photoimmunotherapy

光免疫疗法
  • 文章类型: Journal Article
    cGAS(环GMP-AMP合酶)-STING(干扰素基因的刺激物)途径通过感测从细胞核和线粒体泄漏的胞质DNA片段来促进抗肿瘤免疫反应。在这里,我们设计了一种高电荷的钌光敏剂(Ru1),其β-咔啉生物碱衍生物作为配体,用于光活化cGAS-STING途径。由于多个非共价分子间相互作用的形成,Ru1可以自组装成无载体纳米粒子(NPs)。通过引入三苯基膦取代基,Ru1可以靶向和光损伤线粒体DNA(mtDNA),导致细胞质DNA泄漏,从而激活cGAS-STING途径。最后,Ru1NP显示出有效的抗肿瘤作用,并在体内引发强烈的免疫反应。总之,我们报道了第一个自组装mtDNA靶向光敏剂,可以有效激活cGAS-STING途径,从而为新的光免疫治疗剂的设计提供了创新。
    The cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) pathway promotes antitumor immune responses by sensing cytosolic DNA fragments leaked from nucleus and mitochondria. Herein, we designed a highly charged ruthenium photosensitizer (Ru1) with a β-carboline alkaloid derivative as the ligand for photo-activating of the cGAS-STING pathway. Due to the formation of multiple non-covalent intermolecular interactions, Ru1 can self-assemble into carrier-free nanoparticles (NPs). By incorporating the triphenylphosphine substituents, Ru1 can target and photo-damage mitochondrial DNA (mtDNA) to cause the cytoplasmic DNA leakage to activate the cGAS-STING pathway. Finally, Ru1 NPs show potent antitumor effects and elicit intense immune responses in vivo. In conclusion, we report the first self-assembling mtDNA-targeted photosensitizer, which can effectively activate the cGAS-STING pathway, thus providing innovations for the design of new photo-immunotherapeutic agents.
    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
    目的:乳腺癌(BC)是全球最常见的恶性疾病。局部阶段的BC可以通过手术成功治疗。然而,局部复发发生在约4-10%的患者,需要系统性治疗,损害患者的生活质量,缩短预期寿命。因此,需要新的治疗选择,可以在术中使用,并有助于完全去除手术区域的残留肿瘤细胞。在本研究中,我们描述了抗HER2抗体曲妥珠单抗的半胱氨酸修饰变体,与硅酞菁光敏剂染料WB692-CB1偶联,用于BC的光免疫疗法(PIT)。
    方法:克隆半胱氨酸修饰的曲妥珠单抗变体并在Expi293F细胞中表达。通过固定化亲和层析纯化后,抗体与染料偶联。通过流式细胞术测量抗体和抗体染料缀合物的细胞结合。将BC细胞与缀合物孵育并通过红光照射激活染料后,确定细胞活力。
    结果:抗体和缀合物显示与表达HER2的BC细胞的特异性结合。用缀合物处理HER2highBC细胞系SK-BR-3,然后用32J/cm2的红光剂量照射导致24小时内完全杀死细胞。
    结论:我们的新型抗体染料缀合物代表了局部BC术中治疗的有希望的候选者,旨在消除手术区域残留的肿瘤细胞,并有可能减少局部复发,从而改善BC患者的康复前景。
    OBJECTIVE: Breast cancer (BC) is the most common malignant disease worldwide. Localized stages of BC can be successfully treated by surgery. However, local recurrence occurs in about 4-10% of patients, requiring systemic treatments that impair the patients\' quality of life and shortens life expectancy. Therefore, new therapeutic options are needed, which can be used intraoperatively and contribute to the complete removal of residual tumor cells in the surgical area. In the present study, we describe a cysteine-modified variant of the anti-HER2 antibody trastuzumab, that was coupled to the silicon phthalocyanine photosensitizer dye WB692-CB1 for the photoimmunotherapy (PIT) of BC.
