Multifunctional Nanoparticles

多功能纳米颗粒
  • 文章类型: Journal Article
    眼底新生血管疾病是一系列严重损害视力的致盲眼病。目前,在临床实践中治疗这些疾病的手段在不断发展,并迅速彻底改变了治疗意见。然而,诸如治疗有效性不足等关键问题,高复发率,和不良的患者依从性仍然需要紧急解决。多功能纳米药物可以对内源性和外源性微环境做出特异性反应,有效地将药物递送到特定的靶标,并参与生物成像和小分子检测等活动。纳米在微(NIM)输送系统,如金属,金属氧化物和上转换纳米颗粒(NPs),量子点,和碳材料,在克服眼球内生理屏障的存在方面显示出一定的优势,并被广泛用于眼科疾病的治疗。很少有研究,然而,已经评估了NIM递送系统治疗眼底新生血管性疾病(FND)的疗效。本研究描述了与使用NIM递送系统治疗FND相关的主要临床治疗策略和不良事件,并总结了必须克服的解剖学障碍。在这次审查中,我们希望强调眼内微环境正常化的原理,旨在为设计新的NIM输送系统以治疗特定FND提供更合理的方法。
    Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.
    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
    基于时空控释和肿瘤成像整合的智能药物平台有望克服传统治疗模式的低效率和不确定性。在这项研究中,开发了一种由热敏水凝胶(聚乙烯醇-羧酸水凝胶(PCF))和多功能纳米颗粒(Fe3O4@Au/Mn(Zn)-4-羧基苯基卟啉/聚多巴胺(FAMxP))组成的复合材料,以在磁共振成像(MRI)和荧光成像(FI)的指导下结合肿瘤免疫原性细胞死亡(ICD)/免疫阻滞(ICB)治疗。它不仅可以通过肿瘤细胞的叶酸受体进一步识别靶细胞,而且在暴露于近红外光后也会产生热溶解,从而在原位缓慢释放FAMxP,从而延长治疗时间,避免肿瘤复发。当FAMxP进入肿瘤细胞时,它以pH依赖性方式释放FAMx。化学动力学,光热和光动力疗法可在癌细胞中引起显著的ICD。因此,通过注射抗程序性细胞死亡配体1可以进一步增强ICB,从而提高肿瘤治疗的有效性。开发的PCF-FAMxP复合水凝胶可以代表具有用于肿瘤的协作MRI/FI引导的靶向治疗途径的简单组合物的更新的药物设计方法。
    Smart drug platforms based on spatiotemporally controlled release and integration of tumor imaging are expected to overcome the inefficiency and uncertainty of traditional theranostic modes. In this study, a composite consisting of a thermosensitive hydrogel (polyvinyl alcohol-carboxylic acid hydrogel (PCF)) and a multifunctional nanoparticle (Fe3O4@Au/Mn(Zn)-4-carboxyphenyl porphyrin/polydopamine (FAMxP)) is developed to combine tumor immunogenic cell death (ICD)/immune checkpoint blockade (ICB) therapy under the guidance of magnetic resonance imaging (MRI) and fluorescence imaging (FI). It can not only further recognize the target cells through the folate receptor of tumor cells, but also produce thermal dissolution after exposure to near-infrared light to slowly release FAMxP in situ, thereby prolonging the treatment time and avoiding tumor recurrence. As FAMxP entered the tumor cells, it released FAMx in a pH-dependent manner. Chemodynamic, photothermal and photodynamic therapy can cause significant ICD in cancer cells. ICB can thus be further enhanced by injecting anti-programmed cell death ligand 1, improving the effectiveness of tumor treatment. The developed PCF-FAMxP composite hydrogel may represent an updated drug design approach with simple compositions for cooperative MRI/FI-guided targeted therapeutic pathways for tumors.
