Photothermal therapy (PTT)

光热疗法 (PTT)
  • 文章类型: Journal Article
    通过光学成像引导的癌症光热治疗(PTT)最近显示出精确诊断和有效治疗的巨大潜力。第二个近红外窗口(NIR-II,1000-1700nm)荧光成像(FLI)由于其良好的空间和时间分辨率而非常理想,深层组织穿透,和可忽略的组织毒性。有机小分子由于其低毒性而在生物医学研究中作为成像和治疗剂具有吸引力。快速清除率,不同的结构,易于修改,和优良的生物相容性。已经研究了各种有机小分子用于生物医学应用。然而,很少有关于使用croconaine染料(CRs)的报道,特别是NIR-II发射CRs。据我们所知,以前没有基于CRs的NIR-II发射性小型有机光热剂(SOPTAs)的报道。在这里,我们报告了一种基于croconaine染料(CR-TPE-T)的纳米颗粒(CRNP),在NIR-I和NIR-II窗口中具有吸收和荧光发射,分别。CRNP表现出强烈的近红外吸收,出色的光热性能,良好的生物相容性。体内研究表明,CRNP不仅实现了实时,肿瘤的非侵入性NIR-IIFLI,而且还通过成像引导激光照射诱导了显著的肿瘤消融,没有明显的副作用,并促进结直肠癌模型中抗肿瘤免疫记忆的形成。此外,CRNP显示有效抑制乳腺肿瘤生长,改善小鼠的寿命并引发有效的全身免疫反应,这进一步抑制了肿瘤向肺部的转移。我们的研究证明了CRs作为NIR-II区域癌症诊断的治疗剂的巨大潜力。
    Photothermal therapy (PTT) for cancers guided by optical imaging has recently shown great potential for precise diagnosis and efficient therapy. The second near-infrared window (NIR-II, 1000-1700 nm) fluorescence imaging (FLI) is highly desirable owing to its good spatial and temporal resolution, deep tissue penetration, and negligible tissue toxicity. Organic small molecules are attractive as imaging and treatment agents in biomedical research because of their low toxicity, fast clearance rate, diverse structures, ease of modification, and excellent biocompatibility. Various organic small molecules have been investigated for biomedical applications. However, there are few reports on the use of croconaine dyes (CRs), especially NIR-II emission CRs. To our knowledge, there have been no prior reports of NIR-II emissive small organic photothermal agents (SOPTAs) based on CRs. Herein, we report a croconaine dye (CR-TPE-T)-based nanoparticle (CR NP) with absorption and fluorescence emission in the NIR-I and NIR-II windows, respectively. The CR NPs exhibited intense NIR absorption, outstanding photothermal properties, and good biological compatibility. In vivo studies showed that CR NPs not only achieved real-time, noninvasive NIR-II FLI of tumors, but also induced significant tumor ablation with laser irradiation guided by imaging, without apparent side effects, and promoted the formation of antitumor immune memory in a colorectal cancer model. In addition, the CR NPs displayed efficient inhibition of breast tumor growth, improved longevity of mice and triggered efficient systemic immune responses, which further inhibited tumor metastasis to the lungs. Our study demonstrates the great potential of CRs as therapeutic agents in the NIR-II region for cancer diagnosis.
