Human cancers

人类癌症
  • 文章类型: Journal Article
    在过去的十年里,癌症代谢的重新编程越来越受到关注,特别是葡萄糖代谢的重新编程,也被称为“Warburg效应”。葡萄糖代谢的重编程被认为是人类癌症的新标志。越来越多的研究表明,糖代谢的重编程可以调节癌症的许多生物学过程,包括致癌作用,programming,转移,和抗药性。在这次审查中,我们总结了主要的生物学功能,临床意义,人类癌症中葡萄糖代谢重编程的潜在靶标和信号通路。此外,分析了针对葡萄糖代谢重编程的天然产物和小分子抑制剂的应用,总结了一些针对葡萄糖代谢重编程和试验状态的临床药物,以及靶向葡萄糖代谢重编程用于癌症治疗的利弊分析。总的来说,糖代谢的重编程在预测中起着重要作用,预防,人类癌症的诊断和治疗。葡萄糖代谢重编程相关靶标具有作为改善癌症患者个体预后和预后的生物标志物的巨大潜力。靶向糖代谢重编程相关的临床创新将是未来癌症治疗研究的热点。我们认为,更多高质量的临床试验和更丰富的药物配方和毒理学实验将有利于靶向糖代谢重编程药物的开发和临床应用。这篇综述将为研究人员提供更广阔的视野和全面的了解葡萄糖代谢重编程在人类癌症中的重要意义。
    Reprogramming of cancer metabolism has become increasingly concerned over the last decade, particularly the reprogramming of glucose metabolism, also known as the \"Warburg effect\". The reprogramming of glucose metabolism is considered a novel hallmark of human cancers. A growing number of studies have shown that reprogramming of glucose metabolism can regulate many biological processes of cancers, including carcinogenesis, progression, metastasis, and drug resistance. In this review, we summarize the major biological functions, clinical significance, potential targets and signaling pathways of glucose metabolic reprogramming in human cancers. Moreover, the applications of natural products and small molecule inhibitors targeting glucose metabolic reprogramming are analyzed, some clinical agents targeting glucose metabolic reprogramming and trial statuses are summarized, as well as the pros and cons of targeting glucose metabolic reprogramming for cancer therapy are analyzed. Overall, the reprogramming of glucose metabolism plays an important role in the prediction, prevention, diagnosis and treatment of human cancers. Glucose metabolic reprogramming-related targets have great potential to serve as biomarkers for improving individual outcomes and prognosis in cancer patients. The clinical innovations related to targeting the reprogramming of glucose metabolism will be a hotspot for cancer therapy research in the future. We suggest that more high-quality clinical trials with more abundant drug formulations and toxicology experiments would be beneficial for the development and clinical application of drugs targeting reprogramming of glucose metabolism.This review will provide the researchers with the broader perspective and comprehensive understanding about the important significance of glucose metabolic reprogramming in human cancers.
    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:人内源性逆转录病毒(HERV),在灵长类动物进化过程中整合到人类基因组中,约占人类基因组的8%。尽管大多数HERV由于突变而非蛋白质编码,插入,删除,和截断,最近的研究揭示了它们在生物过程中的不同作用,包括疾病的发病机制。
    目标:尽管许多HERV仍然不活跃,他们与各种疾病有牵连,特别是癌症,促使人们对利用HERV进行治疗的兴趣增加。这篇综述探讨了我们对HERV生物学作用的理解的最新进展,强调它们在癌症治疗中的临床意义。
    方法:这里,我们讨论了如何检测转座因子(TE),包括HERV,通过免疫系统触发人类癌症的先天免疫反应。
    结论:此外,我们概述了在阐明HERV激活对癌症的影响方面的最新进展,以及靶向HERV如何通过调节表观遗传可塑性和破坏癌症的发生和进展而有望用于抗癌治疗.
