TME

TME
  • 文章类型: Journal Article
    肺癌(LC)是全球最常见的癌症之一。肿瘤相关巨噬细胞(TAMs)是肿瘤微环境(TME)的重要组成部分,与肿瘤的发生分期密切相关。发展,和转移。巨噬细胞是可塑的,在TME中不同信号通路的影响下,可以分化为不同的表型和功能。经典活化的(M1样)和交替活化的(M2样)代表巨噬细胞的两种极化状态。M1巨噬细胞表现出抗肿瘤功能,而M2巨噬细胞被认为支持肿瘤细胞存活和转移。巨噬细胞极化涉及复杂的信号通路,而阻断或调节这些信号通路以增强巨噬细胞的抗肿瘤作用已成为近年来的研究热点。同时,关于通过合成和天然药物成分将TAM调节为抗肿瘤表型的新发现。纳米技术能更好地实现药物的联合治疗和靶向给药,最大限度地提高药物的疗效,同时最大限度地减少副作用。到目前为止,纳米药物靶向递送各种活性物质用于重编程TAMs已取得重大进展。在这次审查中,我们主要提供了TAM和LC微环境中各种细胞之间的信号串扰的全面概述。此外,本文还综述了新型药物和靶向巨噬细胞的纳米给药系统(NDDSs)的最新进展.最后,我们讨论了巨噬细胞作为治疗靶点的前景和临床转化的障碍。
    Lung cancer (LC) is one of the most common cancer worldwide. Tumor-associated macrophages (TAMs) are important component of the tumor microenvironment (TME) and are closely related to the stages of tumor occurrence, development, and metastasis. Macrophages are plastic and can differentiate into different phenotypes and functions under the influence of different signaling pathways in TME. The classically activated (M1-like) and alternatively activated (M2-like) represent the two polarization states of macrophages. M1 macrophages exhibit anti-tumor functions, while M2 macrophages are considered to support tumor cell survival and metastasis. Macrophage polarization involves complex signaling pathways, and blocking or regulating these signaling pathways to enhance macrophages\' anti-tumor effects has become a research hotspot in recent years. At the same time, there have been new discoveries regarding the modulation of TAMs towards an anti-tumor phenotype by synthetic and natural drug components. Nanotechnology can better achieve combination therapy and targeted delivery of drugs, maximizing the efficacy of the drugs while minimizing side effects. Up to now, nanomedicines targeting the delivery of various active substances for reprogramming TAMs have made significant progress. In this review, we primarily provided a comprehensive overview of the signaling crosstalk between TAMs and various cells in the LC microenvironment. Additionally, the latest advancements in novel drugs and nano-based drug delivery systems (NDDSs) that target macrophages were also reviewed. Finally, we discussed the prospects of macrophages as therapeutic targets and the barriers to clinical translation.
    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
    胶质母细胞瘤,一种高度恶性的颅内肿瘤,治疗进展缓慢。先前涉及靶向治疗和免疫检查点抑制剂的临床试验显示在治疗胶质母细胞瘤中没有显著益处。这种无效性主要是由于胶质母细胞瘤的复杂免疫抑制环境。胶质母细胞瘤细胞具有较低的免疫原性和较强的异质性,免疫微环境中充斥着抑制性细胞因子,许多免疫抑制细胞,有效T细胞不足。幸运的是,最近CART治疗胶质母细胞瘤的I期临床试验证实了其安全性,一小部分患者获得了生存益处。然而,CART治疗继续面临挑战,包括血脑屏障阻塞,抗原损失,和免疫抑制肿瘤微环境(TME)。本文提供了胶质母细胞瘤的免疫微环境的详细检查,来自内在和外在的肿瘤细胞因子,回顾当前多靶点CART治疗的临床和基础研究,最后概述了使用CART细胞治疗胶质母细胞瘤的关键挑战。
    Glioblastoma, a highly malignant intracranial tumor, has acquired slow progress in treatment. Previous clinical trials involving targeted therapy and immune checkpoint inhibitors have shown no significant benefits in treating glioblastoma. This ineffectiveness is largely due to the complex immunosuppressive environment of glioblastoma. Glioblastoma cells exhibit low immunogenicity and strong heterogeneity and the immune microenvironment is replete with inhibitory cytokines, numerous immunosuppressive cells, and insufficient effective T cells. Fortunately, recent Phase I clinical trials of CART therapy for glioblastoma have confirmed its safety, with a small subset of patients achieving survival benefits. However, CART therapy continues to face challenges, including blood-brain barrier obstruction, antigen loss, and an immunosuppressive tumor microenvironment (TME). This article provides a detailed examination of glioblastoma\'s immune microenvironment, both from intrinsic and extrinsic tumor cell factors, reviews current clinical and basic research on multi-targets CART treatment, and concludes by outlining the key challenges in using CART cells for glioblastoma therapy.
