TAMs

TAM
  • 文章类型: Journal Article
    背景:乳腺癌的内部异质性,特别是由恶性和非恶性细胞组成的肿瘤微环境(TME),近年来进行了广泛的探索。这种复杂的细胞生态系统中的细胞通过表型改变激活或抑制肿瘤免疫,代谢产物的分泌和细胞-细胞通讯网络。巨噬细胞,作为TME中最丰富的免疫细胞,被恶性细胞募集并经历表型重塑。肿瘤相关巨噬细胞(TAMs)表现出多种亚型和功能,在影响肿瘤免疫方面发挥重要作用。然而,它们的精确亚型划分和特定功能仍未充分定义。
    方法:将来自8名具有不同分子亚型和分期的乳腺癌患者的49,141个细胞的公开单细胞转录组纳入我们的研究。采用无监督聚类和手动细胞注释来准确地分类TAM亚型。然后,我们进行了功能分析,并构建了TAM亚型的发展轨迹。随后,以内皮细胞(ECs)和T细胞为关键节点,探讨了TAM亚型在TME内细胞间通讯网络中的作用.最后,在另一个独立发表的scRNA数据集中重复分析,以验证我们对TAM表征的发现.
    结果:TAM被准确地分为7种亚型,显示抗肿瘤或促肿瘤作用。第一次,我们确定了一种新的能够在乳腺癌中增殖和扩增的TAM亚型-TUBA1B+TAM在TAM的多样性和肿瘤进展中起关键作用.发育轨迹说明了TAM如何在TME内重塑并经历表型和功能变化,在初始点使用TUBA1B+TAM。值得注意的是,主要的TAM亚型因乳腺癌的不同分子亚型和分期而异.此外,我们对细胞-细胞通信网络的研究表明,TAM通过直接调节内在免疫而发挥作用,通过T细胞间接调节适应性免疫,以及通过ECs影响肿瘤血管生成和淋巴管生成。
    结论:我们的研究建立了精确的乳腺癌TAMs单细胞图谱,阐明它们在肿瘤生物学中的多方面作用,并为乳腺癌免疫治疗中的靶向TAM提供资源。
    BACKGROUND: The internal heterogeneity of breast cancer, notably the tumor microenvironment (TME) consisting of malignant and non-malignant cells, has been extensively explored in recent years. The cells in this complex cellular ecosystem activate or suppress tumor immunity through phenotypic changes, secretion of metabolites and cell-cell communication networks. Macrophages, as the most abundant immune cells within the TME, are recruited by malignant cells and undergo phenotypic remodeling. Tumor-associated macrophages (TAMs) exhibit a variety of subtypes and functions, playing significant roles in impacting tumor immunity. However, their precise subtype delineation and specific function remain inadequately defined.
    METHODS: The publicly available single-cell transcriptomes of 49,141 cells from eight breast cancer patients with different molecular subtypes and stages were incorporated into our study. Unsupervised clustering and manual cell annotation were employed to accurately classify TAM subtypes. We then conducted functional analysis and constructed a developmental trajectory for TAM subtypes. Subsequently, the roles of TAM subtypes in cell-cell communication networks within the TME were explored using endothelial cells (ECs) and T cells as key nodes. Finally, analyses were repeated in another independent publish scRNA datasets to validate our findings for TAM characterization.
    RESULTS: TAMs are accurately classified into 7 subtypes, displaying anti-tumor or pro-tumor roles. For the first time, we identified a new TAM subtype capable of proliferation and expansion in breast cancer-TUBA1B+ TAMs playing a crucial role in TAMs diversity and tumor progression. The developmental trajectory illustrates how TAMs are remodeled within the TME and undergo phenotypic and functional changes, with TUBA1B+ TAMs at the initial point. Notably, the predominant TAM subtypes varied across different molecular subtypes and stages of breast cancer. Additionally, our research on cell-cell communication networks shows that TAMs exert effects by directly modulating intrinsic immunity, indirectly regulating adaptive immunity through T cells, as well as influencing tumor angiogenesis and lymphangiogenesis through ECs.
