1
|
Zhang J, Zeng F, Li Y, Mu C, Liu C, Wang L, Peng X, He L, Su Y, Li H, Wang A, Feng L, Gao D, Zhang Z, Xu G, Wang Y, Yue R, Si J, Zheng L, Zhang X, He F, Yi H, Tang Z, Li G, Ma K, Li Q. The characterization of technical design of a virus-like structure (VLS) nanodelivery system as vaccine candidate against SARS-CoV-2 variants. Hum Vaccin Immunother 2025; 21:2473183. [PMID: 40045463 PMCID: PMC11901403 DOI: 10.1080/21645515.2025.2473183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The constant mutation of SARS-CoV-2 has led to the continuous appearance of viral variants and their pandemics and has improved the development of vaccines with a broad spectrum of antigens to curb the spread of the virus. The work described here suggested a novel vaccine with a virus-like structure (VLS) composed of combined mRNA and protein that is capable of stimulating the immune system in a manner similar to that of viral infection. This VLS vaccine is characterized by its ability to specifically target dendritic cells and/or macrophages through S1 protein recognition of the DC-SIGN receptor in cells, which leads to direct mRNA delivery to these innate immune cells for activation of robust immunity with a broad spectrum of neutralizing antibodies and immune protective capacity against variants. Research on its composition characteristics and structural features has suggested its druggability. Compared with the current mRNA vaccine, the VLS vaccine was identified as having no cytotoxicity at its effective application dosage, while the results of safety observations in animals revealed fewer adverse reactions during immunization.
Collapse
MESH Headings
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Animals
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Neutralizing/immunology
- Humans
- Antibodies, Viral/immunology
- Mice
- Dendritic Cells/immunology
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/genetics
- Lectins, C-Type/immunology
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- mRNA Vaccines
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Macrophages/immunology
- Mice, Inbred BALB C
- Female
- Cell Adhesion Molecules
Collapse
Affiliation(s)
- Jingjing Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Fengyuan Zeng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanmei Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Changyong Mu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Change Liu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiaowu Peng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Liping He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanrui Su
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongbing Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - An Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lin Feng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Dongxiu Gao
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhixiao Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gang Xu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yixuan Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Rong Yue
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Junbo Si
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Zheng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiong Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Fuyun He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongkun Yi
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhongshu Tang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gaocan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Kaili Ma
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Qihan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| |
Collapse
|
2
|
Zhang T, Jian Z, Chen J, Xu D, Yang X, Lu Y, Yan S, Pan L, Wu Q, Ouyang Z. Efficiency enhancement in main path extraction in mRNA vaccine field: A novel approach leveraging intermediate patents, with shielding origin and terminus patent edges. Hum Vaccin Immunother 2025; 21:2454078. [PMID: 39838876 DOI: 10.1080/21645515.2025.2454078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
mRNA vaccines offer groundbreaking technological advantages and broad application potential. Their rapid advancement, particularly during the COVID-19 pandemic, is the result of decades of research and numerous technological breakthroughs. These discoveries build upon each other, forming dense, interconnected networks of progress. Studying the technological development paths of mRNA vaccines is therefore essential. Main path analysis (MPA) is particularly effective for mapping out development trajectories within complex and interconnected networks, which serves as a powerful tool for identifying key nodes and innovations. This study introduces a novel approach to extracting main paths from a patent citation network in the mRNA vaccine field. Initially, we shielded edges connecting the origin and terminus patents. Subsequently, we extracted the main paths from intermediate patents, and then, we reintegrated the edges connecting the origin and terminus patents based on the citation relationships, resulting in a comprehensive extraction of the main paths. The research findings indicate a consistency among the global main paths, global key-route main paths, local forward main paths, and local key-route main paths within the mRNA vaccine field. The patents on the main paths predominantly focus on nucleic acid modifications and delivery systems. The local backward main paths identify a greater number of patents, especially those related to litigation, offering a richer and more diverse set of technological insights. This study significantly advances the methodology of MPA, with the innovative shielding technique offering a fresh perspective for navigating complex networks and providing a deeper understanding of technological development in the mRNA vaccine domain.
Collapse
Affiliation(s)
- Ting Zhang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhongquan Jian
- Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian, China
| | - Juan Chen
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dongzi Xu
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyi Yang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Lu
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shu Yan
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lizi Pan
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Zhaolian Ouyang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Liu J, Sun J, Hu J, Xue H, Lei L, Pan X. Biomaterial-based drug delivery strategies for oral mucosa. Colloids Surf B Biointerfaces 2025; 251:114604. [PMID: 40081256 DOI: 10.1016/j.colsurfb.2025.114604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Drug therapy is a key field in modern medicine, and the optimization of its delivery method is crucial. Traditional methods are inherently limited by first-pass effects, high-risk adverse reactions, and patient compliance challenges, which significantly restrict the effectiveness and application potential of drugs. Oral mucosal drug delivery has become a minimally invasive and effective drug delivery strategy. The unique anatomical structure of the oral mucosa facilitates the rapid absorption of drugs into the systemic circulation, thus producing rapid therapeutic effects. However, a complex oral microenvironment and mucosal barrier impede drug absorption. Biomaterials have become an important driving force for the innovative development of oral medicine, owing to their unique and excellent properties. They are widely used for preventing, diagnosing, treating, and rehabilitating oral diseases. This review explores recent advancements in biomaterial-enabled oral mucosal drug delivery systems, analyzing key physiological factors and absorption barriers that impact therapeutic outcomes. Focusing on innovative material engineering strategies highlights significant progress in extending drug residence time and improving delivery precision within the oral cavity. Furthermore, the study identifies critical challenges in translating these advancements from research to clinical practice, emphasizing the need for solutions to bridge this gap.
Collapse
Affiliation(s)
- Junhui Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jiao Sun
- Changsha Stomatological Hospital, Changsha 410000, China
| | - Jun Hu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Huaqian Xue
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Xiaoyi Pan
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| |
Collapse
|
4
|
Senapati S, Bertolini TB, Minnier MA, Yazicioglu MN, Markusic DM, Zhang R, Wicks J, Nahvi A, Herzog RW, Walsh MC, Cejas PJ, Armour SM. Inhibition of IFNAR-JAK signaling enhances tolerability and transgene expression of systemic non-viral DNA delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102502. [PMID: 40206655 PMCID: PMC11979999 DOI: 10.1016/j.omtn.2025.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Lipid nanoparticles (LNPs) have demonstrated significant therapeutic value for non-viral delivery of mRNA and siRNA. While there is considerable interest in utilizing LNPs for delivering DNA (DNA-LNPs) to address a broad range of genetic disorders, acute inflammatory responses pose significant safety concerns and limit transgene expression below therapeutically relevant levels. However, the mechanisms and immune signaling pathways underlying DNA-LNP-triggered inflammatory responses are not well characterized. Through the use of gene-targeted mouse models, we have identified cGAS-STING and interferon-α/β receptor (IFNAR) pathways as major mediators of acute inflammation triggered by systemic delivery of DNA-LNPs. cGAS-STING activation induces expression of numerous JAK-STAT-activating cytokines, and we show that treatment of mice with the JAK inhibitors ruxolitinib or baricitinib significantly improves tolerability to systemically delivered DNA-LNPs. Furthermore, specific inhibition of IFNAR signaling enhances both DNA-LNP tolerability and transgene expression. Utilization of JAK inhibitors or IFNAR blockade represent promising strategies for enhancing the safety and efficacy of non-viral DNA delivery for gene therapy.
Collapse
Affiliation(s)
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - David M. Markusic
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rui Zhang
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Joan Wicks
- Gene Therapy Research, Spark Therapeutics, Philadelphia, PA, USA
| | - Ali Nahvi
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Pedro J. Cejas
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Sean M. Armour
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| |
Collapse
|
5
|
Han Z, Huang H, Li B, Zhao R, Wang Q, Liu H, Xue H, Zhou W, Li G. Engineering exosome membrane disguised thermal responsive system for targeted drug delivery and controlled release across the blood-brain barrier. Mater Today Bio 2025; 32:101656. [PMID: 40160247 PMCID: PMC11953974 DOI: 10.1016/j.mtbio.2025.101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/02/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
The blood-brain barrier (BBB) presents a significant challenge for the delivery of chemotherapy drugs to brain tumors, leading to ineffective drug concentrations at the tumor site and contributing to chemotherapy resistance. The hypoxic tumor microenvironment further complicates this process, ultimately resulting in poor patient prognosis. In this study, we developed a thermoresponsive nanocarrier system that incorporates (Ru)(Pt) bimetallic nanoparticles onto defective TiOx nanoparticles with abundant oxygen vacancies, generating composite Ru/Pt-TiOx nanoparticles with photothermal and photocatalytic properties. The Ru and Pt in the nanoparticles enhance the metal-carrier interactions, with Ru increasing both light absorption and photothermal conversion efficiency and Pt catalyzing the conversion of endogenous H2O2 in tumors to produce oxygen. The oxygen produced within the tumor microenvironment reduces HIF-1α, MDR1 and P-gp expression, thereby inhibiting efflux and allowing doxorubicin to accumulate inside the cells. DOX was incorporated into a phase change material and combined with multiple Ru/Pt-TiOx nanoparticles to form composite RPTiOx-DOX particles that can control the release of DOX under near-infrared irradiation. In an effort to overcome the blocking effect of the BBB, we wrapped the RPTiOx-DOX nanoparticles with Angiopep-2-functionalized macrophage exosome membranes. Furthermore, the changes in the internal environment promote macrophage phenotypic transformation (M2→M1) to some extent and further inhibit tumor growth via immunoregulation. In this work, a novel drug delivery system capable of traversing the BBB and exerting synergistic antitumor effects through photostimulated therapeutic agents is described, providing innovative insights for the development of stimulus-responsive composite nanoparticle drug formulations.
Collapse
Affiliation(s)
- Zhe Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| | - Haina Huang
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Ji'nan, 250022, China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| | - RongRong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| | - Qingtong Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| | - Hong Liu
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Ji'nan, 250022, China
- State Key Laboratory of Crystal Materials, Shandong University, Ji'nan, 250100, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| | - Weijia Zhou
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Ji'nan, 250022, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
- Institute of Brain and Brain-Inspired Science, Ji'nan, Shandong, 250012, China
- Shandong Key Laboratory of Brain Function Remodeling, Ji'nan, Shandong, 250012, China
| |
Collapse
|
6
|
Massoud EN, Hebert MK, Siddharthan A, Ferreira T, Neron A, Goodrow M, Ferreira T. Delivery vehicles for light-mediated drug delivery: microspheres, microbots, and nanoparticles: a review. J Drug Target 2025; 33:691-703. [PMID: 39714878 DOI: 10.1080/1061186x.2024.2446636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024]
Abstract
This review delves into the evolving landscape of mediated drug delivery, focusing on the versatility of a variety of drug delivery vehicles such as microspheres, microbots, and nanoparticles (NPs). The review also expounds on the critical components and mechanisms for light-mediated drug delivery, including photosensitizers and light sources such as visible light detectable by the human eye, ultraviolet (UV) light, shorter wavelengths than visible light, and near-infra-red (NIR) light, which has longer wavelength than visible light. This longer wavelength has been implemented in drug delivery for its ability to penetrate deeper tissues and highlighted for its role in precise and controlled drug release. Furthermore, this review discusses the significance of these drug delivery vehicles towards a spectrum of diverse applications spanning gene therapy, cancer treatment, diagnostics, and microsurgery, and the materials used in the fabrication of these vehicles encompassing polymers, ceramics, and lipids. Moreover, the review analyses the challenges and limitations of such drug delivery vehicles as areas of improvement to provide researchers with valuable insights for addressing current obstacles in the progression of drug delivery. Overall, this review underscores the potential of light-mediated drug delivery to revolutionise healthcare and personalised medicine, providing precise, targeted, and effective therapeutic interventions.
