Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (416)

Search Parameters:
Keywords = LNPs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1255 KB  
Article
Development of an mRNA Vaccine for Tick-Borne Encephalitis: Selection of a Prototype Virus Strain
by Maria A. Nikiforova, Vladimir A. Gushchin, Denis A. Kleymenov, Anastasia M. Kocherzhenko, Evgeniia N. Bykonia, Elena P. Mazunina, Sofia R. Kozlova, Leonid I. Russu, Nadezhda A. Kuznetsova, Elena V. Shidlovskaya, Elizaveta V. Marchuk, Evgeny V. Usachev, Olga V. Usacheva, Dmitry V. Shcheblyakov, Irina V. Kozlova, Sergei E. Tkachev, Andrei A. Pochtovyi, Vladimir I. Zlobin, Denis Y. Logunov and Alexander L. Gintsburg
Vaccines 2026, 14(1), 107; https://doi.org/10.3390/vaccines14010107 (registering DOI) - 21 Jan 2026
Abstract
Background/Objectives: While tick-borne encephalitis virus (TBEV) is genetically relatively conserved, the significant antigenic divergence between its main circulating subtypes hinders the development of broadly effective antiviral treatments and vaccines. Current inactivated TBEV vaccines offer limited cross-protection against heterologous strains, as evidenced by [...] Read more.
Background/Objectives: While tick-borne encephalitis virus (TBEV) is genetically relatively conserved, the significant antigenic divergence between its main circulating subtypes hinders the development of broadly effective antiviral treatments and vaccines. Current inactivated TBEV vaccines offer limited cross-protection against heterologous strains, as evidenced by cases among vaccinated individuals in endemic regions. The aim of this study was to design a candidate mRNA vaccine and evaluate the breadth of protective immunity it elicits. Methods: Ten candidate mRNA-PrM/E-LNP vaccines were comparatively evaluated for immunogenicity and protective efficacy in BALB/c mice. Immunogenicity was assessed by measuring antigen-specific IgG titers via ELISA and neutralizing antibody titers against a panel of TBEV strains using a virus-neutralization test. Protective efficiency was determined in a lethal challenge model, where immunized mice were challenged with one of seven distinct TBEV strains. Results: Vaccination with all tested mRNA-PrM/E-LNP candidates conferred 100% survival in mice following a lethal challenge with each of the seven TBEV strains (100 LD50). The construct mRNA-PrM/E—Krasny Yar-8 demonstrated the highest immunogenicity, inducing antigen-specific antibodies with a geometric mean titer (GMT) of 1:6625, as well as the broadest virus-neutralizing activity against both homologous and heterologous TBEV strains in vitro. Conclusions: The mRNA platform represents a promising strategy for developing TBEV vaccines, demonstrating high immunogenicity and cross-protective efficacy against diverse viral strains. Full article
(This article belongs to the Special Issue Feature Papers of DNA and mRNA Vaccines)
22 pages, 3249 KB  
Article
Freeze-Drying in Sucrose Followed by Cryomilling Enables the Formulation of sa-mRNA–LNP Powders for Inhalation
by E. M. Jansen, M. J. R. Ruigrok, M. S. Suh, P. M. Ruppel, Xiaole Cui, L. Opsomer, N. N. Sanders, H. W. Frijlink and W. L. J. Hinrichs
Pharmaceutics 2026, 18(1), 121; https://doi.org/10.3390/pharmaceutics18010121 - 18 Jan 2026
Viewed by 175
Abstract
Background: Self-amplifying mRNA (sa-mRNA) represents a promising platform for vaccines and gene therapies, offering sustained protein expression at low doses through self-replication. For vaccines targeting respiratory pathogens, pulmonary delivery of sa-mRNA lipid nanoparticles (LNPs) is particularly advantageous, enabling direct delivery to the infection [...] Read more.
Background: Self-amplifying mRNA (sa-mRNA) represents a promising platform for vaccines and gene therapies, offering sustained protein expression at low doses through self-replication. For vaccines targeting respiratory pathogens, pulmonary delivery of sa-mRNA lipid nanoparticles (LNPs) is particularly advantageous, enabling direct delivery to the infection site and induction of mucosal immunity. Objective: In this study, we evaluated the stability of sa-mRNA–LNPs under refrigerated and frozen conditions and developed a dry powder formulation suitable for inhalation, produced by freeze-drying followed by cryomilling with leucine. Methods: sa-mRNA–LNPs formulated in HEPES buffer with 20% (w/v) sucrose were stored for up to 8 weeks as liquid or freeze-dried samples at various temperatures (−80 °C, −20 °C, 4 °C, and 20 °C). Biological stability was assessed by transfection efficiency in HeLa cells, while physical stability was characterized by encapsulation efficiency, zeta potential, particle size, and polydispersity index. Results: Liquid formulations remained stable for at least 8 weeks at −80 °C and −20 °C but rapidly lost stability at 4 °C and 20 °C. Freeze-drying effectively preserved sa-mRNA–LNP functionality and structural integrity for up to 8 weeks at 4 °C, with only minor structural changes. Subsequent cryomilling in the presence of 4 wt-% leucine produced a respirable dry powder while retaining approximately 60% of the original sa-mRNA–LNP functionality. Although cryomilling induced some structural alterations, the remaining functional fraction remained stable during storage. The resulting powders displayed favorable aerosol performance for deep lung delivery, as demonstrated by cascade impaction (MMAD = 4.13 ± 0.26 µm). Conclusions: In conclusion, freeze-drying effectively preserved sa-mRNA–LNP integrity at 4 °C, whereas cryomilling with leucine produced a respirable dry powder suitable for pulmonary delivery, providing a foundation for globally accessible, needle-free sa-mRNA vaccines against respiratory diseases. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Graphical abstract

