1
|
Xu Z, Wang Y, Li S, Li Y, Chang L, Yao Y, Peng Q. Advances of functional nanomaterials as either therapeutic agents or delivery systems in the treatment of periodontitis. BIOMATERIALS ADVANCES 2025; 175:214326. [PMID: 40300444 DOI: 10.1016/j.bioadv.2025.214326] [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: 03/14/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Periodontitis is a common chronic inflammatory disease primarily caused by pathogenic microorganisms in the oral cavity. Without appropriate treatments, it may lead to the gradual destruction of the supporting tissues of the teeth. While current treatments can alleviate symptoms, they still have limitations, particularly in eliminating pathogenic bacteria, promoting periodontal tissue regeneration, and avoiding antibiotic resistance. In recent years, functional nanomaterials have shown great potential in the treatment of periodontitis due to their unique physicochemical and biological properties. This review summarizes various functionalization strategies of nanomaterials and explores their potential applications in periodontitis treatment, including metal-based nanoparticles, carbon nanomaterials, polymeric nanoparticles, and exosomes. The mechanisms and advances in antibacterial effects, immune regulation, reactive oxygen species (ROS) scavenging, and bone tissue regeneration are discussed in detail. In addition, the challenges and future directions of applying nanomaterials in periodontitis therapy are also discussed.
Collapse
Affiliation(s)
- Ziyi Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuoshun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuanhong Li
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Tarin M, Oryani MA, Javid H, Hashemzadeh A, Karimi-Shahri M. Advancements in chitosan-based nanocomposites with ZIF-8 nanoparticles: multifunctional platforms for wound healing applications. Carbohydr Polym 2025; 362:123656. [PMID: 40409814 DOI: 10.1016/j.carbpol.2025.123656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/25/2025]
Abstract
The integration of chitosan and zeolitic imidazolate framework-8 (ZIF-8) nanoparticles has demonstrated significant potential in enhancing wound healing through their multifunctional capabilities. This review explores recent developments in chitosan-based nanocomposites incorporating ZIF-8 nanoparticles, emphasizing their antibacterial properties, pH-responsive drug release, angiogenesis promotion, and mechanical stability. Applications span hydrogel scaffolds, electrospun nanofibers, and sprayable membranes, all tailored for addressing challenges such as bacterial resistance, delayed tissue regeneration, and chronic wound management. Key findings highlight the synergistic benefits of ZIF-8's bioactivity with chitosan's biocompatibility, yielding innovative therapeutic strategies for complex wound healing scenarios. The discussed advancements not only underline their clinical relevance but also set a foundation for future explorations in regenerative medicine.
Collapse
Affiliation(s)
- Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of medical sciences, Mashhad. Iran.
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Iran.
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
3
|
Ren H, Xu J, Lai Y, Xu R, Li J, Shen JW, Chen JX. pH sensitive loading and release of doxorubicin by chitosan-graphene quantum dots hybridized material. Int J Biol Macromol 2025:145375. [PMID: 40541879 DOI: 10.1016/j.ijbiomac.2025.145375] [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: 02/04/2025] [Revised: 06/01/2025] [Accepted: 06/17/2025] [Indexed: 06/22/2025]
Abstract
The effective delivery of doxorubicin (DOX) to tumor tissues remains a significant challenge in cancer therapy. In this study, molecular dynamics (MD) simulations were employed to investigate the loading and release mechanisms of DOX in chitosan-graphene quantum dots (GQDs) complexes under varying pH levels and DOX concentrations. The results show that chitosan aggregates at basic pH, thereby enhancing DOX encapsulation via hydrogen bonding. Meanwhile, GQDs exhibit higher DOX capture efficiency at acidic pH via π-π stacking interactions. At higher DOX concentrations, self-aggregation of DOX molecules reduces their availability for interaction with chitosan and GQDs, leading to decreased encapsulation efficiency. Furthermore, our simulations mimicking the pH transition from a neutral to an acidic tumor-like environment show that chitosan disperses in solution, leading to the sensitive release of DOX. These findings demonstrate that the controllable loading and release of DOX by chitosan-GQDs hybridized material can be achieved by adjusting the solution pH (the protonation state of chitosan) and the concentration of DOX.
