Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-04-01DOI: 10.1080/10717544.2025.2482195
Jie Chen, Enhui Ren, Ze Tao, Hongyu Lu, Yunchuan Huang, Jing Li, Yuzhe Chen, Zhuo Chen, Tianshan She, Hao Yang, Hong Zhu, Xiaofeng Lu
{"title":"Orchestrating T and NK cells for tumor immunotherapy via NKG2A-targeted delivery of a de novo designed IL-2Rβγ agonist.","authors":"Jie Chen, Enhui Ren, Ze Tao, Hongyu Lu, Yunchuan Huang, Jing Li, Yuzhe Chen, Zhuo Chen, Tianshan She, Hao Yang, Hong Zhu, Xiaofeng Lu","doi":"10.1080/10717544.2025.2482195","DOIUrl":"10.1080/10717544.2025.2482195","url":null,"abstract":"<p><p>As T and NK cell exhaustion is attributed to increased expression of immune checkpoints and decreased production of proliferative cytokines by these cells, immune checkpoint-targeted delivery of proliferative cytokines might induce robust and sustained antitumor immune responses. Here, the expression profile of NKG2A was first found to be narrower than that of PD-1 in tumor-infiltrated immune cells. Moreover, unlike PD-1, NKG2A was predominantly co-expressed with IL-2Rβγ in tumor-infiltrated CD8<sup>+</sup> T and NK cells, but not in Tregs, suggesting that NKG2A might be an ideal target for delivery of IL-2Rβγ agonists to overcome T and NK exhausting. For NKG2A-targeted delivery of an IL-2Rβγ agonist, a single molecule of de novo designed N215 endowed with Immunoglobin G(IgG)-binding ability was coupled to an antibody against NKG2A (αNKG2A) to produce αNKG2A-N215. NKG2A- and IL-2Rβγ-binding were well preserved in αNKG2A-N215, allowing αNKG2A-N215 to act as both an immune checkpoint inhibitor and a T and NK cell stimulator. Intravenously injected αNKG2A-N215 predominantly induced expansion of tumor-infiltrated CD8<sup>+</sup> T and NK cells while showing little stimulation of Tregs. Compared with the separate combination using αNKG2A and N215, αNKG2A-N215 exerted a greater antitumor effect in mice bearing MC38 or B16/F1 tumors. 50% of mice bearing MC38 tumors were cured by αNKG2A-N215, and long-term immunological memory against the tumor was induced in these mice. These results indicate that NKG2A is another ideal target for delivery of an IL-2Rβγ agonist, and αNKG2A-N215, with specificities for both NKG2A and IL-2Rβγ, might be developed as a novel agent for immunotherapy.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2482195"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11966987/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143763232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Application of chitosan-based drug delivery systems in the treatment of bacterial diseases: a review.","authors":"Huan Huang, Yaxin Zhou, Jiehang Li, Zhijin Zhang, RongJia Han, Jingru Zuo, Yubin Bai, Jiyu Zhang","doi":"10.1080/10717544.2025.2514140","DOIUrl":"10.1080/10717544.2025.2514140","url":null,"abstract":"<p><p>Bacterial diseases are a significant challenge to human and animal health. The current treatment methods still have obvious shortcomings, such as poor targeting, low bioavailability, high side effects and drug resistance. Chitosan, with its outstanding biocompatibility, biodegradability, adhesiveness, antimicrobial properties, and ability to minimize drug side effects while improving bioavailability and therapeutic outcomes, serves as an ideal material for drug delivery systems, presenting a promising strategy for treating bacterial diseases. In this review, we briefly summarize the preparation methods of chitosan-based drug delivery systems and their application in the treatment of bacterial infections. The advantages of preparation of different types of chitosan-based drug delivery systems are discussed, supported by examples demonstrating their ability to improve drug antimicrobial activity, targeting, and bioavailability. Moreover, the current challenges, limitations, and future perspectives in this field were discussed, laying the groundwork for further development of chitosan-based drug delivery systems as high-performance and safe antimicrobial therapeutics.