    BACKGROUND: Human endogenous retroviruses (HERVs), integrated into the human genome during primate evolution, constitute approximately 8% of the human genome. Although most HERVs are non-protein-coding owing to mutations, insertions, deletions, and truncations, recent research has revealed their diverse roles in biological processes, including disease pathogenesis.
    OBJECTIVE: Although many HERVs remain inactive, they have been implicated in various diseases, particularly cancer, prompting an increased interest in harnessing HERVs for therapeutic purposes. This review explores the recent advancements in our understanding of the biological roles of HERVs, emphasizing their clinical relevance in cancer treatment.
    METHODS: Here, we discuss how the detection of transposable elements (TEs), including HERVs, by the immune system triggers innate immune responses in human cancers.
    CONCLUSIONS: Additionally, we outline recent progress in elucidating the implications of HERV activation in cancer and how targeting HERVs holds promise for anti-cancer treatments by modulating epigenetic plasticity and disrupting cancer initiation and progression.
    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
    癌症是一种由异常细胞生长驱动的多层面疾病,对全球健康构成重大威胁。多因素原因,个体对治疗药物敏感性的差异,和诱导的耐药性在有效解决癌症方面构成了重大挑战。使癌症在其生理上高度异质性的最重要方面之一在于所涉及的基因以及在恶性状况下这些基因中的一些发生的变化。遗传因素与肿瘤发生有关,programming,对治疗的反应,和转移。在人类癌症中起关键作用的一种这样的基因是共济失调-毛细血管扩张基因(ATM)的突变形式。ATM基因位于染色体11q23上,在维持基因组稳定性方面起着至关重要的作用。了解A-T的遗传基础对诊断至关重要,管理,和治疗。乳腺癌,肺癌,前列腺癌,和胃癌表现出与ATM基因的不同关系并影响其通路。针对ATM途径证明有希望提高治疗效果,特别是与DNA损伤反应途径有关。分析ATM突变的治疗后果,特别是在这些癌症类型中,促进了早期检测的方法,干预,个性化治疗方法的发展,改善患者预后。这篇综述强调了ATM基因在各种癌症中的作用。强调其对DNA修复途径和治疗反应的影响。
    Cancer is a multifaceted disease driven by abnormal cell growth and poses a significant global health threat. The multifactorial causes, differences in individual susceptibility to therapeutic drugs, and induced drug resistance pose major challenges in addressing cancers effectively. One of the most important aspects in making cancers highly heterogeneous in their physiology lies in the genes involved and the changes occurring to some of these genes in malignant conditions. The Genetic factors have been implicated in the oncogenesis, progression, responses to treatment, and metastasis. One such gene that plays a key role in human cancers is the mutated form of the Ataxia-telangiectasia gene (ATM). ATM gene located on chromosome 11q23, plays a vital role in maintaining genomic stability. Understanding the genetic basis of A-T is crucial for diagnosis, management, and treatment. Breast cancer, lung cancer, prostate cancer, and gastric cancer exhibit varying relationships with the ATM gene and influence their pathways. Targeting the ATM pathway proves promising for enhancing treatment effectiveness, especially in conjunction with DNA damage response pathways. Analyzing the therapeutic consequences of ATM mutations, especially in these cancer types facilitates the approaches for early detection, intervention, development of personalized treatment approaches, and improved patient outcomes. This review emphasizes the role of the ATM gene in various cancers, highlighting its impact on DNA repair pathways and therapeutic responses.