Collapse
Affiliation(s)
- Engi Nadia Massoud
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| | | | | | - Tyler Ferreira
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| | - Abid Neron
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| | - Mary Goodrow
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| | - Tracie Ferreira
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| |
Collapse
|
7
|
Wu Y, Park J, Xu E, Kim D, Lee J, Oh YK. MicroRNA-induced reprogramming of tumor-associated macrophages for modulation of tumor immune microenvironment. J Control Release 2025; 381:113593. [PMID: 40024340 DOI: 10.1016/j.jconrel.2025.113593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment and typically exhibit pro-tumoral phenotypes. TAMs overexpress the signal regulatory protein alpha (SIRPα) receptor on their surface, which interacts with CD47 on tumor cells to inhibit their phagocytic activity. In this study, we developed lipid nanoparticles modified with an anti-SIRPα antibody (aSIRPα) for the targeted delivery of microRNA-155 (miR155@aSIRPα-LNP) to TAMs, aiming to enhance their anti-tumoral phenotypes within the tumor microenvironment. The aSIRPα modification not only facilitated nanoparticle uptake by TAMs rather than B16F10 cells, but also blocked the anti-phagocytosis signal by disrupting the interaction between SIRPα and CD47 on cancer cells. This dual functionality enhanced the expression of anti-tumoral phenotype markers in TAMs and activated macrophage-mediated phagocytosis of tumor cells. In a melanoma model, intratumoral administration of miR155@aSIRPα-LNP to B16F10 tumor-bearing mice reprogrammed TAMs toward anti-tumoral phenotypes. The anti-tumoral cytokines released by these TAMs remodeled the immunosuppressive tumor microenvironment, increasing cytotoxic T cell infiltration and reducing the regulatory T cell population, inhibiting tumor progression. This approach indicates the potential of miRNA-based therapies to overcome the limitations of current immunotherapies in treating cold solid tumors. Overall, the results suggest that delivering miR155 to TAMs by targeting SIRPα is a promising strategy for modulating the immunosuppressive tumor microenvironment in cancer immunotherapy.
Collapse
Affiliation(s)
- Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Moosavi SG, Rahiman N, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in neurodegenerative diseases. J Control Release 2025; 381:113641. [PMID: 40120689 DOI: 10.1016/j.jconrel.2025.113641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases (NDD) are characterized by the progressive loss of neurons and the impairment of cellular functions. Messenger RNA (mRNA) has emerged as a promising therapy for treating NDD, as it can encode missing or dysfunctional proteins and anti-inflammatory cytokines or neuroprotective proteins to halt the progression of these diseases. However, effective mRNA delivery to the central nervous system (CNS) remains a significant challenge due to the limited penetration of the blood-brain barrier (BBB). Lipid nanoparticles (LNPs) offer an efficient solution by encapsulating and protecting mRNA, facilitating transfection and intracellular delivery. This review discusses the pathophysiological mechanisms of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), ischemic stroke, spinal cord injury, and Friedreich's ataxia. Additionally, it explores the potential of LNP-mediated mRNA delivery as a therapeutic strategy for these diseases. Various approaches to overcoming BBB-related challenges and enhancing the delivery and efficacy of mRNA-LNPs are discussed, including non-invasive methods with strong potential for clinical translation. With advancements in artificial intelligence (AI)-guided mRNA and LNP design, targeted delivery, gene editing, and CAR-T cell therapy, mRNA-LNPs could significantly transform the treatment landscape for NDD, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Seyedeh Ghazal Moosavi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Zhao F, Fan M, Jing Z, Zhang Y, Wang Y, Zhou C, Liu Y, Aitken RJ, Xia X. Engineered nanoparticles potentials in male reproduction. Andrology 2025; 13:694-705. [PMID: 39120563 PMCID: PMC12006894 DOI: 10.1111/andr.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The escalating prevalence of fertility problems in the aging population necessitates a comprehensive exploration of contributing factors, extending beyond environmental concerns, work-related stress, and unhealthy lifestyles. Among these, the rising incidence of testicular disorders emerges as a pivotal determinant of fertility issues. Current treatment challenges are underscored by the limitations of high-dose and frequent drug administration, coupled with substantial side effects and irreversible trauma inflicted by surgical interventions on testicular tissue. MATERIAL AND METHODS The formidable barrier posed by the blood-testis barrier compounds the complexities of treating testicular diseases, presenting a significant therapeutic obstacle. The advent of nanocarriers, with their distinctive attributes, holds promise in overcoming this impediment. These nanocarriers exhibit exceptional biocompatibility, and membrane penetration capabilities, and can strategically target the blood-testis barrier through surface ligand modification, thereby augmenting drug bioavailability and enhancing therapeutic efficacy. RESULTS AND DISCUSSION This review concentrates on the transformative potential of nanocarriers in the delivery of therapeutic agents to testicular tissue. By summarizing key applications, we illuminate the strides made in utilizing nanocarriers as a novel avenue to effectively treat testicular diseases. CONCLUSIONS Nanocarriers are critical in delivering therapeutic agents to testicular tissue.
Collapse
Affiliation(s)
- Feifei Zhao
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Mengyu Fan
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
| | - Zhiyang Jing
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
| | - Yanxu Zhang
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
| | - Yanlin Wang
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
| | - Congli Zhou
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
| | - Yang Liu
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
- Department of Radiotherapy and Translational Medicine CenterHuaihe Hospital of Henan University, Henan UniversityKaifengHenanChina
| | - Robert John Aitken
- School of Environmental and Life SciencesCollege of Engineering, Science and Environmental Science, University of NewcastleCallaghanAustralia
| | - Xue Xia
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life SciencesHenan UniversityKaifengHenanChina
- Department of Radiotherapy and Translational Medicine CenterHuaihe Hospital of Henan University, Henan UniversityKaifengHenanChina
| |
Collapse
|
10
|
Chauhan A, Salwa, Shedgaonkar GG, Kumar L, Karmakar A, Khajuria S, Raghavendra AP, Verma R. Antioxidant and anticancer activities of hesperetin and its novel formulations in KB cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5213-5236. [PMID: 39531045 DOI: 10.1007/s00210-024-03581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
This study aimed to formulate the hesperetin nanostructured lipid carriers (NLCs) containing oro-mucosal gel for its activity assessment on the KB cell line. NLCs were prepared with glyceryl monostearate, oleic acid, and lecithin using a modified constant-temperature emulsification technique. The particle size analysis, in vitro drug release studies, etc., of prepared NLCs were evaluated. The formulated gels were analyzed with respect to spreadability, extrudability, swelling index, texture analysis, etc. The particle size, polydispersity index, zeta potential, and drug entrapment of nanocarriers were recorded to be 221.733 ± 61.536 nm, 0.381 ± 0.091, - 51.433 ± 4.143 mV, and 89.29%, respectively. The optimized NLCs in 24 h released 87.14 ± 6.62% of the drug. The round shape of NLCs was noticed with scanning electron microscopy. The pH, spreadability, extrudability, swelling index, content uniformity, and drug release studies of hesperetin NLCs-containing gel (HNG) were found to be 6.81 ± 0.04, 2.49 ± 0.04 cm.mg/s, 539.04 ± 32.88 g/cm2, 4.27 ± 0.47, 107.98 ± 1.93%, and 90.17 ± 6.67% (in 48 h), respectively. The developed formulations showed promising in vitro anticancer and antioxidant activities. HNP results authorize that the formulation may be beneficial for the treatment of oral cancer.
Collapse
Affiliation(s)
- Arunima Chauhan
- Faculty of Dentistry, Melaka-Manipal Medical College, Manipal Academy of Higher Education, Manipal, India
- Faculty of Dentistry, Manipal University College Malaysia, Jalan Batu Hampar, Bukit Baru, Melaka, Malaysia
| | - Salwa
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Gayatri Gopal Shedgaonkar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India.
| | - Arka Karmakar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India
| | - Salil Khajuria
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India
| | | | - Ruchi Verma
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
11
|
Ma Y, Fung V, VanKeulen-Miller R, Tiwade PB, Narasipura EA, Gill NA, Fenton OS. A Metabolite Co-Delivery Strategy to Improve mRNA Lipid Nanoparticle Delivery. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40274610 DOI: 10.1021/acsami.4c22969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Lipid nanoparticles (LNPs) effectively protect mRNA and facilitate its entry into target cells for protein synthesis. Despite these successes, cellular entry alone may not be enough for optimal protein expression, as mRNA translation also depends on the availability of essential metabolites, including metabolic energy sources, coenzymes, and amino acids. Without adequate metabolites, mRNA translation may be less efficient, potentially leading to higher dosing requirements or poorer therapeutic outcomes for LNP therapies. To address this, we develop a metabolite co-delivery strategy by encapsulating essential metabolites within mRNA LNPs, hypothesizing that our approach can uniformly improve mRNA delivery. Instead of adding a fifth component to the organic phase, our strategy involves mixing the metabolite with the mRNA payload in the aqueous phase, while maintaining the molar ratio of the components in the organic phase during LNP formulation. We verify our approach in vitro and in vivo, highlighting the broad applicability of our strategy through mechanism and efficacy studies across multiple cell lines, and physiological conditions, such as normoxia (i.e., 21% oxygen), hypoxia (i.e., 1% oxygen), and in mice. Taken collectively, we anticipate that our metabolite co-delivery strategy may serve as a generalizable strategy to enhance in vitro and in vivo protein expression using mRNA LNPs, potentially offering broad applicability for the study and treatment of disease.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Palas B Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Nicole A Gill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
12
|
Le Ru EC, Böttger R, Andrews D, Baumhof P, Rakonjac JV, Laufersky G, Darby BL. Rapid and Accurate Quantification of RNA in Lipid Nanoparticles by Scatter-Free UV/Visible Spectroscopy. NANO LETTERS 2025; 25:6813-6819. [PMID: 40199736 DOI: 10.1021/acs.nanolett.5c01491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
UV/visible spectroscopy is the method of choice for RNA quantification, thanks to its simplicity and accuracy. However, it cannot be used to quantify RNA in lipid nanoparticles (LNPs), such as those used for drug delivery in mRNA vaccines, because of light scattering by LNPs. Alternative methods such as the RiboGreen fluorescence assay require much more sample preparation and lack reproducibility and accuracy. Here we propose and demonstrate an alternative approach using an integrating sphere setup to measure scatter-free absorption spectra. RNA spectra of RNA-loaded LNPs can then be directly measured, and the total RNA concentration can be deduced. The method shows very good linearity and precision (∼1.5%), and the accuracy is estimated to be ∼5% when applied to various mRNA-LNP formulations. This work paves the way for the routine characterization of payload concentration in LNP formulation research.
