Drug Delivery Systems

药物输送系统
  • 文章类型: Journal Article
    癌症因其每年高死亡率而严重威胁人类健康。它在医疗保健领域引起了极大的关注,确定治疗和缓解癌症疼痛的有效策略需要紧迫性。药物递送系统(DDS)具有优异的疗效,低成本,和低毒性的靶向药物到肿瘤部位。近几十年来,基于聚(苯丙氨酸)(PPhe)和聚(3,4-二羟基-L-苯丙氨酸)(PDopa)的共聚物载体由于其良好的生物相容性而被广泛研究,生物降解性,和可控的刺激反应,这导致了具有加载和目标交付功能的DDS。在这次审查中,我们介绍了PPhe和PDopa的合成,重点介绍最新建议的合成路线,并比较PPhe和PDopa之间的药物递送差异。随后,我们总结了PPhe和PDopa在纳米级靶向DDS中的各种应用,提供基于使用这两种材料的不同刺激响应载体的药物释放行为的综合分析。最后,我们讨论了基于多肽的DDS在癌症治疗领域的挑战和前景,旨在促进其进一步发展,以满足日益增长的治疗需求。
    Cancer is a serious threat to human health because of its high annual mortality rate. It has attracted significant attention in healthcare, and identifying effective strategies for the treatment and relief of cancer pain requires urgency. Drug delivery systems (DDSs) offer the advantages of excellent efficacy, low cost, and low toxicity for targeting drugs to tumor sites. In recent decades, copolymer carriers based on poly(phenylalanine) (PPhe) and poly(3,4-dihydroxy-L-phenylalanine) (PDopa) have been extensively investigated owing to their good biocompatibility, biodegradability, and controllable stimulus responsiveness, which have resulted in DDSs with loading and targeted delivery capabilities. In this review, we introduce the synthesis of PPhe and PDopa, highlighting the latest proposed synthetic routes and comparing the differences in drug delivery between PPhe and PDopa. Subsequently, we summarize the various applications of PPhe and PDopa in nanoscale-targeted DDSs, providing a comprehensive analysis of the drug release behavior based on different stimulus-responsive carriers using these two materials. In the end, we discuss the challenges and prospects of polypeptide-based DDSs in the field of cancer therapy, aiming to promote their further development to meet the growing demands for 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
    mRNA疗法已显示出用于广谱疾病治疗的巨大潜力。然而,mRNA固有的不稳定性和细胞进入困难的挑战阻碍了其在生物医学领域的进展。为了解决细胞屏障并将mRNA递送到感兴趣的细胞,各种交付系统的设计。其中,脂质纳米颗粒(LNPs)是最广泛使用的mRNA递送系统,特别是在冠状病毒病2019(COVID-19)mRNA疫苗的临床批准之后。LNP由可电离的阳离子脂质组成,磷脂,胆固醇,和聚乙二醇衍生的脂质(PEG-脂质)。在这次审查中,我们主要总结了LNPmRNA递送技术的最新进展,重点介绍了四种脂质成分的结构及其生物医学应用。我们深入研究了脂质的结构-活性关系,同时还探索了开发更有效的mRNA递送系统的未来前景和挑战。本文分为:治疗方法和药物发现>新兴技术生物学启发的纳米材料>基于脂质的结构纳米技术生物学方法>生物学中的纳米级系统。
    mRNA therapeutics have shown great potential for a broad spectrum of disease treatment. However, the challenges of mRNA\'s inherent instability and difficulty in cellular entry have hindered its progress in the biomedical field. To address the cellular barriers and deliver mRNA to cells of interest, various delivery systems are designed. Among these, lipid nanoparticles (LNPs) stand out as the most extensively used mRNA delivery systems, particularly following the clinical approvals of corona virus disease 2019 (COVID-19) mRNA vaccines. LNPs are comprised of ionizable cationic lipids, phospholipids, cholesterol, and polyethylene glycol derived lipids (PEG-lipids). In this review, we primarily summarize the recent advancements of the LNP mRNA delivery technology, focusing on the structures of four lipid constituents and their biomedical applications. We delve into structure-activity relationships of the lipids, while also exploring the future prospects and challenges in developing more efficacious mRNA delivery systems. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
