thermo-sensitive liposome

  • 文章类型: Journal Article
    光热疗法(PTT)是乳腺癌的前瞻性治疗方法。然而,PTT诱导的过度炎症反应可能加重肿瘤转移。同时,癌细胞过度表达的热休克蛋白(HSPs)可以保护它们免受PTT期间的高温。因此,减轻PTT诱导的炎症和抑制肿瘤转移,开发了叶酸受体靶向的热敏脂质体(BI-FA-LP)共负载小檗碱(BBR)和吲哚菁绿(ICG)。BI-FA-LP利用增强的通透性和保留(EPR)效应和FA受体介导的内吞作用选择性地在肿瘤中积累,减少治疗期间的脱靶毒性。靶向肿瘤部位后,BBR和ICG在激光照射后从BI-FA-LP释放,ICG表现出良好的光热性能,而BBR在PTT期间抑制HSP70和HSP90的表达,发挥化学-光热协同抗肿瘤作用。此外,BBR可以抑制PTT诱导的炎症,从而抑制肿瘤转移,减轻组织损伤。因此,这种多功能脂质体为乳腺癌治疗提供了一种增强PTT和抗炎作用的新策略.
    Photothermal therapy (PTT) is a prospective therapeutic method for breast cancer. However, excess inflammatory response induced by PTT may aggravate tumor metastasis. Meanwhile, the overexpressed heat shock proteins (HSPs) by cancer cells can protect them from hyperthermia during PTT. Therefore, to attenuate the PTT-induced inflammation and inhibit tumor metastasis, a folate receptor-targeted thermo-sensitive liposome (BI-FA-LP) co-loading Berberine (BBR) and Indocyanine green (ICG) was developed. BI-FA-LP utilized enhanced permeability and retention (EPR) effect and FA receptor-mediated endocytosis to selectively accumulate at tumor, reducing off-target toxicity during the treatment. After targeting to the tumor site, BBR and ICG were released from BI-FA-LP upon laser irradiation, and ICG showed good photothermal performance, while BBR inhibited HSP70 and HSP90 expression during PTT, exerting chemo-photothermal synergetic anti-tumor effect. Moreover, BBR could suppress the PTT induced inflammation, thus inhibiting tumor metastasis and ameliorating tissue injury. Thus, this versatile liposome provided a new strategy to enhance PTT and anti-inflammatory effects for breast 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
    聚焦超声(FUS)触发的纳米级药物输送,作为治疗实体瘤的智能刺激响应系统,进行了计算研究,以增强药物的局部递送和治疗功效。热敏脂质体(TSL)的整合,作为负载多柔比星(DOX)的纳米载体,FUS,提供了一种有前途的药物递送系统。完全耦合的偏微分方程组,包括FUS传播的Helmholtz方程,生物传热,间质液流量,药物在组织和细胞空间的运输,首先提出了这种治疗方法的药效学模型。然后通过有限元方法求解方程以计算细胞内药物浓度和治疗功效。这项研究的主要目的是提出一个多物理和多尺度模型来模拟药物释放,运输,并运送到实体瘤,然后分析FUS暴露时间和药物释放速率如何影响这些过程。我们的发现不仅显示了模型复制这种治疗方法的能力,但也证实了这种治疗的好处,改善了肿瘤中的药物聚集和减少了健康组织中的药物递送。例如,治疗后肿瘤细胞的存活率下降到62.4%,因为大量的药物被输送到癌细胞。接下来,三种释放速率的组合(超快,快,和缓慢)和FUS暴露时间(10、30和60分钟)进行检查。曲线下面积(AUC)结果表明,30分钟FUS暴露和快速药物释放的组合导致实际和有效的治疗反应。
    Focused Ultrasound (FUS)-triggered nano-sized drug delivery, as a smart stimuli-responsive system for treating solid tumors, is computationally investigated to enhance localized delivery of drug and treatment efficacy. Integration of thermosensitive liposome (TSL), as a doxorubicin (DOX)-loaded nanocarrier, and FUS, provides a promising drug delivery system. A fully coupled partial differential system of equations, including the Helmholtz equation for FUS propagation, bio-heat transfer, interstitial fluid flow, drug transport in tissue and cellular spaces, and a pharmacodynamic model is first presented for this treatment approach. Equations are then solved by finite element methods to calculate intracellular drug concentration and treatment efficacy. The main objective of this study is to present a multi-physics and multi-scale model to simulate drug release, transport, and delivery to solid tumors, followed by an analysis of how FUS exposure time and drug release rate affect these processes. Our findings not only show the capability of model to replicate this therapeutic approach, but also confirm the benefits of this treatment with an improvement of drug aggregation in tumor and reduction of drug delivery in healthy tissue. For instance, the survival fraction of tumor cells after this treatment dropped to 62.4%, because of a large amount of delivered drugs to cancer cells. Next, a combination of three release rates (ultrafast, fast, and slow) and FUS exposure times (10, 30, and 60 min) was examined. Area under curve (AUC) results show that the combination of 30 min FUS exposure and rapid drug release leads to a practical and effective therapeutic response.