    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
    生物膜相关感染对医疗保健构成了重大挑战,构成80%的细菌感染,通常导致持久性,慢性疾病。由于由细菌生物膜屏障诱导的高耐受性和抗性,常规抗生素在对抗这些感染的功效上挣扎。二维纳米材料,比如石墨烯家族的那些,氮化硼,二硫化钼(MoS2),MXene,和黑磷,具有对抗生物膜的巨大潜力。这些基于纳米材料的抗菌策略是新颖的工具,在克服耐药细菌和顽固的生物膜方面显示出希望,有能力规避现有的耐药机制。这篇综述全面总结了二维纳米材料的最新进展,作为精确抗生素递送的治疗剂和纳米载体,特别关注纳米平台与光热/光动力疗法相结合,以消除细菌和穿透和/或消融生物膜。这篇综述提供了对当前抗菌纳米治疗方法的最新进展和当前局限性的重要见解,并就该领域的未来发展进行了讨论,为了公共卫生的整体利益。
    Biofilm-associated infections pose a significant challenge in healthcare, constituting 80% of bacterial infections and often leading to persistent, chronic conditions. Conventional antibiotics struggle with efficacy against these infections due to the high tolerance and resistance induced by bacterial biofilm barriers. Two-dimensional nanomaterials, such as those from the graphene family, boron nitride, molybdenum disulfide (MoS2), MXene, and black phosphorus, hold immense potential for combating biofilms. These nanomaterial-based antimicrobial strategies are novel tools that show promise in overcoming resistant bacteria and stubborn biofilms, with the ability to circumvent existing drug resistance mechanisms. This review comprehensively summarizes recent developments in two-dimensional nanomaterials, as both therapeutics and nanocarriers for precision antibiotic delivery, with a specific focus on nanoplatforms coupled with photothermal/photodynamic therapy in the elimination of bacteria and penetrating and/or ablating biofilm. This review offers important insight into recent advances and current limitations of current antibacterial nanotherapeutic approaches, together with a discussion on future developments in the field, for the overall benefit of public health.
    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
    吲哚菁绿是FDA批准的用于确定心输出量的荧光成像染料,肝功能,肝脏血流量,和视网膜灌注。已在光声成像和光热治疗(PTT)中进行了临床前研究;但是,ICG光降解限制了其生物医学应用。ICG的汇总形式,被称为J-骨料(IJA),表现出比单体ICG优异的光声信号和热稳定性。然而,IJA在生物环境中仍然存在低稳定性,体内血液循环短。为了解决这些限制,已经开发了一系列纳米载体来增强IJA的稳定性和性能。这篇综述侧重于IJA的潜力和局限性,除了最近IJA负载纳米载体的发展,特别是癌症成像和治疗。
    Indocyanine green is an FDA-approved fluorescent imaging dye used for determining cardiac output, hepatic function, liver blood flow, and retinal perfusion. It has been investigated preclinically in photoacoustic imaging and photothermal therapy (PTT); however, ICG photodegradation limits its biomedical applications. An aggregated form of ICG, known as J-aggregate (IJA), exhibits superior photoacoustic signals and thermal stability than the monomeric ICG. Nevertheless, IJA still suffers from low stability in the biological milieu, and short in vivo blood circulation. To address these limitations, a range of nanocarriers have been developed to enhance IJA stability and performance. This review focuses on IJA potentials and limitations, besides the recent development of IJA-loaded nanocarriers, particularly for cancer imaging and 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
    干扰素基因的先天免疫刺激因子(STING)途径的刺激已显示可增强抗肿瘤免疫力。然而,STING激动剂向肿瘤的全身递送提出了挑战。因此,我们设计了一种基于环状二核苷酸(CDN)的药物递送系统(DDS)联合光热(PTT)/光动力(PDT)/免疫疗法治疗皮肤黑色素瘤.我们共封装了一种活性氧(ROS)响应性前药硫酮连接的CDN(TK-CDN),线粒体靶向试剂三苯基鳞(TPP)修饰的脂质体(Lipo/TK-CDN/TPP/Y6)中的光响应剂氯E6(Y6)。Lipo/TK-CDN/TPP/Y6表现出与Y6相似的光热效应,并具有较高的细胞摄取率。B16F10细胞胞吞后,Lipo/TK-CDN/TPP/Y6在用于PDT的激光照射下产生大量的ROS。将携带B16F10肿瘤的小鼠静脉注射Lipo/TK-CDN/TPP/Y6并暴露于照射下,导致肿瘤生长的实质性抑制。对抗肿瘤作用机制的探索表明,Lipo/TK-CDN/TPP/Y6与其他组相比,对STING激活和抗肿瘤免疫细胞浸润具有更强的刺激作用。因此,Lipo/TK-CDN/TPP/Y6纳米颗粒作为DDS在肿瘤部位靶向和按需药物释放方面具有巨大潜力。这些纳米颗粒有望成为肿瘤治疗中精确和可控的联合治疗的候选物。
