Immunosuppressive tumor microenvironment

免疫抑制肿瘤微环境
  • 文章类型: Journal Article
    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
    尽管在治疗某些类型的癌症方面取得了重大进展,高级别胶质瘤(HGG)仍然是一个重要的临床问题。在胶质母细胞瘤(GBM)的情况下,成人中枢神经系统最常见的实体瘤,诊断后的平均生存时间仅为15-18个月,尽管使用了强化多模式治疗。表达嵌合抗原受体(CAR)的T细胞,食品和药物管理局已经批准用于治疗某些血液系统恶性肿瘤,是一个新的,有希望的治疗选择。然而,由于免疫抑制性肿瘤微环境(TME),CAR-T细胞在实体瘤中的疗效较低.因此,将免疫抑制性TME重编程为促炎表型似乎特别重要,因为它可以提高CAR-T细胞在实体瘤治疗中的有效性。以下文献综述旨在呈现临床前研究的结果,其显示通过将TME重编程为促炎表型来改善CAR-T在GBM的TME中的功效的可能性。由于在与溶瘤病毒组合的协同疗法中使用CAR-T可能是可以实现的,放射治疗,或表观遗传抑制剂,以及通过支持CAR-T细胞穿越血脑屏障,使TME中受损的血管生成正常化,通过细胞因子信号传导或通过阻断/敲除T细胞抑制剂改善CAR-T效应子功能,和调节microRNA表达。以这种方式修饰的CAR-T细胞在协同治疗中的使用可以通过诱导内源性抗肿瘤反应而导致HGG患者的更长存活。
    Despite significant progress in the treatment of some types of cancer, high-grade gliomas (HGGs) remain a significant clinical problem. In the case of glioblastoma (GBM), the most common solid tumor of the central nervous system in adults, the average survival time from diagnosis is only 15-18 months, despite the use of intensive multimodal therapy. Chimeric antigen receptor (CAR)-expressing T cells, which have already been approved by the Food and Drug Administration for use in the treatment of certain hematologic malignancies, are a new, promising therapeutic option. However, the efficacy of CAR-T cells in solid tumors is lower due to the immunosuppressive tumor microenvironment (TME). Reprogramming the immunosuppressive TME toward a pro-inflammatory phenotype therefore seems particularly important because it may allow for increasing the effectiveness of CAR-T cells in the therapy of solid tumors. The following literature review aims to present the results of preclinical studies showing the possibilities of improving the efficacy of CAR-T in the TME of GBM by reprogramming the TME toward a pro-inflammatory phenotype. It may be achievable thanks to the use of CAR-T in a synergistic therapy in combination with oncolytic viruses, radiotherapy, or epigenetic inhibitors, as well as by supporting CAR-T cells crossing of the blood-brain barrier, normalizing impaired angiogenesis in the TME, improving CAR-T effector functions by cytokine signaling or by blocking/knocking out T-cell inhibitors, and modulating the microRNA expression. The use of CAR-T cells modified in this way in synergistic therapy could lead to the longer survival of patients with HGG by inducing an endogenous anti-tumor response.
