STING activation

STING 激活
  • 文章类型: Journal Article
    干扰素基因刺激因子(STING)激动剂通过刺激先天免疫反应在癌症治疗中显示出希望,然而,它们的临床潜力受到低效的细胞溶质进入和不令人满意的药理活性的限制。此外,具有“冷”和免疫抑制微环境的侵袭性肿瘤可能无法仅通过先天免疫疗法得到有效抑制。在这里,我们提出了一种多面性免疫刺激纳米颗粒(Mn-MCNP),其将锰II(Mn2+)配位的光敏剂(二氢卟啉e6,Ce6)和STING激动剂(MSA-2)整合在聚乙二醇化的纳米结构内。在Mn-MCNP中,Ce6发挥强大的光疗作用,促进肿瘤消融和诱导免疫原性细胞死亡,以引发强大的适应性抗肿瘤免疫。MSA-2激活由Mn2+驱动的STING途径,从而促进先天的抗肿瘤免疫。Mn-MCNP具有高载药量(63.42%),可直接消融肿瘤组织,同时协同增强适应性和先天免疫反应。在皮下肿瘤小鼠模型中,Mn-MCNP不仅在根除原发性肿瘤方面表现出显著的功效,而且通过协同免疫疗法阻止远端和转移性肿瘤的进展。此外,它们通过促进长期免疫记忆有助于预防肿瘤复发。我们的多面免疫刺激纳米颗粒具有克服与抗肿瘤免疫力不足和无效癌症治疗相关的局限性的巨大潜力。
    Stimulator of interferon genes (STING) agonists have shown promise in cancer treatment by stimulating the innate immune response, yet their clinical potential has been limited by inefficient cytosolic entry and unsatisfactory pharmacological activities. Moreover, aggressive tumors with \"cold\" and immunosuppressive microenvironments may not be effectively suppressed solely through innate immunotherapy. Herein, we propose a multifaceted immunostimulating nanoparticle (Mn-MC NP), which integrates manganese II (Mn2+) coordinated photosensitizers (chlorin e6, Ce6) and STING agonists (MSA-2) within a PEGylated nanostructure. In Mn-MC NPs, Ce6 exerts potent phototherapeutic effects, facilitating tumor ablation and inducing immunogenic cell death to elicit robust adaptive antitumor immunity. MSA-2 activates the STING pathway powered by Mn2+, thereby promoting innate antitumor immunity. The Mn-MC NPs feature a high drug-loading capacity (63.42 %) and directly ablate tumor tissue while synergistically boosting both adaptive and innate immune responses. In subsutaneous tumor mouse models, the Mn-MC NPs exhibit remarkable efficacy in not only eradicating primary tumors but also impeding the progression of distal and metastatic tumors through synergistic immunotherapy. Additionally, they contribute to preventing tumor recurrence by fostering long-term immunological memory. Our multifaceted immunostimulating nanoparticle holds significant potential for overcoming limitations associated with insufficient antitumor immunity and ineffective cancer treatment.
