tumor immunotherapy

肿瘤免疫治疗
  • 文章类型: Journal Article
    阻断程序性细胞死亡-1(PD-1)/程序性细胞死亡配体1(PD-L1)途径是一种有吸引力的免疫治疗策略,但小分子PD-1/PD-L1抑制剂的临床应用尚不清楚。在这项工作中,基于BMS-202和我们先前的工作YLW-106,设计并合成了一系列以苯并[d]异噻唑结构为支架的化合物。通过均相时间分辨荧光(HTRF)测定法评估了它们对PD-1/PD-L1相互作用的抑制活性。其中,LLW-018(27c)表现出最有效的抑制活性,IC50值为2.61nM。细胞水平测定表明LLW-018表现出对JurkatT和MDA-MB-231的低细胞毒性。基于PD-1NFAT-LucJurkat细胞和PD-L1TCR激活剂CHO细胞的进一步基于细胞的PD-1/PD-L1阻断生物测定表明,LLW-018可以中断PD-1/PD-L1相互作用,IC50值为0.88μM。多种计算方法,包括分子对接,分子动力学,MM/GBSA,MM/PBSA,元动力学,和QM/MMMD用于PD-L1二聚体复合物,这揭示了LLW-018和C2对称小分子抑制剂LCH1307的结合模式和解离过程。这些结果表明,LLW-018作为PD-1/PD-L1抑制剂表现出有希望的效力,用于进一步研究。
    Blockade of the programmed cell death-1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is an attractive strategy for immunotherapy, but the clinical application of small molecule PD-1/PD-L1 inhibitors remains unclear. In this work, based on BMS-202 and our previous work YLW-106, a series of compounds with benzo[d]isothiazol structure as scaffold were designed and synthesized. Their inhibitory activity against PD-1/PD-L1 interaction was evaluated by a homogeneous time-resolved fluorescence (HTRF) assay. Among them, LLW-018 (27c) exhibited the most potent inhibitory activity with an IC50 value of 2.61 nM. The cellular level assays demonstrated that LLW-018 exhibited low cytotoxicity against Jurkat T and MDA-MB-231. Further cell-based PD-1/PD-L1 blockade bioassays based on PD-1 NFAT-Luc Jurkat cells and PD-L1 TCR Activator CHO cells indicated that LLW-018 could interrupt PD-1/PD-L1 interaction with an IC50 value of 0.88 μM. Multi-computational methods, including molecular docking, molecular dynamics, MM/GBSA, MM/PBSA, Metadynamics, and QM/MM MD were utilized on PD-L1 dimer complexes, which revealed the binding modes and dissociation process of LLW-018 and C2-symmetric small molecule inhibitor LCH1307. These results suggested that LLW-018 exhibited promising potency as a PD-1/PD-L1 inhibitor for further investigation.
    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
    溶瘤腺病毒(OAs)的治疗功效依赖于有效的病毒转导和复制。然而,柯萨奇腺病毒受体在许多肿瘤中的有限表达,随着细胞内的抗病毒信号,对OA感染和溶瘤形成重大障碍。这里,我们介绍了声敏剂武装的OAs(saOAs),它通过声动力疗法增强的病毒复制来增强溶瘤病毒疗法的抗肿瘤功效。saOAs不仅可以通过转铁蛋白受体介导的内吞作用有效感染肿瘤细胞,而且在超声照射下表现出增强的病毒复制和肿瘤溶瘤作用。我们发现,装载在病毒上的超声增敏剂诱导了肿瘤细胞内ROS的产生,触发JNK介导的自噬,最终导致病毒复制增强。在恶性黑色素瘤的小鼠模型中,SAOAs和声动力疗法的结合引发了强大的抗肿瘤免疫反应,导致显著抑制黑素瘤生长并改善宿主存活。这项工作强调了声动力疗法在增强OAs有效性方面的潜力,并为充分利用溶瘤病毒疗法的抗肿瘤功效提供了一个有希望的平台。
    The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.
