Immunosuppressive tumor microenvironment

免疫抑制肿瘤微环境
  • 文章类型: Journal Article
    高度免疫抑制的肿瘤微环境(TME)限制了免疫应答的有效激活。为了恢复对免疫系统的监视以进行强大的激活,致力于使TME正常化的巨大努力。这里,开发了一种锰掺杂的层状双氢氧化物(Mn-LDH),用于通过逆转TME进行有效的抗肿瘤免疫。通过一步水热法合成Mn-LDH。除了LDH固有的质子中和能力外,锰氧化物的引入赋予LDH额外的产生氧的能力。Mn-LDH在暴露于具有高水平H+和H2O2的TME时有效释放Mn2+和Mg2+,分别激活干扰素基因途径的合酶刺激因子并维持CD8+T细胞的细胞毒性。在先天和适应性免疫中实现级联样作用。局部施用的Mn-LDH促进了由成熟树突状细胞组成的“热”网络,M1表型巨噬细胞,以及细胞毒性和辅助性T细胞,显着抑制原发性和远端肿瘤的生长。此外,Mn-LDH的光热转化能力在单次给药和照射的大型肿瘤模型中激发了更强大的治疗效果。总的来说,本研究指导合理设计TME调节免疫疗法以实现稳健的免疫激活,为下一代癌症免疫治疗提供临床候选药物。
    The highly immunosuppressive tumor microenvironment (TME) restricts the efficient activation of immune responses. To restore the surveillance of the immune system for robust activation, vast efforts are devoted to normalizing the TME. Here, a manganese-doped layered double hydroxide (Mn-LDH) is developed for potent anti-tumor immunity by reversing TME. Mn-LDH is synthesized via a one-step hydrothermal method. In addition to the inherent proton neutralization capacity of LDH, the introduction of manganese oxide endows LDH with an additional ability to produce oxygen. Mn-LDH effectively releases Mn2+ and Mg2+ upon exposure to TME with high levels of H+ and H2O2, which activates synthase-stimulator of interferon genes pathway and maintains the cytotoxicity of CD8+ T cells respectively, achieving a cascade-like role in innate and adaptive immunity. The locally administered Mn-LDH facilitated a \"hot\" network consisting of mature dendritic cells, M1-phenotype macrophages, as well as cytotoxic and helper T cells, significantly inhibiting the growth of primary and distal tumors. Moreover, the photothermal conversion capacity of Mn-LDH sparks more robust therapeutic effects in large established tumor models with a single administration and irradiation. Overall, this study guides the rational design of TME-modulating immunotherapeutics for robust immune activation, providing a clinical candidate for next-generation 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
    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
    肿瘤微环境(TME)的内在致癌机制和特性已得到广泛研究。TME的主要特征包括代谢重编程,缺氧,慢性炎症,和肿瘤免疫抑制。先前的研究表明,介导细胞间信息交换的衰老相关分泌表型在TME的动态进化中起作用。具体来说,低氧适应,代谢失调,和细胞衰老调节的免疫细胞的表型变化协同促进免疫抑制微环境和慢性炎症的发展,从而促进肿瘤事件的进展。这篇综述提供了细胞衰老调节肿瘤适应TME动态进化的过程的全面总结。重点研究衰老与肿瘤细胞生物学功能变化之间关系的复杂机制。现有的发现表明,TME的成分共同促进了肿瘤事件的进展。在推进细胞衰老相关研究的背景下,进一步讨论了基于靶向细胞衰老和联合治疗在临床环境中的潜在应用和挑战。
    The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.