    CONCLUSIONS: Our study establishes a precise single-cell atlas of breast cancer TAMs, shedding light on their multifaceted roles in tumor biology and providing resources for targeting TAMs in breast cancer 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
    胶质母细胞瘤(GBM)表现出浸润性生长特征,招募邻近的正常细胞以促进肿瘤生长,维护,侵入大脑。虽然血脑屏障是中枢神经系统的重要自然防御机制,GBM打破了这个屏障,导致巨噬细胞从外周骨髓浸润并激活常驻小胶质细胞。单细胞转录组学和空间转录组学的最新进展已经完善了肿瘤微环境中细胞的分类,以进行精确识别。简要概述了多组学条件下肿瘤微环境中复杂的相互作用和对细胞生长的影响。涉及小胶质细胞的因素和机制,巨噬细胞,内皮细胞,单独检查影响GBM生长的T细胞。肿瘤微环境中肿瘤细胞-免疫细胞相互作用的协同机制协同促进肿瘤细胞的生长,渗透,和胶质瘤的转移,同时也影响肿瘤微环境的免疫状态和治疗反应。随着免疫疗法的不断进步,靶向肿瘤间微环境中的细胞成为GBM的一种有前景的新型治疗方法。通过全面了解和干预肿瘤微环境中复杂的细胞相互作用,可以开发新的治疗方式来提高GBM患者的治疗效果.
    Glioblastoma (GBM) displays an infiltrative growth characteristic that recruits neighboring normal cells to facilitate tumor growth, maintenance, and invasion into the brain. While the blood-brain barrier serves as a critical natural defense mechanism for the central nervous system, GBM disrupts this barrier, resulting in the infiltration of macrophages from the peripheral bone marrow and the activation of resident microglia. Recent advancements in single-cell transcriptomics and spatial transcriptomics have refined the categorization of cells within the tumor microenvironment for precise identification. The intricate interactions and influences on cell growth within the tumor microenvironment under multi-omics conditions are succinctly outlined. The factors and mechanisms involving microglia, macrophages, endothelial cells, and T cells that impact the growth of GBM are individually examined. The collaborative mechanisms of tumor cell-immune cell interactions within the tumor microenvironment synergistically promote the growth, infiltration, and metastasis of gliomas, while also influencing the immune status and therapeutic response of the tumor microenvironment. As immunotherapy continues to progress, targeting the cells within the inter-tumor microenvironment emerges as a promising novel therapeutic approach for GBM. By comprehensively understanding and intervening in the intricate cellular interactions within the tumor microenvironment, novel therapeutic modalities may be developed to enhance treatment outcomes for patients with GBM.
    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
    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
    Lenvatinib,多靶点激酶抑制剂,已被证明是有效的治疗晚期肝细胞癌。以前已经证明,肿瘤组织中的肿瘤相关巨噬细胞(TAMs)可以促进HCC生长,侵袭和转移。此外,lenvatinib对肝癌的治疗具有一定的免疫调节作用。然而,lenvatinib在HCC治疗过程中巨噬细胞极化中的作用尚未得到充分研究.在这项研究中,我们在体外和体内使用了多种实验方法来研究lenvatinib在HCC进展过程中对TAMs的影响.这项研究首次表明lenvatinib可以改变人类和小鼠的巨噬细胞极化。此外,用lenvatinib处理的巨噬细胞在体外显示增强的经典激活的巨噬细胞(M1)活性和抑制肝癌细胞增殖,入侵,和移民。此外,在乐伐替尼诱导的M1巨噬细胞极化过程中,STAT-1是主要的靶转录因子,抑制STAT-1活性逆转了乐伐替尼的作用。总的来说,本研究为乐伐替尼治疗HCC的免疫调节功能提供了理论依据。
