cancer metalloimmunotherapy

  • 文章类型: Journal Article
    原理:手术切除是实体瘤的主要治疗方法,但手术后肿瘤的高复发率和转移率提出了重大挑战。锰(Mn2+),已知通过激活cGAS-STING途径增强树突状细胞介导的癌症免疫治疗,在术后癌症管理方面具有潜力。然而,实现Mn2+的延长和局部递送以刺激免疫应答而没有全身毒性仍然是一个挑战。方法:我们开发了一种嵌入Mn2-果胶微球(MnP@DOP-Gel)的术后微环境响应型石斛多糖水凝胶。此水凝胶系统响应于ROS释放Mn2-果胶微球(MnP),和MnP在体外显示出双重作用:促进免疫原性细胞死亡和激活免疫细胞(树突状细胞和巨噬细胞)。在小鼠皮下和转移性黑色素瘤模型中评估MnP@DOP-Gel作为术后治疗的功效及其免疫激活的潜力,探讨其与抗PD1抗体的协同作用。结果:MnP@DOP-Gel表现出ROS响应性释放MnP,它可以通过诱导肿瘤细胞的免疫原性细胞死亡和激活树突状细胞和巨噬细胞来启动抗肿瘤免疫应答的级联反应来发挥双重作用。体内实验表明,植入的MnP@DOP-Gel可显着抑制残留肿瘤的生长和转移。此外,MnP@DOP-Gel和抗PD1抗体的组合在预防转移或外翻脑肿瘤生长方面显示出优异的治疗效力。结论:MnP@DOP-Gel代表了一种有希望的癌症术后无药治疗策略。利用这种Mn2+嵌入和ROS响应传输系统,它调节手术诱导的免疫反应,促进持续的抗肿瘤反应,有可能提高癌症手术治疗的有效性。
    Rationale: Surgical resection is a primary treatment for solid tumors, but high rates of tumor recurrence and metastasis post-surgery present significant challenges. Manganese (Mn2+), known to enhance dendritic cell-mediated cancer immunotherapy by activating the cGAS-STING pathway, has potential in post-operative cancer management. However, achieving prolonged and localized delivery of Mn2+ to stimulate immune responses without systemic toxicity remains a challenge. Methods: We developed a post-operative microenvironment-responsive dendrobium polysaccharide hydrogel embedded with Mn2+-pectin microspheres (MnP@DOP-Gel). This hydrogel system releases Mn2+-pectin microspheres (MnP) in response to ROS, and MnP shows a dual effect in vitro: promoting immunogenic cell death and activating immune cells (dendritic cells and macrophages). The efficacy of MnP@DOP-Gel as a post-surgical treatment and its potential for immune activation were assessed in both subcutaneous and metastatic melanoma models in mice, exploring its synergistic effect with anti-PD1 antibody. Result: MnP@DOP-Gel exhibited ROS-responsive release of MnP, which could exert dual effects by inducing immunogenic cell death of tumor cells and activating dendritic cells and macrophages to initiate a cascade of anti-tumor immune responses. In vivo experiments showed that the implanted MnP@DOP-Gel significantly inhibited residual tumor growth and metastasis. Moreover, the combination of MnP@DOP-Gel and anti-PD1 antibody displayed superior therapeutic potency in preventing either metastasis or abscopal brain tumor growth. Conclusions: MnP@DOP-Gel represents a promising drug-free strategy for cancer post-operative management. Utilizing this Mn2+-embedding and ROS-responsive delivery system, it regulates surgery-induced immune responses and promotes sustained anti-tumor responses, potentially increasing the effectiveness of surgical cancer treatments.