    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
    MicroRNA-98(miR-98)是肿瘤学复杂环境中的重要分子。作为microRNA的一个子集,这些小的非编码RNA伴随着癌症研究的新时代,支持它们在肿瘤发生中的重要作用,转移,和治疗干预措施。这篇综述提供了对生物发生的全面见解,分子性质,和miR-98的生理事业,突出了其在癌症进展中的双重作用-作为肿瘤的启动子和抑制剂。有趣的是,miR-98对癌症进展的各个方面都有深远的影响。调节关键的细胞功能,包括扩散,凋亡,和细胞周期。鉴于它的表达模式,miR-98作为诊断和预后生物标志物的潜力,尤其是在液体活检和肿瘤组织中,正在探索,强调在临床上翻译这些发现的障碍。该综述通过评估调节miR-98表达的治疗途径来总结,解决治疗抵抗方面的挑战,并评估miR-98干预措施的疗效。总之,虽然miR-98参与癌症展示了有希望的诊断和治疗途径,未来的研究应该转向理解它在肿瘤-基质相互作用中的作用,免疫调节,和代谢调节,从而开启癌症管理的新策略。
    MicroRNA-98 (miR-98) stands as an important molecule in the intricate landscape of oncology. As a subset of microRNAs, these small non-coding RNAs have accompanied a new era in cancer research, underpinning their significant roles in tumorigenesis, metastasis, and therapeutic interventions. This review provides a comprehensive insight into the biogenesis, molecular properties, and physiological undertakings of miR-98, highlighting its double-edged role in cancer progression-acting both as a tumor promoter and suppressor. Intriguingly, miR-98 has profound implications for various aspects of cancer progression, modulating key cellular functions, including proliferation, apoptosis, and the cell cycle. Given its expression patterns, the potential of miR-98 as a diagnostic and prognostic biomarker, especially in liquid biopsies and tumor tissues, is explored, emphasizing the hurdles in translating these findings clinically. The review concludes by evaluating therapeutic avenues to modulate miR-98 expression, addressing the challenges in therapy resistance, and assessing the efficacy of miR-98 interventions. In conclusion, while miR-98\'s involvement in cancer showcases promising diagnostic and therapeutic avenues, future research should pivot towards understanding its role in tumor-stroma interactions, immune modulation, and metabolic regulation, thereby unlocking novel strategies for cancer management.
    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:c-MET是一种受体酪氨酸激酶,通常被HGF激活以激活其下游信号级联,例如MAPK,PI3K/Akt/mTOR,和STAT3。c-MET调节细胞增殖,上皮-间质转化(EMT),免疫反应,形态发生,凋亡,和血管生成。c-MET已被证明在胚胎发生和早期发育中起着重要作用。c-MET途径在广泛的恶性肿瘤中失调,由于配体或受体的过度表达,基因组扩增,和MET突变。c-MET信号的失调与肿瘤进展之间的联系已得到充分证明。c-MET的过表达或过激活与不良的临床结果和对靶向治疗的获得性抗性相关。由于c-MET激活导致致癌途径的触发,废除c-MET途径被认为是癌症治疗中的关键策略。在这里,已经对c-MET通路在人类癌症中的作用及其在骨转移和治疗抗性中的相关性进行了分析.此外,已经尝试总结所选天然化合物对癌症中c-MET信号传导的抑制活性。
    方法:与恶性肿瘤中c-MET途径及其天然化合物调节剂相关的出版物来自PubMed等数据库,Scopus,和谷歌学者,并根据PRISMA指南进行总结。一些用于提取相关文献的关键词是c-MET,c-MET的天然复合抑制剂,肝癌中的c-MET,乳腺癌的c-MET,肺癌的c-MET,胰腺癌中的c-MET,头颈癌的c-MET,c-MET在骨转移中,c-MET在治疗抗性中,以及c-MET抑制剂和化学治疗剂的组合。在PubChem数据库中验证了自然化合物的化学结构。
    结果:搜索产生了3935种出版物,其中195份参考出版物用于我们的分析。使用ClinicalTrials.gov标识符引用临床试验。c-MET途径已被认为是对抗生长的突出目标,转移,和癌症中的化疗抗性。已经阐述了c-MET在骨转移以及治疗抗性中的关键作用。此外,已讨论了某些天然化合物在临床/临床前研究中对c-MET途径的抑制作用。
    BACKGROUND: c-MET is a receptor tyrosine kinase which is classically activated by HGF to activate its downstream signaling cascades such as MAPK, PI3K/Akt/mTOR, and STAT3. The c-MET modulates cell proliferation, epithelial-mesenchymal transition (EMT), immune response, morphogenesis, apoptosis, and angiogenesis. The c-MET has been shown to serve a prominent role in embryogenesis and early development. The c-MET pathway is deregulated in a broad range of malignancies, due to overexpression of ligands or receptors, genomic amplification, and MET mutations. The link between the deregulation of c-MET signaling and tumor progression has been well-documented. Overexpression or overactivation of c-MET is associated with dismal clinical outcomes and acquired resistance to targeted therapies. Since c-MET activation results in the triggering of oncogenic pathways, abrogating the c-MET pathway is considered to be a pivotal strategy in cancer therapeutics. Herein, an analysis of role of the c-MET pathway in human cancers and its relevance in bone metastasis and therapeutic resistance has been undertaken. Also, an attempt has been made to summarize the inhibitory activity of selected natural compounds towards c-MET signaling in cancers.