    Lenvatinib, a multitarget kinase inhibitor, has been proven to be effective in the treatment of advanced hepatocellular carcinoma. It has been previously demonstrated that tumour associated macrophages (TAMs) in tumour tissues can promote HCC growth, invasion and metastasis. Furthermore, lenvatinib has certain immunomodulatory effects on the treatment of HCC. However, the role of lenvatinib in macrophage polarization during HCC treatment has not been fully explored. In this study, we used a variety of experimental methods both in vitro and in vivo to investigate the effect of lenvatinib on TAMs during HCC progression. This study is the first to show that lenvatinib can alter macrophage polarization in both humans and mice. Moreover, macrophages treated with lenvatinib in vitro displayed enhanced classically activated macrophages (M1) activity and suppressed liver cancer cell proliferation, invasion, and migration. Furthermore, during the progression of M1 macrophage polarization induced by lenvatinib, STAT-1 was the main target transcription factor, and inhibiting STAT-1 activity reversed the effect of lenvatinib. Overall, the present study provides a theoretical basis for the immunomodulatory function of lenvatinib in the treatment of 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
    脑转移瘤(BMs)是非小细胞肺癌(NSCLC)患者最常见的转移部位。然而,由于血脑屏障,BMS对免疫疗法没有反应。这是因为颅内免疫细胞如M2肿瘤相关巨噬细胞(TAMs)的积累,创造免疫抑制肿瘤微环境.在这项研究中,我们重点研究了可以穿过血脑屏障并影响颅内免疫微环境的辐射肿瘤细胞释放微粒(RT-MPs).使用BM的动物模型,我们观察到RT-MPs可以穿透血脑屏障并被TAMs吞噬。然后TAMs的微环境从M2表型转变为M1表型,从而调节TAM与肿瘤细胞之间的相互作用。单细胞测序分析表明,TAM,在内化RT-MP之后,激活趋化因子信号通路并分泌更多趋化因子,如CCL5,CXCL2,CXCL1,CCL3,CCL4和CCL22,吸引更多的CD4+T细胞和CD8+T细胞,提高免疫介导的杀伤,并增强随后的联合抗PD-1治疗。这些发现为探索免疫抗性NSCLC患者的替代治疗提供了临床前基础。
    Brain metastases (BMs) are the most common sites of metastasis in patients with non-small cell lung cancer (NSCLC). However, BMs are not responsive to immunotherapy because of the blood-brain barrier. This is because intracranial immune cells such as M2 tumor-associated macrophages (TAMs) accumulate, creating an immunosuppressive tumor microenvironment. In this study, we focused on irradiated tumor cell-released microparticles (RT-MPs) that can cross the blood-brain barrier and influence the intracranial immune microenvironment. Using animal models of BMs, we observed that RT-MPs could penetrate the blood-brain barrier and be swallowed by TAMs. Then the microenvironment of TAMs is shifted from the M2 phenotype to the M1 phenotype, thereby modulating the interactions between TAMs and tumor cells. Single-cell sequencing analysis demonstrated that TAMs, after internalizing RT-MPs, active chemokine signaling pathways and secrete more chemokines, such as CCL5, CXCL2, CXCL1, CCL3, CCL4, and CCL22, attracting more CD4+ T cells and CD8+ T cells, improving immune-mediated killing, and enhancing subsequent combination anti-PD-1 therapy. These findings provide a preclinical foundation for exploring alternative treatments for patients with immunoresistant NSCLC BMs.