    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
    信使RNA(mRNA)已成为一种有前途的治疗分子,在治疗中枢神经系统疾病中具有许多临床应用。肿瘤,COVID-19和其他疾病。mRNA治疗必须封装到安全,稳定,和有效的运载工具,以保护货物免于降解并防止免疫原性。外泌体由于其良好的生物相容性,在mRNA递送中获得了越来越多的关注,低免疫原性,小尺寸,穿过生理屏障的独特能力,和细胞特异性的向性。此外,这些外泌体可以被工程化以利用天然载体靶向特定的细胞或组织。这种靶向方法将增强mRNAs的功效并减少其副作用。然而,必须解决一些困难,例如缺乏一致且可靠的外泌体纯化方法以及将大mRNA有效封装到外泌体中。本文概述了当前在细胞来源的囊泡介导的mRNA递送及其生物医学应用方面的突破。
    Messenger RNA (mRNA) has emerged as a promising therapeutic molecule with numerous clinical applications in treating central nervous system disorders, tumors, COVID-19, and other diseases. mRNA therapies must be encapsulated into safe, stable, and effective delivery vehicles to preserve the cargo from degradation and prevent immunogenicity. Exosomes have gained growing attention in mRNA delivery because of their good biocompatibility, low immunogenicity, small size, unique capacity to traverse physiological barriers, and cell-specific tropism. Moreover, these exosomes can be engineered to utilize the natural carriers to target specific cells or tissues. This targeted approach will enhance the efficacy and reduce the side effects of mRNAs. However, difficulties such as a lack of consistent and reliable methods for exosome purification and the efficient encapsulation of large mRNAs into exosomes must be addressed. This article outlines current breakthroughs in cell-derived vesicle-mediated mRNA delivery and its biomedical 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
    被称为细胞外囊泡(EV)的天然生成的脂质纳米颗粒作为可工程化的治疗递送载体具有显著的前景。然而,以可用于递送的方式将蛋白质货物主动装载到EV中仍然是一个挑战。这里,我们证明,通过合理设计蛋白质运输到质膜并与脂筏相关联,我们可以增强一组结构多样的跨膜和外周膜蛋白的蛋白质货物装载到EV中。然后我们证明了选择的脂质标签介导增加的EV负载和工程化转录因子的功能性递送以调节靶细胞中的基因表达的能力。我们设想可以利用这项技术来开发新的基于EV的疗法,提供广泛的大分子货物。
    Naturally generated lipid nanoparticles termed extracellular vesicles (EVs) hold significant promise as engineerable therapeutic delivery vehicles. However, active loading of protein cargo into EVs in a manner that is useful for delivery remains a challenge. Here, we demonstrate that by rationally designing proteins to traffic to the plasma membrane and associate with lipid rafts, we can enhance loading of protein cargo into EVs for a set of structurally diverse transmembrane and peripheral membrane proteins. We then demonstrate the capacity of select lipid tags to mediate increased EV loading and functional delivery of an engineered transcription factor to modulate gene expression in target cells. We envision that this technology could be leveraged to develop new EV-based therapeutics that deliver a wide array of macromolecular cargo.
    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
    智能纳米药物递送系统(Cu/ZIF-8@GOx-DOX@HA,以下为CZGDH),由掺杂Cu的沸石咪唑酯骨架8(Cu/ZIF-8,以下为CZ)组成,葡萄糖氧化酶(GOx),多柔比星(DOX),透明质酸(HA)用于肿瘤的靶向给药和协同治疗。CZGDH通过HA的靶向作用特异性进入肿瘤细胞,并表现出酸度触发的生物降解作用,随后释放GOx,DOX,和肿瘤微环境(TME)中的Cu2+。GOx氧化肿瘤细胞中的葡萄糖(Glu)以产生H2O2和葡萄糖酸用于饥饿治疗(ST)。DOX进入肿瘤内细胞核进行化疗(CT)。释放的Cu2+消耗肿瘤细胞中过表达的谷胱甘肽(GSH)以产生Cu+。生成的Cu+和H2O2引发类Fenton反应生成有毒的羟基自由基(·OH),这破坏了肿瘤细胞的氧化还原平衡,并有效地杀死了肿瘤细胞进行化学动力学治疗(CDT)。因此,通过TME激活的级联反应实现了协同多峰肿瘤治疗。纳米药物递送系统具有高的载药率(48.3wt%),三模式协同治疗对肿瘤细胞有很强的杀伤作用(67.45%)。
    An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).