    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
    免疫抑制性肿瘤相关巨噬细胞(TAM)占肿瘤组织的高比例,并显着阻碍免疫功效。此外,在TAMs中表达的信号调节蛋白α(SIRPα)与巨噬细胞活化和吞噬作用,导致免疫监视逃逸。为了解决这些困难,甘露糖修饰的,具有瑞喹莫德(R848)和2'的pH响应型纳米平台,3'-环GMP-AMP(cGAMP)共封装(称为M-PNP@R@C)旨在使TAM极化并降低SIRPα表达。R848和cGAMP的共同递送协同促进TAM从抗炎M2表型极化为促炎M1表型,从而增强抗肿瘤免疫疗法。值得注意的是,TAMs中cGAMP介导的干扰素基因刺激因子(STING)的激活显着下调SIRPα的表达,它与分化簇47(CD47)抗体协同作用,以双重阻断CD47-SIRPα轴。单细胞RNA测序的进一步分析表明,STING激活通过调节细胞内脂肪酸氧化代谢来下调SIRPα。体内研究表明,在黑色素瘤移植肿瘤模型中,M-PNP@R@C显着抑制肿瘤生长,具有有效的抗肿瘤免疫反应。在与抗CD47的协同作用之后,SIRPα/CD47轴的双重阻断策略导致肺转移的显著抑制。在用CD47和程序性死亡配体-1抗体组合治疗后观察到用于三重免疫检查点阻断的延长的存活率。总之,我们的研究提供了对STING途径在基于巨噬细胞的免疫疗法中的潜在作用的原始见解,从而为癌症治疗提供了一种潜在的组合策略。
    Immunosuppressive tumor-associated macrophages (TAMs) account for a high proportion of the tumor tissue and significantly impede immunoefficacy. Furthermore, the signal regulatory protein α (SIRPα) expressed in TAMs adversely correlates with macrophage activation and phagocytosis, resulting in immunosurveillance escape. To address these difficulties, a mannose-modified, pH-responsive nanoplatform with resiquimod (R848) and 2\', 3\'-cyclic GMP-AMP (cGAMP) co-encapsulation (named M-PNP@R@C) is designed to polarize TAMs and lower SIRPα expression. The co-delivery of R848 and cGAMP synergistically facilitates the polarization of TAMs from the anti-inflammatory M2 phenotype into the pro-inflammatory M1 phenotype, thereby enhancing antitumor immunotherapy. Remarkably, activation of the cGAMP-mediated stimulator of interferon genes (STING) in TAMs significantly downregulates the expression of SIRPα, which synergizes with the cluster of differentiation 47 (CD47) antibody for the dual blockade of the CD47-SIRPα axis. Further analysis of single-cell RNA sequencing indicates that STING activation downregulates SIRPα by regulating intracellular fatty acid oxidation metabolism. In vivo studies indicate that M-PNP@R@C significantly inhibits tumor growth with a potent antitumor immune response in melanoma graft tumor models. After synergy with anti-CD47, the double blockade strategies of the SIRPα/CD47 axis result in a notable inhibition of lung metastasis. A prolonged survival rate is observed after combination treatment with CD47 and programmed death ligand-1 antibodies for the triple immune checkpoint blockade. In summary, our study provides original insights into the potential role of the STING pathway in macrophage-based immunotherapy, thus offering a potential combinatorial strategy for cancer therapy.
    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
    放射治疗,尽管它的精确性和非侵入性,通常由于癌症干细胞(CSC)的耐药性而失败,其特点是高度的自我更新能力和优越的DNA修复机制。这些细胞可以逃避RT并导致肿瘤复发和转移。为了应对这一挑战,介绍了一种名为PB的新型输送系统。该系统将脂质体与血小板膜结合起来,以包封双-2-(5-苯基乙酰氨基-1,2,4-噻二唑-2-基)乙基硫醚(BPTES),从而增强其在肿瘤部位的递送和释放。此外,该系统不仅有效地靶向CSC,而且在X射线照射下增加BPTES的局部浓度,降低肿瘤细胞中谷胱甘肽的水平,从而增加氧化应激和损伤线粒体。PB引起的线粒体损伤作为STING信号的起始点,其介导cGAS-STING途径相关蛋白表达的显著上调,从而放大STING信号。全身静脉给予PB显著促进DC成熟和CD8+T细胞浸润,从而引发强烈的抗肿瘤作用。总的来说,该PB系统提供了克服CSC相关耐药性的有效方法,并为未来的癌症治疗方案提供了有希望的方法.
