1
|
Zhang M, Zhang R, Feng C, Jiang X, Xu X, Wang J. Ginsenoside compound K-based multifunctional liposomes for the treatment of rheumatoid arthritis. Drug Deliv 2025; 32:2464190. [PMID: 39957241 PMCID: PMC11834820 DOI: 10.1080/10717544.2025.2464190] [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: 06/18/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
The clinical treatment of rheumatoid arthritis (RA) with first-line therapeutic drugs is hindered by the poor solubility, low bioavailability, off-target toxicity, and insufficient accumulation in inflamed joints. Liposomes have been shown to mitigate some of these limitations in drug delivery systems. However, the use of cholesterol to stabilize liposomal structures remains controversial due to its potential association with cardiovascular diseases. Here, we developed a novel liposome based on ginsenoside compound K (CK), which not only serves as an effective therapeutic agent for RA but also replaces cholesterol as a membrane stabilizer to address these challenges. Compared with conventional liposomes, ginsenoside CK Liposomes (CK@Lipo) are excellent nanoparticles, with CK stabilizing the liposomal structure and providing targeting functionality toward inflamed joints. When encapsulated with dexamethasone (Dex), CK@Lipo exhibits a synergistic anti-inflammatory effect, slowing the progression of RA. This study provides a theoretical basis for the future development of multifunctional novel ginsenoside CK@Lipo.
Collapse
Affiliation(s)
- Meng Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| | - Chunbo Feng
- R&D Center, Shanghai Jahwa United Co., Ltd., Shanghai, China
| | - Xinnan Jiang
- R&D Center, Shanghai Jahwa United Co., Ltd., Shanghai, China
| | - Xinchun Xu
- Shanghai Xuhui Central Hospital, Xuhui Hospital Attached to Fudan University, Shanghai, China
| | - Jianxin Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| |
Collapse
|
2
|
Wang Y, He W, Ren P, Zhao L, Zheng D, Jin J. Carthamin yellow-loaded glycyrrhetinic acid liposomes alleviate interstitial fibrosis in diabetic nephropathy. Ren Fail 2025; 47:2459356. [PMID: 39904762 PMCID: PMC11800343 DOI: 10.1080/0886022x.2025.2459356] [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: 05/27/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
OBJECTIVES To investigate the therapeutic efficacy of Carthamin yellow (CY)-loaded glycyrrhetinic acid (GA) liposomes in treating diabetic nephropathy (DN), particularly in alleviating renal interstitial fibrosis and improving kidney function. METHODS CY-loaded GA liposomes were prepared and characterized for structural stability and controlled release. DN rat models were treated with CY-loaded GA liposomes, and kidney pathology, function, collagen deposition, and TGF-β1 expression were evaluated. The effects of CY-loaded GA liposomes were compared to Vitamin E and CY alone. In vitro experiments with TGF-β1-stimulated human renal interstitial fibroblasts (hRIFs) examined the effects of CY-loaded GA liposomes on cell proliferation and the expression of fibrotic markers. Mechanistic studies assessed the role of the TGFBR1/Smad2/Smad3 pathway using TGFBR1 overexpression experiments. RESULTS The CY-loaded GA liposomes exhibited a stable structure and controlled release profile. In DN rats, treatment with CY-loaded GA liposomes significantly alleviated kidney damage, improved kidney function, reduced collagen deposition and fibrosis, and downregulated TGF-β1 expression, showing superior effects compared to Vitamin E or CY alone. In TGF-β1-stimulated hRIFs, CY-loaded GA liposomes effectively suppressed cell proliferation and reduced the expression of Cyclin D1, PCNA, fibronectin, and collagen I. The inhibitory effects were stronger than CY alone and were mediated by the inactivation of the TGFBR1/Smad2/Smad3 pathway, as confirmed by TGFBR1 overexpression studies. CONCLUSIONS CY-loaded GA liposomes demonstrated significant therapeutic efficacy in alleviating renal interstitial fibrosis in DN by targeting the TGFBR1/Smad2/Smad3 pathway. This novel drug delivery system provides a promising approach for the treatment of DN.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wenfang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Peiyao Ren
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Li Zhao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Danna Zheng
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Juan Jin
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Huang H, Zhou Y, Li J, Zhang Z, Han R, Zuo J, Bai Y, Zhang J. Application of chitosan-based drug delivery systems in the treatment of bacterial diseases: a review. Drug Deliv 2025; 32:2514140. [PMID: 40491201 DOI: 10.1080/10717544.2025.2514140] [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: 11/06/2024] [Revised: 05/05/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Bacterial diseases are a significant challenge to human and animal health. The current treatment methods still have obvious shortcomings, such as poor targeting, low bioavailability, high side effects and drug resistance. Chitosan, with its outstanding biocompatibility, biodegradability, adhesiveness, antimicrobial properties, and ability to minimize drug side effects while improving bioavailability and therapeutic outcomes, serves as an ideal material for drug delivery systems, presenting a promising strategy for treating bacterial diseases. In this review, we briefly summarize the preparation methods of chitosan-based drug delivery systems and their application in the treatment of bacterial infections. The advantages of preparation of different types of chitosan-based drug delivery systems are discussed, supported by examples demonstrating their ability to improve drug antimicrobial activity, targeting, and bioavailability. Moreover, the current challenges, limitations, and future perspectives in this field were discussed, laying the groundwork for further development of chitosan-based drug delivery systems as high-performance and safe antimicrobial therapeutics.
Collapse
Affiliation(s)
- Huan Huang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yaxin Zhou
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jiehang Li
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhijin Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - RongJia Han
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jingru Zuo
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yubin Bai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jiyu Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China
- Ministry of Agriculture, Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
4
|
Meng N, Lu J, Zhou J, Yang S, Zhang C, Jia R, Ding Y, Bao Y, Wang J, Ma X, Chen R, Jiang Z, Xie C, Lu L, Lu W. Improved immunocompatibility of active targeting liposomes by attenuating nucleophilic attack of cyclic RGD peptides on complement 3. Biomaterials 2025; 321:123350. [PMID: 40267598 DOI: 10.1016/j.biomaterials.2025.123350] [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: 02/11/2025] [Revised: 03/31/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
One of the challenges for the clinical translation of active targeting nanomedicines is the adverse interactions between targeting ligands and blood components. Herein, a novel regularity, which reveals the interactions between cyclic RGD (Arg-Gly-Asp) peptide-modified liposomes and complement components in blood, is reported. As the nucleophilicity of arginine guanidine group within the cyclic RGD-like peptide increases, targeting liposomes potentiate complement cascade via the amplification loop of complement 3 (C3), ultimately leading to accelerated blood clearance, increased deposition in the reticuloendothelial system (RES) organs, enhanced immune responses, and potential side effects. By appropriately reducing the nucleophilicity of guanidine group, cyclic R2 peptide is designed for modification of liposomes to target integrin αvβ3. Compared to the widely used targeting molecule c(RGDyK), R2 eliminates the negative effects of C3 opsonization and specific antibody production, significantly improves the in vivo immunocompatibility of targeting liposomes, and demonstrates superior anti-tumor efficacy in mouse models of orthotopic breast cancer and glioma. Thus, the proposed regularity of interactions between guanidine nucleophilicity and C3, along with the successful application of the low complement activation capacity targeting ligand R2, provides new insights for addressing challenges related to complement activation in the clinical translation of active targeting nanomedicines.
Collapse
Affiliation(s)
- Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jiasheng Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Shengmin Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Chen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Ruiyi Jia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Xiaopei Ma
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Ruohan Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Zhixuan Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China.
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China.
| |
Collapse
|
5
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 PMCID: PMC11691476 DOI: 10.4103/nrr.nrr-d-24-00048] [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: 01/12/2024] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Chunyu Xiang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Renrui Niu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiaodong He
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wenqi Luo
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wanguo Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Rui Gu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
6
|
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
|
7
|
Hu VT, Ezzatpour S, Selivanovitch E, Sahler J, Pal S, Carter J, Pham QV, Adeleke RA, August A, Aguilar HC, Daniel S, Kamat NP. Cell-Free Expression of Nipah Virus Transmembrane Proteins for Proteoliposome Vaccine Design. ACS NANO 2025; 19:21290-21306. [PMID: 40458951 DOI: 10.1021/acsnano.4c16190] [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: 06/18/2025]
Abstract
Membrane proteins expressed on the surface of enveloped viruses are potent antigens in a vaccine, yet are difficult to produce and present due to their instability without a lipid scaffold. Current vaccination strategies that incorporate viral membrane proteins, such as live attenuated viruses, inactivated viruses, or extracellular vesicles, have limitations including lengthy production time, poor immunogenicity, extensive processing steps, and/or poor stability. Cell-free protein synthesis of viral membrane proteins offers a rapid, one-step method to assemble vaccine nanoparticles via cotranslational folding of membrane proteins into nanoscale liposomes. Here, we develop a vaccine candidate for the deadly Nipah virus (NiV), a highly lethal virus listed by the World Health Organization as a priority pathogen, by cell-free expressing two full-length Nipah virus membrane proteins. We demonstrate that both NiV fusion protein (NiV F) and NiV glycoprotein (NiV G) can be expressed and cotranslationally integrated into liposomes and that they fold into their native conformation. We find that the removal of a signal peptide sequence and the alteration of liposome lipid composition improve viral membrane protein incorporation. Furthermore, a lipid adjuvant, monophosphoryl lipid A (MPLA), can be readily added to liposomes without disrupting protein-vesicle loading or protein folding conformations. Finally, we demonstrate that our generated liposomal formulations lead to enhanced humoral responses in mice compared to empty and single-protein controls. This work establishes a platform to quickly assemble and present membrane antigens as multivalent vaccines that will enable a rapid response to the broad range of emerging pathogenic threats.
