liver targeting

肝脏靶向
  • 文章类型: Journal Article
    酒精性肝损伤是全球肝病负担的主要致病因素。饮酒是全球发病率和死亡率的重要决定因素。鉴于肝脏靶向治疗减轻酒精性肝损伤仍然是一个主要的临床挑战,由于在主动靶向纳米医疗系统中与单一靶向修饰相关的特异性差和不稳定性,双功能靶向修饰可能是一种更有前途的策略。这里,半乳糖官能化透明质酸(Gal-HA)包被的阳离子固体脂质纳米颗粒携带水飞蓟宾(Gal-HA/SIL-SLNPs),具有双靶向透明质酸(HA)和半乳糖(Gal)部分,使能去唾液酸糖蛋白受体(ASGPR)和分化44(CD44)蛋白簇的特异性肝表面靶向,以增强水飞蓟宾的摄取,同时改善了带正电荷的脂质纳米颗粒作为药物载体的缺陷,并保持了它们在血液中的稳定性。根据调查结果,具有优异生物相容性的Gal-HA/SIL-SLNP表现出改善的细胞内化和肝脏分布,同时在酒精诱导的肝损伤小鼠模型中也显示出理想的治疗特性,而不会对其他器官造成损害。这项工作表明,具有双重修饰的Gal-HA/SIL-SLNPs可能代表一种令人鼓舞的方法,用于开发更有效的肝脏靶向纳米药物递送系统,以实现酒精性肝损伤的准确药物治疗。
    Alcoholic liver injury stands as a predominant pathogenic contributor to the global burden of liver diseases, with alcohol consumption serving as a significant determinant of worldwide morbidity and mortality. Given that liver-targeted therapy for mitigating alcoholic liver injury remains to be a major clinical challenge due to the poor specificity and instability associated with single targeting modification in actively targeted nanomedicine systems, bifunctional targeting modification may serve as a more promising strategy. Here, galactose-functionalized hyaluronic acid (Gal-HA) coated cationic solid lipid nanoparticles carrying silybin (Gal-HA/SIL-SLNPs) featuring dual-targeting hyaluronic acid (HA) and galactose (Gal) moieties, enabled specific liver surface targeting of asialoglycoprotein receptor (ASGPR) and cluster of differentiation 44 (CD44) proteins to enhance silybin uptake, while simultaneously ameliorating the deficiencies of positively charged lipid nanoparticles as drug carriers and preserving their stability in the bloodstream. Based on the findings, Gal-HA/SIL-SLNPs with excellent biocompatibility demonstrated improved cellular internalization and liver distribution, while also displaying ideal curative properties in a mouse model of alcohol-induced liver injury without causing damage to other organs. This work suggests that Gal-HA/SIL-SLNPs with dual modification may represent an encouraging approach for developing more effective liver targeted nano-drug delivery systems to achieve accurate medication for alcoholic liver injury.
    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
    这项工作的目的是构建基于氧化还原反应的肝靶向纳米颗粒,以有效地递送大麻二酚(CBD)以预防急性肝损伤(ALI)。使用通过用脱氧胆酸(DA)和α-硫辛酸(α-LA)接枝普鲁兰多糖获得的聚合物DA-PP-LA制备了粒径为126.5±1.56nm的载有CBD的纳米颗粒(CBDNP)。CBDNP表现出典型的氧化还原响应释放行为。有趣的是,CBDNP表现出令人钦佩的肝脏靶向能力,大量积累在肝脏中,并有效促进了CBD在肝细胞中的内在化,从而有效降低H2O2诱导的HepG2细胞氧化损伤,避免细胞凋亡。更重要的是,CBDNP通过保护肝功能有效预防CCl4诱导的ALI,改善氧化应激水平,抑制炎症因子的产生,保护肝脏免受组织学损伤。这项研究为在肝脏中实现CBDNP的靶向递送提供了有希望的策略,从而有效预防ALI。
    The purpose of this work was to construct liver-targeted nanoparticles based on the redox response to effectively deliver cannabidiol (CBD) for the prevention of acute liver injury (ALI). CBD-loaded nanoparticles (CBD NPs) with a particle size of 126.5 ± 1.56 nm were prepared using the polymer DA-PP-LA obtained by grafting pullulan polysaccharide with deoxycholic acid (DA) and α-lipoic acid (α-LA). CBD NPs showed typical redox-response release behavior. Interestingly, CBD NPs exhibited admirable liver targeting ability, significantly accumulated in the liver, and effectively promoted the internalization of CBD in liver cells, thus effectively reducing the H2O2-induced oxidative damage of HepG2 cells and avoiding apoptosis. More importantly, CBD NPs effectively prevented CCl4-induced ALI by protecting liver function, ameliorating oxidative stress levels, inhibiting the production of inflammatory factors, and protecting the liver from histological damage. This study provides a promising strategy for achieving targeted delivery of CBD NPs in the liver, thereby effectively preventing ALI.