    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
    计算模型已被开发为识别实验模型无法正确理解的生物相互作用的潜在平台。在本研究中,一个数学模型已被用于研究载药热敏脂质体(TSL)在血管内释放模式后的治疗反应.由交变磁场产生的热场用于在微血管内释放药物。确定应用磁热疗法所需的时间是本研究的主要目的。结果表明,应用长期连续或脉冲热疗可以影响细胞外间隙中药物的浓度水平。对于所有热疗程序,细胞外空间中游离和结合药物浓度的峰值都相等。此外,与连续模式相比,脉冲模式下游离和结合药物的浓度保持在更高的水平(即,脉冲病例的曲线下面积(AUC)略高于连续病例)。然而,生物利用时间没有显著差异。因此,不同条件下肿瘤生长的开始时间相似。这项研究表明,施加热疗的适当时间是推注后,直到达到细胞外空间的峰值浓度曲线。因此,在与本研究类似的临床应用中,持续热疗30min是较好的选择。这项研究可以成为实验研究的有用指南,以减少体内测试的数量,以及为临床试验做出正确的决定,以提供最佳的药物方案。
    Computational models have been developed as a potential platform to identify bio-interactions that cannot be properly understood by experimental models. In the present study, a mathematical model has been employed to investigate the therapeutic response of drug-loaded thermosensitive liposome (TSL) following intravascular release paradigm. Thermal field created by an alternating magnetic field is utilized to release the drug within microvessels. Determining the time required for the application of magneto-hyperthermia is the main purpose of this study. Results show that applying a long-term continuous or pulsed hyperthermia can affect the concentration level of drugs in the extracellular space. The peak value of free and bound drug concentrations in the extracellular space is equal for all hyperthermia programs. Additionally, the concentrations of free and bound drugs are retained at a higher level in pulsed mode compared to the continuous mode (i.e., area under curve (AUC) of pulsed case is slightly higher than continuous case). However, there is no significant difference in bioavailability time. Hence, onset time of tumor growth is similar for different conditions. This study shows that the appropriate time to apply hyperthermia is post-bolus injection until reaching the peak concentration profile in extracellular space. Therefore, in clinical applications similar to the present study\'s circumstances, continuous hyperthermia for 30 min can be a better choice. This study can be a useful guideline for experimental studies to reduce the number of in vivo tests as well as for clinical trials to make the right decision to provide optimal medication programs.
    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
    Chemotherapy is an important modality available for cancer treatment. However, the present chemotherapy is still far from being satisfactory mainly owing to the severe side effects of the chemotherapeutic agents and drug resistance of cancer cells. Thus, reversing drug resistance by constructing an ideal chemotherapeutic strategy with the least side effects and the best efficacy is greatly needed. Here, we designed a smart nanosystem of thermo-sensitive liposome coated gold nanocages with doxorubicin (DOX) loading (LAD) for near-infrared (NIR)-triggered drug release and chemo-photothermal combination therapy. The biocompatible liposomes coating facilitated the cellular uptake of LAD and meanwhile avoided drug leakage during the circulation. More importantly, LAD exhibited controllable photothermal conversion property and produced mild heat under NIR irradiation, which not only triggered DOX release and transferred DOX from lysosome to nucleus, but also elicited the mild heat cell killing effect to improve the curative efficiency. Further mechanism study revealed that mild heat could reverse drug resistance by down-regulation of the chemoresistance-related markers (e.g., HSF-1, p53, P-gp), and inhibited DOX export and increased drug sensitiveness, thereby prominently increased the anticancer efficiency. This versatile nanoplatform with enhanced curative efficacy and lower side effect is promising to apply in the field of drug controlled release and combination tumor therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    To develop vincristine (VCR) and doxorubicin (DOX) co-encapsulated thermo-sensitive liposomes (VD-TSL) against drug resistance, with increased tumor inhibition rate and decreased system toxicity, improving drug targeting efficiency upon mild hyperthermia (HT) in solid tumor.
    Based on similar physicochemical properties, VCR and DOX were co-loaded in TSL with pH gradient active loading method and characterized. The time-dependent drug release profiles at 37 and 42°C were assessed by HPLC. Then we analysed the phospholipids in filtrate after ultrafiltration and studied VD-TSL stability in mimic in vivo conditions and long-time storage conditions (4°C and -20°C). Cytotoxic effect was studied on PANC and sw-620 using MTT. Intracellular drug delivery was studied by confocal microscopy on HT-1080. In vivo imaging of TSL pharmacokinetic and biodistribution was performed on MCF-7 tumor-bearing nude mice. And therapeutic efficacy on these xenograft models were followed under HT.
    VD-TSL had excellent particle distribution (about 90 nm), high entrapment efficiency (>95%), obvious thermo-sensitive property, and good stability. MTT proved VD-TSL had strongest cell lethality compared with other formulations. Confocal microscopy demonstrated specific accumulation of drugs in tumor cells. In vivo imaging proved the targeting efficiency of TSL under hyperthermia. Then therapeutic efficacy revealed synergism of VCR and DOX co-loaded in TSL, together with HT.
    VD-TSL could increase drug efficacy and decrease system toxicity, by making good use of synergism of VCR and DOX, as well as high targeting efficiency of TSL.
    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