首页 > 最新文献

Journal of Controlled Release最新文献

英文 中文
Engineered lipid nanoparticles enable therapeutic gene silencing of GTSE1 for the treatment of liver fibrosis 经改造的脂质纳米粒子可实现 GTSE1 的治疗性基因沉默,用于治疗肝纤维化。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.jconrel.2024.08.012

Liver fibrosis is characterized by abnormal accumulation of extracellular matrix proteins, disrupting normal liver function. Despite its significant health impact, effective treatments remain limited. Here, we present the development of engineered lipid nanoparticles (LNPs) for targeted RNA therapeutic delivery in the liver. We investigated the therapeutic potential of modulating the G2 and S-phase expressed 1 (GTSE1) protein for treating liver fibrosis. Through screening, we identified P138Y LNP as a potent candidate with superior delivery efficiency and lower toxicity. Using these engineered LNPs, we successfully delivered siGTSE1 to hepatocytes, significantly reducing collagen accumulation and restoring liver function in a fibrosis animal model. Additionally, GTSE1 downregulation altered miRNA expression and upregulated hepatocyte nuclear factor 4 alpha (HNF4α). These findings suggest that therapeutic gene silencing of GTSE1 is a promising strategy for treating liver fibrosis by regenerating liver phenotypes and functions.

肝纤维化的特点是细胞外基质蛋白异常积聚,破坏肝脏的正常功能。尽管肝纤维化对健康有重大影响,但有效的治疗方法仍然有限。在此,我们介绍了用于肝脏靶向 RNA 治疗递送的工程脂质纳米颗粒(LNPs)的开发情况。我们研究了调节 G2 和 S 期表达 1(GTSE1)蛋白治疗肝纤维化的潜力。通过筛选,我们发现 P138Y LNP 是一种有效的候选药物,具有更高的递送效率和更低的毒性。利用这些工程化 LNPs,我们成功地将 siGTSE1 运送到肝细胞中,在肝纤维化动物模型中显著减少了胶原蛋白的积累并恢复了肝功能。此外,GTSE1 的下调改变了 miRNA 的表达,并上调了肝细胞核因子 4 alpha(HNF4α)。这些研究结果表明,治疗性基因沉默GTSE1是通过再生肝脏表型和功能来治疗肝纤维化的一种有前途的策略。
{"title":"Engineered lipid nanoparticles enable therapeutic gene silencing of GTSE1 for the treatment of liver fibrosis","authors":"","doi":"10.1016/j.jconrel.2024.08.012","DOIUrl":"10.1016/j.jconrel.2024.08.012","url":null,"abstract":"<div><p>Liver fibrosis is characterized by abnormal accumulation of extracellular matrix proteins, disrupting normal liver function. Despite its significant health impact, effective treatments remain limited. Here, we present the development of engineered lipid nanoparticles (LNPs) for targeted RNA therapeutic delivery in the liver. We investigated the therapeutic potential of modulating the G2 and S-phase expressed 1 (GTSE1) protein for treating liver fibrosis. Through screening, we identified P13<img>8Y LNP as a potent candidate with superior delivery efficiency and lower toxicity. Using these engineered LNPs, we successfully delivered siGTSE1 to hepatocytes, significantly reducing collagen accumulation and restoring liver function in a fibrosis animal model. Additionally, GTSE1 downregulation altered miRNA expression and upregulated hepatocyte nuclear factor 4 alpha (HNF4α). These findings suggest that therapeutic gene silencing of GTSE1 is a promising strategy for treating liver fibrosis by regenerating liver phenotypes and functions.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142000024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stealth mRNA nanovaccines to control lymph node trafficking 控制淋巴结迁移的隐形 mRNA 纳米疫苗。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.jconrel.2024.08.018

mRNA-based vaccines symbolize a new paradigm shift in personalized medicine for the treatment of infectious and non-infectious diseases. However, the reactogenicity associated with the currently approved formulations limits their applicability in autoinflammatory disorders, such as tumour therapeutics. In this study, we present a delivery system showing controlled immunogenicity and minimal non-specific inflammation, allowing for selective delivery of mRNA to antigen presenting cells (APCs) within the medullary region of the lymph nodes. Our platform offers precise control over the trafficking of nanoparticles within the lymph nodes by optimizing stealth and targeting properties, as well as the subsequent opsonization process. By targeting specific cells, we observed a potent adaptive and humoral immune response, which holds promise for preventive and therapeutic anti-tumoral vaccines. Through spatial programming of nanoparticle distribution, we can promote robust immunization, thus improving and expanding the utilization of mRNA vaccines. This innovative approach signifies a remarkable step forward in the field of targeted nanomedicine.

