controlled release

控释
  • 文章类型: Journal Article
    卵巢癌,最致命的妇科恶性肿瘤,主要用化疗治疗。然而,全身化疗常导致严重的毒副作用和化疗耐药。载药气凝胶已成为一种有前途的药物递送方法,因为它们可以提高药物的溶解度和生物利用度,控制药物释放,减少药物在非靶向组织中的分布,从而最大限度地减少副作用。在这项研究中,使用纳米纤维重组和冷冻干燥技术创建了具有多孔网络结构的壳聚糖寡糖(COS)负载的纳米纤维复合壳聚糖(CS)气凝胶(COS-NFs/CS)。气凝胶的芯层具有60%的COS负载率,使COS-NFs/CS气凝胶能够显着抑制卵巢癌细胞的迁移和增殖(导致48小时后卵巢癌细胞的存活率降低至33.70%)。同轴纤维独特的壳-芯结构和气凝胶的多孔网络结构使COS-NFs/CS气凝胶在30天内稳定缓慢地释放COS,有效减少COS的初始爆发释放。此外,COS-NFs/CS气凝胶表现出良好的生物相容性,降解性(植入6周后仅保留其体重的18.52%),促进血管生成,从而促进卵巢切除术后伤口愈合。总之,COS-NFs/CS气凝胶在卵巢癌治疗中显示出巨大的应用潜力。
    Ovarian cancer, the deadliest gynecological malignancy, primarily treated with chemotherapy. However, systemic chemotherapy often leads to severe toxic side effects and chemoresistance. Drug-loaded aerogels have emerged as a promising method for drug delivery, as they can improve drug solubility and bioavailability, control drug release, and reduce drug distribution in non-targeted tissues, thereby minimizing side effects. In this research, chitosan oligosaccharide (COS)-loaded nanofibers composite chitosan (CS) aerogels (COS-NFs/CS) with a porous network structure were created using nanofiber recombination and freeze-drying techniques. The core layer of the aerogel has a COS loading rate of 60 %, enabling the COS-NFs/CS aerogel to significantly inhibit the migration and proliferation of ovarian cancer cells (resulting in a decrease in the survival rate of ovarian cancer cells to 33.70 % after 48 h). The coaxial fiber\'s unique shell-core structure and the aerogel\'s porous network structure enable the COS-NFs/CS aerogels to release COS steadily and slowly over 30 days, effectively reducing the initial burst release of COS. Additionally, the COS-NFs/CS aerogels exhibit good biocompatibility, degradability (only retaining 18.52 % of their weight after 6 weeks of implantation), and promote angiogenesis, thus promoting wound healing post-oophorectomy. In conclusion, COS-NFs/CS aerogels show great potential for application in the treatment of ovarian 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
    后段疾病是不可逆视力障碍的主要原因。后段疾病的成功治疗需要将治疗物质有效地递送到目标病变。然而,复杂的眼部结构使得局部应用药物的生物利用度非常低。侵入性递送方法如玻璃体内注射可引起不良并发症。为了提高效率,已经开发了几种生物医学工程系统来提高药物在后段的渗透效率和生物利用度。有利的是,发现可生物降解的微球以受控的方式递送治疗剂。由新型生物材料制备的微球可以在后段实现延长释放,副作用最小。此外,它将自动降解为对人体无毒的产品,而无需进行二次操作以去除残留的聚合物基质。此外,可生物降解的微球具有良好的热塑性,可调节的亲水性,控制结晶度,和高抗拉强度,这使得它们适合眼内递送。在这次审查中,我们介绍了微球生产技术的最新进展,并详细介绍了用于制备微球的生物材料。我们系统地讨论了生物可降解微球的药理特性,并比较了它们在治疗后段疾病中的潜在优势和局限性。这些发现将丰富我们对生物可降解微球的知识,并有助于发现有效的眼部药物递送生物材料。
    Posterior segment disease acts as a major cause of irreversible visual impairments. Successful treatment of posterior segment disease requires the efficient delivery of therapeutic substances to the targeted lesion. However, the complex ocular architecture makes the bioavailability of topically applied drugs extremely low. Invasive delivery approaches like intravitreal injection may cause adverse complications. To enhance the efficiency, several biomedical engineering systems have been developed to increase the penetration efficiency and improve the bioavailability of drugs at the posterior segments. Advantageously, biodegradable microspheres are found to deliver the therapeutic agents in a controlled fashion. The microspheres prepared from novel biomaterials can realize the prolonged release at the posterior segment with minimum side effects. Moreover, it will be degraded automatically into products that are non-toxic to the human body without the necessity of secondary operation to remove the residual polymer matrix. Additionally, biodegradable microspheres have decent thermoplasticity, adjustable hydrophilicity, controlled crystallinity, and high tensile strength, which make them suitable for intraocular delivery. In this review, we introduce the latest advancements in microsphere production technology and elaborate on the biomaterials that are used to prepare microspheres. We discuss systematically the pharmacological characteristics of biodegradable microspheres and compare their potential advantages and limitations in the treatment of posterior segment diseases. These findings would enrich our knowledge of biodegradable microspheres and cast light into the discovery of effective biomaterials for ocular drug delivery.