Collapse
Affiliation(s)
- Vivian T Hu
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Ekaterina Selivanovitch
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Julie Sahler
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Sreetama Pal
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Jordan Carter
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Quoc Vinh Pham
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Richard Ayomide Adeleke
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Neha P Kamat
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
8
|
Wang Z, Kulkarni S, Nong J, Zamora M, Ebrahimimojarad A, Hood E, Shuvaeva T, Zaleski M, Gullipalli D, Wolfe E, Espy C, Arguiri E, Wu J, Wang Y, Marcos-Contreras OA, Song W, Muzykantov VR, Fu J, Radhakrishnan R, Myerson JW, Brenner JS. A percolation phase transition controls complement protein coating of surfaces. Cell 2025:S0092-8674(25)00576-8. [PMID: 40516526 DOI: 10.1016/j.cell.2025.05.026] [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: 02/06/2024] [Revised: 12/06/2024] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
When a material enters the body, it is immediately attacked by hundreds of proteins, organized into complex networks of binding interactions and reactions. How do such complex systems interact with a material, "deciding" whether to attack? We focus on the complement system of ∼40 blood proteins that bind microbes, nanoparticles, and medical devices, initiating inflammation. We show a sharp threshold for complement activation upon varying a fundamental material parameter, the surface density of potential complement attachment points. This sharp threshold manifests at scales spanning single nanoparticles to macroscale pathologies, shown here for diverse engineered and living materials. Computational models show these behaviors arise from a minimal subnetwork of complement, manifesting percolation-type critical transitions in the complement response. This criticality switch explains the "decision" of a complex signaling network to interact with a material.
Collapse
Affiliation(s)
- Zhicheng Wang
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sahil Kulkarni
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marco Zamora
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alireza Ebrahimimojarad
- Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, NJ 08103, USA
| | - Elizabeth Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Zaleski
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Wolfe
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Carolann Espy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evguenia Arguiri
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jichuan Wu
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yufei Wang
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenchao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jinglin Fu
- Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, NJ 08103, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob S Brenner
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Shalmani AA, Daware R, Elshafei AS, De Lorenzi F, Moeckel D, Buhl EM, Klinkhammer BM, Boor P, Banala S, Adams A, Kiessling F, Storm G, Metselaar JM, Sofias AM, Shi Y, Lammers T, Peña Q, Ojha T. Hydrophobic ion pairing enables co-loading of water-soluble drugs in polymeric micelles. J Control Release 2025; 382:113748. [PMID: 40252978 DOI: 10.1016/j.jconrel.2025.113748] [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: 10/28/2024] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Polymeric micelles are widely used for the delivery of hydrophobic drugs. However, several highly potent and ubiquitously used anticancer drugs are water-soluble, complicating their (co-)formulation in polymeric micelles without having to chemically modify them. We here explore hydrophobic ion pairing to enable the co-delivery of the water-soluble anthracycline chemotherapy drug doxorubicin and the angiotensin II receptor antagonist telmisartan (a clinically used antihypertensive drug that has shown promising (pre-) clinical outcomes in combination with anthracyclines). We show that hydrophobic ion pairing of doxorubicin and telmisartan promotes the co-encapsulation of both drugs in π electron-stabilized [PEG-b-p(HPMAm-Bz)]-based polymeric micelles. The cytotoxic activity of doxorubicin is retained, and the dual drug-loaded micelles display enhanced antitumor activity in vivo as compared to the combination of the free drugs, while also exhibiting good tolerability. Taken together, this work provides proof-of-concept for hydrophobic ion pairing as a promising formulation strategy to promote multidrug nanomedicine and drug combination therapy.
Collapse
Affiliation(s)
- Armin Azadkhah Shalmani
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Rasika Daware
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Eva M Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Peter Boor
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Srinivas Banala
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alina Adams
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Josbert M Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Quim Peña
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Tarun Ojha
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands.
| |
Collapse
|
10
|
Yin D, Wu X, Chen X, Chen JL, Xia X, Wang J, Chen X, Zhu XM. Enhanced anticancer effect of carfilzomib by codelivery of calcium peroxide nanoparticles targeting endoplasmic reticulum stress. Mater Today Bio 2025; 32:101649. [PMID: 40160245 PMCID: PMC11953955 DOI: 10.1016/j.mtbio.2025.101649] [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: 01/09/2025] [Revised: 02/15/2025] [Accepted: 03/08/2025] [Indexed: 04/02/2025] Open
Abstract
Encouraged by the clinical success of proteasome inhibitors treating hematological malignancy, continuous efforts are being made to improve their efficacy and expand their applications to solid tumor therapy. In this study, liposomes were used to encapsulate the proteasome inhibitor carfilzomib (CFZ) and calcium peroxide (CaO2) nanoparticles for effective combination therapy targeting the interplay between calcium overload and oxidative stress. Low-dose CaO2 synergistically enhances the anticancer effect of CFZ in the human glioblastoma U-87 MG cells. The reactive oxygen species (ROS) generation and glutathione depletion by low-dose CaO2 complement CFZ-induced ubiquitinated protein accumulation further triggering endoplasmic reticulum (ER) stress leading to calcium overload and mitochondrial dysfunction. The liposome-based codelivery system is capable of transporting CFZ and CaO2 simultaneously to the tumor, and results in a superior antitumor effect in U-87 MG tumor-bearing mice compared with monotherapy. Taken together, CaO2 holds great potential to sensitize proteasome inhibitors in the treatment of solid tumors, and this work also presents a new combination therapy strategy targeting the crosstalk between proteasome inhibitors and oxidative stress for future cancer therapy.
Collapse
Affiliation(s)
- Dan Yin
- State Key Laboratory of Quality Research in Chinese Medicines & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Xuan Wu
- State Key Laboratory of Quality Research in Chinese Medicines & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Xu Chen
- State Key Laboratory of Quality Research in Chinese Medicines & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Jian-Li Chen
- State Key Laboratory of Quality Research in Chinese Medicines & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Xinyue Xia
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Xiao-Ming Zhu
- State Key Laboratory of Quality Research in Chinese Medicines & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Zhuhai MUST Science and Technology Research Institute, Macau University of Science and Technology, Zhuhai, Guangdong, 519099, China
| |
Collapse
|
11
|
Qiu C, Zhang W, Zhao Y, Han T, Yang W, Liu Y, Jin P, Chen J, Shuai X, Ren J, Huang P. Reprogramming Glucose Metabolism of Macrophage for Acute Liver Failure Therapy with Itaconate Lipo-Nanodrug. Adv Healthc Mater 2025; 14:e2500019. [PMID: 40249158 DOI: 10.1002/adhm.202500019] [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: 01/02/2025] [Revised: 03/17/2025] [Indexed: 04/19/2025]
Abstract
Acute liver failure (ALF) is a life-threatening disease featuring comprehensive inflammatory response and metabolic disorders in which macrophages exert central roles. A glucose metabolism mediator of macrophages, itaconate, has demonstrated potent anti-inflammatory efficacy in various diseases, implying that itaconate could work in treating ALF. However, systemic administration of itaconate may lead to immune disorder, making targeting the delivery of itaconate to the liver lesion area highly important. Herein, a liposomal nanodrug incorporating itaconate is developed, and its potential in treating acute liver failure in an ALF murine model established by LPS/D-GalN administration is tested. The nanodrug shows preferential liver accumulation to effectively alleviate LPS/D-GalN-induced hepatic histopathological injury by decreasing oxidative stress. Moreover, it reprograms the glucose metabolism of macrophages, resulting in macrophage repolarization toward the anti-inflammatory phenotype. Furthermore, western-blot and immunohistochemical assays verifies that the nanodrug may inhibit aerobic glycolysis of macrophages in an NRF2 and STING-dependent manner. These results underline the promise of the nanodrug for ALF treatment by reprogramming glucose metabolism.
Collapse
Affiliation(s)
- Chen Qiu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wei Zhang
- Department of Medical Ultrasound, Laboratory of Novel Optoacoustic (Ultrasonic) Imaging, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yanan Zhao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Tian Han
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wende Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yajing Liu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jie Ren
- Department of Medical Ultrasound, Laboratory of Novel Optoacoustic (Ultrasonic) Imaging, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
12
|
Cui L, Lou W, Wei X, Li M, Sun M, Wang S, Yang S, Zhang L, Zhou G, Li P, Qu L. Proanthocyanidin-Conjugated NIR-ΙΙ Nano-Prodrugs for Reversing Drug Resistance in Photothermal Therapy. Molecules 2025; 30:2334. [PMID: 40509226 PMCID: PMC12155667 DOI: 10.3390/molecules30112334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 06/18/2025] Open
Abstract
Targeting and multidrug resistance are the significant problems of current antitumor drugs, and these problems become the key factors in the design of nanomedicine. Herein, Au NRs and OPC-Au NPs were prepared via the hydroquinone seedless growth method and proanthocyanidin (OPC) one-pot method, and then pH-GSH-near-infrared ΙΙ (NIR-ΙΙ)-responsive nano-prodrugs Au/DOX-ss LNRs and OPC-Au/DOX-ss LNPs were designed by the encapsulation of doxorubicin prodrug DOX-ss with Au-S affinity and thermal-sensitive liposomes. Interestingly, OPC endowed OPC-Au NPs with reducibility and excellent performance in terms of particle size, zeta potential, encapsulation rate, and drug loading rate. In particular, the photothermal efficiencies of OPC-Au/DOX-ss LNPs increased to 59.22% under the 1064 nm NIR-ΙΙ irradiation. Compared with free DOX-ss and Lipid DOX-ss, the IC50 of OPC-Au/DOX-ss LNPs was decreased by 91.68% and 97.60%, respectively. Furthermore, the expression of P-gp in MCF-7/ADR was significantly inhibited (decreased by 65%). The potential of proanthocyanidin remodels the pH-GSH-NIR-ΙΙ responsiveness and drug resistance of OPC-Au/DOX-ss LNPs for breast cancer treatment in NIR-ΙΙ photodynamic/photothermal therapy.