Collapse
Affiliation(s)
- Eric C Le Ru
- Marama Labs Limited, 32 Salamanca Road, Wellington, 6012, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Roland Böttger
- CureVac SE, Friedrich-Miescher-Str. 15, 72076 Tübingen, Germany
| | - Darren Andrews
- Marama Labs Limited, DCU Alpha, Old Glasnevin Road, Dublin, D11KXN4, Ireland
| | - Patrick Baumhof
- CureVac SE, Friedrich-Miescher-Str. 15, 72076 Tübingen, Germany
| | - Jelena V Rakonjac
- Marama Labs Limited, 32 Salamanca Road, Wellington, 6012, New Zealand
| | - Geoffry Laufersky
- Marama Labs Limited, 32 Salamanca Road, Wellington, 6012, New Zealand
| | - Brendan L Darby
- Marama Labs Limited, DCU Alpha, Old Glasnevin Road, Dublin, D11KXN4, Ireland
| |
Collapse
|
13
|
Tanaka H, Sato Y, Nakabayashi T, Tanaka A, Nishio K, Matsumoto C, Matsumaru A, Yamakawa T, Ishizaki K, Ueda K, Higashi K, Moribe K, Nakai Y, Tange K, Akita H. A Post-Encapsulation Method for the Preparation of mRNA-LNPs via the Nucleic Acid-Bridged Fusion of mRNA-Free LNPs. NANO LETTERS 2025; 25:6445-6453. [PMID: 40219988 DOI: 10.1021/acs.nanolett.4c06643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Lipid nanoparticles with encapsulated mRNA (mRNA-LNPs) have become key modalities for personalized medicines and RNA vaccines. Once the platform technology is established, the mRNA-LNPs could be applicable to a variety of protein-based therapeutic strategies. A post-encapsulation method, in which the mRNA solution is incubated with preformed mRNA-free LNPs to prepare the mRNA-LNPs, would accelerate the development of RNA-based therapeutics since even nonexperts could manufacture the mRNA-LNPs. In this study, we describe that the post-encapsulation of mRNA into mRNA-free LNPs is accompanied by "nucleic acid-bridged fusion" of them. The adsorption of mRNA onto mRNA-free LNPs via electrostatic interactions and the internalization of mRNA into the LNPs via particle-to-particle fusion are two steps that occur at different levels of pH. To complete post-encapsulation using only one-step mixing, the pH must be controlled within a limited region where both processes occur simultaneously. The size of the mRNA-free LNPs determines the effectiveness of mRNA loading.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita 565-0871 Osaka, Japan
| | - Yuka Sato
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Tomoya Nakabayashi
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
| | - Akari Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
| | - Kazuma Nishio
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
| | - Chika Matsumoto
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
| | - Atsuya Matsumaru
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
| | - Takuma Yamakawa
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Kota Ishizaki
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Keisuke Ueda
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Kenjirou Higashi
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Kunikazu Moribe
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba city, Chiba 260-0856, Japan
| | - Yuta Nakai
- Life Science Research Laboratory, NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki city, Kanagawa 210-0865, Japan
| | - Kota Tange
- Life Science Research Laboratory, NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki city, Kanagawa 210-0865, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai city, Miyagi 980-8578, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita 565-0871 Osaka, Japan
| |
Collapse
|
14
|
Zhou Y, Wu Y, Sun S, Wang W, Zhou S, Liu H, Guo Y, Hong S, Ding F, Cai H. Self-Assembled Glycopeptide as a Biocompatible mRNA Vaccine Platform Elicits Robust Antitumor Immunity. ACS NANO 2025; 19:14727-14741. [PMID: 40203215 DOI: 10.1021/acsnano.4c15187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Since the emergence of the COVID-19 pandemic, mRNA vaccines have garnered significant attention. Delivery systems affect the effectiveness of mRNA vaccines, yet there remains a scarcity of vectors that can achieve safe and efficient delivery of mRNA. We took advantage of self-assembled glycopeptides (SAPs) to develop a vector named Man-MPm, which was coupled with mannose and manganese ions to achieve lymph node targeting and STING pathway activation. The Man-MPm-based mRNA vaccine exhibited high biosafety across various administration routes, eliciting robust antigen-specific immune responses within lymph nodes. Due to the elevated antitumor immunity, Man-MPm significantly suppressed tumor growth and extended the survival period of mice in melanoma prevention and treatment models as well as in a colon cancer model. Our findings show that Man-MPm addresses the challenges to safety and effectiveness associated with mRNA delivery by incorporating a lymph node-targeting ligand and a STING pathway agonist onto highly biocompatible SAP, and Man-MPm holds great potential for developing mRNA tumor vaccines.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Wei Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Siai Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Hua Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Yajing Guo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| |
Collapse
|
15
|
Kim M, Choi R, Kim L, Kim YC, Noh I. Cell membrane nanoparticles in cancer therapy: From basic structure to surface functionalization. J Control Release 2025:113752. [PMID: 40254140 DOI: 10.1016/j.jconrel.2025.113752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cell membrane nanoparticles (CNPs) have recently garnered significant attention as effective drug-delivery vehicles. Beyond their simple function of encapsulating cargo within a lipid bilayer structure, the cell membrane is a complex entity derived from biological materials, presenting a variety of surface proteins and glycans. Notable features that enhance their effectiveness as delivery vehicles include the inhibition of protein corona formation in the plasma and the suppression of macrophage phagocytosis, both of which contribute to prolonged blood circulation. Furthermore, CNPs exhibit homotypic targeting effects toward their cells of origin, resulting in reduced side effects, and because they are not xenobiotics, the likelihood of nonspecific immune activation is also minimized. This review focuses on various applications of CNPs in cancer therapeutic studies, examining their structural evolution and surface engineering developments. We introduce studies that leverage the inherent functionality of cell membranes and recent research in functional CNPs synthesized through genetic or chemical engineering methods. Through this review, we aim to trace the progression of CNP research, explore potential directions for their use in biomedical applications, and assess the prospects for clinical trials.
Collapse
Affiliation(s)
- Munsik Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Rohbin Choi
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Lian Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea.
| | - Ilkoo Noh
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
16
|
Peña Á, Heredero J, Blandín B, Mata E, De Miguel D, Toro A, Alejo T, Casabona D, López A, Gallego-Lleyda A, Pérez-Herrán E, Martínez-Oliván J, Giménez-Warren J. Multicomponent thiolactone-based ionizable lipid screening platform for efficient and tunable mRNA delivery to the lungs. Commun Chem 2025; 8:116. [PMID: 40234552 PMCID: PMC12000586 DOI: 10.1038/s42004-025-01516-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/04/2025] [Indexed: 04/17/2025] Open
Abstract
Ionizable lipids are essential components of lipid nanoparticles (LNPs) for efficient mRNA delivery. However, designing them for high protein expression, endosomal escape, and organ targeting is challenging due to complex structure-activity relationships. Here, we present a high-throughput platform for screening ionizable lipids using a two-step, scalable, one-pot reaction. This enabled the synthesis and vivo screening of 91 new lipids, followed by a structure-activity study, leading to the development of CP-LC-0729, which significantly surpasses the MC3 benchmark in protein expression with preliminary studies showing no in vivo toxicity. Additionally, a one-step strategy helped to yield a permanently cationic lipid which was tested in a fifth-lipid formulation, showing a highly selective lung delivery with a 32-fold increase in protein expression in vivo, outperforming current endogenous targeting strategies. All these findings underscore the potential of lipid CP-LC-0729 and our lipid platform in advancing the efficiency and specificity of mRNA delivery systems.
Collapse
Affiliation(s)
- Álvaro Peña
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Juan Heredero
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Beatriz Blandín
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Elena Mata
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Diego De Miguel
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Alfonso Toro
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Teresa Alejo
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Diego Casabona
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Alexandre López
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Ana Gallego-Lleyda
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Esther Pérez-Herrán
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Juan Martínez-Oliván
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain.
| | | |
Collapse
|
17
|
Tan Z, Zheng L, Bo Y, Kambar N, Wang H, Leal C. Click Lipid Nanoparticles for the Delivery of mRNA to Metabolically Labeled Cancer Cells. Biochemistry 2025; 64:1807-1816. [PMID: 40181500 DOI: 10.1021/acs.biochem.4c00699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Lipid nanoparticle (LNP)-based mRNA delivery has a lot of potential in combating a wide range of diseases, but delivering mRNA to specific cell types continues to be challenging. Despite recent advances in organ and cell specificity, the majority of clinical LNP systems cannot fully release their payload to a targeted site. Incorporating active targeting moieties into LNPs is highly desired to expand nanomedicine applications. In this Letter, we developed LNPs that harness the power of bioorthogonal "click" azide-alkyne chemical reactions. We show that the plasma membranes of cancer cells can be labeled with azide groups by metabolic sugar labeling, and these azide groups can react with dibenzocyclooctyne (DBCO) on LNPs to achieve specific binding. To achieve this, we synthesized new and versatile lipids by functionalizing DBCO groups to phospholipids with or without a poly(ethylene glycol) (PEG) linker. The DBCO lipids were successfully formulated into DBCO-LNPs comprising other standard lipid compounds. When using these DBCO-LNPs to deliver mRNA to metabolically labeled cells, DBCO-LNPs showed a remarkable ability to preferentially deliver mRNA to azide-labeled cells. Removing PEG linkers from DBCO lipids enables better integration and retention in the LNP, and the higher the amount of DBCO lipid, the stronger the targeting effect. This work demonstrates that cell-specific targeting can be achieved utilizing azide-alkyne ″click″ chemistry and could inspire the development of the next generation of LNPs for active cyto-tropic nanomedicines.
Collapse
Affiliation(s)
- Zhengzhong Tan
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Lining Zheng
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yang Bo
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nurila Kambar
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hua Wang
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Cecilia Leal
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
18
|
Lim D, Song M, Kim M, Park HK, Kim DW, Pang C. Bioinspired Suction-Driven Strategies with Nanoscale Skin-Controllable Adhesive Architectures for Efficient Liquid Formulated Transdermal Patches. ACS NANO 2025; 19:13567-13590. [PMID: 40170569 DOI: 10.1021/acsnano.5c00585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
For highly efficient and precise drug release, transdermal drug delivery systems (TDDS) have recently evolved through the combination of intelligent material-based structures with various active components. These strategies are an effort to overcome the significant difficulties in delivering large molecule drugs and nanomaterials due to the physical barrier of the skin, especially the stratum corneum, in traditional TDDS. Interestingly, multiscale suction-driven architectures (SDAs) inspired by bioinspired suction adhesion mechanisms have provided innovative solutions to these challenges. These architectures employ negative pressure to enhance nanoscale skin-controllable skin adhesion, temporarily bypass the skin barrier, and facilitate deep penetration of therapeutic agents, thereby, achieving the goals of increasing drug delivery efficiency and maximizing user convenience as a minimal invasive, needle-free platform. This review provides a comprehensive overview of suction-driven transdermal patches and emphasizes their integration with multifunctional materials to achieve stable adhesion and controlled drug release. Next, we present cost-effective and user-friendly suction-driven drug delivery patch devices through optimization of cupping structures without the incorporation of additional devices. Furthermore, we present cost-effective and user-friendly transdermal drug delivery patch devices that optimize multiscale cupping architectures without the need for additional devices. Potential of bioinspired SDAs in localized and systemic drug delivery through challenging and complex skin, as well as future perspectives, are discussed, along with innovative directions for more efficient and patient-centric transdermal drug delivery solutions.
Collapse
Affiliation(s)
- Dohyun Lim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minwoo Song
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minjin Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Hyoung-Ki Park
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Mimetics Co., Ltd, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Da Wan Kim
- Department of Electronic Engineering, Korea National University of Transportation, Chungju-si, Chungbuk 27469, Republic of Korea
| | - Changhyun Pang
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
19
|
Guo SH, Yu XK, Zhu YL, Zhang LL, Huang YN. Controlled release mechanism of drugs from onion-like dendrimersomes: insight from dissipative particle dynamics simulations. Phys Chem Chem Phys 2025. [PMID: 40227623 DOI: 10.1039/d4cp04780j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Compared with current lipid nanoparticle delivery systems, a new drug delivery system that can simultaneously achieve high stability toward temperature and time, and controllable release of drugs will be smart and next-generation. However, designing such systems for the complex human body environment remains a daunting challenge. Herein, we use highly stable multilayer dendrimersomes as a model to study the mechanism of controlled release of drugs through stimulus-response by dissipative particle dynamics simulations. The results show that when the dendrimersomes remain intact, the release of encapsulated hydrophilic, hydrophobic, and neutral drugs is minimal. Once the amphiphilic dendrimers in the dendrimersomes are decomposed beyond a threshold by cleaving the linkers connecting hydrophobic and hydrophilic segments, which can be achieved by exogenous perturbations, a significant or complete release of the drugs occurs. The introduction of liquid flow will remarkably enhance the release capability of drugs in decomposed dendrimersomes. These insights into the controlled release of drugs at the microscopic level offer helpful guidance for the development of advanced drug delivery vehicles.