基于 mRNA 的疫苗标志着治疗传染性和非传染性疾病的个性化医疗模式的新转变。然而,目前已获批准的制剂所具有的致反应性限制了其在自身炎症性疾病(如肿瘤治疗)中的适用性。在这项研究中,我们提出了一种可控免疫原性和最小非特异性炎症的递送系统,可将 mRNA 选择性地递送至淋巴结髓质区的抗原呈递细胞(APC)。我们的平台通过优化隐身和靶向特性以及随后的疏松过程,对纳米粒子在淋巴结内的运输进行了精确控制。通过靶向特定细胞,我们观察到了强大的适应性和体液免疫反应,这为预防性和治疗性抗肿瘤疫苗带来了希望。通过对纳米粒子分布的空间编程,我们可以促进强效免疫,从而提高和扩大 mRNA 疫苗的利用率。这一创新方法标志着靶向纳米医学领域向前迈出了重要一步。
{"title":"Stealth mRNA nanovaccines to control lymph node trafficking","authors":"","doi":"10.1016/j.jconrel.2024.08.018","DOIUrl":"10.1016/j.jconrel.2024.08.018","url":null,"abstract":"<div><p>mRNA-based vaccines symbolize a new paradigm shift in personalized medicine for the treatment of infectious and non-infectious diseases. However, the reactogenicity associated with the currently approved formulations limits their applicability in autoinflammatory disorders, such as tumour therapeutics. In this study, we present a delivery system showing controlled immunogenicity and minimal non-specific inflammation, allowing for selective delivery of mRNA to antigen presenting cells (APCs) within the medullary region of the lymph nodes. Our platform offers precise control over the trafficking of nanoparticles within the lymph nodes by optimizing stealth and targeting properties, as well as the subsequent opsonization process. By targeting specific cells, we observed a potent adaptive and humoral immune response, which holds promise for preventive and therapeutic anti-tumoral vaccines. Through spatial programming of nanoparticle distribution, we can promote robust immunization, thus improving and expanding the utilization of mRNA vaccines. This innovative approach signifies a remarkable step forward in the field of targeted nanomedicine.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0168365924005571/pdfft?md5=3668865831021e3eea666698f4124a7d&pid=1-s2.0-S0168365924005571-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142000025","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fluorogenic RNA-based biomaterials for imaging and tracking the cargo of extracellular vesicles 基于荧光 RNA 的生物材料,用于成像和跟踪细胞外囊泡的货物。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.jconrel.2024.07.043

Extracellular vesicles (EVs), or exosomes, play important roles in physiological and pathological cellular communication and have gained substantial traction as biological drug carriers. EVs contain both short and long non-coding RNAs that regulate gene expression and epigenetic processes. To fully capitalize on the potential of EVs as drug carriers, it is important to study and understand the intricacies of EV function and EV RNA-based communication. Here we developed a genetically encodable RNA-based biomaterial, termed EXO-Probe, for tracking EV RNAs. The EXO-Probe comprises an EV-loading RNA sequence (EXO-Code), fused to a fluorogenic RNA Mango aptamer for RNA imaging. This fusion construct allowed the visualization and tracking of EV RNA and colocalization with markers of multivesicular bodies; imaging RNA within EVs, and non-destructive quantification of EVs. Overall, the new RNA-based biomaterial provides a useful and versatile means to interrogate the role of EVs in cellular communication via RNA trafficking to EVs and to study cellular sorting decisions. The system will also help lay the foundation to further improve the therapeutic efficacy of EVs as drug carriers.

细胞外囊泡(EVs)或外泌体(exosomes)在生理和病理细胞通讯中发挥着重要作用,并作为生物药物载体得到了广泛关注。EVs含有调节基因表达和表观遗传过程的长短非编码RNA。要充分利用 EVs 作为药物载体的潜力,就必须研究和了解 EV 功能和 EV RNA 通信的复杂性。在这里,我们开发了一种基于 RNA 的基因编码生物材料,称为 EXO-探针,用于追踪 EV RNA。EXO-Probe由EV载体RNA序列(EXO-Code)和用于RNA成像的荧光RNA Mango适配体融合而成。这种融合构建物允许可视化和跟踪 EV RNA,并与多囊体标记物共定位;对 EV 内的 RNA 进行成像,并对 EV 进行非破坏性量化。总之,这种基于 RNA 的新型生物材料提供了一种有用的多功能手段,可通过 RNA 向 EVs 的迁移来探究 EVs 在细胞通讯中的作用,并研究细胞的分拣决策。该系统还将有助于为进一步提高 EVs 作为药物载体的治疗效果奠定基础。
{"title":"Fluorogenic RNA-based biomaterials for imaging and tracking the cargo of extracellular vesicles","authors":"","doi":"10.1016/j.jconrel.2024.07.043","DOIUrl":"10.1016/j.jconrel.2024.07.043","url":null,"abstract":"<div><p>Extracellular vesicles (EVs), or exosomes, play important roles in physiological and pathological cellular communication and have gained substantial traction as biological drug carriers. EVs contain both short and long non-coding RNAs that regulate gene expression and epigenetic processes. To fully capitalize on the potential of EVs as drug carriers, it is important to study and understand the intricacies of EV function and EV RNA-based communication. Here we developed a genetically encodable RNA-based biomaterial, termed EXO-Probe, for tracking EV RNAs. The EXO-Probe comprises an EV-loading RNA sequence (EXO-Code), fused to a fluorogenic RNA Mango aptamer for RNA imaging. This fusion construct allowed the visualization and tracking of EV RNA and colocalization with markers of multivesicular bodies; imaging RNA within EVs, and non-destructive quantification of EVs. Overall, the new RNA-based biomaterial provides a useful and versatile means to interrogate the role of EVs in cellular communication via RNA trafficking to EVs and to study cellular sorting decisions. The system will also help lay the foundation to further improve the therapeutic efficacy of EVs as drug carriers.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of helper lipids in optimising nanoparticle formulations of self-amplifying RNA 辅助脂质在优化自扩增 RNA 纳米粒子配方中的作用。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.jconrel.2024.08.016