Collapse
Affiliation(s)
- Lan Cui
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, China
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China;
| | - Weishuang Lou
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Xin Wei
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Mengdi Li
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Mengyao Sun
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Siyue Wang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Shuoye Yang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Lu Zhang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Guangzhou Zhou
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (L.C.); (W.L.); (X.W.); (M.L.); (M.S.); (S.W.); (S.Y.); (L.Z.)
| | - Peng Li
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, China
| | - Lingbo Qu
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China;
| |
Collapse
|
13
|
Wang X, Wang H, Dai Y, Lu R, Chen J, Kong Q. Application potential of injectable hydrogels in the post-surgical window period following tumor surgery. Int J Pharm 2025; 679:125754. [PMID: 40425056 DOI: 10.1016/j.ijpharm.2025.125754] [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: 04/12/2025] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Chemotherapy is one of the primary modalities for the treatment of malignant diseases. The outcomes, however, are different between tumors of various origins, which hinder clinical applications. The advantages of chemotherapies in patients with hematological lesions are more obvious than those seen in solid tumors. This might be attributed to the availability of drug concentration and exposure time. Based on this phenomenon, we hypothesis that localized drug administration is expected to be more potential for solid tumors, particularly for the residual tumors in post-operative "window period". The presence of residual tumors after surgical resection are the major factors leading to tumor recurrence after surgery. The methods of dealing with this problem are yet to be found. Conventional chemotherapies are scarcely applied in the post-surgical window period due to their unselected and unexpected side effects. This article studied the advantages and disadvantages of prominent formulations currently utilized in the field of local implantation in cancer treatment, with the notable superiority of injectable hydrogel platforms being most appealing. These platforms not only enhance wound healing of the patients with less side effects, during the "window period" following tumor surgery, but also effectively eradicate residual tumors by facilitating the establishment of a favorable microenvironment. Additionally, the challenges seen in this field and future directions are discussed, which is expected to provide insights for pharmaceutical professionals and clinical applications.
Collapse
Affiliation(s)
- Xilei Wang
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China; Taizhong Pharmaceutical Co., Ltd, No. 799 Yaocheng Avenue, Medical High-Tech Zone (Fudan University Taizhou Institute of Health Sciences), Taizhou 225326, China.
| | - Huan Wang
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Yue Dai
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Rong Lu
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Jingdi Chen
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Qingzhong Kong
- Taizhong Pharmaceutical Co., Ltd, No. 799 Yaocheng Avenue, Medical High-Tech Zone (Fudan University Taizhou Institute of Health Sciences), Taizhou 225326, China.
| |
Collapse
|
14
|
Yao ZW, Zhu H. Pharmacological mechanisms and drug delivery systems of Ginsenoside Rg3: a comprehensive review. Pharmacol Res 2025; 216:107799. [PMID: 40414584 DOI: 10.1016/j.phrs.2025.107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Ginsenoside Rg3, as one of the major active components of Panax ginseng, exhibits significant anti-tumor, anti-inflammatory, antioxidant, antidiabetic, hepatoprotective, wound healing and immunomodulatory pharmacological effects and has been developed as an adjuvant therapy in clinical practice. However, its poor water solubility and low permeability result in limited bioavailability, restricting its clinical application. This review systematically summarizes the pharmacological mechanisms of ginsenoside Rg3, including its anti-tumor effects through multiple signaling pathways that inhibit cancer cell proliferation, induce apoptosis, and suppress tumor angiogenesis; anti-inflammatory properties via the inhibition of NF-κB and related factors; antioxidant effects by increasing antioxidant enzyme levels and regulating the Nrf2 pathway; antidiabetic effects via the promotion of insulin secretion by inhibiting the MAPK pathway; hepatoprotective effects via the attenuation of hepatic inflammation through suppressing NF-κB phosphorylation; wound-healing-promoting effects via modulating the TGF-β/SMAD signaling pathway, and immunomodulatory activities through immune cell regulation and inhibition of PD-L1 glycosylation. Additionally, this review discusses the pharmacokinetic properties of Rg3, such as rapid oral absorption but low plasma concentration and bioavailability. Furthermore, this review highlights various drug delivery systems, including liposomes, solid dispersions, cyclodextrin inclusion complexes, microspheres, electrospun nanofiber membranes, hydrogels, nanoparticles, micelles, and microneedles, which have been developed to improve its physicochemical properties and enhance its therapeutic efficacy. By systematically summarizing the pharmacological mechanisms and formulation optimization strategies of Rg3, this review provides theoretical insights and technical support for future research and clinical translation.
Collapse
Affiliation(s)
- Zhong-Wei Yao
- Drug Clinical Trial Center, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, China
| | - He Zhu
- Drug Clinical Trial Center, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, China; Phase I Clinical Research Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| |
Collapse
|
15
|
Wang A, Wang X, Li D, Li A, He M, Yuan Y, Ye L, Liu J. A superior method for antitumor therapy and application: dual-ligand nanomedicines. J Mater Chem B 2025. [PMID: 40396464 DOI: 10.1039/d5tb00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Currently, nanomedicines have been widely applied in the treatment of various types of tumors. However, due to the complexity of the tumor microenvironment, conventional nanomedicines often exhibit poor efficacy, insufficient site specificity, and susceptibility to off-target effects. In contrast, dual-ligand nanomedicines demonstrate superior targeting ability and drug penetration in tumor therapy. These nanomedicines are equipped with two ligands on their surface, enabling targeting of specific receptors on the same or different cells. The specific binding between ligands and receptors significantly enhances the selectivity and targeting of dual-ligand nanomedicines towards tumors. This review systematically describes the preparation of dual-ligand nanomedicines, the influencing factors, and the types of delivered drugs, focusing on the application of dual-ligand nanomedicines in targeting the treatment of various tumors. We highlight the comprehensiveness of dual-ligand nanomedicines for the treatment of tumors, including glioblastoma, lung cancer, breast cancer, gastric cancer, and many other types of tumors. Finally, the possible challenges for the future development of dual-ligand nanomedicines in terms of preparation, clinic, and safety are further analyzed. We look forward to exploring dual-ligand nanomedicines in greater depth to provide references for their future development and clinical applications.
Collapse
Affiliation(s)
- Ailing Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Xuejun Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Mengyuan He
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Yingying Yuan
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
- College of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Ye
- School of Pharmacy and (R & D Center) Lab. for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau SAR, 999078, China.
| | - Jiyong Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| |
Collapse
|
16
|
Muteeb G, El-Morsy MT, Abo-Taleb MA, Mohamed SK, Khafaga DSR. Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems. Pharmaceutics 2025; 17:673. [PMID: 40430962 PMCID: PMC12114929 DOI: 10.3390/pharmaceutics17050673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive and prevalent liver cancer with a poor prognosis. Nanotechnology combined with natural products has emerged as a promising strategy to enhance HCC treatment efficacy. This review assesses the current literature on the application of nanotechnology in delivering natural products for HCC therapy. A comprehensive search was conducted in PubMed, Science Direct, Web of Science, and Google Scholar to identify relevant studies published up to the present articles focusing on nanotechnology-based drug delivery systems using natural products for HCC therapy, including different nanoparticle (NP) formulations and therapeutic interventions, were included. Natural products with anticancer properties have been encapsulated using various nanocarriers such as liposomes, polymeric nanoparticles, and quantum dots, which have improved drug stability, prolonged circulation time, and enhanced targeted delivery to HCC cells. These advancements have led to increased therapeutic efficacy and reduced side effects. Additionally, combining multiple natural products or integrating them with conventional therapies via nanocarriers enables personalized treatment approaches based on patient characteristics and molecular profiles. The integration of nanotechnology with natural products shows great potential for improving HCC treatment outcomes, representing a significant advancement in precision medicine for liver cancer and paving the way for more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Manar T. El-Morsy
- Bio-Nanotechnology Department, Faculty of Nanotechnology, Cairo University, Giza 12613, Egypt;
| | | | | | | |
Collapse
|
17
|
Xu C, Chen L, Liu G, Xu J, Lv W, Gao X, Xu P, Tang M, Wang Y, Zhao X, Nie G, Cheng K, Liu F. Tailoring an intravenously injectable oncolytic virus for augmenting radiotherapy. Cell Rep Med 2025; 6:102078. [PMID: 40233744 DOI: 10.1016/j.xcrm.2025.102078] [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: 08/23/2024] [Revised: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Oncolytic viruses (OVs) combined with radiotherapy (RT) show promise but are limited by challenges such as poor intravenous delivery and insufficient RT-induced DNA damage. In this study, an oncolytic adenovirus (AD) formulation, RadioOnco (AD@PSSP), is developed to improve delivery, infectivity, immune response, and RT efficacy. The multifunctional polyethylenimine (PEI)-selenium-polyethylene glycol (PEG) (PSSP) enhances intravenous delivery, shields the virus from rapid clearance, and enables targeted delivery to tumor sites after RT. The exposed PEI enhances the infectivity of AD through electrostatic interactions, thereby increasing DNA damage after RT by inhibiting the expression of DNA repair proteins, such as CHEK1 and CDK1. Furthermore, AD-PEI captures and delivers RT-induced tumor-released antigens to lymph nodes, activating robust anti-tumor immune responses. Animal model data demonstrate that RadioOnco overcomes RT resistance, targets distant metastases, and promotes long-term immunity, addressing metastasis and recurrence. In summary, this intravenously injectable OV enhances RT synergy through surface modification with multifunctional materials.
Collapse
Affiliation(s)
- Chen Xu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Peijun Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ming Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China.
| |
Collapse
|
18
|
Li X, Zhao X, Wang J, Xu B, Feng J, Huang W. High-Pressure Microfluidic Homogenization Improves the Stability and Antioxidant Properties of Coenzyme Q10 Nanoliposomes. BIOLOGY 2025; 14:568. [PMID: 40427757 PMCID: PMC12108558 DOI: 10.3390/biology14050568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025]
Abstract
Coenzyme Q10 is a natural antioxidant with anti-tumor and mitochondrial protective effects. However, its unstable physicochemical properties and large molecular weight result in low bioavailability. This study aimed to develop an effective technique for constructing nanoliposomes to improve the physicochemical properties of CoQ10 by using high-pressure microfluidic homogenization. Liposomes were prepared using the ethanol injection method and homogenized by high-pressure microfluidics to optimize their physicochemical properties. Liposome morphology and microstructure were observed via transmission electron microscopy (TEM). The particle size distribution, polydispersity index (PDI), and encapsulation efficiency were assessed, while effects on cell viability and antioxidant properties were investigated in HepG2 cells. The results indicate that the prepared liposomes exhibit favorable characteristics, including high encapsulation efficiency (>96%) and low PDI (<0.3), indicating uniform particle size distribution and good stability. The storage stability of liposomes at room temperature was significantly enhanced compared to liposomes not subjected to high pressure homogenization. In vitro cell experiments confirmed the liposomes' non-cytotoxicity and substantial antioxidant activity, ensuring their safety for biomedical applications. This study introduced a liposome preparation method combining ethanol injection and high-pressure microfluidic homogenization, offering a novel approach for liposome modification with potential for development and application in innovative drug delivery systems and antioxidant therapy.
