photoimmunotherapy

光免疫疗法
  • 文章类型: Journal Article
    冷肿瘤缺乏T细胞浸润,免疫原性低,导致免疫疗法反应不足。因此,如何实现从冷肿瘤到热肿瘤的转化是一个亟待解决的问题。光动力疗法可以引诱肿瘤细胞免疫原性逝世亡(ICD)和激活T淋巴细胞产生肿瘤免疫反响。然而,冷肿瘤微环境中的缺氧限制了光动力疗法的有效性。所以在这篇文章中,基于克服上述问题,构建了作为功能性共递送纳米脂质体的MET-HMME/CAT-HMME@Nlip。首先,缺氧状态可以通过以下两种方式得到改善,一种是CAT-HMME@Nlip中负载的过氧化氢酶可以分解高浓度的过氧化氢产生氧气,另一种是在MET-HMME@Nlip中加载二甲双胍可以通过抑制线粒体呼吸来减少氧气消耗。然后随着底物氧浓度的增加,光动力疗法的敏感性可以大大提高,PDT诱导的ICD的抗肿瘤免疫反应也可以明显增强。此外,二甲双胍可以作为一种小分子免疫检查点抑制剂,降低肿瘤细胞表面PD-L1的表达,从而有效提高细胞毒性T细胞对肿瘤细胞的特异性杀伤能力,而且还通过免疫记忆功能抑制模拟远处肿瘤的生长。本研究为提高低氧冷肿瘤的临床治疗效果提供了新思路,特别是对于由于肿瘤细胞表面PD-L1蛋白的低表达或无表达而无法从免疫治疗中获益的肿瘤。
    Cold tumors lack T cells infiltration and have low immunogenicity, resulting insufficient immunotherapy response. Therefore, how to realize the transformation from cold tumor to hot tumor is an urgent problem to be solved. Photodynamic therapy can induce immunogenic death of tumor cells (ICD) and activate T lymphocytes to produce tumor immune response. However, hypoxia in the cold tumor microenvironment limits the effectiveness of photodynamic therapy. So in this article, MET-HMME/CAT-HMME@Nlip as a functional co-delivery nanoliposomes was constructed based on overcoming the above problems. Firstly, the oxygen-deficient state could be improved by the following two ways, one is catalase loaded in CAT-HMME@Nlip can decompose high concentration hydrogen peroxide to produce oxygen, and the other is metformin loaded in MET-HMME@Nlip can decrease oxygen consumption by inhibiting of mitochondrial respiration. And then with the increase of substrate oxygen concentration, the sensitivity of photodynamic therapy can be greatly improved and the anti-tumor immune response by PDT-induced ICD can also be enhanced obviously. In addition, metformin could act as a small molecule immune checkpoint inhibitor to reduce the expression of PD-L1 on the surface of tumor cells, thereby effectively improving the specific killing ability of cytotoxic T cells to tumor cells which could not only erasing the primary tumor, but also inhibiting the growth of simulated distant tumors through the immune memory function. This study provides a new idea for improving the clinical treatment effect of hypoxic cold tumors, especially for tumors that could not benefit from immunotherapy due to low or no expression of PD-L1 protein on the surface of tumor cells.
    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
    与常规疗法相比,光免疫疗法提供精确的靶向癌症治疗,对健康组织的损害最小,副作用减少,但其疗效可能受到浅层光穿透和潜在肿瘤耐药的限制。这里,一种受体-供体-受体(A-D-A)结构的纳米聚集体是通过双重光疗开发的,包括光动力疗法(PDT)和光热疗法(PTT),由单个近红外(NIR)光触发。受益于强大的分子内电荷转移(ICT),A-D-A结构的纳米聚集体表现出广泛的吸收延伸到NIR区域并有效抑制荧光,这使得深穿透和有效的光热转化(η=67.94%)。合适的HOMO-LUMO分布有助于充分的系统间交叉(ISC),以将基态氧(3O2)转化为单线态氧(1O2)和超氧化物阴离子(·O2-),并催化羟基自由基(·OH)的产生。增强的ICT和ISC效应赋予A-D-A结构化纳米聚集体有效的PTT和PDT治疗宫颈癌,诱导有效的免疫原性细胞死亡。结合临床铝佐剂凝胶,在原位和腹膜内转移宫颈癌动物模型中,开发了一种新的宫颈癌光免疫治疗策略,并证明该策略可显著抑制原发性和转移性肿瘤。无创治疗策略为临床早期和晚期宫颈癌治疗提供了新的见解。
    Compared with conventional therapies, photoimmunotherapy offers precise targeted cancer treatment with minimal damage to healthy tissues and reduced side effects, but its efficacy may be limited by shallow light penetration and the potential for tumor resistance. Here, an acceptor-donor-acceptor (A-D-A)-structured nanoaggregate is developed with dual phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), triggered by single near-infrared (NIR) light. Benefiting from strong intramolecular charge transfer (ICT), the A-D-A-structured nanoaggregates exhibit broad absorption extending to the NIR region and effectively suppressed fluorescence, which