    Stimulation of the innate immune stimulator of interferon genes (STING) pathway has been shown to boost anti-tumour immunity. Nevertheless, the systemic delivery of STING agonists to the tumour presents challenges. Therefore, we designed a cyclic dinucleotide (CDN)-based drug delivery system (DDS) combined photothermal therapy (PTT)/photodynamic therapy (PDT)/immunotherapy for cutaneous melanoma. We coencapsulated a reactive oxygen species (ROS)-responsive prodrug thioketone-linked CDN (TK-CDN), and photoresponsive agents chlorin E6 (Y6) within mitochondria-targeting reagent triphenylphosphonium (TPP)-modified liposomes (Lipo/TK-CDN/TPP/Y6). Lipo/TK-CDN/TPP/Y6 exhibited a photothermal effect similar to Y6, along with a superior cellular uptake rate. Upon endocytosis by B16F10 cells, Lipo/TK-CDN/TPP/Y6 generated large amounts of ROS under laser irradiation for PDT. Mice bearing B16F10 tumours were intravenously injected with Lipo/TK-CDN/TPP/Y6 and exposed to irradiation, resulting in a substantial inhibition of tumour growth. Exploration of the mechanism of anti-tumour action showed that Lipo/TK-CDN/TPP/Y6 had a stronger stimulation of STING activation and anti-tumour immune cell infiltration compared to other groups. Hence, the Lipo/TK-CDN/TPP/Y6 nanoparticles offer great potential as a DDS for targeted and on-demand drug release at tumour sites. These nanoparticles exhibit promise as a candidate for precise and controllable combination therapy in the treatment of tumours.
    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
    乳腺癌是女性死亡率较高的恶性肿瘤。因此,有必要开发新的疗法来有效治疗这种疾病。在这项研究中,硒化铁纳米棒(FeSe2NRs)被设计用于磁热,光热,乳腺癌的化疗动力学治疗(MHT/PTT/CDT)。为了说明它们的功效,用化疗剂甲氨蝶呤(MTX)修饰FeSe2NRs。MTX修饰的FeSe2(FeSe2-MTX)表现出优异的受控药物释放特性。从FeSe2NRs释放的Fe2通过类Fenton/Fenton反应诱导H2O2释放·OH,增强CDT的疗效。在交变磁场(AMF)刺激和808nm激光照射下,FeSe2-MTX通过抑制乳腺癌裸鼠模型中的肿瘤生长而发挥有效的高温和光热效应。此外,FeSe2NRs可以通过将它们的超顺磁性特征结合到单个纳米材料中来用于体内磁共振成像。总的来说,我们提出了一种将功能性纳米系统精确递送至肿瘤的新技术,该技术可增强乳腺癌治疗的疗效.
    Breast cancer is a malignant tumor with a high mortality rate among women. Therefore, it is necessary to develop novel therapies to effectively treat this disease. In this study, iron selenide nanorods (FeSe2 NRs) were designed for use in magnetic hyperthermic, photothermal, and chemodynamic therapy (MHT/PTT/CDT) for breast cancer. To illustrate their efficacy, FeSe2 NRs were modified with the chemotherapeutic agent methotrexate (MTX). MTX-modified FeSe2 (FeSe2-MTX) exhibited excellent controlled drug release properties. Fe2+ released from FeSe2 NRs induced the release of •OH from H2O2 via a Fenton/Fenton-like reaction, enhancing the efficacy of CDT. Under alternating magnetic field (AMF) stimulation and 808 nm laser irradiation, FeSe2-MTX exerted potent hyperthermic and photothermal effects by suppressing tumor growth in a breast cancer nude mouse model. In addition, FeSe2 NRs can be used for magnetic resonance imaging in vivo by incorporating their superparamagnetic characteristics into a single nanomaterial. Overall, we presented a novel technique for the precise delivery of functional nanosystems to tumors that can enhance the efficacy of breast cancer treatment.