Collapse
Affiliation(s)
- Xinyu Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Xingyu Zhao
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Jing Wang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China;
| | - Baoshun Xu
- Kangcare Bioindustry Co., Ltd., Nanjing 210006, China;
| | - Jin Feng
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Wuyang Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China;
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
19
|
Zheng H, Chen Y, Luo W, Han S, Sun M, Lin M, Wu C, Gao L, Xie T, Kong N. Integration of active ingredients from traditional Chinese medicine with nano-delivery systems for tumor immunotherapy. J Nanobiotechnology 2025; 23:357. [PMID: 40382641 PMCID: PMC12085060 DOI: 10.1186/s12951-025-03378-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Tumor immune escape presents a significant challenge in cancer treatment, characterized by the upregulation of immune inhibitory molecules and dysfunction of immune cells. Tumor immunotherapy seeks to restore normal anti-tumor immune responses to control and eliminate tumors effectively. The active ingredients of traditional Chinese medicine (TCM) demonstrate a variety of anti-tumor activities and mechanisms, including the modulation of immune cell functions and inhibiting tumor-related suppressive factors, thereby potentially enhancing anti-tumor immune responses. Furthermore, nano-delivery systems function as efficient carriers to enhance the bioavailability and targeted delivery of TCM active ingredients, augmenting therapeutic efficacy. This review comprehensively analyzes the impact of TCM active ingredients on the immune system and explores the synergistic application of nano-delivery systems in combination with TCM active ingredients for enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Mengjuan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Min Lin
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Chenghan Wu
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Lili Gao
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China.
| | - Tian Xie
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
20
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025; 17:11864-11893. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
21
|
Lin Y, Wu H, Wang J, He W, Hou J, Martin VT, Zhu C, Chen Y, Zhong J, Yu B, Lu A, Guan D, Qin G, Chen W. Nicotinamide Adenine Dinucleotide-Loaded Lubricated Hydrogel Microspheres with a Three-Pronged Approach Alleviate Age-Related Osteoarthritis. ACS NANO 2025; 19:17606-17626. [PMID: 40315404 PMCID: PMC12080321 DOI: 10.1021/acsnano.5c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
Chondrocyte senescence, synovitis, and decreased level of lubrication play pivotal roles in the pathogenesis of age-related osteoarthritis (AROA). However, there are currently no effective therapeutic interventions capable of altering the progression of OA until it reaches advanced stages, necessitating joint replacement. In this study, lubricious and drug-loaded hydrogel microspheres were designed and fabricated by utilizing microfluidic technology for radical polymerization of chondroitin sulfate methacrylate and incorporating nicotinamide adenine dinucleotide (NAD)-loaded liposomes modified with lactoferrin that are positively charged. Mechanical, tribological, and drug release analyses demonstrated enhanced lubrication properties and an extended drug dissemination time for the NAD@NPs@HM microspheres. In vitro assays unveiled the ability of NAD@NPs@HM to counteract chondrocyte senescence. RNA sequencing analysis, untargeted metabolomics analysis, and in vitro experiments on macrophages revealed that NAD@NPs@HM can regulate the metabolic reprogramming of synovial macrophages, promoting their repolarization from the M1 to M2 phenotype, thereby alleviating synovitis. Intra-articular injection of NAD@NPs@HM in aged mice reduced the mechanisms associated with AROA. These results suggest that NAD@NPs@HM may provide extended drug release, improved joint lubrication leading to better gait, and attenuation of AROA pathogenic processes, indicating its potential as a therapeutic approach for AROA.
Collapse
Affiliation(s)
- Yanpeng Lin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hangtian Wu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Jun Wang
- School
of Animal Science and Technology, Foshan
University, Foshan, Guangdong 528231, People’s Republic of China
| | - Wanling He
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jiahui Hou
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Vidmi Taolam Martin
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Chencheng Zhu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yupeng Chen
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Junyuan Zhong
- Department
of Medical Imaging, Ganzhou People’s
Hospital, Ganzhou, Jiangxi 341000, P. R. China
| | - Bin Yu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Aiping Lu
- Institute
of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong 999077, P. R. China
- Guangdong-Hong
Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510515, P. R. China
| | - Daogang Guan
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Genggeng Qin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Weiguo Chen
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
22
|
Yadavalli VK. The convergence of nanomanufacturing and artificial intelligence: trends and future directions. NANOTECHNOLOGY 2025; 36:222001. [PMID: 40311640 DOI: 10.1088/1361-6528/add304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 05/01/2025] [Indexed: 05/03/2025]
Abstract
The integration of nanoscale production processes with Artificial intelligence (AI) algorithms has the potential to open new frontiers in nanomanufacturing by accelerating development timelines, optimizing production, reducing costs, enhancing quality control, and improving sustainability. Such changes are already underway with digital and cyber-physical technologies becoming increasingly intertwined with 'smart' manufacturing and industrial processes today. With the nanomanufacturing sector focused on the scalable production of complex (nano)materials, (nano)devices, and biologics, AI and its sub-fields, including machine learning (ML), are positioned to be key enablers of efficiency and innovation. In this topical review, we briefly explore the current state-of-the-art of how AI and ML techniques can be employed within nanomanufacturing. We discuss from a birds-eye perspective, the impact of AI/ML on various stages of the production lifecycle, and examine future opportunities and challenges. Key areas include computational design and discovery, process optimization, predictive maintenance, and quality assurance/defect detection. Further, challenges in implementation, process complexity, and ethical and regulatory considerations are explored in light of the increasing reliance on data-driven approaches for manufacturing.
Collapse
Affiliation(s)
- Vamsi K Yadavalli
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W Main Street, Richmond, VA 23284, United States of America
| |
Collapse
|
23
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [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: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
24
|
Kocas M, Yamashita F, Comoglu T, Zhang Q. Enhancing Intracellular Uptake of Ivermectin through Liposomal Encapsulation. AAPS PharmSciTech 2025; 26:123. [PMID: 40316874 DOI: 10.1208/s12249-025-03113-8] [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: 11/19/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025] Open
Abstract
Ivermectin (IVM), an antiparasitic drug approved by the Food and Drug Administration (FDA), is widely used to treat several neglected tropical diseases, including onchocerciasis, helminthiases, and scabies. Additionally, IVM has shown potential as a potent inhibitor of certain RNA viruses, such as SARS-CoV-2. However, IVM is highly hydrophobic, essentially insoluble in water, which limits its bioavailability and therapeutic effectiveness. The use of liposomes as drug carriers offers several advantages, including enhanced solubility for lipophilic drugs, passive targeting of immune system cells, sustained release, and improved tissue penetration. To address the limitations of IVM, including its poor solubility and bioavailability, liposomal formulations were developed using a combination of soyphosphatidylcholine (SPC), dioleylphosphatidylcholine (DOPC), cholesterol (Ch), and diethylphosphate (DCP) in two distinct molar ratios (1.85:1:0.15 and 7:2:1) via the ethanol injection method. The physicochemical properties of the placebo and IVM-loaded liposomes were extensively characterized in our earlier study, including the particle size, polydispersity index, and zeta potential. The present work adds a deeper level of investigation into how to effect cellular uptake and cytotoxicity in vitro of both free IVM and IVM-loaded liposomes in Vero E6 cells. The half-maximal cytotoxic concentrations (CC50) for free IVM and IVM-loaded liposomes were 10 μM and > 110 μM, respectively and the cellular uptake of IVM-loaded liposomes ranged from 13 to 60%, whereas free IVM showed a significantly lower uptake of only 2%. These results demonstrate that liposomal encapsulation effectively enhances IVM's cellular uptake while reducing its cytotoxicity, thus offering a promising strategy for improving the effectiveness of IVM.
Collapse
Affiliation(s)
- Meryem Kocas
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Selcuk University, Selçuklu, Konya, 42130, Turkey
- Graduate School of Health Sciences, Ankara University, Dışkapı, Ankara, 06610, Turkey
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara University, Yenimahalle, Ankara, 06560, Turkey
- Graduate School of Pharmaceutical Sciences, Department of Drug Delivery Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Fumiyoshi Yamashita
- Graduate School of Pharmaceutical Sciences, Department of Quantitative Pharmaceutics, Kyoto University, Kyoto, 606-8501, Japan
- Graduate School of Pharmaceutical Sciences, Department of Drug Delivery Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Tansel Comoglu
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara University, Yenimahalle, Ankara, 06560, Turkey.
| | - Qiyue Zhang
- Graduate School of Pharmaceutical Sciences, Department of Quantitative Pharmaceutics, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
25
|
Xiao Y, Tong Z, Xu H, Jia Z, Wang C, Cao Y, Song L, Hao S, Yang J, Zhou Y, Xie Y, Wu P, He T, Wu Y, Petersen RB, Peng A, Zhang C, Chen H, Zheng L, Huang K. A rationally designed injury kidney targeting peptide library and its application in rescuing acute kidney injury. SCIENCE ADVANCES 2025; 11:eadt3943. [PMID: 40315322 PMCID: PMC12047437 DOI: 10.1126/sciadv.adt3943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Acute kidney injury (AKI) has high incidence and mortality rates. Present treatments are mostly symptomatic and cause side effects due to systemic distribution; thus, targeted kidney drug delivery is desired. Transmembrane kidney injury molecule-1 (KIM1) is expressed at low levels in normal kidneys but markedly up-regulated following injury, making it an ideal marker/target for injured kidneys. Here, assisted by AlphaFold, we constructed a library of 1885 peptides that target the extracellular Ig V domain of KIM1 based on interacting fragments from 47 potential KIM1 binding proteins followed by systemic optimization according to their binding energies with KIM1. Experimental validation of top candidates (TKP 1-5) demonstrated that TKP 4 efficiently targeted injured renal cells/kidneys, with its specificity demonstrated in KIM1 knockout cells/mice. TKP 4-decorating liposomes were loaded with nystatin, a renal-protective compound with systemic side effects, and efficiently targeted injured mouse kidneys and alleviated AKI. This work establishes a virtual platform to screen/identify drug delivery candidates with broad research/therapeutic potentials.
