Targeted delivery

有针对性的交付
  • 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
    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
    外来体是由几乎所有类型的细胞分泌的纳米级膜结合囊泡。它们独特的属性,如最小的免疫原性和与生物系统的兼容性,使它们成为新型的药物输送载体。这些天然的外泌体含有蛋白质,核酸,小分子化合物,和荧光剂。此外,通过化学和生物工程方法的结合,外泌体被定制为将精确的治疗有效载荷运输到指定的细胞或组织。在这次审查中,我们总结了工程外泌体中外泌体修饰和药物装载方式的策略。此外,我们概述了工程化外泌体用于靶向药物递送的研究进展.最后,我们讨论了化学工程与生物工程外泌体介导的靶向治疗的优点和局限性.这些见解为在药物开发中改进工程外泌体提供了额外的选择,并有望加快工程外泌体从工作台到床边的成功翻译。
    Exosomes are nanoscale membrane bound vesicles secreted by almost all types of cells. Their unique attributes, such as minimal immunogenicity and compatibility with biological systems, make them novel carriers for drug delivery. These native exosomes harbor proteins, nucleic acids, small molecule compounds, and fluorogenic agents. Moreover, through a combination of chemical and bioengineering methodologies, exosomes are tailored to transport precise therapeutic payloads to designated cells or tissues. In this review, we summarize the strategies for exosome modification and drug loading modalities in engineered exosomes. In addition, we provide an overview of the advances in the use of engineered exosomes for targeted drug delivery. Lastly, we discuss the merits and limitations of chemically engineered versus bioengineered exosome-mediated target therapies. These insights offer additional options for refining engineered exosomes in pharmaceutical development and hold promise for expediting the successful translation of engineered exosomes from the bench to the bedside.
    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
    佐剂在疫苗诱导有效免疫应答中起关键作用。这里,制备将佐剂功能整合到递送载体中的自组装纳米疫苗平台。将阳离子香菇多糖(CLNT)与卵清蛋白(OVA)混合以获得自组装纳米疫苗(CLNTO纳米疫苗),通过toll样受体2/4(TLR2/4)诱导骨髓树突状细胞(BMDC)的摄取和成熟,以产生有效的抗原交叉呈递。CLNTO纳米疫苗靶向淋巴结(LN),并通过TLR和肿瘤坏死因子(TNF)信号通路诱导强大的OVA特异性免疫反应,视黄酸诱导基因I(RIG-I)受体,和细胞因子-细胞因子受体相互作用。此外,发现CLNTO纳米疫苗可促进滤泡辅助性T(Tfh)细胞的激活,并诱导生发中心(GC)B细胞分化为记忆B细胞和浆细胞,从而增强免疫反应。用CLNTO纳米疫苗接种显着抑制表达卵清蛋白(OVA)的B16黑色素瘤细胞(B16-OVA)肿瘤的生长,表明其在癌症免疫疗法方面的巨大潜力。因此,这项研究提出了一个简单的,安全,和有效的自组装纳米疫苗,诱导辅助性T细胞1(Th1)和辅助性T细胞(Th2)免疫反应,使其成为有效的疫苗输送系统。
    Adjuvants play a critical role in the induction of effective immune responses by vaccines. Here, a self-assembling nanovaccine platform that integrates adjuvant functions into the delivery vehicle is prepared. Cationic Lentinan (CLNT) is mixed with ovalbumin (OVA) to obtain a self-assembling nanovaccine (CLNTO nanovaccine), which induces the uptake and maturation of bone marrow dendritic cells (BMDCs) via the toll-like receptors 2/4 (TLR2/4) to produce effective antigen cross-presentation. CLNTO nanovaccines target lymph nodes (LNs) and induce a robust OVA-specific immune response via TLR and tumor necrosis factor (TNF) signaling pathways, retinoic acid-inducible gene I (RIG-I) receptor, and cytokine-cytokine receptor interactions. In addition, CLNTO nanovaccines are found that promote the activation of follicular helper T (Tfh) cells and induce the differentiation of germinal center (GC) B cells into memory B cells and plasma cells, thereby enhancing the immune response. Vaccination with CLNTO nanovaccine significantly inhibits the growth of ovalbumin (OVA)-expressing B16 melanoma cell (B16-OVA) tumors, indicating its great potential for cancer immunotherapy. Therefore, this study presents a simple, safe, and effective self-assembling nanovaccine that induces helper T cell 1 (Th1) and helper T cell (Th2) immune responses, making it an effective vaccine delivery system.