    METHODS: The cysteine modified trastuzumab variant was cloned and expressed in Expi293F cells. After purification via immobilized affinity chromatography, the antibody was coupled to the dye. Cell binding of the antibody and the antibody dye conjugate was measured by flow cytometry. After incubation of BC cells with the conjugate and activation of the dye by irradiation with red light, cell viability was determined.
    RESULTS: The antibody and the conjugate showed specific binding to HER2-expressing BC cells. Treatment of the HER2high BC cell line SK-BR-3 with the conjugate followed by irradiation with a red light dose of 32 J/cm2 led to complete cell killing within 24 h.
    CONCLUSIONS: Our novel antibody dye conjugate represents a promising candidate for intraoperative treatment of localized BC, aiming to eliminate residual tumor cells in the surgical area and potentially reduce local recurrence, thereby improving recovery prospects for BC patients.
    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
    由于肿瘤内光热剂的积累不足以及治疗期间癌症免疫循环的逆转,光免疫疗法面临挑战。在这项研究中,一种抗PD-L1固定化磁性金纳米小屋,AuNH-2-Ab,光响应,热敏,和免疫调节特性,以有效抑制原发性肿瘤的生长,提高免疫原性细胞死亡(ICD)水平,逆转肿瘤免疫微环境(TIME),并因此抑制转移发展。AuNH-2-Ab在全身给药后实现高肿瘤积累(9.54%注射剂量),允许在肿瘤中调节超过50°C的热疗剂量。通过优化热疗剂量,AuNH-2-Ab同时靶向和消除癌细胞和肿瘤相关巨噬细胞,从而激活有效的抗肿瘤免疫力,而不会受到免疫抑制元件的损害。高温/pH诱导的AuNH-2-Ab的形态转化,涉及表面抗体的分离以抑制原位PD-L1,和内部岩藻依聚糖层的暴露,以激活自然杀伤(NK)细胞。这种精确的光免疫疗法重新编程时间,显著延长小鼠肝细胞癌模型(Hep55.1c)的生存期,并利用单一纳米平台利用ICD生产和检查点抑制剂的协同作用。
    Photoimmunotherapy faces challenges due to insufficient intratumoral accumulation of photothermal agents and the reversion of the cancer-immunity cycle during treatment. In this study, an anti-PD-L1-immobilized magnetic gold nanohut, AuNH-2-Ab, with photoresponsive, thermosensitive, and immunomodulatory properties to effectively suppress the growth of primary tumors, elevate immunogenic cell death (ICD) levels, reverse the tumor immune microenvironment (TIME), and consequently inhibit metastases are developed. AuNH-2-Ab achieves high tumor accumulation (9.54% injected dose) following systemic administration, allowing the modulation of hyperthermia dose of over 50 °C in the tumor. By optimizing the hyperthermia dose, AuNH-2-Ab simultaneously target and eliminate cancer cells and tumor-associated macrophages, thereby activating potent antitumor immunity without being compromised by immunosuppressive elements. Hyperthermia/pH induced morphological transformation of AuNH-2-Ab involving the detachment of the surface antibody for in situ PD-L1 inhibition, and exposure of the inner fucoidan layer for natural killer (NK) cell activation. This precision photoimmunotherapy approach reprograms the TIME, significantly prolongs survival in a murine hepatocellular carcinoma model (Hep55.1c), and harnesses the synergistic effects of ICD production and checkpoint inhibitors by utilizing a single nanoplatform.
