首页 > 最新文献

Journal of Controlled Release最新文献

英文 中文
High-throughput, AI-assisted design and optimization of lipid nanoparticles for drug delivery 高通量,人工智能辅助设计和优化脂质纳米颗粒给药
IF 10.8 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114573
Junjie Zeng, Runlin Chen, Shun He, Chong Zhang, Wen-Che Liu, Hao Song, Shuai Liu, Kewang Nan
Lipid nanoparticles (LNPs) have demonstrated great potential in drug delivery. To fully unlock the therapeutic effect for various diseases, specific design and optimization of the LNP are necessary, which entails an efficient workflow to navigate the LNP design space and tailor the critical attributes. The convergence of high-throughput technology and artificial intelligence (AI) may provide a transformative paradigm to address this challenge. While high-throughput technology serves as a crucial strategy to generate the reliable, large-scale datasets, AI can correlate the critical biological attributes with the LNP structures by learning from the experimental data obtained through the high-throughput method, enabling efficient LNP discovery. In this review, we will discuss the establishment of datasets through high-throughput technology for AI training. AI-assisted LNP design and optimization will be then summarized. Finally, we give an outlook and challenge to discuss the applications of AI for future clinical LNP development.
脂质纳米颗粒(LNPs)在药物传递方面显示出巨大的潜力。为了充分发挥各种疾病的治疗效果,有必要对LNP进行特定的设计和优化,这需要一个有效的工作流程来导航LNP设计空间并定制关键属性。高通量技术和人工智能(AI)的融合可能为应对这一挑战提供一种变革性的范例。虽然高通量技术是生成可靠的大规模数据集的关键策略,但人工智能可以通过从高通量方法获得的实验数据中学习,将关键生物学属性与LNP结构关联起来,从而实现高效的LNP发现。在这篇综述中,我们将讨论通过高通量技术建立用于人工智能训练的数据集。然后总结ai辅助LNP的设计和优化。最后,对人工智能在未来临床LNP发展中的应用进行了展望和挑战。
{"title":"High-throughput, AI-assisted design and optimization of lipid nanoparticles for drug delivery","authors":"Junjie Zeng, Runlin Chen, Shun He, Chong Zhang, Wen-Che Liu, Hao Song, Shuai Liu, Kewang Nan","doi":"10.1016/j.jconrel.2025.114573","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.114573","url":null,"abstract":"Lipid nanoparticles (LNPs) have demonstrated great potential in drug delivery. To fully unlock the therapeutic effect for various diseases, specific design and optimization of the LNP are necessary, which entails an efficient workflow to navigate the LNP design space and tailor the critical attributes. The convergence of high-throughput technology and artificial intelligence (AI) may provide a transformative paradigm to address this challenge. While high-throughput technology serves as a crucial strategy to generate the reliable, large-scale datasets, AI can correlate the critical biological attributes with the LNP structures by learning from the experimental data obtained through the high-throughput method, enabling efficient LNP discovery. In this review, we will discuss the establishment of datasets through high-throughput technology for AI training. AI-assisted LNP design and optimization will be then summarized. Finally, we give an outlook and challenge to discuss the applications of AI for future clinical LNP development.","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"29 1","pages":""},"PeriodicalIF":10.8,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813844","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
Nanochirality-programmed type-I photosensitizer enables deep-tumor photodynamic therapy by reducing extracellular-matrix adhesion 纳米手性编程i型光敏剂通过减少细胞外基质粘连实现深部肿瘤光动力治疗
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114572
Yichen Liu, Gaeun Kim, Runyao Zhu, Hyunsu Jeon, Yichun Wang
Tumor hypoxia and poor penetration of therapeutics across tumor-microenvironment barriers remain major obstacles to effective cancer therapy, including photodynamic therapy (PDT). Here we introduce a nanochirality-programmed assembly (L-Chi-GAIN) in which nanochirality drives site-selective assembly that activates Type-I reactive oxygen species (ROS) generation with significantly reduced oxygen dependence and diminishes hyaluronan-mediated matrix adhesion, thereby enabling deep intratumoral therapy. Glycosylation imparts structural chirality to graphene quantum dots (GQDs), directing site-selective assembly of indocyanine green (ICG) that turns on photoinduced electron transfer (PET), producing a 64-fold increase in ROS relative to free ICG. Nanochirality also modulates assembly–extracellular matrix (ECM) interactions. L-GQDs show a less favorable hyaluronan binding free energy (ΔGbind), thereby accelerating interstitial transport and resulting in ∼3-fold greater penetration depth and ∼ 21-fold higher mean intratumoral ICG signal within the penetrating area relative to liposomal carriers. Under near-infrared irradiation, L-Chi-GAIN elicits strong oxidative stress and triggers Gasdermin-D (GSDMD)-dependent pyroptosis, leading to significant suppression of tumor growth. This work offers a nanochirality-guided design strategy for PDT in deep tumors by coupling site-selective assembly with stereoselective navigation of the ECM.
