外和体内得到证实。
    结果:SWD剂量依赖性抑制小鼠原位生长肿瘤中的HCC生长和肺转移,无明显毒性和不良副作用。SWD诱导肝癌细胞坏死,但不直接抑制MDSCs的体外培养,相反,SWD处理的HCC细胞培养上清液通过诱导其细胞凋亡来抑制MDSCs。HCC细胞的坏死反应还可以抑制TME中的MDSC群体而不减少循环MDSC浸润到肿瘤中。MDSCs的过继转移在SWD处理的小鼠中恢复了HCC的肿瘤生长和肺转移。在HCC细胞中,SWD诱导了坏死反应,和肝细胞癌细胞坏死反应的阻断恢复MDSCs群体在体外和体内,并在SWD处理的小鼠中恢复肿瘤生长和肺转移。SWD的组合改善索拉非尼的抗HCC功效而不诱导不良副作用。Albiflorin,SWD的有效化合物,其抗HCC方式已被证实与SWD一致。
    结论:我们的研究首次观察到SWD可以通过TME中肿瘤细胞的坏死调节MDSCs来抑制HCC。SWD的主要有效化合物,albiflorin可以作为人类HCC临床治疗的潜在辅助治疗。
    BACKGROUND: Human hepatocellular carcinoma (HCC) acquired resistance to anti-cancer agents due to the presence of immunosuppressive tumour microenvironment (TME) established by the interaction between tumour cells and immune populations. New treatment targeting the interaction is urgently needed and clinically beneficial to patients with HCC. This study aims to explore the anti-tumour effect of a Traditional Chinese Medicine formula Siwu Decoction (SWD) and its potential mechanism.
    METHODS: The chemical profile of SWD was determined by high-performance liquid chromatography coupled with mass spectrometry. In vitro and in vivo effects of SWD in regressing HCC were assessed. The role of myeloid-derived suppressor cells (MDSCs) in mediating SWD-induced HCC inhibition was determined by adoptive transfer assay. The regulation of SWD-induced interaction between HCC cells and MDSCs was also confirmed both in vitro and in vivo.
    RESULTS: SWD dose-dependent inhibited the HCC growth and lung metastasis in an orthotopic growth tumour in mice, without significant toxicity and adverse side effect. SWD induced necroptosis in HCC cells, but did not directly inhibit in vitro culture of MDSCs, instead, SWD-treated HCC cell culture supernatant suppressed MDSCs by inducing its cell apoptosis. The necroptotic response of HCC cells can also suppress the MDSCs population in the TME without reducing circulating MDSCs infiltration into the tumours. Adoptive transfer of MDSCs recovered tumour growth and lung metastasis of HCC in SWD-treated mice. In HCC cells, SWD induced a necroptotic response, and blockade of necroptotic response in HCC cells recovered the MDSCs population in vitro and in vivo, and restored tumour growth and lung metastasis in SWD-treated mice. A combination of SWD improves the anti-HCC efficacy of sorafenib without inducing adverse side effects. Albiflorin, the effective compound of SWD, its anti-HCC manner has been verified to be consistent with that of SWD.
    CONCLUSIONS: Our study observed for the first time that SWD can suppress HCC by regulating MDSCs through necroptosis of tumour cells in the TME. The main effective compound of SWD, albiflorin can be a potential adjuvant therapy in the clinical management of human HCC.
