mRNA delivery

mRNA 递送
  • 文章类型: Journal Article
    将mRNA递送到肺中是解决经常发生在肺中的感染性和难治性疾病的关键。由于使用雾化器吸入是将mRNA递送到肺部的最有前途的方法,已经有许多尝试使脂质纳米颗粒适应mRNA吸入。然而,常规脂质纳米粒,它们对mRNA的全身递送和肌内疫苗接种显示出巨大的有效性,由于雾化过程中的结构不稳定性以及无法适应肺微环境,因此对肺部输送无效。为了解决这些问题,我们开发了一种可电离的脂质体-mRNA脂质复合物(iLPX)。iLPX具有高度有序的脂质双层结构,这增加了雾化过程中的稳定性,其不含聚(乙二醇)的组合物允许其渗入低血清环境和肺中的肺表面活性剂层。我们选择了吸入优化的iLPX(IH-iLPX),使用模拟吸入纳米颗粒的肺部递送过程的多步骤筛选程序。与吸入后的常规脂质纳米颗粒相比,IH-iLPX在肺中显示出更高的转染效率,在体内没有观察到毒性。此外,肺分布分析显示在肺深处甚至有蛋白质表达,有效递送到上皮细胞。这项研究提供了与吸入mRNA肺部疗法发展相关的挑战和解决方案的见解。
    The delivery of mRNA into the lungs is the key to solving infectious and intractable diseases that frequently occur in the lungs. Since inhalation using a nebulizer is the most promising method for mRNA delivery into the lungs, there have been many attempts toward adapting lipid nanoparticles for mRNA inhalation. However, conventional lipid nanoparticles, which have shown great effectiveness for systemic delivery of mRNA and intramuscular vaccination, are not effective for pulmonary delivery due to their structural instability during nebulization and their inability to adapt to the pulmonary microenvironment. To address these issues, we developed an ionizable liposome-mRNA lipocomplex (iLPX). iLPX has a highly ordered lipid bilayer structure, which increases stability during nebulization, and its poly(ethylene glycol)-free composition allows it to infiltrate the low serum environment and the pulmonary surfactant layer in the lungs. We selected an inhalation-optimized iLPX (IH-iLPX) using a multistep screening procedure that mimics the pulmonary delivery process of inhaled nanoparticles. The IH-iLPX showed a higher transfection efficiency in the lungs compared to conventional lipid nanoparticles after inhalation with no observed toxicity in vivo. Furthermore, analysis of lung distribution revealed even protein expression in the deep lungs, with effective delivery to epithelial cells. This study provides insights into the challenges and solutions related to the development of inhaled mRNA pulmonary therapeutics.
    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
    大小依赖性吞噬作用是单核细胞和巨噬细胞中充分表征的现象。然而,这种优先基因递送到这些重要细胞靶标的大小效应尚未得到充分利用,因为静电复合核酸纳米颗粒通常采用的稳定方法,如聚乙二醇化和电荷排斥,通常将车辆尺寸限制在200nm以下。这里,我们通过带电纳米粒子的静电自组装来弥合可扩展合成较大亚微米基因载体的技术差距,由结构上设计用于调节纳米粒子间库仑力和范德华力的聚合物促进。具体来说,我们的策略允许将小的聚(β-氨基酯)/信使核糖核酸(mRNA)纳米颗粒受控组装成颗粒,其大小在200至1,000nm之间动力学可调,在生理介质中具有高胶体稳定性。我们发现,平均尺寸为400nm的组装颗粒在静脉内给药后最安全,最有效地转染单核细胞,并介导其分化为外周巨噬细胞。当CpG佐剂与抗原mRNA共同加载到颗粒中时,单核细胞分化成炎性树突状细胞,并在肿瘤引流淋巴结中引发适应性抗癌免疫。这种平台技术提供了一个独特的配体无关,颗粒大小介导的优先mRNA递送策略,并通过单核细胞编程实现治疗范例。
    Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(β-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.