肿瘤缺氧和治疗药物穿透肿瘤微环境屏障的能力差仍然是有效治疗癌症的主要障碍,包括光动力治疗(PDT)。在这里,我们介绍了一种纳米手性编程组装(L-Chi-GAIN),其中纳米手性驱动位点选择性组装,激活i型活性氧(ROS)的产生,显著降低氧依赖性,减少透明质酸介导的基质粘附,从而实现深部肿瘤内治疗。糖基化赋予石墨烯量子点(GQDs)结构手性,指导吲哚菁绿(ICG)的位点选择性组装,从而开启光诱导电子转移(PET),产生相对于自由ICG增加64倍的ROS。纳米手性也调节组装-细胞外基质(ECM)的相互作用。L-GQDs显示出不太有利的透明质酸结合自由能(ΔGbind),从而加速了间质运输,导致穿透深度增加~3倍,穿透区域内肿瘤内ICG信号相对于脂质体携带者增加 ~ 21倍。在近红外照射下,L-Chi-GAIN引发强烈的氧化应激,触发Gasdermin-D (GSDMD)依赖性焦亡,从而显著抑制肿瘤生长。这项工作提供了一种纳米手性引导的深部肿瘤PDT设计策略,通过耦合位点选择性组装和ECM的立体选择性导航。
{"title":"Nanochirality-programmed type-I photosensitizer enables deep-tumor photodynamic therapy by reducing extracellular-matrix adhesion","authors":"Yichen Liu,&nbsp;Gaeun Kim,&nbsp;Runyao Zhu,&nbsp;Hyunsu Jeon,&nbsp;Yichun Wang","doi":"10.1016/j.jconrel.2025.114572","DOIUrl":"10.1016/j.jconrel.2025.114572","url":null,"abstract":"<div><div>Tumor hypoxia and poor penetration of therapeutics across tumor-microenvironment barriers remain major obstacles to effective cancer therapy, including photodynamic therapy (PDT). Here we introduce a nanochirality-programmed assembly (<em>L</em>-Chi-GAIN) in which nanochirality drives site-selective assembly that activates Type-I reactive oxygen species (ROS) generation with significantly reduced oxygen dependence and diminishes hyaluronan-mediated matrix adhesion, thereby enabling deep intratumoral therapy. Glycosylation imparts structural chirality to graphene quantum dots (GQDs), directing site-selective assembly of indocyanine green (ICG) that turns on photoinduced electron transfer (PET), producing a 64-fold increase in ROS relative to free ICG. Nanochirality also modulates assembly–extracellular matrix (ECM) interactions. <em>L</em>-GQDs show a less favorable hyaluronan binding free energy (ΔG<sub>bind</sub>), thereby accelerating interstitial transport and resulting in ∼3-fold greater penetration depth and ∼ 21-fold higher mean intratumoral ICG signal within the penetrating area relative to liposomal carriers. Under near-infrared irradiation, <em>L</em>-Chi-GAIN elicits strong oxidative stress and triggers Gasdermin-D (GSDMD)-dependent pyroptosis, leading to significant suppression of tumor growth. This work offers a nanochirality-guided design strategy for PDT in deep tumors by coupling site-selective assembly with stereoselective navigation of the ECM.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114572"},"PeriodicalIF":11.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145812893","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
Hepatocyte-specific partial cellular reprogramming via selective OSK mRNA lipid nanoparticle attenuates liver fibrosis 肝细胞特异性部分细胞重编程通过选择性OSK mRNA脂质纳米颗粒减轻肝纤维化
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114569
Chenchen Zhang , Yuerong Bai , Qimeng Yin , Jiahao Li , Kun Huang , Min Qiu
Liver fibrosis is a major global health burden with no approved therapies. Transient expression of reprogramming factors Oct4, Sox2, and Klf4 (OSK) promotes tissue regeneration without inducing full pluripotency, which represents an attractive regenerative therapy. Here, we introduce a hepatocyte-specific mRNA delivery strategy for in vivo partial cellular reprogramming using a chemically defined lipid nanoparticle (LNP) platform. We synthesized a series of natural unsaturated fatty alcohol-based ionizable lipids and identified a lead compound, H4T3, with mRNA delivery efficacy comparable to SM102. Further formulation optimization led to a simplified, phospholipid-free, three-component LNP formulation, H4T3_F6 that exhibits high potency and enhanced hepatocyte selectivity, alongside minimal immunogenicity and an overall favorable safety profile. Hepatocyte-specific delivery of OSK mRNA via H4T3_F6 LNPs transiently reprogrammed fibrotic hepatocytes into progenitor-like cells, rejuvenated hepatic gene expression, and promoted functional regeneration. This rejuvenation process downregulates fibrogenic mediators (Tgfb1, Pdgfb), disrupting hepatocyte-stellate cell signaling and halting extracellular matrix (ECM) deposition. The integrated reprogramming and paracrine modulation collectively shift the liver microenvironment from a fibrotic to a regenerative state in a CCl4-induced liver fibrosis mouse model. This study provides proof-of-concept for rejuvenating fibrotic livers via selective mRNA-based in vivo cellular reprogramming, offering a promising strategy for liver fibrosis or age-related tissue repair.
肝纤维化是全球主要的健康负担,目前尚无批准的治疗方法。瞬时表达重编程因子Oct4、Sox2和Klf4 (OSK)可促进组织再生,但不诱导完全多能性,这是一种有吸引力的再生疗法。在这里,我们介绍了一种肝细胞特异性mRNA递送策略,使用化学定义的脂质纳米颗粒(LNP)平台进行体内部分细胞重编程。我们合成了一系列天然不饱和脂肪醇基可电离脂质,并鉴定了一种先导化合物H4T3,其mRNA递送效果与SM102相当。进一步的配方优化得到了一种简化的、无磷脂的三组分LNP配方H4T3_F6,它具有高效能和增强的肝细胞选择性,同时具有最小的免疫原性和总体良好的安全性。通过H4T3_F6 LNPs向肝细胞特异性递送OSK mRNA可瞬间将纤维化肝细胞重编程为祖细胞样细胞,恢复肝脏基因表达,促进功能再生。这个恢复过程下调了纤维化介质(Tgfb1, Pdgfb),破坏肝细胞-星状细胞信号传导并阻止细胞外基质(ECM)沉积。在ccl4诱导的肝纤维化小鼠模型中,整合的重编程和旁分泌调节共同将肝脏微环境从纤维化状态转移到再生状态。该研究提供了通过基于选择性mrna的体内细胞重编程使纤维化肝脏恢复活力的概念证明,为肝纤维化或与年龄相关的组织修复提供了一种有希望的策略。
{"title":"Hepatocyte-specific partial cellular reprogramming via selective OSK mRNA lipid nanoparticle attenuates liver fibrosis","authors":"Chenchen Zhang ,&nbsp;Yuerong Bai ,&nbsp;Qimeng Yin ,&nbsp;Jiahao Li ,&nbsp;Kun Huang ,&nbsp;Min Qiu","doi":"10.1016/j.jconrel.2025.114569","DOIUrl":"10.1016/j.jconrel.2025.114569","url":null,"abstract":"<div><div>Liver fibrosis is a major global health burden with no approved therapies. Transient expression of reprogramming factors <em>Oct4</em>, <em>Sox2,</em> and <em>Klf4</em> (<em>OSK</em>) promotes tissue regeneration without inducing full pluripotency, which represents an attractive regenerative therapy. Here, we introduce a hepatocyte-specific mRNA delivery strategy for <em>in vivo</em> partial cellular reprogramming using a chemically defined lipid nanoparticle (LNP) platform. We synthesized a series of natural unsaturated fatty alcohol-based ionizable lipids and identified a lead compound, H4T3, with mRNA delivery efficacy comparable to SM102. Further formulation optimization led to a simplified, phospholipid-free, three-component LNP formulation, H4T3_F6 that exhibits high potency and enhanced hepatocyte selectivity, alongside minimal immunogenicity and an overall favorable safety profile. Hepatocyte-specific delivery of <em>OSK</em> mRNA <em>via</em> H4T3_F6 LNPs transiently reprogrammed fibrotic hepatocytes into progenitor-like cells, rejuvenated hepatic gene expression, and promoted functional regeneration. This rejuvenation process downregulates fibrogenic mediators (<em>Tgfb1</em>, <em>Pdgfb</em>), disrupting hepatocyte-stellate cell signaling and halting extracellular matrix (ECM) deposition. The integrated reprogramming and paracrine modulation collectively shift the liver microenvironment from a fibrotic to a regenerative state in a CCl<sub>4</sub>-induced liver fibrosis mouse model. This study provides proof-of-concept for rejuvenating fibrotic livers <em>via</em> selective mRNA-based <em>in vivo</em> cellular reprogramming, offering a promising strategy for liver fibrosis or age-related tissue repair.