Lipid nanoparticle (LNP) formulation plays a vital role in RNA vaccine delivery. However, further optimisation of self-amplifying RNA (saRNA) vaccine formulation could help enhance seroconversion rates in humans and improve storage stability. Altering either the ionisable or helper lipid can alter the characteristics and performance of formulated saRNA through the interplay of the phospholipid's packing parameter and the geometrical shape within the LNP membrane. In this study, we compared the impact of three helper lipids (DSPC, DOPC, or DOPE) used with two different ionisable lipids (MC3 and C12–200) on stability, transfection efficiency and the inflammation and immunogenicity of saRNA. While helper lipid identity altered saRNA expression across four cell lines in vitro, this was not predictive of an ex vivo or in vivo response. The helper lipid used influenced LNP storage where DSPC provided the best stability profile over four weeks at 2–8 °C. Importantly, helper lipid impact on LNP storage stability was the best predictor of expression in human skin explants, where C12–200 in combination with DSPC provided the most durable expression. C12–200 LNPs also improved protein expression (firefly luciferase) and humoral responses to a SARS-CoV-2 spike saRNA vaccine compared to MC3 LNPs, where the effect of helper lipids was less apparent. Nevertheless, the performance of C12–200 in combination with DSPC appears optimal for saRNA when balancing preferred storage stability requirements against in vivo and ex vivo potency. These data suggest that helper lipid influences the stability and functionality of ionisable lipid nanoparticle-formulated saRNA.

脂质纳米粒子(LNP)配方在 RNA 疫苗递送中发挥着重要作用。然而,进一步优化自扩增 RNA(saRNA)疫苗的配方有助于提高人体血清转换率并改善储存稳定性。通过磷脂的堆积参数和 LNP 膜内的几何形状的相互作用,改变可电离脂质或辅助脂质可改变配制 saRNA 的特性和性能。在这项研究中,我们比较了三种辅助脂质(DSPC、DOPC 或 DOPE)与两种不同的可电离脂质(MC3 和 C12-200)一起使用对 saRNA 的稳定性、转染效率以及炎症和免疫原性的影响。虽然辅助脂质的特性会改变四种细胞系的体外 saRNA 表达,但这并不能预测体内外反应。所使用的辅助脂质影响了 LNP 的储存,其中 DSPC 在 2 2-8 °C 下储存四周的稳定性最好。重要的是,辅助脂质对 LNP 储存稳定性的影响是预测在人体皮肤外植体中表达的最佳指标,其中 C12-200 与 DSPC 结合可提供最持久的表达。与辅助脂质作用不明显的 MC3 LNPs 相比,C12-200 LNPs 还能改善蛋白质表达(萤火虫荧光素酶)和对 SARS-CoV-2 穗状 saRNA 疫苗的体液反应。不过,在平衡首选储存稳定性要求与体内外效力的情况下,C12-200 与 DSPC 的组合似乎是 saRNA 的最佳选择。这些数据表明,辅助脂会影响可电离脂质纳米粒子配制的 saRNA 的稳定性和功能性。
{"title":"The role of helper lipids in optimising nanoparticle formulations of self-amplifying RNA","authors":"","doi":"10.1016/j.jconrel.2024.08.016","DOIUrl":"10.1016/j.jconrel.2024.08.016","url":null,"abstract":"<div><p>Lipid nanoparticle (LNP) formulation plays a vital role in RNA vaccine delivery. However, further optimisation of self-amplifying RNA (saRNA) vaccine formulation could help enhance seroconversion rates in humans and improve storage stability. Altering either the ionisable or helper lipid can alter the characteristics and performance of formulated saRNA through the interplay of the phospholipid's packing parameter and the geometrical shape within the LNP membrane. In this study, we compared the impact of three helper lipids (DSPC, DOPC, or DOPE) used with two different ionisable lipids (MC3 and C12–200) on stability, transfection efficiency and the inflammation and immunogenicity of saRNA. While helper lipid identity altered saRNA expression across four cell lines <em>in vitro</em>, this was not predictive of an <em>ex vivo</em> or <em>in vivo</em> response. The helper lipid used influenced LNP storage where DSPC provided the best stability profile over four weeks at 2–8 °C. Importantly, helper lipid impact on LNP storage stability was the best predictor of expression in human skin explants, where C12–200 in combination with DSPC provided the most durable expression. C12–200 LNPs also improved protein expression (firefly luciferase) and humoral responses to a SARS-CoV-2 spike saRNA vaccine compared to MC3 LNPs, where the effect of helper lipids was less apparent. Nevertheless, the performance of C12–200 in combination with DSPC appears optimal for saRNA when balancing preferred storage stability requirements against <em>in vivo</em> and <em>ex vivo</em> potency. These data suggest that helper lipid influences the stability and functionality of ionisable lipid nanoparticle-formulated saRNA.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0168365924005558/pdfft?md5=fcc163df6f7b460d08e58c43a6583e11&pid=1-s2.0-S0168365924005558-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141982392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhanced delivery of CRISPR/Cas9 system based on biomimetic nanoparticles for hepatitis B virus therapy 基于仿生纳米颗粒的 CRISPR/Cas9 系统在乙型肝炎病毒治疗中的强化传递。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.jconrel.2024.08.019