19 pages, 4966 KB  
Article
Self-Multimerization of mRNA LNP-Derived Antigen Improves Antibody Responses
by Cody A. Despins, James Round, Lisa Dreolini, Tracy S. Lee, Scott D. Brown and Robert A. Holt
Vaccines 2026, 14(1), 80; https://doi.org/10.3390/vaccines14010080 - 12 Jan 2026
Viewed by 329
Abstract
Background: mRNA LNP technology is now being widely applied as a highly effective vaccine platform. Antigen multimerization is a well-established approach to enhance the antibody titers and protective efficacy of several protein subunit vaccines. However, this approach has been less explored for [...] Read more.
Background: mRNA LNP technology is now being widely applied as a highly effective vaccine platform. Antigen multimerization is a well-established approach to enhance the antibody titers and protective efficacy of several protein subunit vaccines. However, this approach has been less explored for mRNA LNP vaccines. Methods: Here, within the context of mRNA LNP vaccination, we used mStrawberry (mSb) as a model antigen to conduct a comprehensive, head-to-head comparison of the ability of the foldon (3-mer), IMX313 (7-mer), and ferritin (24-mer) multimerization domains to enhance immunogenicity in mice. Results: We compared multimerized antigen to monomeric secreted antigen and monomeric surface-displayed antigen and observed that the IMX313 domain efficiently multimerized mSb protein and significantly enhanced anti-mSb antibody titers, whereas the foldon and ferritin domains failed to multimerize or improve antibody levels. Conclusions: Our results extend the observation of improved immunogenicity from antigen multimerization to mRNA LNP vaccines and indicate that the 7-mer forming IMX313 multimerization domain may be an ideal candidate for multimer formation in the context of mRNA LNP vaccination. Future studies are needed to evaluate the multimerization of pathogen-derived antigens, in the mRNA LNP format, for the enhancement of neutralization and protective efficacy. Full article
(This article belongs to the Special Issue Feature Papers of DNA and mRNA Vaccines)
Show Figures

Figure 1

38 pages, 2595 KB  
Review
Gene Editing Therapies Targeting Lipid Metabolism for Cardiovascular Disease: Tools, Delivery Strategies, and Clinical Progress
by Zhuoying Ren, Jun Zhou, Dongshan Yang, Yanhong Guo, Jifeng Zhang, Jie Xu and Y Eugene Chen
Cells 2026, 15(2), 134; https://doi.org/10.3390/cells15020134 - 12 Jan 2026
Viewed by 513
Abstract
Gene editing technologies have revolutionized therapeutic development, offering potentially curative and preventative strategies for cardiovascular disease (CVD), which remains a leading global cause of morbidity and mortality. This review provides an introduction to the state-of-the-art gene editing tools—including ZFNs, TALENs, CRISPR/Cas9 systems, base [...] Read more.
Gene editing technologies have revolutionized therapeutic development, offering potentially curative and preventative strategies for cardiovascular disease (CVD), which remains a leading global cause of morbidity and mortality. This review provides an introduction to the state-of-the-art gene editing tools—including ZFNs, TALENs, CRISPR/Cas9 systems, base editors, and prime editors—and evaluates their application in lipid metabolic pathways central to CVD pathogenesis. Emphasis is placed on targets such as PCSK9, ANGPTL3, CETP, APOC3, ASGR1, LPA, and IDOL, supported by findings from human genetics, preclinical models, and recent first-in-human trials. Emerging delivery vehicles (AAVs, LNPs, lentivirus, virus-like particles) and their translational implications are discussed. The review highlights ongoing clinical trials employing liver-targeted in vivo editing modalities (LivGETx-CVD) and provides insights into challenges in delivery, off-target effects, genotoxicity, and immunogenicity. Collectively, this review captures the rapid progress of LivGETx-CVD from conceptual innovation to clinical application, and positions gene editing as a transformative, single-dose strategy with the potential to redefine prevention and long-term management of dyslipidemia and atherosclerotic cardiovascular disease. Full article
(This article belongs to the Special Issue CRISPR-Based Genome Editing in Translational Research—Third Edition)
Show Figures