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114569"},"PeriodicalIF":11.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813845","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
Multifunctional microfluidic-directed polymer/hydrogel fabrics towards pH-responsive drug delivery, wound monitoring and wearable sensing 多功能微流体定向聚合物/水凝胶织物用于ph响应药物递送,伤口监测和可穿戴传感
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114565
Xiang-Yun Du, Yin-Jian Song, Chen Du, Ji-Dong Liu
Delayed skin wound healing caused by bacterial infection and persistent oxidative stress has been considered as a frequently encountered clinical problem that significantly affects patients' health. However, the real-time monitoring and on-demand treatment of non-healing wounds remain a great challenge. Herein, a feasible strategy is developed to core-shell polymer/hydrogel microfibers by integrating microfluidic spinning with shear-flow-induced coating technology. The fabricated self-healing microfibers are facilely assembled into nonwoven fabrics through reversible physical interactions. The core polycaprolactone (PCL) acting as a skeleton endows fabrics with structural and mechanical robustness (4.67 MPa), while the hydrogel shell brings functional diversity including great flexibility, self-healing, self-adhesion and pH-responsive swelling capability. By introducing epigallocatechin gallate (EGCG) in core PCL phase, the remarkable reactive oxygen species (ROS) cleaning capacity (93.8 % for 0.5 h) and antibacterial activity against E. coli (98.7 %) and S. aureus (99.6 %) are achieved. Particularly, the pH-regulated drug release of fabric in a prolonged form is achieved owing to the reversibly swelling/shrinking behavior of hydrogel shell with the changed environmental pH, which is expected to realize on-demand drug delivery and treatment of wound. Additionally, the pH-responsive curcumin-loaded mesoporous microparticles are incorporated into hydrogel shell, aiming to visually real-time detect pH level in a high-efficiency and reversible manner. Moreover, the fabric can serve as a flexible wearable sensor for precisely monitoring motions of patients. This strategy explores a feasible solution to diligently tracking wound pH and movements, along with inhibiting wound infection, substantiating a great foundation for application in personalized, intelligent and precise theranostic wound dressings.
细菌感染和持续氧化应激引起的皮肤伤口延迟愈合一直被认为是一个常见的临床问题,严重影响患者的健康。然而,对未愈合伤口的实时监测和按需治疗仍然是一个巨大的挑战。本文将微流控纺丝技术与剪切流诱导涂层技术相结合,提出了一种可行的核壳聚合物/水凝胶微纤维制备策略。制备的自修复微纤维通过可逆的物理相互作用,可以很容易地组装成非织造布。核心聚己内酯(PCL)作为骨架赋予织物结构和机械稳健性(4.67 MPa),而水凝胶壳则带来了功能多样性,包括巨大的柔韧性、自修复、自粘附和ph响应膨胀能力。通过在核心PCL相中引入表没食子儿茶素没食子酸酯(EGCG),获得了显著的活性氧(ROS)净化能力(93.8 %,0.5 h)和对大肠杆菌(98.7 %)和金黄色葡萄球菌(99.6 %)的抑菌活性。特别是,由于水凝胶壳随着环境pH的变化而发生可逆的膨胀/收缩行为,从而实现pH调节的织物药物长时间释放,有望实现按需给药和伤口治疗。此外,将负载姜黄素的pH响应介孔微粒子整合到水凝胶壳中,旨在以高效、可逆的方式可视化实时检测pH水平。此外,该织物还可以作为灵活的可穿戴传感器,用于精确监测患者的运动。该策略探索了一种切实可行的解决方案,既能跟踪伤口pH值和运动,又能抑制伤口感染,为个性化、智能化、精准化治疗性伤口敷料的应用奠定了坚实的基础。
{"title":"Multifunctional microfluidic-directed polymer/hydrogel fabrics towards pH-responsive drug delivery, wound monitoring and wearable sensing","authors":"Xiang-Yun Du,&nbsp;Yin-Jian Song,&nbsp;Chen Du,&nbsp;Ji-Dong Liu","doi":"10.1016/j.jconrel.2025.114565","DOIUrl":"10.1016/j.jconrel.2025.114565","url":null,"abstract":"<div><div>Delayed skin wound healing caused by bacterial infection and persistent oxidative stress has been considered as a frequently encountered clinical problem that significantly affects patients' health. However, the real-time monitoring and on-demand treatment of non-healing wounds remain a great challenge. Herein, a feasible strategy is developed to core-shell polymer/hydrogel microfibers by integrating microfluidic spinning with shear-flow-induced coating technology. The fabricated self-healing microfibers are facilely assembled into nonwoven fabrics through reversible physical interactions. The core polycaprolactone (PCL) acting as a skeleton endows fabrics with structural and mechanical robustness (4.67 MPa), while the hydrogel shell brings functional diversity including great flexibility, self-healing, self-adhesion and pH-responsive swelling capability. By introducing epigallocatechin gallate (EGCG) in core PCL phase, the remarkable reactive oxygen species (ROS) cleaning capacity (93.8 % for 0.5 h) and antibacterial activity against <em>E. coli</em> (98.7 %) and <em>S. aureus</em> (99.6 %) are achieved. Particularly, the pH-regulated drug release of fabric in a prolonged form is achieved owing to the reversibly swelling/shrinking behavior of hydrogel shell with the changed environmental pH, which is expected to realize on-demand drug delivery and treatment of wound. Additionally, the pH-responsive curcumin-loaded mesoporous microparticles are incorporated into hydrogel shell, aiming to visually real-time detect pH level in a high-efficiency and reversible manner. Moreover, the fabric can serve as a flexible wearable sensor for precisely monitoring motions of patients. This strategy explores a feasible solution to diligently tracking wound pH and movements, along with inhibiting wound infection, substantiating a great foundation for application in personalized, intelligent and precise theranostic wound dressings.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114565"},"PeriodicalIF":11.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145801408","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