    METHODS: The publications related to c-MET pathway in malignancies and its natural compound modulators were obtained from databases such as PubMed, Scopus, and Google Scholar and summarized based on PRISMA guidelines. Some of the keywords used for extracting relevant literature are c-MET, natural compound inhibitors of c-MET, c-MET in liver cancer, c-MET in breast cancer, c-MET in lung cancer, c-MET in pancreatic cancer, c-MET in head and neck cancer, c-MET in bone metastasis, c-MET in therapeutic resistance, and combination of c-MET inhibitors and chemotherapeutic agents. The chemical structure of natural compounds was verified in PubChem database.
    RESULTS: The search yielded 3935 publications, of which 195 reference publications were used for our analysis. Clinical trials were referenced using ClinicalTrials.gov identifier. The c-MET pathway has been recognized as a prominent target to combat the growth, metastasis, and chemotherapeutic resistance in cancers. The key role of the c-MET in bone metastasis as well as therapeutic resistance has been elaborated. Also, suppressive effect of selected natural compounds on the c-MET pathway in clinical/preclinical studies has been discussed.
    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:细胞周期蛋白依赖性激酶16(CDK16)在各种疾病中起着重要的生物学作用。尽管如此,其在不同癌症类型中的功能及其与肿瘤免疫微环境(TIME)的关系仍未得到很好的理解。
    方法:我们分析了表达谱,遗传改变,临床特征,使用来自癌症基因组图谱的数据,以及CDK16在泛癌症中的预后价值,基因型-组织表达数据库,和体外实验。此外,我们利用TIMER2和ImmuCellAI数据库评估CDK16表达与免疫细胞浸润水平之间的相关性.最后,我们使用收集的免疫疗法数据检查了CDK16与免疫疗法应答之间的相关性.
    结果:CDK16在泛癌症中明显过表达,是各种癌症预后不良的危险因素。我们的发现表明,CDK16不仅调节细胞周期相关功能以促进细胞增殖,而且还调节先天和适应性免疫系统的自身免疫相关功能。以及其他免疫相关的信号通路。此外,CDK16过表达有助于免疫抑制肿瘤微环境,广泛抑制免疫相关特征,如免疫相关基因和途径的表达,以及免疫浸润细胞的计数。我们的分析表明,与具有高CDK16表达的个体相比,具有低CDK16表达的个体对免疫检查点抑制剂的反应率更高,总体生存期更长。
    结论:本研究确立了CDK16作为预测多种癌症预后不良的潜在生物标志物。它在塑造免疫抑制肿瘤微环境和影响免疫疗法疗效方面的作用突出了开发针对CDK16的靶向疗法的迫切需要,为癌症治疗和管理提供了新的途径。
    Cyclin-Dependent Kinase 16 (CDK16) plays significant biological roles in various diseases. Nonetheless, its function in different cancer types and its relationship with the Tumor Immune Microenvironment (TIME) are still not well-understood.