    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
    低级别浆液性卵巢癌(LGSC)是一种罕见且致命的卵巢癌亚型。LGSC是病态的,生物学,在临床上与更常见的高级别浆液性卵巢癌(HGSC)不同。LGSC起源于浆液性交界性卵巢肿瘤(SBT)。SBT转化为LGSC的机制仍然知之甚少。为了更好地理解LGSC的生物学,我们进行了福尔马林固定的全蛋白质组分析,石蜡包埋的LGSC组织块(n=11),HGSC(n=19),和SBT(n=26)。我们确定整个蛋白质组能够区分卵巢上皮性肿瘤的组织型。与肿瘤微环境相关的蛋白质在LGSC和SBT之间差异表达。成纤维细胞活化蛋白(FAP),在癌症相关的成纤维细胞中表达的蛋白质,是与SBT相比,LGSC中差异最丰富的蛋白质。免疫标志物的多重免疫组织化学(IHC)(CD20,CD79a,进行CD3,CD8和CD68)以确定B细胞的存在,T细胞,和巨噬细胞。LGSCFAP+基质与更丰富的Tregs和M2巨噬细胞相关,SBT中不存在的特征。我们的蛋白质组学队列显示,LGSC的肿瘤微环境与其推定的前体病变相比有变化,SBT。这些变化表明肿瘤微环境为LGSC肿瘤发生和发展提供了支持环境。因此,靶向LGSC的肿瘤微环境可能是一种可行的治疗策略。©2024英国和爱尔兰病理学会。
    Low-grade serous ovarian carcinoma (LGSC) is a rare and lethal subtype of ovarian cancer. LGSC is pathologically, biologically, and clinically distinct from the more common high-grade serous ovarian carcinoma (HGSC). LGSC arises from serous borderline ovarian tumours (SBTs). The mechanism of transformation for SBTs to LGSC remains poorly understood. To better understand the biology of LGSC, we performed whole proteome profiling of formalin-fixed, paraffin-embedded tissue blocks of LGSC (n = 11), HGSC (n = 19), and SBTs (n = 26). We identified that the whole proteome is able to distinguish between histotypes of the ovarian epithelial tumours. Proteins associated with the tumour microenvironment were differentially expressed between LGSC and SBTs. Fibroblast activation protein (FAP), a protein expressed in cancer-associated fibroblasts, is the most differentially abundant protein in LGSC compared with SBT. Multiplex immunohistochemistry (IHC) for immune markers (CD20, CD79a, CD3, CD8, and CD68) was performed to determine the presence of B cells, T cells, and macrophages. The LGSC FAP+ stroma was associated with greater abundance of Tregs and M2 macrophages, features not present in SBTs. Our proteomics cohort reveals that there are changes in the tumour microenvironment in LGSC compared with its putative precursor lesion, SBT. These changes suggest that the tumour microenvironment provides a supportive environment for LGSC tumourigenesis and progression. Thus, targeting the tumour microenvironment of LGSC may be a viable therapeutic strategy. © 2024 The Pathological Society of Great Britain and Ireland.
    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
    ARHGAP家族基因常被用作胶质瘤致癌因子,它们的作用机制仍然无法解释。我们的研究需要对各种神经胶质瘤亚型的免疫微环境和富集途径进行彻底检查。使用CGGA队列开发了一个独特的6基因签名,导致对临床特征差异的洞察,突变模式,和不同风险类别中的免疫细胞浸润。此外,建立了一个独特的列线图,在ARHGAP上接地,DCA曲线说明了该模型在指导治疗策略方面的前瞻性临床实用性。强调ARHGAP30的作用,它是我们模型不可或缺的,通过RT-qPCR证实了其对胶质瘤严重程度的影响和我们的风险评估模型的可信度,蛋白质印迹分析,和细胞功能测定。我们确定了6个与胶质瘤预后相关的ARHGAP家族基因。使用Kaplan-Meier方法的分析表明,神经胶质瘤患者的风险水平升高与不良结局之间存在相关性。风险评分,与肿瘤分期和IDH突变状态有关,成为预测预后的独立因素。高风险类别的患者表现出增加的免疫细胞浸润,增强的肿瘤突变负担,免疫检查点基因更明显的表达,对ICB治疗有更好的反应。一个列线图,将风险评分与胶质瘤患者的病理特征相结合,已开发。DCA分析和细胞研究证实了该模型改善患者临床治疗结果的潜力。一个新的ARHGAP家族基因标记揭示了神经胶质瘤的预后。
    ARHGAP family genes are often used as glioma oncogenic factors, and their mechanism of action remains unexplained. Our research entailed a thorough examination of the immune microenvironment and enrichment pathways across various glioma subtypes. A distinctive 6-gene signature was developed employing the CGGA cohort, leading to insights into the disparities in clinical characteristics, mutation patterns, and immune cell infiltration among distinct risk categories. Additionally, a unique nomogram was established, grounded on ARHGAPs, with DCA curves illustrating the model\'s prospective clinical utility in guiding therapeutic strategies. Emphasizing the role of ARHGAP30, integral to our model, its impact on glioma severity and the credibility of our risk assessment model were substantiated through RT-qPCR, Western blot analysis, and cellular functional assays. We identified 6 ARHGAP family genes associated with glioma prognosis. Analysis using the Kaplan-Meier method indicated a correlation between elevated risk levels and adverse outcomes in glioma patients. The risk score, linked with tumour staging and IDH mutation status, emerged as an independent factor predicting prognosis. Patients in the high-risk category exhibited increased immune cell infiltration, enhanced tumour mutational burden, more pronounced expression of immune checkpoint genes, and a better response to ICB therapy. A nomogram, integrating the risk score with the pathological features of glioma patients, was developed. DCA analysis and cellular studies confirmed the model\'s potential to improve clinical treatment outcomes for patients. A novel ARHGAP family gene signature reveals the prognosis of glioma.