Collapse
Affiliation(s)
- Hao Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiahao Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuanqiu Lai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ruru Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiachen Li
- School of Physics, HangZhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jiang-Xing Chen
- School of Physics, HangZhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
4
|
Chandra DK, Kumar A, Mahapatra C. High-Precision Multistage Molecular Dynamics Simulations and Quantum Mechanics Investigation of Adsorption Mechanisms of Cerium-Based H 3BTC MOF (Ce-H 3BTC-MOF) on Pristine and Functionalized Carbon Nanotubes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:14889-14914. [PMID: 40407229 DOI: 10.1021/acs.langmuir.5c01011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Metal-organic frameworks (MOFs) and carbon nanotubes (CNTs) exhibit exceptional physicochemical properties, making them promising candidates for nanotherapeutics, catalysis, and drug delivery. However, CNTs face inherent limitations in solubility and functionalization, which hinder their practical applications. This study systematically investigates the adsorption mechanisms of cerium-based benzene-1,3,5-tricarboxylate MOFs (Ce-H3BTC-MOF) on pristine (PCNT), mildly functionalized (MFCNT), and densely functionalized (DFCNT) CNTs using multistage molecular dynamics simulations and quantum mechanics calculations. Functionalization was achieved by grafting 5 (-COOH) groups on the MFCNT and 20 (-COOH) groups on the DFCNT, enhancing interfacial interactions. Loading configurations of 1-5 Ce-H3BTC MOF molecules were analyzed to understand binding stability and molecular dispersion. Simulation results demonstrated enhanced adsorption on functionalized CNTs due to π-π stacking and electrostatic interactions, with 5CeBTC-1DFCNT exhibiting the highest cohesive energy (311.01 kcal/mol) and optimized solubility (1.13 MPa1/2). Density functional theory calculations revealed a HOMO-LUMO energy gap of 0.21029 eV for BTC and 0.23089 eV for CeO2, indicating electronic stability. Root mean square deviation (rmsd) and radius of gyration (Rg) analyses confirmed structural stability, with 1CeBTC-1MFCNT maintaining the lowest rmsd (∼2.47 Å) and 5CeBTC-1DFCNT exhibiting significant fluctuations (∼6.87 Å) due to steric hindrance. This study advances the understanding of CNT-MOF hybrid systems by elucidating their interfacial dynamics, providing a foundation for future applications in nanomedicine and catalysis.
Collapse
Affiliation(s)
- Dilip Kumar Chandra
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh 492010, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh 492010, India
| | - Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh 492010, India
| |
Collapse
|
5
|
Boyuklieva R, Zahariev N, Simeonov P, Penkov D, Katsarov P. Next-Generation Drug Delivery for Neurotherapeutics: The Promise of Stimuli-Triggered Nanocarriers. Biomedicines 2025; 13:1464. [PMID: 40564183 PMCID: PMC12191273 DOI: 10.3390/biomedicines13061464] [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: 05/16/2025] [Revised: 06/10/2025] [Accepted: 06/11/2025] [Indexed: 06/28/2025] Open
Abstract
Nanotherapeutics have emerged as novel unparalleled drug delivery systems (DDSs) for the treatment of neurodegenerative disorders. By applying different technological approaches, nanoparticles can be engineered to possess different functionalities. In recent years, the developed, stimuli-responsive nanocarriers stand out as novel complex DDSs ensuring selective and specific drug delivery in response to different endogenous and exogenous stimuli. Due to the multifaceted pathophysiology of the nervous system, a major challenge in modern neuropharmacology is the development of effective therapies ensuring high efficacy and low toxicity. Functionalization of the nanocarriers to react to specific microenvironmental changes in the nervous system tissues or external stimulations significantly enhances the efficacy of drug delivery. This review discusses the microenvironmental characteristics of some common neurological diseases in-depth and provides a comprehensive overview on the progress of the development of exogenous and endogenous stimuli-sensitive nanocarriers for the treatment of Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Radka Boyuklieva
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (R.B.); (N.Z.); (P.S.); (D.P.)