    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
    脂肪组织功能障碍,这与结直肠癌(CRC)的风险增加有关,是肥胖病理生理学的重要因素。肥胖相关的炎症和细胞外基质(ECM)重塑通过塑造肿瘤微环境(TME)促进结直肠癌转移(CRCM)。当CRC发生时,肿瘤细胞的代谢共生将邻近的脂肪细胞募集到TME中提供能量。同时,丰富的免疫细胞,来自脂肪组织和血液,被招募加入TME,它受到促炎因子的刺激,并引发慢性局部促炎TME。异常调节的ECM蛋白和细胞表面粘附分子增强ECM重塑并进一步增加肿瘤和基质细胞之间的收缩性。促进上皮-间质转化(EMT)。EMT增加肿瘤迁移和侵入周围组织或血管并加速CRCM。结直肠共生菌群在促进CRCM方面也起着重要作用。在这次审查中,我们提供脂肪组织及其对CRC的贡献,特别强调脂肪细胞的作用,巨噬细胞,中性粒细胞,T细胞,ECM,和共生肠道菌群在CRC进展中的作用及其对CRC微环境的贡献。我们强调脂肪细胞和肿瘤细胞之间的相互作用,以及针对这些相互作用的潜在治疗方法。
    Adipose tissue dysfunction, which is associated with an increased risk of colorectal cancer (CRC), is a significant factor in the pathophysiology of obesity. Obesity-related inflammation and extracellular matrix (ECM) remodeling promote colorectal cancer metastasis (CRCM) by shaping the tumor microenvironment (TME). When CRC occurs, the metabolic symbiosis of tumor cells recruits adjacent adipocytes into the TME to supply energy. Meanwhile, abundant immune cells, from adipose tissue and blood, are recruited into the TME, which is stimulated by pro-inflammatory factors and triggers a chronic local pro-inflammatory TME. Dysregulated ECM proteins and cell surface adhesion molecules enhance ECM remodeling and further increase contractibility between tumor and stromal cells, which promotes epithelial-mesenchymal transition (EMT). EMT increases tumor migration and invasion into surrounding tissues or vessels and accelerates CRCM. Colorectal symbiotic microbiota also plays an important role in the promotion of CRCM. In this review, we provide adipose tissue and its contributions to CRC, with a special emphasis on the role of adipocytes, macrophages, neutrophils, T cells, ECM, and symbiotic gut microbiota in the progression of CRC and their contributions to the CRC microenvironment. We highlight the interactions between adipocytes and tumor cells, and potential therapeutic approaches to target these interactions.