Figure 1

20 pages, 6259 KB  
Article
Plant-Derived miR-55 Alleviates Liver Fibrosis by Disrupting the CK2α/SMO Complex and Promoting SMO Ubiquitination
by Lei Wu, Jing Yang, Anqi Li, Yuqiang Zhao, Qing Liu, Zhenbo Li, Yihan Liu, Peng Tang and Rui Wang
Int. J. Mol. Sci. 2026, 27(2), 748; https://doi.org/10.3390/ijms27020748 - 12 Jan 2026
Viewed by 136
Abstract
The development of RNA-based drugs for MAFLD-related fibrosis is severely hampered by the poor oral bioavailability of nucleic acids. This study employed a novel, patent-protected LNP formulation to orally deliver plant-derived miR-55 and investigate its therapeutic potential, focusing on its novel mechanism of [...] Read more.
The development of RNA-based drugs for MAFLD-related fibrosis is severely hampered by the poor oral bioavailability of nucleic acids. This study employed a novel, patent-protected LNP formulation to orally deliver plant-derived miR-55 and investigate its therapeutic potential, focusing on its novel mechanism of action via the CK2α/SMO interaction. In a rat model established with a methionine-choline-deficient diet, orally administered miR-55 markedly improved liver injury, lipid dysregulation, oxidative stress, and pathological collagen deposition. The anti-fibrotic efficacy was quantitatively confirmed by a significant reduction in hepatic hydroxyproline content and downregulation of key fibrogenic genes (Col1a1, Col3a1, TIMP-1, TGF-β1, CTGF) and pro-inflammatory cytokines (TNF-α, IL-6), achieving effects comparable to the full Ge Xia Zhu Yu Decoction. Mechanistically, both bioinformatic prediction and in vivo validation indicated that miR-55 is predicted to target CK2α. This targeting suppressed CK2α expression and disrupted the endogenous CK2α-SMO complex, thereby promoting the ubiquitin-mediated degradation of SMO—a previously unreported mechanism. This cascade inhibited the downstream Gli1 pathway and downregulated pro-fibrotic and pro-angiogenic factors (VEGF, PDGF), thereby providing a comprehensive mechanistic basis for the therapeutic effects. This study is the first to provide evidence that orally delivered, plant-derived miR-55 may act as a natural modulator that potentially through disrupting the CK2α/SMO interaction via a unique complex disruption-promoted degradation mechanism, attenuating Hedgehog signaling and alleviating liver fibrosis. These findings offer important insights into cross-kingdom regulation and highlight miR-55 as a potential targeted therapeutic candidate. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