Collapse
Affiliation(s)
- Yushuo Xiao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhijian Tong
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huidie Xu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhouyan Jia
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Wang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Cao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liangliang Song
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Siyu Hao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yihao Zhou
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yunhao Xie
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Peng Wu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Tong He
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yancai Wu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Robert B. Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI 48859, USA
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan 430070, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zheng
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
26
|
Zhang H, Wang Y, Geng X, Dong M, Liu Z, Sun C, Yu K, Xin W, Xu Y, Xu N, Liu W. ANG-Modified Liposomes Coloaded With α-Melittin and Resveratrol Induce Apoptosis and Pyroptosis in Glioblastoma Cells by Impeding Wnt/β-Catenin Signaling. CNS Neurosci Ther 2025; 31:e70437. [PMID: 40400263 PMCID: PMC12095925 DOI: 10.1111/cns.70437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025] Open
Abstract
MAIN PROBLEM Glioblastoma (GB) is one of the most prevalent and devastating types of brain cancer for which efficient treatments are currently lacking because of limitations such as antitumor efficacy, brain delivery, tumor selectivity, and drug resistance. A promising strategy to overcome these obstacles is developing anticancer agents that can be delivered to GB tissues to inhibit tumors with low toxicity to normal brain tissue. METHODS We developed liposomes encapsulating resveratrol (RES), a polyphenolic compound, and α-melittin (α-MEL), which is composed of melittin conjugated with an amphiphilic α-helical peptide at its N-terminus. RES-, α-MEL-, and α-MEL-RES-loaded liposomes (Lips) were modified with Angiopep-2 (ANG). The effects of the above liposomes on GB cells were assessed, and the possible mechanisms were analyzed. RESULTS ANG-modified α-MEL-RES-Lips treatment facilitated the passage of these agents through the blood-brain barrier (BBB), increased tumor targeting, and significantly reduced α-MEL-associated hemolysis. The combined management of α-MEL with RES impeded GB cell growth and prolonged the lifespan of GB tumor-bearing model mice. α-MEL-RES-Lips treatment triggered GB cell apoptosis and induced pyroptosis-associated protein expressions of gasdermin-D (GSDMD), gasdermin E (GSDME), cleaved caspase 3, and NLR family pyrin domain containing 3 (NLRP3), and inhibited epithelial-mesenchymal transition (EMT) by modulating the Wnt/β-catenin signaling pathway. CONCLUSION ANG-modified α-MEL-RES-Lips might be a potential nanosystem for GB therapy, and polyphenolic compounds combined with antimicrobial peptides may promote the induction of apoptosis, pyroptosis, and the apoptosis-pyroptosis switch in GB.
Collapse
Affiliation(s)
- Hai‐Qian Zhang
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Yan Wang
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Xiao Geng
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Mingxin Dong
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Ziwei Liu
- The Second Hospital of Jilin UniversityChangchunChina
| | - Chengbiao Sun
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Kaikai Yu
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| | - Wenwen Xin
- State Key Laboratory of Pathogen and BiosecurityInstitute of Microbiology and Epidemiology, AMMSBeijingChina
| | - Ye Xu
- Basic College of MedicineJilin Medical UniversityJilinChina
| | - Na Xu
- Basic College of MedicineJilin Medical UniversityJilinChina
| | - Wensen Liu
- Changchun Veterinary Research InstituteChinese Academy of Agricultural ScienceChangchunChina
| |
Collapse
|
27
|
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
|
28
|
Deng L, Liao J, Liu W, Liang X, Zhou R, Jiang Y. Research Advances for Protein-Based Pickering Emulsions as Drug Delivery Systems. Pharmaceutics 2025; 17:587. [PMID: 40430878 PMCID: PMC12114756 DOI: 10.3390/pharmaceutics17050587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Nanotechnologically engineered protein-based carriers have attracted considerable attention in the pharmaceutical field due to the advantages of superior biocompatibility, tunability and good emulsifying properties. Recently, protein-based Pickering emulsions (PPEs) systems with multi-level structures have been introduced as innovative colloidal delivery systems for advanced drug encapsulation, protection, delivery and controlled release. Natural source protein nanoparticles are promising candidates to provide a wide range of functional performances and interfacial properties in the preparation and stabilization of Pickering emulsions. Herein, this review summarizes the development of PPEs in drug delivery systems, focusing on the research progress concerning the aspects of protein particle preparation methods, formation mechanisms and rational design principles, emphasizing the relationship between protein particle structure and functional properties. To further understand the interfacial behavior in protein nanoparticle stabilized emulsion, the mesoscopic dissipative particle dynamics (DPD) simulations were discussed, which bridges the gaps between macroscopic time and length scales, as well as molecular-scale simulations on particles and oil/water interface systems. The structure-effect relationship between the tunable physicochemical properties of protein-based interface design, which leads to the effective loading, stimuli-responsiveness for the controlled release and multiple delivery, was then summarized. Finally, the opportunities and challenges for the future development of PPEs for drug delivery are discussed. This review aims to provide a reference for the further application of PPEs as advanced drug delivery systems.
Collapse
Affiliation(s)
- Long Deng
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
- School of Materials Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Junqiu Liao
- School of Materials Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Weiqi Liu
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xiaoxiao Liang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Rujin Zhou
- School of Chemical Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Yanbin Jiang
- School of Chemical Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China
| |
Collapse
|
29
|
Zhou Q, Ma H, Xu H, Bai Y, Tan C, Su Y, Li Y, Ying X, Zheng Y. Effect of Prodrug Activation Rate on In Vivo Drug Release and Antitumor Efficacy of SN38-Prodrug-Entrapped Liposomes. Mol Pharm 2025. [PMID: 40293415 DOI: 10.1021/acs.molpharmaceut.5c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The aim of this study is to investigate the effects of prodrug activation rates on the in vivo drug release and antitumor activity of prodrug-entrapped liposomes. We performed such an investigation using two liposomes encapsulating hydrophilic SN38-glutathione (GSH) prodrugs (linked at 10- and 20-hydroxy of SN38) with different activation rates. The results showed that SN38-GSH was first released from liposomes and, consequently, activated into SN38. The SN38-GSH release rate from liposomes was similar, but the 10SN38-GSH activated much faster than 20SN38-GSH (t1/2, < 1 min versus ∼60 min) in plasma. Such different activation rates did not influence the prodrug's pharmacokinetics and biodistribution, but the fast prodrug activation rate resulted in 4-6-fold higher SN38 concentration in various organs and led to more potent antitumor efficacy. By contrast, the slowly activated SN38-GSH liposomes failed to exhibit potent antitumor activity, even at the maximum tolerance dose. Our data illustrated how the prodrug activation rate influences in vivo drug release and antitumor activity of prodrug-loaded liposomes, suggesting that sufficient prodrug activation is a prerequisite for potent prodrug-loaded liposomes.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu 610051, China
| | - Hailong Ma
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Hongling Xu
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yuyang Bai
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Chang Tan
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yangxue Su
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xue Ying
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yaxin Zheng
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
30
|
Qiu X, Xie J, Mei J. Recent Advances in the Applications and Studies of Polysaccharide-, Protein-, and Lipid-Based Delivery Systems in Enhancing the Bioavailability of Capsaicin-A Review. Polymers (Basel) 2025; 17:1196. [PMID: 40362978 PMCID: PMC12073809 DOI: 10.3390/polym17091196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
The primary active ingredient in capsicum is capsaicin. However, capsaicin bioavailability is low due to its restricted water solubility, and its potent spicy flavor will further restrict its use in food. This paper provides a complete overview of capsaicin. The biological activity of capsaicin and its impacts on metabolism in vivo are described. To increase capsaicin stability and bioavailability, several capsaicin-based delivery systems, including liposomes, double emulsions, nanoparticle mesosystems, and multiple systems made of distinct hydrocolloids, are covered in this review. Finally, potential uses for food preservation are introduced in line with this. Numerous delivery systems introduced in this review have effectively solved the problems of poor water solubility and poor bioavailability of capsaicin. Although capsaicin has potential uses in food preservation, there is little research on its application in functional food development. More innovative capsaicin-based delivery methods should be established, and more capsaicin-based applications should be developed in the future.
Collapse
Affiliation(s)
- Xiang Qiu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China;
| | - Jing Xie
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China;
- National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic Product Processing and Preservation, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Professional Technology Service Platform on Cold Chain Equipment Performance and Energy Saving Evaluation, Shanghai Ocean University, Shanghai 201306, China
| | - Jun Mei
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China;
- National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic Product Processing and Preservation, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Professional Technology Service Platform on Cold Chain Equipment Performance and Energy Saving Evaluation, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
31
|
Zhou Q, Jin M, Cui Y, Jiang S, Shang P, Li L. Advances in pharmacological activity and drug delivery systems of vinca alkaloids. Nat Prod Res 2025:1-21. [PMID: 40276897 DOI: 10.1080/14786419.2025.2494625] [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: 09/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
Vinca alkaloids (VAs), derived from the Catharanthus roseus, are naturally occurring or semi-synthetic alkaloids primarily used in the treatment approach for diverse types of cancer. They have shown significant efficacy in treating leukaemia, Hodgkin's lymphoma. Nevertheless, their clinical application is considerably limited owing to the severe side effects, low bioavailability, and multidrug resistance (MDR). Over the past few years, drug delivery systems such as nanoparticles, liposomes, and solid lipid nanoparticles (SLN) have been shown to improve the pharmacokinetic properties and tumour targeting of VAs. The use of multiple drugs in combination can also reduce the adverse reactions of VAs and significantly enhance their efficacy, thereby broadening their application. This review introduces the main pharmacologically active components of VAs, summarises their chemotherapeutic effects, and provides a statistical overview and analysis of recent research progress in VAs drug delivery technologies, offering a reference for further research and clinical application of VAs in cancer treatment.