    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
    协同癌症疗法由于其多模式肿瘤抑制特性而受到广泛关注。尤其是,光响应光免疫疗法证明了一种新兴的癌症治疗范式,显着提高了治疗效率。在这里,近红外-II反应性卵清蛋白功能化金-京尼平纳米系统(Au-G-OVANRs)被设计用于乳腺癌的免疫治疗和深层光热治疗.采用简便的合成方法制备了具有良好分散性的均相Au纳米棒(AuNRs)。纳米疫苗是通过Au-NRs的化学交联进一步开发的,京尼平和卵清蛋白。Au-G-OVANRs突出的水溶性,以及对正常细胞和癌细胞的生物相容性。所设计的NRs具有增强的局域表面等离子体共振(LSPR)效应,扩大了第二个窗口的近红外吸收,实现有前途的光热特性。此外,京尼平涂层提供了互补的红色荧光,制备的Au-G-OVANRs显示出有效的光免疫疗法结果的显着细胞内封装。设计的纳米系统具有乳腺癌的深层光热疗法,在1064nm激光照射后,Au-G-OVANRs(80μgmL-1浓度)消融了90%的4T1细胞。此外,Au-G-OVANRs通过促进OVA递送表现出突出的疫苗接种现象,抗原摄取,骨髓树突状细胞(BMDCs)的成熟,和细胞因子IFN-γ分泌用于肿瘤免疫监视。上述优点允许利用荧光成像引导的光免疫疗法治疗癌症。展示了开发适合精确肿瘤治疗的纳米疫苗的简单方法。
    Synergistic cancer therapies have attracted wide attention owing to their multi-mode tumor inhibition properties. Especially, photo-responsive photoimmunotherapy demonstrates an emerging cancer treatment paradigm that significantly improved treatment efficiency. Herein, near-infrared-II responsive ovalbumin functionalized Gold-Genipin nanosystem (Au-G-OVA NRs) was designed for immunotherapy and deep photothermal therapy of breast cancer. A facile synthesis method was employed to prepare the homogeneous Au nanorods (Au NRs) with good dispersion. The nanovaccine was developed further by the chemical cross-linking of Au-NRs, genipin and ovalbumin. The Au-G-OVA NRs outstanding aqueous solubility, and biocompatibility against normal and cancer cells. The designed NRs possessed enhanced localized surface plasmon resonance (LSPR) effect, which extended the NIR absorption in the second window, enabling promising photothermal properties. Moreover, genipin coating provided complimentary red fluorescent and prepared Au-G-OVA NRs showed significant intracellular encapsulation for efficient photoimmunotherapy outcomes. The designed nanosystem possessed deep photothermal therapy of breast cancer and 90% 4T1 cells were ablated by Au-G-OVA NRs (80μg ml-1concentration) after 1064 nm laser irradiation. In addition, Au-G-OVA NRs demonstrated outstanding vaccination phenomena by facilitating OVA delivery, antigen uptake, maturation of bone marrow dendritic cells, and cytokine IFN-γsecretion for tumor immunosurveillance. The aforementioned advantages permit the utilization of fluorescence imaging-guided photo-immunotherapy for cancers, demonstrating a straightforward approach for developing nanovaccines tailored to precise tumor 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
    目的:虽然一些研究报道了唾液腺癌(SGC)中表皮生长因子受体(EGFR)的表达,由于缺乏EGFR阳性的统一定义,结果各不相同.在这项研究中,我们使用EGFR阳性评分和EGFR累积评分评估了SGC患者的EGFR表达水平.
    方法:2010年1月至2021年4月,对102例接受手术切除的SGC患者进行免疫组织化学回顾性分析。膜染色强度评分如下:无染色(0),弱染色(1+),中间染色(2+),和强染色(3+)。使用公式:IX(1+:弱染色细胞的百分比)+2X(2+:中度染色细胞的百分比)+3X(3+:强染色细胞的百分比)在0-300的连续标度上确定累积EGFR得分。
    结果:EGFR在SGC中的表达差异很大,即使在相同和不同的组织病理学类型中也是如此。平均EGFR阳性评分为46.0%,55.7%,51.6%,1.0%,26.8%,50%,粘液表皮样癌(MEC)占76.8%,涎管癌(SDC),腺样囊性癌(AdCC),腺泡细胞癌(AcCC),腺癌NOS(ACNOS),多形性腺瘤(CAexPA),鳞状细胞癌(SqCC),分别。MEC的平均累积EGFR评分分别为82、91、80、1、52、93和185。SDC,AdCC,AcCC,ACNOS,CAexPA,和SqCC,分别。
    结论:SGC的EGFR阳性评分和累积EGFR评分在不同组织学类型之间有所不同,甚至在相同的组织学类型。这些评分可以预测EGFR靶向治疗的SGC的临床结果。例如头颈部光免疫疗法,并需要在未来的研究中进行评估。
    OBJECTIVE: While several studies reported epidermal growth factor receptor (EGFR) expression in salivary gland cancer (SGC), results varied due to a lack of unified definition of EGFR positivity. In this study, we assessed the EGFR expression level using both EGFR positive score and cumulative EGFR score in the patients with SGC.