The persistent presence of covalently closed circular DNA (cccDNA) in hepatocyte nuclei poses a significant obstacle to achieving a comprehensive cure for hepatitis B virus (HBV). Current applications of CRISPR/Cas9 for targeting and eliminating cccDNA have been confined to in vitro studies due to challenges in stable cccDNA expression in animal models and the limited non-immunogenicity of delivery systems. This study addresses these limitations by introducing a novel non-viral gene delivery system utilizing Gemini Surfactant (GS). The developed system creates stable and targeted CRISPR/Cas9 nanodrugs with a negatively charged surface through modification with red blood cell membranes (RBCM) or hepatocyte membranes (HCM), resulting in GS-pDNA@Cas9-CMs complexes. These GS-pDNA complexes demonstrated complete formation at a 4:1 w/w ratio. The in vitro transfection efficiency of GS-pDNA-HCM reached 54.61%, showing homotypic targeting and excellent safety. Additionally, the study identified the most effective single-guide RNA (sgRNA) from six sequences delivered by GS-pDNA@Cas9-HCM. Using GS-pDNA@Cas9-HCM, a significant reduction of 96.47% in in vitro HBV cccDNA and a 52.34% reduction in in vivo HBV cccDNA were observed, along with a notable decrease in other HBV-related markers. The investigation of GS complex uptake by AML-12 cells under varied time and temperature conditions revealed clathrin-mediated endocytosis (CME) for GS-pDNA and caveolin-mediated endocytosis (CVME) for GS-pDNA-HCM and GS-pDNA-RBCM. In summary, this research presents biomimetic gene-editing nanovectors based on GS (GS-pDNA@Cas9-CMs) and explores their precise and targeted clearance of cccDNA using CRISPR/Cas9, demonstrating good biocompatibility both in vitro and in vivo. This innovative approach provides a promising therapeutic strategy for advancing the cure of HBV.

肝细胞核中持续存在的共价闭合环状 DNA(cccDNA)是实现全面治愈乙型肝炎病毒(HBV)的一大障碍。由于在动物模型中稳定表达cccDNA所面临的挑战以及递送系统有限的非免疫原性,目前CRISPR/Cas9在靶向和消除cccDNA方面的应用仅限于体外研究。本研究利用双子表面活性剂(GS)引入了一种新型非病毒基因递送系统,从而解决了这些局限性。所开发的系统通过与红细胞膜(RBCM)或肝细胞膜(HCM)进行修饰,生成表面带负电荷的稳定、靶向 CRISPR/Cas9 纳米药物,从而形成 GS-pDNA@Cas9-CMs 复合物。这些 GS-pDNA 复合物以 4:1 的重量比完全形成。GS-pDNA-HCM 的体外转染效率达到 54.61%,显示了同型靶向性和出色的安全性。此外,研究还从 GS-pDNA@Cas9-HCM 提供的六个序列中鉴定出了最有效的单导 RNA(sgRNA)。使用 GS-pDNA@Cas9-HCM 后,体外 HBV cccDNA 显著减少 96.47%,体内 HBV cccDNA 减少 52.34%,其他 HBV 相关标记物也明显减少。对 AML-12 细胞在不同时间和温度条件下摄取 GS 复合物的调查显示,GS-pDNA 由凝集素介导内吞(CME),GS-pDNA-HCM 和 GS-pDNA-RBCM 由洞穴素介导内吞(CVME)。总之,这项研究提出了基于 GS 的仿生基因编辑纳米载体(GS-pDNA@Cas9-CMs),并利用 CRISPR/Cas9 探索了它们对cccDNA 的精确和靶向清除,在体外和体内均显示出良好的生物相容性。这种创新方法为推进 HBV 治疗提供了一种前景广阔的治疗策略。
{"title":"Enhanced delivery of CRISPR/Cas9 system based on biomimetic nanoparticles for hepatitis B virus therapy","authors":"","doi":"10.1016/j.jconrel.2024.08.019","DOIUrl":"10.1016/j.jconrel.2024.08.019","url":null,"abstract":"<div><p>The persistent presence of covalently closed circular DNA (cccDNA) in hepatocyte nuclei poses a significant obstacle to achieving a comprehensive cure for hepatitis B virus (HBV). Current applications of CRISPR/Cas9 for targeting and eliminating cccDNA have been confined to in vitro studies due to challenges in stable cccDNA expression in animal models and the limited non-immunogenicity of delivery systems. This study addresses these limitations by introducing a novel non-viral gene delivery system utilizing Gemini Surfactant (GS). The developed system creates stable and targeted CRISPR/Cas9 nanodrugs with a negatively charged surface through modification with red blood cell membranes (RBCM) or hepatocyte membranes (HCM), resulting in GS-pDNA@Cas9-CMs complexes. These GS-pDNA complexes demonstrated complete formation at a 4:1 <em>w</em>/<em>w</em> ratio. The in vitro transfection efficiency of GS-pDNA-HCM reached 54.61%, showing homotypic targeting and excellent safety. Additionally, the study identified the most effective single-guide RNA (sgRNA) from six sequences delivered by GS-pDNA@Cas9-HCM. Using GS-pDNA@Cas9-HCM, a significant reduction of 96.47% in in vitro HBV cccDNA and a 52.34% reduction in in vivo HBV cccDNA were observed, along with a notable decrease in other HBV-related markers. The investigation of GS complex uptake by AML-12 cells under varied time and temperature conditions revealed clathrin-mediated endocytosis (CME) for GS-pDNA and caveolin-mediated endocytosis (CVME) for GS-pDNA-HCM and GS-pDNA-RBCM. In summary, this research presents biomimetic gene-editing nanovectors based on GS (GS-pDNA@Cas9-CMs) and explores their precise and targeted clearance of cccDNA using CRISPR/Cas9, demonstrating good biocompatibility both in vitro and in vivo. This innovative approach provides a promising therapeutic strategy for advancing the cure of HBV.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995781","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Spatiotemporal coordination of antigen presentation and co-stimulatory signal for enhanced anti-tumor vaccination 时空协调抗原递呈和协同刺激信号,增强抗肿瘤疫苗接种。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.jconrel.2024.08.025