    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
    肝细胞癌(HCC)是一种发病率高、异质性高的恶性肿瘤。复发,和转移率。免疫疗法的出现改善了晚期肝癌的治疗,但临床上仍存在耐药和免疫相关不良事件等问题。HCC的免疫抑制性肿瘤微环境(TME)限制了免疫治疗的疗效,对HCC的进展和转移至关重要。因此,有必要阐明免疫抑制TME背后的机制以开发和应用免疫疗法.本文系统总结了肝癌的发病机制,高度异质的TME的形成,以及免疫抑制TME加速HCC进展和转移的机制。我们还回顾了HCC免疫治疗的现状,并进一步讨论了针对免疫抑制性TME的现有挑战和潜在治疗策略。我们希望通过全面了解肝癌免疫抑制TME的结构和功能,为优化和创新免疫治疗策略提供启发。
    Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with high incidence, recurrence, and metastasis rates. The emergence of immunotherapy has improved the treatment of advanced HCC, but problems such as drug resistance and immune-related adverse events still exist in clinical practice. The immunosuppressive tumor microenvironment (TME) of HCC restricts the efficacy of immunotherapy and is essential for HCC progression and metastasis. Therefore, it is necessary to elucidate the mechanisms behind immunosuppressive TME to develop and apply immunotherapy. This review systematically summarizes the pathogenesis of HCC, the formation of the highly heterogeneous TME, and the mechanisms by which the immunosuppressive TME accelerates HCC progression and metastasis. We also review the status of HCC immunotherapy and further discuss the existing challenges and potential therapeutic strategies targeting immunosuppressive TME. We hope to inspire optimizing and innovating immunotherapeutic strategies by comprehensively understanding the structure and function of immunosuppressive TME in 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
    角化是一种以铜依赖性方式调节细胞死亡的新类型,并已被报道参与各种恶性肿瘤的发生和发展。然而,透明细胞肾细胞癌(ccRCC)的角化和肿瘤微环境(TME)之间的关系尚不清楚.为了解决这个问题,我们整合了ccRCC不同阶段的单细胞RNA测序(scRNA-seq)数据集,系统地检查了ccRCC的TME中角化相关基因(CRGs)的独特表达模式,并使用空间转录组测序(ST-seq)数据集探索了关键特征。随着ccRCC的发展,癌组织中的细胞凋亡活性降低,治疗后恢复。我们确定了HILPDA+ccRCC1亚型,以缺氧为特征,作为易感细胞与更好的预后相关。HILPDA+ccRCC1亚型的主要共表达模块强调了在阴离子转运中的作用,对氧和PD-L1-PD-1途径的反应。此外,免疫抑制细胞可能通过HAVCR2-LGALS9,C3-C3AR1,HLA-A-CD8B和HLA-C-CD8A轴突与HILPDA+ccRCC1亚型相互作用,形成角化相关的TME景观.总之,我们预计这项研究将为ccRCC的治疗提供有价值的见解和潜在策略。
    Cuproptosis is a novel type to regulate cell death with copper-dependent manner, and has been reported to involve in the occurrence and development of various malignant tumors. However, the association between cuproptosis and the tumor microenvironment (TME) of clear cell renal cell carcinoma (ccRCC) remained unclear. To address this question, we integrated the single cell RNA sequencing (scRNA-seq) datasets of ccRCC across different stages, systematically examined the distinctive expression patterns of cuproptosis-related genes (CRGs) within the TME of ccRCC, and explored the crucial signatures using the spatial transcriptome sequencing (ST-seq) dataset. The cuproptosis activities reduced in cancer tissues along with the ccRCC development, and recovered after therapy. We identified HILPDA+ ccRCC1 subtype, characterized with hypoxia, as cuproptosis susceptible cells associated with a better prognosis. The main co-expression modules of HILPDA+ ccRCC1 subtype highlighted the role in anion transport, response to oxygen species and PD-L1-PD-1 pathway. Furthermore, the immunosuppressive cells might interact with HILPDA+ ccRCC1 subtype via HAVCR2-LGALS9, C3-C3AR1, HLA-A-CD8B and HLA-C-CD8A axises to shape the cuproptosis-related TME landscape. In summary, we anticipate that this study will offer valuable insights and potential strategies of cuproptosis for therapy of ccRCC.