    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
    乳腺癌骨转移是一种晚期疾病,通常采用放疗和化疗治疗,这导致严重的副作用和有限的有效性。为了改善这一点,声动力疗法可能是未来更安全有效的方法。细菌外膜囊泡(OMV)具有优异的免疫调节特性,包括调节巨噬细胞极化,促进DC细胞成熟,增强抗肿瘤作用。将OMV与声动力疗法结合可以产生协同抗肿瘤作用。因此,我们构建了多功能纳米颗粒用于治疗乳腺癌骨转移。我们将乳腺癌细胞膜和细菌外膜囊泡融合形成杂化膜(HM),然后将负载IR780的PLGA与HM封装在一起以产生纳米颗粒,IR780@PLGA@HM,具有肿瘤靶向性,免疫调节,和声动力学能力。实验表明,IR780@PLGA@HM纳米粒子具有良好的生物相容性,有效靶向4T1肿瘤,促进巨噬细胞I型极化和DC细胞活化,抗肿瘤炎症因子表达增强,并表现出在体外和体内有效杀死肿瘤的能力,对乳腺癌骨转移有很好的治疗效果。因此,我们构建的纳米颗粒为有效治疗乳腺癌骨转移提供了新的策略。
    Breast cancer bone metastasis is a terminal-stage disease and is typically treated with radiotherapy and chemotherapy, which causes severe side effects and limited effectiveness. To improve this, Sonodynamic therapy may be a more safe and effective approach in the future. Bacterial outer membrane vesicles (OMV) have excellent immune-regulating properties, including modulating macrophage polarization, promoting DC cell maturation, and enhancing anti-tumor effects. Combining OMV with Sonodynamic therapy can result in synergetic anti-tumor effects. Therefore, we constructed multifunctional nanoparticles for treating breast cancer bone metastasis. We fused breast cancer cell membranes and bacterial outer membrane vesicles to form a hybrid membrane (HM) and then encapsulated IR780-loaded PLGA with HM to produce the nanoparticles, IR780@PLGA@HM, which had tumor targeting, immune regulating, and Sonodynamic abilities. Experiments showed that the IR780@PLGA@HM nanoparticles had good biocompatibility, effectively targeted to 4T1 tumors, promoted macrophage type I polarization and DC cells activation, strengthened anti-tumor inflammatory factors expression, and presented the ability to effectively kill tumors both in vitro and in vivo, which showed a promising therapeutic effect on breast cancer bone metastasis. Therefore, the nanoparticles we constructed provided a new strategy for effectively treating breast cancer bone metastasis.
    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
    尽管出现了新的诊断,药理学,干预和预防策略,动脉粥样硬化性心血管疾病仍然是发病率和死亡率的重要原因。基于纳米粒子的平台包括不同的成像,递送和药理学特性,为在细胞和分子水平上完善动脉粥样硬化的诊断和治疗干预措施提供了新的机会。巨噬细胞在动脉粥样硬化中起关键作用,因此是一个重要的疾病相关诊断和治疗靶点。特别是考虑到它们固有的被动和主动纳米粒子摄取能力。在这次审查中,我们讨论一系列无机物,碳基和脂基纳米颗粒提供磁性,射线照相和荧光成像能力,用于动脉粥样硬化的一系列非常有前途的研究和临床应用。我们讨论了针对一系列巨噬细胞相关功能的纳米颗粒的设计,例如脂蛋白氧化,胆固醇流出,血管炎症和有缺陷的红细胞增多症。我们还提供了为其他病理如癌症开发的纳米颗粒系统的例子,并强调了它们在心血管疾病中的再利用潜力。最后,我们讨论了目前的状态和纳米粒子的未来。虽然这并非没有挑战,纳米颗粒设计中可能具有的多功能功能阵列确保了它们将成为下一个令人兴奋的新疗法前沿的一部分,这些新疗法可以同时提高斑块诊断的准确性,并在副作用有限的情况下更有效地减少动脉粥样硬化.
    Despite the emergence of novel diagnostic, pharmacological, interventional, and prevention strategies, atherosclerotic cardiovascular disease remains a significant cause of morbidity and mortality. Nanoparticle (NP)-based platforms encompass diverse imaging, delivery, and pharmacological properties that provide novel opportunities for refining diagnostic and therapeutic interventions for atherosclerosis at the cellular and molecular levels. Macrophages play a critical role in atherosclerosis and therefore represent an important disease-related diagnostic and therapeutic target, especially given their inherent ability for passive and active NP uptake. In this review, we discuss an array of inorganic, carbon-based, and lipid-based NPs that provide magnetic, radiographic, and fluorescent imaging capabilities for a range of highly promising research and clinical applications in atherosclerosis. We discuss the design of NPs that target a range of macrophage-related functions such as lipoprotein oxidation, cholesterol efflux, vascular inflammation, and defective efferocytosis. We also provide examples of NP systems that were developed for other pathologies such as cancer and highlight their potential for repurposing in cardiovascular disease. Finally, we discuss the current state of play and the future of theranostic NPs. Whilst this is not without its challenges, the array of multifunctional capabilities that are possible in NP design ensures they will be part of the next frontier of exciting new therapies that simultaneously improve the accuracy of plaque diagnosis and more effectively reduce atherosclerosis with limited side effects.