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2514140"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12152997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144257600","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Ionic liquid-iontophoresis mediates transdermal delivery of sparingly soluble drugs.","authors":"Wenyan Gao, Wenmin Xing, Zhan Tang, Qiao Wang, Wenying Yu, Qi Zhang","doi":"10.1080/10717544.2025.2489730","DOIUrl":"https://doi.org/10.1080/10717544.2025.2489730","url":null,"abstract":"<p><p>Low solubility restricted transdermal penetration of drugs. We aimed to develop a novel ionic liquid-iontophoresis (IL-IS) technology and assess their efficacy and primary factors in facilitating transdermal drug delivery. Five choline-based ILs with different chain length were synthesized and validated, and the impact of IL and/or IS technology on transdermal penetration of model drugs were investigated. The results indicated that five groups of ILs synthesized in this study exhibited minimal level of toxicity, and the longer the chain of acid ligands of ILs, the greater the cytotoxicity. The longer chain of acid ligand was demonstrated superior solubilizing capabilities compared to the shorter chain. Cinnamic acid-choline-based IL ([Cho] [Cin]) significantly improved permeation of all three model drugs, and permeation quantity was linearly positively associated with the concentration of ILs. The 10 h cumulative permeation of aripiprazole applied with ILs alone was enhanced by about 14-fold when paired with IS, and the penetration was linearly positively associated with the concentration and current strength of the ILs. <i>In vivo</i> results indicated that IL and/or IS technology primarily facilitated drug penetration into the skin, with potential involvement of endocytosis in this process. This study demonstrated that [Cho] [Cin] exhibited a significant enhancement in the transdermal delivery of three sparingly soluble drugs. It further enhanced the transdermal permeation of weak base drug following with the combining IL and IS technology. These findings highlighted that the IL-IS technology holded promise for facilitating the transdermal delivery of sparingly soluble and weak base drugs.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2489730"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12013143/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143997039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-06-12DOI: 10.1080/10717544.2025.2509969
Ryouken Kimura, Tomoyoshi Yamano, Uryo Onishi, Xiabing Lyu, Kanto Nagamori, Toan Van Le, Mitsutoshi Nakada, Rikinari Hanayama
{"title":"Selective expansion and differentiation of antigen-specific CD4<sup>+</sup> T-helper cells by engineered extracellular vesicles.","authors":"Ryouken Kimura, Tomoyoshi Yamano, Uryo Onishi, Xiabing Lyu, Kanto Nagamori, Toan Van Le, Mitsutoshi Nakada, Rikinari Hanayama","doi":"10.1080/10717544.2025.2509969","DOIUrl":"https://doi.org/10.1080/10717544.2025.2509969","url":null,"abstract":"<p><p>Extracellular vesicles (EVs), particularly small EVs (sEVs), are lipid bilayer vesicles secreted by various cell types and play a key role in intercellular communication. These vesicles are promising tools for cancer immunotherapy owing to their biocompatibility, low immunogenicity, and capacity for targeted drug delivery. In this study, we aimed to assess the potential of engineered antigen-presenting EVs (AP-EVs) to selectively expand and differentiate antigen-specific CD4<sup>+</sup> T cells. We engineered two types of AP-EVs: AP-EVs-Th1 expressing MHC class II, CD80, and interleukin (IL)-12 on their surface to promote Th1 differentiation, and AP-EVs-Th2 expressing MHC class II, CD80, and IL-4 to induce Th2 differentiation. <i>In vitro</i> experiments demonstrated that AP-EVs successfully induced the antigen-specific proliferation and differentiation of Th1 and Th2 cells, respectively. Notably, <i>in vivo</i> administration of AP-EVs-Th1 significantly enhanced the proliferation and differentiation of tumor antigen-specific Th1 cells, leading to robust anti-tumor effects in a murine melanoma model. These findings highlight the potential of AP-EVs-Th1 for cancer immunotherapy, particularly in augmenting CD4<sup>+</sup> T cell responses. Furthermore, the versatility and adaptability of EV-based therapies make them beneficial for the development of personalized immunotherapeutic strategies for various cancer types, offering the advantages of targeted immune modulation, ease of use, and reduced risk compared to cell-based therapies.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2509969"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144274460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-04-23DOI: 10.1080/10717544.2025.2494775