    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
    一种创新的纳米酶,铁掺杂聚多巴胺(Fe-PDA),将铁离子整合到PDA基质中,赋予过氧化物酶模拟活性,并实现了43.5%的实质性光热转化效率。Fe-PDA介导H2O2催化产生毒性羟基自由基(•OH),从而促进肿瘤细胞中的脂质过氧化并诱导铁凋亡。溶质载体家族7号的下调。11(SLC7A11)和溶质载体家族3号。系统Xc-中的2(SLC3A2)导致细胞内谷胱甘肽(GSH)产生减少和核因子红系2相关因子2(NRF2)-谷胱甘肽过氧化物酶4(GPX4)途径的失活,有助于铁性凋亡。此外,光热疗法(PTT)的应用增强了化学动力学疗法(CDT)的有效性,加速Fenton反应以靶向肿瘤根除,同时保留邻近的非癌组织。体内实验表明,Fe-PDA显著阻碍了小鼠的肿瘤进展,强调结合CDT和PTT的双模式治疗在未来临床肿瘤学应用中的潜力。
    An innovative nanozyme, iron-doped polydopamine (Fe-PDA), which integrates iron ions into a PDA matrix, conferred peroxidase-mimetic activity and achieved a substantial photothermal conversion efficiency of 43.5 %. Fe-PDA mediated the catalysis of H2O2 to produce toxic hydroxyl radicals (•OH), thereby facilitating lipid peroxidation in tumour cells and inducing ferroptosis. Downregulation of solute carrier family 7 no. 11 (SLC7A11) and solute carrier family 3 no. 2 (SLC3A2) in System Xc- resulted in decreased intracellular glutathione (GSH) production and inactivation of the nuclear factor erythroid 2-related factor 2 (NRF2)-glutathione peroxidase 4 (GPX4) pathway, contributing to ferroptosis. Moreover, the application of photothermal therapy (PTT) enhanced the effectiveness of chemodynamic therapy (CDT), accelerating the Fenton reaction for targeted tumour eradication while sparing adjacent non-cancerous tissues. In vivo experiments revealed that Fe-PDA significantly hampered tumour progression in mice, emphasizing the potential of the dual-modality treatment combining CDT and PTT for future clinical oncology applications.
    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
    光疗有望用于侵袭性肿瘤的非侵入性治疗,特别是当结合热感应和氧化过程时。在这里,我们显示了与甲苯胺蓝-O(AuSHINRs@TBO)缀合的金壳分离纳米棒对人类结直肠肿瘤细胞(Caco-2)的光毒性增强,并具有光热(PTT)和光动力疗法(PDT)的协同作用。对Caco-2细胞培养物进行的线粒体代谢活性测试(MTT)表明,由于局部表面等离子体共振(LSPR)的光吸收增强,AuSHINR产生了光热效应。AuSHINRs@TBO对Caco-2细胞的光毒性进一步增加,其中氧化过程,如氢过氧化,也在场,导致细胞活力从85.5降低到39.0%。使用Caco-2脂质提取物的Langmuir单层在生物启发的肿瘤膜上研究了负责这些作用的分子水平机制。偏振调制红外反射吸收光谱(PM-IRRAS)表明,AuSHINRs@TBO掺入是由于与Caco-2脂质提取物的带负电荷基团的有吸引力的静电相互作用,导致表面压力等温线膨胀。照射后,含有AuSHINRs@TBO(1:1v/v)的Caco-2脂质提取物单层显示出大约。表面积增加1.0%。这归因于活性氧(ROS)的产生及其与Caco-2脂质提取物单层的相互作用,导致氢过氧化物的形成。AuSHINRs@TBO渗透到提取物的极性基团中促进了氧化作用,允许碳链不饱和的氧化反应。这些机制与共聚焦荧光显微镜的发现一致,其中Caco-2质膜是细胞死亡诱导过程的主要部位。
    Phototherapies are promising for noninvasive treatment of aggressive tumors, especially when combining heat induction and oxidative processes. Herein, we show enhanced phototoxicity of gold shell-isolated nanorods conjugated with toluidine blue-O (AuSHINRs@TBO) against human colorectal tumor cells (Caco-2) with synergic effects of photothermal (PTT) and photodynamic therapies (PDT). Mitochondrial metabolic activity tests (MTT) performed on Caco-2 cell cultures indicated a photothermal effect from AuSHINRs owing to enhanced light absorption from the localized surface plasmon resonance (LSPR). The phototoxicity against