enables deep penetration and efficient photothermal conversion (η = 67.94%). A suitable HOMO-LUMO distribution facilitates sufficient intersystem crossing (ISC) to convert ground-state oxygen (3O2) to singlet oxygen (1O2) and superoxide anions (·O2 -), and catalyze hydroxyl radical (·OH) generation. The enhanced ICT and ISC effects endow the A-D-A structured nanoaggregates with efficient PTT and PDT for cervical cancer, inducing efficient immunogenic cell death. In combination with clinical aluminum adjuvant gel, a novel photoimmunotherapy strategy for cervical cancer is developed and demonstrated to significantly inhibit primary and metastatic tumors in orthotopic and intraperitoneal metastasis cervical cancer animal models. The noninvasive therapy strategy offers new insights for clinical early-stage and advanced cervical 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
    光免疫疗法代表了提高癌症治疗中免疫疗法效率的创新方法。这种方法涉及免疫疗法和光疗的融合(包括光动力疗法(PDT)和光热疗法(PTT)等技术)。硼-二吡咯亚甲基(BODIPY)由于其出色的PD和PT效率而具有引发免疫疗法的潜力。然而,水溶性的改善,生物利用度,PD/PT综合效率,BODIPY和肿瘤组织靶向需要引入合适的载体以进行潜在的实际应用。在这里,以生物相容性和GSH响应降解性能优异的二硫键基中空介孔有机二氧化硅(HMON)为载体负载联噻吩Aza-BODIPY染料(B5),构建无样品化疗试剂的B5@HMON纳米平台,实现三重协同光免疫疗法。亲爱的,涉及二硫键,用于提高水溶性,肿瘤组织靶向,通过耗尽GSH和增强B5和HMO之间的主客体相互作用来提高PD效率。研究表明,HMON的大比表面积和多孔特性显着增强了光的收集和氧吸附能力。HMON富介孔结构和内腔的B5负载率为11%。研究发现,三重协同纳米平台引发了更强的抗肿瘤免疫反应,包括肿瘤侵袭,细胞因子产生,钙网蛋白易位,和树突状细胞成熟,在体内和体外引发特异性肿瘤特异性免疫反应。使用具有4T1肿瘤的BALB/c小鼠模型来评估体内肿瘤抑制效率,显示B5@HMON组的几乎所有肿瘤在14天后消失。这种简单的无化疗试剂的B5@HMON纳米平台实现了三重协同光免疫疗法。
    Photoimmunotherapy represents an innovative approach to enhancing the efficiency of immunotherapy in cancer treatment. This approach involves the fusion of immunotherapy and phototherapy (encompassing techniques like photodynamic therapy (PDT) and photothermal therapy (PTT)). Boron-dipyrromethene (BODIPY) has the potential to trigger immunotherapy owing to its excellent PD and PT efficiency. However, the improvements in water solubility, bioavailability, PD/PT combined efficiency, and tumor tissue targeting of BODIPY require introduction of suitable carriers for potential practical application. Herein, a disulfide bond-based hollow mesoporous organosilica (HMON) with excellent biocompatibility and GSH-responsive degradation properties was used as a carrier to load a bithiophene Aza-BODIPY dye (B5), constructing a sample chemotherapy reagent-free B5@HMON nanoplatform achieving triple-synergistic photoimmunotherapy. HMON, involving disulfide bond, is utilized to improve water solubility, tumor tissue targeting, and PD efficiency by depleting GSH and enhancing host-guest interaction between B5 and HMO. The study reveals that HMON\'s large specific surface area and porous properties significantly enhance the light collection and oxygen adsorption capacity. The HMON\'s rich mesoporous structure and internal cavity achieved a loading rate of B5 at 11 %. It was found that the triple-synergistic nanoplatform triggered a stronger anti-tumor immune response, including tumor invasion, cytokine production, calreticulin translocation, and dendritic cell maturation, eliciting specific tumor-specific immunological responses in vivo and in vitro. The BALB/c mouse model with 4T1 tumors was used to assess tumor suppression efficiency in vivo, showing that almost all tumors in the B5@HMON group disappeared after 14 days. Such a simple chemotherapy reagent-free B5@HMON nanoplatform achieved triple-synergistic 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
    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

公众号