drug delivery systems

药物输送系统
  • 文章类型: Journal Article
    Prostate cancer is a prevalent carcinoma among males, and conventional treatment options are often limited. Cytotoxic chemotherapy, despite its drawbacks, remains a mainstay. We propose a targeted co-delivery approach using nanoscale delivery units for Oncolytic measles virus (OMV) and vincristine (VC) to enhance treatment efficacy. The HA-coated OMV + VC-loaded TCs nanoformulation is designed for targeted oncolytic activity in prostate cancer. The CD44 expression analysis in prostate cancer cell lines indicates a significantly high expression in PC3 cells. The optimization of nanoformulations using Design of Expert (DOE) is performed, and the preparation and characterization of HA-coated OMV + VC-loaded TCs nanoformulations are detailed showing average particle size 397.2 ± 0.01 nm and polydispersity index 0.122 with zeta potential 19.7 + 0.01 mV. Results demonstrate successful encapsulation efficiency with 2.4 × 106 TCID50/Ml and sustained release of OMV and VC from the nanoformulation for up to 72 h. In vitro, assays reveal potent anticancer activity at 10 ± 0.71% cell viability in PC3 cells compared to 73 ± 0.66% in HPrEC and significant morphological changes at 90 µg/ml in dose and time-dependent manner. The co-formulation showed positive cell death 49.5 ± 0.02% at 50 µg PI/ml in PBS and 54.3% cell cycle arrest at the G2/M phase, 8.1% G0/G1 and 5.7% at S phase, with significant mitochondrial membrane potential (MMP) at 50 µg/ml, as assessed by flow cytometry (FACS). The surface-integrating ligand approach enhances the targeted delivery of the oncolytic virus and chemotherapeutic drug, presenting a potential alternative for prostate cancer treatment and suggested that co-administering VC and OMV in a nanoformulation could improve therapeutic outcomes while reducing chemotherapeutic drug doses.
    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
    The high malignant degree and poor prognosis of pancreatic cancer (PC) pose severe challenges to the basic research and clinical translation of next-generation therapies. The rise of immunotherapy has improved the treatment of a variety of solid tumors, while the application in PC is highly restricted by the challenge of immunosuppressive tumor microenvironment. The latest progress of nanotechnology as drug delivery platform and immune adjuvant has improved drug delivery in a variety of disease backgrounds and enhanced tumor therapy based on immunotherapy. Based on the immune loop of PC and the status quo of clinical immunotherapy of tumors, this article discussed and critically analyzed the key transformation difficulties of immunotherapy adaptation to the treatment of PC, and then proposed the rational design strategies of new nanocarriers for drug delivery and immune regulation, especially the design of combined immunotherapy. This review also put forward prospective views on future research directions, so as to provide information for the new means of clinical treatment of PC combined with the next generation of nanotechnology and immunotherapy.
    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
    Following nerve injury, growth factors (GFs) are transiently upregulated in injured neurons, proliferating Schwann cells, and denervated muscle and skin. They act on these same cells and tissues to promote nerve regeneration and end-organ reinnervation. Consequently, much attention has been focused on developing GF-based therapeutics. A major barrier to clinical translation of GFs is their short half-life. To provide sustained GF treatment to the affected nerve, muscle, and skin in a safe and practical manner, engineered drug delivery systems are needed. This review highlights recent advancements in GF-based therapeutics and discusses the remaining hurdles for clinical translation.
    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
    白血病是一种起源于骨髓的血液系统恶性肿瘤,它提供了启动的基本信号,programming,和白血病复发。然而,如何特异性地将药物递送到骨髓仍然难以捉摸。这里,我们通过用脂质体(HSPC脂质体)输注造血干细胞和祖细胞(HSPC)膜来开发仿生囊泡,通过透明质酸-CD44轴迁移到白血病小鼠的骨髓。此外,仿生囊泡通过细胞间粘附分子-1(ICAM-1)/整联蛋白β2(ITGB2)相互作用对白血病细胞表现出优异的结合亲和力。进一步的实验证实,携带化疗药物阿糖胞苷(Ara-C@HSPC-Lipo)的囊泡明显抑制细胞增殖,诱导白血病细胞凋亡和分化,减少白血病干细胞的数量。机械上,RNA-seq显示Ara-C@HSPC-Lipo处理诱导细胞凋亡和分化并抑制致癌途径。最后,我们证实HSPC脂质体在小鼠体内是安全的。本研究为靶向骨髓和治疗白血病提供了一种方法。
    Leukemia is a kind of hematological malignancy originating from bone marrow, which provides essential signals for initiation, progression, and recurrence of leukemia. However, how to specifically deliver drugs to the bone marrow remains elusive. Here, we develop biomimetic vesicles by infusing hematopoietic stem and progenitor cell (HSPC) membrane with liposomes (HSPC liposomes), which migrate to the bone marrow of leukemic mice via hyaluronic acid-CD44 axis. Moreover, the biomimetic vesicles exhibit superior binding affinity to leukemia cells through intercellular cell adhesion molecule-1 (ICAM-1)/integrin β2 (ITGB2) interaction. Further experiments validate that the vesicles carrying chemotherapy drug cytarabine (Ara-C@HSPC-Lipo) markedly inhibit proliferation, induce apoptosis and differentiation of leukemia cells, and decrease number of leukemia stem cells. Mechanically, RNA-seq reveals that Ara-C@HSPC-Lipo treatment induces apoptosis and differentiation and inhibits the oncogenic pathways. Finally, we verify that HSPC liposomes are safe in mice. This study provides a method for targeting bone marrow and treating leukemia.
    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
    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
    骨关节炎(OA),影响全球约2.4亿人是一个主要威胁。目前,现有的药物只能治疗OA的症状;它们不能逆转疾病的进展。将药物递送至患病的关节是具有挑战性的,因为关节软骨的不良脉管系统导致它们较低的生物利用度和从关节的快速消除。最近批准的药物如KGN和IL-1受体拮抗剂由于制剂不足也面临挑战。因此,微球由于其优异的理化性质,可能是干预OA的潜在参与者。这篇综述主要关注不同生物材料的微球,这些微球通过不同的递送途径在OA的有效管理中充当药物和生物制品的货物。微球可以通过在特定身体位置的靶向策略来提高治疗剂的功效。这篇综述还强调了过去几十年进行的临床试验。
    Osteoarthritis (OA), affecting around 240 million people globally is a major threat. Currently, available drugs only treat the symptoms of OA; they cannot reverse the disease\'s progression. The delivery of drugs to afflicted joints is challenging because of poor vasculature of articular cartilage results in their less bioavailability and quick elimination from the joints. Recently approved drugs such as KGN and IL-1 receptor antagonists also encounter challenges because of inadequate formulations. Therefore, microspheres could be a potential player for the intervention of OA owing to its excellent physicochemical properties. This review primarily focuses on microspheres of distinct biomaterials acting as cargo for drugs and biologicals via different delivery routes in the effective management of OA. Microspheres can improve the efficacy of therapeutics by targeting strategies at specific body locations. This review also highlights clinical trials conducted in the last few decades.
    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

       PDF(Pubmed)

  • 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

       PDF(Pubmed)

  • 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

公众号