Dmitri Simberg, Yechezkel Barenholz, Steve R Roffler, Katharina Landfester, Alexander V Kabanov, Seyed M Moghimi
{"title":"PEGylation technology: addressing concerns, moving forward.","authors":"Dmitri Simberg, Yechezkel Barenholz, Steve R Roffler, Katharina Landfester, Alexander V Kabanov, Seyed M Moghimi","doi":"10.1080/10717544.2025.2494775","DOIUrl":"https://doi.org/10.1080/10717544.2025.2494775","url":null,"abstract":"<p><p>PEGylation technology, that is grafting of poly(ethylene glycol)(PEG) to biologics, vaccines and nanopharmaceuticals, has become a cornerstone of modern medicines with over thirty products used in the clinic. PEGylation of therapeutic proteins, nucleic acids and nanopharmaceuticals improves their stability, pharmacokinetic and biodistribution. While PEGylated medicines are safe in the majority of patients, there are growing concerns about the emergence of anti-PEG antibodies and their impact on the therapeutic efficacy of PEGylated medicines as well as broader immune responses, particularly in complement activation and hypersensitivity reactions. These concerns are beginning to scrutinize the future viability of PEGylation technology in medicine design. Here, we outline these concerns, encourage more efforts into looking for comprehensive scientific evidence on the role of anti-PEG antibodies in hypersensitivity reactions, discuss alternatives to PEG and propose strategies for moving PEGylation technology forward.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2494775"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12020137/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143983775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-05-29DOI: 10.1080/10717544.2025.2509962
Xiao-Yun Hou, Xiao-Ling Zhang, An-Kang Ying, Yu-Xin Yue, Tao Yang, Dong-Sheng Guo, Zhi-Qing Li
{"title":"Ophthalmic formulation of methotrexate: a strategy of using the self-assembled LacAC4A nanoparticles for non-invasive drug delivery to the ocular posterior segment.","authors":"Xiao-Yun Hou, Xiao-Ling Zhang, An-Kang Ying, Yu-Xin Yue, Tao Yang, Dong-Sheng Guo, Zhi-Qing Li","doi":"10.1080/10717544.2025.2509962","DOIUrl":"10.1080/10717544.2025.2509962","url":null,"abstract":"<p><p>Drug delivery to ocular posterior segment remains difficult due to the challenges imposed by dynamic and static ocular barriers, lesion point targeting, and off-target effect. In this study, a novel approach is demonstrated for non-invasive drug delivery to the ocular posterior segments using lactose-modified azocalix[4] arene (LacAC4A) as a supramolecular ocular drug delivery platform. LacAC4A contains azo groups and is covalently modified by lactose groups, which confers active targeting to the retina, and induces a hypoxic response. The immunomodulator methotrexate (MTX), which is commonly used in ophthalmology to treat immune system diseases such as uveitis, was also selected as a guest to prepare MTX@LacAC4A. The prepared LacAC4A and MTX@LacAC4A systems were characterized, then the internalization mechanisms and hypoxia response abilities were determined through flow cytometry and fluorescence imaging, respectively. Besides, the delivery route and efficiency were verified, and the safety profile of MTX@LacAC4A was evaluated in multiple dimensions. Importantly, it was found that the prepared MTX@LacAC4A exhibits good biocompatibility, can effectively reach the posterior segment, and demonstrates potential ophthalmic applications. These findings lay the grounds for the future development of non-invasive ocular posterior segment disease treatments based on the advanced use of LacAC4A as a drug delivery platform.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2509962"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12123953/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173020","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-05-29DOI: 10.1080/10717544.2025.2509973
Ravi Parashar, Preeti K Suresh