Deciphering the in vivo fate and biomacromolecular interactions of DSPE-PEG2000 破译DSPE-PEG2000的体内命运和生物大分子相互作用
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114571
Deqi Cai , Chong Su , Mingyang Zhao , Huan Yang , Zhe Zhang , Yingze Liu , Jingkai Gu
DSPE-PEG2000 (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-poly(ethylene glycol)-2000), a key excipient of stealth nano-formulations like Doxil®, prolongs nano-carrier blood residence by forming steric hydrophilic barriers. However, its assumed biological inertness faces challenges from emerging PEG-related risks. Limited by analytical methods, its in vivo fate and biomacromolecular interactions remain uncharacterized. Here, Liquid Chromatography-Quadrupole/Time-of-Flight Mass Spectrometry (LC-Q-TOF MS) and anti-PEG single-chain variable fragment (PEG-scFv) strategy were innovatively employed to decode DSPE-PEG2000's biological properties post-intravenous administration. Results reveal a short plasma half-life, tissue-specific distribution in lung, liver, and spleen, complement activation without adaptive antibody response, cleavage to DSPE/PEG2000 with renal excretion/degradation of PEG2000, and a plasma protein binding rate (PPB) of 63.2 ± 7.11 % that competitively displaces 50 % of doxorubicin binding. These findings establish its non-inert profile of tissue-specific distribution, innate immune activation, complex metabolism, and plasma protein-based excipient-drug interactions, highlighting DSPE-PEG2000's potential safety risks. The innovative methodology developed herein provides a paradigm for polymer tracking and offers critical insights to guide excipient safety assessments and optimization.
DSPE-PEG2000(1,2-二硬脂酰- n-甘油-3-磷酸乙醇胺- n-聚乙二醇-2000)是隐形纳米制剂如Doxil®的关键赋形剂,通过形成立体亲水性屏障延长纳米载体血液的停留时间。然而,其假定的生物惰性面临着来自peg相关风险的挑战。受分析方法的限制,其在体内的命运和生物大分子相互作用仍未表征。本研究创新性地采用液相色谱-四极杆/飞行时间质谱(LC-Q-TOF MS)和抗peg单链可变片段(PEG-scFv)策略来解码DSPE-PEG2000静脉给药后的生物学特性。结果显示,血浆半衰期短,在肺、肝脏和脾脏有组织特异性分布,补体活化而无适应性抗体反应,对DSPE/PEG2000的裂解与肾排泄/PEG2000的降解,血浆蛋白结合率(PPB)为63.2 ± 7.11 %,竞争性地取代了50% %的阿霉素结合。这些发现建立了其组织特异性分布、先天免疫激活、复杂代谢和基于血浆蛋白的赋形剂-药物相互作用的非惰性特征,突出了DSPE-PEG2000的潜在安全风险。本文开发的创新方法为聚合物跟踪提供了范例,并为指导赋形剂安全性评估和优化提供了关键见解。
{"title":"Deciphering the in vivo fate and biomacromolecular interactions of DSPE-PEG2000","authors":"Deqi Cai ,&nbsp;Chong Su ,&nbsp;Mingyang Zhao ,&nbsp;Huan Yang ,&nbsp;Zhe Zhang ,&nbsp;Yingze Liu ,&nbsp;Jingkai Gu","doi":"10.1016/j.jconrel.2025.114571","DOIUrl":"10.1016/j.jconrel.2025.114571","url":null,"abstract":"<div><div>DSPE-PEG2000 (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-poly(ethylene glycol)-2000), a key excipient of stealth nano-formulations like Doxil®, prolongs nano-carrier blood residence by forming steric hydrophilic barriers. However, its assumed biological inertness faces challenges from emerging PEG-related risks. Limited by analytical methods, its <em>in vivo</em> fate and biomacromolecular interactions remain uncharacterized. Here, Liquid Chromatography-Quadrupole/Time-of-Flight Mass Spectrometry (LC-Q-TOF MS) and anti-PEG single-chain variable fragment (PEG-scFv) strategy were innovatively employed to decode DSPE-PEG2000's biological properties post-intravenous administration. Results reveal a short plasma half-life, tissue-specific distribution in lung, liver, and spleen, complement activation without adaptive antibody response, cleavage to DSPE/PEG2000 with renal excretion/degradation of PEG2000, and a plasma protein binding rate (PPB) of 63.2 ± 7.11 % that competitively displaces 50 % of doxorubicin binding. These findings establish its non-inert profile of tissue-specific distribution, innate immune activation, complex metabolism, and plasma protein-based excipient-drug interactions, highlighting DSPE-PEG2000's potential safety risks. The innovative methodology developed herein provides a paradigm for polymer tracking and offers critical insights to guide excipient safety assessments and optimization.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114571"},"PeriodicalIF":11.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145801382","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
Alanine-serine-cysteine transporter-targeted small-molecule boron carriers for neutron capture therapy of L-4‑boronophenylalanine-refractory tumors 以丙氨酸-丝氨酸-半胱氨酸转运蛋白为靶点的小分子硼载体用于L-4 -硼苯丙氨酸难治肿瘤的中子俘获治疗
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-22 DOI: 10.1016/j.jconrel.2025.114566
Kazuki Miura , Tomoyuki Araki , Taiki Morita , Kai Nishimura , Satoshi Okada , Minoru Suzuki , Hiroyuki Nakamura
Boron neutron capture therapy (BNCT) requires the selective delivery of sufficient amounts of 10B to tumors. However, its clinical efficacy remains limited because the approved 10B agent L-4‑boronophenylalanine (BPA) relies primarily on the L-type amino acid transporter 1 (LAT1), leaving LAT1-low tumors refractory to treatment. Here, we report GluB-2, a water-soluble, alanine-serine-cysteine transporter 2 (ASCT2)-targeted small-molecule 10B carrier that expands BNCT applicability beyond BPA. GluB-2 exhibited high solubility, low cytotoxicity, and preferential ASCT2-mediated uptake in multiple cancer cell lines, particularly those with low LAT1 and high ASCT2 expression. In vivo, GluB-2 achieved tumor boron concentrations exceeding the therapeutic threshold (>20 μg [10B]/g tissue) after both intravenous and intraperitoneal administration, representing the first non-BPA small-molecule 10B carrier to reach this level. Upon thermal neutron irradiation, GluB-2 induced pronounced tumor suppression in both the CT26 allograft and the BPA-refractory U87MG xenograft models without evidence of systemic toxicity. These findings demonstrate that GluB-2 enables therapeutic small-molecule delivery of 10B to tumors through multiple dosing routes and expands the clinical applicability of BNCT beyond BPA, highlighting its translational potential.