    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
    全球范围内,非小细胞肺癌(NSCLC)是对人类健康的重大威胁,占肺癌病例的80%以上。顺铂(CDDP),临床治疗中常用的药物,一直是旨在通过包封在脂质体中减轻其有效毒性的研究重点。然而,挑战,例如降低的药物装载效率和非特异性释放,已经成为障碍。本研究旨在通过预先制备CDDP并通过掺入花生凝集素(PNA)作为配体[CDDP负载的PNA修饰的脂质体(CDDP-PNA-Lip)]修饰脂质体表面来提高CDDP在脂质体中的包封效率。该策略旨在增强CDDP向肿瘤组织的递送,从而减少相关的副作用。通过体外研究阐明了CDDP-PNA-Lip对MUC1高表达NSCLC细胞系增殖和迁移的影响。此外,通过异种移植肿瘤实验评估了PNA修饰增强脂质体靶向抗肿瘤功效的能力.结果表明,在体外摄取测定中,负载罗丹明B(RhB)的PNA修饰的脂质体被细胞吸收,与游离RhB相比,效率提高了约50%。此外,与游离CDDP相比,CDDP-PNA-Lip在体内的肿瘤抑制作用增强了2.65倍。这些发现表明,CDDP在配体修饰的脂质体中的封装可以显着提高其肿瘤靶向能力,为临床药物开发提供有价值的见解。
    Globally, non‑small cell lung cancer (NSCLC) is a significant threat to human health, and constitutes >80% of lung cancer cases. Cisplatin (CDDP), a commonly used drug in clinical treatment, has been the focus of research aiming to mitigate its potent toxicity through encapsulation within liposomes. However, challenges, such as a reduced drug loading efficiency and nonspecific release, have emerged as obstacles. The present study aimed to improve the encapsulation efficiency of CDDP within liposomes by pre‑preparation of CDDP and modifying the liposome surface through the incorporation of peanut agglutinin (PNA) as a ligand [CDDP‑loaded PNA‑modified liposomes (CDDP‑PNA‑Lip)]. This strategy was designed to enhance the delivery of CDDP to tumour tissues, thereby reducing associated side effects. The effect of CDDP‑PNA‑Lip on the proliferation and migration of NSCLC cell lines with high MUC1 expression was elucidated through in vitro studies. Additionally, the capacity of PNA modification to augment the targeted anti‑tumour efficacy of liposomes was assessed through xenograft tumour experiments. The results indicated that in an in vitro uptake assay Rhodamine B (RhB)‑loaded PNA‑modified liposomes were taken up by cells with ~50% higher efficiency compared with free RhB. In addition, CDDP‑PNA‑Lip resulted in a 2.65‑fold enhancement of tumour suppression in vivo compared with free CDDP. These findings suggested that the encapsulation of CDDP within ligand‑modified liposomes may significantly improve its tumour‑targeting capabilities, providing valuable insights for clinical drug development.
    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
    痤疮是毛囊皮脂腺常见的慢性炎症性疾病。常用的外用药物会引起皮肤刺激,透皮能力较弱,使其难以穿透角质层皮肤屏障。毛囊可以帮助打破这种屏障。随着纳米材料的进步,基于聚合物的纳米载体通常用于毛囊药物递送以治疗痤疮和其他皮肤问题。根据毛囊的生理解剖特征,本文讨论了影响聚合物纳米载体毛囊传递的因素,总结了提高载体对毛囊靶向性的常用组合技术,最后综述了不同聚合物纳米给药系统靶向毛囊治疗痤疮的最新研究进展。
    Acne is a common chronic inflammatory disorder of the sebaceous gland in the hair follicle. Commonly used external medications cause skin irritation, and the transdermal capacity is weak, making it difficult to penetrate the cuticle skin barrier. Hair follicles can aid in the breakdown of this barrier. As nanomaterials progress, polymer-based nanocarriers are routinely used for hair follicle drug delivery to treat acne and other skin issues. Based on the physiological and anatomical characteristics of hair follicles, this paper discusses factors affecting hair follicle delivery by polymer nanocarriers, summarizes the common combination technology to improve the targeting of hair follicles by carriers, and finally reviews the most recent research progress of different polymer nanodrug-delivery systems for the treatment of acne by targeting hair follicles.