    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
    信使核糖核酸(mRNA)于1961年被发现,用于将遗传信息从DNA转移到核糖体以进行蛋白质合成。COVID-19大流行引起了全球对mRNA疫苗的关注。两种COVID-19mRNA疫苗的紧急使用授权,BNT162b2和mRNA-1273是疫苗发展史上的重大成就。脂质纳米颗粒(LNP),最优秀的非病毒递送载体之一,作为COVID-19疫苗的一部分,在临床翻译方面取得了许多令人兴奋的进展,因此有可能加速许多基因药物的临床翻译。此外,由于这些小尺寸,生物相容性和优异的生物降解性,LNP可以有效地将核酸递送到细胞中,这对于目前的mRNA治疗方案尤为重要。LNP由阳离子或pH依赖性可电离脂质双层组成,聚乙二醇(PEG),磷脂,和胆固醇,代表用于递送mRNA疫苗的先进系统。此外,对构成LNP的这四种组分的优化已经证明了增强的疫苗效力和减少的副作用。生物可降解脂质的掺入增强了LNP的生物相容性,从而提高其作为一种有效的治疗方法的潜力,为广泛的具有挑战性和复杂的疾病,包括传染病,肝脏疾病,癌症,心血管疾病,脑血管条件,在其他人中。因此,本综述旨在为科学界提供有关mRNA疫苗和LNP递送系统的最新信息.
    Messenger ribonucleic acid (mRNA) was discovered in 1961 as an intermediary for transferring genetic information from DNA to ribosomes for protein synthesis. The COVID-19 pandemic brought worldwide attention to mRNA vaccines. The emergency use authorization of two COVID-19 mRNA vaccines, BNT162b2 and mRNA-1273, were major achievements in the history of vaccine development. Lipid nanoparticles (LNPs), one of the most superior non-viral delivery vectors available, have made many exciting advances in clinical translation as part of the COVID-19 vaccine and therefore has the potential to accelerate the clinical translation of many gene drugs. In addition, due to these small size, biocompatibility and excellent biodegradability, LNPs can efficiently deliver nucleic acids into cells, which is particularly important for current mRNA therapeutic regimens. LNPs are composed cationic or pH-dependent ionizable lipid bilayer, polyethylene glycol (PEG), phospholipids, and cholesterol, represents an advanced system for the delivery of mRNA vaccines. Furthermore, optimization of these four components constituting the LNPs have demonstrated enhanced vaccine efficacy and diminished adverse effects. The incorporation of biodegradable lipids enhance the biocompatibility of LNPs, thereby improving its potential as an efficacious therapeutic approach for a wide range of challenging and intricate diseases, encompassing infectious diseases, liver disorders, cancer, cardiovascular diseases, cerebrovascular conditions, among others. Consequently, this review aims to furnish the scientific community with the most up-to-date information regarding mRNA vaccines and LNP delivery systems.
    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
    改造和重编程细胞在治疗多种疾病方面具有巨大的治疗潜力,通过替换缺失或有缺陷的蛋白质,提供转录因子(TFs)来重新编程细胞表型,或提供用于基于CRISPR的细胞工程的RNA指导的Cas9衍生物等酶。而病毒载体介导的基因转移在这一领域发挥了重要作用,mRNA的使用避免了与DNA整合到宿主细胞基因组相关的安全问题,使mRNA对体内应用特别有吸引力。mRNA在细胞工程中的广泛应用受到其在生物环境中的不稳定性和mRNA递送至其靶位点所涉及的挑战的限制。在这次审查中,我们研究开发有效疗法必须克服的挑战。
    Engineering and reprogramming cells has significant therapeutic potential to treat a wide range of diseases, by replacing missing or defective proteins, to provide transcription factors (TFs) to reprogram cell phenotypes, or to provide enzymes such as RNA-guided Cas9 derivatives for CRISPR-based cell engineering. While viral vector-mediated gene transfer has played an important role in this field, the use of mRNA avoids safety concerns associated with the integration of DNA into the host cell genome, making mRNA particularly attractive for in vivo applications. Widespread application of mRNA for cell engineering is limited by its instability in the biological environment and challenges involved in the delivery of mRNA to its target site. In this review, we examine challenges that must be overcome to develop effective therapeutics.