    We analyzed the expression profile, genetic alterations, clinical features, and prognostic value of CDK16 in pan-cancer using data from The Cancer Genome Atlas, Genotype-Tissue Expression databases, and in vitro experiments. Additionally, the TIMER2 and ImmuCellAI databases were utilized to assess the correlation between CDK16 expression and immune cell infiltration levels. Finally, we examined the correlation between CDK16 and the response to immunotherapy using collected immunotherapy data.
    CDK16 is notably overexpressed in pan-cancer and is a risk factor for poor prognosis in various cancers. Our findings reveal that CDK16 regulates not only cell cycle-related functions to promote cell proliferation but also the autoimmunity-related functions of the innate and adaptive immune systems, along with other immune-related signaling pathways. Moreover, CDK16 overexpression contributes to an immunosuppressive tumor microenvironment, extensively suppressing immune-related features such as the expression of immune-related genes and pathways, as well as the count of immune-infiltrating cells. Our analysis indicated that individuals with low CDK16 expression showed higher response rates to immune checkpoint inhibitors and longer overall survival compared to those with high CDK16 expression.
    This study establishes CDK16 as a potential biomarker for predicting poor prognosis in a wide range of cancers. Its role in shaping the immunosuppressive tumor microenvironment and influencing the efficacy of immunotherapy highlights the urgent need for developing targeted therapies against CDK16, offering new avenues for cancer treatment and management.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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:Prostein(P501S),也称为溶质载体家族45成员3(SLC45A3)是在前列腺上皮细胞中优先表达的雄激素调节蛋白。由于其在前列腺癌中的频繁表达,prostein被认为是诊断前列腺癌的标志物.
    方法:为了全面评估Prostein免疫组织化学的诊断效用,通过免疫组织化学分析了一个组织微阵列,该芯片包含来自152种不同肿瘤类型和亚型的19,202个样本以及来自76种不同正常组织类型的608个样本.
    结果:Prostein免疫染色通常是细胞质,颗粒状和核周。在419例前列腺癌中,有96.7%的前列腺癌呈阳性,其中78.3%具有强染色。在16,709个前列腺外肿瘤中,在所有病例中观察到7.2%的患者为prostein阳性,但只有0.3%的患者具有强染色.总的来说,50个不同的前列腺外肿瘤类别为prostein阳性,其中12例包括至少一例强烈阳性病例。前列腺癌阳性率最高的前列腺外肿瘤包括不同亚型的唾液腺肿瘤(7.6-44.4%),神经内分泌肿瘤(15.8-44.4%),胃肠道腺癌(7.3-14.8%),胆胰腺腺癌(3.6-38.7%),肝细胞癌(8.1%),和其他器官的腺癌(高达21%)。
    结论:我们的数据提供了关于人类癌症中Prostein表达的全面概述。Prostein是在>96%的前列腺癌中发生的高度敏感的前列腺癌标志物。因为prostein也可以在各种其他肿瘤实体中表达,将肿瘤块分类为前列腺癌不应仅基于prostein阳性。
    BACKGROUND: Prostein (P501S), also termed solute carrier family 45 member 3 (SLC45A3) is an androgen regulated protein which is preferentially expressed in prostate epithelial cells. Because of its frequent expression in prostate cancer, prostein was suggested a diagnostic prostate cancer marker.
    METHODS: In order to comprehensively assess the diagnostic utility of prostein immunohistochemistry, a tissue microarray containing 19,202 samples from 152 different tumor types and subtypes as well as 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry.
    RESULTS: Prostein immunostaining was typically cytoplasmic, granular and perinuclear. Prostein positivity was seen in 96.7% of 419 prostate cancers including 78.3% with strong staining. In 16,709 extra-prostatic tumors, prostein positivity was observed in 7.2% of all cases but only 0.3% had a strong staining. Overall, 50 different extra-prostatic tumor categories were prostein positive, 12 of which included at least one strongly positive case. Extra-prostatic tumors with highest rates of prostein positivity included different subtypes of salivary gland tumors (7.6-44.4%), neuroendocrine neoplasms (15.8-44.4%), adenocarcinomas of the gastrointestinal tract (7.3-14.8%), biliopancreatic adenocarcinomas (3.6-38.7%), hepatocellular carcinomas (8.1%), and adenocarcinomas of other organs (up to 21%).