    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
    纳米凝胶为精确的药物输送提供了希望,而解决药物输送障碍对于有效的前列腺癌(PCa)管理至关重要。我们开发了一种可注射的弹性蛋白纳米凝胶(ENG),用于有效的药物递送系统,通过递送Decursin来克服去势抵抗性前列腺癌(CRPC),一种阻断PCa的Wnt/β连环蛋白途径的小分子抑制剂。ENG表现出良好的特性,如生物相容性,灵活性,和低毒性。在这项研究中,尺寸,形状,表面电荷,化学成分,热稳定性,和ENG的其他特性用于确认成功合成并将Decursin(DEC)掺入到弹性蛋白纳米凝胶(ENG)中用于前列腺癌治疗。体外研究表明,DEC从ENG持续释放超过120小时,具有pH依赖性释放模式。DU145细胞系诱导DEC-ENG的中等细胞毒性表明纳米药物对细胞活力有影响,并有助于在治疗功效和安全性之间取得平衡,而与游离DEC相比,EPR效应能够靶向药物递送至前列腺肿瘤部位。形态学分析进一步支持DEC-ENG诱导细胞死亡的有效性。总的来说,这些发现强调了ENG封装的decurin作为CRPC靶向给药系统的有希望的作用。
    Nanogels offer hope for precise drug delivery, while addressing drug delivery hurdles is vital for effective prostate cancer (PCa) management. We developed an injectable elastin nanogels (ENG) for efficient drug delivery system to overcome castration-resistant prostate cancer (CRPC) by delivering Decursin, a small molecule inhibitor that blocks Wnt/βcatenin pathways for PCa. The ENG exhibited favourable characteristics such as biocompatibility, flexibility, and low toxicity. In this study, size, shape, surface charge, chemical composition, thermal stability, and other properties of ENG were used to confirm the successful synthesis and incorporation of Decursin (DEC) into elastin nanogels (ENG) for prostate cancer therapy. In vitro studies demonstrated sustained release of DEC from the ENG over 120 h, with a pH-dependent release pattern. DU145 cell line induces moderate cytotoxicity of DEC-ENG indicates that nanomedicine has an impact on cell viability and helps strike a balance between therapeutics efficacy and safety while the EPR effect enables targeted drug delivery to prostate tumor sites compared to free DEC. Morphological analysis further supported the effectiveness of DEC-ENG in inducing cell death. Overall, these findings highlight the promising role of ENG-encapsulated decursin as a targeted drug delivery system for CRPC.
    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
    Primary liver cancer is one of the most common malignant tumors of the digestive system,of which hepatocellular carcinoma (HCC) accounts for more than 90% of the total cases.The patients with early HCC treated by surgical resection generally demonstrate good prognosis.However,due to the insidious onset,HCC in the vast majority of patients has progressed to the mid-to-late stage when being diagnosed.As a result,surgical treatment has unsatisfactory effects,and non-surgical treatment methods generally have severe side effects and low tumor selectivity.Nanoparticles (NP) with small sizes,large specific surface areas,and unique physical and chemical properties have become potential carriers for the delivery of therapeutic agents such as drugs,genes,and cytokines.The nano-delivery systems with NP as the carrier can regulate the metabolism and transformation of drugs,genes,and cytokines in vivo from time,space,and dose via functional modification,showing great potential in the treatment of HCC.This paper introduces the current status and advantages of several common nano-delivery systems,including organic nano-carriers,inorganic nano-carriers,and exosomes,in the treatment of HCC.Furthermore,this paper summarizes the mechanisms of NP-based nano-carriers in treating HCC and provides reference for the development of new nano-delivery systems.
    原发性肝癌是消化系统最常见的恶性肿瘤之一,其中肝细胞癌(HCC)占90%以上。早期HCC以手术切除为主,且预后较好,然而因HCC起病隐匿,绝大多数患者确诊时已进展至中晚期,手术治疗效果较差,而非手术治疗方式因为普遍存在不良反应大,肿瘤选择性低等问题,疗效也不理想,所以目前中晚期HCC治疗仍是临床工作的难点。纳米粒(NP)尺寸小、比表面积大,具有多种独特的理化性质,成为输送药物、基因及细胞活性因子等治疗剂的潜在载体。纳米递送系统以NP为载体,通过功能化修饰,从时间、空间及剂量上调控药物、基因及细胞活性因子等在体内的代谢及转化,在HCC治疗中展现出巨大的潜力。本文主要介绍了几种常见纳米递送系统,包括有机纳米载体、无机纳米载体、外泌体等在HCC治疗中的现状和优势,总结了基于NP的纳米载体治疗HCC的机制,为新型纳米递送系统的研发提供参考。.