    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疫苗已经彻底改变了疾病的预防和治疗。然而,它们的进一步应用受到炎症副作用的阻碍,主要由递送系统如脂质纳米颗粒(LNP)引起。针对这个问题,我们制备了源自米膦酸盐的阳离子脂质(MLPs),一种小分子药物,随后开发了包含低剂量mLP的LNP(mLNP-69)。与基于SM-102的LNP(sLNP)相比,这是一种市售的可电离脂质,mLNP-69确保有效的mRNA递送,同时显著减少局部炎症。在临床前预防性和治疗性B16-OVA黑色素瘤模型中,mLNP-69证明了在体内成功的mRNA癌症疫苗递送,有效预防肿瘤发生或阻碍肿瘤进展。结果表明,源自米膦酸盐的阳离子脂质,表现出有效的输送能力和最小的炎症副作用,临床应用前景广阔。
    mRNA vaccines have been revolutionizing disease prevention and treatment. However, their further application is hindered by inflammatory side effects, primarily caused by delivery systems such as lipid nanoparticles (LNPs). In response to this issue, we prepared cationic lipids (mLPs) derived from mildronate, a small-molecule drug, and subsequently developed the LNP (mLNP-69) comprising a low dose of mLP. Compared with the LNP (sLNP) based on SM-102, a commercially available ionizable lipid, mLNP-69 ensures effective mRNA delivery while significantly reducing local inflammation. In preclinical prophylactic and therapeutic B16-OVA melanoma models, mLNP-69 demonstrated successful mRNA cancer vaccine delivery in vivo, effectively preventing tumor occurrence or impeding tumor progression. The results suggest that the cationic lipids derived from mildronate, which exhibit efficient delivery capabilities and minimal inflammatory side effects, hold great promise for clinical application.
    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
    脂质纳米颗粒(LNP)设计的进步显着促进了当前临床批准的基于mRNA的疫苗的出现,并且对于递送mRNA以对抗治疗替代品稀疏的疾病具有高度相关性。LNP辅助的mRNA递送利用由内体环境的酸化驱动的可电离的脂质介导的货物转运穿过内体膜。然而,这个过程的效率很低,最好的百分之几。利用具有单一LNP和mRNA分辨率的表面敏感荧光显微镜,我们研究了单个LNP和在纳米多孔二氧化硅上形成的平面阴离子负载脂质双层(SLB)之间的pH控制相互作用,模仿早期内体膜的静电条件。对于平均直径为140nm的LNP,当pH从6.6降低到6.0时,优先发生与阴离子SLB的融合。此外,pH下降后,LNP融合的开始延迟,融合后,显著部分(>70%)的mRNA被释放到代表内体腔的酸性溶液中,而即使在将pH逆转至中性胞质条件后,一部分mRNA仍与SLB结合。最后,两种LNP制剂与不同表面浓度的凝胶形成脂质的融合效率的比较与先前在人原代细胞转染研究中观察到的蛋白质翻译效率的差异相关。一起,这些发现强调了包含可电离脂质的LNP辅助mRNA递送机制的生物物理研究的相关性,同时也可能提供优化具有增强内体逃逸能力的LNP设计的方法.
    Advances in lipid nanoparticle (LNP) design have contributed notably to the emergence of the current clinically approved mRNA-based vaccines and are of high relevance for delivering mRNA to combat diseases where therapeutic alternatives are sparse. LNP-assisted mRNA delivery utilizes ionizable lipid-mediated cargo translocation across the endosomal membrane driven by the acidification of the endosomal environment. However, this process occurs at a low efficiency, a few percent at the best. Utilizing surface-sensitive fluorescence microscopy with a single LNP and mRNA resolution, we have investigated pH-controlled interactions between individual LNPs and a planar anionic supported lipid bilayer (SLB) formed on nanoporous silica, mimicking the electrostatic conditions of the early endosomal membrane. For LNPs with an average diameter of 140 nm, fusion with the anionic SLB preferentially occurred when the pH was reduced from 6.6 to 6.0. Furthermore, there was a delay in the onset of LNP fusion after the pH drop, and upon fusion, a significant fraction (>70%) of mRNA was released into the acidic solution representing the endosomal lumen, while a fraction of mRNA remained bound to the SLB even after reversing the pH to neutral cytosolic conditions. Finally, a comparison of the fusion efficiency of two LNP formulations with different surface concentrations of gel-forming lipids correlated with differences in the protein translation efficiency previously observed in human primary cell transfection studies. Together, these findings emphasize the relevance of biophysical investigations of ionizable lipid-containing LNP-assisted mRNA delivery mechanisms while potentially also offering means to optimize the design of LNPs with enhanced endosomal escape capabilities.