    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
    脂肪来源的干细胞(ASC)显著影响肿瘤微环境(TME)内的肿瘤进展。这篇综述探讨了ASC的促肿瘤发生作用,专注于旁分泌信号,直接的细胞-细胞相互作用,和免疫调节。ASC介导的通过隧道纳米管(TNTs)和间隙连接(GJs)的线粒体转移在增强癌细胞存活和代谢中起着重要作用。具有功能失调的线粒体的癌细胞从ASC获得线粒体以满足其代谢需求并在TME中茁壮成长。靶向线粒体转移,调节ASC功能,和影响代谢途径是潜在的治疗策略。然而,像TME复杂性这样的挑战,特异性,安全问题,必须解决抵抗机制。破坏ASC-癌细胞-线粒体轴提供了一种有希望的癌症治疗方法。
    Adipose-derived stem cells (ASCs) significantly influence tumor progression within the tumor microenvironment (TME). This review examines the pro-tumorigenic roles of ASCs, focusing on paracrine signaling, direct cell-cell interactions, and immunomodulation. ASC-mediated mitochondrial transfer through tunneling nanotubes (TNTs) and gap junctions (GJs) plays a significant role in enhancing cancer cell survival and metabolism. Cancer cells with dysfunctional mitochondria acquire mitochondria from ASCs to meet their metabolic needs and thrive in the TME. Targeting mitochondrial transfer, modulating ASC function, and influencing metabolic pathways are potential therapeutic strategies. However, challenges like TME complexity, specificity, safety concerns, and resistance mechanisms must be addressed. Disrupting the ASC-cancer cell-mitochondria axis offers a promising approach to cancer 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
    布法林,来自中国蟾蜍(Bufogargarizans)的强心类固醇,已经在各种癌症类型中表现出有效的抗癌特性,将其定位为有前途的治疗候选。然而,缺乏针对头颈部癌症的全面机制研究。我们的研究旨在通过研究bufalin在头颈部癌细胞中的作用机制来弥合这一差距。使用几种方法,比如西方印迹,免疫荧光,和流式细胞术,我们观察到bufalin的剂量依赖性细胞活力降低,细胞膜完整性的破坏,以及抑制HPV阳性和HPV阴性细胞系中的集落形成。Bufalin通过调节凋亡相关蛋白诱导凋亡,线粒体功能,和活性氧的产生。它还可以在G2/M期阻止细胞周期并减弱细胞迁移,同时影响上皮-间质转化标志物并靶向关键信号通路,包括Wnt/β-catenin,EGFR,和NF-κB。此外,bufalin通过将巨噬细胞极化为M1表型而发挥免疫调节作用,增强抗肿瘤免疫反应。这些发现强调了bufalin作为针对头颈部癌症的多方面治疗剂的潜力,靶向参与增殖的必要途径,凋亡,细胞周期调节,转移,和免疫调节。需要进一步的研究来验证这些机制并优化bufalin的临床应用。
    Bufalin, a cardiotonic steroid derived from the Chinese toad (Bufo gargarizans), has demonstrated potent anticancer properties across various cancer types, positioning it as a promising therapeutic candidate. However, comprehensive mechanistic studies specific to head and neck cancers have been lacking. Our study aimed to bridge this gap by investigating bufalin\'s mechanisms of action in head and neck cancer cells. Using several methods, such as Western blotting, immunofluorescence, and flow cytometry, we observed bufalin\'s dose-dependent reduction in cell viability, disruption of cell membrane integrity, and inhibition of colony formation in both HPV-positive and HPV-negative cell lines. Bufalin induces apoptosis through the modulation of apoptosis-related proteins, mitochondrial function, and reactive oxygen species production. It also arrests the cell cycle at the G2/M phase and attenuates cell migration while affecting epithelial-mesenchymal transition markers and targeting pivotal signaling pathways, including Wnt/β-catenin, EGFR, and NF-κB. Additionally, bufalin exerted immunomodulatory effects by polarizing macrophages toward the M1 phenotype, bolstering antitumor immune responses. These findings underscore bufalin\'s potential as a multifaceted therapeutic agent against head and neck cancers, targeting essential pathways involved in proliferation, apoptosis, cell cycle regulation, metastasis, and immune modulation. Further research is warranted to validate these mechanisms and optimize bufalin\'s clinical application.
