anticancer immunity

抗癌免疫
  • 文章类型: Journal Article
    DNA错配修复(MMRd)的缺陷在癌症基因组中留下了微卫星不稳定性(MSI)的特征性足迹。我们使用来自癌症基因组图谱和国际癌症基因组联盟的数据对MSI相关癌症进行了全面分析,专注于indel突变签名。我们根据其indel谱将MSI高基因组分为两个亚型:缺失显性(MMRd-del)和插入显性(MMRd-ins)。与MMRd-del基因组相比,MMRd-ins基因组表现出不同的突变和转录组特征,包括T>C取代和相关突变特征的患病率较高。MMRd-ins和MMRd-del基因组中的短插入和缺失靶向不同的基因集,导致两个亚型之间不同的indel配置文件。此外,MMRd-ins基因组中的indel富含亚克隆改变,这些亚克隆改变提供了有关MMRd-ins和MMRd-del基因组之间独特进化关系的线索。值得注意的是,转录组分析表明MMRd-ins癌症上调免疫相关基因,显示高水平的免疫细胞浸润,并显示出升高的新抗原负荷。两种类型的MMRd基因组之间的基因组和转录组差异突出了遗传机制的异质性,以及导致癌症的基因组足迹和转录组变化。具有潜在的临床意义。
    Deficiencies in DNA mismatch repair (MMRd) leave characteristic footprints of microsatellite instability (MSI) in cancer genomes. We used data from the Cancer Genome Atlas and International Cancer Genome Consortium to conduct a comprehensive analysis of MSI-associated cancers, focusing on indel mutational signatures. We classified MSI-high genomes into two subtypes based on their indel profiles: deletion-dominant (MMRd-del) and insertion-dominant (MMRd-ins). Compared with MMRd-del genomes, MMRd-ins genomes exhibit distinct mutational and transcriptomic features, including a higher prevalence of T>C substitutions and related mutation signatures. Short insertions and deletions in MMRd-ins and MMRd-del genomes target different sets of genes, resulting in distinct indel profiles between the two subtypes. In addition, indels in the MMRd-ins genomes are enriched with subclonal alterations that provide clues about a distinct evolutionary relationship between the MMRd-ins and MMRd-del genomes. Notably, the transcriptome analysis indicated that MMRd-ins cancers upregulate immune-related genes, show a high level of immune cell infiltration, and display an elevated neoantigen burden. The genomic and transcriptomic distinctions between the two types of MMRd genomes highlight the heterogeneity of genetic mechanisms and resulting genomic footprints and transcriptomic changes in cancers, which has potential clinical implications.
    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
    受体相互作用丝氨酸/苏氨酸蛋白激酶1(RIPK1)作为一个关键的应激前哨,协调细胞存活,炎症,和免疫原性细胞死亡(ICD)。尽管RIPK1的催化功能是触发细胞死亡所必需的,其非催化支架功能介导强大的促生存信号。因此,癌细胞可以劫持RIPK1来阻止坏死和逃避免疫检测。我们产生了选择性降解人和鼠RIPK1的小分子蛋白水解靶向嵌合体(PROTAC)。PROTAC介导的RIPK1耗尽下调TNFR1和TLR3/4信号中枢,增强NF-κB的输出,MAPK,和IFN信号。此外,RIPK1降解同时促进RIPK3活化和凋亡诱导。我们进一步证明,RIPK1降解通过使癌细胞对治疗诱导的TNF和干扰素敏感来增强放射疗法和免疫疗法的免疫刺激作用。这促进了ICD,抗肿瘤免疫,和持久的治疗反应。因此,通过PROTACs靶向RIPK1是克服放射疗法或免疫疗法耐药性并增强抗癌疗法的一种有前景的方法。
    Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.