Caco-2 cells was further increased with AuSHINRs@TBO where oxidative processes, such as hydroperoxidation, were also present, leading to a cell viability reduction from 85.5 to 39.0%. The molecular-level mechanisms responsible for these effects were investigated on bioinspired tumor membranes using Langmuir monolayers of Caco-2 lipid extract. Polarization-modulation infrared reflection-absorption spectroscopy (PM-IRRAS) revealed that the AuSHINRs@TBO incorporation is due to attractive electrostatic interactions with negatively charged groups of the Caco-2 lipid extract, resulting in the expansion of surface pressure isotherms. Upon irradiation, Caco-2 lipid extract monolayers containing AuSHINRs@TBO (1:1 v/v) exhibited ca. 1.0% increase in surface area. This is attributed to the generation of reactive oxygen species (ROS) and their interaction with Caco-2 lipid extract monolayers, leading to hydroperoxide formation. The oxidative effects are facilitated by AuSHINRs@TBO penetration into the polar groups of the extract, allowing oxidative reactions with carbon chain unsaturations. These mechanisms are consistent with findings from confocal fluorescence microscopy, where the Caco-2 plasma membrane was the primary site of the cell death induction process.
    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
    基于单分子的协同光疗具有抗微生物治疗的巨大潜力。在这里,我们报告了一种正交分子阳离子化策略,以改善七甲菁(Cy7)的活性氧(ROS)和高温生成,用于细菌感染的光动力和光热治疗。阳离子吡啶(Py)通过分子内电荷转移(ICT)在不对称Cy7的内消旋位置引入,以构建非典型的电子转移三联体,这减少了ΔES1-S0,避免了快速的电荷重组,并同时基于自旋轨道电荷转移ISC(SOCT-ISC)机制增强系统间交叉(ISC)。这种独特的分子结构产生反斯托克斯发光(ASL),因为富含高振动旋转能级的可旋转C-N键提高了热带吸收(HBA)效率。在800nm以上的辐照下,与吲哚菁绿(ICG)相比,获得的三联体具有更高的单线态氧量子产率和光热转换效率。Py的阳离子化使三合会通过强烈的静电吸引作用靶向细菌,以及在黑暗中对广谱细菌的杀生物特性。此外,辐射下的三联征可以增强体外生物膜根除性能,并在统计学上改善小鼠MRSA感染伤口的愈合效果。因此,这项工作为设计用于细菌感染的协同光疗的小分子光敏剂提供了一种简单而有效的策略.重要声明:我们开发了一种正交分子阳离子化策略,以增强七甲基花青(Cy7)的活性氧和热效应,用于细菌感染的光动力和光热处理。具体来说,阳离子吡啶(Py)在不对称Cy7的内消旋位置引入,以构建非典型电子转移三联体,这减少了ΔES1-S0,避免了快速的电荷重组,并同时增强系统间交叉(ISC)。这个三合会,具有可旋转的C-N键,由于热带吸收而产生反斯托克斯发光。三联组可增强抗微生物性能,并在统计学上改善小鼠MRSA感染伤口的愈合功效。这种位点特异性阳离子化策略可以提供对用于细菌感染的协同光疗的基于小分子的光敏剂的设计的见解。
    Single-molecule-based synergistic phototherapy holds great potential for antimicrobial treatment. Herein, we report an orthogonal molecular cationization strategy to improve the reactive oxygen species (ROS) and hyperthermia generation of heptamethine cyanine (Cy7) for photodynamic and photothermal treatments of bacterial infections. Cationic pyridine (Py) is introduced at the meso‑position of the asymmetric Cy7 with intramolecular charge transfer (ICT) to construct an atypical electron-transfer triad, which reduces ΔES1-S0, circumvents rapid charge recombination, and simultaneously enhances intersystem crossing (ISC) based on spin-orbit charge-transfer ISC (SOCT-ISC) mechanism. This unique molecular construction produces anti-Stokes luminescence (ASL) because the rotatable CN bond enriched