    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
    皮肤黑素瘤是所有皮肤肿瘤中最致命的。最近,突起,一种新形式的细胞死亡与氧化磷酸化有关,已经成为一个重要因素。然而,角化瘤在黑色素瘤中的确切作用尚不清楚.我们的研究探索了角化相关基因之间的潜在联系,预后,免疫微环境,和黑色素瘤治疗。重要的是,在黑素瘤和正常组织之间显示出显著的差异,确定它们与黑色素瘤的相关性。新开发的角化相关基因标签(CGS)显示出预测黑色素瘤总生存期(OS)的强大能力。我们构建了一个新的列线图,将临床特征与CGS相结合,以提高预测准确性。此外,这项研究揭示了CGS和免疫细胞群之间的相关性,包括CD8+T细胞,Tfh细胞,B细胞,和骨髓来源的抑制细胞。在CGS内,在黑色素瘤中,肽基丙氨酰异构酶C(PPIC)与不良预后和耐药性最密切相关。PPIC被确定为黑色素瘤进展的启动子,增强细胞侵袭力,同时抑制CD8+T细胞活化。这项全面的研究不仅阐明了CGS之间的复杂联系,黑色素瘤预后,免疫微环境,和耐药性,但也提供了令人信服的证据支持PPIC作为预测黑色素瘤治疗中OS的有希望的生物标志物。
    Cutaneous melanoma is the most lethal of all skin tumors. Recently, cuproptosis, a novel form of cell death linked to oxidative phosphorylation, has emerged as an important factor. However, the precise role of cuproptosis in melanoma remains unclear. Our research explored the potential links between cuproptosis-related genes, prognosis, immune microenvironments, and melanoma treatments. Significantly, cuproptosis regulators showed remarkable differences between melanoma and normal tissues, establishing their relevance to melanoma. The newly developed cuproptosis-related gene signature (CGS) demonstrated a robust ability to predict overall survival (OS) in melanoma. We constructed a novel nomogram that combined clinical features with CGS to improve predictive accuracy. In addition, the study revealed correlations between CGS and immune cell populations, including CD8+T cells, Tfh cells, B cells, and myeloid-derived suppressor cells. Within the CGS, Peptidylprolyl isomerase C (PPIC) emerged as the most strongly associated with poor prognosis and drug resistance in melanoma. PPIC was identified as a promoter of melanoma progression, enhancing cell invasiveness while concurrently suppressing CD8+T cell activation. This comprehensive study not only elucidated the intricate connections between CGS, melanoma prognosis, immune microenvironment, and drug resistance but also provided compelling evidence supporting PPIC as a promising biomarker for predicting OS in melanoma treatment.
    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
    HIGD1B(HIG1缺氧诱导域家族成员1B)是与各种疾病的发生和发展有关的蛋白质编码基因。然而,其在胃癌(GC)中的确切功能尚不清楚。
    通过TCGA和GEO数据库确定HIGD1B的表达,并使用实验进行验证。通过Kaplan-Meier(K-M)曲线分析HIGD1B与GC患者预后之间的关联。随后,研究人员利用ROC曲线评估HIGD1B的诊断能力,并利用COX分析调查GC的危险因素.然后对差异表达基因(DEGs)进行功能富集分析,并生成列线图来预测GC患者的生存结局和概率。此外,我们评估了HIGD1B与免疫细胞浸润之间的相互作用,并预测了GC患者对治疗的敏感性。
    HIGD1B在GC组织和细胞系中明显升高,HIGD1B高表达的患者预后较差。此外,HIGD1B与不同的等级有关,阶段,T阶段。HIGD1B和列线图5年的生存ROC曲线分别为0.741和0.735,提示诊断效能水平合适。根据Cox回归分析,HIGD1B代表胃癌预后的单独风险因素(p<0.01)。GSEA分析表明HIGD1B与癌症形成和晚期通路密切相关。此外,HIGD1B高表达的患者表现出更高水平的肿瘤浸润免疫细胞(TIIC),化疗和免疫治疗后更有可能出现免疫逃逸和耐药.
    本研究探讨了HIGD1B在GC中的潜在机制以及诊断和预后用途,以及将HIGD1B鉴定为有价值的生物标志物和可能的GC治疗靶标。
    UNASSIGNED: HIGD1B (HIG1 Hypoxia Inducible Domain Family Member 1B) is a protein-coding gene linked to the occurrence and progression of various illnesses. However, its precise function in gastric cancer (GC) remains unclear.