Collapse
Affiliation(s)
- Quanying Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Jin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yulong Cui
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siqi Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizhao Shang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lingjun Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
32
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
33
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
34
|
Al-Shihabi AM, Al-Mohaya M, Haider M, Demiralp B. Exploring the promise of lipoplexes: From concept to clinical applications. Int J Pharm 2025; 674:125424. [PMID: 40043964 DOI: 10.1016/j.ijpharm.2025.125424] [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: 10/07/2024] [Revised: 02/02/2025] [Accepted: 02/28/2025] [Indexed: 03/09/2025]
Abstract
Lipoplexes are non-viral lipid vectors that effectively form complexes with genetic material, positioning them as promising alternatives to viral vectors in gene therapy. Their advantages include lower toxicity, reduced immunogenicity, improved targetability, and ease of large-scale production. A typical lipoplex is composed of cationic lipids, neutral lipids, and anionic nucleic acids (e.g., DNA, mRNA, miRNA, siRNA, shRNA). Neutral lipids play an auxiliary role and are often used as transfection enhancers. Enhancing lipoplex efficiency often involves modifying the cationic lipid structure through functional groups like PEG polymers and targeting ligands. The assembly of lipoplexes occurs spontaneously. This process involves the binding of the positively charged polar head group of the cationic lipid to the negatively charged DNA spontaneously as a result of electrostatic interaction, then irreversible rearrangement and condensation of the lipoplex occurs to form either lamellar or hexagonal structures. The transfection process encompasses several steps: cellular entry, endosomal escape and cargo release, cytoplasmic trafficking, and nuclear entry. The physicochemical and biological properties of lipoplexes are influenced by factors such as lipid structure, charge ratio, and environmental conditions. Despite certain limitations like low gene transfer efficiency and rapid clearance by serum proteins, lipoplexes show promise for clinical applications. They can be administered through various routes, offering potential treatments for diseases such as cancer, bone damage, infection, and cystic fibrosis. The study aims to examine the potential of lipoplexes as a promising vehicle for delivering therapeutic agents and their progression from theoretical concepts to practical clinical applications.
Collapse
Affiliation(s)
- Alaa M Al-Shihabi
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mazen Al-Mohaya
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates.
| | - Burcu Demiralp
- Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey.
| |
Collapse
|
35
|
Wei HX, Liu MH, Wang TY, Shih MH, Yu J, Yeh YC. Fabrication of pH- and Ultrasound-Responsive Polymeric Micelles: The Effect of Amphiphilic Block Copolymers with Different Hydrophilic/Hydrophobic Block Ratios for Self-Assembly and Controlled Drug Release. Biomacromolecules 2025; 26:2116-2130. [PMID: 40067950 PMCID: PMC12004527 DOI: 10.1021/acs.biomac.4c01202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Stimuli-responsive polymeric vehicles can change their physical or chemical properties when exposed to internal or external triggers, enabling precise spatiotemporal control of drug release. Nevertheless, systematic research is lacking in preparing dual stimuli-responsive amphiphilic block copolymers with different hydrophilic/hydrophobic block ratios in forming self-assembled structures. Here, we synthesized two types of block copolymers consisting of the hydrophobic segments (i.e., pH-responsive 2-(diethylamino)ethyl methacrylate (DEA) and ultrasound-responsive 2-methoxyethyl methacrylate (MEMA)) and hydrophilic poly(ethylene glycol) methyl ether (mPEG) segments, forming mPEGX-b-P(DEAY-co-MEMAZ). These amphiphilic block copolymers can self-assemble to form polymeric micelles, and their structures (e.g., size) and properties (e.g., critical vesicle concentration, stability, stimuli-responsiveness to pH and ultrasound, drug loading efficiency, and controlled drug release performance) were thoroughly investigated. In vitro cell studies further demonstrate that ultrasound can efficiently trigger drug release from polymeric micelles, emphasizing their potential for controlled drug delivery in therapeutic applications.
Collapse
Affiliation(s)
- Hong-Xiang Wei
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| | - Ming-Hsin Liu
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Tzu-Ying Wang
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| | - Meng-Hsiu Shih
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Jiashing Yu
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Yi-Cheun Yeh
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
36
|
Tang Y, Chen Y, Qi YD, Yan HY, Peng WA, Wang YQ, Huang QX, Liu XH, Ye JJ, Yu Y, Zhang XZ, Huang C. Engineered Bdellovibrio bacteriovorus enhances antibiotic penetration and biofilm eradication. J Control Release 2025; 380:283-296. [PMID: 39894266 DOI: 10.1016/j.jconrel.2025.01.075] [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: 08/31/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Biofilms increase bacterial resistance to antibiotics, as conventional antibiotic doses are often ineffective at penetrating the biofilm matrix to eliminate bacteria. Recent research has shown that the Gram-negative predator bacterium Bdellovibrio bacteriovorus can penetrate Gram-positive bacterial biofilms during its predation phase and benefit from them without direct predation. Here, based on the penetration ability of B. bacteriovorus, we constructed antibiotic-loaded liposome-engineered B. bacteriovorus as a drug delivery strategy for biofilm-related diseases. As a "living antibiotic," B. bacteriovorus can prey on Gram-negative bacteria, penetrate biofilms, and disrupt their dense structure. During this process, the rapid movement of B. bacteriovorus enhances the delivery of antibiotic-loaded liposomes into the biofilm, promoting efficient antibiotic release and improving biofilm eradication. Our findings demonstrate that this engineered living antibiotic strategy significantly improves the control and removal of bacterial biofilms, accelerates the elimination of dental plaque, promotes wound healing, and holds promise as a novel platform for treating biofilm-related infections.
Collapse
Affiliation(s)
- Ying Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China
| | - Yong-Dan Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Hui-Yi Yan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China
| | - Wen-An Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China
| | - Yu-Qiang Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China
| | - Qian-Xiao Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xin-Hua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yun Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University. Wuhan 430079, PR China.
| |
Collapse
|
37
|
Li X, Wu F, Yu D, Su X, Wang K, Huang Z, Lu Z. Archaea-inspired deoxyribonuclease I liposomes prevent multiple organ dysfunction in sepsis. J Control Release 2025; 380:1109-1126. [PMID: 39986474 DOI: 10.1016/j.jconrel.2025.02.050] [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: 11/23/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Neutrophil extracellular traps (NETs) and circulating cell-free DNA (cfDNA) are pivotal in driving excessive inflammation and organ damage during sepsis, with their levels correlating positively with sepsis severity in both patients and murine models. Despite the ability of deoxyribonuclease I (DNase I) to degrade NETs and cfDNA, its short half-life and rapid degradation limit its therapeutic effectiveness. To address this challenge, we developed a methyl-branched liposome fused with a red blood cell membrane for the systemic delivery of DNase I (DNase I/Rm-Lipo). The efficacy of DNase I/Rm-Lipo was evaluated in the stimulated immune cells and septic model. The data confirmed that DNase I/Rm-Lipo efficiently removed excess NETs and cfDNA in activated neutrophils. Following injection, DNase I/Rm-Lipo exhibited an extended circulation time, effectively suppressing neutrophil activation and regulating macrophage polarization to mitigate inflammation and prevent organ dysfunction in septic mice. These findings highlight the therapeutic potential of DNase I/Rm-Lipo as a promising candidate for sepsis management by targeting the degradation of NETs and cfDNA.
Collapse
Affiliation(s)
- Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Fan Wu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Dedong Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiayi Su
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Kaikai Wang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Zhiwei Huang
- Central Laboratory, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China.
| |
Collapse
|
38
|
Lin M, Tang K, Zheng W, Zheng S, Hu K. Curcumin delivery system based on a chitosan-liposome encapsulated zeolitic imidazolate framework-8: a potential treatment antioxidant and antibacterial treatment after phacoemulsification. Biomed Mater 2025; 20:035013. [PMID: 40081008 DOI: 10.1088/1748-605x/adc05c] [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: 12/25/2024] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
Curcumin is a natural polyphenol extracted from plants that can interact with various molecular targets, including antioxidant, antibacterial, anticancer, and anti-aging activities. Due to its variety of pharmacological activities and large margin pf safety, curcumin has been used in the prevention and treatment of various diseases, such as Alzheimer's, heart, and rheumatic immune diseases. To develop curcumin eye drops that can be used as antioxidant and antibacterial agents after phacoemulsification, we have designed a nano-based drug delivery system to improve curcumin bioavailability and duration of action. We successfully prepared zeolitic imidazolate framework-8 (ZIF-8) coated with chitosan-liposome (Cur@ZIF-8/CS-Lip) for curcumin delivery. It can release curcumin for over 20 hin vitroand exhibits excellent biosafety, antioxidant, and antibacterial activities. Therefore, we hypothesized that Cur@ZIF-8/CS-Lip could reduce the incidence of oxidative stress and infection after cataract surgery.