    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
    背景:根尖周炎治疗过程中面临的主要问题是细菌感染的处理和促进牙槽骨缺损的修复以缩短病程。传统的根管冲洗剂的功效有限,并伴有多种副作用。本研究介绍了一种基于一氧化氮(NO)和抗菌光动力疗法(aPDT)的协同疗法,用于治疗根尖周炎。
    结果:这项研究开发了一种多功能纳米粒子,CGP,以胍基聚乙二醇-聚ε-己内酯聚合物为载体,内部装有光敏剂二氢萘e6。在根管冲洗期间,CGP表面的胍基能够有效地穿透生物膜。这些基团在aPDT过程中被过氧化氢氧化,触发NO的释放而不阻碍单线态氧的产生。产生的NO显著增强了aPDT的抗菌能力和生物膜根除功效。此外,CGP不仅在根除生物膜方面优于常规aPDT,而且还能有效促进根除后牙槽骨缺损的修复。重要的是,我们的研究结果表明,与次氯酸钠相比,CGP表现出更高的生物安全性,在根尖周炎大鼠模型中具有出色的治疗效果。
    结论:这项研究表明,CGP,基于aPDT和NO的有效根系灌溉系统,在根管治疗中具有广阔的应用前景。
    BACKGROUND: The main issues faced during the treatment of apical periodontitis are the management of bacterial infection and the facilitation of the repair of alveolar bone defects to shorten disease duration. Conventional root canal irrigants are limited in their efficacy and are associated with several side effects. This study introduces a synergistic therapy based on nitric oxide (NO) and antimicrobial photodynamic therapy (aPDT) for the treatment of apical periodontitis.
    RESULTS: This research developed a multifunctional nanoparticle, CGP, utilizing guanidinylated poly (ethylene glycol)-poly (ε-Caprolactone) polymer as a carrier, internally loaded with the photosensitizer chlorin e6. During root canal irrigation, the guanidino groups on the surface of CGP enabled effective biofilm penetration. These groups undergo oxidation by hydrogen peroxide in the aPDT process, triggering the release of NO without hindering the production of singlet oxygen. The generated NO significantly enhanced the antimicrobial capability and biofilm eradication efficacy of aPDT. Furthermore, CGP not only outperforms conventional aPDT in eradicating biofilms but also effectively promotes the repair of alveolar bone defects post-eradication. Importantly, our findings reveal that CGP exhibits significantly higher biosafety compared to sodium hypochlorite, alongside superior therapeutic efficacy in a rat model of apical periodontitis.
    CONCLUSIONS: This study demonstrates that CGP, an effective root irrigation system based on aPDT and NO, has a promising application in root canal therapy.