硼中子俘获疗法(BNCT)需要选择性地向肿瘤输送足够量的硼。然而,其临床疗效仍然有限,因为批准的10B药物L-4 -硼苯丙氨酸(BPA)主要依赖于l型氨基酸转运蛋白1 (LAT1),导致LAT1低的肿瘤难以治疗。在这里,我们报道了GluB-2,一种水溶性,丙氨酸-丝氨酸-半胱氨酸转运体(ASCT2)靶向小分子10B载体,将BNCT的适用性扩展到BPA之外。GluB-2在多种癌细胞系中表现出高溶解度、低细胞毒性和ASCT2介导的优先摄取,特别是那些低LAT1和高ASCT2表达的癌细胞系。在体内,经静脉和腹腔给药后,GluB-2的肿瘤硼浓度均超过治疗阈值(>;20 μg [10B]/g组织),是第一个达到这一水平的非bpa小分子10B载体。在热中子照射下,GluB2在CT26同种异体移植物和bpa难治性U87MG异种移植物模型中均诱导了明显的肿瘤抑制,无系统性毒性证据。这些发现表明,GluB-2可以通过多种给药途径将10B治疗性小分子递送到肿瘤中,并将BNCT的临床适用性扩展到BPA之外,突出了其转化潜力。
{"title":"Alanine-serine-cysteine transporter-targeted small-molecule boron carriers for neutron capture therapy of L-4‑boronophenylalanine-refractory tumors","authors":"Kazuki Miura ,&nbsp;Tomoyuki Araki ,&nbsp;Taiki Morita ,&nbsp;Kai Nishimura ,&nbsp;Satoshi Okada ,&nbsp;Minoru Suzuki ,&nbsp;Hiroyuki Nakamura","doi":"10.1016/j.jconrel.2025.114566","DOIUrl":"10.1016/j.jconrel.2025.114566","url":null,"abstract":"<div><div>Boron neutron capture therapy (BNCT) requires the selective delivery of sufficient amounts of <sup>10</sup>B to tumors. However, its clinical efficacy remains limited because the approved <sup>10</sup>B agent L-4‑boronophenylalanine (BPA) relies primarily on the L-type amino acid transporter 1 (LAT1), leaving LAT1-low tumors refractory to treatment. Here, we report <strong>GluB-2</strong>, a water-soluble, alanine-serine-cysteine transporter 2 (ASCT2)-targeted small-molecule <sup>10</sup>B carrier that expands BNCT applicability beyond BPA. <strong>GluB-2</strong> exhibited high solubility, low cytotoxicity, and preferential ASCT2-mediated uptake in multiple cancer cell lines, particularly those with low LAT1 and high ASCT2 expression. <em>In vivo</em>, <strong>GluB-2</strong> achieved tumor boron concentrations exceeding the therapeutic threshold (&gt;20 μg [<sup>10</sup>B]/g tissue) after both intravenous and intraperitoneal administration, representing the first non-BPA small-molecule <sup>10</sup>B carrier to reach this level. Upon thermal neutron irradiation, <strong>GluB-2</strong> induced pronounced tumor suppression in both the CT26 allograft and the BPA-refractory U87MG xenograft models without evidence of systemic toxicity. These findings demonstrate that <strong>GluB-2</strong> enables therapeutic small-molecule delivery of <sup>10</sup>B to tumors through multiple dosing routes and expands the clinical applicability of BNCT beyond BPA, highlighting its translational potential.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114566"},"PeriodicalIF":11.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145801493","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
Engineered nanoparticles for subconjunctival delivery to the retinal pigment epithelium: A multi-target therapy for dry AMD 用于结膜下视网膜色素上皮的工程纳米颗粒:干性AMD的多靶点治疗。
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-21 DOI: 10.1016/j.jconrel.2025.114563
Yao Li , Shiman Yuan , Cong Zhang , Pengcheng Hu , Xiaoying Huang , Jialin Zhou , Danning Liu , Xiyuan Zhou
Dry age-related macular degeneration (dAMD) is a leading cause of irreversible blindness, driven by oxidative stress-induced retinal pigment epithelial (RPE) cell degeneration. Existing therapies suffer from poor bioavailability and insufficient multi-pathway modulation. To address this, we developed P(R)/T-Lf nanoparticles, a subconjunctivally administered nanotherapy co-loaded with resveratrol (Res) and dual-functionalized with trimethyl chitosan (TMC) and lactoferrin (Lf). The P(R)/T-Lf NPs exhibited: (1) prolonged ocular retention via TMC-mediated mucoadhesion and enhanced RPE targeting through Lf receptor binding; (2) sustained Res release over 35 days, effectively scavenging reactive oxygen species and inhibiting ferroptosis by downregulating NOX2, ACSL4, and COX2 while restoring GPX4; (3) superior therapeutic outcomes in NaIO₃-induced dAMD models, preserving retinal morphology and function. Comparative studies demonstrated that P(R)/T-Lf NPs outperformed non-targeted controls. This nanoplatform provides a translation-ready strategy to concurrently tackle oxidative stress, inflammation, and ferroptosis via sustained, targeted delivery, representing a transformative approach for dAMD therapy.