    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
    YTHDF蛋白是哺乳动物RNAN6-甲基腺苷(m6A)甲基化的主要胞质读码蛋白。它们通过控制mRNA翻译和降解在细胞胞质溶胶中主要负责m6A介导的调节。最近对YTHDF蛋白的功能和机制研究表明,这些蛋白具有不同的功能,可以实现对表位转录组的通用调节。它们的不同功能很大程度上源于它们在低复杂度N末端的不同氨基酸序列。因此,它们具有不同的相分离倾向并且具有不同的翻译后修饰(PTM)。不同的PTM,亚细胞定位,和伴侣蛋白之间的竞争已成为控制这些YTHDF蛋白功能的三种主要机制。我们还总结了这些YTHDF蛋白在抗癌免疫中的关键作用的最新进展,以及靶向这些蛋白开发新的抗癌疗法的潜力。
    YTHDF proteins are main cytoplasmic \'reader\' proteins of RNA N6-methyladenosine (m6A) methylation in mammals. They are largely responsible for m6A-mediated regulation in the cell cytosol by controlling both mRNA translation and degradation. Recent functional and mechanistic investigations of the YTHDF proteins revealed that these proteins have different functions to enable versatile regulation of the epitranscriptome. Their divergent functions largely originate from their different amino acid sequences in the low-complexity N termini. Consequently, they have different phase separation propensities and possess distinct post-translational modifications (PTMs). Different PTMs, subcellular localizations, and competition among partner proteins have emerged as three major mechanisms that control the functions of these YTHDF proteins. We also summarize recent progress on critical roles of these YTHDF proteins in anticancer immunity and the potential for targeting these proteins for developing new anticancer therapies.
    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
    尽管癌症治疗取得了进展,肝细胞癌(HCC),肝癌最常见的形式,仍然是世界范围内的主要公共卫生问题。免疫微环境在调节肿瘤进展和治疗抵抗中起着至关重要的作用。在HCC中,肿瘤微环境(TME)的特征是大量的免疫抑制细胞和促进免疫逃避和转移的信号。最近,抗癌免疫疗法,旨在调节免疫系统以识别和消除癌症的治疗干预措施,已成为癌症治疗的重要基石。免疫疗法已经证明了在某些HCC患者组中提高生存率和提供持久的癌症控制的能力。同时减少不良副作用。这些发现代表了改善癌症治疗结果的重要一步。正如临床试验所证明的那样,免疫检查点抑制剂(ICIs)的施用,特别是与抗血管生成剂和酪氨酸激酶抑制剂联合使用,延长了一部分HCC患者的生存期,为一线治疗进展的患者提供替代方案。在这次审查中,我们旨在提供HCC的概述和免疫系统在其发展中的作用,总结涉及ICI的临床试验的结果,作为单一疗法或与其他药物联合治疗该疾病。还概述了在HCC治疗中施用ICIs的挑战和考虑因素。
    Despite advances in cancer treatment, hepatocellular carcinoma (HCC), the most common form of liver cancer, remains a major public health problem worldwide. The immune microenvironment plays a critical role in regulating tumor progression and resistance to therapy, and in HCC, the tumor microenvironment (TME) is characterized by an abundance of immunosuppressive cells and signals that facilitate immune evasion and metastasis. Recently, anti-cancer immunotherapies, therapeutic interventions designed to modulate the immune system to recognize and eliminate cancer, have become an important cornerstone of cancer therapy. Immunotherapy has demonstrated the ability to improve survival and provide durable cancer control in certain groups of HCC patients, while reducing adverse side effects. These findings represent a significant step toward improving cancer treatment outcomes. As demonstrated in clinical trials, the administration of immune checkpoint inhibitors (ICIs), particularly in combination with anti-angiogenic agents and tyrosine kinase inhibitors, has prolonged survival in a subset of patients with HCC, providing an alternative for patients who progress on first-line therapy. In this review, we aimed to provide an overview of HCC and the role of the immune system in its development, and to summarize the findings of clinical trials involving ICIs, either as monotherapies or in combination with other agents in the treatment of the disease. Challenges and considerations regarding the administration of ICIs in the treatment of HCC are also outlined.
    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
    免疫细胞死亡(ICD)正在成为抗肿瘤治疗的关键组成部分,它利用患者的免疫系统来对抗癌症。近年来,在改进基于ICD的治疗方面,我们做出了几项努力.这里,我们讨论了基于纳米材料的策略如何提高ICD的疗效,并强调了它们的益处和挑战.
    Immunogenic cell death (ICD) is emerging as a key component of antitumor therapy that harnesses the immune system of the patient to combat cancer. In recent years, several efforts were made to improve the ICD-based therapies. Here, we discuss how nanomaterial-based strategies increase the efficacy of ICD and highlight their benefits and challenges.