in high vibrational-rotational energy levels improves hot-band absorption (HBA) efficiency. The obtained triad exhibits higher singlet oxygen quantum yield and photothermal conversion efficiency compared to indocyanine green (ICG) under irradiation above 800 nm. Cationization with Py enables the triad to target bacteria via intense electrostatic attractions, as well as biocidal property against a broad spectrum of bacteria in the dark. Moreover, the triad under irradiation can enhance biofilm eradication performance in vitro and statistically improve healing efficacy of MRSA-infected wound in mice. Thus, this work provides a simple but effective strategy to design small-molecule photosensitizers for synergistic phototherapy of bacterial infections. STATEMENT OF SIGNIFICANCE: We developed an orthogonal molecular cationization strategy to enhance the reactive oxygen species and thermal effects of heptamethine cyanine (Cy7) for photodynamic and photothermal treatments of bacterial infections. Specifically, cationic pyridine (Py) was introduced at the meso‑position of the asymmetric Cy7 to construct an atypical electron-transfer triad, which reduced ΔES1-S0, circumvented rapid charge recombination, and simultaneously enhanced intersystem crossing (ISC). This triad, with a rotatable CN bond, produced anti-Stokes luminescence due to hot-band absorption. The triad enhanced antimicrobial performance and statistically improved the healing efficacy of MRSA-infected wounds in mice. This site-specific cationization strategy may provide insights into the design of small molecule-based photosensitizers for synergistic phototherapy of bacterial infections.
    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
    将化学动力疗法(CDT)与光热疗法(PTT)相结合已成为一种有希望的癌症治疗方法。因为它通过氧化还原反应和外部激光感应增强治疗效率。在这项研究中,我们通过碳化工艺设计了金属有机骨架(MOF)衍生的Cu5Zn8/HPCNC,并用金纳米颗粒(Au@Cu5Zn8/HPCNC)装饰它们。所得纳米颗粒用作光热剂和Fenton催化剂。Fenton反应通过与局部H2O2的反应促进了Cu2与Cu的转化,产生了具有有效细胞毒性作用的反应性羟基自由基(·OH)。增强Fenton样反应,实现综合治疗,Au@Cu5Zn8/HPCNC的激光照射通过产生局部热来诱导有效的光热治疗。随着Au@Cu5Zn8/HPCNC在808nm处的吸收显著增加,光热效率为57.45%。此外,Au@Cu5Zn8/HPCNC显示出作为癌症磁共振成像(MRI)造影剂的潜力。此外,PTT和CDT的协同组合显著抑制肿瘤生长。这种PTT和CDT的综合方法为癌症治疗带来了巨大的希望,提供增强的CDT和肿瘤微环境(TME)的调节,为抗击癌症开辟新的途径。
    Combining chemodynamic therapy (CDT) with photothermal therapy (PTT) has developed as a promising approach for cancer treatment, as it enhances therapeutic efficiency through redox reactions and external laser induction. In this study, we designed metal organic framework (MOF) -derived Cu5Zn8/HPCNC through a carbonization process and decorated them with gold nanoparticles (Au@Cu5Zn8/HPCNC). The resulting nanoparticles were employed as a photothermal agent and Fenton catalyst. The Fenton reaction facilitated the conversation of Cu2+ to Cu+ through reaction with local H2O2, generating reactive hydroxyl radicals (·OH) with potent cytotoxic effects. To enhance