    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
    金属离子通过在肿瘤微环境(TME)中传递细胞内和细胞外信号,在调节免疫细胞功能中起着至关重要的作用。在这些免疫细胞中,我们关注金属离子对T细胞的影响,因为它们可以识别和杀死癌细胞,在基于免疫的癌症治疗中发挥重要作用.金属离子通常用于肿瘤免疫治疗的纳米药物中。在这次审查中,我们讨论了与抗肿瘤免疫有关的七种金属离子,阐明它们在免疫治疗中的作用,并为肿瘤免疫治疗和临床应用提供新的见解。
    Metal ions play an essential role in regulating the functions of immune cells by transmitting intracellular and extracellular signals in tumor microenvironment (TME). Among these immune cells, we focused on the impact of metal ions on T cells because they can recognize and kill cancer cells and play an important role in immune-based cancer treatment. Metal ions are often used in nanomedicines for tumor immunotherapy. In this review, we discuss seven metal ions related to anti-tumor immunity, elucidate their roles in immunotherapy, and provide novel insights into tumor immunotherapy and clinical applications.
    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
    杀伤细胞凝集素样受体G1(KLRG1)是主要在NK和T细胞亚群中表达的免疫检查点受体,其下调免疫细胞的活化和增殖并参与细胞介导的免疫应答。越来越多的证据表明,KLRG1作为一个值得注意的疾病标志物和治疗靶点的重要性,可以影响疾病的发作。programming,和预后。阻断KLRG1已被证明可以有效减轻各种小鼠肿瘤模型的下调效应,包括实体瘤和血液恶性肿瘤。然而,KLRG1抑制剂尚未被批准用于人类使用,对KLRG1的表达及其在各种疾病中的作用机制的认识尚不完全。在这次审查中,我们探索分布的变化,结构,和KLRG1在免疫细胞中的信号通路,并总结其在自身免疫性疾病发生发展中的表达模式和作用,传染病,和癌症。此外,我们讨论了KLRG1作为肿瘤免疫治疗工具的潜在应用。
    Killer cell lectin-like receptor G1 (KLRG1) is an immune checkpoint receptor expressed predominantly in NK and T-cell subsets that downregulates the activation and proliferation of immune cells and participates in cell-mediated immune responses. Accumulating evidence has demonstrated the importance of KLRG1 as a noteworthy disease marker and therapeutic target that can influence disease onset, progression, and prognosis. Blocking KLRG1 has been shown to effectively mitigate the effects of downregulation in various mouse tumor models, including solid tumors and hematologic malignancies. However, KLRG1 inhibitors have not yet been approved for human use, and the understanding of KLRG1 expression and its mechanism of action in various diseases remains incomplete. In this review, we explore alterations in the distribution, structure, and signaling pathways of KLRG1 in immune cells and summarize its expression patterns and roles in the development and progression of autoimmune diseases, infectious diseases, and cancers. Additionally, we discuss the potential applications of KLRG1 as a tool for 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
    肿瘤的低免疫原性对有效肿瘤免疫治疗的发展提出了挑战。然而,新出现的证据表明,某些治疗方法,比如化疗,放射治疗,和光疗,可以诱导不同程度的免疫原性细胞死亡(ICD)。这种ICD现象导致肿瘤抗原的释放和树突状细胞(DC)的成熟,从而增强肿瘤的免疫原性和促进免疫应答。然而,使用单一常规ICD诱导剂通常无法实现原位肿瘤消融并建立长期抗肿瘤免疫反应。此外,ICD诱导的诱导因不同的方法而异,以及治疗剂在体内的分布影响ICD的水平和毒副作用的发生。为了应对这些挑战并进一步提高肿瘤免疫力,研究人员已经探索了纳米系统作为ICD与肿瘤免疫治疗的诱导剂。这篇综述探讨了ICD的机制和不同的诱导方法,特别关注ICD与肿瘤免疫的关系。目的是探索利用各种纳米材料诱导ICD增强机体抗肿瘤作用的研究进展。本文旨在为基于纳米材料的ICD诱导剂在肿瘤免疫治疗领域的开发和临床应用提供重要的理论指导和实践参考。
    Low immunogenicity of tumors poses a challenge in the development of effective tumor immunotherapy. However, emerging evidence suggests that certain therapeutic approaches, such as chemotherapy, radiotherapy, and phototherapy, can induce varying degrees of immunogenic cell death (ICD). This ICD phenomenon leads to the release of tumor antigens and the maturation of dendritic cells (DCs), thereby enhancing tumor immunogenicity and promoting immune responses. However, the use of a single conventional ICD inducer often fails to achieve in situ tumor ablation and establish long-term anti-tumor immune responses. Furthermore, the induction of ICD induction varies among different approaches, and the distribution of the therapeutic agent within the body influences the level of ICD and the occurrence of toxic side effects. To address these challenges and further boost tumor immunity, researchers have explored nanosystems as inducers of ICD in combination with tumor immunotherapy. This review examines the mechanisms of ICD and different induction methods, with a specific focus on the relationship between ICD and tumor immunity. The aim is to explore the research advancements utilizing various nanomaterials to enhance the body\'s anti-tumor effects by inducing ICD. This paper aims to contribute to the development and clinical application of nanomaterial-based ICD inducers in the field of cancer immunotherapy by providing important theoretical guidance and practical references.