    METHODS: Between January 2010 and April 2021, 102 patients with SGC who underwent surgical resection were reviewed retrospectively by immunohistochemistry. The membrane staining intensity was scored as follows: no staining (0), weak staining (1+), intermediate staining (2+), and strong staining (3+). The cumulative EGFR score was determined on a continuous scale of 0-300 using the formula:1 × (1+: percentage of weakly stained cells) + 2 × (2+: percentage of moderately stained cells) + 3 × (3+: percentage of strongly stained cells).
    RESULTS: EGFR expression in SGC varied widely even among the same as well as different histopathological types. The average EGFR positive scores were 46.0 %, 55.7 %, 51.6 %, 1.0 %, 26.8 %, 50 %, and 76.8 % for mucoepidermoid carcinoma (MEC), salivary duct carcinoma (SDC), adenoid cystic carcinoma (AdCC), acinic cell carcinoma (AcCC), adenocarcinoma NOS (ACNOS), carcinoma ex pleomorphic adenoma (CAexPA), and squamous cell carcinoma (SqCC), respectively. The average cumulative EGFR scores were 82, 91, 80, 1, 52, 93, and 185 for MEC, SDC, AdCC, AcCC, ACNOS, CAexPA, and SqCC, respectively.
    CONCLUSIONS: EGFR positive scores and cumulative EGFR scores in SGCs varied among the various histological types, and even in the same histological type. These scores may predict the clinical outcome of SGC treated with EGFR-targeting therapies, such as head and neck photoimmunotherapy, and need to be evaluated in future studies.
    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
    目的:局限性前列腺癌的标准治疗包括手术切除前列腺。然而,当肿瘤细胞持续存在于手术部位时,局部复发和肿瘤扩散的风险很高,导致紧张的后续治疗,生活质量受损,降低总体生存率。这项研究检查了光免疫疗法(PIT)作为前列腺癌细胞的新治疗选择。
    方法:我们产生了由靶向前列腺特异性膜抗原(PSMA)的人源化抗体或其Fab片段组成的缀合物,以及我们的硅酞菁光敏剂染料WB692-CB1。将表达PSMA的前列腺癌细胞与抗体染料或Fab染料缀合物一起孵育,并使用流式细胞术测量细胞结合。用不同剂量的红光照射细胞以激活染料,和细胞毒性通过红色素B染色和随后使用Neubauer计数室的分析来确定。
    结果:用抗体染料缀合物在前列腺癌细胞中以光剂量依赖性方式诱导特异性细胞毒性。用Fab染料缀合物处理细胞导致较低的细胞毒性,这可归因于Fab片段的降低的结合亲和力和降低的染料摄取。
    结论:我们的新抗体染料和Fab染料缀合物提供了未来局部前列腺癌患者术中PIT的潜力,为了确保从手术区域完全去除肿瘤细胞,为了避免局部复发,并改善临床结果。
    OBJECTIVE: The standard treatment for localized prostate cancer involves surgical removal of the prostate with curative intent. However, when tumor cells persist in the operation site, there is high risk of local recurrence and tumor spread, leading to stressful follow-up treatments, impaired quality of life, and reduced overall survival. This study examined photoimmunotherapy (PIT) as a new treatment option for prostate cancer cells.
    METHODS: We generated conjugates consisting of either a humanized antibody or Fab fragments thereof targeting the prostate specific membrane antigen (PSMA), along with our silicon phthalocyanine photosensitizer dye WB692-CB1. PSMA-expressing prostate cancer cells were incubated with the antibody dye or Fab dye conjugates and cell binding was measured using flow cytometry. Cells were irradiated with varying doses of red light for dye activation, and cytotoxicity was determined by erythrosin B staining and subsequent analysis using a Neubauer counting chamber.