    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
    The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
    Lung cancer remains a significant global health concern, and researchers are actively exploring innovative approaches to boost the immune system’s ability to recognize and destroy cancer cells. Boosting the immune system responses against the lung tumor microenvironment is one of promising approaches for lung cancer therapy. The lung tumor microenvironment refers to the complex network of cells, proteins, and molecules that surround and support the growth of lung tumors. Unfortunately, this environment often hinders the body’s immune response, allowing cancer cells to evade detection and destruction. By comprehending the cellular and molecular factors at play, researchers can devise novel strategies to tip the balance in favor of the immune system. Cancer cells often employ various mechanisms to suppress the immune system within the lung tumor microenvironment. One approach to combating this suppression is the use of adjuvants, substances that enhance the immune response. Adjuvants can be administered alongside cancer vaccines or other immunotherapies to strengthen the immune system’s ability to recognize and attack tumor cells. The recent progresses have shown the potential of some products, adjuvants, immunotherapy drugs, vaccines, and nanoparticles. This article aims to discuss recent advancements in the field of cancer immunotherapy, specifically focusing on strategies to strengthen the body’s immune response against lung tumors.
    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: Meta-Analysis
    目的:探讨性别与免疫检查点抑制剂(ICIs)的关系。材料与方法:我们使用交互测试评估了性别之间ICI的生存结果差异。结果:共纳入108项研究,代表70,243例患者。在第一行设置中,合并交互作用HR为0.97(95%CI:0.91-1.04).在后续行设置中,合并交互作用HR为0.85(95%CI:0.77-0.95).当ICIs作为围手术期治疗或全身治疗给予PD-L1表达阳性的患者时,男女都获得了平等的生存福利。结论:两性,线的治疗,在决定向患者提供ICI时,在评估风险与获益时,应考虑癌症(亚)类型和PD-L1状态.
    男人和女人之间有很多区别,这些差异是否也影响免疫检查点抑制剂的疗效是重要的,因为免疫检查点抑制剂已被广泛接受为恶性肿瘤患者的标准化治疗.先前的研究报告了相互矛盾的结果。进行这项研究是为了通过其他可用报告重新评估这种性别差异。我们发现男性和女性都可以从免疫检查点抑制剂中受益,而在决定向已经接受大量治疗的患者提供免疫检查点抑制剂时,应考虑性别。
    Aim: To explore the association between sex and immune checkpoint inhibitors (ICIs). Materials & methods: We assessed the difference in survival outcomes from ICIs between sexes using an interaction test. Results: 108 studies representing 70,243 patients were included. In the first-line setting, the pooled interaction HR was 0.97 (95% CI: 0.91-1.04). In the subsequent-line setting, the pooled interaction HR was 0.85 (95% CI: 0.77-0.95). When ICIs were given as perioperative therapy or as systemic therapy in patients with positive PD-L1 expression, both men and women obtained equal survival benefits. Conclusion: Both sex, line of therapy, cancer (sub)type and PD-L1 status should be taken into account in the assessment of risk versus benefit when deciding to offer ICIs to patients.
    There are many differences between men and women, and whether these differences also affect the efficacy of immune checkpoint inhibitors is important because immune checkpoint inhibitors have been widely accepted as standardized treatments for patients with malignant tumors. Previous studies have reported conflicting results. This research was conducted to reassess this sex disparity with additional available reports. We found that both men and women can benefit from immune checkpoint inhibitors, while sex should be taken into account when deciding to offer immune checkpoint inhibitors to patients who have already been heavily treated.