    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
    牙周炎是由宿主免疫系统和牙菌斑微生物群之间复杂的相互作用引起的炎症性疾病。牙周炎引起的氧化应激和炎症微环境是导致疾病进展的主要因素。此外,牙菌斑微生物群的存在在影响病情中起着重要作用。因此,牙周炎的治疗策略应该是多方面的。在这项研究中,通过用苯基硼酸频哪醇酯(PBAP)在结构上修饰透明质酸(HA),开发了一种活性氧(ROS)响应的药物递送系统。将姜黄素(CUR)包封在该药物递送系统中以形成载有姜黄素的纳米颗粒(HA@CUR纳米颗粒)。释放结果表明,CUR可以在ROS环境中快速释放,达到治疗所需的浓度。就吸收而言,HA可以有效地增强NPs的细胞摄取,因为它特异性识别正常细胞表达的CD44。此外,HA@CURNP不仅保留了CUR的抗菌功效,而且在体内和体外都表现出更明显的抗炎和抗氧化应激功能。这为牙周炎的治疗提供了良好的潜在药物递送系统,并且可以为针对牙周病的牙科治疗提供有价值的见解。
    Periodontitis is an inflammatory disease induced by the complex interactions between the host immune system and the microbiota of dental plaque. Oxidative stress and the inflammatory microenvironment resulting from periodontitis are among the primary factors contributing to the progression of the disease. Additionally, the presence of dental plaque microbiota plays a significant role in affecting the condition. Consequently, treatment strategies for periodontitis should be multi-faceted. In this study, a reactive oxygen species (ROS)-responsive drug delivery system was developed by structurally modifying hyaluronic acid (HA) with phenylboronic acid pinacol ester (PBAP). Curcumin (CUR) was encapsulated in this drug delivery system to form curcumin-loaded nanoparticles (HA@CUR NPs). The release results indicate that CUR can be rapidly released in a ROS environment to reach the concentration required for treatment. In terms of uptake, HA can effectively enhance cellular uptake of NPs because it specifically recognizes CD44 expressed by normal cells. Moreover, HA@CUR NPs not only retained the antimicrobial efficacy of CUR, but also exhibited more pronounced anti-inflammatory and anti-oxidative stress functions both in vivo and in vitro. This provides a good potential drug delivery system for the treatment of periodontitis, and could offer valuable insights for dental therapeutics targeting periodontal diseases.
    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
    多功能纳米颗粒对于协同多峰抗肿瘤活性具有重要意义。在这里,氧化锌(ZnO)被用作pH敏感的纳米颗粒,用于加载化疗剂阿霉素(DOX)和光敏剂吲哚菁绿(ICG),生物相容性低分子量肝素(LMHP)被用作协同光热疗法/光动力疗法/化学疗法/免疫疗法的看门人。ZnO被分解为细胞毒性的Zn2+离子,导致ICG和DOX的肿瘤特异性释放。ZnO同时产生氧气(O2)和活性氧(ROS)用于光动力疗法(PDT)。在激光照射下释放的ICG产生了用于PDT的ROS,并提高了用于光热治疗(PTT)的肿瘤温度。释放的DOX直接导致化疗的肿瘤细胞死亡。DOX和ICG也诱导免疫疗法的免疫原性细胞死亡(ICD)。体内和体外结果对肿瘤进展具有优异的抑制作用,转移和复发。因此,本研究可为设计用于协同多模式抗肿瘤治疗的多功能纳米颗粒提供有效方法。
    Multifunctional nanoparticles are of significant importance for synergistic multimodal antitumor activity. Herein, zinc oxide (ZnO) was used as pH-sensitive nanoparticles for loading the chemotherapy agent doxorubicin (DOX) and the photosensitizer agent indocyanine green (ICG), and biocompatible low-molecular-weight heparin (LMHP) was used as the gatekeepers for synergistic photothermal therapy/photodynamic therapy/chemotherapy/immunotherapy. ZnO was decomposed into cytotoxic Zn2+ ions, leading to a tumor-specific release of ICG and DOX. ZnO simultaneously produced oxygen (O2) and reactive oxygen species (ROS) for photodynamic therapy (PDT). The released ICG under laser irradiation produced ROS for PDT and raised the tumor temperature for photothermal therapy (PTT). The released DOX directly caused tumor cell death for chemotherapy. Both DOX and ICG also induced immunogenic cell death (ICD) for immunotherapy. The in vivo and in vitro results presented a superior inhibition of tumor progression, metastasis and recurrence. Therefore, this study could provide an efficient approach for designing multifunctional nanoparticles for synergistic multimodal antitumor therapy.