{"title":"Emerging liposomal therapies for diabetic retinopathy: a review of novel targeting approaches and advances in retinal health outcomes.","authors":"Ravi Parashar, Preeti K Suresh","doi":"10.1080/10717544.2025.2509973","DOIUrl":"10.1080/10717544.2025.2509973","url":null,"abstract":"<p><p>Diabetic retinopathy (DR), which affects over millions of individuals globally, is the leading cause of permanent visual loss. Current therapies, including as intravitreal anti-vascular endothelial growth factor (VEGF) medications and laser photocoagulation, are limited by frequent dosing and side effects. Liposomes, with their ability to encapsulate hydrophilic and hydrophobic medications, offer tailored delivery, prolonged release, and low systemic toxicity. This study looks at advances in liposomal formulations that address DR's multifactorial etiology, including as anti-angiogenic, anti-inflammatory, and antioxidant processes. We assess new preparation methods (e.g. supercritical CO<sub>2</sub>, microfluidics) and clinical considerations, including stability and cost-effectiveness. To address the heterogeneity of DR, future endeavors will prioritize combinatorial medications and customized therapy.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2509973"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12123966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173016","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Constructed transferrin receptor-targeted liposome for the delivery of fluvoxamine to improve prognosis in a traumatic brain injury mouse model.","authors":"Liang Mi, Jiangyuan Yuan, Yuxing Jiang, Yuqian Hu, Chuanxiang Lv, Yongqiang Xu, Mingqi Liu, Tao Liu, Xuanhui Liu, Jinhao Huang, Rongcai Jiang, Wei Quan","doi":"10.1080/10717544.2025.2486840","DOIUrl":"https://doi.org/10.1080/10717544.2025.2486840","url":null,"abstract":"<p><p>The dysregulation of blood-brain barrier (BBB) activates pathological mechanisms such as neuroinflammation after traumatic brain injury (TBI), and glymphatic system dysfunction accelerates toxic waste accumulation after TBI. It is essential to find an effective way to inhibit inflammation and repair BBB and glymphatic system after TBI; however, effective and lasting drug therapy remains challenging because BBB severely prevents drugs from being delivered to central nervous system. Transferrin receptors (TfRs) are mainly expressed on brain capillary endothelial cells. Here, we report a TfR-targeted nanomedicine for TBI treatment by penetrating BBB and delivering fluvoxamine (Flv). The TfR-targeted polypeptide liposome loaded with Flv (TPL-Flv) implements cell targeting ability on human umbilical vein endothelial cells (HUVECs) <i>in vitro</i> detected by flow cytometry, and drug safety was proved through cell viability analysis and blood routine and biochemistry analysis. Afterwards, we established a controlled cortical impact model to explore TPL-Flv administration effects on TBI mice. We confirmed that TPL-Flv could stimulate CXCR4/SDF-1 signaling pathway, activate Treg cells, and inhibit inflammation after TBI. TPL-Flv treatment also alleviated BBB disruption and restored aquaporin-4 (AQP4) polarization, as well as reversed glymphatic dysfunction. Furthermore, TPL-Flv accomplished remarkable improvement of motor and cognitive functions. These findings demonstrate that TPL-Flv can effectively cross BBB and achieve drug delivery to cerebral tissue, validating its potential to improve therapeutic outcomes for TBI.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2486840"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12001850/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143977814","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-05-26DOI: 10.1080/10717544.2025.2486340
Barnabas Nyesiga, Karin Hägerbrand, Laura Varas, Anette Gjörloff Wingren, Mats Ohlin, Peter Ellmark, Laura von Schantz