    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
    骨再生是一个协调良好的过程,涉及炎症,血管生成,和成骨。因此,有效的骨移植物应设计为针对骨愈合过程中的多种分子事件和生物学需求。在这项研究中,基于天然生物事件开发了一种基于明胶甲基丙烯酰(GelMA)的可生物降解Janus微球递送系统,该系统包含磷酸钙低聚物(CPO)和骨形态发生蛋白2(BMP-2)。GelMA的特殊可调性促进了生物分子的受控释放和按需应用,并探索了CPO和BMP-2的优化递送曲线。在初始愈合阶段持续释放CPO有助于早期免疫调节并促进晚期矿化。同时,在治疗范围内以相对较高的浓度施用BMP-2增强了骨诱导性质。这个传送系统,通过微调的释放模式,诱导M2巨噬细胞极化并创建有益的免疫微环境,这反过来又促进了体内有效的骨再生。总的来说,这项研究提出了一个自下而上的概念,旨在开发针对单个生物事件的用户友好且易于控制的递送系统,这可能为开发用于临床的功能优化的生物材料提供新的视角。
    Bone regeneration is a well-orchestrated process synergistically involving inflammation, angiogenesis, and osteogenesis. Therefore, an effective bone graft should be designed to target multiple molecular events and biological demands during the bone healing process. In this study, a biodegradable gelatin methacryloyl (GelMA)-based Janus microsphere delivery system containing calcium phosphate oligomer (CPO) and bone morphogenetic protein-2 (BMP-2) is developed based on natural biological events. The exceptional adjustability of GelMA facilitates the controlled release and on-demand application of biomolecules, and optimized delivery profiles of CPO and BMP-2 are explored. The sustained release of CPO during the initial healing stages contributes to early immunomodulation and promotes mineralization in the late stage. Meanwhile, the administration of BMP-2 at a relatively high concentration within the therapeutic range enhances the osteoinductive property. This delivery system, with fine-tuned release patterns, induces M2 macrophage polarization and creates a conducive immuno-microenvironment, which in turn facilitates effective bone regeneration in vivo. Collectively, this study proposes a bottom-up concept, aiming to develop a user-friendly and easily controlled delivery system targeting individual biological events, which may offer a new perspective on developing function-optimized biomaterials for clinical use.