Collapse
Affiliation(s)
- Meiting Lin
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Kunyuan Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Wendi Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Shijie Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| |
Collapse
|
39
|
Yun Y, An J, Kim HJ, Choi HK, Cho HY. Recent advances in functional lipid-based nanomedicines as drug carriers for organ-specific delivery. NANOSCALE 2025; 17:7617-7638. [PMID: 40026004 DOI: 10.1039/d4nr04778h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Lipid-based nanoparticles have emerged as promising drug delivery systems for a wide range of therapeutic agents, including plasmids, mRNA, and proteins. However, these nanoparticles still encounter various challenges in drug delivery, including drug leakage, poor solubility, and inadequate target specificity. In this comprehensive review, we present an in-depth investigation of four distinct drug delivery methods: liposomes, lipid nanoparticle formulations, solid lipid nanoparticles, and nanoemulsions. Moreover, we explore recent advances in lipid-based nanomedicines (LBNs) for organ-specific delivery, employing ligand-functionalized particles that specifically target receptors in desired organs. Through this strategy, LBNs enable direct and efficient drug delivery to the intended organs, leading to superior DNA or mRNA expression outcomes compared to conventional approaches. Importantly, the development of novel ligands and their judicious combination holds promise for minimizing the side effects associated with nonspecific drug delivery. By leveraging the unique properties of lipid-based nanoparticles and optimizing their design, researchers can overcome the limitations associated with current drug delivery systems. In this review, we aim to provide valuable insights into the advancements, challenges, and future directions of lipid-based nanoparticles in the field of drug delivery, paving the way for enhanced therapeutic strategies with improved efficacy and reduced adverse effects.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Jeongmin An
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hyun Joong Kim
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, USA
| | - Hyeon-Yeol Cho
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| |
Collapse
|
40
|
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
|
41
|
Hua Q, Wang Q, Wang X, Jiang X, Gong M, Li J, Li T, Wang X, Cao X, Yu J, Toreniyazov E, Zong B, Xu X, Shi F, Adu-Frimpong M. Preparation of PEG-modified isoquercitrin liposomes and anti-chronic kidney disease research. J Liposome Res 2025:1-15. [PMID: 40125927 DOI: 10.1080/08982104.2025.2480782] [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: 12/01/2024] [Revised: 01/27/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
The clinical application of Isoquercitrin (IQ) is limited by its low water solubility and short retention time in the body, despite its diverse pharmacological effects. To address these issues, we prepared polyethylene glycol (PEG)-modified IQ liposomes (IQ-L) using the thin film dispersion method and optimized the formulation through a combination of One Factor at a Time (OFAT) method and response surface experiments. Characterization of the IQ-L that was prepared using the optimal formulation revealed a particle size of 185.48 nm, a polydispersity index of 0.252, a zeta potential of -33.88 mV, and an impressive encapsulation efficiency of 97.84%. In vitro release studies showed a significantly higher cumulative release rate for IQ-L compared to free IQ. Pharmacokinetic evaluations in rats demonstrated a 4.54-fold increase in the area under the concentration-time curve, a 1.63-fold prolongation of the half-life, and a 2.07-fold increase in peak concentration for IQ-L compared to unmodified IQ. Moreover, assessments of renal function in a mouse model indicated promising therapeutic effects. In summary, the PEG-modified liposome system greatly improved the solubility and in vivo retention time of IQ, thus making it a potential clinical agent for the treatment of chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Qinyang Hua
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xue Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xia Jiang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mingjie Gong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiaying Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tingyuan Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaowen Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | | | - Bin Zong
- Hospital of Chinese Traditional and Western Medicine, Zhenjiang, Jiangsu, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feng Shi
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, UK Ghana
| |
Collapse
|
42
|
Liu Y, Xie Y, Chen Y, Duan J, Bao C, Wang J, Feng H, Wang M, Ren Y, Li P, Luo Q, Xu J, Jiang M, Men Y, Wu Y, Li J, Wang G, Lu W. A protease-cleavable liposome for co-delivery of anti-PD-L1 and doxorubicin for colon cancer therapy in mice. Nat Commun 2025; 16:2854. [PMID: 40128211 PMCID: PMC11933685 DOI: 10.1038/s41467-025-57965-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Immune checkpoint blockade therapy using programmed cell death 1 (PD1) or programmed death ligand 1 (PD-L1) has made significant progress in the treatment of advanced cancers, with some patients achieving long-term remission without clinical recurrence. However, only a minority of colon cancer patients respond to the therapy. Here, we report a protease-cleavable anti-PD-L1 antibody liposome, eLipo anti-PD-L1, for enhancing colon cancer therapy. In vivo, eLipo anti-PD-L1 is cleaved by legumain at colon cancer site into pegylated anti-PD-L1 and cancer-homing doxorubicin liposome. Functional assessments show cancer-targeting, legumain-responding, tumor-penetrating, and immune-activating effects, as well as efficacy in treating colon cancer-bearing mice in vivo. Further mechanistic analysis implicates genes related to T cell differentiation and T cell receptor signaling as potential molecular mediators. Lastly, human colorectal cancer tissue evaluations verify expressions of PD-L1 and legumain, hinting a potential translatability. Our study thus suggests that eLipo anti-PD-L1 may be a feasible vector for co-delivery of immunochemotherapy for colon cancer.
Collapse
Affiliation(s)
- Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jinling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hexuan Feng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Mengjie Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuxin Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jiarui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Min Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yanchen Men
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yang Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Guiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
| |
Collapse
|
43
|
Mlachkova A, Dosseva-Panova V, Maynalovska H, Pashova-Tasseva Z. Nanoparticles as Strategies for Modulating the Host's Response in Periodontitis Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:476. [PMID: 40214523 PMCID: PMC11990483 DOI: 10.3390/nano15070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025]
Abstract
Periodontitis is a widespread disease, associated with challenges both in its diagnosis and in selecting from various therapeutic approaches, which do not always yield the expected success. This literature review was conducted to explore diverse therapeutic approaches, especially those focused on nanotechnologies, and their potential contribution to the successful modulation of the host's response. The effects of the existing microbial diversity and the imbalance of key microbial species in contributing to the progression and worsening of the host's response in periodontitis are well known. It is essential to understand the role of a well-structured treatment plan for periodontitis, providing opportunities for new research and innovative treatment strategies aimed at reducing the impact of periodontitis on oral and overall systemic health. This will be beneficial for dental professionals, enabling them to effectively prevent and treat periodontitis, ultimately improving the overall health and well-being of patients.
Collapse
Affiliation(s)
| | | | | | - Zdravka Pashova-Tasseva
- Department of Periodontology, Faculty of Dental Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (A.M.); (V.D.-P.); (H.M.)
| |
Collapse
|
44
|
Du J, Liu Y, Dong Z, Huang Y, An Y, Cheng X, Sun G, Du C, Nie G, Hou X, Zhang Y. Cationic liposomes as broad-spectrum antidotes for heparin-based anticoagulants. Acta Biomater 2025; 195:283-296. [PMID: 39983854 DOI: 10.1016/j.actbio.2025.02.037] [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: 10/25/2024] [Revised: 01/18/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Heparin-based anticoagulants have been widely used for the prevention and treatment of venous thrombotic diseases, as well as for anticoagulation during cardiopulmonary bypass and hemodialysis. However, excessive heparin usage brings serious bleeding risk, necessitating immediate reversal of their anticoagulant activity. Additionally, to prevent bleeding during surgery and restore hemostatic function post-cardiopulmonary bypass and hemodialysis, it is also crucial to reverse heparin's anticoagulant effects. Currently, protamine sulfate (PS) is the only clinically approved antidote for heparin. However, its effectiveness against low molecular weight heparin (LMWH) and fondaparinux sodium is limited. Moreover, PS has great potential to trigger fatal allergic reactions. Despite these concerns, no successful clinical substitutes for PS have been developed. In the current work, drawing inspiration from the mechanism by which PS efficiently reverses heparin, we modified the cationic liposome with cationic amino acids, arginine and lysine, to serve as a broad-spectrum antidote (CRKRK-Lipo) for heparin-based anticoagulants. This modification not only enhances their reversal efficiency but also reduces the overall surface charge, potentially improving their biocompatibility. In the tail bleeding and liver injury mouse models, CRKRK-Lipo demonstrated reversal efficiency comparable to PS for heparin and superior reversal efficiency for LMWH and fondaparinux sodium. Notably, CRKRK-Lipo exhibited a wider therapeutic dose window and did not exhibit severe cytotoxicity or immunogenicity, in contrast to PS. It is worth noting that cationic liposomes without polypeptide modification also displayed a significant heparin reversal effect. Our findings not only offer a potential alternative for PS but also broaden the application fields of cationic liposome. STATEMENT OF SIGNIFICANCE: This study introduces the cationic liposomes as a novel and effective alternative to protamine sulfate (PS) for the functional reversal of heparin-based anticoagulants. The results reveal that both CRKRK-modified cationic liposomes (CRKRK-Lipo) and unmodified cationic liposomes (Lipo) showed comparable reversal efficiency to PS for UFH and superior reversal efficiency for LMWH and fondaparinux sodium, with a wider therapeutic dose window and reduced toxicity. This work offers an alternative strategy for detoxifying heparin-based anticoagulants and expands the biomedical applications of cationic liposomes.
Collapse
Affiliation(s)
- Jiarui Du
- College of Pharmacy, Shandong First Medical University, Shandong 250117, PR China
| | - Yang Liu
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China
| | - Zhenzhen Dong
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China
| | - Yubiao Huang
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China
| | - Yang An
- College of Pharmacy, Jilin University, Jilin 132000, PR China
| | - Xiaoyu Cheng
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China
| | - Ge Sun
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China
| | - Chong Du
- The Comprehensive Breast Care Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Guangjun Nie
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, PR China.
| | - Xueqin Hou
- College of Pharmacy, Shandong First Medical University, Shandong 250117, PR China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, PR China; The Comprehensive Breast Care Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
45
|
Yu N, Xu Y, Sun Q, Ge Y, Guo Y, Chen M, Shan H, Zheng M, Chen Z, Zhao S, Chen X. Size-specific clonidine-loaded liposomes: Advancing melanoma microenvironment suppression with safety and precision. J Control Release 2025; 379:120-134. [PMID: 39756687 DOI: 10.1016/j.jconrel.2025.01.001] [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: 09/02/2024] [Revised: 11/26/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) plays a crucial role in the progression and treatment resistance of melanoma. Modulating the TME is thus a key strategy for enhancing therapeutic outcomes. Previousstudies have identified clonidine (CLD), an α2-adrenergic receptor agonist, as a promising agent that enhances T lymphocyte infiltration and reduces myeloid-derived suppressor cells within the TME, thereby promoting antitumor immune responses. In this study, we discovered that CLD reshaped the melanoma immune microenvironment, facilitating T-cell activation and exerting antitumor effects. However, the high doses of CLD required for effective TME modulation pose significant toxicity concerns, limiting its clinical applicability. To address this, we employed the controllable cavitation-on-a-chip (CCC) platform to formulate CLD-loaded liposomes and optimize their size. This approach aimed to enhance the precision and efficacy of drug delivery while reducing systemic side effects. Our results demonstrated that size-specific CLD liposomes, particularly those at 50 nm, significantly improved tumor growth inhibition and immune cell infiltration within the TME. Moreover, these optimized liposomes mitigate adverse effects associated with high-dose CLD treatment. This study indicates the potential of CCC-optimized CLD liposomes as a safer and more effective melanoma therapy, highlighting the critical interplay between liposome size control and therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Nianzhou Yu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Sun
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Yi Ge
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yeye Guo
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Maike Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Han Shan
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Mingde Zheng
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zeyu Chen
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China.