    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
    肝纤维化(LF)是由慢性肝损伤引起的病理修复反应,影响全球数百万人的健康,在没有及时干预的情况下进展为危及生命的肝硬化和肝癌。由于LF病理学的复杂性,多种病因学特征,和沉积的细胞外基质,传统药物无法以时空匹配的方式达到适当的目标,从而降低治疗效果。纳米颗粒药物递送系统(NDDS)实现多药联合治疗,并开发针对病理过程的多因子递送策略,在LF治疗中显示出巨大的潜力。根据LF的发病机制和目前的临床治疗现状,我们系统地阐明了NDDS用于治疗LF的靶向机制。随后,我们专注于LF给药应用的进展,包括肝纤维化病理环境的联合递送,克服生物障碍,精确的细胞内调节,和智能响应递送肝纤维化微环境。希望本文能对未来LFNDDS的合理设计有所启发,为促进LF回归和治愈提供思路和方法。
    Liver fibrosis (LF) is a pathological repair reaction caused by a chronic liver injury that affects the health of millions of people worldwide, progressing to life-threatening cirrhosis and liver cancer without timely intervention. Due to the complexity of LF pathology, multiple etiological characteristics, and the deposited extracellular matrix, traditional drugs cannot reach appropriate targets in a time-space matching way, thus decreasing the therapeutic effect. Nanoparticle drug delivery systems (NDDS) enable multidrug co-therapy and develop multifactor delivery strategies targeting pathological processes, showing great potential in LF therapy. Based on the pathogenesis and the current clinical treatment status of LF, we systematically elucidate the targeting mechanism of NDDS used in the treatment of LF. Subsequently, we focus on the progress of drug delivery applications for LF, including combined delivery for the liver fibrotic pathological environment, overcoming biological barriers, precise intracellular regulation, and intelligent responsive delivery for the liver fibrotic microenvironment. We hope that this review will inspire the rational design of NDDS for LF in the future in order to provide ideas and methods for promoting LF regression and cure.