    CONCLUSIONS: Our data provide a comprehensive overview on prostein expression in human cancers. Prostein is a highly sensitive prostate cancer marker occurring in > 96% of prostate cancers. Because prostein can also be expressed in various other tumor entities, classifying of a tumor mass as a prostate cancer should not be based on prostein positivity alone.
    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
    Toll样受体(TLRs)作为人体的第一道防线,识别从受损或死亡细胞释放的病原体表达的分子和宿主来源的分子。不同细胞类型的广泛分布,从上皮细胞到免疫细胞,强调了TLRs在连接先天免疫和适应性免疫中的关键作用。在刺激时,TLRs结合介导几种衔接蛋白和下游激酶的表达,这导致了几种其他信号分子的诱导,如关键的促炎介质。的确,免疫生物学研究的非凡进展表明,TLRs可能是炎症相关疾病治疗干预的有希望的靶标,自身免疫性疾病,微生物感染以及人类癌症。到目前为止,用于预防和可能治疗炎症性疾病,各种TLR拮抗剂/抑制剂已显示在从临床前评估到临床试验的几个阶段是有效的。因此,TLRs在调节先天和适应性水平的人类免疫反应中的迷人作用指导科学家选择这些免疫传感器蛋白作为开发针对癌症的化学疗法和免疫疗法的合适靶标。到目前为止,几种TLR靶向小分子(例如,Pam3CSK4,聚(I:C),Poly(A:U)),化合物,植物化合物(例如,姜黄素),肽,和抗体已被发现对几种类型的癌症提供保护。然而,据报道,施用不适当剂量的此类TLR调节治疗剂或错误输注施用会引起有害结果。这篇综述总结了TLRs分子和结构生物学的最新发现,并通过讨论已建立和管道发现的发现,概述了TLR针对癌症的治疗策略的效力和前景。
    Toll-like receptors (TLRs) serve as the body\'s first line of defense, recognizing both pathogen-expressed molecules and host-derived molecules released from damaged or dying cells. The wide distribution of different cell types, ranging from epithelial to immune cells, highlights the crucial roles of TLRs in linking innate and adaptive immunity. Upon stimulation, TLRs binding mediates the expression of several adapter proteins and downstream kinases, that lead to the induction of several other signaling molecules such as key pro-inflammatory mediators. Indeed, extraordinary progress in immunobiological research has suggested that TLRs could represent promising targets for the therapeutic intervention of inflammation-associated diseases, autoimmune diseases, microbial infections as well as human cancers. So far, for the prevention and possible treatment of inflammatory diseases, various TLR antagonists/inhibitors have shown to be efficacious at several stages from pre-clinical evaluation to clinical trials. Therefore, the fascinating role of TLRs in modulating the human immune responses at innate as well as adaptive levels directed the scientists to opt for these immune sensor proteins as suitable targets for developing chemotherapeutics and immunotherapeutics against cancer. Hitherto, several TLR-targeting small molecules (e.g., Pam3CSK4, Poly (I:C), Poly (A:U)), chemical compounds, phytocompounds (e.g., Curcumin), peptides, and antibodies have been found to confer protection against several types of cancers. However, administration of inappropriate doses of such TLR-modulating therapeutics or a wrong infusion administration is reported to induce detrimental outcomes. This review summarizes the current findings on the molecular and structural biology of TLRs and gives an overview of the potency and promises of TLR-directed therapeutic strategies against cancers by discussing the findings from established and pipeline discoveries.