    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
    结合现有的药物治疗对于开发新的疾病预防和治疗治疗剂至关重要。在临床前调查中,某些已知药物的联合作用已经在治疗广泛的人类疾病中得到了很好的证实。归因于通过靶向各种疾病途径和优势的协同作用,例如减少给药剂量,降低毒性,并减轻耐药性,目前正在通过提供治疗药物来对抗重大临床疾病,比如癌症,动脉粥样硬化,肺动脉高压,心肌炎,类风湿性关节炎,炎症性肠病,代谢紊乱和神经退行性疾病。组合疗法涉及组合或共同递送两种或更多种用于治疗特定疾病的药物。纳米颗粒(NP)介导的药物递送系统,即,脂质体NP,聚合物NP和纳米晶体,对靶向药物递送引起的各种疾病的组合疗法非常感兴趣,延长药物释放,和更高的药物稳定性,以避免在感染区域快速清除。这篇综述总结了各种疾病的目标,临床前或临床批准的药物组合以及用于组合治疗的多功能NP的开发,并强调基于药物递送的组合治疗策略,以治疗严重的临床疾病。最终,我们讨论了开发NP-共同交付和翻译的挑战,并提供了解决这些限制的潜在方法。这篇综述为开发NP介导的人类疾病联合疗法提供了最新的前沿和挑战性的全面概述。
    Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
    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
    近年来,光热疗法(PTT)作为一种新兴的癌症治疗方式获得了相当大的兴趣。放射治疗(RT)作为一种传统的治疗方法已广泛应用于临床。然而,RT和PTT治疗受到副作用和穿透深度的限制,分别。此外,肿瘤内的缺氧会导致对治疗的抵抗力增加。
    我们通过调节反应条件合成了多种尺寸的AuPt。选择最小尺寸的AuPt并用叶酸(FA)修饰用于PTT和RT协同治疗。各种方法,包括透射电子显微镜(TEM),X射线光电子能谱(XPS),X射线衍射(XRD)和傅里叶变换红外光谱(FITR)用于确定AuPt-FA(AF)的结构和组成。此外,我们用红外相机和红外激光器研究了AF的光热特性。流式细胞术,集落形成试验,CCK8,并进行荧光染色以探查体外处理效果。此外,我们通过TEM和体内成像系统(IVIS)探索了AF的靶向性。体内实验,我们记录肿瘤体积和重量的变化以及肿瘤切片的染色(ROS,Ki67和苏木精和伊红)。
    与其他尺寸的AuPt(30nm和100nm)相比,粒径为16nm的AuPt赋予其显著高的光热转化效率(46.84%)和过氧化氢酶活性。AF缓解肿瘤微环境中的缺氧,导致在治疗过程中产生更多的活性氧(ROS)。此外,联合RT和PTT治疗效果显着增强,体外凋亡率为81.1%,体内肿瘤体积缩小率为94.0%。
    这些结果表明,AF增强了PTT和RT的协同作用,并具有临床转化的潜力。
    UNASSIGNED: Photothermal therapy (PTT) has gained considerable interest as an emerging modality for cancer treatment in recent years. Radiation therapy (RT) has been widely used in the clinic as a traditional treatment method. However, RT and PTT treatments are limited by side effects and penetration depth, respectively. In addition, hypoxia within the tumor can lead to increased resistance to treatment.
    UNASSIGNED: We synthesized multiple sizes of AuPt by modulating the reaction conditions. The smallest size of AuPt was selected and modified with folic acid (FA) for PTT and RT synergy therapy. Various methods including transmission electron microscope (TEM), X-ray photoelectron spectroscopy (XPS), X-ray diffraction (XRD), and Fourier transform infrared spectroscopy (FITR) are used to determine the structure and composition of AuPt-FA (AF). In addition, we researched the photothermal properties of AF with IR cameras and infrared lasers. Flow cytometry, colony formation assays, CCK8, and fluorescent staining for probing the treatment effect in vitro. Also, we explored the targeting of AF by TEM and In Vivo Imaging Systems (IVIS). In vivo experiments, we record changes in tumor volume and weight as well as staining of tumor sections (ROS, Ki67, and hematoxylin and eosin).
    UNASSIGNED: The AuPt with particle size of 16 nm endows it with remarkably high photothermal conversion efficiency (46.84%) and catalase activity compared to other sizes of AuPt (30 nm and 100 nm). AF alleviates hypoxia in the tumor microenvironment, leading to the production of more reactive oxygen species (ROS) during the treatment. In addition, the therapeutic effect was significantly enhanced by combining RT and PTT, with an apoptosis rate of 81.1% in vitro and an in vivo tumor volume reduction rate of 94.0% in vivo.
    UNASSIGNED: These results demonstrate that AF potentiates the synergistic effect of PTT and RT and has the potential for clinical translation.