    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
    不完全手术切除(SR)后的促肿瘤小生境可导致血管生成免疫抑制肿瘤微环境(TME)增强的局部进展和远处转移。在这里,中性粒细胞胞外诱捕网(NETs)和癌症相关神经递质(CANTs,例如,儿茶酚胺)首先被确定为两种主要诱因。Further,具有高组织粘附性的可注射纤维蛋白-藻酸盐水凝胶已被构建用于特异性共递送NETs抑制剂(DNaseI)包封的PLGA纳米颗粒和非选择性β-肾上腺素能受体阻滞剂(普萘洛尔).这两个组件(即,纤维蛋白和藻酸盐)可以对两种触发因素(凝血酶和Ca2+,分别)在术后出血凝胶化,成型为具有高强度的互穿网络(IPN)。DNaseI和PR的持续释放可以破坏NETs并拮抗儿茶酚胺以降低微血管密度,阻断骨髓来源的抑制细胞,分泌各种促炎细胞因子,增强自然杀伤细胞功能并阻止细胞毒性T细胞耗尽。重新编程的TME显著抑制局部残留和远处肿瘤,诱导强烈的免疫记忆效应,从而抑制肺转移。因此,通过这种基于原位IPN的水凝胶药物储库实现的靶向降解NETs和阻断CANTs提供了一种针对SR诱导的癌症复发和转移的简单有效的方法。
    Tumor-promoting niche after incomplete surgery resection (SR) can lead to more aggressive local progression and distant metastasis with augmented angiogenesis-immunosuppressive tumor microenvironment (TME). Herein, elevated neutrophil extracellular traps (NETs) and cancer-associated neurotransmitters (CANTs, e.g., catecholamines) are firstly identified as two of the dominant inducements. Further, an injectable fibrin-alginate hydrogel with high tissue adhesion has been constructed to specifically co-deliver NETs inhibitor (DNase I)-encapsulated PLGA nanoparticles and an unselective β-adrenergic receptor blocker (propranolol). The two components (i.e., fibrin and alginate) can respond to two triggers (thrombin and Ca2+, respectively) in postoperative bleeding to gelate, shaping into an interpenetrating network (IPN) featuring high strength. The continuous release of DNase I and PR can wreck NETs and antagonize catecholamines to decrease microvessel density, blockade myeloid-derived suppressor cells, secrete various proinflammatory cytokines, potentiate natural killer cell function and hamper cytotoxic T cell exhaustion. The reprogrammed TME significantly suppress locally residual and distant tumors, induce strong immune memory effects and thus inhibit lung metastasis. Thus, targetedly degrading NETs and blocking CANTs enabled by this in-situ IPN-based hydrogel drug depot provides a simple and efficient approach against SR-induced cancer recurrence and metastasis.
    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
    不仅刺激免疫循环而且调节免疫抑制肿瘤微环境的新策略对于有效的癌症免疫治疗至关重要。这里,一种新型的时空仿生“双子纳米免疫调节剂”被设计为通过整合扩增的免疫原性细胞死亡(ICD)的三重穿孔来激活强大的全身光免疫疗法,肿瘤相关巨噬细胞(TAMs)表型重编程和程序性细胞死亡配体1(PD-L1)降解。以生物相容性介孔聚多巴胺(mPDA)为纳米载体,将二甲双胍(Met)和Toll样受体7/8激动剂resiquimod(R848)通过包裹嵌入T7或M2肽的红细胞膜(RM)进行特异性生物识别,构建了“Gemini纳米免疫调节剂”PM@RM-T7和PR@RM-M2。mPDA/Met@RM-T7(缩写为PM@RM-T7)被构建以通过靶向PTT引起扩增的原位ICD效应并有效地刺激抗癌免疫。同时,剩余癌细胞上的PD-L1被爆发的二甲双胍降解以防止免疫逃避。随后,mPDA/R848@RM-M2(缩写为PR@RM-M2)特异性识别TAM,并将表型从M2状态重置为M1状态,从而破坏免疫抑制微环境并进一步增强细胞毒性T淋巴细胞的功能。这对姐妹纳米免疫调节剂共同协调了全面的抗癌活性,显著抑制原发和远处4T1肿瘤的生长,防止恶性转移。这项研究强调了使用多种纳米药物的时空合作模式,并为针对易转移肿瘤的有效癌症免疫治疗提供了新的范例。
    A novel strategy of not only stimulating the immune cycle but also modulating the immunosuppressive tumor microenvironment is of vital importance to efficient cancer immunotherapy. Here, a new type of spatiotemporal biomimetic \"Gemini nanoimmunoregulators\" was engineered to activate robust systemic photoimmunotherapy by integrating the triple-punch of amplified immunogenic cell death (ICD), tumor-associated macrophages (TAMs) phenotype reprogramming and programmed cell death ligand 1 (PD-L1) degradation. The \"Gemini nanoimmunoregulators\" PM@RM-T7 