    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
    自然杀伤(NK)细胞,作为先天淋巴细胞,具有细胞毒性能力,并通过激活和抑制受体的一系列功能吸引靶细胞。特别是,NK细胞上的自然杀伤组2成员D(NKG2D)受体识别应激诱导的配体-肿瘤细胞上呈递的MHCI类链相关分子A和B(MICA/B),是触发NK细胞溶细胞反应的关键。然而,肿瘤已经发展出复杂的策略来逃避NK细胞监测,导致肿瘤免疫治疗失败。在本文中,我们总结了这些免疫逃避策略,包括激活受体的配体的下调,抑制性受体的配体上调,分泌免疫抑制化合物,和凋亡抵抗的发展。然后,我们专注于NK细胞免疫治疗的最新进展,其中包括激活NK细胞受体,上调NKG2D配体MICA/B表达,阻断抑制性NK细胞受体,过继NK细胞疗法,嵌合抗原受体(CAR)-工程NK细胞(CAR-NK),和NKG2DCAR-T细胞,特别是针对MICA/B的几种疫苗。这篇综述将启发肿瘤中NK细胞生物学的研究,并通过利用NK细胞的强大细胞毒活性为改善癌症治疗结果提供重要希望。
    Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
    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
    原发性肝癌是全球癌症相关死亡的第二大原因。目前,一旦被诊断出肝癌通常处于晚期,治疗效果一般较差。因此,迫切需要其他有效的治疗方法。巨噬细胞是肿瘤微环境的重要组成部分,巨噬细胞极化对肿瘤的增殖和分化至关重要。巨噬细胞亚型之间的调节相互作用,如M1和M2,导致许多临床结果,包括肿瘤进展和转移。所以,研究这一过程的驱动因素非常重要。长链非编码RNA已被广泛证明在肿瘤的早期诊断和治疗中具有重要价值。许多研究表明,长链非编码RNA通过其驱动M1或M2极化的能力参与巨噬细胞极化,从而参与肝癌的发生发展。在这篇文章中,我们系统地阐述了参与肝癌巨噬细胞极化的长链非编码RNA,希望能为肝癌的早期诊断和治疗提供新的思路。从PubMed检索肝癌相关研究。基于我们对LncRNA和巨噬细胞极化作为肝癌有效疗法的鉴定,我们分析了PubMed系统在过去十年中关于LncRNA和巨噬细胞极化之间的串扰的研究文章。通过靶向M1/M2巨噬细胞极化,LncRNA可能促进或抑制肝癌,参考文献主要由文章的影响因子决定。因此,探讨了LncRNA与M1/M2巨噬细胞极化的具体作用机制,随着它们在串扰发生中的作用,扩散,和肝癌转移。lncRNA在肝癌中双向表达,可以靶向巨噬细胞极化来调节肿瘤行为。lncRNA主要起ceRNA的作用,可通过细胞外囊泡参与肝癌细胞与巨噬细胞的串扰。lncRNA可能通过靶向巨噬细胞参与肝癌的免疫治疗,成为肝癌的一种新的生物分子标志物。
    Primary liver cancer is the second leading cause of cancer-related death worldwide. At present, liver cancer is often in an advanced stage once diagnosed, and treatment effects are generally poor. Therefore, there is an urgent need for other powerful treatments. Macrophages are an important component of the tumor microenvironment, and macrophage polarization is crucial to tumor proliferation and differentiation. Regulatory interactions between macrophage subtypes, such as M1 and M2, lead to a number of clinical outcomes, including tumor progression and metastasis. So, it is important to study the drivers of this process. Long non-coding RNA has been widely proven to be of great value in the early diagnosis and treatment of tumors. Many studies have shown that long non-coding RNA participates in macrophage polarization through its ability to drive M1 or M2 polarization, thereby participating in the occurrence and development of liver cancer. In this article, we systematically elaborated on the long non-coding RNAs involved in the polarization of liver cancer macrophages, hoping to provide a new idea for the early diagnosis and treatment of liver cancer. Liver cancer- related studies were retrieved from PubMed. Based on our identification of LncRNA and macrophage polarization as powerful therapies for liver cancer, we analyzed research articles in the PubMed system in the last ten years on the crosstalk between LncRNA and macrophage polarization. By targeting M1/M2 macrophage polarization, LncRNA may promote or suppress liver cancer, and the references are determined primarily by the