    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
    尽管免疫检查点阻断(ICB)疗法在治疗各种癌症类型方面具有相当大的潜力,它面临着几个挑战,其中在癌症患者中观察到的受约束的客观反应率和相对较短的反应持续时间是最重要的。这项研究介绍了一种可注射的温度敏感水凝胶,PluronicF-127(PF-127)@MnCl2/藻酸盐微球(ALG-MS)@MgCl2,可增强癌细胞中程序性细胞死亡配体1(PD-L1)的治疗效果。本研究中使用的水凝胶材料促进了肿瘤微环境中大量锰离子(Mn2)的快速释放以及镁离子(Mg2)的逐渐和持续释放。这种分阶段的释放曲线促进有利于CD8+T细胞和自然杀伤细胞的细胞毒性的免疫微环境,从而提高ICB治疗的疗效。此外,PF-127@MnCl2/ALG-MS@MgCl2复合水凝胶具有将低PD-L1反应的耐药肿瘤(“冷肿瘤”)转化为高PD-L1反应的“热肿瘤”的能力。总之,PF-127@MnCl2/ALG-MS@MgCl2水凝胶通过Mg2+和Mn2+的精确放电来操纵免疫微环境,因此,增强ICB治疗的疗效。
    Despite the considerable potential of immune checkpoint blockade (ICB) therapy in treating various cancer types, it faces several challenges, of which the constrained objective response rate and relatively short duration of response observed in patients with cancer are the most important. This study introduces an injectable temperature-sensitive hydrogel, Pluronic F-127 (PF-127)@MnCl2/ alginate microspheres (ALG-MS)@MgCl2, that enhances the therapeutic efficacy of programmed cell death-ligand 1 (PD-L1) in cancer cells. The hydrogel material used in this study facilitated the rapid release of a significant amount of manganese ions (Mn2+) and the gradual and sustained release of magnesium ions (Mg2+) within the tumor microenvironment. This staged release profile promotes an immune microenvironment conducive to the cytotoxicity of CD8+ T cells and natural killer cells, thereby enhancing the efficacy of ICB therapy. Furthermore, the PF-127@MnCl2/ALG-MS@MgCl2 composite hydrogel exhibits the ability to convert drug-resistant tumor (\"cold tumor\") with a low PD-L1 response to a \"hot tumor\" with a high PD-L1 response. In summary, the PF-127@MnCl2/ALG-MS@MgCl2 hydrogel manipulates the immune microenvironment through the precise discharge of Mg2+ and Mn2+, thus, augmenting the efficacy of ICB 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
    莱茵,一种天然的蒽醌化合物,来源于中药,通过调节活性氧或氮(RONS)的水平表现出有效的抗炎特性。然而,它在水中的溶解度有限,血浆存在的短暂持续时间,以及其显著的全身毒性,对其在体内的使用构成障碍,需要创建可靠的药物递送平台来规避这些困难。在这项研究中,通过将大黄酸与聚乙二醇(PEG)修饰的三苯基鳞(TPP)分子结合,合成了酯酶响应性和线粒体靶向的纳米前药,形成TPP-PEG-RH,当分散在水介质中时,它可以自发地自组装成RPTNP。这些纳米颗粒的TPP外层增强了它们的药代动力学特征,促进向线粒体的有效递送,并促进细胞摄取,从而能够增强线粒体中的积累并改善体外治疗效果。在RPTNP治疗后,在IL-1β刺激的软骨细胞中观察到RONS的下降。RPTNP也发挥优异的抗炎作用(IL-1β,TNF-α,IL-6和MMP-13)和抗氧化作用(Cat和Sod)通过Nrf2信号通路,软骨相关基因(Col2a1和Acan)的上调。此外,RPTNP显示线粒体膜电位的保护和软骨细胞凋亡的抑制。此外,这些发现表明,线粒体靶向聚合物-大黄酸缀合物可能为患有慢性关节疾病的患者提供治疗解决方案,通过减轻骨关节炎(OA)的进展。
    Rhein, a natural anthraquinone compound derived from traditional Chinese medicine, exhibits potent anti-inflammatory properties via modulating the level of Reactive oxygen or nitrogen species (RONS). Nevertheless, its limited solubility in water, brief duration of plasma presence, as well as its significant systemic toxicity, pose obstacles to its in vivo usage, necessitating the creation of a reliable drug delivery platform to circumvent these difficulties. In this study, an esterase-responsive and mitochondria-targeted nano-prodrug was synthesized by conjugating Rhein with the polyethylene glycol (PEG)-modified triphenyl phosphonium (TPP) molecule, forming TPP-PEG-RH, which could spontaneously self-assemble into RPT NPs when dispersed in aqueous media. The TPP outer layer of these nanoparticles enhances their pharmacokinetic profile, facilitates efficient delivery to mitochondria, and promotes cellular uptake, thereby enabling enhanced accumulation in mitochondria and improved therapeutic effects in vitro. The decline in RONS was observed in IL-1β-stimulated chondrocyte after RPT NPs treating. RPT NPs also exert excellent anti-inflammatory (IL-1β, TNF-α, IL-6 and MMP-13) and antioxidative effects (Cat and Sod) via the Nrf2 signalling pathway, upregulation of cartilage related genes (Col2a1 and Acan). Moreover, RPT NPs shows protection of mitochondrial membrane potential and inhibition of chondrocyte apoptosis. Moreover, These findings suggest that the mitochondria-targeted polymer-Rhein conjugate may offer a therapeutic solution for patients suffering from chronic joint disorders, by attenuating the progression of osteoarthritis (OA).