| | - Shuang Zhao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
46
|
Tang J, Li D, Wang R, Li S, Xing Y, Yu F. Engineered extracellular vesicles: an emerging nanomedicine therapeutic platform. Chem Commun (Camb) 2025; 61:4123-4146. [PMID: 39969526 DOI: 10.1039/d4cc06501h] [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: 02/20/2025]
Abstract
The intercellular communication role of extracellular vesicles has been widely proved in various organisms. Compelling evidence has illustrated the involvement of these vesicles in both physiological and pathological processes. Various studies indicate that extracellular vesicles surpass conventional synthetic drug carriers, owing to their abundance in organisms, enhanced targeting ability and low immunogenicity. Therefore, extracellular vesicles have been deemed to be potential drug carriers for the treatment of various diseases, and related studies have increased rapidly. Here, we intend to provide a comprehensive and in-depth review of recent advances in the sources, delivery function, extraction and cargo-loading technologies of extracellular vesicles, as well as their clinical potential in constructing emerging nanomedicine therapeutic platforms. In particular, microfluidic-based isolation and drug-loading technologies, as well as the treatment of various diseases, are highlighted. We also make comparisons between extracellular vesicles and other conventional drug carriers and discuss the challenges in developing drug delivery platforms for clinical translation.
Collapse
Affiliation(s)
- Jingshi Tang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Dezhong Li
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Shiwei Li
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Yanlong Xing
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
47
|
Hou Y, Gao X, Gong J, Dong X, Hao Y, Zhai Z, Zhang H, Zhang M, Liu R, Wang R, Zhao L. Targeted Sodium Acetate Liposomes for Hepatocytes and Kupffer Cells: An Oral Dual-Targeted Therapeutic Approach for Non-Alcoholic Fatty Liver Disease Alleviation. Nutrients 2025; 17:930. [PMID: 40077800 PMCID: PMC11901740 DOI: 10.3390/nu17050930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Sodium acetate (NaA) has demonstrated potential in improving non-alcoholic fatty liver disease (NAFLD) by targeting hepatocytes and Kupffer cells. However, its clinical application is hindered by low oral bioavailability and insufficient liver concentrations. Liposomes, with their capacity to encapsulate water-soluble drugs and be surface-modified, offer a promising solution for targeted oral drug delivery. Methods: We designed NaA-loaded liposomes modified with sodium cholate (SC) and mannose (MAN) (NaA@SC/MAN-LPs) to target hepatocytes and Kupffer cells. Results: The NaA@SC/MAN-LPs had a mean diameter of approximately 100 nm with a positive surface charge. Compared to free NaA, NaA@SC/MAN-LPs significantly extended the serum half-life from 2.85 h to 15.58 h, substantially improving in vivo bioavailability. In vivo distribution studies revealed that NaA@SC/MAN-LPs extended the acetate peak time in the liver from 15 min to 60 min and increased hepatic acetate accumulation to 3.75 times that of free NaA. In in vitro cell experiments, NaA@SC/MAN-LPs significantly reduced the lipid droplet, triglycerides (TG), and total cholesterol (TC) in a fatty acid-induced hepatocyte steatosis model and suppressed proinflammation in a lipopolysaccharide (LPS)-activated Kupffer cell inflammation model. Free NaA effectively improved hepatic lipid deposition in NAFLD mice. Furthermore, NaA@SC/MAN-LPs decreased hepatic TG, TC, and the relative area of lipid droplets by 30.44%, 15.26%, and 55.83%, compared to free NaA. Furthermore, the liposomes reduced macrophage infiltration and pro-inflammatory response. Conclusions: The NaA@SC/MAN-LPs demonstrated effective dual targeting effects on hepatocytes and Kupffer cells, significantly improving the pathogenesis of NAFLD, compared to free NaA. This study provides a new strategy for developing effective and safe oral drugs for NAFLD.
Collapse
Affiliation(s)
- Yichao Hou
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Xilong Gao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Jiahui Gong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Xinrui Dong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Zhengyuan Zhai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Hao Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China;
| | - Rong Liu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| |
Collapse
|
48
|
Wang Y, Yuan M, Li S, Tang J, Wan Y, Liang X, Guo Y, Guo L. Multifunctional Liposome Delivery System Based on Ursodeoxycholic Acid Sodium for the Encapsulation of Silibinin and Combined Treatment of Alcoholic Liver Injury. Mol Pharm 2025; 22:1480-1497. [PMID: 39931930 DOI: 10.1021/acs.molpharmaceut.4c01197] [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: 03/04/2025]
Abstract
Alcohol liver disease (ALD) is a chronic liver disorder resulting from long-term heavy alcohol consumption. The pathogenesis of ALD is multifactorial, and existing therapeutic agents primarily target specific aspects of the disease while presenting significant side effects, including drug-induced liver injury and hepatobiliary disease. Silibinin (SLB) has attracted widespread attention for its hepatoprotective effects and favorable safety profile. However, inherent limitations associated with SLB, such as poor solubility and bioavailability, have significantly limited its clinical application. Drug delivery systems, including liposomes, offer promising potential for the delivery of hydrophobic drugs. However, the selection of an appropriate delivery vehicle requires optimization. Ursodeoxycholic acid sodium (UAS) serves as a promising alternative to cholesterol in liposomal formulations, offering a potential strategy to mitigate the health risks associated with cholesterol. In this study, UAS was employed as the liposomal membrane material to prepare a UAS liposome loaded with SLB (SUL), and its efficacy and mechanism of action in alcoholic-induced liver injury were subsequently evaluated. The experimental results demonstrated that SUL exhibited a uniform particle size distribution, good stability, and an effective release profile in vitro. Following oral administration, SUL effectively inhibited alcohol-induced liver damage, oxidative stress, and fat accumulation. In addition, SUL regulated the expression of the kelch-1ike ECH- associated protein l (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1) proteins, thereby exerting antioxidative stress effects. Furthermore, it also modulated apoptosis-related factors, including B-cell lymphoma-2 (Bcl-2), BCL-2-associated X (Bax), cysteinyl aspartate specific proteinase-3 (Caspase-3), and cleaved caspase-3, to mitigate hepatocyte apoptosis. In summary, SUL demonstrates enhanced therapeutic efficacy against ALD, offering a novel approach for the clinical application of SLB in the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| |
Collapse
|
49
|
Gandhi N, Modi S, Soni S, Andey T. Modular self-emulsifying drug delivery platform to enhance cellular uptake activity in triple-negative breast cancer. Eur J Pharm Sci 2025; 206:106993. [PMID: 39708842 DOI: 10.1016/j.ejps.2024.106993] [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: 07/22/2023] [Revised: 11/08/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Triple-negative breast cancer (TNBC) presents with resistance phenotypes to certain therapies, such as cisplatin, often requiring higher dosing, with associated acquired tumor resistance, renal toxicity, and variable patient responses. A self-emulsifying drug delivery (SEDD) formulation approach was proposed to overcome the limitations of cisplatin in TNBC, focusing on improving intracellular cisplatin and control siRNA uptake as a proof-of-principle of dual drug delivery. Four SEDD formulations were prepared and optimized for cisplatin (o/w) emulsion and FITC-siRNA (w/o) emulsion using pseudo-ternary phase diagrams to facilitate the formation of water-in-oil-water (w/o/w) emulsions. Formulations were characterized by size, polydispersity (PDI), and surface charge and tested in vitro. Cellular uptake via triplex staining of drug-loaded SEDDs was investigated. SEDDs showed enhanced internalization and promoted selective TNBC cellular uptake. The current study is a proof-of-principle for the successful co-delivery of cisplatin (small molecule) and siRNA (large molecule) via the SEDDs platform.
Collapse
Affiliation(s)
- Nandini Gandhi
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Shail Modi
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Shailvi Soni
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Terrick Andey
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA.
| |
Collapse
|
50
|
Zhang X, Zhang L, Tian J, Li Y, Wu M, Zhang L, Qin X, Gong L. The application and prospects of drug delivery systems in idiopathic pulmonary fibrosis. BIOMATERIALS ADVANCES 2025; 168:214123. [PMID: 39615374 DOI: 10.1016/j.bioadv.2024.214123] [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: 06/11/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease primarily affecting elderly individuals aged >65 years and has a poor prognosis. No effective treatment is currently available for IPF. The two antipulmonary fibrosis drugs nintedanib and pirfenidone approved by the FDA in the United States have somewhat decelerated IPF progression. However, the side effects of these drugs can lead to poor patient tolerance and compliance with the medications. Researchers have recently developed various methods for IPF treatment, such as gene silencing and pathway inhibitors, which hold great promise in IPF treatment. Nevertheless, the nonselectivity and nonspecificity of drugs often affect their efficacies. Drug delivery systems (DDS) are crucial for delivering drugs to specific target tissues or cells, thereby minimizing potential side effects, enhancing drug bioavailability, and reducing lung deposition. This review comprehensively summarizes the current state of DDS and various delivery strategies for IPF treatment (e.g., nano-delivery, hydrogel delivery, and biological carrier delivery) to completely expound the delivery mechanisms of different drug delivery carriers. Subsequently, the advantages and disadvantages of different DDS are fully discussed. Finally, the challenges and difficulties associated with the use of different DDS are addressed so as to accelerate their rapid clinical translation.
Collapse
Affiliation(s)
- Xi Zhang
- School of Biological Engineering, Zunyi Medical University, Guangdong 519000, China; Department of Clinical Medicine, The Fifth Clinical Institution, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Jiahua Tian
- Department of Clinical Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yunfei Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Manli Wu
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Longju Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Xiaofei Qin
- School of Biological Engineering, Zunyi Medical University, Guangdong 519000, China.
| | - Ling Gong
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China.
| |
Collapse
|