the Fenton-like reaction and achieve combined therapy, laser irradiation of the Au@Cu5Zn8/HPCNC induced efficient photothermal therapy by generating localized heat. With a significantly increased absorption of Au@Cu5Zn8/HPCNC at 808 nm, the photothermal efficiency was determined to be 57.45 %. Additionally, Au@Cu5Zn8/HPCNC demonstrated potential as a contrast agent for magnetic resonance imaging (MRI) of cancers. Furthermore, the synergistic combination of PTT and CDT significantly inhibited tumor growth. This integrated approach of PTT and CDT holds great promise for cancer therapy, offering enhanced CDT and modulation of the tumor microenvironment (TME), and opening new avenues in the fight against 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
    协同光热免疫疗法由于对原发性和转移性肿瘤的相互增强的治疗作用而引起了广泛的关注。然而,缺乏用于空间的临床批准纳米药物,temporal,和药物共同给药的剂量控制强调了这一领域面临的挑战。这里,光热剂(Cy7-TCF)和免疫检查点阻断剂(NLG919)通过二硫键缀合,构建肿瘤特异性小分子前药(Cy7-TCF-SS-NLG),其自组装成自递送和自配制的类前药纳米组装体(PNA)。在肿瘤细胞中,过量产生的GSH裂解二硫键以释放Cy7-TCF-OH,它重新组装成纳米颗粒以增强光热转化,同时在激光照射下产生活性氧(ROS),然后与内源性白蛋白结合以激活近红外荧光,支持多模式成像引导的光疗,用于原发性肿瘤消融和随后的肿瘤相关抗原(TAA)释放。这些TAAs,与共释放的NLG919组合,有效激活效应T细胞并抑制Tregs,从而增强抗肿瘤免疫力以防止肿瘤转移。这项工作提供了一种简单而有效的策略,该策略将超分子动力学和可逆性与刺激响应性共价键合相结合,以设计具有协同多模态成像引导的光疗和免疫疗法级联的简单小分子,用于具有高临床价值的癌症治疗。
    Synergistic photothermal immunotherapy has attracted widespread attention due to the mutually reinforcing therapeutic effects on primary and metastatic tumors. However, the lack of clinical approval nanomedicines for spatial, temporal, and dosage control of drug co-administration underscores the challenges facing this field. Here, a photothermal agent (Cy7-TCF) and an immune checkpoint blocker (NLG919) are conjugated via disulfide bond to construct a tumor-specific small molecule prodrug (Cy7-TCF-SS-NLG), which self-assembles into prodrug-like nano-assemblies (PNAs) that are self-delivering and self-formulating. In tumor cells, over-produced GSH cleaves disulfide bonds to release Cy7-TCF-OH, which re-assembles into nanoparticles to enhance photothermal conversion while generate reactive oxygen species (ROSs) upon laser irradiation, and then binds to endogenous albumin to activate near-infrared fluorescence, enabling multimodal imaging-guided phototherapy for primary tumor ablation and subsequent release of tumor-associated antigens (TAAs). These TAAs, in combination with the co-released NLG919, effectively activated effector T cells and suppressed Tregs, thereby boosting antitumor immunity to prevent tumor metastasis. This work provides a simple yet effective strategy that integrates the supramolecular dynamics and reversibility with stimuli-responsive covalent bonding to design a simple small molecule with synergistic multimodal imaging-guided phototherapy and immunotherapy cascades for cancer treatment with high clinical value.
    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