    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
    中枢神经系统(CNS)肿瘤是严重的难治性疾病,预后不良。特别是恶性胶质母细胞瘤和脑转移患者。目前,许多研究已经探索了细菌和肠道菌群在肿瘤发展和治疗中的潜在作用。细菌可以穿透血脑屏障(BBB),靶向肿瘤核心的缺氧微环境,从而消除肿瘤并激活先天和适应性免疫反应,使它们成为中枢神经系统肿瘤的有希望的治疗剂。此外,工程细菌和衍生物,如细菌膜蛋白和细菌孢子,还可以作为靶向给药的良好候选载体。此外,肠道菌群可以通过“肠-脑轴”调节中枢神经系统肿瘤代谢并影响免疫微环境。因此,细菌抗肿瘤治疗,工程化细菌靶向药物递送,和肠道菌群的干预为CNS肿瘤的治疗提供了治疗方式。在本文中,我们对细菌治疗中枢神经系统肿瘤的机制和治疗实践进行了全面回顾,并讨论了该领域未来潜在的研究方向.
    Tumors of the central nervous system (CNS) are severe and refractory diseases with poor prognosis, especially for patients with malignant glioblastoma and brain metastases. Currently, numerous studies have explored the potential role of bacteria and intestinal flora in tumor development and treatment. Bacteria can penetrate the blood-brain barrier (BBB), targeting the hypoxic microenvironment at the core of tumors, thereby eliminating tumors and activating both the innate and adaptive immune responses, rendering them promising therapeutic agents for CNS tumors. In addition, engineered bacteria and derivatives, such as bacterial membrane proteins and bacterial spores, can also be used as good candidate carriers for targeted drug delivery. Moreover, the intestinal flora can regulate CNS tumor metabolism and influence the immune microenvironment through the \"gut-brain axis\". Therefore, bacterial anti-tumor therapy, engineered bacterial targeted drug delivery, and intervention of the intestinal flora provide therapeutic modalities for the treatment of CNS tumors. In this paper, we performed a comprehensive review of the mechanisms and therapeutic practices of bacterial therapy for CNS tumors and discussed potential future research directions in this field.
    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
    纳米工程已经成为癌症治疗的一种渐进方法,提供精确和靶向的治疗剂递送,同时降低整体毒性。这篇学术文章探讨了纳米工程的创新策略和进步,弥合了临床实践和癌症治疗领域研究之间的差距。各种纳米工程平台,如纳米颗粒,脂质体,和树枝状聚合物被仔细审查它们封装药物的能力,增强药物功效,并增强药代动力学。此外,这篇文章调查了推动纳米工程疗法进步和增强的研究突破,包括生物标志物的识别,建立临床前模型,和生物材料的进步,所有这些对于将实验室研究结果转化为实际的医疗干预措施都是必不可少的。此外,纳米技术与成像模式的整合,放大了癌症的检测,治疗监测,和反应评估,被彻底检查。最后,纳米工程的障碍和未来方向,包括与可扩展性相关的监管挑战和问题,被检查。这强调了促进各种实体之间合作的重要性,以便有效地将纳米工程干预措施转化为增强的癌症治疗和患者管理。
    Nanoengineering has emerged as a progressive method in cancer treatment, offering precise and targeted delivery of therapeutic agents while concurrently reducing overall toxicity. This scholarly article delves into the innovative strategies and advancements in nanoengineering that bridge the gap between clinical practice and research in the field of cancer treatment. Various nanoengineered platforms such as nanoparticles, liposomes, and dendrimers are scrutinized for their capacity to encapsulate drugs, augment drug efficacy, and enhance pharmacokinetics. Moreover, the article investigates research breakthroughs that drive the progression and enhancement of nanoengineered remedies, encompassing the identification of biomarkers, establishment of preclinical models, and advancement of biomaterials, all of which are imperative for translating laboratory findings into practical medical interventions. Furthermore, the integration of nanotechnology with imaging modalities, which amplify cancer detection, treatment monitoring, and response assessment, is thoroughly examined. Finally, the obstacles and prospective directions in nanoengineering, including regulatory challenges and issues related to scalability, are examined. This underscores the significance of fostering collaboration among various entities in order to efficiently translate nanoengineered interventions into enhanced cancer therapies and patient management.