    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
    肿瘤缺氧,高氧化应激,低免疫原性创造了一个根深蒂固的免疫抑制微环境,对肿瘤免疫治疗实体瘤的疗效提出了重大挑战。在这里,能够缓解缺氧和免疫刺激的对肿瘤微环境(TME)敏感的智能纳米平台已被设计用于有效的实体瘤免疫治疗.MnO2@OxA@OMV纳米反应器,包裹细菌来源的外膜囊泡(OMV)包裹的MnO2纳米酶和免疫原性细胞死亡诱导剂奥沙利铂(OxA),在TME内表现出固有的过氧化氢酶样活性,有效地将内源性H2O2催化成O2,从而延长氧气供应,从而减轻肿瘤的氧化应激和缺氧TME,加快OxA的释放。OxA引起的ICD效应和来自纳米反应器的Mn2+的组合作用使cGAS-STING途径的动机能够显着提高STING的激活和树突状细胞(DC)成熟,导致金属免疫疗法。此外,免疫刺激剂OMV在促进活化的CD8+T细胞浸润到实体瘤中起着至关重要的作用。总的来说,纳米反应器为实体瘤治疗提供了一个强大的平台,强调了将缓解肿瘤缺氧和免疫刺激相结合用于金属免疫疗法的巨大潜力。重要声明:通过将细菌来源的外膜囊泡(OMV)封闭在MnO2纳米酶上并加载免疫原性细胞死亡诱导剂奥沙利铂(OxA)用于肿瘤金属免疫疗法,制造了量身定制的纳米反应器。纳米反应器在肿瘤微环境中具有固有的过氧化氢酶样活性,通过催化内源性H2O2转化为O2,从而有效地延长了氧气供应,从而减轻了肿瘤缺氧并加快了OxA的释放。此外,营养Mn2+的TME响应释放使cGAS-STING途径敏感,并与OxA诱导的免疫原性细胞死亡(ICD)协作。与免疫刺激性OMV结合增强DC对纳米反应器的摄取并促进活化的CD8+T细胞的浸润。这种纳米反应器为实体瘤治疗提供了一个强大的平台,强调了将缓解肿瘤缺氧和免疫刺激相结合用于金属免疫疗法的巨大潜力。
    Tumor hypoxia, high oxidative stress, and low immunogenic create a deep-rooted immunosuppressive microenvironment, posing a major challenge to the therapeutic efficiency of cancer immunotherapy for solid tumor. Herein, an intelligent nanoplatform responsive to the tumor microenvironment (TME) capable of hypoxia relief and immune stimulation has been engineered for efficient solid tumor immunotherapy. The MnO2@OxA@OMV nanoreactor, enclosing bacterial-derived outer membrane vesicles (OMVs)-wrapped MnO2 nanoenzyme and the immunogenic cell death inducer oxaliplatin (OxA), demonstrated intrinsic catalase-like activity within the TME, which effectively catalyzed the endogenous H2O2 into O2 to enable a prolonged oxygen supply, thereby alleviating the tumor\'s oxidative stress and hypoxic TME, and expediting OxA release. The combinational action of OxA-caused ICD effect and Mn2+ from nanoreactor enabled the motivation of the cGAS-STING pathway to significantly improve the activation of STING and dendritic cells (DCs) maturation, resulting in metalloimmunotherapy. Furthermore, the immunostimulant OMVs played a crucial role in promoting the infiltration of activated CD8+T cells into the solid tumor. Overall, the nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy. STATEMENT OF SIGNIFICANCE: A tailor-made nanoreactor was fabricated by enclosing bacterial-derived outer membrane vesicles (OMVs) onto MnO2 nanoenzyme and loading with immunogenic cell death inducer oxaliplatin (OxA) for tumor metalloimmunotherapy. The nanoreactor possesses intrinsic catalase-like activity within the tumor microenvironment, which effectively enabled a prolonged oxygen supply by catalyzing the conversion of endogenous H2O2 into O2, thereby alleviating tumor hypoxia and expediting OxA release. Furthermore, the TME-responsive release of nutritional Mn2+ sensitized the cGAS-STING pathway and collaborated with OxA-induced immunogenic cell death (ICD). Combing with immunostimulatory OMVs enhances the uptake of nanoreactors by DCs and promotes the infiltration of activated CD8+T cells. This nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy.