    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:癌症是严重威胁人类生命的疾病,健康和社会发展。近年来,纳米胶束,作为一种新兴的药物载体材料,因其提高生物利用度的优势,逐渐进入人们的视野,维持药物水平,减少全身副作用和增加药物在靶位点的积累。方法:在本研究中,以马钱子碱为模型药物,甘草次酸-聚乙二醇-3-亚甲基二醇-二硫代二丙酸-单硬脂酸甘油酯为载体,采用薄膜分散水化法制备了B-GPSG纳米胶束。制备过程,表征,体外药物释放,研究了药代动力学和肝脏靶向。结果:结果表明,在粒径范围内,多分散指数和Zeta电位为102.7±1.09nm,分别为0.201±0.02和-24.5±0.19mV。包封率和载药量分别为83.79±2.13%和12.56±0.09%,分别。体外释药实验和药动学实验表明其具有明显的缓释作用。对于药代动力学研究,表明B-GPSG溶液组和B-PSG溶液组均改变了马钱子碱的代谢动力学参数,但B-GPSG溶液组效果较好。与B-PSG溶液组相比,这种药物在大鼠体内的时间更长。B-GPSG在体内的半衰期和在体内的保留时间更有助于提高药物的生物利用度,发挥长效作用。小鼠尾静脉注射结果表明,B-GPSG可以靶向并在肝脏中积累马钱子碱,而不会影响其他关键器官。体内细胞摄取实验和组织分布实验表明,甘草次酸修饰的纳米胶束可以增加马钱子碱在肝细胞中的积累,具有良好的肝脏靶向作用,并可作为治疗肝癌的新制剂。结论:本实验制备的B-SPSG可为肝癌的治疗提供新的治疗方法和研究思路。
    Background: Cancer is a serious threat to human life, health and social development. In recent years, nanomicelles, as an emerging drug carrier material, have gradually entered people\'s field of vision because of their advantages of improving bioavailability, maintaining drug levels, reducing systemic side effects and increasing drug accumulation at target sites. Methods: In this study, B-GPSG nano-micelles were prepared by film dispersion hydration method using brucine as model drug and glycyrrhetinic acid-polyethylene glycol-3-methylene glycol-dithiodipropionic acid-glycerol monostearate polymer as nano-carrier. The preparation process, characterization, drug release in vitro, pharmacokinetics and liver targeting were investigated. Results: The results showed that the range of particle size, polydispersion index and Zeta potential were 102.7 ± 1.09 nm, 0.201 ± 0.02 and -24.5 ± 0.19 mV respectively. The entrapment efficiency and drug loading were 83.79 ± 2.13% and 12.56 ± 0.09%, respectively. The drug release experiments in vitro and pharmacokinetic experiments showed that it had obvious sustained release effect. For pharmacokinetics study, it shows that both the B-GPSG solution group and the B-PSG solution group changed the metabolic kinetic parameters of brucine, but the B-GPSG solution group had a better effect. Compared with the B-PSG solution group, the drug was more prolonged in rats. The half-life in the body and the retention time in the body of B-GPSG are more helpful to improve the bioavailability of the drug and play a long-term effect. The tail vein injection results of mice indicate that B-GPSG can target and accumulate brucine in the liver without affecting other key organs. Cell uptake experiments and tissue distribution experiments in vivo show that glycyrrhetinic acid modified nano-micelles can increase the accumulation of brucine in hepatocytes, has a good liver targeting effect, and can be used as a new preparation for the treatment of liver cancer. Conclusion: The B-SPSG prepared in this experiment can provide a new treatment method and research idea for the treatment of liver 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
    通过受体介导将基因靶向递送到肝细胞中的新型递送方案的制定对于肝脏特异性疾病的治疗是重要的。包括癌症.非病毒递送方法已被广泛研究用于基因治疗。金纳米粒子(AuNPs)由于其生物相容性而在纳米医学中受到关注。在这项研究中,合成了AuNP并用聚合物:壳聚糖(CS),和聚乙二醇(PEG)。靶向部分,乳糖酸(LA),添加用于肝细胞特异性递送。物理化学表征表明,所有纳米制剂都是球形和单分散的,流体动力学尺寸在70和250nm之间。与pCMV-LucDNA(pDNA)的纳米复合物证实了NP可以结合,紧凑型,并保护pDNA免受核酸酶降解。细胞毒性研究表明,AuNPs在人肝细胞癌(HepG2)中具有良好的耐受性(细胞活力>70%),胚胎肾(HEK293),和结肠直肠腺癌(Caco-2)细胞,在所有细胞中具有增强的转基因活性。在NP制剂中包含LA在HepG2细胞中是显著的,在其细胞表面过度表达脱唾液酸糖蛋白受体。与未靶向的AuNP相比,对于靶向LA的AuNP,荧光素酶基因表达增加5倍是明显的。这些AuNP已显示出作为肝定向基因治疗的安全和合适的靶向递送载体的潜力。