25 pages, 351 KB  
Review
Advances in mRNA-Based Melanoma Vaccines: A Narrative Review of Lipid Nanoparticle and Dendritic Cell Delivery Platforms
by Connor K. Sisk, Laci M. Turner, Shafkat Meraj and Nabiha Yusuf
Cells 2026, 15(2), 99; https://doi.org/10.3390/cells15020099 - 6 Jan 2026
Viewed by 519
Abstract
Melanoma remains one of the deadliest cutaneous malignancies worldwide, and despite advances in systemic therapy, recurrence and treatment resistance remain frequent challenges. Following the success of COVID-19 mRNA vaccines, mRNA-based cancer vaccines targeting melanoma antigens have emerged as a promising therapeutic direction. This [...] Read more.
Melanoma remains one of the deadliest cutaneous malignancies worldwide, and despite advances in systemic therapy, recurrence and treatment resistance remain frequent challenges. Following the success of COVID-19 mRNA vaccines, mRNA-based cancer vaccines targeting melanoma antigens have emerged as a promising therapeutic direction. This review summarizes current evidence on mRNA melanoma vaccines, focusing on two leading delivery platforms: lipid nanoparticles (LNPs) and dendritic cell (DC) vaccines. A comprehensive search of MEDLINE, Embase, and Scopus from 2015 to 2025 identified clinical trials, preclinical studies, and review articles evaluating mRNA vaccine constructs and delivery strategies. Completed clinical studies demonstrate that personalized LNP-formulated mRNA vaccines can enhance neoantigen-specific T-cell responses and improve recurrence-free survival, particularly when combined with immune checkpoint inhibitors. DC-based mRNA vaccines also show potent immunogenicity, with stronger responses observed when DC maturation is optimized. Ongoing trials continue to investigate next-generation LNP formulations, DC priming strategies, and personalized neoantigen approaches. Overall, current evidence indicates that both LNP and DC platforms can augment antitumor immunity by broadening T-cell responses and enhancing checkpoint inhibition. Continued refinement of delivery vehicles, neoantigen selection, and scalable manufacturing processes will be essential to realizing the full clinical potential of mRNA vaccines in melanoma. Full article
Show Figures

Graphical abstract

29 pages, 5336 KB  
Review
Lipid-Based Colloidal Nanocarriers for Site-Specific Drug Delivery
by Kamyar Shameli, Behnam Kalali, Hassan Moeini and Aras Kartouzian
Colloids Interfaces 2026, 10(1), 7; https://doi.org/10.3390/colloids10010007 - 4 Jan 2026
Viewed by 590
Abstract
Lipid nanoparticles (LNPs) are now the go-to method for delivering genetic medicines, backed by real-world use in patients. Things like which fats they are made of, their shape at the molecular level, how ingredients mix, and how they are built, matter a lot. [...] Read more.
Lipid nanoparticles (LNPs) are now the go-to method for delivering genetic medicines, backed by real-world use in patients. Things like which fats they are made of, their shape at the molecular level, how ingredients mix, and how they are built, matter a lot. This review attempts to take a close look at how different components, such as ionizable lipids, auxiliary lipids (DSPC, DOPE), cholesterol, and PEG-based lipids, affect the bioavailability of LNPs. It also focuses on key functions of LNPs, including packaging genetic material, escaping cellular traps, spreading in the body, and remaining active in the blood. New data show that lipids with the right handedness and highly sensitive chiroptical quality control can sharpen delivery accuracy and boost transport rates, turning stereochemistry into a practical design knob. Rather than simply listing results, we examine real-world examples that are already used to regulate gene expression, enhance mRNA expression, splenic targeting, and show great potential for gene repair, protein replacement, and DNA base-editing applications. Also, recent advances in AI-based designs for LNPs that take molecular shape into account and help speed up modifications to lipid arrangements and mixture configurations are highlighted. In summary, this paper presents a practical and scientific blueprint to support smarter production of advanced LNPs used in genetic medicine, addressing existing obstacles, balanced with future opportunities. Full article
(This article belongs to the Special Issue Feature Reviews in Colloids and Interfaces)
Show Figures

Figure 1

26 pages, 4811 KB  
Article
Boosting LNP Performance: Higher Concentrations of Lipid Mixtures Improve In Vivo Gene Expression and Storage Stability
by Blerina Shkodra, Ashish Muglikar, Janani Thangapandian, Matthias Schumacher, Burcu Binici and Yvonne Perrie
Pharmaceutics 2026, 18(1), 50; https://doi.org/10.3390/pharmaceutics18010050 - 30 Dec 2025
Viewed by 1080
Abstract
Background: An efficient formulation of lipid nanoparticles (LNPs) is often considered crucial in the successful development of nucleic acid therapeutics. This study explores the impact of varying the lipid and payload concentrations as starting materials on key LNP properties. Results: The [...] Read more.
Background: An efficient formulation of lipid nanoparticles (LNPs) is often considered crucial in the successful development of nucleic acid therapeutics. This study explores the impact of varying the lipid and payload concentrations as starting materials on key LNP properties. Results: The outcomes of the study revealed that the desired particle properties could be retained even at a starting lipid mixture concentration of 70 mg/mL. Particle size remained largely unchanged despite changes in lipid mixture concentration, with polydispersity index values below 0.2. CryoTEM analysis revealed that LNPs prepared using higher lipid mixture concentrations were more uniform and more abundant in solid core morphologies. Buffer composition was shown to influence the LNP particle size, surface charge, and gene expression, as well as storage stability. In vivo studies in mice showed enhanced gene expression and biodistribution for LNPs formulated at higher lipid and RNA concentrations, with LNPs in Tris-sucrose eliciting superior gene expression compared to LNPs in PBS. Conclusions: This study demonstrated that intensified mixing processes based on confined jet-impingement allow the use of elevated starting material concentrations in LNP formulations, resulting in improved biological performance and stability of mRNA-LNPs, as well as enhanced scalability and throughput. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Graphical abstract