Collapse
Affiliation(s)
- Sheng-Hong Guo
- Xinjiang Laboratory of Phase Transitions and Microstructures in Condensed Matter Physics, College of Physical Science and Technology, Yili Normal University, Yining, Xinjiang 835000, China.
| | - Xiang-Kun Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Chang-chun, 130012, China.
| | - You-Liang Zhu
- Xinjiang Laboratory of Phase Transitions and Microstructures in Condensed Matter Physics, College of Physical Science and Technology, Yili Normal University, Yining, Xinjiang 835000, China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Chang-chun, 130012, China.
| | - Li-Li Zhang
- Xinjiang Laboratory of Phase Transitions and Microstructures in Condensed Matter Physics, College of Physical Science and Technology, Yili Normal University, Yining, Xinjiang 835000, China.
| | - Yi-Neng Huang
- Xinjiang Laboratory of Phase Transitions and Microstructures in Condensed Matter Physics, College of Physical Science and Technology, Yili Normal University, Yining, Xinjiang 835000, China.
- School of Physics, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing 210093, China
| |
Collapse
|
20
|
Nagaraj K, Kamalesu S. State-of-the-art surfactants as biomedical game changers: unlocking their potential in drug delivery, diagnostics, and tissue engineering. Int J Pharm 2025; 676:125590. [PMID: 40228615 DOI: 10.1016/j.ijpharm.2025.125590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
This review presents a comprehensive analysis of surfactant-based medicinal formulations, highlighting both their advantages and disadvantages. Surfactants enhance drug solubility, enhance targeted delivery, and facilitate controlled release of drugs. Their antimicrobial action is a result of their ability to disrupt microbial membranes, and their application in the delivery of genes and proteins involves stabilizing lipid nanoparticles for messenger ribonucleic acid (mRNA) vaccines and clustered regularly interspaced short palindromic repeats (CRISPR). Surfactants also assist in biomedical imaging and theranostics by enhancing magnetic resonance imaging (MRI) contrast, fluorescence bioimaging, and cancer diagnosis. In tissue engineering, they assist in the manufacturing of scaffolds and coatings of biomaterials. In spite of their broad application, cytotoxicity concerns, environmental impact, and regulatory constraints bar clinical use. Biodegradable biosurfactants, stimuli-responsive intelligent surfactants, and AI-driven formulation design are areas that future studies can focus on to enhance safety and effectiveness in current healthcare applications.
Collapse
Affiliation(s)
- Karuppiah Nagaraj
- Biomedical & Nano-Drug Formulation Laboratory, Department of General Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Nagar, Thandalam, Kanchipuram - Chennai Rd, Chennai 602105 Tamil Nadu, India.
| | - S Kamalesu
- Division of Chemistry, School of Science, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Tiruchirappalli 621105 Tamil Nadu, India
| |
Collapse
|
21
|
Tao J, Ning W, Lu W, Wang R, Zhou H, Zhang H, Xu J, Wang S, Teng Z, Wang L. Smart self-transforming nano-systems for overcoming biological barrier and enhancing tumor treatment efficacy. J Control Release 2025; 380:85-107. [PMID: 39880041 DOI: 10.1016/j.jconrel.2025.01.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Nanomedicines need to overcome multiple biological barriers in the body to reach the target area. However, traditional nanomedicines with constant physicochemical properties are not sufficient to meet the diverse and sometimes conflicting requirements during in vivo transport, making it difficult to penetrate various biological barriers, resulting in suboptimal drug delivery efficiency. Smart self-transforming nano-systems (SSTNs), capable of altering their own physicochemical properties (including size, charge, hydrophobicity, stiffness, morphology, etc.) under different physiological conditions, hold the potential to break through multiple biological barriers, thereby improving drug delivery efficiency and the efficacy of cancer treatment. In this review, we first summarize the design strategies of five most popular SSTNs (such as size-, charge-, hydrophilicity-, stiffness-, and morphology-self-transforming nano-systems), and then delve into their biomedical applications in enhancing circulation time, tissue penetration, and cellular uptake. Finally, we discuss the opportunities and challenges that SSTNs face in the future for cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Jun Tao
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Weiqing Ning
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Wei Lu
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Rui Wang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Hui Zhou
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Hongru Zhang
- Key Laboratory of Agricultural Product Processing and Quality Control, Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, PR China
| | - Jiayi Xu
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China
| | - Shouju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 211166, PR China.
| | - Zhaogang Teng
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China.
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, PR China.
| |
Collapse
|
22
|
Zhang L, Seow BYL, Bae KH, Zhang Y, Liao KC, Wan Y, Yang YY. Role of PEGylated lipid in lipid nanoparticle formulation for in vitro and in vivo delivery of mRNA vaccines. J Control Release 2025; 380:108-124. [PMID: 39875076 DOI: 10.1016/j.jconrel.2025.01.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
mRNA-loaded lipid nanoparticles (mRNA-LNPs) hold great potential for disease treatment and prevention. LNPs are normally made from four lipids including ionizable lipid, helper lipid, cholesterol, and PEGylated lipid (PEG-lipid). Although PEG-lipid has the lowest content, it plays a crucial role in the effective delivery of mRNA-LNPs. However, previous studies have yet to elucidate the key factors of PEG-lipid that influence the properties of LNPs. This study reported how PEG-lipid content, lipid tail length, and chemical linkage between PEG and lipid affected in vitro and in vivo properties of mRNA-LNPs. Forty-eight LNP formulations were prepared and characterized. The results revealed that a PEG-lipid molar content exceeding 3.0 % significantly reduced the encapsulation efficiency of mRNA in LNPs via manual mixing. An increased PEG-lipid content also significantly decreased mRNA translation efficiency. Although the chemical linkage had minimal impact, the lipid tail length of PEG-lipid significantly affected the properties of mRNA-LNPs, irrespective of whether the LNPs were prepared using manual or microfluidic mixing. mRNA-LNPs made from ALC-0159 with C14 lipid tails, which is used in Pfizer/BioNTech COVID-19 mRNA vaccines, or C16-Ceramide-PEG preferably accumulated in the liver, while mRNA-LNPs prepared from C8-Ceramide-PEG were largely found in the lymph nodes. In a mouse SARS-CoV-2 Delta variant spike protein-encoded mRNA vaccine model, mRNA-LNPs made from either C8-Ceramide-PEG or C16-Ceramide-PEG yielded comparable vaccination efficacy to mRNA-LNPs made from ALC-0159, while mRNA-LNPs formulated with DSPE-PEG with C18 lipid tails mediated lower vaccination efficacy. C16-Ceramide-PEG LNPs and DSPE-PEG LNPs induced higher anti-PEG antibody response than C8-Ceramide-PEG and ALC-0159 LNPs. All the LNPs tested did not cause significant toxicity in mice. These results offer valuable insights into the use of PEG-lipid in LNP formulations and suggest that C8-Ceramide-PEG holds potential for use in the formulation of mRNA vaccine-loaded LNPs.
Collapse
Affiliation(s)
- Li Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Brandon Yi Loong Seow
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Yue Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore
| | - Kuo-Chieh Liao
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Yue Wan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Republic of Singapore.
| |
Collapse
|
23
|
Wu H, Chen Z, Jiang Z, Chen M, Shan H, Lin Q, Sun Q, Lyu C, Ye K, Chen Z, Tan W. Ultrasonic Hollow Microneedle Array (USHM) for Androgenetic Alopecia Treatment through Modulating the Expression of Hair-Growth-Associated Genes. ACS APPLIED MATERIALS & INTERFACES 2025; 17:21419-21430. [PMID: 40145651 DOI: 10.1021/acsami.5c00379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
The application of finasteride through the skin to treat androgenetic alopecia shows promise, as it avoids side effects on the prostate and sexual function. However, the effectiveness has been limited due to the structure of the skin. In this study, we developed a drug delivery system called an ultrasound hollow microneedle array (USHM) to enhance the transdermal delivery of finasteride for the treatment of androgenetic alopecia. This technique successfully used ultrasonic cavitation effects in the skin to improve the delivery and therapeutic effects of finasteride. Hair regeneration experiments in mice showed that USHM accelerated hair regrowth by one to two treatment cycles compared to microneedle delivery and by three cycles compared to topical application. Analysis based on RNA sequencing showed that USHM-mediated finasteride delivery upregulated genes related to keratin filaments, intermediate filaments, and the intermediate filament cytoskeleton, enhancing hair regeneration. This study expands the potential of finasteride treatment for androgenetic alopecia and offers an effective strategy for transdermal drug delivery.
Collapse
Affiliation(s)
- Huayi Wu
- Department of Plastic and Aesthetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Ziyan Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zixi Jiang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Maike Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Han Shan
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Qibo Lin
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Qi Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Changling Lyu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Kai Ye
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zeyu Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Wuyuan Tan
- Department of Plastic and Aesthetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
24
|
Zaccariotto GDC, Bistaffa MJ, Zapata AMM, Rodero C, Coelho F, Quitiba JVB, Lima L, Sterman R, Cardoso VMDO, Zucolotto V. Cancer Nanovaccines: Mechanisms, Design Principles, and Clinical Translation. ACS NANO 2025. [PMID: 40202241 DOI: 10.1021/acsnano.4c15765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Cancer immunotherapy has transformed the landscape of oncological treatment by employing various strategies to teach the immune system to eliminate tumors. Among these, cancer nanovaccines are an emerging strategy that utilizes nanotechnology to enhance immune activation in response to tumor antigens. This review addresses the principles behind the different technologies in this field aimed at generating a robust and effective immune response. The diversity of strategies adopted for the design of nanovaccines is discussed, including the types of active agents, nanocarriers, their functionalizations, and the incorporation of adjuvants. Furthermore, strategies to optimize nanoparticle formulations to enhance the antigen presentation, target immune cells, and organs and promote strong and durable antitumor responses are explored. Finally, we analyze the current state of clinical application, highlighting ongoing clinical trials and the future potential of cancer nanovaccines. The insights presented in this review aim to guide future research and development efforts in the field, contributing to the advancement of more effective and targeted nanovaccines in the fight against cancer.
Collapse
Affiliation(s)
- Gabriel de Camargo Zaccariotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Angelica Maria Mazuera Zapata
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Camila Rodero
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Fernanda Coelho
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - João Victor Brandão Quitiba
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Lorena Lima
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Raquel Sterman
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | | | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| |
Collapse
|
25
|
Choi AS, Moon TJ, Bhalotia A, Rajan A, Ogunnaike L, Hutchinson DW, Hwang I, Gokhale A, Kim JN, Ma T, Karathanasis E. Lipid Nanoparticles and PEG: Time Frame of Immune Checkpoint Blockade Can Be Controlled by Adjusting the Rate of Cellular Uptake of Nanoparticles. Mol Pharm 2025; 22:1859-1868. [PMID: 40035231 PMCID: PMC11975481 DOI: 10.1021/acs.molpharmaceut.4c01039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The engineerability of lipid nanoparticles (LNPs) and their ability to deliver nucleic acids make LNPs attractive tools for cancer immunotherapy. LNP-based gene delivery can be employed for various approaches in cancer immunotherapy, including encoding tumor-associated antigens and silencing of negative immune checkpoint proteins. For example, LNPs carrying small interfering RNAs can offer several advantages, including sustained and durable inhibition of an immune checkpoint protein. Due to their tunable design, modifying the lipid composition of LNPs can regulate the rate of their uptake by immune cells and the rate of gene silencing. Controlling the kinetics of LNP uptake provides additional flexibility and strategies to generate appropriate immunomodulation in the tumor microenvironment. Here, we evaluated the effects of polyethylene glycol (PEG) content ranging from 0.5 to 6 mol % on the cellular uptake of LNPs by immune cells and gene silencing of PD-L1 after intratumoral administration. We evaluated the cellular uptake and PD-L1 blockade in vitro in cell studies and in vivo using the YUMM1.7 melanoma tumor model. Cell studies showed that the rate of cell uptake was inversely correlated to an increasing mol % of PEG in a linear relationship. In the in vivo studies, 0.5% PEG LNP initiated an immediate effect in the tumor with a significant decrease in the PD-L1 expression of immune cells observed within 24 h. In comparison, the gene silencing effect of 6% PEG LNP was delayed, with a significant decrease of PD-L1 expression in immune cell subsets being observed 72 h after administration. Notably, performance of the 6% PEG LNP at 72 h was comparable to that of the 0.5% PEG LNP at 24 h. Overall, this study suggests that PEG modifications and intratumoral administration of LNPs can be a promising strategy for an effective antitumor immune response.