干性年龄相关性黄斑变性(dAMD)是不可逆失明的主要原因,由氧化应激诱导的视网膜色素上皮(RPE)细胞变性驱动。现有的治疗方法存在生物利用度差和多途径调节不足的问题。为了解决这个问题,我们开发了P(R)/T-Lf纳米颗粒,这是一种结膜下给药的纳米疗法,共负载白藜芦醇(Res),并与三甲基壳聚糖(TMC)和乳铁蛋白(Lf)双功能化。P(R)/T-Lf NPs表现出:(1)通过tmc介导的黏附延长眼潴留,并通过Lf受体结合增强RPE靶向性;(2) Res持续释放超过35 d,通过下调NOX2、ACSL4和COX2有效清除活性氧,抑制铁下沉,同时恢复GPX4;(3) NaIO₃诱导的dAMD模型具有良好的治疗效果,保留了视网膜的形态和功能。比较研究表明,P(R)/T-Lf NPs优于非靶向对照。该纳米平台提供了一种可翻译的策略,通过持续的靶向递送来同时解决氧化应激、炎症和铁下沉,代表了dAMD治疗的一种变革性方法。
{"title":"Engineered nanoparticles for subconjunctival delivery to the retinal pigment epithelium: A multi-target therapy for dry AMD","authors":"Yao Li ,&nbsp;Shiman Yuan ,&nbsp;Cong Zhang ,&nbsp;Pengcheng Hu ,&nbsp;Xiaoying Huang ,&nbsp;Jialin Zhou ,&nbsp;Danning Liu ,&nbsp;Xiyuan Zhou","doi":"10.1016/j.jconrel.2025.114563","DOIUrl":"10.1016/j.jconrel.2025.114563","url":null,"abstract":"<div><div>Dry age-related macular degeneration (dAMD) is a leading cause of irreversible blindness, driven by oxidative stress-induced retinal pigment epithelial (RPE) cell degeneration. Existing therapies suffer from poor bioavailability and insufficient multi-pathway modulation. To address this, we developed P(R)/T-Lf nanoparticles, a subconjunctivally administered nanotherapy co-loaded with resveratrol (Res) and dual-functionalized with trimethyl chitosan (TMC) and lactoferrin (Lf). The P(R)/T-Lf NPs exhibited: (1) prolonged ocular retention via TMC-mediated mucoadhesion and enhanced RPE targeting through Lf receptor binding; (2) sustained Res release over 35 days, effectively scavenging reactive oxygen species and inhibiting ferroptosis by downregulating NOX2, ACSL4, and COX2 while restoring GPX4; (3) superior therapeutic outcomes in NaIO₃-induced dAMD models, preserving retinal morphology and function. Comparative studies demonstrated that P(R)/T-Lf NPs outperformed non-targeted controls. This nanoplatform provides a translation-ready strategy to concurrently tackle oxidative stress, inflammation, and ferroptosis via sustained, targeted delivery, representing a transformative approach for dAMD therapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114563"},"PeriodicalIF":11.5,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813471","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
Octopus-inspired polymeric nanovaccine enables high antigen loading and robust T cell activation for cancer immunotherapy 章鱼启发的聚合纳米疫苗为癌症免疫治疗提供了高抗原负载和强大的T细胞激活
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-20 DOI: 10.1016/j.jconrel.2025.114562
Zijuan Wang , Yuanzhen Su , Shucheng Zhang , Bingzheng Yu , Dongbo Chen , Xiang Gao , Yan Wei , Irina A. Veselova , Mingqiang Li , Shixian Lv
Tumor vaccines hold significant promise for immunotherapy, but are limited by low antigen loading capacity, inefficient cytosolic delivery, and suboptimal T cell activation. Here, we present an octopus-inspired polymeric nanovaccine that integrates high antigen-loading capacity and effective cytosolic delivery within a single polymeric platform. The nanovaccine is constructed by encapsulating antigens with an imidazole-functionalized fluorinated polyethyleneimine and Mn2+ ions, forming a structure that mimics octopus tentacles and suction cups, where the PEI backbone acts as tentacle-like arms and the imidazole-Mn2+ units serve as suction cups. This multivalent interface enables robust antigen binding through electrostatic, coordination, and hydrophobic interactions. Beyond stabilizing the antigen payload, the amphiphilic cationic design of the polymers offers efficient cytosolic delivery of antigens into dendritic cells (DCs). Meanwhile, the intracellular release of Mn2+ activates the STING pathway, promoting innate immune responses. Consequently, the vaccine elicits robust antigen-specific CD8+ T cell responses and durable antitumor immunity in multiple tumor models. This work presents a streamlined, multifunctional strategy to overcome delivery barriers in cancer vaccines.
肿瘤疫苗在免疫治疗方面具有重要的前景,但由于抗原负荷能力低、细胞质递送效率低和T细胞活化不理想而受到限制。在这里,我们提出了一种受章鱼启发的聚合物纳米疫苗,它在单个聚合物平台内整合了高抗原负载能力和有效的细胞质递送。纳米疫苗是通过用咪唑功能化的氟化聚乙烯亚胺和Mn2+离子包裹抗原,形成模仿章鱼触手和吸盘的结构,其中PEI主链充当触手状臂,咪唑-Mn2+单元充当吸盘。这种多价界面通过静电、配位和疏水相互作用使抗原结合更加牢固。除了稳定抗原负载外,聚合物的两亲性阳离子设计提供了有效的细胞质递送抗原到树突状细胞(dc)。同时,细胞内释放Mn2+激活STING通路,促进先天免疫应答。因此,该疫苗在多种肿瘤模型中引发了强大的抗原特异性CD8+ T细胞反应和持久的抗肿瘤免疫。这项工作提出了一种简化的多功能策略,以克服癌症疫苗的递送障碍。
{"title":"Octopus-inspired polymeric nanovaccine enables high antigen loading and robust T cell activation for cancer immunotherapy","authors":"Zijuan Wang ,&nbsp;Yuanzhen Su ,&nbsp;Shucheng Zhang ,&nbsp;Bingzheng Yu ,&nbsp;Dongbo Chen ,&nbsp;Xiang Gao ,&nbsp;Yan Wei ,&nbsp;Irina A. Veselova ,&nbsp;Mingqiang Li ,&nbsp;Shixian Lv","doi":"10.1016/j.jconrel.2025.114562","DOIUrl":"10.1016/j.jconrel.2025.114562","url":null,"abstract":"<div><div>Tumor vaccines hold significant promise for immunotherapy, but are limited by low antigen loading capacity, inefficient cytosolic delivery, and suboptimal T cell activation. Here, we present an octopus-inspired polymeric nanovaccine that integrates high antigen-loading capacity and effective cytosolic delivery within a single polymeric platform. The nanovaccine is constructed by encapsulating antigens with an imidazole-functionalized fluorinated polyethyleneimine and Mn<sup>2+</sup> ions, forming a structure that mimics octopus tentacles and suction cups, where the PEI backbone acts as tentacle-like arms and the imidazole-Mn<sup>2+</sup> units serve as suction cups. This multivalent interface enables robust antigen binding through electrostatic, coordination, and hydrophobic interactions. Beyond stabilizing the antigen payload, the amphiphilic cationic design of the polymers offers efficient cytosolic delivery of antigens into dendritic cells (DCs). Meanwhile, the intracellular release of Mn<sup>2+</sup> activates the STING pathway, promoting innate immune responses. Consequently, the vaccine elicits robust antigen-specific CD8<sup>+</sup> T cell responses and durable antitumor immunity in multiple tumor models. This work presents a streamlined, multifunctional strategy to overcome delivery barriers in cancer vaccines.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114562"},"PeriodicalIF":11.5,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145796069","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