    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
    心肌梗塞(MI)已成为全球心血管疾病发病率的主要原因。MI的发病机制是一个渐进的过程,包括三个关键阶段:炎症,扩散,和重塑。智能刺激响应性水凝胶因其在MI部位精确且可控地递送治疗药物的能力而引起了相当大的关注。这里,设计了一种具有双重交联网络结构的智能刺激响应水凝胶,这使得治疗药物在不同病理阶段的精确和受控释放能够治疗MI。水凝胶可以在MI的炎症阶段快速释放姜黄素(Cur)以发挥抗凋亡/抗炎作用。将重组人源化III型胶原(rhColIII)加载到水凝胶中,并在增殖期期间随着水凝胶膨胀/降解而释放,以促进新血管形成。RepSox(选择性TGF-β抑制剂)在重塑阶段从移植有醛纳米颗粒(PF127-CHO@RepSoxNP)的PluronicF-127释放以对抗纤维化。体外和体内结果表明,水凝胶通过抑制炎症和细胞凋亡改善心脏功能并减轻心脏重塑,促进新生血管形成,抑制心肌纤维化.全程修复系统,利用刺激响应多功能水凝胶,在增强MI后心脏功能和促进受损心肌组织的恢复方面表现出显著的有效性。
    Myocardial infarction (MI) has emerged as the predominant cause of cardiovascular morbidity globally. The pathogenesis of MI unfolds as a progressive process encompassing three pivotal phases: inflammation, proliferation, and remodeling. Smart stimulus-responsive hydrogels have garnered considerable attention for their capacity to deliver therapeutic drugs precisely and controllably at the MI site. Here, a smart stimulus-responsive hydrogel with a dual-crosslinked network structure is designed, which enables the precise and controlled release of therapeutic drugs in different pathological stages for the treatment of MI. The hydrogel can rapidly release curcumin (Cur) in the inflammatory phase of MI to exert anti-apoptotic/anti-inflammatory effects. Recombinant humanized collagen type III (rhCol III) is loaded in the hydrogel and released as the hydrogel swelled/degraded during the proliferative phase to promote neovascularization. RepSox (a selective TGF-β inhibitor) releases from Pluronic F-127 grafted with aldehyde nanoparticles (PF127-CHO@RepSox NPs) in the remodeling phase to against fibrosis. The results in vitro and in vivo suggest that the hydrogel improves cardiac function and alleviates cardiac remodeling by suppressing inflammation and apoptosis, promoting neovascularization, and inhibiting myocardial fibrosis. A whole-course-repair system, leveraging stimulus-responsive multifunctional hydrogels, demonstrates notable effectiveness in enhancing post-MI cardiac function and facilitating the restoration of damaged myocardial tissue.