    Radiotherapy, despite its precision and non-invasiveness, often fails due to the resistance of cancer stem cells (CSCs), which are characterized by high self-renewal capabilities and superior DNA repair mechanisms. These cells can evade RT and lead to tumor recurrence and metastasis. To address this challenge, a novel delivery system named PB has been introduced. This system combines liposomes with platelet membranes to encapsulate Bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES), thus enhancing its delivery and release specifically at tumor sites. In addition, this system not only targets CSCs effectively but also increases the local concentration of BPTES upon X-ray irradiation, which reduces glutathione levels in tumor cells, thereby increasing oxidative stress and damaging mitochondria. PB-elicited mitochondrial damage as the STING signal initiator, which mediated significant upregulation in the expression of a cGAS-STING pathway-related protein thereby amplifying the STING signal. Systemic intravenous administration of PB remarkably promoted DC maturation and CD8+ T cell infiltration, thus eliciting strong antitumor effects. Overall, this PB system presents a potent method to overcome CSC-related resistance and offers a promising approach for future cancer treatment protocols.
    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
    我们评估了局部和肝转移性结直肠癌(LMCC)中免疫抑制肿瘤微环境的调节,专注于肿瘤相关的巨噬细胞,它们是LMCC中主要的免疫抑制细胞。我们开发了一种口服节拍化疗方案,口服CAPOX。该方案结合了卡培他滨和纳米胶束封装,赖氨酸连接的脱氧胆酸盐和奥沙利铂复合物(OPT/LDC-NM)。该治疗通过激活cGAS-STING途径和诱导免疫原性细胞死亡来有效调节肿瘤微环境内的免疫细胞。这种疗法比卡培他滨单一疗法更有效地调节免疫细胞,目前标准的大肠癌维持化疗。口服CAPOX的巨噬细胞修饰作用是通过cGAS-STING途径介导的。这是新发现的由节拍化疗诱导的免疫细胞活化模式。此外,口服CAPOX与抗PD-1抗体(αPD-1)协同增强T细胞介导的抗肿瘤免疫应答。在CT26。CL25皮下模型,联合治疗获得了91%的完全缓解率,并且对肿瘤有明确的记忆效应.这种组合也改变了LMCC的免疫抑制肿瘤微环境,其中αPD-1单药治疗无法实现。口服CAPOX和αPD-1联合疗法优于治疗LMCC的最大耐受剂量,建议将节拍疗法作为一种有前途的策略。
    We evaluated modulation of the immunosuppressive tumor microenvironment in both local and liver metastatic colorectal cancer (LMCC), focusing on tumor-associated macrophages, which are the predominant immunosuppressive cells in LMCC. We developed an orally administered metronomic chemotherapy regimen, oral CAPOX. This regimen combines capecitabine and a nano-micelle encapsulated, lysine-linked deoxycholate and oxaliplatin complex (OPt/LDC-NM). The treatment effectively modulated immune cells within the tumor microenvironment by activating the cGAS-STING pathway and inducing immunogenic cell death. This therapy modulated immune cells more effectively than did capecitabine monotherapy, the current standard maintenance chemotherapy for colorectal cancer. The macrophage-modifying effect of oral CAPOX was mediated via the cGAS-STING pathway. This is a newly identified mode of immune cell activation induced by metronomic chemotherapy. Moreover, oral CAPOX synergized with anti-PD-1 antibody (αPD-1) to enhance the T-cell-mediated antitumor immune response. In the CT26. CL25 subcutaneous model, combination therapy achieved a 91 % complete response rate with a confirmed memory effect against the tumor. This combination also altered the immunosuppressive tumor microenvironment in LMCC, which αPD-1 monotherapy could not achieve. Oral CAPOX and αPD-1 combination therapy outperformed the maximum tolerated dose for treating LMCC, suggesting metronomic therapy as a promising strategy.