Antigen-conjugated scaffolds enable sustained delivery of antigen and enrichment of antigen-specific T-cells 抗原偶联支架能够持续递送抗原和富集抗原特异性t细胞
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-20 DOI: 10.1016/j.jconrel.2025.114564
Sydney N. Wheeler , Mary E. Dickenson , Connor N. Joyce , Samantha N. Lukpat , Leon J.M.W. Wagner , Andrés R. Muñoz-Rojas , Aaron H. Morris
A thorough understanding of T-cell dynamics and interactions could improve patient care in autoimmunity, cancer immunotherapy, and myriad other conditions, yet monitoring antigen-specific T-cell clones is challenging. T-cells recognize antigens presented by antigen-presenting cells (APCs) in the context of major histocompatibility complexes (MHCs). Specific T-cell clones are rare in the blood (<1 in 100,000), and thus cell expansion which consequently alters cell phenotype and function is typically necessary before analysis. This motivates the development of new methods for enriching T-cell populations of interest without phenotypically altering them. Recent work has demonstrated that implantable biomaterial systems can recruit disease-relevant cells in autoimmune conditions, and that if antigens are present, antigen-specific T-cells become enriched in these materials. To date, antigen-loaded materials have exhibited uncontrolled loading, burst release, and subsequent T-cell exhaustion. In this report, we engineer a novel biomaterial antigen delivery system by conjugating antigens to the polymer backbone prior to porous scaffold fabrication. We demonstrate that this technique enables precise antigen loading via ratiometric mixing of modified and unmodified polymer. We show controlled release of antigen into the microenvironment and demonstrate that released antigen is processed and presented by APCs. Using this fabrication method, we achieve sustained release of peptide antigens over a period of 3 weeks in vitro. When implanted in healthy mice, these antigen-conjugated scaffolds are invaded by host myeloid and lymphoid cells and exhibit a dose-dependent enrichment of systemically circulating antigen-specific T-cell populations, while avoiding significant T-cell exhaustion. Finally, we apply this system to an autoantigen from multiple sclerosis (MS) and show release and interaction with autoantigen-specific T-cells. Using this technique, disease-relevant T-cells can be recruited for diagnostic assessment or for immunological research. Future work will investigate the potential of these systems to monitor disease onset and progression in vivo, co-deliver multiple antigens for assessment of epitope spreading, therapeutically target disease-relevant cells within a local niche in situ, and expand the platform for controlled delivery of therapeutic peptides in models beyond autoimmunity.
彻底了解t细胞动力学和相互作用可以改善自身免疫、癌症免疫治疗和无数其他疾病的患者护理,但监测抗原特异性t细胞克隆是具有挑战性的。t细胞识别抗原呈递细胞(APCs)在主要组织相容性复合体(MHCs)的背景下呈递的抗原。特异性t细胞克隆在血液中是罕见的(100,000分之一),因此在分析之前通常需要细胞扩增,从而改变细胞表型和功能。这促使开发新的方法来丰富感兴趣的t细胞群而不改变它们的表型。最近的研究表明,可植入的生物材料系统可以在自身免疫性疾病中招募与疾病相关的细胞,如果存在抗原,抗原特异性t细胞会在这些材料中富集。迄今为止,抗原装载材料已经表现出不受控制的装载、爆裂释放和随后的t细胞衰竭。在本报告中,我们设计了一种新的生物材料抗原递送系统,在多孔支架制造之前,将抗原偶联到聚合物骨架上。我们证明这种技术可以通过比例混合修饰和未修饰的聚合物来精确地装载抗原。我们展示了抗原在微环境中的受控释放,并证明释放的抗原由apc加工和提呈。使用这种制造方法,我们在体外实现了3 周内肽抗原的持续释放。当植入健康小鼠体内时,这些抗原结合支架受到宿主髓细胞和淋巴细胞的侵袭,并表现出系统性循环抗原特异性t细胞群的剂量依赖性富集,同时避免显著的t细胞衰竭。最后,我们将该系统应用于多发性硬化症(MS)的自身抗原,并显示其释放和与自身抗原特异性t细胞的相互作用。利用这种技术,疾病相关的t细胞可以用于诊断评估或免疫学研究。未来的工作将研究这些系统在体内监测疾病发生和进展的潜力,共同递送多种抗原以评估表位扩散,治疗性地靶向局部生态位内的疾病相关细胞,并扩大在自身免疫以外的模型中控制治疗肽递送的平台。
{"title":"Antigen-conjugated scaffolds enable sustained delivery of antigen and enrichment of antigen-specific T-cells","authors":"Sydney N. Wheeler ,&nbsp;Mary E. Dickenson ,&nbsp;Connor N. Joyce ,&nbsp;Samantha N. Lukpat ,&nbsp;Leon J.M.W. Wagner ,&nbsp;Andrés R. Muñoz-Rojas ,&nbsp;Aaron H. Morris","doi":"10.1016/j.jconrel.2025.114564","DOIUrl":"10.1016/j.jconrel.2025.114564","url":null,"abstract":"<div><div>A thorough understanding of T-cell dynamics and interactions could improve patient care in autoimmunity, cancer immunotherapy, and myriad other conditions, yet monitoring antigen-specific T-cell clones is challenging. T-cells recognize antigens presented by antigen-presenting cells (APCs) in the context of major histocompatibility complexes (MHCs). Specific T-cell clones are rare in the blood (&lt;1 in 100,000), and thus cell expansion which consequently alters cell phenotype and function is typically necessary before analysis. This motivates the development of new methods for enriching T-cell populations of interest without phenotypically altering them. Recent work has demonstrated that implantable biomaterial systems can recruit disease-relevant cells in autoimmune conditions, and that if antigens are present, antigen-specific T-cells become enriched in these materials. To date, antigen-loaded materials have exhibited uncontrolled loading, burst release, and subsequent T-cell exhaustion. In this report, we engineer a novel biomaterial antigen delivery system by conjugating antigens to the polymer backbone prior to porous scaffold