    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
    肿瘤相关巨噬细胞(TAMs)是肿瘤微环境中最丰富的免疫成分之一。又分为抗肿瘤M1亚型和促肿瘤M2亚型。最近的研究发现,TAM在肿瘤发生的调节和发展中起着至关重要的作用。此外,TAMs促进肿瘤血管化,并支持肿瘤细胞的存活,对肿瘤生长和患者预后有影响。大量研究表明,降低TAM的密度,或调节TAM的极化,可以抑制肿瘤生长,表明TAMs是肿瘤免疫治疗的有希望的靶标。最近,临床试验发现,针对TAM的治疗取得了令人鼓舞的结果,美国食品和药物管理局已经批准了许多用于癌症治疗的药物。在这次审查中,我们总结了起源,极化,和TAM的功能,并在临床研究和科学研究中强调针对TAMs在癌症治疗中的治疗策略,显示了TAMs靶向治疗在肿瘤免疫治疗中的广阔前景。
    Tumor-associated macrophages (TAMs) are one of the most plentiful immune compositions in the tumor microenvironment, which are further divided into anti-tumor M1 subtype and pro-tumor M2 subtype. Recent findings found that TAMs play a vital function in the regulation and progression of tumorigenesis. Moreover, TAMs promote tumor vascularization, and support the survival of tumor cells, causing an impact on tumor growth and patient prognosis. Numerous studies show that reducing the density of TAMs, or modulating the polarization of TAMs, can inhibit tumor growth, indicating that TAMs are a promising target for tumor immunotherapy. Recently, clinical trials have found that treatments targeting TAMs have achieved encouraging results, and the U.S. Food and Drug Administration has approved a number of drugs for use in cancer treatment. In this review, we summarize the origin, polarization, and function of TAMs, and emphasize the therapeutic strategies targeting TAMs in cancer treatment in clinical studies and scientific research, which demonstrate a broad prospect of TAMs-targeted therapies in tumor 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
    免疫检查点阻断(ICB)治疗的主要挑战在于免疫抑制肿瘤微环境(TME)。将M2样肿瘤相关巨噬细胞(TAM)重新极化为炎性M1表型是一种有前途的癌症免疫治疗策略。这里,这项研究表明,肿瘤抑制蛋白SHISA3调节TAMs的抗肿瘤功能。编码Shisa3的mRNA的局部递送使癌症免疫治疗能够通过将TAMs重编程为抗肿瘤表型,从而增强程序性细胞死亡1(PD-1)抗体的功效。Shisa3在TAM中的强制表达增加了它们的吞噬作用和抗原呈递能力,并促进CD8+T细胞介导的抗肿瘤免疫。SHISA3的表达是由巨噬细胞中的损伤/病原体相关分子模式(DAMPs/PAMPs)通过核因子-κB(NF-κB)转录因子诱导的。相互,SHISA3与热休克蛋白家族A成员8(HSPA8)形成复合物,以激活NF-κB信号,从而维持巨噬细胞的M1极化。敲除Shisa3在很大程度上消除了Toll样受体4(TLR4)激动剂单磷酰脂质A(MPLA)和PD-1抗体的联合免疫疗法的抗肿瘤功效。进一步发现SHISA3在抗肿瘤TAMs中的较高表达与肺癌患者的较好总体生存率相关。一起来看,研究结果描述了SHISA3在改善癌症免疫治疗的TAM重编程中的作用.
    The main challenge for immune checkpoint blockade (ICB) therapy lies in immunosuppressive tumor microenvironment (TME). Repolarizing M2-like tumor-associated macrophages (TAMs) into inflammatory M1 phenotype is a promising strategy for cancer immunotherapy. Here, this study shows that the tumor suppressive protein SHISA3 regulates the antitumor functions of TAMs. Local delivery of mRNA encoding Shisa3 enables cancer immunotherapy by reprogramming TAMs toward an antitumoral phenotype, thus enhancing the efficacy of programmed cell death 1 (PD-1) antibody. Enforced expression of Shisa3 in TAMs increases their phagocytosis and antigen presentation abilities and promotes CD8+ T cell-mediated antitumor immunity. The expression of SHISA3 is induced by damage/pathogen-associated molecular patterns (DAMPs/PAMPs) in macrophages via nuclear factor-κB (NF-κB) transcription factors. Reciprocally, SHISA3 forms a complex with heat shock protein family A member 8 (HSPA8) to activate NF-κB signaling thus maintaining M1 polarization of macrophages. Knockout Shisa3 largely abolishes the antitumor efficacy of combination immunotherapy with Toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) and PD-1 antibody. It further found that higher expression of SHISA3 in antitumoral TAMs is associated with better overall survival in lung cancer patients. Taken together, the findings describe the role of SHISA3 in reprogramming TAMs that ameliorate 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