    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
    疫苗接种对于预防和控制传染病至关重要,同时降低死亡率。开发安全和通用的佐剂以增强对疫苗的体液和细胞免疫应答仍然是疫苗开发中的关键挑战。这里,我们在水相系统中使用椰油酰胺丙基甜菜碱(CAPB)和PluronicsF127设计了分层介孔MOF-801(HM801)。同时,我们合成了一种新型的SARS-CoV-2纳米疫苗(R@M@HM801),具有对STING激动剂(MSA-2)和Delta受体结合域(Delta-RBD)抗原的高负载能力。R@M@HM801增强了MSA-2和RBD的利用,并有效地将MSA-2和RBD抗原共同递送至引流淋巴结中的抗原呈递细胞,从而促进T和B细胞的活化。淋巴细胞单细胞分析显示R@M@HM801刺激稳健的CD11b+CD4+T细胞,CXCR5+CD4+T滤泡辅助细胞(Tfh),和持久的CD4+CD44+CD62L-,CD8+CD44+CD62L-效应记忆T细胞(TEM)免疫应答,并促进体内CD26+B细胞的增殖活化。同时,与RBD+MAS-2和RBD+MAS-2+明矾疫苗相比,R@M@HM801诱导针对Delta的更强特异性抗体和假病毒的中和。我们的研究证明了分级介孔HM801的功效及其潜在的免疫激活机制在增强针对病毒和其他疾病的适应性免疫应答中的作用。
    Vaccination is essential for preventing and controlling infectious diseases, along with reducing mortality. Developing safe and versatile adjuvants to enhance humoral and cellular immune responses to vaccines remains a key challenge in vaccine development. Here, we designed hierarchical mesoporous MOF-801 (HM801) using a Cocamidopropyl betaine (CAPB) and a Pluronics F127 in an aqueous phase system. Meanwhile, we synthesized a novel SARS-CoV-2 nanovaccine (R@M@HM801) with a high loading capacity for both the STING agonist (MSA-2) and the Delta receptor binding domain (Delta-RBD) antigen. R@M@HM801 enhanced MSA-2 and RBD utilization and effectively co-delivered MSA-2 and RBD antigens to antigen-presenting cells in the draining lymph nodes, thereby promoting the activation of both T and B cells. Lymphocyte single-cell analysis showed that R@M@HM801 stimulated robust CD11b+CD4+ T cells, CXCR5+CD4+ T follicular helper (Tfh), and durable CD4+CD44+CD62L-, CD8+CD44+CD62L- effector memory T cell (TEM) immune responses, and promoted the proliferative activation of CD26+ B cells in vivo. Meanwhile, R@M@HM801 induced stronger specific antibodies and neutralization of pseudovirus against Delta compared to the RBD + MAS-2 and RBD + MAS-2 + Alum vaccines. Our study demonstrated the efficacy of a hierarchical mesoporous HM801 and its potential immune activation mechanism in enhancing adaptive immune responses against viruses and other diseases.
    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
    树突状细胞(DC)成熟和抗原呈递是成功的基于疫苗的癌症免疫治疗的关键因素。这项研究开发了锰基层状双氢氧化物(Mn-LDH)纳米颗粒作为自佐剂疫苗载体,不仅通过协同消耗内源性谷胱甘肽(GSH)和激活STING信号通路促进DC成熟,但也促进了模型抗原卵清蛋白(OVA)进入淋巴结和随后在DC中的抗原呈递。负载OVA的Mn-LDH(OVA/Mn-LDH)纳米疫苗的显着治疗预防功效是通过对携带B16-OVA肿瘤的小鼠的肿瘤生长抑制来确定的。我们的结果表明,OVA/Mn-LDH纳米颗粒可以在不需要佐剂的情况下成为癌症疫苗开发的有效递送系统。因此,GSH耗竭和STING途径激活的组合可能是促进DC成熟和抗原呈递的可取方法,最终提高癌症疫苗的疗效。
    Dendritic cell (DC) maturation and antigen presentation are key factors for successful vaccine-based cancer immunotherapy. This study developed manganese-based layered double hydroxide (Mn-LDH) nanoparticles as a self-adjuvanted vaccine carrier that not only promoted DC maturation through synergistically depleting endogenous glutathione (GSH) and activating STING signaling pathway, but also facilitated the delivery of model antigen ovalbumin (OVA) into lymph nodes and subsequent antigen presentation in DCs. Significant therapeutic-prophylactic efficacy of the OVA-loaded Mn-LDH (OVA/Mn-LDH) nanovaccine was determined by the tumor growth inhibition in the mice bearing B16-OVA tumor. Our results showed that the OVA/Mn-LDH nanoparticles could be a potent delivery system for cancer vaccine development without the need of adjuvant. Therefore, the combination of GSH exhaustion and STING pathway activation might be an advisable approach for promoting DC maturation and antigen presentation, finally improving cancer vaccine efficacy.