19 pages, 1625 KB  
Article
Enhanced Antitumor Effect of Oncolytic Virotherapy Combined with mRNA-Encoded Immunoadjuvants in Colorectal Carcinoma (CT26) Tumor Model
by Anastasia Ryapolova, Daniil Shevyrev, Angela Tsvetkova, Olesya Sokolova, Olga Vasileva, Vladimir Andriianov, Bogdan Krapivin, Alena Kozlova, Kristina Vorona, Sofia Kondratieva, Roman A. Ivanov, Alexander Karabelsky, Vasiliy Reshetnikov and Ekaterina Minskaia
Appl. Biosci. 2026, 5(1), 1; https://doi.org/10.3390/applbiosci5010001 - 22 Dec 2025
Viewed by 492
Abstract
The search for effective anti-cancer therapies is one of the most significant goals of modern medicine. The combination of oncolytic viruses (OV) and mRNA immunoadjuvants can significantly improve the outcome or even substitute traditional immunotherapy. In addition to the direct OV-mediated cytotoxic elimination [...] Read more.
The search for effective anti-cancer therapies is one of the most significant goals of modern medicine. The combination of oncolytic viruses (OV) and mRNA immunoadjuvants can significantly improve the outcome or even substitute traditional immunotherapy. In addition to the direct OV-mediated cytotoxic elimination of tumor cells, both OV and mRNA immunoadjuvants can significantly alter the immunosuppressive tumor microenvironment (TME) supporting cancer cells and unleash the immune response against the malignant cells. The present study is aimed at assessing the therapeutic effects of recombinant vesicular stomatitis virus (rVSV) and lipid nanoparticles (LNP) delivering mRNA coding for murine interleukin-12 (mIL12) and granulocyte-macrophage colony-stimulating factor (mGMCSF) (mRNA-LNP) in colorectal carcinoma CT26-induced tumors both as independent therapies and in combination with each other. The results of the in vivo experiment on BALB/c mice demonstrated that rVSV monotherapy did not have a significant effect, with the tumor growth inhibition index (TGII) ranging from 13.7 to 29.8% on days 6–10 after the therapy start. While monotherapy with mRNA-LNP was more effective (TGII of 48.6–53.7%), it was the therapy combining the two approaches (rVSV and mRNA-LNP) that resulted in the highest TGII of 66.7% on day 10. While these results can be further improved by optimizing the experimental design, they show the great potential of combination immunotherapy for the treatment of oncological diseases. Full article
Show Figures