    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
    纳米颗粒介导的mRNA递送已成为一种有前途的治疗方式,但它的增长仍然受到有效和耐受性良好的交付策略的发现和优化的限制。含有带电或可电离脂质的脂质纳米颗粒是体内mRNA递送的新兴标准,所以创造便利,合成这些关键类脂分子的可调策略对于推进该领域至关重要。这里,我们产生了一个N-取代的甘氨酸寡聚体文库,peptoids,并进行多阶段向下选择过程,以在我们的果壳纳米颗粒平台中确定前导候选肽作为电离成分。首先,我们通过基于预测的物理性质对>200个分子的文库进行聚类来鉴定有希望的类肽结构基序,并评估每个簇的成员在体内的报告基因表达。然后,铅类肽基序使用实验设计方法进行优化,以探索类肽的带电和亲脂性部分的变化,有利于发现结构元素和纳米颗粒性质之间的趋势。我们进一步证明,与基准可电离脂质相比,基于类肽的果壳导致小鼠中治疗相关水平的抗呼吸道合胞病毒抗体表达,耐受性问题或诱导的免疫反应最小。DLin-MC3-DMA。通过这项工作,我们提出了基于类肽的纳米颗粒作为一个可调的递送平台,可以针对一系列治疗方案进行优化。
    Nanoparticle-mediated mRNA delivery has emerged as a promising therapeutic modality, but its growth is still limited by the discovery and optimization of effective and well-tolerated delivery strategies. Lipid nanoparticles containing charged or ionizable lipids are an emerging standard for in vivo mRNA delivery, so creating facile, tunable strategies to synthesize these key lipid-like molecules is essential to advance the field. Here, we generate a library of N-substituted glycine oligomers, peptoids, and undertake a multistage down-selection process to identify lead candidate peptoids as the ionizable component in our Nutshell nanoparticle platform. First, we identify a promising peptoid structural motif by clustering a library of >200 molecules based on predicted physical properties and evaluate members of each cluster for reporter gene expression in vivo. Then, the lead peptoid motif is optimized using design of experiments methodology to explore variations on the charged and lipophilic portions of the peptoid, facilitating the discovery of trends between structural elements and nanoparticle properties. We further demonstrate that peptoid-based Nutshells leads to expression of therapeutically relevant levels of an anti-respiratory syncytial virus antibody in mice with minimal tolerability concerns or induced immune responses compared to benchmark ionizable lipid, DLin-MC3-DMA. Through this work, we present peptoid-based nanoparticles as a tunable delivery platform that can be optimized toward a range of therapeutic programs.
    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
    使用信使RNA(mRNA)作为癌症疫苗和基因治疗需要靶向载体递送到疾病部位。这里,我们设计了一种包裹mRNA的脂质纳米颗粒(LNP)与抗程序性死亡配体1(PD-L1)DNA适体结合,提供编码肿瘤抑制基因的mRNA,即磷酸酶和张力蛋白同源物(PTEN),去势抵抗性前列腺癌(CRPC)细胞在细胞表面表达PD-L1。与没有靶向配体的LNP相比,DNA适体缀合的基于LNP的mRNA递送系统(Apt-LNP[PTENmRNA])在CRPC细胞中介导有效的mRNA递送和转染。与非靶向LNP递送相比,使用Apt-LNP的癌症靶向PTENmRNA递送通过适体介导的内吞作用在靶癌细胞中实现了显著更高的PTEN表达。导致AKT磷酸化显著下调。这增强了PI3K/AKT通路的调控,两天后又减少了细胞迁移,细胞活力下降了70%,导致有效的凋亡细胞死亡。在CRPC异种移植模型中,Apt-LNP[PTENmRNA]导致肿瘤生长减少约60%,这归因于有效的PTEN恢复和PI3K/AKT信号通路调节。PTEN在CRPC肿瘤组织中表达显著增强,消除了癌细胞的致瘤性。这些发现证明了Apt-LNPs靶向mRNA递送到癌细胞的潜力。从而为使用常规LNP系统将mRNA靶向递送至一系列癌症和组织提供了有希望的工具。
    The use of messenger RNA (mRNA) as a cancer vaccine and gene therapy requires targeted vehicle delivery to the site of disease. Here, we designed a mRNA-encapsulating lipid nanoparticle (LNP) conjugated with anti-programmed death-ligand 1 (PD-L1) DNA aptamer that delivers mRNA encoding a tumor suppressor gene, namely phosphatase and tensin homolog (PTEN), to castration-resistant prostate cancer (CRPC) cells expressing PD-L1 on the cell surface. The DNA aptamer-conjugated LNP-based mRNA delivery system (Apt-LNP[PTEN mRNA]) mediated efficient mRNA delivery and transfection in CRPC cells than LNPs without targeting ligands. Cancer-targeted PTEN mRNA delivery using Apt-LNPs achieved significantly higher PTEN expression via aptamer-mediated endocytosis in target cancer cells compared with non-targeted LNP delivery, resulting in significant downregulation of AKT phosphorylation. This enhanced PI3K/AKT pathway regulation, and in turn reduced cell migration after two days along with a 70 % decrease in cell viability, leading to effective apoptotic cell death. In a CRPC xenograft model, Apt-LNP[PTEN mRNA] led to an approximate 60 % reduction in tumor growth, which was attributable to the effective PTEN restoration and PI3K/AKT signaling pathway regulation. PTEN expression was significantly enhanced in CRPC tumor tissues, which abolished cancer cell tumorigenicity. These findings demonstrated the potential of Apt-LNPs for targeted mRNA delivery to cancer cells, thus providing a promising tool for targeted mRNA delivery to a range of cancers and tissues using a conventional LNP systems.