    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
    现有癌症疫苗的关键挑战是协调抗原富集的供应和抗原呈递细胞(APC)内的最佳抗原呈递功能的需求。这里,使用负载有干扰素基因刺激因子(STING)激动剂(DT-Exo-STING)的树突状细胞(DC)-肿瘤杂交细胞衍生的嵌合外来体,开发了一种互补的免疫治疗策略,用于最大化肿瘤特异性T细胞免疫。这些嵌合载体配备有广谱抗原复合物,以通过直接自我呈递和间接DC-T免疫刺激途径引发强大的T细胞介导的炎症程序。这种嵌合外泌体辅助递送策略在明亮的组织归巢能力方面与现成的环状二核苷酸(CDN)递送技术相比具有优点,即使穿过棘手的血脑屏障(BBB),和用于增强的STING激活信号传导的期望的细胞溶质进入。这种纳米疫苗驱动的STING激活的改进的抗原呈递性能进一步增强了肿瘤特异性T细胞免疫应答。因此,DT-Exo-STING将免疫抑制性胶质母细胞瘤微环境逆转为促炎,杀肿瘤的状态,导致颅内原发性病变几乎消失。重要的是,利用自体肿瘤组织进行个性化DT-Exo-STING疫苗的升级选项可提高对免疫检查点阻断(ICB)治疗的敏感性,并发挥针对术后神经胶质瘤复发的全身免疫记忆.这些发现代表了胶质母细胞瘤免疫治疗的一种新兴方法,保证临床领域的进一步探索性发展。
    A critical challenge of existing cancer vaccines is to orchestrate the demands of antigen-enriched furnishment and optimal antigen-presentation functionality within antigen-presenting cells (APCs). Here, a complementary immunotherapeutic strategy is developed using dendritic cell (DC)-tumor hybrid cell-derived chimeric exosomes loaded with stimulator of interferon genes (STING) agonists (DT-Exo-STING) for maximized tumor-specific T-cell immunity. These chimeric carriers are furnished with broad-spectrum antigen complexes to elicit a robust T-cell-mediated inflammatory program through direct self-presentation and indirect DC-to-T immunostimulatory pathway. This chimeric exosome-assisted delivery strategy possesses the merits versus off-the-shelf cyclic dinucleotide (CDN) delivery techniques in both the brilliant tissue-homing capacity, even across the intractable blood-brain barrier (BBB), and the desired cytosolic entry for enhanced STING-activating signaling. The improved antigen-presentation performance with this nanovaccine-driven STING activation further enhances tumor-specific T-cell immunoresponse. Thus, DT-Exo-STING reverses immunosuppressive glioblastoma microenvironments to pro-inflammatory, tumoricidal states, leading to an almost obliteration of intracranial primary lesions. Significantly, an upscaling option that harnesses autologous tumor tissues for personalized DT-Exo-STING vaccines increases sensitivity to immune checkpoint blockade (ICB) therapy and exerts systemic immune memory against post-operative glioma recrudesce. These findings represent an emerging method for glioblastoma immunotherapy, warranting further exploratory development in the clinical realm.