Controlled-release systems enhance anti-tumor effects by leveraging local antigen persistence for antigen-presenting cells (APCs) recruitment and T cell engagement. However, constant antigen presentation alone tends to induce dysfunction in tumor-specific CD8+ T cells, neglecting the synergistic effects of co-stimulatory signal. To address this, we developed a soft particle-stabilized emulsion (SPE) to deliver lipopeptides with controlled release profiles by adjusting their hydrophobic chain lengths: C6-SPE (fast release), C10-SPE (medium release), and C16-SPE (slow release). Following administration, C6-SPE release antigen rapidly, inducing early antigen presentation, whereas C16-SPE's slow-release delays antigen presentation. Both scenarios missed the critical window for coordinating with the expression of CD86, leading to either T cell apoptosis or suboptimal activation. In contrast, C10-SPE achieved a spatiotemporally synergetic effect of the MHC-I-peptide complex and co-stimulatory signal (CD86), leading to effective dendritic cell (DC) activation, enhanced T cell activation, and tumor regression in EG7-OVA bearing mice. Additionally, co-delivery of cytosine-phosphate-guanine (CpG) with SPE provided a sustained expression of the CD86 window for DC activation, improving the immune response and producing robust anti-tumor effects with C6-SPE comparable to C10-SPE. These findings highlight that synchronizing the spatiotemporal dynamics of antigen presentation and APC activation may confer an optimal strategy for enhanced vaccinations.

控释系统利用局部抗原的持久性促进抗原递呈细胞(APCs)的招募和T细胞的参与,从而增强抗肿瘤效果。然而,单纯的持续抗原递呈往往会诱发肿瘤特异性 CD8+ T 细胞的功能障碍,而忽略了协同刺激信号的协同效应。为了解决这个问题,我们开发了一种软颗粒稳定乳液(SPE),通过调整疏水链长度来递送具有可控释放特性的脂肽:C6-SPE(快速释放)、C10-SPE(中等释放)和C16-SPE(缓慢释放)。给药后,C6-SPE 会快速释放抗原,诱导早期抗原呈递,而 C16-SPE 的缓释则会延迟抗原呈递。这两种情况都错过了与 CD86 表达协调的关键窗口期,导致 T 细胞凋亡或激活效果不佳。相比之下,C10-SPE 实现了 MHC-I 肽复合物和共刺激信号(CD86)的时空协同效应,从而有效激活了树突状细胞(DC),增强了 T 细胞的激活,并使携带 EG7-OVA 的小鼠肿瘤消退。此外,与 SPE 共同递送胞嘧啶-磷酸鸟嘌呤(CpG)为 DC 激活提供了一个持续表达 CD86 的窗口,改善了免疫反应,C6-SPE 产生的抗肿瘤效果与 C10-SPE 不相上下。这些发现突出表明,使抗原呈递和APC活化的时空动态同步可能是增强疫苗接种的最佳策略。
{"title":"Spatiotemporal coordination of antigen presentation and co-stimulatory signal for enhanced anti-tumor vaccination","authors":"","doi":"10.1016/j.jconrel.2024.08.025","DOIUrl":"10.1016/j.jconrel.2024.08.025","url":null,"abstract":"<div><p>Controlled-release systems enhance anti-tumor effects by leveraging local antigen persistence for antigen-presenting cells (APCs) recruitment and T cell engagement. However, constant antigen presentation alone tends to induce dysfunction in tumor-specific CD8<sup>+</sup> T cells, neglecting the synergistic effects of co-stimulatory signal. To address this, we developed a soft particle-stabilized emulsion (SPE) to deliver lipopeptides with controlled release profiles by adjusting their hydrophobic chain lengths: C<sub>6</sub>-SPE (fast release), C<sub>10</sub>-SPE (medium release), and C<sub>16</sub>-SPE (slow release). Following administration, C<sub>6</sub>-SPE release antigen rapidly, inducing early antigen presentation, whereas C<sub>16</sub>-SPE's slow-release delays antigen presentation. Both scenarios missed the critical window for coordinating with the expression of CD86, leading to either T cell apoptosis or suboptimal activation. In contrast, C<sub>10</sub>-SPE achieved a spatiotemporally synergetic effect of the MHC-I-peptide complex and co-stimulatory signal (CD86), leading to effective dendritic cell (DC) activation, enhanced T cell activation, and tumor regression in EG7-OVA bearing mice. Additionally, co-delivery of cytosine-phosphate-guanine (CpG) with SPE provided a sustained expression of the CD86 window for DC activation, improving the immune response and producing robust anti-tumor effects with C<sub>6</sub>-SPE comparable to C<sub>10</sub>-SPE. These findings highlight that synchronizing the spatiotemporal dynamics of antigen presentation and APC activation may confer an optimal strategy for enhanced vaccinations.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995793","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An in situ bioadhesive foam as a large intestinal delivery platform for antibody fragment to treat inflammatory bowel disease 将原位生物黏性泡沫作为治疗炎症性肠病的抗体片段的大肠给药平台。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.jconrel.2024.08.023

Biologics have been widely used as injectables in the treatment of inflammatory bowel disease (IBD). Different local treatment attempts have been developed in recent years. However, maintaining systemic levels of biologics is still crucial for achieving colitis remission. An equilibrium between systemic and local concentrations of biologics is therefore essential for treatment of colitis. Current formulations struggle to create optimal balance between drug concentrations in plasma and the colonic wall. Addressing this challenge, we developed a rectally delivered in situ foam that generates CO2 via a reaction between potassium bicarbonate (PB) and citric acid (CA) without the aid of an external device. An anti-TNF-α antibody fragment (Fab) was loaded into the foam formulation, which promoted prolonged colon retention and improved Fab distribution up to proximal colon following rectal administration to mice. In addition, we observed increased plasma Fab concentrations in mice receiving the rectal Fab foam compared to a Fab solution. In a non-everted rat gut ex vivo model, a single exposure to the CO2-containing foam improved macromolecule transepithelial flux across colonic tissue by over ten-fold. Foam efficacy for Fab was investigated in a range of colitis mouse models, from acute to chronic. This non-invasive formulation platform demonstrates potential to overcome existing limitations in delivering biologics to inflamed colonic tissue.