    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
    过度活跃的肿瘤微环境(TME)驱动不受限制的癌细胞存活,耐药性,卵巢癌(OC)的转移。然而,OC的TME内的治疗目标仍然难以捉摸,和量化TME活性的有效方法仍然有限。在这里,我们采用整合的生物信息学方法来确定哪些免疫相关基因(IRGs)调节TME,并进一步评估其在OC进展中的潜在治疗意义(治疗+诊断).使用稳健的方法,我们从癌症基因组图谱(TCGA)数据库中建立了一个预测风险模型,以回顾性分析OC患者的临床病理参数.来自国际癌症基因组联盟(ICGC)队列的数据证实了预后模型的有效性。我们的方法确定了九个IRG,AKT2,FGF7,FOS,IL27RA,LRP1,OBP2A,PAEP,PDGFRA,和PI3,形成OC进展的预后模型,在低危组中区分临床结局明显更好的患者.我们验证了该模型作为一个独立的预后指标,并证明当与临床列线图一起使用时具有增强的预后意义,以进行准确的预测。LRP1表达升高,这表明膀胱癌(BLCA)预后不良,OC,低级别胶质瘤(LGG),和胶质母细胞瘤(GBM),在其他几种癌症中也与免疫浸润有关。与免疫检查点基因(ICG)的显着相关性突出了LRP1作为生物标志物和治疗靶标的潜在重要性。此外,基因集富集分析强调了LRP1参与代谢相关途径,在BLCA中也支持其预后和治疗相关性,OC,低级别胶质瘤(LGG),GBM,肾癌,OC,BLCA,肾透明细胞癌(KIRC),胃腺癌(STAD),和胃和食道癌(STES)。我们的研究在TME中产生了9个IRG在癌症中的新特征,这可能是潜在的预后预测因子,并为改善OC的预后提供了宝贵的资源。
    A hyperactive tumour microenvironment (TME) drives unrestricted cancer cell survival, drug resistance, and metastasis in ovarian carcinoma (OC). However, therapeutic targets within the TME for OC remain elusive, and efficient methods to quantify TME activity are still limited. Herein, we employed an integrated bioinformatics approach to determine which immune-related genes (IRGs) modulate the TME and further assess their potential theragnostic (therapeutic + diagnostic) significance in OC progression. Using a robust approach, we developed a predictive risk model to retrospectively examine the clinicopathological parameters of OC patients from The Cancer Genome Atlas (TCGA) database. The validity of the prognostic model was confirmed with data from the International Cancer Genome Consortium (ICGC) cohort. Our approach identified nine IRGs, AKT2, FGF7, FOS, IL27RA, LRP1, OBP2A, PAEP, PDGFRA, and PI3, that form a prognostic model in OC progression, distinguishing patients with significantly better clinical outcomes in the low-risk group. We validated this model as an independent prognostic indicator and demonstrated enhanced prognostic significance when used alongside clinical nomograms for accurate prediction. Elevated LRP1 expression, which indicates poor prognosis in bladder cancer (BLCA), OC, low-grade gliomas (LGG), and glioblastoma (GBM), was also associated with immune infiltration in several other cancers. Significant correlations with immune checkpoint genes (ICGs) highlight the potential importance of LRP1 as a biomarker and therapeutic target. Furthermore, gene set enrichment analysis highlighted LRP1\'s involvement in metabolism-related pathways, supporting its prognostic and therapeutic relevance also in BLCA, OC, low-grade gliomas (LGG), GBM, kidney cancer, OC, BLCA, kidney renal clear cell carcinoma (KIRC), stomach adenocarcinoma (STAD), and stomach and oesophageal carcinoma (STES). Our study has generated a novel signature of nine IRGs within the TME across cancers, that could serve as potential prognostic predictors and provide a valuable resource to improve the prognosis of OC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号