    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
    胶质母细胞瘤,可怕的脑癌,由于其侵略性和对常规治疗的抵抗力,仍然是治疗挑战。最近的数据表明适体,短的合成DNA或RNA分子由于其更好的肿瘤渗透性,可用于抗癌治疗,特异性结合亲和力,在肿瘤部位保留更长的时间和它们穿过血脑屏障的能力。通过选择过程修饰这些寡核苷酸的能力,并使用合理的设计来修改它们,后SELEX适体在胶质母细胞瘤治疗中提供了几个优势,包括精确靶向癌细胞,同时保留健康组织。这篇综述讨论了适体在胶质母细胞瘤治疗和诊断中的关键作用。强调它们提高治疗疗效的潜力,并强调基于适体的疗法的最新进展,这些疗法可以改变胶质母细胞瘤治疗的前景,给患者和临床医生带来新的希望。
    Glioblastoma, a formidable brain cancer, has remained a therapeutic challenge due to its aggressive nature and resistance to conventional treatments. Recent data indicate that aptamers, short synthetic DNA or RNA molecules can be used in anti-cancer therapy due to their better tumour penetration, specific binding affinity, longer retention in tumour sites and their ability to cross the blood-brain barrier. With the ability to modify these oligonucleotides through the selection process, and using rational design to modify them, post-SELEX aptamers offer several advantages in glioblastoma treatment, including precise targeting of cancer cells while sparing healthy tissue. This review discusses the pivotal role of aptamers in glioblastoma therapy and diagnosis, emphasising their potential to enhance treatment efficacy and also highlights recent advancements in aptamer-based therapies which can transform the landscape of glioblastoma treatment, offering renewed hope to patients and clinicians alike.
    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
    靶向端粒维持已成为肝细胞癌(HCC)治疗的有希望的策略。然而,考虑到端粒-端粒酶轴在端粒维持中的二重性,迫切需要一项全面的战略。在这里,我们开发了一种基于聚(氨基酸)(D-PAAs)的策略,用于端粒酶抑制剂的时空共递送,BIBR1523和AKT抑制剂,异叶瓦查酮。通过利用D-PAA的可修改性,我们合成了聚合物-抑制剂缀合物(PB和PI)和叶酸修饰的肿瘤靶向载体(PF)。这些结构单元通过利用D-PAAs的非共价组装进行胶束化,以制造共递送纳米药物(P-BI@P-FA)。P-BI@P-FA改善了药代动力学,肿瘤选择性,和小分子抑制剂的生物利用度,并通过结合端粒酶失活和端粒破坏启动双重端粒特异性抑制。此外,使用D-PAAs和二氧化锰设计了一种混合肿瘤靶向磁性纳米系统,以展示磁共振成像能力。我们基于D-PAA的策略解决了端粒特异性HCC治疗的迫切需要,同时允许诊断应用。为纳米医学设计提供了一条有希望的途径。
    Targeting telomere maintenance has emerged as a promising strategy for hepatocellular carcinoma (HCC) treatment. However, given the duality of the telomere-telomerase axis in telomere maintenance, a comprehensive strategy is urgently needed. Herein, we develop a poly(amino acid) (D-PAAs)-based strategy for spatiotemporal codelivery of telomerase inhibitor, BIBR1523, and AKT inhibitor, isobavachalcone. By leveraging D-PAAs\' modifiability, we synthesize polymer-inhibitor conjugates (PB and PI) and a folic acid-decorated tumor-targeting vector (PF). These building blocks undergo micellization to fabricate a codelivery nanomedicine (P-BI@P-FA) by exploiting D-PAAs\' noncovalent assembly. P-BI@P-FA improves the pharmacokinetics, tumor selectivity, and bioavailability of small molecule inhibitors and initiates a dual telomere-specific inhibition by combining telomerase deactivation with telomere disruption. Furthermore, a hybrid tumor-targeting magnetic nanosystem is designed using D-PAAs and manganese dioxide to showcase magnetic resonance imaging capacities. Our D-PAAs-based strategy addresses the pressing need for telomere-specific HCC treatment while allowing for diagnostic application, presenting a promising avenue for nanomedicine design.