    将肿瘤相关巨噬细胞(TAM)重新极化为抑制肿瘤的M1巨噬细胞被认为是增强癌症免疫疗法的有希望的策略。然而,几种免疫抑制配体(例如,LSECtin)仍可在M1巨噬细胞上高表达,诱导不满意的治疗结果。我们在本文中开发了一种抗体修饰的纳米平台,该平台由PEG化的氧化铁纳米颗粒(IONP)和LSECtin抗体通过腙键缀合到IONP的表面上,用于增强的癌症免疫疗法。静脉给药后,肿瘤微环境(TME)pH可触发腙键断裂并诱导纳米平台解离成游离的LSECtin抗体和IONP。因此,IONP可以将TAM重新极化为M1巨噬细胞,以重塑免疫抑制性TME,并通过分泌杀肿瘤因子提供额外的抗癌作用(例如,interlukin-12)。同时,LSECtin抗体可进一步阻断M1巨噬细胞表达的LSECtin活性,减轻其对CD8+T细胞的免疫抑制作用,最终导致肿瘤生长的显著抑制。
    Repolarizing tumor-associated macrophages (TAMs) into tumor-inhibiting M1 macrophages has been considered a promising strategy for enhanced cancer immunotherapy. However, several immunosuppressive ligands (e.g., LSECtin) can still be highly expressed on M1 macrophages, inducing unsatisfactory therapeutic outcomes. We herein developed an antibody-decorated nanoplatform composed of PEGylated iron oxide nanoparticles (IONPs) and LSECtin antibody conjugated onto the surface of IONPs via the hydrazone bond for enhanced cancer immunotherapy. After intravenous administration, the tumor microenvironment (TME) pH could trigger the hydrazone bond breakage and induce the disassociation of the nanoplatform into free LSECtin antibodies and IONPs. Consequently, the IONPs could repolarize TAMs into M1 macrophages to remodel immunosuppressive TME and provide an additional anticancer effect via secreting tumoricidal factors (e.g., interlukin-12). Meanwhile, the LSECtin antibody could further block the activity of LSECtin expressed on M1 macrophages and relieve its immunosuppressive effect on CD8+ T cells, ultimately leading to significant inhibition of tumor growth.
    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:乳腺癌是全球女性癌症相关死亡的主要原因,对女性健康构成重大威胁。因此,寻找新的乳腺癌治疗靶点和预后生物标志物至关重要.方法:生物信息学分析,实时定量PCR(qRT-PCR),采用荧光原位杂交(FISH)技术研究hsa_circ_002144在乳腺癌中的表达。Transwell分析,西方印迹,和细胞活力测定用于评估hsa_circ_002144对增殖的影响,迁移,和乳腺癌细胞的侵袭。此外,建立小鼠模型以验证其功能.流式细胞术,WB分析,酶联免疫吸附测定(ELISA),qRT-PCR,外泌体分离,和共培养系统用于阐明巨噬细胞极化的分子机制。结果:我们首次发现hsa_circ_002144在乳腺癌中高表达。它影响肿瘤的生长和转移,并可能通过糖酵解途径影响巨噬细胞的极化。结论:这一发现为乳腺癌的治疗和预后评估提供了新的方向。
    Background: Breast cancer is a leading cause of cancer-related deaths in women worldwide, posing a significant threat to female health. Therefore, it is crucial to search for new therapeutic targets and prognostic biomarkers for breast cancer patients. Method: Bioinformatics analysis, quantitative real-time PCR (qRT-PCR), and fluorescence in situ hybridization (FISH) were employed to investigate the expression of hsa_circ_002144 in breast cancer. Transwell assay, Western blotting, and cell viability assay were utilized to assess the impact of hsa_circ_002144 on the proliferation, migration, and invasion of breast cancer cells. Additionally, a mouse model was established to validate its functionality. Flow cytometry, WB analysis, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, exosomes isolation, and co-culture system were employed to elucidate the molecular mechanism underlying macrophage polarization. Result: we have discovered for the first time that hsa_circ_002144 is highly expressed in breast cancer. It affected tumor growth and metastasis and could influence macrophage polarization through the glycolytic pathway. Conclusion: This finding provides a new direction for breast cancer treatment and prognosis assessment.