Figure 1

29 pages, 972 KB  
Systematic Review
A Systematic Review of Advanced Drug Delivery Systems: Engineering Strategies, Barrier Penetration, and Clinical Progress (2016–April 2025)
by Assem B. Uzakova, Elmira M. Yergaliyeva, Azamat Yerlanuly and Zhazira S. Mukatayeva
Pharmaceutics 2026, 18(1), 11; https://doi.org/10.3390/pharmaceutics18010011 - 22 Dec 2025
Viewed by 1023
Abstract
Background/Objectives: Advanced drug delivery systems (DDSs) are essential for targeted delivery, controlled release, and reduced systemic toxicity, but their clinical adoption is limited by biological barriers, manufacturing complexities, and cost. The aim of this systematic review is to critically evaluate the quantitative relationships [...] Read more.
Background/Objectives: Advanced drug delivery systems (DDSs) are essential for targeted delivery, controlled release, and reduced systemic toxicity, but their clinical adoption is limited by biological barriers, manufacturing complexities, and cost. The aim of this systematic review is to critically evaluate the quantitative relationships between platform design, overcoming biological barriers, and clinical translation outcomes for DDS developed between 2016 and 2025. Methods: A comprehensive literature search was conducted in PubMed/MEDLINE, Scopus, and Web of Science (January 2016–April 2025) in accordance with the PRISMA 2020 guidelines. Included studies focused on experimental or clinical data for nanocarrier platforms (liposomes, lipid nanoparticles, polymer systems, biomimetic carriers, extracellular vesicles). Data on platform characteristics, interactions with barriers, pharmacokinetics, manufacturing, and clinical outcomes were extracted and synthesized in narrative form due to the significant methodological heterogeneity. Results: An analysis of 77 included studies confirms that successful clinical translation depends on matching the physicochemical properties of the carrier (size, surface chemistry, material) to specific biological barriers. Liposomes and lipid nanoparticles (LNPs) remain the most clinically validated platforms, exploiting the EPR effect and liver tropism, respectively. Key engineering solutions include stealth coatings, ligand-mediated targeting, and stimulus-responsive materials to overcome barriers such as mononuclear phagocyte system clearance, the blood–brain barrier, and mucosal barriers. Microfluidic and continuous manufacturing processes enable reproducibility, but scalability, cost, and immunogenicity (e.g., anti-PEG responses) remain key translational challenges. Engineered extracellular vesicles, biomimetic carriers, and 3D/4D-printed systems combined with AI-driven design demonstrate the potential for personalized, adaptive delivery. Conclusions: Cutting-edge DDSs have validated their clinical value, but realizing their full potential requires a holistic, patient-centered design approach integrating barrier-specific engineering, scalable manufacturing, and rigorous safety assessment from the earliest stages of development. Further progress will depend on standardizing methods for new platforms (e.g., extracellular vesicles), implementing digital and AI tools, and ensuring translational feasibility as a fundamental principle. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

37 pages, 3930 KB  
Review
Targeted Hepatic Delivery of Bioactive Molecules via Nanovesicles: Recent Developments and Emerging Directions
by Alessia Rita Canestrale, Sharad Kholia, Veronica Dimuccio and Maria Beatriz Herrera Sanchez
J. Pers. Med. 2026, 16(1), 1; https://doi.org/10.3390/jpm16010001 - 19 Dec 2025
Viewed by 566
Abstract
Liver diseases, including fibrosis, viral hepatitis, hepatocellular carcinoma, and monogenic genetic disorders, represent a major global health burden with limited therapeutic options and frequent systemic toxicity from conventional treatments. Nanovesicle-based drug and gene delivery systems offer targeted approaches that may improve therapeutic precision [...] Read more.
Liver diseases, including fibrosis, viral hepatitis, hepatocellular carcinoma, and monogenic genetic disorders, represent a major global health burden with limited therapeutic options and frequent systemic toxicity from conventional treatments. Nanovesicle-based drug and gene delivery systems offer targeted approaches that may improve therapeutic precision and reduce off-target effects. This review aims to evaluate the promise and comparative potential of three key nanovesicle platforms—lipid nanoparticles (LNPs), extracellular vesicles (EVs) and liposomes—for drug and gene delivery in liver disease therapy. A systematic search of peer-reviewed studies published in electronic databases was performed, focusing on preclinical and clinical research investigating the use of LNPs, EVs and liposomes for hepatic drug or gene delivery. Studies were analyzed for vesicle composition, targeting efficiency, payload capacity, therapeutic outcomes, and reported limitations. The analysis indicates that LNPs demonstrate strong efficiency in nucleic acid encapsulation and delivery, supported by growing clinical translation. EVs show promising biocompatibility and innate targeting to hepatic cells but face challenges in large-scale production and standardization. Liposomes remain versatile and well-characterized platforms capable of carrying diverse therapeutic molecules, though rapid clearance can limit their efficacy. Together, these nanovesicle systems hold considerable potential for advancing targeted drug and gene therapies in liver disease. Future work should focus on improving stability, manufacturing scalability, and cell-specific targeting to support clinical translation. Full article
(This article belongs to the Section Omics/Informatics)
Show Figures