article\'s impact factor. Consequently, the specific mechanism of action between LncRNA and M1/M2 macrophage polarization was explored, along with the role of their crosstalk in the occurrence, proliferation, and metastasis of liver cancer. lncRNA is bidirectionally expressed in liver cancer and can target macrophage polarization to regulate tumor behavior. lncRNA mainly functions as ceRNA and can participate in the crosstalk between liver cancer cells and macrophages through extracellular vesicles. lncRNA can potentially participate in the immunotherapy of liver cancer by targeting macrophages and becoming a new biomolecular marker of liver cancer.
    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
    自然杀伤(NK)细胞,作为免疫系统的前线防御,并且能够监视和消除肿瘤细胞。近年来,它们在肿瘤免疫治疗中的意义引起了广泛关注。然而,NK细胞和肿瘤细胞之间缺乏特异性受体-配体相互作用阻碍了它们的选择性,从而限制了基于NK细胞的肿瘤免疫疗法的治疗效果。在这里,我们通过代谢糖工程和无铜点击化学构建多甘露糖工程NK(pM-NK)细胞。合成了含有二苯并环辛炔末端基团(pM-DBCO)的聚甘露糖,并在叠氮标记的NK细胞表面上进行了共价修饰。与未经处理的NK细胞相比,pM-NK细胞和MDA-MB-231细胞之间的相互作用,具有甘露糖受体过表达的乳腺肿瘤细胞系,显着增加,并导致杀死效力显著增强。因此,静脉内给予pM-NK细胞可以有效抑制肿瘤生长,并延长携带MDA-MB-231肿瘤的小鼠的生存期。因此,这项工作为肿瘤靶向NK细胞的肿瘤免疫治疗提供了一种新策略.本文受版权保护。保留所有权利。
    Natural killer (NK) cells, serve as the frontline defense of the immune system, and are capable of surveilling and eliminating tumor cells. Their significance in tumor immunotherapy has garnered considerable attention in recent years. However, the absence of specific receptor-ligand interactions between NK cells and tumor cells hampers their selectivity, thereby limiting the therapeutic effectiveness of NK cell-based tumor immunotherapy. Herein, this work constructs polymannose-engineered NK (pM-NK) cells via metabolic glycoengineering and copper-free click chemistry. Polymannose containing dibenzocyclooctyne terminal groups (pM-DBCO) is synthesized and covalently modified on the surface of azido-labeled NK cells. Compared to the untreated NK cells, the interactions between pM-NK cells and MDA-MB-231 cells, a breast tumor cell line with overexpression of mannose receptors (MRs), are significantly increased, and lead to significantly enhanced killing efficacy. Consequently, intravenous administration of pM-NK cells will effectively inhibit the tumor growth and will prolong the survival of mice bearing MDA-MB-231 tumors. Thus, this work presents a novel strategy for tumor-targeting NK cell-based tumor 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
    乳腺癌具有高发病率和高转移倾向。缺乏有效干预的精确目标,因此必须制定增强的治疗策略。外泌体,以脂双层为特征,尺寸范围从30到150nm,可以被各种细胞主动释放,包括那些肿瘤。来自不同免疫细胞亚群的外来体已经显示出调节肿瘤内的免疫微环境并影响乳腺癌进展。此外,肿瘤来源的外泌体已被证明有助于乳腺癌的发生和发展,并可能成为乳腺癌免疫治疗的新靶点.肿瘤免疫疗法已成为治疗肿瘤的一种选择,和外来体已成为可用于各种病理状况的治疗载体。编辑的外泌体可用作乳腺癌治疗的纳米级药物递送系统,有助于免疫抑制肿瘤微环境的重塑并影响免疫治疗的疗效。本文综述了来自不同细胞的外泌体在乳腺癌中的调节作用以及外泌体作为纳米药物递送系统和免疫治疗剂在乳腺癌中的最新应用。显示外泌体在乳腺癌临床治疗中的发展前景。