    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
    microRNAs(miRNAs)是19-23个核苷酸长,在转录后水平调节基因表达的进化保守的非编码RNA分子。在这次审查中,miRNA参与免疫细胞的分化和功能,在抗感染免疫反应中,免疫缺陷和自身免疫性疾病。还讨论了miRNA在抗癌免疫以及实体器官和造血干细胞移植中的作用。主要关注miRNAs的翻译临床应用,包括建立用于鉴别诊断和预测预后的无创生物标志物。对生物治疗的患者选择和反应预测是最有前途的应用领域之一。miRNA的替代或抑制具有巨大的治疗潜力,不断扩大的可能性。尽管重要的挑战仍在等待解决方案,miRNA指纹图谱的评估可能有助于免疫失调的日益个性化管理,并显著降低毒性和治疗副作用。关于体育锻炼和营养对免疫系统的分子影响的更详细的知识可能有助于自我定制的生活方式建议和预防进展。
    MicroRNAs (miRNAs) are 19-23 nucleotide long, evolutionarily conserved noncoding RNA molecules that regulate gene expression at the post-transcriptional level. In this review, involvement of miRNAs is summarized in the differentiation and function of immune cells, in anti-infective immune responses, immunodeficiencies and autoimmune diseases. Roles of miRNAs in anticancer immunity and in the transplantation of solid organs and hematopoietic stem cells are also discussed. Major focus is put on the translational clinical applications of miRNAs, including the establishment of noninvasive biomarkers for differential diagnosis and prediction of prognosis. Patient selection and response prediction to biological therapy is one of the most promising fields of application. Replacement or inhibition of miRNAs has enormous therapeutic potential, with constantly expanding possibilities. Although important challenges still await solutions, evaluation of miRNA fingerprints may contribute to an increasingly personalized management of immune dysregulation with a remarkable reduction in toxicity and treatment side effects. More detailed knowledge of the molecular effects of physical exercise and nutrition on the immune system may facilitate self-tailored lifestyle recommendations and advances in prevention.
    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
    简介:尽管在理解癌症生物学方面取得了进展,恶性胶质瘤仍然无法治愈。本工作进行了一项多组学分析,以研究神经胶质瘤中氯化物细胞内通道1(CLIC1)的重要性。方法:覆盖转录组学的胶质瘤的多组学数据,基因组学,来自多个公共队列的DNA甲基化和单细胞转录组学被登记用于分析CLIC1。在CLIC1siRNA转染后,进行体外实验以测量U251和U373神经胶质瘤细胞中的凋亡和细胞迀移率。结果:CLIC1表达升高被证明可以稳定且独立地估计较差的生存结果。CLIC1表达在更晚期阶段更高,野生型IDH和未甲基化MGMT样品。在CLIC1上调的肿瘤中,致瘤性和抗癌免疫途径显着丰富。此外,CLIC1与癌症免疫周期呈正相关,基质激活,DNA损伤修复和细胞周期。抑制CLIC1导致神经胶质瘤细胞的凋亡和细胞运动减弱。在CLIC1上调的肿瘤中发现了更频繁的基因组改变。CLIC1表达与DNA甲基化呈显著负相关。CLIC1高表达样本对喜树碱较敏感,顺铂,阿霉素,厄洛替尼,紫杉醇,雷帕霉素,氯法拉滨,坦司匹霉素,甲氨蝶呤,依维莫司,TAK-733、曲美替尼和AZD8330。CLIC1上调的肿瘤表现出丰富的免疫细胞浸润,免疫检查点和调节剂的更高表达和类似的转录组分析,表明对免疫检查点阻断(ICB)的反应良好。然而,由于潮汐分数升高,具有CLIC1上调的肿瘤似乎对ICB有抗性。单细胞分析揭示了CLIC1在肿瘤细胞和肿瘤微环境中普遍表达。结论:总体而言,CLIC1是神经胶质瘤的一个有希望的治疗弱点。
    Introduction: Despite advances in comprehending cancer biology, malignant gliomas remain incurable. The present work conducted a multi-omics analysis for investigating the significance of chloride intracellular channel 1 (CLIC1) in gliomas. Methods: Multi-omics data of glioma covering transcriptomics, genomics, DNA methylation and single-cell transcriptomics from multiple public cohorts were enrolled for analyzing CLIC1. In vitro experiments were conducted to measure apoptosis and cell mobility in U251 and U373 glioma cells following transfection of CLIC1 siRNAs. Results: Elevated CLIC1 expression was proven to stably and independently estimate worse survival outcomes. CLIC1 expression was higher in more advanced stage, wild-type IDH and unmethylated MGMT samples. Tumorigenic and anticancer immunity pathways were remarkably enriched in CLIC1-up-regulated tumors. Additionally, CLIC1 was positively linked with cancer-immunity cycle, stromal