fabrication. We demonstrate that this technique enables precise antigen loading <em>via</em> ratiometric mixing of modified and unmodified polymer. We show controlled release of antigen into the microenvironment and demonstrate that released antigen is processed and presented by APCs. Using this fabrication method, we achieve sustained release of peptide antigens over a period of 3 weeks <em>in vitro</em>. When implanted in healthy mice, these antigen-conjugated scaffolds are invaded by host myeloid and lymphoid cells and exhibit a dose-dependent enrichment of systemically circulating antigen-specific T-cell populations, while avoiding significant T-cell exhaustion. Finally, we apply this system to an autoantigen from multiple sclerosis (MS) and show release and interaction with autoantigen-specific T-cells. Using this technique, disease-relevant T-cells can be recruited for diagnostic assessment or for immunological research. Future work will investigate the potential of these systems to monitor disease onset and progression <em>in vivo</em>, co-deliver multiple antigens for assessment of epitope spreading, therapeutically target disease-relevant cells within a local niche <em>in situ</em>, and expand the platform for controlled delivery of therapeutic peptides in models beyond autoimmunity.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114564"},"PeriodicalIF":11.5,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145796070","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
Localized inflammasome inhibition mitigates foreign body response to subcutaneous long-acting antiretroviral therapy for HIV 局部炎性体抑制减轻了皮下长效抗逆转录病毒治疗HIV的异物反应
IF 11.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-12-20 DOI: 10.1016/j.jconrel.2025.114559
Ilaria Facchi , Nicola Di Trani , Camden Caffey , Thi Thao Linh Nguyen , Yongbin Liu , Junjun Zheng , Junhua Mai , Fernanda P. Pons-Faudoa , Yitian Xu , Shu-Hsia Chen , Jason T. Kimata , Joan E. Nichols , Corrine Ying Xuan Chua , Alessandro Grattoni
Long-acting antiretroviral therapy (LA-ART) holds promise for improving adherence and viral suppression in human immunodeficiency virus (HIV) prevention and treatment, respectively. These LA-ART encompass different delivery modalities such as intravaginal rings, subcutaneous implants, and intramuscular or subcutaneous injectables. However, subcutaneous implants, especially those containing tenofovir alafenamide (TAF), can trigger local inflammation. In this study, we incorporated MCC950, a selective NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inhibitor, into a subcutaneous nanofluidic implant co-delivering TAF and bictegravir (BIC). In a rodent model, MCC950 reduced local inflammation, fibrotic capsule formation, and inflammatory cell infiltration without affecting the antiviral activity of TAF or BIC. Sustained plasma levels of both drugs were maintained for up to 45 days, and imaging mass cytometry and histological analyses confirmed localized immunomodulation. These findings establish inflammasome inhibition as a viable strategy to improve the safety and tolerability of subcutaneous LA-ART and lay the groundwork for future immunomodulatory-enhanced drug delivery systems.
长效抗逆转录病毒疗法(LA-ART)有望分别改善人类免疫缺陷病毒(HIV)预防和治疗中的依从性和病毒抑制。这些LA-ART包括不同的递送方式,如阴道内环、皮下植入、肌肉注射或皮下注射。然而,皮下植入物,特别是那些含有替诺福韦阿拉芬胺(TAF)的植入物,可引发局部炎症。在这项研究中,我们将选择性NLRP3 (NOD-、LRR-和pyrin结构域蛋白3)抑制剂MCC950加入到皮下纳米流体植入物中,共同递送TAF和比替替韦(BIC)。在啮齿动物模型中,MCC950减少了局部炎症、纤维化囊形成和炎症细胞浸润,而不影响TAF或BIC的抗病毒活性。两种药物的持续血浆水平维持了45 天,成像细胞术和组织学分析证实了局部免疫调节。这些发现证实了炎症小体抑制是提高皮下LA-ART安全性和耐受性的可行策略,并为未来免疫调节增强的药物传递系统奠定了基础。
{"title":"Localized inflammasome inhibition mitigates foreign body response to subcutaneous long-acting antiretroviral therapy for HIV","authors":"Ilaria Facchi ,&nbsp;Nicola Di Trani ,&nbsp;Camden Caffey ,&nbsp;Thi Thao Linh Nguyen ,&nbsp;Yongbin Liu ,&nbsp;Junjun Zheng ,&nbsp;Junhua Mai ,&nbsp;Fernanda P. Pons-Faudoa ,&nbsp;Yitian Xu ,&nbsp;Shu-Hsia Chen ,&nbsp;Jason T. Kimata ,&nbsp;Joan E. Nichols ,&nbsp;Corrine Ying Xuan Chua ,&nbsp;Alessandro Grattoni","doi":"10.1016/j.jconrel.2025.114559","DOIUrl":"10.1016/j.jconrel.2025.114559","url":null,"abstract":"<div><div>Long-acting antiretroviral therapy (LA-ART) holds promise for improving adherence and viral suppression in human immunodeficiency virus (HIV) prevention and treatment, respectively. These LA-ART encompass different delivery modalities such as intravaginal rings, subcutaneous implants, and intramuscular or subcutaneous injectables. However, subcutaneous implants, especially those containing tenofovir alafenamide (TAF), can trigger local inflammation. In this study, we incorporated MCC950, a selective NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inhibitor, into a subcutaneous nanofluidic implant co-delivering TAF and bictegravir (BIC). In a rodent model, MCC950 reduced local inflammation, fibrotic capsule formation, and inflammatory cell infiltration without affecting the antiviral activity of TAF or BIC. Sustained plasma levels of both drugs were maintained for up to 45 days, and imaging mass cytometry and histological analyses confirmed localized immunomodulation. These findings establish inflammasome inhibition as a viable strategy to improve the safety and tolerability of subcutaneous LA-ART and lay the groundwork for future immunomodulatory-enhanced drug delivery systems.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"390 ","pages":"Article 114559"},"PeriodicalIF":11.5,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145796073","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:604180095
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