    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
    直接向肿瘤微环境(TME)供应抗癌药物的药物局部递送系统导致优异的肿瘤控制并使与抗癌药物相关的副作用最小化。免疫检查点抑制剂(ICIs)一直是癌症免疫治疗的支柱。然而,ICIs的全身给药伴随着相当大的免疫治疗相关毒性.探讨通过缓释凝胶形成载体局部施用的抗PD-L1抗体是否保留其有效的抗癌功能,同时引起较少的结肠炎样副作用,CT,以前报道的仓库系统,用于将抗PD-L1抗体与姜黄素一起局部递送至患有膀胱癌的溃疡性结肠炎模型小鼠中的TME。我们表明,CT介导的抗PD-L1抗体和姜黄素的肿瘤内共注射能够持续释放负载的抗PD-L1抗体和姜黄素,这有助于对UC模型小鼠的结肠具有可忽略的副作用的实质性抗癌作用。然而,尽管抗PD-L1抗体与CT介导的姜黄素的肿瘤内递送在抑制肿瘤生长方面具有系统性协同作用,通过腹膜内施用抗PD-L1抗体,结肠炎显著恶化。这些发现表明,CT是抗癌药物局部递送的有前途的药物,因为它可以保留有效的抗癌功能,同时大大减少与这些药物的全身给药相关的不良副作用。
    Drug local delivery system that directly supply anti-cancer drugs to the tumor microenvironment (TME) results in excellent tumor control and minimizes side effects associated with the anti-cancer drugs. Immune checkpoint inhibitors (ICIs) have been the mainstay of cancer immunotherapy. However, the systemic administration of ICIs is accompanied by considerable immunotherapy-related toxicity. To explore whether an anti-PD-L1 antibody administered locally via a sustained-release gel-forming carrier retains its effective anticancer function while causing fewer colitis-like side effects, CT, a previously reported depot system, was used to locally deliver an anti-PD-L1 antibody together with curcumin to the TME in bladder cancer-bearing ulcerative colitis model mice. We showed that CT-mediated intratumoral coinjection of an anti-PD-L1 antibody and curcumin enabled sustained release of both the loaded anti-PD-L1 antibody and curcumin, which contributed to substantial anticancer effects with negligible side effects on the colons of the UC model mice. However, although the anti-PD-L1 antibody administered systemically synergized with the CT-mediated intratumoral delivery of curcumin in inhibiting tumour growth, colitis was significantly worsened by intraperitoneal administration of anti-PD-L1 antibody. These findings suggested that CT is a promising agent for the local delivery of anticancer drugs, as it can allow effective anticancer functions to be retained while sharply reducing the adverse side effects associated with the systemic administration of these drugs.
    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
    肝纤维化是一种缺乏特异性临床治疗的慢性病理状况。干细胞,在再生医学方面具有显著的潜力,提供在治疗肝纤维化的承诺。然而,干细胞治疗受到潜在免疫排斥反应的阻碍,致癌风险,功效变异,和高成本。基于干细胞分泌组的无细胞治疗为应对这些挑战提供了潜在的解决方案。但它是有限的低交货效率和快速清除。在这里,开发了一种用于原位植入智能微针(MN)阵列的创新方法,该方法能够精确控制地将多种治疗剂直接递送到纤维化肝组织中。通过整合无细胞和铂基纳米催化联合治疗,MN阵列可以使肝星状细胞失活。此外,它们促进超过75%的细胞外基质过度降解,接近正常水平。此外,智能MN阵列可以提供肝细胞保护,同时将炎症水平降低约70-90%。它们还可以在清除几乎100%的活性氧和缓解缺氧方面表现出非凡的能力。最终,这种治疗策略可以有效抑制纤维化进展。全面的体外和体内实验,补充蛋白质组和转录组分析,证实该方法治疗肝纤维化的有效性,拥有巨大的临床应用前景。
    Liver fibrosis is a chronic pathological condition lacking specific clinical treatments. Stem cells, with notable potential in regenerative medicine, offer promise in treating liver fibrosis. However, stem cell therapy is hindered by potential immunological rejection, carcinogenesis risk, efficacy variation, and high cost. Stem cell secretome-based cell-free therapy offers potential solutions to address these challenges, but it is limited by low delivery efficiency and rapid clearance. Herein, an innovative approach for in situ implantation of smart microneedle (MN) arrays enabling precisely controlled delivery of multiple therapeutic agents directly into fibrotic liver tissues is developed. By integrating cell-free and platinum-based nanocatalytic combination therapy, the MN arrays can deactivate hepatic stellate cells. Moreover, they promote excessive extracellular matrix degradation by more than 75%, approaching