Collapse
Affiliation(s)
- Andrew S Choi
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Taylor J Moon
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Anubhuti Bhalotia
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aarthi Rajan
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Laolu Ogunnaike
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Diarmuid W Hutchinson
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Inga Hwang
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aaditya Gokhale
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Justin N Kim
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Timothy Ma
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Efstathios Karathanasis
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
26
|
Telkoparan-Akillilar P, Chichiarelli S, Tucci P, Saso L. Integration of MicroRNAs with nanomedicine: tumor targeting and therapeutic approaches. Front Cell Dev Biol 2025; 13:1569101. [PMID: 40260417 PMCID: PMC12009947 DOI: 10.3389/fcell.2025.1569101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a pivotal role in the post-transcriptional regulation of gene expression. Over the past decade, they have emerged as key regulators in cancer progression, influencing different cellular processes such as proliferation, apoptosis, metastasis, and immune evasion. Their unique ability to target multiple genes simultaneously makes miRNAs highly attractive as potential therapeutic agents in oncology. However, several challenges have hindered their direct clinical application, most notably their inherent instability in biological fluids, rapid degradation by nucleases, and inefficient delivery to specific tumor sites. Additionally, off-target effects and the potential for toxicity further complicate the therapeutic use of miRNAs. Nanomedicine offers a promising solution to these challenges by enabling the development of advanced platforms for the stable, safe, and targeted delivery of miRNAs. Nanoparticle-based delivery systems, such as liposomes, polymeric nanoparticles, and inorganic nanocarriers, can protect miRNAs from degradation, improve their bioavailability, and allow for precise tumor targeting through passive or active targeting mechanisms. These nanocarriers can also be engineered to release miRNAs in response to specific stimuli within the tumor microenvironment, enhancing therapeutic efficacy while minimizing side effects. This review will explore the integration of miRNAs with nanotechnology, focusing on various nanoparticle formulations and their roles in enhancing miRNA stability, specificity, and function in cancer treatment. In addition, we will discuss current advances in preclinical and clinical applications, highlight promising tumor-targeting strategies, and address the remaining challenges such as toxicity, immunogenicity, and scalability. Future research should focus on overcoming these barriers, ultimately paving the way for the widespread adoption of personalized miRNA-based nanomedicine in cancer therapy.
Collapse
Affiliation(s)
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, Rome, Italy
| |
Collapse
|
27
|
Li M, Schroder R, Ozuguzel U, Corts TM, Liu Y, Zhao Y, Xu W, Ling J, Templeton AC, Chaudhuri B, Gindy M, Wagner A, Su Y. Molecular Insight into Lipid Nanoparticle Assembly from NMR Spectroscopy and Molecular Dynamics Simulation. Mol Pharm 2025; 22:2193-2212. [PMID: 40135901 DOI: 10.1021/acs.molpharmaceut.4c01437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as the premier drug delivery system for oligonucleotide vaccines and therapeutics in recent years. Despite their prosperous advancement in research and clinical applications, there is a significant lack of mechanistic understanding of the assembly of lipid particles at the molecular level. In our study, we utilized a combination of solution and solid-state NMR, together with molecular dynamics simulations, to elucidate local structures and interactions of chemical components across multiple motional regimes. Our results comprehensively evaluated the impact of formulation components and engineering process factors on the particle formation and identified the interplay of phospholipids (DSPC), poly(ethylene glycol) (PEG) lipid conjugates, and cholesterol in governing the particle size and lipid dynamics from a structural perspective, using static 31P NMR techniques. These studies provide novel insights into the impact of particle engineering on the molecular properties of the LNP envelope membrane. Additionally, molecular interactions and compositional distribution play a critical role in particle engineering and the consequent stability and potency. In this study, we have identified intermolecular contacts among the lipid components using one-dimensional 1H-13C cross-polarization magic angle spinning experiments, 1H relaxation measurements, and two-dimensional 1H-1H correlation methods, providing a structural basis for the lipid assembly. Interestingly, the cationic and ionizable lipids, conventionally regarded as stabilizing agents primarily located within the core of LNPs, were found to interact with PEG lipids and coexist in the outer layer of the particles. We suggest that LNPs examined here are comprised of an outer layer rich in lipid components surrounding a core region. Our high-resolution findings offer insightful structural and dynamic details pertaining to the individual chemical components in the lipid particles and their interactions influence lipid complex structure and stability in particle engineering.
Collapse
Affiliation(s)
- Mingyue Li
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ryan Schroder
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Umut Ozuguzel
- Department of Chemistry, University of Connecticut, Stamford, Connecticut 06901, United States
| | - Tyler M Corts
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yong Liu
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yuejie Zhao
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Wei Xu
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jing Ling
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Allen C Templeton
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Bodhisattwa Chaudhuri
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
- Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Marian Gindy
- Small Molecule Science and Technology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Angela Wagner
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
28
|
García Rodríguez B, Olsén E, Skärberg F, Volpe G, Höök F, Midtvedt DS. Optical label-free microscopy characterization of dielectric nanoparticles. NANOSCALE 2025; 17:8336-8362. [PMID: 40079204 PMCID: PMC11904879 DOI: 10.1039/d4nr03860f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
In order to relate nanoparticle properties to function, fast and detailed particle characterization is needed. The ability to characterize nanoparticle samples using optical microscopy techniques has drastically improved over the past few decades; consequently, there are now numerous microscopy methods available for detailed characterization of particles with nanometric size. However, there is currently no "one size fits all" solution to the problem of nanoparticle characterization. Instead, since the available techniques have different detection limits and deliver related but different quantitative information, the measurement and analysis approaches need to be selected and adapted for the sample at hand. In this tutorial, we review the optical theory of single particle scattering and how it relates to the differences and similarities in the quantitative particle information obtained from commonly used label-free microscopy techniques, with an emphasis on nanometric (submicron) sized dielectric particles. Particular emphasis is placed on how the optical signal relates to mass, size, structure, and material properties of the detected particles and to its combination with diffusivity-based particle sizing. We also discuss emerging opportunities in the wake of new technology development, including examples of adaptable python notebooks for deep learning image analysis, with the ambition to guide the choice of measurement strategy based on various challenges related to different types of nanoparticle samples and associated analytical demands.
Collapse
Affiliation(s)
| | - Erik Olsén
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden.
| | - Fredrik Skärberg
- Department of Physics, University of Gothenburg, Gothenburg, Sweden.
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden.
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden.
| | | |
Collapse
|
29
|
Kim J. Smart Nanocarriers in Cosmeceuticals Through Advanced Delivery Systems. Biomimetics (Basel) 2025; 10:217. [PMID: 40277615 DOI: 10.3390/biomimetics10040217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Nanomaterials have revolutionized various biological applications, including cosmeceuticals, enabling the development of smart nanocarriers for enhanced skin delivery. This review focuses on the role of nanotechnologies in skincare and treatments, providing a concise overview of smart nanocarriers, including thermo-, pH-, and multi-stimuli-sensitive systems, focusing on their design, fabrication, and applications in cosmeceuticals. These nanocarriers offer controlled release of active ingredients, addressing challenges like poor skin penetration and ingredient instability. This work discusses the unique properties and advantages of various nanocarrier types, highlighting their potential in addressing diverse skin concerns. Furthermore, we address the critical aspect of biocompatibility, examining potential health risks associated with nanomaterials. Finally, this review highlights current challenges, including the precise control of drug release, scalability, and the transition from in vitro to in vivo applications. We also discuss future perspectives such as the integration of digital technologies and artificial intelligence for personalized skincare to further advance the technology of smart nanocarriers in cosmeceuticals.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
30
|
George M, Boukherroub R, Sanyal A, Szunerits S. Treatment of lung diseases via nanoparticles and nanorobots: Are these viable alternatives to overcome current treatments? Mater Today Bio 2025; 31:101616. [PMID: 40124344 PMCID: PMC11930446 DOI: 10.1016/j.mtbio.2025.101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Challenges Respiratory diseases remain challenging to treat, with current efforts primarily focused on managing symptoms rather than maintaining overall lung health. Traditional treatment methods, such as oral or parenteral administration of antiviral, antibacterial, and anti-inflammatory drugs, face limitations. These include difficulty in delivering therapeutic agents to pathogens residing deep in the airways and the risk of severe side effects due to high systemic drug concentrations. The growing threat of drug-resistant pathogens further complicates infection management. Advancements The lung's large surface area offers an attractive target for inhalation-based drug delivery. Nanoparticles (NP) enable uniform and sustained drug distribution across the alveolar network, overcoming challenges posed by complex lung anatomy. Recent breakthroughs in nanorobots (NR) have demonstrated precise navigation through biological environments, delivering therapies directly to affected lung areas with enhanced accuracy. Nanotechnology has also shown promise in treating lung cancer, with nanoparticles engineered to overcome biological barriers, improve drug solubility, and enable controlled drug release. Future scope This review explores the progress of NP and NR in addressing challenges in pulmonary drug delivery. These innovations allow targeted delivery of nucleic acids, drugs, or peptides to the pulmonary epithelium with unprecedented accuracy, offering significant potential for improving therapeutic effectiveness in respiratory disorders.