    The formulation of novel delivery protocols for the targeted delivery of genes into hepatocytes by receptor mediation is important for the treatment of liver-specific disorders, including cancer. Non-viral delivery methods have been extensively studied for gene therapy. Gold nanoparticles (AuNPs) have gained attention in nanomedicine due to their biocompatibility. In this study, AuNPs were synthesized and coated with polymers: chitosan (CS), and polyethylene glycol (PEG). The targeting moiety, lactobionic acid (LA), was added for hepatocyte-specific delivery. Physicochemical characterization revealed that all nano-formulations were spherical and monodispersed, with hydrodynamic sizes between 70 and 250 nm. Nanocomplexes with pCMV-Luc DNA (pDNA) confirmed that the NPs could bind, compact, and protect the pDNA from nuclease degradation. Cytotoxicity studies revealed that the AuNPs were well tolerated (cell viabilities > 70%) in human hepatocellular carcinoma (HepG2), embryonic kidney (HEK293), and colorectal adenocarcinoma (Caco-2) cells, with enhanced transgene activity in all cells. The inclusion of LA in the NP formulation was notable in the HepG2 cells, which overexpress the asialoglycoprotein receptor on their cell surface. A five-fold increase in luciferase gene expression was evident for the LA-targeted AuNPs compared to the non-targeted AuNPs. These AuNPs have shown potential as safe and suitable targeted delivery vehicles for liver-directed gene 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
    内分泌治疗是激素受体阳性(HR)乳腺癌治疗的标准。然而,目前针对雌激素信号的策略很少关注肝脏中的雌二醇代谢,并且通常受到治疗失败的挑战。在之前的研究中,我们证明了天然化合物柚皮素(NAR)通过激活肝脏中雌激素磺基转移酶(EST)的表达来抑制HR乳腺癌的生长。然而,水溶性差,低生物屏障渗透性,非特异性分布限制了其临床应用,特别是口服给药。这里,报道了一种新型的纳米内分泌药物NAR-细胞穿透肽-半乳糖纳米颗粒(NCG)。我们证明了NCG在细胞和斑马鱼异种移植模型中均具有特异性的肝靶向性和增加的肠屏障通透性。此外,口服后,NCG在小鼠乳腺癌异种移植物中显示出肝靶向和肠肝循环。值得注意的是,NCG的抑癌效果优于NAR和阳性对照他莫昔芬,并伴有肝脏EST表达增加和肝脏中雌二醇水平降低,血,和肿瘤组织。此外,NCG治疗后几乎没有副作用。我们的发现表明NCG是内分泌治疗的有希望的候选药物,并强调肝EST靶向作为HR+乳腺癌的新治疗策略。
    Endocrine therapy is standard for hormone receptor-positive (HR+) breast cancer treatment. However, current strategies targeting estrogen signaling pay little attention to estradiol metabolism in the liver and is usually challenged by treatment failure. In a previous study, we demonstrated that the natural compound naringenin (NAR) inhibited HR+ breast cancer growth by activating estrogen sulfotransferase (EST) expression in the liver. Nevertheless, the poor water solubility, low bio-barrier permeability, and non-specific distribution limited its clinical application, particularly for oral administration. Here, a novel nano endocrine drug NAR-cell penetrating peptide-galactose nanoparticles (NCG) is reported. We demonstrated that NCG presented specific liver targeting and increased intestinal barrier permeability in both cell and zebrafish xenotransplantation models. Furthermore, NCG showed liver targeting and enterohepatic circulation in mouse breast cancer xenografts following oral administration. Notably, the cancer inhibition efficacy of NCG was superior to that of both NAR and the positive control tamoxifen, and was accompanied by increased hepatic EST expression and reduced estradiol levels in the liver, blood, and tumor tissue. Moreover, few side effects were observed after NCG treatment. Our findings reveal NCG as a promising candidate for endocrine therapy and highlight hepatic EST targeting as a novel therapeutic strategy for HR+ breast cancer.