normal levels. Additionally, the smart MN arrays can provide hepatocyte protection while reducing inflammation levels by ≈70-90%. They can also exhibit remarkable capability in scavenging almost 100% of reactive oxygen species and alleviating hypoxia. Ultimately, this treatment strategy can effectively restrain fibrosis progression. The comprehensive in vitro and in vivo experiments, supplemented by proteome and transcriptome analyses, substantiate the effectiveness of the approach in treating liver fibrosis, holding immense promise for clinical 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
    霉菌毒素是导致全球食品损失和食品安全问题的代表性污染物。百里酚能有效抑制粮食贮藏过程中病原菌的侵染和黄曲霉毒素的积累,但高波动性限制了它的应用。这里,通过研磨诱导的自组装合成了百里酚-甜菜碱共晶体系。由于丰富的分子间相互作用,THY-TMG共晶体表现出优异的热稳定性,熔点为91.2°C。值得注意的是,在30°C下15天后,百里酚从共晶体中的释放率仅为55%,远远超过纯百里酚。值得注意的是,共晶显示了在控制百里酚释放的同时在环境中结合H2O的能力,本质上充当干燥剂。此外,该共晶体有效抑制了黄曲霉的生长和黄曲霉毒素B1的生物合成。实际上,THY-TMG共晶成功地防止了花生中的AFB1污染和营养流失,从而延长它们在28°C和70%RH条件下的保质期。
    Mycotoxins are representative contaminants causing food losses and food safety problems worldwide. Thymol can effectively inhibit pathogen infestation and aflatoxin accumulation during grain storage, but high volatility limits its application. Here, a thymol-betaine co-crystal system was synthesized through grinding-induced self-assembly. The THY-TMG co-crystal exhibited excellent thermal stability with melting point of 91.2 °C owing to abundant intermolecular interactions. Remarkably, after 15 days at 30 °C, the release rate of thymol from co-crystal was only 55%, far surpassing that of pure thymol. Notably, the co-crystal demonstrated the ability to bind H2O in the environment while controlling the release of thymol, essentially acting as a desiccant. Moreover, the co-crystals effectively inhibited the growth of Aspergillus flavus and the biosynthesis of aflatoxin B1. In practical terms, the THY-TMG co-crystal was successful in preventing AFB1 contamination and nutrients loss in peanuts, thereby prolonging their shelf-life under conditions of 28 °C and 70% RH.
    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
    本研究的重点是有效构建药物载体并在体外模拟中评估其动态释放的挑战。通过采用酸洗乳液和层层自组装的方法。微胶囊以茶树油为核心材料,SiO2纳米粒子作为稳定剂,壳聚糖和透明质酸作为外壳材料。微囊化机理,以及核壳质量比和搅拌的影响,进行了讨论。具体来说,设计并制作了基于3D打印技术的动态循环模拟微通道系统。在这个仿真系统中,微胶囊的释放速度加快,趋势发生变化,其行为与玻尔兹曼模型一致。该研究证明了自组装的无机-有机载药微胶囊在可控制备和易于功能修饰方面的优势。并显示了3D打印循环微通道系统在药物和生理分析中的可操作性和模拟保真度方面的潜力。
    This research focuses on the challenges of efficiently constructing drug carriers and evaluating their dynamic release in vitro simulation. By using pickering emulsion and layer-by-layer self-assembly methods. The microcapsules had tea tree oil as the core material, SiO2 nanoparticles as stabilizers, and chitosan and hyaluronic acid as shell materials. The microencapsulation mechanism, as well as the effects of core-shell mass ratio and stirring, were discussed. Specifically, a dynamic circulation simulation microchannel system was designed and manufactured based on 3D printing technology. In this simulation system, the release rate of microcapsules is accelerated and the trend changes, with its behavior aligning with the Boltzmann model. The study demonstrates the advantages of self-assembled inorganic-organic drug-loaded microcapsules in terms of controllable fabrication and ease of functional modification, and shows the potential of 3D printed cyclic microchannel systems in terms of operability and simulation fidelity in drug and physiological analysis.
    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