    RESULTS: Specific cytotoxicity was induced with the antibody dye conjugate in the prostate cancer cells in a light dose-dependent manner. Treatment of the cells with the Fab dye conjugate resulted in lower cytotoxicity, which could be attributed to a reduced binding affinity and a reduced dye uptake of the Fab fragment.
    CONCLUSIONS: Our new antibody dye and Fab dye conjugates offer potential for future intraoperative PIT in patients with localized prostate cancer, with the aim to ensure complete removal of tumor cells from the surgical area, to avoid local recurrence, and to improve clinical outcome.
    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:光敏剂和吲哚胺-2,3双加氧酶(IDO)抑制剂的组合为黑素瘤治疗提供了有前途的光免疫疗法(PIT)策略。双重药物递送系统为优化PIT对黑素瘤增殖和转移的抑制作用提供了潜在的方法。
    目的:开发基于PIT的双重给药系统,并研究其抑制黑色素瘤增殖和转移的功效。
    方法:我们使用光敏剂-紫癜18(P18)构建了多功能纳米卟啉材料(P18-APBA-HA),透明质酸(HA),和4-(氨基甲基)苯基硼酸(APBA)。将所得的P18-APBA-HA插入磷脂膜,并将IDO抑制剂epacadostat(EPA)加载到内相中,以制备双重药物递送系统(Lip\\EPA\\P18-APBA-HA)。此外,我们还研究了它的物理化学性质,瞄准,抗肿瘤免疫,以及抗肿瘤增殖和转移的作用。
    结果:设计的系统利用硼酸酯的pH敏感性来实现增强靶向策略,以促进药物在肿瘤病变中的分布和有效的受体介导的细胞内吞作用。从Lip\\EPA\\P18-APBA-HA的细胞内释放EPA是由热辐射触发的,从而抑制肿瘤微环境中的IDO活性,并促进免疫反应的激活。静脉给药Lip\\EPA\\P18-APBA-HA通过促进树突状细胞成熟有效诱导抗肿瘤免疫,细胞毒性T细胞活化,和调节性T细胞抑制,调节细胞因子分泌,抑制黑色素瘤的增殖和肺转移。
    结论:所提出的纳米药物递送系统有望为增强EPA和P18组合对黑色素瘤增殖和转移的抑制作用提供有希望的策略。
    BACKGROUND: The combination of a photosensitizer and indoleamine-2,3 dioxygenase (IDO) inhibitor provides a promising photoimmunotherapy (PIT) strategy for melanoma treatment. A dual drug delivery system offers a potential approach for optimizing the inhibitory effects of PIT on melanoma proliferation and metastasis.
    OBJECTIVE: To develop a dual drug delivery system based on PIT and to study its efficacy in inhibiting melanoma proliferation and metastasis.
    METHODS: We constructed a multifunctional nano-porphyrin material (P18-APBA-HA) using the photosensitizer-purpurin 18 (P18), hyaluronic acid (HA), and 4-(aminomethyl) phenylboronic acid (APBA). The resulting P18-APBA-HA was inserted into a phospholipid membrane and the IDO inhibitor epacadostat (EPA) was loaded into the internal phase to prepare a dual drug delivery system (Lip\\EPA\\P18-APBA-HA). Moreover, we also investigated its physicochemical properties, targeting, anti-tumor immunity, and anti-tumor proliferation and metastasis effects.
    RESULTS: The designed system utilized the pH sensitivity of borate ester to realize an enhanced-targeting strategy to facilitate the drug distribution in tumor lesions and efficient receptor-mediated cellular endocytosis. The intracellular release of EPA from Lip\\EPA\\P18-APBA-HA was triggered by thermal radiation, thereby inhibiting IDO activity in the tumor microenvironment, and promoting activation of the immune response. Intravenous administration of Lip\\EPA\\P18-APBA-HA effectively induced anti-tumor immunity by promoting dendritic cell maturation, cytotoxic T cell activation, and regulatory T cell suppression, and regulating cytokine secretion, to inhibit the proliferation of melanoma and lung metastasis.