Graphical abstract

22 pages, 5738 KB  
Review
Probing Membrane Structure of Lipid Nanomedicines Using Solution Small-Angle X-Ray Scattering: Applications and Prospects
by Ke-Meng Li, Panqi Song, Xiao-Peng He and Na Li
Membranes 2025, 15(12), 382; https://doi.org/10.3390/membranes15120382 - 16 Dec 2025
Viewed by 810
Abstract
Lipid-based nanomedicines are already widely used in antitumor therapy and gene delivery. However, their complex structural features demand advanced mesoscopic structural characterization tools for effective research and development (R&D) and quality control. Synchrotron small-angle X-ray scattering (SAXS) is a powerful, non-invasive technique for [...] Read more.
Lipid-based nanomedicines are already widely used in antitumor therapy and gene delivery. However, their complex structural features demand advanced mesoscopic structural characterization tools for effective research and development (R&D) and quality control. Synchrotron small-angle X-ray scattering (SAXS) is a powerful, non-invasive technique for probing nanoscale membrane organizations, monitoring in situ dynamic membrane assembly, and exploring the interactions of components in lipid-based drug delivery systems, including liposomes, lipoplexes, lipid nanoparticles (LNPs), and lyotropic liquid crystals (LLCs). Recent advances in high-flux synchrotron facilities, high-frequency detectors, and automated SAXS data processing pipelines permit a detailed structural characterization of lamellarity, bilayer spacing, internal phases, core–shell morphology, as well as “pump-probe” dynamic process studies for lipid nanomedicines. Though major challenges remain in sample polydispersity and model fitting, the advances in time-resolved synchrotron SAXS, high-throughput automation, and artificial intelligence (AI)-assisted modeling are rapidly reducing this barrier. This review summarizes SAXS methodology and introduces representative case studies in the field of lipid nanomedicines. The performance of BioSAXS beamline BL19U2 in the Shanghai synchrotron radiation facility (SSRF) and prospects of AI-guided drug screening at BL19U2 are highlighted to advance intelligent R&D and quality control for lipid nanomedicines. Full article
Show Figures

Graphical abstract

24 pages, 7331 KB  
Article
Coarse-Grained Molecular Dynamics Simulations of Lipid Nanodroplets and Endosomal Membranes: Focusing on the Fusion Mechanisms
by Yeon Ju Go, Erkhembayar Jadamba and Hyunjin Shin
Int. J. Mol. Sci. 2025, 26(24), 11960; https://doi.org/10.3390/ijms262411960 - 11 Dec 2025
Viewed by 1106
Abstract
Lipid nanoparticles (LNPs) have received significant attention as effective RNA carriers in RNA-based therapeutics and vaccines. Particularly, ionizable lipids (ILs) of LNPs play a crucial role in endosomal escape and lipid-mediated RNA delivery owing to their pH-dependent molecular characteristics. Therefore, it is essential [...] Read more.
Lipid nanoparticles (LNPs) have received significant attention as effective RNA carriers in RNA-based therapeutics and vaccines. Particularly, ionizable lipids (ILs) of LNPs play a crucial role in endosomal escape and lipid-mediated RNA delivery owing to their pH-dependent molecular characteristics. Therefore, it is essential to enhance the endosomal escape efficiency of ILs, which is primary bottleneck in the successful cytoplasmic delivery of RNA. However, the molecular-level understanding of the roles and dynamics of ILs during the endosomal escape stage remains unclear. To elucidate this, we utilized coarse-grained (CG) molecular dynamics (MD) simulations. In this simulation, we designed lipid nanodroplets (LNDs) containing D-Lin-MC3-DMA (MC3) and ALC-0315, which have proven effective as LNPs in RNA-based therapeutics and vaccines, respectively, while accounting for the pH environments of early and late endosomes. Also, we formulated lipid bilayers reflecting the composition of early and late endosomal membranes to investigate the fusion process between LNDs and endosomal membranes. Our findings reveal that, irrespective of endosomal membrane composition and LNP types, ILs are the first lipids to enter the endosomal membrane during the fusion, and the flip-flop process of ILs from the inner leaflet to the outer leaflet of the endosomal membrane is a critical step for LNP endosomal escape. More specifically, we observed that protonated ILs predominantly participate in the flip-flop process, while many deprotonated ILs remain clustered and disordered within the intermediate layer of the endosomal membrane. Furthermore, we found that the extent of IL flip-flop varies with the cholesterol content of the endosomal membrane. Additionally, under identical pH conditions, MC3-containing LNDs exhibited a more active IL flip-flop process toward the outer leaflet than ALC-0315-containing LNDs. This observation supports experimental findings that MC3-containing LNPs manifest higher endosomal escape efficiency than ALC-0315-containing LNPs in mRNA delivery studies. The mechanistic insights into the endosomal escape mechanism demonstrated by our simulations could aid in the development of effective ILs. Full article
Show Figures