    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
    背景:基于Fenton/Fenton样反应的化学动力学治疗(CDT)已成为一种有前途的癌症治疗策略。然而,内源性谷胱甘肽(GSH)引起的肿瘤微环境(TME)的强抗氧化特性仍然严重阻碍CDT的有效性。基于核@壳结构的传统CDT纳米平台具有不同亚基的固有干扰,从而阻碍了整体的治疗效率。因此,迫切需要构建具有分离的功能单元和GSH消耗能力的新型结构以实现期望的联合CDT治疗效率。
    结果:这里,提出了一种表面曲率诱导的定向组装策略,以合成由两个功能单元组成的类似寿司的新型Janus治疗纳米平台,作为CDT亚基的FeOOH纳米纺锤体和作为载药亚基的mSiO2纳米棒。FeOOHCDT亚基沿其长轴被mSiO2纳米棒覆盖了一半,形成寿司状结构。FeOOH纳米轴的长度约为400nm,直径为50nm,mSiO2纳米棒的长度约为550nm,直径为100nm。mSiO2亚基的长度和直径可以在宽范围内调节,同时保持寿司状的Janus结构,这归因于吉布斯自由能主导的表面曲率诱导的定向装配过程。在这个Janus治疗纳米平台中,FeOOH中的Fe3+首先被内源性GSH还原为Fe2+,然后,生成的Fe2有效地催化TME中过表达的H2O2转化为高致死性·OH,以实现有效的CDT。负载在mSiO2亚基中的多柔比星(DOX)可以被释放以实现联合化疗。利用与Fe3相关的GSH消耗,与Fe2相关的增强·OH生成,和DOX诱导的化疗,合成的纳米平台具有优异的治疗效率,肿瘤细胞的体外清除效率高达约87%。体内实验也显示了对肿瘤的有效抑制,验证合成的寿司状Janus纳米颗粒作为有前途的治疗纳米平台。
    结论:一般来说,我们的工作为构建新型治疗性纳米平台以实现有效的肿瘤抑制提供了成功的范例.
    BACKGROUND: Chemodynamic therapy (CDT) based on Fenton/Fenton-like reaction has emerged as a promising cancer treatment strategy. Yet, the strong anti-oxidation property of tumor microenvironment (TME) caused by endogenous glutathione (GSH) still severely impedes the effectiveness of CDT. Traditional CDT nanoplatforms based on core@shell structure possess inherent interference of different subunits, thus hindering the overall therapeutic efficiency. Consequently, it is urgent to construct a novel structure with isolated functional units and GSH depletion capability to achieve desirable combined CDT therapeutic efficiency.
    RESULTS: Herein, a surface curvature-induced oriented assembly strategy is proposed to synthesize a sushi-like novel Janus therapeutic nanoplatform which is composed of two functional units, a FeOOH nanospindle serving as CDT subunit and a mSiO2 nanorod serving as drug-loading subunit. The FeOOH CDT subunit is half covered by mSiO2 nanorod along its long axis, forming sushi-like structure. The FeOOH nanospindle is about 400 nm in length and 50 nm in diameter, and the mSiO2 nanorod is about 550 nm in length and 100 nm in diameter. The length and diameter of mSiO2 subunit can be tuned in a wide range while maintaining the sushi-like Janus structure, which is attributed to a Gibbs-free-energy-dominating surface curvature-induced oriented assembly process. In this Janus therapeutic nanoplatform, Fe3+ of FeOOH is firstly reduced to Fe2+ by endogenous GSH, the as-generated Fe2+ then effectively catalyzes overexpressed H2O2 in TME into highly lethal ·OH to achieve efficient CDT. The doxorubicin (DOX) loaded in the mSiO2 subunit can be released to achieve combined chemotherapy. Taking advantage of Fe3+-related GSH depletion, Fe2+-related enhanced ·OH generation, and DOX-induced chemotherapy, the as-synthesized nanoplatform possesses excellent therapeutic efficiency, in vitro eliminating efficiency of tumor cells is as high as ~ 87%. In vivo experiments also show the efficient inhibition of tumor, verifying the synthesized sushi-like Janus nanoparticles as a promising therapeutic nanoplatform.
    CONCLUSIONS: In general, our work provides a successful paradigm of constructing novel therapeutic nanoplatform to achieve efficient tumor inhibition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号