Collapse
Affiliation(s)
- Meekha George
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Sabine Szunerits
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| |
Collapse
|
31
|
Skwarczynski M, Alharbi N, Nahar UJ, Shalash AO, Azuar A, Koirala P, Khisty SJ, Wang J, Marasini N, Hussein WM, Khalil ZG, Toth I. Influence of component structural arrangement on cholesterol-antigen conjugate immunogenicity and antisera bactericidal activity against group A Streptococcus. Bioorg Chem 2025; 157:108248. [PMID: 39952060 DOI: 10.1016/j.bioorg.2025.108248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Immune stimulants (adjuvants) are essential vaccine components; however, clinically approved adjuvants are limited with the majority being derived from pathogenic components. In this study, the adjuvanting capacity of cholesterol, a natural human lipid, was explored following conjugation with peptide antigens. A structure-activity relationship study was conducted to compare linear and branched cholesterol conjugates with other lipopeptide vaccines and commercial adjuvants. Group A Streptococcus (GAS) M protein-derived J8 B-cell epitope and a universal helper T-cell epitope P25 were selected as an antigen. In addition, liposomal formulations of the cholesterol-based vaccines were also evaluated in the mouse model. Following subcutaneous and intranasal administration, conjugates comprised of cholesterol, P25 and J8 induced the highest antibody production. Linear cholesterol peptide vaccines triggered strong antibody responses that killed GAS clinical isolates as effectively as responses triggered by commercial adjuvants. The immunogenicity of the vaccines was greatly influenced by the structural arrangement of the vaccine conjugate components. The lead cholesterol conjugate was self-adjuvanting and induced the desired immune response without any exogenous immune stimulation.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Nedaa Alharbi
- Applied College at Khulais, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Ummey J Nahar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Armira Azuar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Shefali J Khisty
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jingwen Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nirmal Marasini
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Waleed M Hussein
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zeinab G Khalil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
32
|
Meier N, Berten‐Schunk L, Roger Y, Hänsch R, Hoffmann A, Bunjes H, Dempwolf W, Menzel H. Characterization of Thin Polymer Layer Prepared from Liposomes and Polyelectrolytes for TGF-β 3 Release in Tissue Engineering. Macromol Biosci 2025; 25:e2400447. [PMID: 39803844 PMCID: PMC11995839 DOI: 10.1002/mabi.202400447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/09/2024] [Indexed: 04/15/2025]
Abstract
Implant-integrated drug delivery systems that enable the release of biologically active factors can be part of an in situ tissue engineering approach to restore biological function. Implants can be functionalized with drug-loaded nanoparticles through a layer-by-layer assembly. Such coatings can release biologically active levels of growth factors. Sustained release is desired for many in vivo applications. The layer-by-layer technique also allows for the addition of extra layers, which can serve as "barriers" to delay the release. Electrospun Polycaprolactone (PCL) fiber mats are modified with a Chitosan (CS) grafted with PCL sidechains (CS-g-PCL24) and coated with transforming growth factor beta 3 (TGF-β3) loaded Chitosan/tripolyphosphate nanoparticles as a drug delivery system. Additional layers including polystyrene sulfonate, alginate, carboxymethyl cellulose, and liposomes (phosphatidylcholine) are applied. Streaming potential and X-ray photoelectron spectroscopy (XPS) measurements indicated a strong interpenetration of the chitosan and polyanion layers, while liposomes formed separate layers, which are more promising for sustained release. All samples release TGF-β3 at different cumulative levels without altering release kinetics. Variations in layer structure, interpenetration, and stability depending on the chitosan used are observed, which ultimately has minimal impact on the release kinetics. Polyelectrolyte layers strongly interpenetrated the active layers and therefore do not act as effective diffusion barriers, while the liposome layer, though separated, lacked sufficient stability.
Collapse
Affiliation(s)
- Nils Meier
- Institute for Technical ChemistryMacromolecular ChemistryTU BraunschweigHagenring 3038106BraunschweigGermany
| | - Leonie Berten‐Schunk
- Institute of Pharmaceutical Technology and BiopharmaceuticsTU BraunschweigMendelssohnstrasse 138106BraunschweigGermany
| | - Yvonne Roger
- Department of Orthopedic SurgeryHannover Medical SchoolCarl‐Neuberg‐Straße 130625HannoverGermany
- Graded Implants and Regenerative StrategiesNiedersächsisches Zentrum für BiomedizintechnikImplantatforschung und Entwicklung (NIFE)Stadtfelddamm 3430625HannoverGermany
| | - Robert Hänsch
- Institute of Plant BiologyTU BraunschweigHumboldtstraße 138106BraunschweigGermany
| | - Andrea Hoffmann
- Department of Orthopedic SurgeryHannover Medical SchoolCarl‐Neuberg‐Straße 130625HannoverGermany
- Graded Implants and Regenerative StrategiesNiedersächsisches Zentrum für BiomedizintechnikImplantatforschung und Entwicklung (NIFE)Stadtfelddamm 3430625HannoverGermany
| | - Heike Bunjes
- Institute of Pharmaceutical Technology and BiopharmaceuticsTU BraunschweigMendelssohnstrasse 138106BraunschweigGermany
| | - Wibke Dempwolf
- Institute for Technical ChemistryMacromolecular ChemistryTU BraunschweigHagenring 3038106BraunschweigGermany
| | - Henning Menzel
- Institute for Technical ChemistryMacromolecular ChemistryTU BraunschweigHagenring 3038106BraunschweigGermany
| |
Collapse
|
33
|
Gautam K, Singh H, Sinha AK. Nanotechnology in Plant Nanobionics: Mechanisms, Applications, and Future Perspectives. Adv Biol (Weinh) 2025; 9:e2400589. [PMID: 39936866 DOI: 10.1002/adbi.202400589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/04/2025] [Indexed: 02/13/2025]
Abstract
Plants are vital to ecosystems and human survival, possessing intricate internal and inter-plant signaling networks that allow them to adapt quickly to changing environments and maintain ecological balance. The integration of engineered nanomaterials (ENMs) with plant systems has led to the emergence of plant nanobionics, a field that holds the potential to enhance plant capabilities significantly. This integration may result in improved photosynthesis, increased nutrient uptake, and accelerated growth and development. Plants treated with ENMs can be stress mitigators, pollutant detectors, environmental sensors, and even light emitters. This review explores recent advancements in plant nanobionics, focusing on nanoparticle (NP) synthesis, adhesion, uptake, transport, fate, and application in enhancing plant physiological functioning, stress mitigation, plant health monitoring, energy production, environmental sensing, and overall plant growth and productivity. Potential research directions and challenges in plant nanobionics are highlighted, and how material optimization and innovation are propelling the growth in the field of smart agriculture, pollution remediation, and energy/biomass production are discussed.
Collapse
Affiliation(s)
- Kajal Gautam
- Department of Chemistry, School of advanced Engineering, UPES, Dehradun, India
| | - Hukum Singh
- Plant Physiology, Genetics and Tree Improvement Division, Forest Research institute (FRI), Dehradun, India
| | - A K Sinha
- Department of Physics, School of advanced Engineering, UPES, Dehradun, India
| |
Collapse
|
34
|
Mathiesen H, Juul-Madsen K, Tramm T, Vorup-Jensen T, Møller HJ, Etzerodt A, Andersen MN. Prognostic value of CD163 + macrophages in solid tumor malignancies: A scoping review. Immunol Lett 2025; 272:106970. [PMID: 39778658 DOI: 10.1016/j.imlet.2025.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Tumor-associated macrophages (TAMs) play crucial roles in development and progression of malignant diseases. Notably, CD163+ TAMs likely perform specific pro-tumorigenic functions, suggesting that this subset may serve as both prognostic biomarkers and targets for future anti-cancer therapy. We conducted a scoping review to map the current knowledge on the prognostic role of CD163+ TAMs in the five most lethal cancers worldwide: Lung, colorectal, gastric, liver, and breast cancer. For all cancer types, most studies showed that high tumoral presence of CD163+ cells was associated with poor patient outcome, and this association was more frequently observed when CD163+ cells were measured at the tumor periphery compared to more central parts of the tumor. These results support that CD163+ TAMs represent a biomarker of poor patient outcome across a variety of solid tumors, and highlight the relevance of further investigations of CD163+ TAMs as targets of future immunotherapies.
Collapse
Affiliation(s)
- Henriette Mathiesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Juul-Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Trine Tramm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Morten Nørgaard Andersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
35
|
Brako F, Boateng J. Transmucosal drug delivery: prospects, challenges, advances, and future directions. Expert Opin Drug Deliv 2025; 22:525-553. [PMID: 39976299 DOI: 10.1080/17425247.2025.2470224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Traditional administration routes have limitations including first-pass metabolism and gastrointestinal degradation for sensitive drugs (oral) and pain associated with parenteral injections, which also require trained personnel and refrigeration, making them expensive. This has increased interest in alternative routes, with mucosal surfaces being of high priority. AREAS COVERED Mucosal routes include ocular, oral (buccal/sublingual), nasal and vaginal mucosae which avoid the limitations of the oral and parenteral routes. Though mucosal routes show great potential, they are still hindered by several barriers, especially for systemic absorption, resulting in the development of more advanced novel drug delivery systems to overcome these limitations and achieve therapeutic actions both locally and systemically, similar to or exceeding the oral route. This paper systematically reviews and compares the different mucosal routes, challenges, and recent advances in advanced novel drug delivery system design for emerging clinical challenges including the advent of large biological macromolecules (proteins, peptides, and RNA) for treatment and prevention of diseases. The review also focuses on current challenges and future perspectives. EXPERT OPINION Among the various transmucosal routes discussed, nose-to-brain drug delivery has the greatest translational potential to go beyond the current state of the art and achieve significant clinical impact for neurological diseases.
Collapse
Affiliation(s)
- Francis Brako
- School of Science, Faculty of Engineering and Science, University of Greenwich, Chatham, Maritime, UK
| | - Joshua Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Chatham, Maritime, UK
| |
Collapse
|
36
|
Lam CD, Park S. Nanomechanical characterization of soft nanomaterial using atomic force microscopy. Mater Today Bio 2025; 31:101506. [PMID: 40018054 PMCID: PMC11867545 DOI: 10.1016/j.mtbio.2025.101506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 03/01/2025] Open
Abstract
Atomic force microscopy (AFM) is a promising method for generating high-spatial-resolution images, providing insightful perspectives on the nanomechanical attributes of soft matter, including cells, bacteria, viruses, proteins, and nanoparticles. AFM is widely used in biological and pharmaceutical sciences because it can scrutinize mechanical properties under physiological conditions. We comprehensively reviewed experimental techniques and fundamental mathematical models to investigate the mechanical properties, including elastic moduli and binding forces, of soft materials. To determine these mechanical properties, two-dimensional arrays of force-distance (f-d) curves are obtained through AFM indentation experiments using the force volume technique. For elasticity determination, models are divided into approach f-d curve-based models, represented by the Hertz model, and retract f-d curve-based models, exemplified by the Johnson-Kendall-Roberts and Derjaguin-Müller-Toporov models. Especially, the Chen, Tu, and Cappella models, developed from the Hertz model, are used for thin samples on hard substrates. Additionally, the establishment of physical or chemical bonds during indentation experiments, observable in retract f-d curves, is crucial for the adhesive properties of samples and binding affinity between antibodies (receptors) and antigens (ligands). Chemical force microscopy, single-molecule force spectroscopy, and single-cell force spectroscopy are primary AFM methods that provide a comprehensive view of such properties through retract curve analysis. Furthermore, this paper, structured into key thematic sections, also reviews the exemplary application of AFM across multiple scientific disciplines. Notably, cancer cells are softer than healthy cells, although more sophisticated investigations are required for prognostic applications. AFM also investigates how bacteria adapt to antibiotics, addressing antimicrobial resistance, and reveals that stiffer virus capsids indicate reduced infectivity, aiding in the development of new strategies to combat viral infections. Moreover, AFM paves the way for innovative therapeutic approaches in designing effective drug delivery systems by providing insights into the physical properties of soft nanoparticles and the binding affinity of target moieties. Our review provides researchers with representative studies applying AFM to a wide range of cross-disciplinary research.