    UNASSIGNED: The expression of HIGD1B is determined through the TCGA and GEO databases and verified using experiments. The association between HIGD1B and GC patients\' prognosis was analyzed via the Kaplan-Meier (K-M) curve. Subsequently, the researchers utilized ROC curves to assess the diagnostic capacity of HIGD1B and employed COX analysis to investigate risk factors for GC. The differentially expressed genes (DEGs) were then subjected to functional enrichment analysis, and a nomogram was generated to forecast the survival outcome and probability of GC patients. Additionally, we evaluated the interaction between HIGD1B and the immune cell infiltration and predicted the susceptibility of GC patients to therapy.
    UNASSIGNED: HIGD1B is markedly elevated in GC tissue and cell lines, and patients with high HIGD1B expression have a poorer outcome. In addition, HIGD1B is related to distinct grades, stages, and T stages. The survival ROC curves of HIGD1B and nomogram for five years were 0.741 and 0.735, suggesting appropriate levels of diagnostic efficacy. According to Cox regression analysis, HIGD1B represents a separate risk factor for the prognosis of gastric cancer (p<0.01). GSEA analysis demonstrated that the HIGD1B is closely related to cancer formation and advanced pathways. Moreover, patients with high HIGD1B expression exhibited a higher level of Tumor-infiltration immune cells (TIICs) and were more likely to experience immune escape and drug resistance after chemotherapy and immunotherapy.
    UNASSIGNED: This study explored the potential mechanisms and diagnostic and prognostic utility of HIGD1B in GC, as well as identified HIGD1B as a valuable biomarker and possible therapeutic target for GC.
    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:炎症在肿瘤发展中起关键作用。炎症细胞浸润和炎症介质合成导致几种癌症中肿瘤微环境(TME)的变化,尤其是在肝内胆管细胞癌(ICC)。然而,使用可靠的生物标志物确定患者炎症状态的方法仍在探索中。
    方法:我们从已发表的研究中检索了244例ICC患者的RNA测序和体细胞突变分析结果以及临床特征。我们进行了一致聚类以鉴定与炎症相关的分子亚型。我们比较了预后模式,临床特征,体细胞突变谱,和炎症亚型的免疫细胞浸润模式。我们进行了定量实时聚合酶链反应(qRT-PCR)和免疫组织化学(IHC)以确认基因表达。我们进行了逻辑回归分析,以构建预测ICC患者炎症状态的列线图。
    结果:我们的结果证实ICC可以分为炎症高亚型(IHS)和炎症低亚型(ILS)。每个组的患者都有不同的预后,临床特征,和TME组成。IHS组的ICC患者由于免疫抑制微环境以及KRAS和TP53突变的频率高,预后较差。癌相关成纤维细胞(CAF)衍生的COLEC11减少髓样炎症细胞浸润并减弱炎症反应。qRT-PCR和IHC实验的结果证实,与癌旁组织相比,COLEC11在肿瘤组织中的表达水平显着降低。IHS组的ICC患者更有可能对免疫检查点抑制剂(ICIs)治疗产生反应,因为他们的肿瘤突变负担(TMB)评分较高。肿瘤新抗原负荷(TNB)评分,新抗原计数,和免疫检查点表达水平。最后,我们根据ICC患者的临床特征和炎症基因表达水平,建立了一个有效预测ICC患者炎症状态的列线图.我们评估了校准结果,潜在的歧视,和列线图的临床实用性。
    结论:IHS的炎症反应主要由髓系细胞诱导。COLEC11可以降低这组细胞的浸润水平,髓样炎症细胞可能是ICC治疗的新靶点。我们开发了一种新的列线图,可以有效地预测ICC患者的炎症状态,这将有助于指导个性化治疗计划。
    BACKGROUND: Inflammation plays a critical role in tumor development. Inflammatory cell infiltration and inflammatory mediator synthesis cause changes in the tumor microenvironment (TME) in several cancers, especially in intrahepatic cholangiocellular carcinoma (ICC). However, methods to ascertain the inflammatory state of patients using reliable biomarkers are still being explored.