生物制剂作为注射剂已被广泛用于治疗炎症性肠病(IBD)。近年来,已开发出不同的局部治疗尝试。然而,维持生物制剂的全身水平对于实现结肠炎缓解仍然至关重要。因此,平衡生物制剂在全身和局部的浓度对于治疗结肠炎至关重要。目前的配方很难在血浆和结肠壁的药物浓度之间达到最佳平衡。为了应对这一挑战,我们开发了一种直肠给药原位泡沫,它可以通过碳酸氢钾(PB)和柠檬酸(CA)之间的反应生成二氧化碳,而无需借助外部设备。在泡沫配方中加入抗肿瘤坏死因子-α抗体片段(Fab),可促进小鼠直肠给药后延长结肠保留时间并改善 Fab 在结肠近端的分布。此外,与 Fab 溶液相比,我们观察到直肠给药泡沫小鼠的血浆 Fab 浓度有所提高。在非退行性大鼠肠道体外模型中,单次接触含二氧化碳的泡沫可使大分子通过结肠组织的上皮通量提高 10 倍以上。在一系列结肠炎小鼠模型(从急性到慢性)中对泡沫法布的功效进行了研究。这种非侵入性制剂平台展示了克服将生物制剂输送到发炎结肠组织的现有限制的潜力。
{"title":"An in situ bioadhesive foam as a large intestinal delivery platform for antibody fragment to treat inflammatory bowel disease","authors":"","doi":"10.1016/j.jconrel.2024.08.023","DOIUrl":"10.1016/j.jconrel.2024.08.023","url":null,"abstract":"<div><p>Biologics have been widely used as injectables in the treatment of inflammatory bowel disease (IBD). Different local treatment attempts have been developed in recent years. However, maintaining systemic levels of biologics is still crucial for achieving colitis remission. An equilibrium between systemic and local concentrations of biologics is therefore essential for treatment of colitis. Current formulations struggle to create optimal balance between drug concentrations in plasma and the colonic wall. Addressing this challenge, we developed a rectally delivered <em>in situ</em> foam that generates CO<sub>2</sub> <em>via</em> a reaction between potassium bicarbonate (PB) and citric acid (CA) without the aid of an external device. An anti-TNF-α antibody fragment (Fab) was loaded into the foam formulation, which promoted prolonged colon retention and improved Fab distribution up to proximal colon following rectal administration to mice. In addition, we observed increased plasma Fab concentrations in mice receiving the rectal Fab foam compared to a Fab solution. In a non-everted rat gut <em>ex vivo</em> model, a single exposure to the CO<sub>2</sub>-containing foam improved macromolecule transepithelial flux across colonic tissue by over ten-fold. Foam efficacy for Fab was investigated in a range of colitis mouse models, from acute to chronic. This non-invasive formulation platform demonstrates potential to overcome existing limitations in delivering biologics to inflamed colonic tissue.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0168365924005625/pdfft?md5=dc60066f7f941959bbc1d801a0e2e3ca&pid=1-s2.0-S0168365924005625-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies. 增强 RNA 脂质纳米粒子的传递:器官和细胞特异性及条形码策略
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.jconrel.2024.08.030
Pu-Sheng Wei, Nagasri Thota, Gresham John, Evelyn Chang, Sunjae Lee, Yuanjun Wang, Zitao Ma, Yu-Husan Tsai, Kuo-Ching Mei

Recent advancements in RNA therapeutics highlight the critical need for precision gene delivery systems that target specific organs and cells. Lipid nanoparticles (LNPs) have emerged as key vectors in delivering mRNA and siRNA, offering protection against enzymatic degradation, enabling targeted delivery and cellular uptake, and facilitating RNA cargo release into the cytosol. This review discusses the development and optimization of organ- and cell-specific LNPs, focusing on their design, mechanisms of action, and therapeutic applications. We explore innovations such as DNA/RNA barcoding, which facilitates high-throughput screening and precise adjustments in formulations. We address major challenges, including improving endosomal escape, minimizing off-target effects, and enhancing delivery efficiencies. Notable clinical trials and recent FDA approvals illustrate the practical applications and future potential of LNP-based RNA therapies. Our findings suggest that while considerable progress has been made, continued research is essential to resolve existing limitations and bridge the gap between pre-clinical and clinical evaluation of the safety and efficacy of RNA therapeutics. This review highlights the dynamic progress in LNP research. It outlines a roadmap for future advancements in RNA-based precision medicine.