    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
    众所周知,破骨细胞活性受到细胞内pH波动的显著影响。因此,pH敏感的门控纳米药物递送系统代表了减轻破骨细胞过度活性的有希望的治疗方法。我们之前的研究表明,柚皮苷,一种天然类黄酮,有效减轻破骨细胞活性。然而,柚皮苷的口服利用率低,半衰期短,阻碍了其临床应用。我们开发了一种药物递送系统,其中壳聚糖,作为看门人,包覆载有柚皮苷(CS@MSNs-柚皮苷)的介孔二氧化硅纳米颗粒。然而,CS@MSNs-柚皮苷对破骨细胞的抑制作用和潜在机制尚不清楚,保证进一步的研究。
    首先,我们合成了CS@MSNs-柚皮苷,并进行了全面表征。我们还测量了pH梯度溶液中的药物释放速率并验证了其生物安全性。随后,我们研究了CS@MSNs-柚皮苷对骨髓源性巨噬细胞诱导的破骨细胞的影响,在探索潜在机制的同时,重点关注分化和骨吸收活性。最后,我们建立了大鼠双侧临界大小的颅骨缺损模型,其中CS@MSNs-柚皮苷分散在GelMA水凝胶中以实现原位药物递送。我们观察到CS@MSNs-柚皮苷在体内促进骨再生和抑制破骨细胞活性的能力。
    CS@MSNs-柚皮苷表现出高的均匀性和分散性,低细胞毒性(浓度≤120μg/mL),和显著的pH敏感性。体外,与Naringin和MSNs-Naringin相比,CS@MSNs-柚皮苷更有效地抑制破骨细胞的形成和骨吸收活性。这种作用伴随着NF-κB和MAPK信号通路中关键因子的磷酸化减少,细胞凋亡水平增加,以及随后的破骨细胞特异性基因和蛋白质的产生减少。在体内,CS@MSNs-Naringin的表现优于Naringin和MSNs-Naringin,促进新骨形成,同时更大程度地抑制破骨细胞活性。
    我们的研究表明,CS@MSNs-Naringin在体外和体内表现出惊人的抗破骨细胞能力,而且促进颅骨缺损的骨再生。
    UNASSIGNED: It is well-established that osteoclast activity is significantly influenced by fluctuations in intracellular pH. Consequently, a pH-sensitive gated nano-drug delivery system represents a promising therapeutic approach to mitigate osteoclast overactivity. Our prior research indicated that naringin, a natural flavonoid, effectively mitigates osteoclast activity. However, naringin showed low oral availability and short half-life, which hinders its clinical application. We developed a drug delivery system wherein chitosan, as gatekeepers, coats mesoporous silica nanoparticles loaded with naringin (CS@MSNs-Naringin). However, the inhibitory effects of CS@MSNs-Naringin on osteoclasts and the underlying mechanisms remain unclear, warranting further research.
    UNASSIGNED: First, we synthesized CS@MSNs-Naringin and conducted a comprehensive characterization. We also measured drug release rates in a pH gradient solution and verified its biosafety. Subsequently, we investigated the impact of CS@MSNs-Naringin on osteoclasts induced by bone marrow-derived macrophages, focusing on differentiation and bone resorption activity while exploring potential mechanisms. Finally, we established a rat model of bilateral critical-sized calvarial bone defects, in which CS@MSNs-Naringin was dispersed in GelMA hydrogel to achieve in situ drug delivery. We observed the ability of CS@MSNs-Naringin to promote bone regeneration and inhibit osteoclast activity in vivo.
    UNASSIGNED: CS@MSNs-Naringin exhibited high uniformity and dispersity, low cytotoxicity (concentration≤120 μg/mL), and significant pH sensitivity. In vitro, compared to Naringin and MSNs-Naringin, CS@MSNs-Naringin more effectively inhibited the formation and bone resorption activity of osteoclasts. This effect was accompanied by decreased phosphorylation of key factors in the NF-κB and MAPK signaling pathways, increased apoptosis levels, and a subsequent reduction in the production of osteoclast-specific genes and proteins. In vivo, CS@MSNs-Naringin outperformed Naringin and MSNs-Naringin, promoting new bone formation while inhibiting osteoclast activity to a greater extent.
    UNASSIGNED: Our research suggested that CS@MSNs-Naringin exhibited the strikingly ability to anti-osteoclasts in vitro and in vivo, moreover promoted bone regeneration in the calvarial bone defect.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号