    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
    卵巢癌的低免疫原性和免疫抑制微环境严重限制了免疫介导的肿瘤杀伤能力。免疫原性细胞死亡(ICD)通过增加抗原暴露和呈递引入了抗肿瘤免疫的理论原理。尽管最近的研究进展,目前可用的ICD诱导剂仍然非常有限,基于传统的内质网(ER)应激,其中许多很难诱导足够的ICD。越来越多的证据表明,诱导线粒体应激通常在诱发大规模ICD方面比通过内质网应激表现出更高的效率。受此启发,在这里,线粒体靶向的多前药纳米颗粒(名为Mito-CMPN)作为一个非常优越的ICD诱导物,有效诱导化学光动力疗法引起的肿瘤细胞线粒体应激。合理设计的刺激反应性聚前药,可以自组装成纳米粒子,用罗丹明B功能化用于线粒体靶向,顺铂和米托蒽醌(MTO)用于协同化学免疫疗法,和MTO还用作光动力免疫疗法的光敏剂。在卵巢癌皮下模型和高级别浆液性卵巢癌模型中都验证了Mito-CMPN逆转免疫抑制微环境的有效性和鲁棒性。我们的结果支持,通过集中的线粒体应激诱导丰富的ICD是提高免疫抑制性卵巢癌治疗效果的高度有效策略。
    Hypoimmunogenicity and the immunosuppressive microenvironment of ovarian cancer severely restrict the capability of immune-mediated tumor killing. Immunogenic cell death (ICD) introduces a theoretical principle for antitumor immunity by increasing antigen exposure and presentation. Despite recent research progress, the currently available ICD inducers are still very limited, and many of them can hardly induce sufficient ICD based on traditional endoplasmic reticulum (ER) stress. Accumulating evidence indicates that inducing mitochondrial stress usually shows a higher efficiency in evoking large-scale ICD than that via ER stress. Inspired by this, herein, a mitochondria-targeted polyprodrug nanoparticle (named Mito-CMPN) serves as a much superior ICD inducer, effectively inducing chemo-photodynamic therapy-caused mitochondrial stress in tumor cells. The rationally designed stimuli-responsive polyprodrugs, which can self-assemble into nanoparticles, were functionalized with rhodamine B for mitochondrial targeting, cisplatin and mitoxantrone (MTO) for synergistic chemo-immunotherapy, and MTO also serves as a photosensitizer for photodynamic immunotherapy. The effectiveness and robustness of Mito-CMPNs in reversing the immunosuppressive microenvironment is verified in both an ovarian cancer subcutaneous model and a high-grade serous ovarian cancer model. Our results support that the induction of abundant ICD by focused mitochondrial stress is a highly effective strategy to improve the therapeutic efficacy of immunosuppressive ovarian 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
    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
    溶瘤病毒(OVs)代表了一种新兴的免疫治疗策略,因为它们具有直接溶解肿瘤和诱导抗肿瘤免疫的能力。然而,短暂的持久性和适度的功效等障碍需要创新的方法。代谢重塑最近作为一种战略干预措施获得了突出的地位,其中OV或组合方案可以重新编程肿瘤和免疫细胞代谢以增强病毒复制和溶瘤作用。在这次审查中,我们总结了肿瘤和免疫细胞代谢的战略性重编程以增强基于OV的免疫疗法的最新进展。具体的策略包括工程病毒靶向糖酵解,谷氨酰胺分解,和癌细胞中的核苷酸合成途径,促进病毒复制和肿瘤细胞死亡。此外,重新连接T细胞和NK细胞的脂质代谢,氨基酸,和碳水化合物显示出增强抗肿瘤作用的希望。讨论了进一步的见解,为代谢增强的溶瘤平台的临床实施铺平道路。包括平衡代谢调节,以限制抗病毒反应,同时促进病毒的持久性和肿瘤清除。
    Oncolytic viruses (OVs) represent an emerging immunotherapeutic strategy owing to their capacity for direct tumor lysis and induction of antitumor immunity. However, hurdles like transient persistence and moderate efficacy necessitate innovative approaches. Metabolic remodeling has recently gained prominence as a strategic intervention, wherein OVs or combination regimens could reprogram tumor and immune cell metabolism to enhance viral replication and oncolysis. In this review, we summarize recent advances in strategic reprogramming of tumor and immune cell metabolism to enhance OV-based immunotherapies. Specific tactics include engineering viruses to target glycolytic, glutaminolytic, and nucleotide synthesis pathways in cancer cells, boosting viral replication and tumor cell death. Additionally, rewiring T cell and NK cell metabolism of lipids, amino acids, and carbohydrates shows promise to enhance antitumor effects. Further insights are discussed to pave the way for the clinical implementation of metabolically enhanced oncolytic platforms, including balancing metabolic modulation to limit antiviral responses while promoting viral persistence and tumor clearance.