RNA 疗法的最新进展凸显了对针对特定器官和细胞的精准基因递送系统的迫切需要。脂质纳米颗粒(LNPs)已成为递送 mRNA 和 siRNA 的关键载体,可防止酶降解,实现靶向递送和细胞摄取,并促进 RNA 货物释放到细胞质中。本综述讨论了器官和细胞特异性 LNPs 的开发和优化,重点关注其设计、作用机制和治疗应用。我们探讨了 DNA/RNA 条形码等创新技术,这些技术有助于高通量筛选和精确调整配方。我们解决了一些主要难题,包括改善内体逃逸、最大限度地减少脱靶效应以及提高递送效率。著名的临床试验和最近获得的 FDA 批准说明了基于 LNP 的 RNA 疗法的实际应用和未来潜力。我们的研究结果表明,虽然已经取得了相当大的进展,但要解决现有的局限性并弥合 RNA 疗法的安全性和有效性的临床前评估与临床评估之间的差距,继续研究是必不可少的。本综述强调了 LNP 研究的动态进展。它为基于 RNA 的精准医学的未来发展勾画了路线图。
{"title":"Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies.","authors":"Pu-Sheng Wei, Nagasri Thota, Gresham John, Evelyn Chang, Sunjae Lee, Yuanjun Wang, Zitao Ma, Yu-Husan Tsai, Kuo-Ching Mei","doi":"10.1016/j.jconrel.2024.08.030","DOIUrl":"https://doi.org/10.1016/j.jconrel.2024.08.030","url":null,"abstract":"<p><p>Recent advancements in RNA therapeutics highlight the critical need for precision gene delivery systems that target specific organs and cells. Lipid nanoparticles (LNPs) have emerged as key vectors in delivering mRNA and siRNA, offering protection against enzymatic degradation, enabling targeted delivery and cellular uptake, and facilitating RNA cargo release into the cytosol. This review discusses the development and optimization of organ- and cell-specific LNPs, focusing on their design, mechanisms of action, and therapeutic applications. We explore innovations such as DNA/RNA barcoding, which facilitates high-throughput screening and precise adjustments in formulations. We address major challenges, including improving endosomal escape, minimizing off-target effects, and enhancing delivery efficiencies. Notable clinical trials and recent FDA approvals illustrate the practical applications and future potential of LNP-based RNA therapies. Our findings suggest that while considerable progress has been made, continued research is essential to resolve existing limitations and bridge the gap between pre-clinical and clinical evaluation of the safety and efficacy of RNA therapeutics. This review highlights the dynamic progress in LNP research. It outlines a roadmap for future advancements in RNA-based precision medicine.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142046685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lipiodol emulsion as a dual chemoradiation-sensitizer for pancreatic cancer treatment 脂碘乳剂作为胰腺癌治疗的双重化疗增敏剂。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-20 DOI: 10.1016/j.jconrel.2024.08.020

Pancreatic ductal adenocarcinoma (PDAC) has a low survival rate and limited treatment options. Concurrent chemoradiotherapy is considered beneficial to improve tumor control, but the low drug bioavailability at tumor site and the low radiation tolerance of surrounding healthy organs greatly limits its effectiveness. Lipiodol, a natural drug carrier used in clinical transarterial chemoembolization, has shown potential as a radiosensitizer due to its high Z element iodine composition. Thus, this study aims to repurpose lipiodol as a sensitizer to simultaneously enhance chemo- and radiotherapy for PDAC. To this end, a stable lipiodol emulsion (IOE) loaded with gemcitabine is designed using clinically approved surfactants. At in vivo level, IOE demonstrates better radiotherapeutic effect than existing nanoradiosensitizers and enhanced drug bioavailability over free drug, leading to significant tumor inhibition and improved survival rates under concurrent chemo-radiotherapy. This may due to the sustained drug release, homogenous spatial distribution, and long-term retention ability of IOE in solid PDAC tumor. Furthermore, to better understand the functioning mechanism of drug-loaded IOE, in vitro study is conducted to reveal the ROS- and DNA damage-related therapeutic pathways. Lastly, a comprehensive toxicity assessment also proves the good biocompatibility and safety of as-prepared IOE. This study offers a clinically feasible sensitizer for simultaneous chemoradiotherapy and holds potential for other types of cancer treatment in clinics.

胰腺导管腺癌(PDAC)生存率低,治疗方案有限。同期化放疗被认为有利于提高肿瘤控制率,但由于肿瘤部位的药物生物利用度低以及周围健康器官的辐射耐受性低,大大限制了其有效性。经动脉化疗栓塞术中使用的天然药物载体 Lipiodol 因其高 Z 元素碘成分而具有放射增敏剂的潜力。因此,本研究旨在重新利用脂碘作为增敏剂,同时加强对 PDAC 的化疗和放疗。为此,我们使用临床认可的表面活性剂设计了一种负载吉西他滨的稳定的脂碘乳剂(IOE)。在体内,IOE 比现有的纳米放射增敏剂具有更好的放射治疗效果,而且比游离药物具有更高的药物生物利用度,从而在同时进行化疗和放疗的情况下显著抑制肿瘤并提高生存率。这可能得益于 IOE 在 PDAC 实体瘤中的持续药物释放、均匀空间分布和长期滞留能力。此外,为了更好地了解药物负载 IOE 的作用机制,还进行了体外研究,以揭示与 ROS 和 DNA 损伤相关的治疗途径。最后,全面的毒性评估也证明了制备的 IOE 具有良好的生物相容性和安全性。这项研究为同步放化疗提供了一种临床上可行的增敏剂,并为临床上其他类型的癌症治疗提供了潜力。
{"title":"Lipiodol emulsion as a dual chemoradiation-sensitizer for pancreatic cancer treatment","authors":"","doi":"10.1016/j.jconrel.2024.08.020","DOIUrl":"10.1016/j.jconrel.2024.08.020","url":null,"abstract":"<div><p>Pancreatic ductal adenocarcinoma (PDAC) has a low survival rate and limited treatment options. Concurrent chemoradiotherapy is considered beneficial to improve tumor control, but the low drug bioavailability at tumor site and the low radiation tolerance of surrounding healthy organs greatly limits its effectiveness. Lipiodol, a natural drug carrier used in clinical transarterial chemoembolization, has shown potential as a radiosensitizer due to its high Z element iodine composition. Thus, this study aims to repurpose lipiodol as a sensitizer to simultaneously enhance chemo- and radiotherapy for PDAC. To this end, a stable lipiodol emulsion (IOE) loaded with gemcitabine is designed using clinically approved surfactants. At in vivo level, IOE demonstrates better radiotherapeutic effect than existing nanoradiosensitizers and enhanced drug bioavailability over free drug, leading to significant tumor inhibition and improved survival rates under concurrent chemo-radiotherapy. This may due to the sustained drug release, homogenous spatial distribution, and long-term retention ability of IOE in solid PDAC tumor. Furthermore, to better understand the functioning mechanism of drug-loaded IOE, in vitro study is conducted to reveal the ROS- and DNA damage-related therapeutic pathways. Lastly, a comprehensive toxicity assessment also proves the good biocompatibility and safety of as-prepared IOE. This study offers a clinically feasible sensitizer for simultaneous chemoradiotherapy and holds potential for other types of cancer treatment in clinics.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chimeric peptide-engineered immunostimulant for endoplasmic reticulum targeted photodynamic immunotherapy against metastatic tumor 用于内质网靶向光动力免疫治疗转移性肿瘤的嵌合肽工程免疫刺激剂。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-20 DOI: 10.1016/j.jconrel.2024.08.013