    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递送系统,如脂质纳米颗粒(LNP),在改善mRNA表达方面取得了显著进展,而免疫系统激活作用在一个阈值上。维持抗原表达和树突状细胞(DC)活化之间的微妙平衡对于有效的免疫识别至关重要。这里,开发了用磷酸钙纳米颗粒(CaP-PME)稳定的水包油包水(w/o/w)皮克林乳液,用于癌症疫苗接种中的mRNA递送。CaP-PME有效地将mRNA转运到细胞质中,诱导促炎反应并通过破坏细胞内钙/钾离子平衡激活DC。与LNP不同,CaP-PME显示出对DC的偏好,增强它们的激活和向淋巴结的迁移。它引发干扰素-γ介导的CD8+T细胞反应,促进NK细胞增殖和活化,导致明显的NK细胞浸润和改善肿瘤微环境。制备的w/o/wPickering乳剂在E.G7和B16-OVA肿瘤模型中表现出优异的抗肿瘤效果,作为癌症疫苗接种的增强mRNA递送载体,提供了有希望的前景。
    mRNA delivery systems, such as lipid nanoparticle (LNP), have made remarkable strides in improving mRNA expression, whereas immune system activation operates on a threshold. Maintaining a delicate balance between antigen expression and dendritic cell (DC) activation is vital for effective immune recognition. Here, a water-in-oil-in-water (w/o/w) Pickering emulsion stabilized with calcium phosphate nanoparticles (CaP-PME) is developed for mRNA delivery in cancer vaccination. CaP-PME efficiently transports mRNA into the cytoplasm, induces pro-inflammatory responses and activates DCs by disrupting intracellular calcium/potassium ions balance. Unlike LNP, CaP-PME demonstrates a preference for DCs, enhancing their activation and migration to lymph nodes. It elicits interferon-γ-mediated CD8+ T cell responses and promotes NK cell proliferation and activation, leading to evident NK cells infiltration and ameliorated tumor microenvironment. The prepared w/o/w Pickering emulsion demonstrates superior anti-tumor effects in E.G7 and B16-OVA tumor models, offering promising prospects as an enhanced mRNA delivery vehicle for cancer vaccinations.
    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
    通过靶向细胞本身来调节免疫细胞的功能已成为免疫疗法对抗癌症等各种疾病的关键策略。自身免疫性疾病,和传染病。mRNA(mRNA)的使用是短暂诱导蛋白质表达的强大工具,通常用于遗传操作。然而,其固有的不稳定性和无法精确地到达靶细胞需要使用生物材料来安全和有效地递送。此外,由于其抗性机制,转染免疫细胞是困难和复杂的,信号通路,和细胞相互作用。本文综述了用于将mRNA传递到免疫细胞的生物材料的最新发展。包括脂质纳米颗粒和聚合物载体。它还概述了靶向和向免疫细胞提供治疗有效载荷的挑战,对下一代材料的设计进行评论和展望。最后,这种方法有可能显着提高各种疾病治疗干预措施的准确性和有效性,塑造免疫疾病mRNA递送的未来。
    Modulating the function of immune cells by targeting the cells themselves has become a key strategy in immunotherapy for combating various diseases such as cancer, autoimmune disorders, and infectious ailments. The use of mRNA (mRNA) is a powerful tool for transiently inducing protein expression, which is often used for genetic manipulation. However, its inherent instability and inability to precisely reach target cells necessitate the use of biomaterials for safe and effective delivery. Additionally, transfecting immune cells is difficult and complex due to their resistance mechanisms, signaling pathways, and cellular interactions. This review focuses on the recent development of biomaterials for mRNA delivery to immune cells, including lipid nanoparticles and polymeric carriers. It also outlines the challenges of targeting and delivering therapeutic payloads to immune cells, providing commentary and outlook on the design of next-generation materials. Finally, this approach has the potential to significantly enhance the precision and effectiveness of therapeutic interventions for various diseases, shaping the future of mRNA delivery for immune conditions.
    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