    CONCLUSIONS: The proposed nano-drug delivery system holds promise as offers a promising strategy to enhance the inhibitory effects of the combination of EPA and P18 on melanoma proliferation and metastasis.
    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
    肿瘤免疫疗法已成为一种有效的治疗方法,可以动员患者的免疫系统来实现持久的肿瘤抑制。这里,我们设计了一种光动力疗法驱动的纳米疫苗(Dex-HDL/ALA-Fe3O4)共递送5-氨基乙酰丙酸和Fe3O4纳米酶,证明了一种长期持久的免疫治疗策略.接种疫苗后,纳米疫苗表现出明显的肿瘤部位积累,淋巴结归巢,和特异性和记忆抗肿瘤免疫唤起。激光照射后,Dex-HDL/ALA-Fe3O4在肿瘤部位有效地产生活性氧,不仅诱导免疫原性细胞死亡级联反应,而且触发全类型肿瘤抗原的按需释放。有趣的是,Fe3O4纳米酶催化过氧化氢产生的氧气用于缓解肿瘤缺氧和改变抑制性肿瘤微环境,从而表现出作为敏化剂的显著潜力。在不同的临床前癌症模型中静脉内施用纳米疫苗已经证明了显著的肿瘤消退和术后肿瘤复发和转移的抑制,从而实现针对高度异质性肿瘤的个性化治疗策略。
    Tumor immunotherapy has emerged as an efficacious therapeutic approach that mobilizes the patient\'s immune system to achieve durable tumor suppression. Here, we design a photodynamic therapy-motivated nanovaccine (Dex-HDL/ALA-Fe3O4) co-delivering 5-aminolevulinic acid and Fe3O4 nanozyme that demonstrate a long-term durable immunotherapy strategy. After vaccination, the nanovaccine exhibits obvious tumor site accumulation, lymph node homing, and specific and memory antitumor immunity evocation. Upon laser irradiation, Dex-HDL/ALA-Fe3O4 effectively generates reactive oxygen species at the tumor site not only to induce the immunogenic cell death-cascade but also to trigger the on-demand release of full types of tumor antigens. Intriguingly, Fe3O4 nanozyme-catalyzed hydrogen peroxide generated oxygen for alleviating tumor hypoxia and modifying the inhibitory tumor microenvironment, thereby exhibiting remarkable potential as a sensitizer. The intravenous administration of nanovaccines in diverse preclinical cancer models has demonstrated remarkable tumor regression and inhibition of postoperative tumor recurrence and metastasis, thereby enabling personalized treatment strategies against highly heterogeneous tumors.
    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
    光疗通过诱导免疫原性细胞死亡(ICD)促进抗肿瘤免疫,然而,伴随的炎症反应也会引发免疫抑制,减弱光免疫疗法的疗效。在这里,他们共同组装了靶向细胞膜的嵌合肽C16-Cypate-RRKK-PEG8-COOH(CCP)和抗炎双氯芬酸(DA),以开发一种纳米药物(CCP@DA),该药物既可以增强光疗的免疫效果,又可以减弱炎症介导的免疫抑制。CCP@DA实现了细胞膜靶向光动力和光热协同治疗,以损伤程序性死亡配体1(PD-L1),并诱导强烈的ICD激活抗肿瘤反应。同时,释放的DA抑制肿瘤细胞中的环过氧化物酶-2(COX-2)/前列腺素E2(PGE2)通路以抑制肿瘤炎症,并进一步下调PD-L1表达以缓解免疫抑制微环境。CCP@DA在体外和体内均能显著抑制肿瘤生长和炎症反应,同时保持有效的抗肿瘤免疫反应。此外,它表现出优异的抗转移能力,并通过单剂量和低水平的近红外(NIR)光暴露延长小鼠的存活时间。这项工作为控制高效光免疫疗法的治疗诱导的炎症提供了有价值的策略。
    Phototherapy promotes anti-tumor immunity by inducing immunogenic cell death (ICD), However, the accompanying inflammatory responses also trigger immunosuppression, attenuating the efficacy of photo-immunotherapy. Herein, they co-assembled a cell-membrane targeting chimeric peptide C16-Cypate-RRKK-PEG8-COOH (CCP) and anti-inflammatory diclofenac (DA) to develop a nanodrug (CCP@DA) that both enhances the immune effect of phototherapy and weakens the inflammation-mediated immunosuppression. CCP@DA achieves cell membrane-targeting photodynamic and photothermal