    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:肺腺癌(LUAD)是最常见的肺癌组织学类型,生存率较低。靶向治疗和免疫检查点免疫疗法的最新进展取得了显著的成功。仍有很大比例的LUAD缺乏可用的治疗选择.由于肿瘤的异质性,LUAD的诊断和治疗具有挑战性。探索LUAD的生物学并确定新的生物标志物和治疗靶标选择至关重要。
    方法:我们对6个配对的初级和邻近LUAD组织进行了单细胞RNA测序(scRNA-seq),和scRNA-seq的整合组学分析,大量RNA-seq和全外显子组测序数据揭示了分子亚型特征。我们的实验结果证实CDC25C基因可以作为LUAD不良预后的潜在标志物。
    结果:我们通过scRNA-seq数据研究了LUAD中不同细胞类型中的异常基因表达。此外,多组聚类揭示了由转录谱和分子亚型4(MS4)定义的四个亚组,具有较差的生存概率,免疫细胞浸润特征显示MS4倾向于免疫抑制亚型。我们的研究表明,CDC25C基因可能是一个独特的预后生物标志物,表明LUAD患者的免疫浸润水平和对免疫疗法的反应。我们的实验结果得出结论,CDC25C表达影响肺癌细胞的侵袭和迁移,可能在调节上皮-间充质转化(EMT)途径中起关键作用。
    结论:我们的多组学结果揭示了在细胞和组织水平上与LUAD预后相关基因相关的一组全面的分子属性。免疫抑制性TME亚型的鉴定和LUAD的预后特征。我们发现细胞周期调控基因CDC25C影响肺癌细胞的侵袭和迁移,可作为LUAD的潜在生物标志物。
    BACKGROUND: Lung adenocarcinoma (LUAD) is the most common histological type of lung cancer with lower survival rates. Recent advancements in targeted therapies and immunotherapies targeting immune checkpoints have achieved remarkable success, there is still a large percentage of LUAD that lacks available therapeutic options. Due to tumor heterogeneity, the diagnosis and treatment of LUAD are challenging. Exploring the biology of LUAD and identifying new biomarker and therapeutic targets options are essential.
    METHODS: We performed single-cell RNA sequencing (scRNA-seq) of 6 paired primary and adjacent LUAD tissues, and integrative omics analysis of the scRNA-seq, bulk RNA-seq and whole-exome sequencing data revealed molecular subtype characteristics. Our experimental results confirm that CDC25C gene can serve as a potential marker for poor prognosis in LUAD.
    RESULTS: We investigated aberrant gene expression in diverse cell types in LUAD via the scRNA-seq data. Moreover, multi-omics clustering revealed four subgroups defined by transcriptional profile and molecular subtype 4 (MS4) with poor survival probability, and immune cell infiltration signatures revealed that MS4 tended to be the immunosuppressive subtype. Our study revealed that the CDC25C gene can be a distinct prognostic biomarker that indicates immune infiltration levels and response to immunotherapy in LUAD patients. Our experimental results concluded that CDC25C expression affects lung cancer cell invasion and migration, might play a key role in regulating Epithelial-Mesenchymal Transition (EMT) pathways.
    CONCLUSIONS: Our multi-omics result revealed a comprehensive set of molecular attributes associated with prognosis-related genes in LUAD at the cellular and tissue level. Identification of a subtype of immunosuppressive TME and prognostic signature for LUAD. We identified the cell cycle regulation gene CDC25C affects lung cancer cell invasion and migration, which can be used as a potential biomarker for LUAD.
    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
    近年来,癌症免疫疗法已成为一种有前途的临床治疗策略。不幸的是,复杂的免疫抑制肿瘤微环境(ITM)限制了免疫治疗的令人满意的抗肿瘤疗效。为了重塑ITM并减轻免疫逃避,我们构建了FA-PEG修饰的脂质体来递送质粒IL-15(pIL-15)和吉西他滨(GEM)(FPCL@pIL-15+FPGL),分别。取决于叶酸(FA)专门的靶向功能,FPCL@pIL-15(150nm)和FPGL(120nm)表现出对称的球形结构以及在肿瘤组织上的期望的渗透和积累。IL-15的转染表达通过与IL-15R结合有效促进自然杀伤(NK)细胞和CD8+T细胞的增殖和共活化。FPGL上调自然杀伤组2成员D配体(NKG2DLs)的表达,增强NK细胞的识别,以减轻免疫逃避,同时通过免疫原性细胞死亡(ICD)作用促进CD8+T细胞的活化。更重要的是,联合给药在皮下4T1肿瘤模型中实现了预期的抗肿瘤功效。实质上,我们证明了FPCL@pIL-15与FPGL的结合协同刺激和动员免疫系统以逆转ITM并触发抗肿瘤免疫反应,表明在免疫疗法中应用的巨大潜力。
    Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in 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
    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)

公众号