    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
    益生菌是生活在胃肠道中并为消费者提供健康益处的活微生物。然而,必须将足够数量的活益生菌细胞递送到胃肠道中的特定目的部位以发挥这些益处。对于口服消耗的益生菌,使用适当的包衣材料和食物基质可以实现对亚致死胃肠应激的增强的活力和耐受性。迄今为止,文献中已经对益生菌微胶囊的释放机制和与胃肠道的相互作用进行了最低限度的探索。就作者所知,目前还没有发表综述来讨论益生菌的释放性质和靶向递送面临的挑战.这篇综述讨论了益生菌菌株靶向递送和控释制剂中胃肠道相关的并发症。它研究了在过渡阶段和向胃肠道中的目标区域递送的环境压力的影响。pH值等因素的影响,酶促降解,以及氧化还原条件对益生菌释放机制的影响。最后,评估益生菌递送系统效率的可用方法,包括体外和体内,进行审查和评估。本文最后讨论了该领域的新兴技术,并强调了未来需要研究的关键领域。
    Probiotics are live microorganisms that inhabit the gastrointestinal tract and confer health benefits to consumers. However, a sufficient number of viable probiotic cells must be delivered to the specific site of interest in the gastrointestinal tract to exert these benefits. Enhanced viability and tolerance to sublethal gastrointestinal stress can be achieved using appropriate coating materials and food matrices for orally consumed probiotics. The release mechanism and interaction of probiotic microcapsules with the gastrointestinal tract have been minimally explored in the literature to date. To the authors\' knowledge, no review has been published to discuss the nature of release and the challenges in the targeted delivery of probiotics. This review addresses gastrointestinal-related complications in the formulation of targeted delivery and controlled release of probiotic strains. It investigates the impacts of environmental stresses during the transition stage and delivery to the target region in the gastrointestinal tract. The influence of factors such as pH levels, enzymatic degradation, and redox conditions on the release mechanisms of probiotics is presented. Finally, the available methods to evaluate the efficiency of a probiotic delivery system, including in vitro and in vivo, are reviewed and assessed. The paper concludes with a discussion highlighting the emerging technologies in the field and emphasising key areas in need of future study.
    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
    生物正交反应通过实现活生物体和细胞内的选择性化学转化而彻底改变了化学生物学。这篇综述全面探讨了生物正交化学,强调四嗪和应变亲二烯体之间的反电子需求Diels-Alder(IEDDA)反应及其在化学生物学和人体各种应用中的关键作用。这种高度反应性和选择性的反应发现不同的应用,包括裂解抗体-药物缀合物,前药,蛋白质,肽抗原,和酶底物。多功能性延伸到水凝胶化学,这对生物医学应用至关重要,然而,它在实现精确细胞化方面面临挑战。来自可注射生物聚合物的细胞毒性化合物的原位激活属于点击激活的抗癌原药(CAPAC)平台,肿瘤靶向前药递送和激活的创新方法。CAPAC平台,依靠反式环辛烯(TCO)和四嗪修饰的生物聚合物之间的点击化学,在不同的肿瘤特征中表现出模块化,在抗癌治疗中提出了一种有希望的方法。该综述强调了生物正交反应在开发用于正电子发射断层扫描(PET)成像和治疗的放射性药物中的重要性,为多样化的治疗应用提供了一个有希望的途径。
    Bioorthogonal reactions have revolutionized chemical biology by enabling selective chemical transformations within living organisms and cells. This review comprehensively explores bioorthogonal chemistry, emphasizing inverse-electron-demand Diels-Alder (IEDDA) reactions between tetrazines and strained dienophiles and their crucial role in chemical biology and various applications within the human body. This highly reactive and selective reaction finds diverse applications, including cleaving antibody-drug conjugates, prodrugs, proteins, peptide antigens, and enzyme substrates. The versatility extends to hydrogel chemistry, which is crucial for biomedical applications, yet it faces challenges in achieving precise cellularization. In situ activation of cytotoxic compounds from injectable biopolymer belongs to the click-activated protodrugs against cancer (CAPAC) platform, an innovative approach to tumor-targeted prodrug delivery and activation. The CAPAC platform, relying on click chemistry between trans-cyclooctene (TCO) and tetrazine-modified biopolymers, exhibits modularity across diverse tumor characteristics, presenting a promising approach in anticancer therapeutics. The review highlights the importance of bioorthogonal reactions in developing radiopharmaceuticals for positron emission tomography (PET) imaging and theranostics, offering a promising avenue for diverse therapeutic applications.
    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