and PR@RM-M2 were constructed by taking the biocompatible mesoporous polydopamine (mPDA) as nanovectors to deliver metformin (Met) and toll-like receptor 7/8 agonist resiquimod (R848) to cancer cells and TAMs by specific biorecognition via wrapping of red blood cell membrane (RM) inlaid with T7 or M2 peptides. mPDA/Met@RM-T7 (abbreviated as PM@RM-T7) was constructed to elicit an amplified in situ ICD effect through the targeted PTT and effectively stimulated the anticancer immunity. Meanwhile, PD-L1 on the remaining cancer cells was degraded by the burst metformin to prevent immune evasion. Subsequently, mPDA/R848@RM-M2 (abbreviated as PR@RM-M2) specifically recognized TAMs and reset the phenotype from M2 to M1 state, thus disrupting the immunosuppressive microenvironment and further boosting the function of cytotoxic T lymphocytes. This pair of sister nanoimmunoregulators cooperatively orchestrated the comprehensive anticancer activity, which remarkably inhibited the growth of primary and distant 4T1 tumors and prevented malignant metastasis. This study highlights the spatiotemporal cooperative modalities using multiple nanomedicines and provides a new paradigm for efficient cancer immunotherapy against metastatic-prone tumors.
    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: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: Journal Article
    子宫内膜癌(EC)是影响妇女的三种最常见的妇科肿瘤之一,并且是发达国家中最常见的妇科恶性肿瘤。它的发病率在世界范围内迅速增加,主要影响绝经后妇女,而最近它在年轻人中的患病率有所增加。EC是一种免疫基因疾病,许多研究表明肿瘤免疫抑制微环境在癌症进展中起重要作用。近年来,关于EC的免疫抑制肿瘤微环境(ITME)的发现包括免疫逃避机制和免疫疗法,主要是EC的免疫检查点抑制剂(ICI)。最近对不同分子类型EC的ITME的研究发现,不同分子类型可能具有不同的ITME。随着对EC免疫微环境的研究,近年来,基于免疫微环境的免疫表型分类已经开始进行。然而,ITME对EC的影响尚不清楚,EC的免疫表型仍局限于研究阶段。我们的综述描述了有关不同EC分子类型的ITME特征的最新发现。免疫治疗的出现为晚期或复发性EC患者带来了改善疗效和预后的希望。ICIs联合治疗的有效性和安全性仍是未来研究的重点。
    Endometrial cancer (EC) is one of the three most prevalent gynecological tumors affecting women and is the most prevalent gynecological malignancy in the developed world. Its incidence is rapidly increasing worldwide, mostly affecting postmenopausal women, whereas recently its prevalence has increased in younger people. EC is an immune gene disease and many studies have shown that the tumor-immunosuppressive microenvironment plays an important role in cancer progression. In recent years, findings regarding the immunosuppressive tumor microenvironment (ITME) of EC have included immune evasion mechanisms and immunotherapy, which are mostly immune checkpoint inhibitors (ICI) for EC. Recently studies on the ITME of different molecular types of EC have found that different molecular types may have different ITME. With the research on the immune microenvironment of EC, a new immunophenotype classification based on the immune microenvironment has been carried out in recent years. However, the impact of the ITME on EC remains unclear, and the immunophenotype of EC remains limited to the research stage. Our review describes recent findings regarding the ITME features of different EC molecular types. The advent of immunotherapy has brought hope for improved efficacy and prognosis in patients with advanced or recurrent EC. The efficacy and safety of ICIs combination therapy remains the focus of future research.