synergistic therapies to damage programmed death ligand 1 (PD-L1) and induce a strong ICD to activate anti-tumor response. Simultaneously, the released DA inhibits the cycoperoxidase-2 (COX-2)/prostaglandin E2 (PGE2) pathway in tumor cells to inhibit pro-tumor inflammation and further down-regulate PD-L1 expression to relieve the immunosuppressive microenvironment. CCP@DA significantly inhibited tumor growth and inflammation both in vitro and in vivo, while maintaining a potent anti-tumor immune response. Additionally, it exhibits excellent anti-metastatic capabilities and prolongs mouse survival time with a single dose and low levels of near-infrared (NIR) light exposure. This work provides a valuable strategy to control the therapy-induced inflammation for high-efficiency photoimmunotherapy.
    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
    癌症免疫疗法在解决各种恶性肿瘤方面具有重要的前景。然而,其疗效仍然受到复杂的肿瘤免疫抑制微环境的限制。在这里,设计了光触发的纳米酶Fe-TCPP-R848-PEG(Fe-MOF-RP)用于重塑免疫抑制微环境。Fe-TCPP-MOFs不仅用作对抗肿瘤破坏的核心催化成分,而且还用作免疫激动剂的生物相容性递送载体。改善其长期循环和肿瘤富集。同时,它催化肿瘤内H2O2的分解,产生氧气以增强光动力疗法。诱导的铁性凋亡,与光动力疗法协同作用,促使肿瘤相关抗原从肿瘤细胞中释放,诱导免疫原性细胞死亡。R848激动剂的光触发按需释放刺激树突状细胞的成熟,并将促进肿瘤的M2表型恢复为过继性M1巨噬细胞,这进一步重塑了肿瘤免疫抑制微环境。值得注意的是,纳米酶有效地抑制成熟的肿瘤,如B16F10黑色素瘤。此外,它证明了对原位光治疗的远端肿瘤抑制作用。更重要的是,在肺转移模型中,它引发强大的免疫记忆,赋予对肿瘤再攻击的持久保护。我们的研究提出了一种简单而广泛适用的策略,用于制作具有有效阻止癌症复发和转移潜力的纳米酶。
    Cancer immunotherapy holds significant promise for addressing diverse malignancies. Nevertheless, its efficacy remains constrained by the intricate tumor immunosuppressive microenvironment. Herein, a light-triggered nanozyme Fe-TCPP-R848-PEG (Fe-MOF-RP) was designed for remodeling the immunosuppressive microenvironment. The Fe-TCPP-MOFs were utilized not only as a core catalysis component against tumor destruction but also as a biocompatible delivery vector of an immunologic agonist, improving its long circulation and tumor enrichment. Concurrently, it catalyzes the decomposition of H2O2 within the tumor, yielding oxygen to augment photodynamic therapy. The induced ferroptosis, in synergy with photodynamic therapy, prompts the liberation of tumor-associated antigens from tumor cells inducing immunogenic cell death. Phototriggered on-demand release of R848 agonists stimulated the maturation of dendritic cells and reverted the tumor-promoting M2 phenotypes into adoptive M1 macrophages, which further reshaped the tumor immunosuppressive microenvironment. Notably, the nanozyme effectively restrains well-established tumors, such as B16F10 melanoma. Moreover, it demonstrates a distal tumor-inhibiting effect upon in situ light treatment. What is more, in a lung metastasis model, it elicits robust immune memory, conferring enduring protection against tumor rechallenge. Our study presents a straightforward and broadly applicable strategy for crafting nanozymes with the potential to effectively thwart cancer recurrence and metastasis.
    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