activation, DNA damage repair and cell cycle. Suppressing CLIC1 resulted in apoptosis and attenuated cell motility of glioma cells. More frequent genomic alterations were found in CLIC1-up-regulated tumors. CLIC1 expression presented a remarkably negative connection to DNA methylation. High CLIC1 expression samples were more sensitive to camptothecin, cisplatin, doxorubicin, erlotinib, paclitaxel, rapamycin, clofarabine, tanespimycin, methotrexate, everolimus, TAK-733, trametinib and AZD8330. Tumors with upregulated CLIC1 presented abundant immune cell infiltration, higher expression of immune-checkpoints and -modulators and similar transcriptome profiling, indicative of well response to immune-checkpoint blockade (ICB). Nevertheless, due to elevated TIDE score, tumors with CLIC1 upregulation appeared to be resistant to ICB. Single-cell analysis unveiled that CLIC1 was expressed ubiquitously in tumor cells and tumor microenvironment. Conclusions: Overall, CLIC1 was a promising treatment vulnerability in 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
    炎性细胞死亡是程序性细胞死亡(PCD)的一种形式,其在该过程中诱导炎性介质。细胞死亡期间炎症介质的产生在标准癌症治疗中是有益的,因为它可以打破癌症中的免疫沉默并诱导抗癌免疫。光动力疗法(PDT)是一种使用光敏剂分子和光源破坏癌细胞的癌症疗法,目前用于治疗临床上不同类型的癌症。在这项研究中,我们调查了使用5-氨基乙酰丙酸(5-ALA-PDT)的PDT是否会导致炎性细胞死亡,随后,增加癌细胞的免疫原性。
    用5-ALA处理小鼠乳腺癌(4T1)和人结肠癌(DLD-1)细胞4小时,然后用光源照射。通过蛋白质印迹分析和FACS测量PCD诱导。通过透射电子显微镜(TEM)确定形态变化。BALB/c小鼠注射无细胞培养基,冷冻/解冻细胞的上清液或PDT细胞的上清液每周肌内注射4周,然后在BALB/c的右后侧翼用4T1细胞攻击。监测肿瘤生长12天。
    我们发现5-ALA-PDT诱导炎性细胞死亡,但不是凋亡,在体外4T1细胞和DLD-1细胞中。此外,当小鼠用5-ALA-PDT培养上清液预处理时,与用冷冻和解冻(F/T)4T1培养上清液预处理的那些相比,4T1肿瘤的生长被显著抑制。
    这些结果表明5-ALA-PDT诱导炎性细胞死亡,其促进体内抗癌免疫。
    UNASSIGNED: Inflammatory cell death is a form of programmed cell death (PCD) that induces inflammatory mediators during the process. The production of inflammatory mediators during cell death is beneficial in standard cancer therapies as it can break the immune silence in cancers and induce anticancer immunity. Photodynamic therapy (PDT) is a cancer therapy with photosensitizer molecules and light sources to destroy cancer cells, which is currently used for treating different types of cancers in clinical settings. In this study, we investigated if PDT using 5-aminolevulinic (5-ALA-PDT) causes inflammatory cell death and, subsequently, increases the immunogenicity of cancer cells.
    UNASSIGNED: Mouse breast cancer (4T1) and human colon cancer (DLD-1) cells were treated with 5-ALA for 4 hours and then irradiated with a light source. PCD induction was measured by western blot analysis and FACS. Morphological changes were determined by transmission electron microscopy (TEM). BALB/c mice were injected with cell-free media, supernatant of freeze/thaw cells or supernatant of PDT cells intramuscular every week for 4 weeks and then challenged with 4T1 cells at the right hind flank of BALB/c. Tumor growth was monitored for 12 days.
    UNASSIGNED: We found that 5-ALA-PDT induces inflammatory cell death, but not apoptosis, in 4T1 cells and DLD-1 cells in vitro. Moreover, when mice were pretreated with 5-ALA-PDT culture supernatant, the growth of 4T1 tumors was significantly suppressed compared to those pretreated with freeze and thaw (F/T) 4T1 culture supernatant.
    UNASSIGNED: These results indicate that 5-ALA-PDT induces inflammatory cell death which promotes anticancer immunity in vivo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号