    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
    人类生长激素1(HGH1),蛋白质编码基因,在人类的成长和发展中起着至关重要的作用。然而,其在人类癌症中的作用尚不清楚。第一次,本研究全面评估了HGH1对癌症预后和免疫功能的潜在影响。为了实现这一点,来自各种数据库的数据,包括癌症基因组图谱,基因型组织表达,癌细胞谱系百科全书,人类蛋白质图谱,cBioPortal,肿瘤免疫评估资源和免疫细胞丰度标识符,被整理,以及一项大型临床研究,三个免疫治疗队列和体外实验。本研究旨在阐明HGH1表达在癌症预后和免疫应答中的作用。我们的发现揭示了HGH1表达增加与各种癌症类型预后恶化之间的显著关联。主要是,拷贝数变异被确定为最常见的基因突变.此外,观察到HGH1不仅调节细胞周期相关功能以促进细胞增殖,而且还影响先天和适应性免疫系统内的自身免疫相关功能。以及其他相关的免疫相关信号通路。使用基因集富集分析和基因集变异分析来证实这些发现。HGH1过表达导致免疫缺陷(免疫沙漠)肿瘤微环境,其特征是免疫相关特征的显着表达,例如免疫相关基因和途径表达以及免疫浸润细胞的数量。此外,观察到HGH1表达与四种癌症中肿瘤突变负荷和八种癌症中微卫星不稳定性之间的相关性。这表明HGH1具有作为免疫治疗靶标的潜力。免疫治疗数据分析支持这个概念,证明与HGH1高表达患者相比,接受免疫检查点抑制剂治疗的HGH1低表达患者的生存率提高,免疫疗法应答的可能性更高.总的来说,这些发现强调了HGH1在人类癌症中的重要作用,阐明其参与肿瘤发生和癌症免疫。鉴定升高的HGH1表达指示免疫沙漠肿瘤微环境。因此,HGH1的靶向,特别是与免疫检查点抑制剂治疗相结合,有望提高癌症患者的治疗效果。
    HGH1 homolog, a protein-coding gene, plays a crucial role in human growth and development. However, its role in human cancer remains unclear. For the first time, this study comprehensively evaluated the potential involvement of HGH1 in cancer prognosis and immunological function. To achieve this, data from various databases, including The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, cBioPortal, Tumor Immune Estimation Resource and Immune Cell Abundance Identifier, were collated, as well as from one large clinical study, three immunotherapy cohorts and in vitro experiments. This study aims to elucidate the role of HGH1 expression in cancer prognosis and immune response. Our findings revealed a significant association between increased HGH1 expression and a worse prognosis across various cancer types. Predominantly, copy number variations were identified as the most common genetic mutations. Additionally, HGH1 was observed to not only regulate cell cycle-related functions to promote cell proliferation but also influence autoimmunity-related functions within both the innate and adaptive immune systems, along with other relevant immune-related signaling pathways. Gene set enrichment analysis and gene set variation analysis were used to substantiate these findings. HGH1 overexpression contributed to an immune-deficient (immune-desert) tumor microenvironment, which was characterized by a significant expression of immune-related features such as immune-related gene and pathway expression and the number of immune-infiltrating cells. Furthermore, the correlation between HGH1 expression and tumor mutational burden in four cancers and microsatellite instability in eight cancers was observed. This suggests that HGH1 has potential as an immunotherapeutic target. Immunotherapy data analysis supports this notion, demonstrating that patients with low HGH1 expression treated with immune checkpoint inhibitors exhibit improved survival rates and a higher likelihood of responding to immunotherapy than patients with high HGH1 expression. Collectively, these findings highlight the significant role of HGH1 in human cancers, illuminating its involvement in tumorigenesis and cancer immunity. Elevated HGH1 expression was identified to be indicative of an immune-desert tumor microenvironment. Consequently, the targeting of HGH1, particularly in combination with immune checkpoint inhibitor therapy, holds promise for enhancing therapeutic outcomes in patients with cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号