    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
    改善纳米医学中的靶标与脱靶比率仍然是提高药物生物利用度和降低毒性的主要挑战。在纳米颗粒表面上使用配体的主动靶向是关键方法,但临床成功有限。潜在的问题是靶向配体的整合也改变了纳米颗粒的物理化学性质(被动靶向)。了解体内主动靶向和脱靶机制的直接研究由于缺乏合适的工具而受到限制。这里,分析了代表性活性靶向脂质体的生物分布,用与低密度脂蛋白受体(LDLR)结合的载脂蛋白E(ApoE)肽修饰,使用斑马鱼胚胎。ApoE脂质体显示了预期的肝靶向作用,但也在肾小球中积累。开发了ldlra-/-斑马鱼以探索ApoE脂质体的LDLR特异性。有趣的是,肝脏靶向取决于LDLR特异性相互作用,而肾小球的积累与LDLR和肽序列无关。发现肽的阳离子电荷和脂质体的大小控制肾小球靶向。增加ApoE脂质体的大小可以避免这种脱靶。一起来看,这项研究表明,斑马鱼胚胎模型具有理解主动和被动靶向机制的潜力,可用于优化纳米粒子的设计。
    Improving target versus off-target ratio in nanomedicine remains a major challenge for increasing drug bioavailability and reducing toxicity. Active targeting using ligands on nanoparticle surfaces is a key approach but has limited clinical success. A potential issue is the integration of targeting ligands also changes the physicochemical properties of nanoparticles (passive targeting). Direct studies to understand the mechanisms of active targeting and off-targeting in vivo are limited by the lack of suitable tools. Here, the biodistribution of a representative active targeting liposome is analyzed, modified with an apolipoprotein E (ApoE) peptide that binds to the low-density lipoprotein receptor (LDLR), using zebrafish embryos. The ApoE liposomes demonstrated the expected liver targeting effect but also accumulated in the kidney glomerulus. The ldlra-/- zebrafish is developed to explore the LDLR-specificity of ApoE liposomes. Interestingly, liver targeting depends on the LDLR-specific interaction, while glomerular accumulation is independent of LDLR and peptide sequence. It is found that cationic charges of peptides and the size of liposomes govern glomerular targeting. Increasing the size of ApoE liposomes can avoid this off-targeting. Taken together, the study shows the potential of the zebrafish embryo model for understanding active and passive targeting mechanisms, that can be used to optimize the design of nanoparticles.
    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: English Abstract
    将3-琥珀酸-30-硬脂酰甘草次酸(18-GA-Suc)插入甘草次酸(GA)-丹参酮Ⅱ_A(TSN)-丹酚酸B(SalB)脂质体(GTS-lip)中,制备由GA受体介导的肝靶向复合脂质体(Suc-GTS-lip)。接下来,通过UPLC比较Suc-GTS-lip和GTS-lip的药代动力学和组织分布,并对Suc-GTS-lip进行体内成像追踪。作者研究了Suc-GTS-lip对肝星状细胞(HSC)增殖的抑制作用,并探讨了其改善肝纤维化的分子机制。药代动力学结果表明,AUC_(SalB)从(636.06±27.73)μg·h·mL〜(-1)降低到(550.39±12.34)μg·h·mL〜(-1),AUC_(TSN)由(1.08±0.72)μg·h·mL~(-1)降至(0.65±0.04)μg·h·mL~(-1),但AUC_(GA)从(43.64±3.10)μg·h·mL〜(-1)增加到(96.21±3.75)μg·h·mL〜(-1)。组织分布结果表明,Suc-GTS-lip组肝脏中SalB的AUC_(SalB)和C_(max)分别是GTS-lip组的10.21和4.44倍,分别。SalB的肝靶向效率,TSN,Suc-GTS-lip组的GA为40.66%,3.06%,和22.08%,分别。体内成像研究表明,修饰的脂质体倾向于在肝脏中积累。MTT结果显示Suc-GTS-lip能显著抑制HSC的增殖,RT-PCR结果显示各组MMP-1的表达均显著升高,但TIMP-1和TIMP-2明显下降。各组胶原-I和胶原-Ⅲ的mRNA表达均显著降低。实验结果表明Suc-GTS-lip具有肝靶向性,抑制HSC的增殖并诱导其凋亡,为Suc-GTS-lip靶向治疗肝纤维化提供实验依据。