Figure 1

17 pages, 4997 KB  
Article
Construction of Spleen-Accumulated Polysorbate 20-Containing Ionizable Lipid Nanoparticles for mRNA Delivery
by Hanyu Liu, Siqi Li, Kexin Chen, Shuyi Yao, Xuefeng Tang and Xiaojun Han
Nanomaterials 2025, 15(24), 1844; https://doi.org/10.3390/nano15241844 - 8 Dec 2025
Viewed by 503
Abstract
Messenger RNA therapy represents a transformative therapeutic in vaccine development, tumor immunotherapy, and genetic disease intervention. Polyethylene glycol (PEG) lipid, a key component of ionizable lipid nanoparticles (iLNPs) for mRNA delivery, and the PEG antibodies induced by PEG are associated with hypersensitivity and [...] Read more.
Messenger RNA therapy represents a transformative therapeutic in vaccine development, tumor immunotherapy, and genetic disease intervention. Polyethylene glycol (PEG) lipid, a key component of ionizable lipid nanoparticles (iLNPs) for mRNA delivery, and the PEG antibodies induced by PEG are associated with hypersensitivity and accelerated blood clearance. To address the above PEG-associated challenges, we systematically investigated polysorbate 20 (PS20) as an alternative for constructing PEG-free iLNPs. The formulation of PS20-incorporated iLNPs (PS20-iLNPs) for carrying mRNA was systematically investigated by analyzing the particle size, pKa, endosomal escape efficiency, and cellular internalization efficiency of iLNPs with different molar ratio PS20 content (0.5–5.0%). iLNPs with a relative molar ratio of 2.5% were identified as the optimal mRNA delivery carrier. This carrier exhibited excellent resistance to serum protein adsorption capacity, with serum stability 1.3-fold higher than PEG-iLNPs. At high lipid concentrations (2.7 mg/mL), the cell viability of PS20-iLNPs was maintained at 91.1%, which was 1.07-fold higher than PEG-iLNPs. Under serum interference, PS20-iLNPs achieved a transfection efficiency of 46.5%, marking a 10.6% improvement over PEG-iLNPs under identical conditions. Notably, PS20-iLNPs exhibited 24 times higher spleen accumulation than PEG-iLNPs. These findings highlight PS20 as a viable PEG substitute for developing PEG-free spleen-accumulated mRNA delivery platforms with enhanced therapeutic potential. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

16 pages, 1805 KB  
Article
MERS-CoV RBD-mRNA Presents Better Immunogenicity and Protection than the Spike-mRNA
by Qian Liu, Abhishek K. Verma, Xiaoqing Guan, Shengnan Qian, Stanley Perlman and Lanying Du
Cells 2025, 14(23), 1928; https://doi.org/10.3390/cells14231928 - 4 Dec 2025
Viewed by 636
Abstract
Pathogenic Middle East respiratory syndrome CoV (MERS-CoV), first identified in Saudi Arabia in 2012, continues to pose a threat to public health. The trimeric spike (S) protein of MERS-CoV binds to the cellular receptor through the receptor-binding domain (RBD) in the S1 subunit [...] Read more.
Pathogenic Middle East respiratory syndrome CoV (MERS-CoV), first identified in Saudi Arabia in 2012, continues to pose a threat to public health. The trimeric spike (S) protein of MERS-CoV binds to the cellular receptor through the receptor-binding domain (RBD) in the S1 subunit to initiate virus entry and infection. Therefore, both the S protein and its RBD are targets for the development of MERS-CoV vaccines. Nevertheless, a direct comparison of the immune efficiency of S- and RBD-based MERS-CoV vaccines has not been made. Here, we compared two mRNA vaccines, respectively, targeting the S (S-mRNA) and RBD (RBD-mRNA) of MERS-CoV for their durable immunogenicity, neutralizing activity, and protective efficacy in a mouse model. Both mRNAs encapsulated with lipid nanoparticles (LNPs) maintained strong stability at various temperatures during the detection period. LNP-encapsulated RBD-mRNA elicited significantly higher and more durable antibodies than LNP-encapsulated S-mRNA, maintaining stronger and broadly neutralizing activity against the MERS-CoV original strain, as well as multiple variants containing key mutations within the RBD region. Importantly, RBD-mRNA provided durable protective efficacy against MERS-CoV infection in middle-aged mice, and this protection was associated positively with serum neutralizing antibody titers. Overall, this study identifies RBD-mRNA as an effective vaccine against MERS-CoV, with great potential for further development. Full article
Show Figures

Figure 1

Back to TopTop