    Breast cancer has a high incidence and a heightened propensity for metastasis. The absence of precise targets for effective intervention makes it imperative to devise enhanced treatment strategies. Exosomes, characterized by a lipid bilayer and ranging in size from 30 to 150 nm, can be actively released by various cells, including those in tumors. Exosomes derived from distinct subsets of immune cells have been shown to modulate the immune microenvironment within tumors and influence breast cancer progression. In addition, tumor-derived exosomes have been shown to contribute to breast cancer development and progression and may become a new target for breast cancer immunotherapy. Tumor immunotherapy has become an option for managing tumors, and exosomes have become therapeutic vectors that can be used for various pathological conditions. Edited exosomes can be used as nanoscale drug delivery systems for breast cancer therapy, contributing to the remodeling of immunosuppressive tumor microenvironments and influencing the efficacy of immunotherapy. This review discusses the regulatory role of exosomes from different cells in breast cancer and the latest applications of exosomes as nanoscale drug delivery systems and immunotherapeutic agents in breast cancer, showing the development prospects of exosomes in the clinical treatment of breast cancer.
    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)使T细胞免于耗尽并增强对肿瘤的T细胞应答。然而,大多数患者对ICB治疗没有反应,在具有少量浸润淋巴细胞的“冷”肿瘤中,只能实现有限的反应。合成生物学可用于工程化细菌作为可控生物反应器以原位合成生物治疗剂。我们设计了具有合成基因回路的减毒沙门氏菌VNP20009,以产生PD-1和Tim-3scFv,以阻断耗尽的T细胞上的免疫抑制受体,以恢复其抗肿瘤反应。分泌的PD-1和Tim-3scFv通过它们的靶向受体在体外结合PD-1+Tim-3+T细胞并增强IFN-γ的T细胞分泌。工程化细菌定植于肿瘤的缺氧核心,并原位合成PD-1和Tim-3scFv,恢复CD4+T细胞和CD8+T细胞以执行抗肿瘤反应。这种细菌还引发了强烈的先天免疫反应,其刺激肿瘤内IFN-γ+CD4+T细胞的扩增以诱导直接和间接的抗肿瘤免疫。
    Immune-checkpoint blockade (ICB) reinvigorates T cells from exhaustion and potentiates T-cell responses to tumors. However, most patients do not respond to ICB therapy, and only a limited response can be achieved in a \"cold\" tumor with few infiltrated lymphocytes. Synthetic biology can be used to engineer bacteria as controllable bioreactors to synthesize biotherapeutics in situ. We engineered attenuated Salmonella VNP20009 with synthetic gene circuits to produce PD-1 and Tim-3 scFv to block immunosuppressive receptors on exhausted T cells to reinvigorate their antitumor response. Secreted PD-1 and Tim-3 scFv bound PD-1+ Tim-3+ T cells through their targeting receptors in vitro and potentiated the T-cell secretion of IFN-γ. Engineered bacteria colonized the hypoxic core of the tumor and synthesized PD-1 and Tim-3 scFv in situ, reviving CD4+ T cells and CD8+ T cells to execute an antitumor response. The bacteria also triggered a strong innate immune response, which stimulated the expansion of IFN-γ+ CD4+ T cells within the tumors to induce direct and indirect antitumor immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号