    The 3-succinate-30-stearyl glycyrrhetinic acid(18-GA-Suc) was inserted into glycyrrhetinic acid(GA)-tanshinone Ⅱ_A(TSN)-salvianolic acid B(Sal B) liposome(GTS-lip) to prepare liver targeting compound liposome(Suc-GTS-lip) mediated by GA receptors. Next, pharmacokinetics and tissue distribution of Suc-GTS-lip and GTS-lip were compared by UPLC, and in vivo imaging tracking of Suc-GTS-lip was conducted. The authors investigated the effect of Suc-GTS-lip on the proliferation inhibition of hepatic stellate cells(HSC) and explored their molecular mechanism of improving liver fibrosis. Pharmacokinetic results showed that the AUC_(Sal B) decreased from(636.06±27.73) μg·h·mL~(-1) to(550.39±12.34) μg·h·mL~(-1), and the AUC_(TSN) decreased from(1.08±0.72) μg·h·mL~(-1) to(0.65±0.04) μg·h·mL~(-1), but the AUC_(GA) increased from(43.64±3.10) μg·h·mL~(-1) to(96.21±3.75) μg·h·mL~(-1). The results of tissue distribution showed that the AUC_(Sal B) and C_(max) of Sal B in the liver of the Suc-GTS-lip group were 10.21 and 4.44 times those of the GTS-lip group, respectively. The liver targeting efficiency of Sal B, TSN, and GA in the Suc-GTS-lip group was 40.66%, 3.06%, and 22.08%, respectively. In vivo imaging studies showed that the modified liposomes tended to accumulate in the liver. MTT results showed that Suc-GTS-lip could significantly inhibit the proliferation of HSC, and RT-PCR results showed that the expression of MMP-1 was significantly increased in all groups, but that of TIMP-1 and TIMP-2 was significantly decreased. The mRNA expressions of collagen-I and collagen-Ⅲ were significantly decreased in all groups. The experimental results showed that Suc-GTS-lip had liver targeting, and it could inhibit the proliferation of HSC and induce their apoptosis, which provided the experimental basis for the targeted treatment of liver fibrosis by Suc-GTS-lip.
    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
    肝纤维化主要是由肝脏细胞外基质的过度积累和结构改变引起的,最终导致肝硬化,如果不及时治疗。通过抑制透明质酸沉积或调节透明质酸合酶的表达来减少透明质酸合成可以改善肝纤维化症状。在这项研究中,我们的目的是提高羟甲基香豆素(4-MU)的生物利用度和肝脏靶向能力,使用新开发的磷脂复合物壳聚糖纳米颗粒(4-MUPC/CNP),优化Box-Behnken设计.采用溶剂蒸发技术配制了复合纳米载体递送体系,并对配方和工艺参数进行了评价。此外,对4-MUPC/CNPs及其药代动力学进行了表征。建立的4-MUPC/CNPs的平均粒径为153.07±0.29nm,多分散指数值为0.383,正zeta电位为~35.4mV。与4-MU相比,4-MUPC/CNPs的水溶性显著提高,更快的血浆清除和组织分布,和更好的肝脏靶向。药代动力学分析显示4-MU在4-MUPC/CNPs中的口服生物利用度显著高于简单4-MU。总之,4-MUPC改善了药物脂质(油水分配系数为1.31±0.03)和水溶解度(药物的2.05倍)。4-MUPC/CNPs显著提高4-MU口服生物利用度,代表了一种有希望的提高药物溶解度的方法。这项研究表明,4-MUPC/CNPs在肝脏中的靶向参数均大于1,表明它们特异性靶向肝脏,从而潜在地减轻肝纤维化。
    Liver fibrosis is mainly caused by excessive accumulation of extracellular matrix and structural changes in the liver, ultimately leading to cirrhosis if left untreated. Reducing hyaluronan synthesis by inhibiting hyaluronic acid deposition or regulating the expression of hyaluronic synthase can ameliorate liver fibrosis symptoms. In this study, we aimed to improve the bioavailability and liver-targeting capacity of hydroxymethyl coumarin (4-MU) using a newly developed phospholipid complex chitosan nanoparticle (4-MU PC/CNP) optimized using the Box-Behnken design. The composite nanocarrier delivery system was formulated using solvent evaporation technology, and formulation and process parameters were evaluated. Furthermore, 4-MU