{"title":"Antigenic peptide delivery to antigen-presenting cells using a CD40-coiled coil affinity-based platform.","authors":"Barnabas Nyesiga, Karin Hägerbrand, Laura Varas, Anette Gjörloff Wingren, Mats Ohlin, Peter Ellmark, Laura von Schantz","doi":"10.1080/10717544.2025.2486340","DOIUrl":"10.1080/10717544.2025.2486340","url":null,"abstract":"<p><p>Delivery of antigenic peptides to antigen presenting cells (APCs) such as dendritic cells (DCs) using monoclonal antibodies (mAbs) is an attractive approach to evoke antigen-specific T cell activation and improve drug efficacy. Peptide linkage to mAbs has previously been achieved through genetic fusion, chemical conjugation, nano-engineered platforms and high affinity peptides. In this study, we have developed a flexible antibody-peptide linking technology using oppositely charged coiled coil domains to non-covalently link peptides to mAbs. The technology comprises (1) an anti-CD40 mAb connected with negatively charged E domains and (2) an immunogenic OVA peptide (SIINFEKL) from ovalbumin used as a model antigenic peptide fused with positively charged K domains. Combining these constructs leads to the formation of complexes that can be targeted to CD40 expressed on cells. Proof of concept antibody constructs connected with E domains generated from transient expressions exhibited good manufacturability, binding, and stability attributes comparable to a control mAb. Also, optimal repeat lengths for coiled-coil oligomerization domains were identified in these studies. Binding kinetics studies showed that connecting E domains to mAbs do not impede Fc gamma and neonatal receptor interactions. Additionally, formation of stable complexes capable of binding CD40 expressing cells was demonstrated <i>in vitro. In vivo</i> functionality evaluations showed that treatment of human CD40 transgenic mice with complexes elicited expansion of OVA peptide-specific CD8+ T cells and potent antitumor effects superior to peptide monotherapies. Overall, these findings demonstrate that the technology has great potential for application as an <i>in vivo</i> tool for antigenic peptide delivery.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2486340"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12107651/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-03-03DOI: 10.1080/10717544.2025.2465909
Mehrnoosh Rafiei, Akbar Shojaei, Ying Chau
{"title":"Machine learning-assisted design of immunomodulatory lipid nanoparticles for delivery of mRNA to repolarize hyperactivated microglia.","authors":"Mehrnoosh Rafiei, Akbar Shojaei, Ying Chau","doi":"10.1080/10717544.2025.2465909","DOIUrl":"10.1080/10717544.2025.2465909","url":null,"abstract":"<p><p>Regulating inflammatory microglia presents a promising strategy for treating neurodegenerative and autoimmune disorders, yet effective therapeutic agents delivery to these cells remains a challenge. This study investigates modified lipid nanoparticles (LNP) for mRNA delivery to hyperactivated microglia, particularly those with pro-inflammatory characteristics, utilizing supervised machine learning (ML) classifiers. We developed and screened a library of 216 LNP formulations with varying lipid compositions, N/P ratios, and hyaluronic acid (HA) modifications. The transfection efficiency of eGFP mRNA was assessed in the BV-2 murine microglia cell line under different immunological states, including resting and activated conditions (LPS-activated and IL4/IL13-activated). ML-guided morphometric analysis tracked the phenotypes of various microglia subtypes before and after transfection. Four supervised ML classifiers were investigated to predict transfection efficiency and phenotypic changes based on LNP design parameters. The Multi-Layer Perceptron (MLP) neural network emerged as the best-performing model, achieving weighted F1-scores ≥0.8. While it accurately predicted responses from LPS-activated and resting cells, it struggled with IL4/IL13-activated cells. The MLP model was validated by predicting the performance of four unseen LNP formulations delivering eGFP mRNA to LPS-activated BV2 cells. HA-LNP2 emerged as optimal formulation for delivering target IL10 mRNA, effectively suppressing inflammatory phenotypes, evidenced by shifts in cell morphology, increased IL10 expression, and reduced TNF-α levels. We also evaluated HA-LNP2 on LPS-activated human iPSC-derived microglia, confirming its efficacy in modulating inflammatory responses. This study highlights the potential of tailored LNP design and ML techniques to enhance mRNA therapy for neuroinflammatory disorders by leveraging carrier's immunogenic properties to modulate microglial responses.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2465909"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11878168/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143540603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}