    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
    外周炎症细胞的存在与癌症的预后有关。本研究旨在探讨中性粒细胞绝对计数(ANC)和单核细胞绝对计数(AMC)在肾细胞癌(RCC)与肾血管平滑肌脂肪瘤(RAML)鉴别诊断中的作用。以及它们在RCC中的预后意义。
    我们对外周免疫细胞数据进行了全面分析,临床病理资料,2015年1月至2021年12月诊断为RCC或RAML的患者的肿瘤特征。接收器工作特性(ROC)曲线,以及单变量和多变量分析,用于评估AMC和ANC在区分RCC和RAML中的诊断效用。采用Kaplan-Meier曲线分析研究不同AMC和ANC的RCC患者的生存情况。使用COX单因素和多因素分析研究AMC和ANC在RCC中的预后价值。使用癌症基因组图谱(TCGA)和基因表达综合(GEO)数据库进行生物信息学相关性分析。
    总共1120名符合条件的患者被纳入研究。与RAML患者相比,RCC患者的术前平均AMC和ANC明显更高(分别为P=0.001和P<0.001)。术前AMC和ANC高与吸烟史显著相关,肿瘤长度,肉眼血尿,高T级,N级,和病理分级。在多变量分析中,ANC>3.205*10^9/L与RCC的存在独立相关(HR=1.618,P=0.008)。高AMC和ANC与OS和PFS降低显著相关(P<0.05)。ANC可能是一个独立的预后因素。公共数据库分析表明,肿瘤相关巨噬细胞(TAMs)和肿瘤相关中性粒细胞(TANs)的特征基因在ccRCC中高表达。
    术前升高的ANC和AMC可以区分RCC和RAML,并预测RCC患者的不良预后。此外,TAM和TAN的特征基因在透明细胞RCC中表现出高表达水平。
    UNASSIGNED: The presence of peripheral inflammatory cells has been linked to the prognosis of cancer. This study aims to investigate the distinct roles of absolute neutrophil count (ANC) and absolute monocyte count (AMC) in differentiating renal cell carcinoma (RCC) from renal angiomyolipoma (RAML), as well as their prognostic significance in RCC.
    UNASSIGNED: We conducted a comprehensive analysis of peripheral immune cell data, clinicopathological data, and tumor characteristics in patients diagnosed with RCC or RAML from January 2015 to December 2021. Receiver operating characteristic (ROC) curves, as well as univariate and multivariate analyses, were employed to assess the diagnostic utility of AMC and ANC in differentiating between RCC and RAML. Kaplan-Meier curve analysis was used to study the survival of RCC patients with different AMC and ANC. The prognostic value of AMC and ANC in RCC was investigated using COX univariate and multivariate analysis. The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were used for bioinformatic correlation analysis.
    UNASSIGNED: A total of 1120 eligible patients were included in the study. The mean preoperative AMC and ANC in patients with RCC were found to be significantly higher compared to those in patients with RAML (P = 0.001 and P < 0.001, respectively). High preoperative AMC and ANC significantly correlated with smoking history, tumor length, gross hematuria, and high T Stage, N stage, and pathological grade. In multivariate analyses, an ANC> 3.205 *10^9/L was identified to be independently associated with the presence of RCC (HR = 1.618, P = 0.008). High AMC and ANC were significantly associated with reduced OS and PFS (P < 0.05), and ANC may be an independent prognostic factor. Public database analysis showed that signature genes of tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) were highly expressed in ccRCC.
    UNASSIGNED: Elevated preoperative ANC and AMC can distinguish RCC from RAML and predict poor prognosis in patients with RCC. Furthermore, the signature genes of TAMs and TANs exhibit high expression levels in clear cell RCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号