PC/CNPs and their pharmacokinetics were characterized. The established 4-MU PC/CNPs had an average particle size of 153.07 ± 0.29 nm, a polydispersity index value of 0.383, and a positive zeta potential of ∼35.4 mV. Compared with 4-MUs, 4-MU PC/CNPs exhibited significantly improved water solubility, faster plasma clearance and tissue distribution, and better liver targeting. Pharmacokinetic analysis showed that the oral bioavailability of 4-MU in 4-MU PC/CNPs was significantly higher than that of simple 4-MU. In conclusion, 4-MU PC improved drug lipid (oil-water distribution coefficient of 1.31 ± 0.03) and water solubilities (2.05 times the drug substance). 4-MU PC/CNPs significantly improved 4-MU oral bioavailability, representing a promising approach for enhancing drug solubility. This study demonstrates that the targeting parameters of 4-MU PC/CNPs in the liver were all greater than 1, indicating that they specifically targeted the liver, thereby potentially alleviating liver fibrosis.
    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
    索拉非尼是一种口服治疗肝细胞癌(HCC)。然而,水溶性差,恶劣的胃肠道环境和脱靶效应导致口服索拉非尼的生物利用度较低。植物来源的细胞外囊泡(PDEV)是具有各种生物活性功能的生物纳米囊泡,其在口服药物递送领域提供显著的优势:防止被胃肠液降解;穿过肠上皮屏障;特异性靶向;安全性;和丰富的产量。然而,有较少的研究应用PDEVs抗肿瘤药物递送到消化外组织.在这项研究中,从猕猴桃中分离纯化猕猴桃来源的细胞外囊泡(KEV),和它们的自然肝脏积累特性被用于靶向递送索拉非尼(KEVs-SFB)。有证据表明,包封KEVs可减少索拉非尼在胃肠道环境中的渗漏,增强其穿过肠上皮的能力;KEVs-SFB能够实现肝脏蓄积,并主要被HepG2细胞摄取;KEVs-SFB有效抑制4T1细胞增殖;在原位肝癌模型中,口服KEVs-SFB抑制肿瘤生长,改善SFB的副作用。这种基于PDEVs的口服药物递送平台对于提高口服生物利用度和减少药物副作用是重要的。
    Sorafenib is an oral treatment for hepatocellular carcinoma (HCC). However, poor water solubility, harsh gastrointestinal environment and off-target effects contribute to the low bioavailability of oral sorafenib. Plant-derived extracellular vesicles (PDEVs) are biological nanovesicles with various bioactive functions that offer significant advantages in the field of oral drug delivery: protection from degradation by gastrointestinal fluids; crossing the intestinal epithelial barrier; specific targeting; safety; and abundant yield. However, there are fewer studies applying PDEVs for anti-tumor drug delivery to extra-digestive tissues. In this study, kiwifruit-derived extracellular vesicles (KEVs) were isolated and purified from kiwifruit, and their natural hepatic accumulation properties were exploited for targeted delivery of sorafenib (KEVs-SFB). Evidence showed that encapsulation of KEVs reduced the leakage of sorafenib in the gastrointestinal environment and enhanced the ability to cross the intestinal epithelium; KEVs-SFB was able to achieve liver accumulation and was predominantly taken up by HepG2 cells; KEVs-SFB was effective in inhibiting 4T1 cell proliferation; in the orthotopic liver cancer model, oral administration of KEVs-SFB inhibited tumor growth and improved the side effects of SFB. This PDEVs-based oral drug delivery platform is important for improving oral bioavailability and reducing drug side effects.
    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