    METHODS: We retrieved the RNA sequencing and somatic mutation analyses results and the clinical characteristics of 244 patients with ICC from published studies. We performed consensus clustering to identify the molecular subtypes associated with inflammation. We compared the prognostic patterns, clinical characteristics, somatic mutation profiles, and immune cell infiltration patterns across inflammatory subtypes. We performed quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) to confirm gene expression. We performed logistic regression analyses to construct a nomogram predicting the inflammatory status of patients with ICC.
    RESULTS: Our results confirmed that ICC can be categorized into an inflammation-high subtype (IHS) and an inflammation-low subtype (ILS). Patients from each group had distinct prognosis, clinical characteristics, and TME composition. Patients with ICC in the IHS group showed poorer prognosis owing to the immunosuppressive microenvironment and high frequency of KRAS and TP53 mutations. Cancer-associated fibroblast (CAF)-derived COLEC11 reduced myeloid inflammatory cell infiltration and attenuated inflammatory responses. The results of qRT-PCR and IHC experiments confirmed that COLEC11 expression levels were significantly reduced in tumor tissues compared to those in paracancerous tissues. Patients with ICC in the IHS group were more likely to respond to treatment with immune checkpoint inhibitors (ICIs) owing to their higher tumor mutational burden (TMB) scores, tumor neoantigen burden (TNB) scores, neoantigen counts, and immune checkpoint expression levels. Finally, we developed a nomogram to effectively predict the inflammatory status of patients with ICC based on their clinical characteristics and inflammatory gene expression levels. We evaluated the calibration, discrimination potential, and clinical utility of the nomogram.
    CONCLUSIONS: The inflammatory response in IHS is primarily induced by myeloid cells. COLEC11 can reduce the infiltration level of this group of cells, and myeloid inflammatory cells may be a novel target for ICC treatment. We developed a novel nomogram that could effectively predict the inflammatory state of patients with ICC, which will be useful for guiding individualized treatment plans.
    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:过度的周细胞覆盖促进肿瘤生长,而下调可能会解决这一困境。由于血管周细胞在肿瘤微环境(TME)中的双刃剑作用,伊马替尼不加选择地降低周细胞覆盖率会导致不良的治疗结局.这里,我们优化了在高周细胞覆盖状态的结直肠癌(CRC)模型中使用伊马替尼,并揭示了9.4T时多参数磁共振成像(mpMRI)在监测与治疗相关的周细胞覆盖率和TME变化中的价值。
    方法:通过组织学血管表征和mpMRI评估CRC异种移植模型。周细胞覆盖率最高的小鼠用伊马替尼或盐水治疗;然后,血管特征,对肿瘤细胞凋亡和HIF-1α水平进行组织学分析,通过qPCR评估Bcl-2/bax通路表达的改变。通过动态对比增强(DCE)监测伊马替尼的效果-,扩散加权成像(DWI)-和酰胺质子转移化学交换饱和转移(APTCEST)-MRI在9.4T。
    结果:DCE参数提供了与肿瘤血管特征良好的组织学匹配。在高周细胞覆盖率状态下,伊马替尼表现出显著的肿瘤生长抑制,坏死增加和周细胞覆盖率下调,这些变化伴随着血管渗透性的增加,微血管密度(MVD)降低,肿瘤细胞凋亡增加,凋亡相关Bcl-2/bax通路基因表达改变。战略上,4天伊马替尼有效降低周细胞覆盖率和HIF-1α水平,连续治疗导致周细胞覆盖率下降不明显,HIF-1α水平再次升高。相关性分析证实了使用mpMRI参数监测伊马替尼治疗的可行性,DCE衍生的Ve和Ktrans与周细胞覆盖率最相关,Ve与血管渗透性,AUC与微血管密度(MVD),DWI衍生的ADC与肿瘤凋亡,和APTCEST衍生的MTRasym在1µT与HIF-1α。
    结论:这些结果提供了优化的伊马替尼方案,以在高周细胞覆盖率CRC模型中降低周细胞覆盖率和HIF-1α水平,并提供了一种超高场多参数MRI方法,用于监测周细胞覆盖率和TME对治疗的动力学反应。
    BACKGROUND: Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME.