Collapse
Affiliation(s)
- Chi-Dat Lam
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Soyeun Park
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| |
Collapse
|
37
|
Xiao W, Geng R, Bi D, Sun Y, Li Z, Liu Y, Zhu J. Responsive boronate ester lipid nanoparticles for enhanced delivery of veliparib and platinum (IV) prodrug in chemotherapy. J Colloid Interface Sci 2025; 683:375-386. [PMID: 39693876 DOI: 10.1016/j.jcis.2024.12.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
The chemotherapeutic effectiveness of breast cancer treatment is currently unsatisfactory due to inadequate drug delivery, suboptimal drug release, and drug inactivation. Herein, we present an innovative boronate ester lipid nanoformulation to improve the delivery of a platinum (IV) prodrug (Pt-C12) and veliparib (Veli), aiming to increase their therapeutic efficacy through a synergistic effect. We identify the optimal ratio of Pt-C12 to Veli for achieving synergy in vitro, followed by the encapsulation of Pt-C12 and Veli in lipid nanoparticles (NPs) incorporating responsive boronate ester lipids (LPC-PPE) to produce responsive lipid NPs (LPV NPs). These LPV NPs demonstrate high sensitivity to low levels of hydrogen peroxide (H2O2), enabling efficient drug release. In contrast, the nonresponsive lipid NP (DPV NP) control shows minimal responsiveness to H2O2. Furthermore, acidic tumor microenvironments trigger phenylboronic acid (PBA) generation from LPC-PPE in the LPV NPs. Compared with DPV NPs, the interaction between PBA on the LPV NPs and sugar components on tumor cells significantly improves LPV NP cellular uptake and lysosomal escape in vitro. Due to the enhanced cellular delivery and the synergistic drug combination, the LPV NPs induce an increase in apoptosis in 4 T1 cells compared with control groups. Moreover, the LPV NPs exhibit greater efficiency of drug delivery to tumors than the DPV NPs, and have a greater inhibitory effect on tumors than the controls. Overall, our findings highlight the potential of functional lipids and synergistic drug combinations in overcoming obstacles in breast cancer treatment and advancing the development of responsive delivery systems.
Collapse
Affiliation(s)
- Wanyue Xiao
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Rui Geng
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Duohang Bi
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Yufeng Sun
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Zhilang Li
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Yijing Liu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518057, China.
| | - Jintao Zhu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| |
Collapse
|
38
|
Yan J, Peng Y, Huang S, Fei W, Chen Y, Zheng C, Zhang M, Zhao M. Treatment of endometriosis with mifepristone mediated by nanostructured lipid carriers. Drug Deliv Transl Res 2025; 15:1181-1192. [PMID: 38992183 DOI: 10.1007/s13346-024-01661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 07/13/2024]
Abstract
Mifepristone, a progesterone receptor antagonist, was initially used to terminate early pregnancy. As scientific research advanced, it emerged to be effective in the treatment of various tumors and tumor-like conditions such as endometriosis. Despite the therapeutic potential of mifepristone, its therapeutic effect is still far from ideal because the drug is difficult to dissolve and to accumulate in the target tissue sites. To address this issue, mifepristone-loaded nanostructured lipid carriers (Mif-NLC) were prepared by a simple solvent diffusion method and their anti-endometriosis performance and mechanisms were initially investigated. By optimizing the preparation protocol, we obtained uniform and spheroidal Mif-NLC with an average particle size of 280 nm. The encapsulation rate and drug loading capacity were 64.67% ± 0.15% and 2.7% ± 0.014%, respectively, as measured by UV spectrophotometry. The in vitro release kinetics indicated that mifepristone was released from NLC in a sustained-release manner. Compared with free mifepristone, Mif-NLC exhibited enhanced cellular uptake and inhibition of invasion activity in primary mesenchymal cells of endometriosis. A certain reduction in the size of endometriotic cysts was observed in animals compared to controls. The induction of autophagy via Mif-NLC may serve as the molecular mechanism underlying this effect. Furthermore, observation of uterine structures showed negligible toxic effects. This suggested that mifepristone encapsulated in NLC can improve its bioavailability and anti-endometriosis efficacy, which provided a new strategy for the treatment of endometriosis.
Collapse
Affiliation(s)
- Jingjing Yan
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yujie Peng
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shiyao Huang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Mengdan Zhao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
39
|
Azouz AA, El komy MH, Elmowafy M, Mahmoud MO, Ali FE, Aboud HM. Crafting cationic lecithmer nanocomposites as promising wagons for brain targeting of cinnamaldehyde: Accentuated neuroprotection via downregulation of Aβ1-42/p-tau crosstalk. J Drug Deliv Sci Technol 2025; 106:106664. [DOI: 10.1016/j.jddst.2025.106664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2025]
|
40
|
Meng S, Hara T, Miura Y, Arao Y, Saito Y, Inoue K, Hirotsu T, Vecchione A, Satoh T, Ishii H. In Vivo Engineered CAR-T Cell Therapy: Lessons Built from COVID-19 mRNA Vaccines. Int J Mol Sci 2025; 26:3119. [PMID: 40243757 PMCID: PMC11988490 DOI: 10.3390/ijms26073119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized cancer immunotherapy but continues to face significant challenges that limit its broader application, such as antigen targeting, the tumor microenvironment, and cell persistence, especially in solid tumors. Meanwhile, the global implementation of mRNA vaccines during the COVID-19 pandemic has highlighted the transformative potential of mRNA and lipid nanoparticle (LNP) technologies. These innovations, characterized by their swift development timelines, precise antigen design, and efficient delivery mechanisms, provide a promising framework to address some limitations of CAR-T therapy. Recent advancements, including mRNA-based CAR engineering and optimized LNP delivery, have demonstrated the capacity to enhance CAR-T efficacy, particularly in the context of solid tumors. This review explores how mRNA-LNP technology can drive the development of in vivo engineered CAR-T therapies to address current limitations and discusses future directions, including advancements in mRNA design, LNP optimization, and strategies for improving in vivo CAR-T functionality and safety. By bridging these technological insights, CAR-T therapy may evolve into a versatile and accessible treatment paradigm across diverse oncological landscapes.
Collapse
Grants
- grant nos. 19K22658, 20H00541, 21K19526, 22H03146, 22K19559, 23K19505, 23K18313, 23KK0153, 24K22144, and 16H06279 (PAGS) Ministry of Education, Culture, Sports, Science and Technology
- grant nos. JP23ym0126809 and JP24ym0126809 Japan Agency for Medical Research and Development
- 23-255001 Princess Takamatsu Cancer Research Fund
- G-2024-3-00 IFO Research Communications
- 2024 Oceanic Wellness Foundation
- 2024 Suzuken Memorial Foundation
Collapse
Affiliation(s)
- Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Integrated Research, Institute of Science Tokyo, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501, Japan
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yoshiko Saito
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Kana Inoue
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | | | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035, 00189 Rome, Italy
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
41
|
Costa M, Soares C, Silva A, Barroso MF, Simões P, Ferreira M, Gameiro P, Grosso C, Delerue-Matos C. Optimization of Nanoencapsulation of Codium tomentosum Extract and Its Potential Application in Yogurt Fortification. Mar Drugs 2025; 23:147. [PMID: 40278268 DOI: 10.3390/md23040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Marine macroalgae are excellent sources of bioactive compounds recognized by their pharmaceutical and biomedical potential. A subcritical water extraction (SWE) was applied to the macroalga Codium tomentosum, and the extract was used to prepare phytosomes. A Box-Behnken design was applied to optimize the entrapment efficiency. These phytosomes were further modified with DSPE-PEG (2000)-maleimide and apolipoprotein E and characterized by dynamic light scattering, UV spectrophotometry, octanol/water partition coefficient, differential scanning calorimetry, and Fourier transform infrared spectroscopy. As proof of concept, prototypes of functional food tailored to the elderly were produced. Yogurts were fortified with seaweed extract or phytosomes, and physicochemical properties and proximal composition (pH, acidity, syneresis, moisture, peroxides, proteins, total lipids, sugar content, ash, and mineral composition) were analyzed. The antioxidant and the inhibition capacity of two brain enzymes, cholinesterases (AChE and BuChE), involved in the pathogenesis of Alzheimer's disease, were also evaluated in the final prototypes. Despite their unappealing sensory characteristics, the results are promising for integrating marine extracts with potential neuroprotective effects into functional foods.
Collapse
Affiliation(s)
- Micaela Costa
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
| | - Cristina Soares
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
| | - Aurora Silva
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
- Department of Analytical Chemistry and Food Science, Nutrition and Food Group (NuFoG), Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Universidade de Vigo, 36310 Vigo, Spain
| | - Maria Fátima Barroso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
| | - Pedro Simões
- LAQV, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal
| | - Mariana Ferreira
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Paula Gameiro
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Clara Grosso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
| |
Collapse
|
42
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
43
|
Du J, Wang H, Zhong L, Wei S, Min X, Deng H, Zhang X, Zhong M, Huang Y. Bioactivity and biomedical applications of pomegranate peel extract: a comprehensive review. Front Pharmacol 2025; 16:1569141. [PMID: 40206073 PMCID: PMC11979244 DOI: 10.3389/fphar.2025.1569141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Pomegranate peel is a by-product generated during the processing of pomegranate (Punica granatum L.) fruit, accounting for approximately 50% of the total mass of the fruit. Although pomegranate peel is usually regarded as waste, it is rich in various bioactive metabolites such as polyphenols, tannins, and flavonoids, demonstrating significant medicinal and nutritional value. In recent years, Pomegranate peel extract (PPE) has shown broad application prospects in the biomedical field due to its multiple effects, including antioxidant, anti-inflammatory, antibacterial, anti-apoptotic properties, and promotion of cell regeneration. This review consolidates the major bioactive metabolites of PPE and explores its applications in biomedical materials, including nanodrug carriers, hydrogels, and tissue engineering scaffolds. By synthesizing the existing literature, we delve into the potential value of PPE in biomedicine, the challenges currently encountered, and the future directions for research. The aim of this review is to provide a scientific basis for optimizing the utilization of PPE and to facilitate its broader application in the biomedical field.
Collapse
Affiliation(s)
- Jinsong Du
- School of Health Management, Zaozhuang University, Zaozhuang, China
- Department of Teaching and Research, Shandong Coal Health School, Zaozhuang, China
| | - Heming Wang
- School of Nursing, Jilin University, Jilin, China
| | - Lingyun Zhong
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Shujie Wei
- Image Center, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Xiaoqiang Min
- Department of Teaching and Research, Shandong Coal Health School, Zaozhuang, China
- Department of Geriatics, Shandong Healthcare Group Xinwen Central Hospital, Taian, China
| | - Hongyan Deng
- School of Health Management, Zaozhuang University, Zaozhuang, China
| | - Xiaoyan Zhang
- Magnetic Resonance Imaging Department, Shandong Healthcare Group Zaozhuang Central Hospital, Zaozhuang, China
| | - Ming Zhong
- Lanshu Cosmetics Co., Ltd., Huzhou, Zhejiang, China
| | - Yi Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
44
|
Lee J, Jung W, Lee D, Kang M, Chung HJ, Yeom J. Chirality-Controlled Lipid Nanoparticles for mRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18150-18159. [PMID: 40096648 DOI: 10.1021/acsami.5c00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
mRNA therapeutics present a promising strategy for the treatment of human diseases, which requires a carrier to protect the mRNA and ensure its effective cellular delivery. However, research on how the surface physicochemical properties of the mRNA carriers affect the efficiency of delivery of mRNA into cells has been limited thus far. Here, we report that chirality control of lipid nanoparticles (LNPs) can enhance the delivery and transfection efficiency of cargo mRNA into cells. We prepared chiral LNPs by surface modification of LNPs with chiral cobalt oxide nanoparticles. The chiral LNPs with d- or l-chirality resulted in different cellular uptake and transfection efficiencies of luciferase or EGFP mRNA. Chiral LNPs with d-chirality showed significant enhancement in cellular delivery as well as transfection (5.3-fold) of mRNA compared to the control LNPs. This study suggests a novel strategy of using chirality for the delivery of nucleic acid drugs and provides a more in-depth understanding of the effect of chirality on cells.