    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

  • 文章类型: Journal Article
    联合免疫疗法的功效受到肿瘤特异性和免疫相关不良事件(irAEs)的限制。在这里,我们报道了聚合物STING前激动剂(PSPA)的发展,其声免疫治疗功效由肿瘤微环境(TME)内的声照射和谷胱甘肽(GSH)升高激活。PSPA经由GSH可活化接头由声敏剂(半导体聚合物)和STING激动剂(MSA-2)组成。在声波照射下,PSPA用作超声增敏剂以产生IO2并诱导恶性肿瘤细胞的免疫原性细胞死亡(ICD)。此外,MSA-2在高表达GSH的肿瘤微环境中特异性释放,最小化脱靶副作用。STING途径的激活升高干扰素-β水平并与SDT协同以增强抗肿瘤应答。因此,这项工作为癌症声免疫疗法的时空调控提出了一种通用方法。
    The efficacy of combination immunotherapy has been limited by tumor specificity and immune-related adverse events (irAEs). Herein, we report the development of polymeric STING pro-agonists (PSPA), whose sono-immunotherapeutic efficacy is activated by sono-irradiation and elevated glutathione (GSH) within the tumor microenvironment (TME). PSPA is composed of sonosensitizers (semiconducting polymer) and STING agonists (MSA-2) via the GSH-activatable linkers. Under sono-irradiation, PSPA serves as a sonosensitizer to generate 1 O2 and induce immunogenic cell death (ICD) of malignant tumor cells. Furthermore, MSA-2 is released specifically in tumor microenvironment with highly expressed GSH, minimizing off-target side effects. The activation of the STING pathway elevates the interferon-β level and synergizes with SDT to enhance the anti-tumor response. Therefore, this work proposes a universal approach for spatiotemporal regulation of cancer sono-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
    免疫疗法为治疗头颈部鳞状细胞癌(HNSCC)提供了新的机会;然而,它的临床应用受到适度的治疗结果和免疫调节剂的“始终在线”药理活性的阻碍。精确时空激活抗肿瘤免疫的策略可以解决这些问题,但仍然具有挑战性。在这里,据报道,一种半导体聚合物纳米激动剂(SPNM)具有可原位激活的免疫治疗作用,可用于HNSCC的精确声免疫疗法。SPNM由与干扰素基因刺激物(STING)激动剂(MSA-2)通过单线态氧可裂解接头缀合的声动力半导体聚合物核心自组装。在声波照射下,SPNM不仅产生单线态氧以根除肿瘤细胞以引发免疫原性细胞死亡,而且还通过二苯氧基乙烯键的裂解释放笼中的STING激动剂,以在肿瘤区域原位激活STING途径。这种由SPNM介导的超声驱动的STING激活促进效应T细胞浸润并增强全身抗肿瘤免疫,最终导致肿瘤生长抑制和长期免疫记忆。因此,这项研究为癌症免疫治疗的精确时空激活提供了有希望的策略。
    Immunotherapy has offered new opportunities to treat head and neck squamous cell carcinoma (HNSCC); however, its clinical applications are hindered by modest therapeutic outcomes and the \"always-on\" pharmacological activity of immunomodulatory agents. Strategies for precise spatiotemporal activation of antitumor immunity can tackle these issues but remain challenging. Herein, a semiconducting polymeric nanoagonist (SPNM) with in situ sono-activatable immunotherapeutic effects for precision sono-immunotherapy of HNSCC is reported. SPNM is self-assembled from a sonodynamic semiconducting polymer core conjugated with a stimulator of interferon genes (STING) agonist (MSA-2) via a singlet oxygen cleavable linker. Under sono-irradiation, SPNM produces singlet oxygen not only to eradicate tumor cells to trigger immunogenic cell death but also to unleash caged STING agonists via the cleavage of diphenoxyethene bonds for in situ activation of the STING pathway in the tumor region. Such sono-driven STING activation mediated by SPNM promotes effector T cell infiltration and potentiates systemic antitumor immunity, eventually leading to tumor growth inhibition and long-term immunological memory. This study thus presents a promising strategy for the precise spatiotemporal activation of cancer immunotherapy.
    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