    METHODS: CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T.
    RESULTS: The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α.
    CONCLUSIONS: These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to 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
    结肠直肠癌(CRC)对公共卫生构成挑战,其特征是发病率高。本研究探讨了CRC患者肿瘤微环境(TME)中铁死亡与脂肪酸代谢之间的关系,以确定这些相互作用如何影响免疫治疗的预后和有效性。关注患者预后和预测治疗反应的潜力。
    使用来自多个队列的数据集,包括癌症基因组图谱(TCGA)和基因表达综合(GEO),我们进行了一项深入的多组学研究,以揭示铁凋亡调节因子与CRC脂肪酸代谢之间的关系.通过无监督聚类,我们发现了将铁死亡和脂肪酸代谢联系起来的独特模式,并在免疫细胞浸润和途径分析的背景下进一步研究了它们。我们开发了FeFAMscore,使用机器学习算法的组合创建的预后模型,并评估其对患者预后和治疗反应性的预测能力。使用RT-PCR确认FeFAMscore签名表达水平,ACAA2在癌症中的进展得到进一步证实。
    这项研究揭示了铁凋亡调节因子和脂肪酸代谢相关基因在肿瘤进展方面的显著相关性。确定了具有不同预后和免疫细胞浸润的三个不同患者群。FeFAMscore显示出比现有模型更高的预后准确性,训练队列的C指数为0.689,四个独立验证队列的值范围为0.648至0.720。它也对免疫疗法和化疗有反应,表明特殊疗法的敏感反应(例如,抗PD-1,抗CTLA4,奥希替尼)在高FeFAM评分患者中。
    铁凋亡调节因子和脂肪酸代谢相关基因不仅增强免疫激活,但也有助于免疫逃逸。因此,FeFAMscore,一种新的预后工具,对于预测CRC患者的免疫治疗策略的预后和疗效是有希望的。
    UNASSIGNED: Colorectal cancer (CRC) poses a challenge to public health and is characterized by a high incidence rate. This study explored the relationship between ferroptosis and fatty acid metabolism in the tumor microenvironment (TME) of patients with CRC to identify how these interactions impact the prognosis and effectiveness of immunotherapy, focusing on patient outcomes and the potential for predicting treatment response.
    UNASSIGNED: Using datasets from multiple cohorts, including The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we conducted an in-depth multi-omics study to uncover the relationship between ferroptosis regulators and fatty acid metabolism in CRC. Through unsupervised clustering, we discovered unique patterns that link ferroptosis and fatty acid metabolism, and further investigated them in the context of immune cell infiltration and pathway analysis. We developed the FeFAMscore, a prognostic model created using a combination of machine learning algorithms, and assessed its predictive power for patient outcomes and responsiveness to treatment. The FeFAMscore signature expression level was confirmed using RT-PCR, and ACAA2 progression in cancer was further verified.
    UNASSIGNED: This study revealed significant correlations between ferroptosis regulators and fatty acid metabolism-related genes with respect to tumor progression. Three distinct patient clusters with varied prognoses and immune cell infiltration were identified. The FeFAMscore demonstrated superior prognostic accuracy over existing models, with a C-index of 0.689 in the training cohort and values ranging from 0.648 to 0.720 in four independent validation cohorts. It also responses to immunotherapy and chemotherapy, indicating a sensitive response of special therapies (e.g., anti-PD-1, anti-CTLA4, osimertinib) in high FeFAMscore patients.
    UNASSIGNED: Ferroptosis regulators and fatty acid metabolism-related genes not only enhance immune activation, but also contribute to immune escape. Thus, the FeFAMscore, a novel prognostic tool, is promising for predicting both the prognosis and efficacy of immunotherapeutic strategies in patients with CRC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号