- Research Institute at Medical University of Plovdiv (RIMU), 4002 Plovdiv, Bulgaria
| | - Nikolay Zahariev
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (R.B.); (N.Z.); (P.S.); (D.P.)
- Research Institute at Medical University of Plovdiv (RIMU), 4002 Plovdiv, Bulgaria
| | - Plamen Simeonov
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (R.B.); (N.Z.); (P.S.); (D.P.)
| | - Dimitar Penkov
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (R.B.); (N.Z.); (P.S.); (D.P.)
| | - Plamen Katsarov
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (R.B.); (N.Z.); (P.S.); (D.P.)
- Research Institute at Medical University of Plovdiv (RIMU), 4002 Plovdiv, Bulgaria
| |
Collapse
|
6
|
Lee YJ, Kim MS. Advances in drug-loaded microspheres for targeted, controlled, and sustained drug delivery: Potential, applications, and future directions. Biomed Pharmacother 2025; 189:118244. [PMID: 40516334 DOI: 10.1016/j.biopha.2025.118244] [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: 04/23/2025] [Revised: 05/23/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025] Open
Abstract
Drug-loaded microspheres are an innovative technology in drug delivery systems (DDS), addressing many limitations of conventional methods. Their ability to enable controlled release, precise targeting, and broad drug compatibility makes them a versatile platform with significant potential in modern medicine. This review explores the unique properties of microspheres, including their biocompatibility, biodegradability, and customizable architecture, positioning them as promising candidates for therapeutic use in cancer, diabetes, and rheumatoid arthritis. These characteristics enhance drug stability and bioavailability while reducing systemic side effects, improving patient outcomes. The key findings discussed in this review highlight critical factors influencing microsphere performance, including material selection, particle size, surface modification, and multi-drug loading strategies. Particularly, the integration of nanoscale materials and the combination of microsphere technology with gene therapy and immunotherapy have shown great potential to improve treatment precision and efficacy. However, challenges such as large-scale production, reproducibility, and optimization of drug release profiles remain significant hurdles. Large-scale manufacturing of microspheres with consistent size, efficient drug loading, and predictable release patterns is technically complex, and optimizing release, especially for drugs with narrow therapeutic windows, requires a deeper understanding of the interactions between drugs and polymers. Future advances in microsphere technology are expected to leverage innovations in nanotechnology, gene therapy, and immunotherapy. These advancements may enable more efficient and personalized treatments for diseases that were previously difficult to treat. The findings presented in this review emphasize the transformative potential of microspheres in revolutionizing drug delivery, offering safer, more effective, and patient-specific therapies.
Collapse
Affiliation(s)
- Ye Jin Lee
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea.
| |
Collapse
|
7
|
Zhao D, Zheng S, Zuo X, Xu H, Ding Y, Liang F. Light-Activated Hypoxia-Responsive Nanoparticles for Photodynamic Chemotherapy. ACS OMEGA 2025; 10:22719-22724. [PMID: 40521527 PMCID: PMC12163835 DOI: 10.1021/acsomega.4c11283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 04/24/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025]
Abstract
Hypoxia is a characteristic of solid tumors, and it significantly impedes cancer treatment. Here, we report light-activated hypoxia-responsive nanoparticles NPs-TPZ consisting of 5,10,5,20-tetrakis-(4-aminophenyl)-porphine (TAPP) modified with four azobenzene groups, cyclodextrin (CD), and 3-aminobenzotriazine-1,4-di-N-oxide tirapazamine (TPZ) by the synergy of π-π stacking, host-guest, and hydrophobic interactions for synergistic photodynamic chemotherapy (PDT-CT). Under near-infrared (NIR) irradiation, the process of PDT depletes oxygen and generates singlet oxygen (1O2). The induced hypoxia exacerbation further accelerates the release and activation of TPZ. As a result, this hypoxia-responsive nanoparticle provides an effective strategy for the ablation of hypoxic solid tumors by synergistic PDT-CT.