The combination of therapy-induced immunogenic cell death (ICD) and immune checkpoint blockade can provide a mutually reinforced strategy to reverse the poor immunogenicity and immune escape behavior of tumors. In this work, a chimeric peptide-engineered immunostimulant (ER-PPB) is fabricated for endoplasmic reticulum (ER)-targeted photodynamic immunotherapy against metastatic tumors. Among which, the amphiphilic chimeric peptide (ER-PP) is composed of ER-targeting peptide FFKDEL, hydrophilic PEG8 linker and photosensitizer protoporphyrin IX (PpIX), which could be assembled with a PD-1/PD-L1 blocker (BMS-1) to prepare ER-PPB. After passively targeting at tumor tissues, ER-PPB will selectively accumulate in the ER. Next, the localized PDT of ER-PPB will produce a lot of ROS to destroy the primary tumor cells, while increasing the ER stress to initiate a robust ICD cascade. Moreover, the concomitant delivery of BMS-1 can impede the immune escape of tumor cells through PD-1/PD-L1 blockade, thus synergistically activating the immune system to combat metastatic tumors. In vitro and in vivo results demonstrate the robust immune activation and metastatic tumor inhibition characteristics of ER-PPB, which may offer a promising strategy for spatiotemporally controlled metastatic tumor therapy.

治疗诱导的免疫原性细胞死亡(ICD)与免疫检查点阻断相结合,可以提供一种相互促进的策略,扭转肿瘤的免疫原性差和免疫逃逸行为。本研究制备了一种嵌合肽工程免疫刺激剂(ER-PPB),用于内质网(ER)靶向光动力免疫治疗转移性肿瘤。其中,两亲嵌合肽(ER-PP)由ER靶向肽FFKDEL、亲水性PEG8连接体和光敏剂原卟啉IX(PpIX)组成,可与PD-1/PD-L1阻断剂(BMS-1)组装制备ER-PPB。ER-PPB被动靶向肿瘤组织后,会选择性地聚集在ER中。接下来,ER-PPB 的局部光动力疗法会产生大量的 ROS 来破坏原发肿瘤细胞,同时增加 ER 应激,启动强大的 ICD 级联反应。此外,同时给药的 BMS-1 还能通过 PD-1/PD-L1 阻断作用阻碍肿瘤细胞的免疫逃逸,从而协同激活免疫系统来对抗转移性肿瘤。体外和体内研究结果表明,ER-PPB 具有强大的免疫激活和转移性肿瘤抑制特性,可为时空控制的转移性肿瘤治疗提供一种前景广阔的策略。
{"title":"Chimeric peptide-engineered immunostimulant for endoplasmic reticulum targeted photodynamic immunotherapy against metastatic tumor","authors":"","doi":"10.1016/j.jconrel.2024.08.013","DOIUrl":"10.1016/j.jconrel.2024.08.013","url":null,"abstract":"<div><p>The combination of therapy-induced immunogenic cell death (ICD) and immune checkpoint blockade can provide a mutually reinforced strategy to reverse the poor immunogenicity and immune escape behavior of tumors. In this work, a chimeric peptide-engineered immunostimulant (ER-PPB) is fabricated for endoplasmic reticulum (ER)-targeted photodynamic immunotherapy against metastatic tumors. Among which, the amphiphilic chimeric peptide (ER-PP) is composed of ER-targeting peptide FFKDEL, hydrophilic PEG<sub>8</sub> linker and photosensitizer protoporphyrin IX (PpIX), which could be assembled with a PD-1/PD-L1 blocker (BMS-1) to prepare ER-PPB. After passively targeting at tumor tissues, ER-PPB will selectively accumulate in the ER. Next, the localized PDT of ER-PPB will produce a lot of ROS to destroy the primary tumor cells, while increasing the ER stress to initiate a robust ICD cascade. Moreover, the concomitant delivery of BMS-1 can impede the immune escape of tumor cells through PD-1/PD-L1 blockade, thus synergistically activating the immune system to combat metastatic tumors. <em>In vitro</em> and <em>in vivo</em> results demonstrate the robust immune activation and metastatic tumor inhibition characteristics of ER-PPB, which may offer a promising strategy for spatiotemporally controlled metastatic tumor therapy.</p></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":null,"pages":null},"PeriodicalIF":10.5,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Controlled Release
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1