    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
    胰腺导管腺癌(PDAC),全世界最致命的恶性肿瘤之一,是包含许多成纤维细胞和免疫抑制细胞的肿瘤微环境(TME)的特征。PDAC的常规疗法通常受到有限的药物递送效率的限制。免疫抑制TME,和不利影响。因此,PDAC治疗迫切需要有效和安全的治疗方法。近年来,水凝胶,具有优异的生物相容性,高药物负荷能力,和可持续的发布概况,已被开发为有效的药物递送系统,为PDAC提供潜在的治疗选择。这篇综述总结了PDAC免疫抑制TME的独特特征,并讨论了基于水凝胶的疗法在PDAC中的应用。重点关注这些水凝胶如何重塑TME并以受控方式输送不同类型的货物。此外,我们还讨论了潜在的候选药物以及基于水凝胶的PDAC疗法的挑战和前景。通过全面概述基于水凝胶的PDAC治疗方法,这篇综述旨在为参与制定针对PDAC微环境的治疗策略的研究人员和临床医生提供参考.
    Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies worldwide, is characteristic of the tumor microenvironments (TME) comprising numerous fibroblasts and immunosuppressive cells. Conventional therapies for PDAC are often restricted by limited drug delivery efficiency, immunosuppressive TME, and adverse effects. Thus, effective and safe therapeutics are urgently required for PDAC treatment. In recent years, hydrogels, with their excellent biocompatibility, high drug load capacity, and sustainable release profiles, have been developed as effective drug-delivery systems, offering potential therapeutic options for PDAC. This review summarizes the distinctive features of the immunosuppressive TME of PDAC and discusses the application of hydrogel-based therapies in PDAC, with a focus on how these hydrogels remodel the TME and deliver different types of cargoes in a controlled manner. Furthermore, we also discuss potential drug candidates and the challenges and prospects for hydrogel-based therapeutics for PDAC. By providing a comprehensive overview of hydrogel-based therapeutics for PDAC treatment, this review seeks to serve as a reference for researchers and clinicians involved in developing therapeutic strategies targeting the PDAC microenvironment.
    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
    免疫检查点抑制剂(ICI)的引入彻底改变了转移性肾细胞癌(mRCC)的治疗方法,并显着改善了患者的预后。使用靶向PD-1/PD-L1和CTLA-4的ICIs的单一疗法或组合,以及与酪氨酸激酶抑制剂一起添加ICIs,显著提高了mRCC患者的总体生存率。尽管这些有希望的结果,仍有一部分患者对治疗无反应(原发性耐药)或随着时间的推移对治疗产生耐药(获得性耐药).了解ICI治疗耐药发展的潜在机制对mRCC的管理至关重要。因为它们可用于确定创新治疗策略的新目标。目前,目前尚未满足开发新的预测和预后生物标志物的需求,这些生物标志物可以帮助mRCC患者制定个性化治疗方案.在这次审查中,我们总结了RCC中ICI抗性的几种机制,包括肿瘤微环境的改变,替代免疫检查点途径的上调,遗传和表观遗传变化。此外,我们强调可以用来克服阻力的潜在策略,如联合治疗,靶向治疗,和免疫调节。
    The introduction of immune checkpoint inhibitor (ICI) has revolutionized the treatment of metastatic renal cell carcinoma (mRCC) and has dramatically improved the outcomes of patients. The use of monotherapy or combinations of ICIs targeting PD-1/PD-L1 and CTLA-4, as well as the addition of ICIs with tyrosine kinase inhibitors, has significantly enhanced the overall survival of mRCC patients. Despite these promising results, there remains a subset of patients who either do not respond to treatment (primary resistance) or develop resistance to therapy over time (acquired resistance). Understanding the mechanisms underlying the development of resistance to ICI treatment is crucial in the management of mRCC, as they can be used to identify new targets for innovative therapeutic strategies. Currently, there is an unmet need to develop new predictive and prognostic biomarkers that can aid in the development of personalized treatment options for mRCC patients. In this review, we summarize several mechanisms of ICI resistance in RCC, including alterations in tumor microenvironment, upregulation of alternative immune checkpoint pathways, and genetic and epigenetic changes. Additionally, we highlight potential strategies that can be used to overcome resistance, such as combination therapy, targeted therapy, and immune modulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号