Collapse
Affiliation(s)
- Juhee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Wookjin Jung
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Dongkyu Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Mihyun Kang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jihyeon Yeom
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
45
|
Gomes F, Wasserberg D, Edelbroek R, van Weerd J, Jonkheijm P, Leijten J. OPSALC: On-Particle Solvent-Assisted Lipid Coating to Create Erythrocyte Membrane-like Coatings with Improved Hemocompatibility. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18179-18193. [PMID: 40079786 PMCID: PMC11955951 DOI: 10.1021/acsami.5c02103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Particles are essential building blocks in nanomedicine and cell engineering. Their administration often involves blood contact, which demands a hemocompatible material profile. Coating particles with isolated cell membranes is a common strategy to improve hemocompatibility, but this solution is nonscalable and potentially immunogenic. Cell membrane-like lipid coatings are a promising alternative, as lipids can be synthesized on a large scale and used to create safe cell membrane-like supported bilayers. However, a method to controllably and scalably lipid-coat a wide range of particles has remained elusive. Here, an on-particle solvent-assisted lipid coating (OPSALC) method is introduced as an innovative technique to endow various types of particles with cell membrane-like coatings. Coating formation efficiency is shown to depend on lipid concentration, buffer addition rate, and solvent:buffer ratio, as these parameters determine lipid assembly and lipid-surface interactions. Four lipid formulations with various levels of erythrocyte membrane mimicry are explored in terms of hemocompatibility, demonstrating a reduced particle-induced hemolysis and plasma coagulation time. Interestingly, formulations with higher mimicry levels show the lowest levels of complement activation and highest colloidal stability. Overall, OPSALC represents a simple yet scalable strategy to endow particles with cell membrane-like lipid coatings to facilitate blood-contact applications.
Collapse
Affiliation(s)
- Francisca
L. Gomes
- Department
of Bioengineering Technologies, Leijten Laboratory, Faculty of Science
and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
- Department
of Molecules and Materials, Laboratory of Biointerface Chemistry,
Faculty of Science and Technology, Technical Medical Centre and MESA+
Institute, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
| | - Dorothee Wasserberg
- Department
of Molecules and Materials, Laboratory of Biointerface Chemistry,
Faculty of Science and Technology, Technical Medical Centre and MESA+
Institute, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
- LipoCoat
BV, Hengelosestraat 535, Enschede 7521AG, The Netherlands
| | - Rick Edelbroek
- Department
of Molecules and Materials, Laboratory of Biointerface Chemistry,
Faculty of Science and Technology, Technical Medical Centre and MESA+
Institute, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
| | - Jasper van Weerd
- LipoCoat
BV, Hengelosestraat 535, Enschede 7521AG, The Netherlands
| | - Pascal Jonkheijm
- Department
of Molecules and Materials, Laboratory of Biointerface Chemistry,
Faculty of Science and Technology, Technical Medical Centre and MESA+
Institute, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
| | - Jeroen Leijten
- Department
of Bioengineering Technologies, Leijten Laboratory, Faculty of Science
and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, Enschede 7522NB, The Netherlands
| |
Collapse
|
46
|
Yang LX, Li H, Cheng ZH, Sun HY, Huang JP, Li ZP, Li XX, Hu ZG, Wang J. The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases. Int J Mol Sci 2025; 26:3055. [PMID: 40243658 PMCID: PMC11988403 DOI: 10.3390/ijms26073055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Non-coding RNAs (ncRNAs) are a class of RNAs that largely lack the capacity to encode proteins. They have garnered significant attention due to their central regulatory functions across numerous cellular and physiological processes at transcriptional, post-transcriptional, and translational levels. Over the past decade, ncRNA-based therapies have gained considerable attention in the diagnosis, treatment, and prevention of diseases, and many studies have revealed a significant relationship between ncRNAs and diseases. At the same time, due to their tissue specificity, an increasing number of projects have focused on the application of ncRNAs as biomarkers in diseases, as well as the design and development of novel ncRNA-based vaccines and therapies for clinical use. These ncRNAs may also drive research into the potential molecular mechanisms and complex pathogenesis of related diseases. However, new biomarkers need to be validated for their clinical effectiveness. Additionally, to produce safe and stable RNA products, factors such as purity, precise dosage, and effective delivery methods must be ensured to achieve optimal bioactivity. These challenges remain key issues in the clinical application of ncRNAs. This review summarizes the prospects of ncRNAs as potential biomarkers, as well as the current research status and clinical applications of ncRNAs in therapies and vaccines, and discusses the challenges and expectations of ncRNAs in disease diagnosis and drug therapy.
Collapse
Affiliation(s)
- Lu-Xuan Yang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - Hui Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - Zhi-Hui Cheng
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - He-Yue Sun
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - Jie-Ping Huang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - Zhi-Peng Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| | - Xin-Xin Li
- Institute of Scientific Research, Guangxi University, Nanning 530004, China;
| | - Zhi-Gang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jian Wang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (L.-X.Y.); (H.L.); (Z.-H.C.); (H.-Y.S.); (J.-P.H.); (Z.-P.L.)
| |
Collapse
|
47
|
Kaps FT, Ziegler AL, Fritsche P, Takmakova E, Kerr A, Boye S, Lederer A, Luxenhofer R. Electron-Deficient Alkyne Lipids Enable Efficient Synthesis of Comparable Polymer Lipids via Copper-Free Azide-Alkyne Cycloaddition. Angew Chem Int Ed Engl 2025:e202501262. [PMID: 40129178 DOI: 10.1002/anie.202501262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Polymer lipids (PLs) are essential components of liposomes and lipid nanoparticles (LNPs) for drug and gene delivery, providing colloidal stabilization and defining the biological interface. While poly(ethylene glycol) (PEG)-based PLs are the current standard, they are suspected to be responsible for rare adverse reactions, e. g. to LNP-based Covid-19 vaccines. Therefore, PLs based on alternative stealth polymers are being intensively investigated for their use in LNPs. However, alternative PLs often lack comparability due to different synthesis protocols and are often not easily accessible. Herein we present a catalyst-free, efficient and versatile coupling procedure for PL synthesis based on azide-functionalized polymers and electron-deficient acetylene dicarboxylate lipids. To highlight the versatility of this approach, we prepared PLs based on PEG and 4 alternative stealth polymers with quantitative coupling efficiencies. The linker structure showed appropriate pH stability and biocompatibility. All PLs enabled the preparation of well-defined liposomes with excellent stability. Our facile and versatile approach yields comparable PLs with minimized linker size, making them promising candidates for future comparative studies, and biomedical applications.
Collapse
Affiliation(s)
- Florian T Kaps
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| | - Anna-Lena Ziegler
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| | - Paul Fritsche
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| | - Ekaterina Takmakova
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| | - Andrew Kerr
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| | - Susanne Boye
- Leibniz-Institut für Polymerforschung, Center Macromolecular Structure Analysis, Hohe Str. 6, 01069, Dresden, Germany
| | - Albena Lederer
- Leibniz-Institut für Polymerforschung, Center Macromolecular Structure Analysis, Hohe Str. 6, 01069, Dresden, Germany
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Robert Luxenhofer
- Soft Matter Chemistry, Department of Chemistry, and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, 00014, Finland
| |
Collapse
|
48
|
Fu G, Zhao Y, Mao C, Liu Y. Enhancing nano-immunotherapy of cancer through cGAS-STING pathway modulation. Biomater Sci 2025. [PMID: 40111213 DOI: 10.1039/d4bm01532k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a critical role in cancer immunotherapy due to the secretion of multiple pro-inflammatory cytokines and chemokines. Numerous cGAS-STING agonists have been developed for preclinical and clinical trials in tumor immunity. However, several obstacles, such as agonist molecules requiring multiple doses, rapid degradation and poor targeting, weaken STING activation at the tumor site. The advancement of nanotechnology provides an optimized platform for the clinical application of STING agonists. In this review, we summarize events of cGAS-STING pathway activation, the dilemma of delivering STING agonists, and recent advances in the nano-delivery of cGAS-STING agonist formulations for enhancing tumor immunity. Furthermore, we address the future challenges associated with STING-based therapies and offer insights to guide subsequent clinical applications.
Collapse
Affiliation(s)
- Gaohong Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Yanan Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Chengqiong Mao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, P. R. China
| | - Yang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
49
|
Laturski AE, Dulay MT, Perry JL, DeSimone JM. Transfection via RNA-Based Nanoparticles: Comparing Encapsulation vs Adsorption Approaches of RNA Incorporation. Bioconjug Chem 2025; 36:367-376. [PMID: 39999074 DOI: 10.1021/acs.bioconjchem.5c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Historically, RNA delivery via nanoparticles has primarily relied on encapsulation, as demonstrated by lipid nanoparticles in SARS-CoV-2 vaccines. Concerns about RNA degradation on nanoparticle surfaces initially limited the exploration of adsorption-based approaches. However, recent advancements have renewed interest in adsorption as a viable alternative. This Viewpoint explores the approaches of RNA incorporation in nanoparticles, comparing encapsulation, adsorption, and the combination of encapsulation and adsorption, and presents a framework to guide the selection of the most suitable strategy based on general characteristics.
Collapse
Affiliation(s)
- Amy E Laturski
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Maria T Dulay
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Jillian L Perry
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7575, United States
| | - Joseph M DeSimone
- Department of Chemical Engineering and Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
50
|
Zhang Y, Zeng R, Xia Y, Han W, Luan Y, Zhang Y, Wu S, Wang S, Wang J, Chen Y, Chen D. Visualization of protrusion-localized STAT3 mRNA using a self-powered lipidic nanoflare for predicting hepatocellular carcinoma metastasis. Mikrochim Acta 2025; 192:241. [PMID: 40102274 PMCID: PMC11920302 DOI: 10.1007/s00604-025-06988-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/15/2025] [Indexed: 03/20/2025]
Abstract
Cancer cell metastasis is one of the major causes of patients death with hepatocellular carcinoma (HCC). Previous findings demonstrated that protrusion-accumulated STAT3 mRNA is highly related to HCC cell metastasis, making protrusion-localized STAT3 mRNA an ideal biomarker for evaluating HCC cell initiation and progression. A self-powered lipidic nanoflare (SLNF) has been developed for detecting the expression level of protrusion-accumulated STAT3 mRNA in individual HCC cells, which enables accurate prediction of HCC metastasis. The LNF system is a cholesterol micelle decorated with two kinds of DNA probes, a double-stranded response DNA and a single-stranded fuel probe. The cholesterol micelle can be easily assembled from an amphipathic cholesterol-conjugated DNA via hydrophobicity-mediated aggregation, exhibiting a highly efficient cell internalization. Moreover, the compact and high-density arrangement of DNA probes on the surface of cholesterol micelle enhances their biostability. All the above features make the LNF system an ideal approach for intracellular RNA imaging. The assay commences with the binding of STAT3 mRNA to the response DNA, which peels off the waste DNA and exposes the toehold domain. This domain serves as the proximal holding point for the fuel probe to initiate a strand displacement amplification, which is a crucial step in enabling the detection of targets expressed at trace levels, yielding a limit of detection (LOD) of 100 pM at 37 °C within 1.5 h. The SLNF system is expected to provide useful insight into the development of simple and degradation-resistant DNA probes for visual prediction of HCC metastasis, showing potential applications in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Ya Zhang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China
- Hepatology Institute of Wenzhou Medical University, Wenzhou, 325035, China
| | - Ruichao Zeng
- School of Clinical Medicine, The First People's Hospital of Lin'an Distract, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yuanhang Xia
- School of Clinical Medicine, The First People's Hospital of Lin'an Distract, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, 310053, China
| | - Wei Han
- Center of Laboratory Animal, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yifei Luan
- School of Innovation and Entrepreneurship, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yuheng Zhang
- School of Clinical Medicine, The First People's Hospital of Lin'an Distract, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, 310053, China
| | - Shijia Wu
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China
- Hepatology Institute of Wenzhou Medical University, Wenzhou, 325035, China
| | - Shouhao Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China
- Hepatology Institute of Wenzhou Medical University, Wenzhou, 325035, China
| | - Jinyong Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China
- Hepatology Institute of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China.
- Hepatology Institute of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Dazhi Chen
- School of Clinical Medicine, The First People's Hospital of Lin'an Distract, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, 310053, China.
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, 325035, China.
| |
Collapse
|