    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
    癌症治疗性疫苗是免疫系统激活和引发针对肿瘤的保护性应答的有力工具。然而,它们的功效经常受到弱和慢的免疫反应的阻碍。这里,作者介绍了一种采用衰老红细胞的免疫策略,通过暂时饱和肝巨噬细胞,促进免疫调节锌-明矾/卵白蛋白(ZAlum/OVA)纳米疫苗在脾脏和实体瘤中的积累.这种方法通过级联免疫激活为加强癌症金属免疫疗法奠定了基础。ZAlum/OVA纳米疫苗在脾脏中的积累基本上增强了树突状细胞中自噬依赖性抗原呈递,快速启动针对实体瘤的OVA特异性T细胞反应。同时,在肿瘤微环境中积累的ZAlum/OVA纳米疫苗引发免疫原性细胞死亡,导致诱导个体化的肿瘤相关抗原特异性T细胞反应和增加的T细胞浸润。这种使用ZAlum/OVA纳米疫苗的红细胞辅助级联免疫激活导致快速和强大的抗肿瘤免疫诱导。临床癌症金属免疫疗法具有巨大潜力。
    Cancer therapeutic vaccines are powerful tools for immune system activation and eliciting protective responses against tumors. However, their efficacy has often been hindered by weak and slow immune responses. Here, the authors introduce an immunization strategy employing senescent erythrocytes to facilitate the accumulation of immunomodulatory zinc-Alum/ovalbumin (ZAlum/OVA) nanovaccines within both the spleen and solid tumors by temporarily saturating liver macrophages. This approach sets the stage for boosted cancer metalloimmunotherapy through a cascade immune activation. The accumulation of ZAlum/OVA nanovaccines in the spleen substantially enhances autophagy-dependent antigen presentation in dendritic cells, rapidly initiating OVA-specific T-cell responses against solid tumors. Concurrently, ZAlum/OVA nanovaccines accumulated in the tumor microenvironment trigger immunogenic cell death, leading to the induction of individualized tumor-associated antigen-specific T cell responses and increased T cell infiltration. This erythrocyte-assisted cascade immune activation using ZAlum/OVA nanovaccines results in rapid and robust antitumor immunity induction, holding great potential for clinical cancer metalloimmunotherapy.
    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
    金属有机框架(MOF)由于其结构和功能的多功能性而显示出巨大的药物递送前景。然而,在大多数情况下,MOF通常用作生物惰性载体。内在免疫刺激MOF的创建仍然具有挑战性。在这项研究中,提出了一种简便,绿色的合成方法,用于制备用于癌症金属免疫疗法的基于锰离子(Mn2)的免疫刺激MOF(ISAMn-MOF)。ISAMn-MOF显著促进骨髓来源的树突状细胞(BMDC)中干扰素基因(cGAS-STING)相关基因和信号通路的环GMP-AMP合酶-刺激物的激活。用ISAMn-MOF处理的BMDC分泌的I型干扰素和促炎细胞因子比用同等MnCl2处理的BMDC高4倍。ISAMn-MOF单独或其与免疫检查点抗体的组合显著抑制肿瘤生长和转移并延长小鼠存活。机制研究表明,ISAMn-MOF治疗促进了肿瘤和淋巴器官中刺激性免疫细胞的浸润。这项研究为改善癌症金属免疫疗法的生物活性MOFs设计提供了见解。
    Metal-organic frameworks (MOFs) show tremendous promise for drug delivery due to their structural and functional versatility. However, MOFs are usually used as biologically inert carriers in most cases. The creation of intrinsically immunostimulatory MOFs remains challenging. In this study, a facile and green synthesis method is proposed for the preparation of a manganese ion (Mn2+)-based immunostimulatory MOF (ISAMn-MOF) for cancer metalloimmunotherapy. ISAMn-MOF significantly facilitates the activation of cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) related genes and signaling pathways in bone-marrow-derived dendritic cells (BMDCs). BMDCs treated with ISAMn-MOF secrete 4-fold higher type I interferon and 2- to 16-fold higher proinflammatory cytokines than those treated with equivalent MnCl2. ISAMn-MOF alone or its combination with immune checkpoint antibodies significantly suppresses tumor growth and metastasis and prolongs mouse survival. Mechanistic studies indicate that ISAMn-MOF treatment facilitates the infiltration of stimulatory immune cells in tumors and lymphoid organs. This study provides insight into the design of bioactive MOFs for improved cancer metalloimmunotherapy.
    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