Collapse
Affiliation(s)
- Dan Zhao
- Department
of Intensive Care Unit, The Affiliated Wuxi
People’s Hospital of Nanjing Medical University, Wuxi Medical
Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi214023, Jiangsu, China
| | - Shunliang Zheng
- School
of Chemistry and Chemical Engineering, Nantong
University, Nantong226019, P. R. China
| | - Xinyi Zuo
- School
of Chemistry and Chemical Engineering, Nantong
University, Nantong226019, P. R. China
| | - Hongyang Xu
- Department
of Intensive Care Unit, The Affiliated Wuxi
People’s Hospital of Nanjing Medical University, Wuxi Medical
Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi214023, Jiangsu, China
| | - Yue Ding
- School
of Chemistry and Chemical Engineering, Nantong
University, Nantong226019, P. R. China
| | - Fengming Liang
- Department
of Intensive Care Unit, The Affiliated Wuxi
People’s Hospital of Nanjing Medical University, Wuxi Medical
Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi214023, Jiangsu, China
| |
Collapse
|
8
|
Jian X, Liu D, Liu X, Zhao X, Zhu Y, Xia S, Peng E, Zhao M, Yi J, Jiang G, Xu D, Cheng K, Weng W, Zhu Z, Shi B, Tang B. Thiol-ene photoclick hydrogels reinforced with poly(protocatechualdehyde)-coated gallium doped bioactive glass nanoparticles for scarless healing of infected wound. Biomaterials 2025; 324:123469. [PMID: 40489905 DOI: 10.1016/j.biomaterials.2025.123469] [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: 01/15/2025] [Revised: 05/16/2025] [Accepted: 06/03/2025] [Indexed: 06/11/2025]
Abstract
Complex pathological microenvironment mainly characterized by excessive inflammation, reactive oxygen species (ROS) production, drug-resistant bacterial infection makes wound rapid scarless healing still a major clinical challenge. Inspired by skin composed principally of collagen and mucopolysaccharides, an injectable nanocomposite hydrogel with immunoregulation, antioxidant and antibacterial activity based on allyl glycidyl ether grafted gelatin (Gel-AGE), cysteamine grafted hyaluronic acid (HA-CSA) and poly(protocatechualdehyde)-coated gallium doped bioactive glass (PPA@GaBG) nanoparticles, was designed for treatment of infected wound. The PPA@GaBG/Gel-AGE/HA-CSA (PGBGH) hydrogel can be rapidly formed via thiol-ene photo-click chemistry between Gel-AGE and HA-CSA. In PGBGH hydrogel, the presence of core-shell PPA@GaBG significantly enhanced the antioxidant ability of hydrogels and M2 polarization of macrophages, as well as improving mechanical and tissue adhesive properties of the hydrogels via the formation of Schiff base bonds between PPA and gelatin/skin tissue. Moreover, the PPA shell effectively inhibited the ion burst release of Ga3+ observed in GaBG/GH hydrogels, which improved the biocompatibility of the PGBGH hydrogel while maintaining excellent antibacterial activity. As a result of multiple functions of the PPA@GaBG, the PGBGH hydrogel promoted angiogenesis, granulation tissue formation and re-epithelialization with improved deposition ratio of collagen III/I, which achieved the rapid scarless healing of infected wound.
Collapse
Affiliation(s)
- Xinyi Jian
- School of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Dun Liu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xuexue Liu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Xingping Zhao
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yongjie Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Sanqiang Xia
- The Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - Enuo Peng
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Mengran Zhao
- School of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Jie Yi
- School of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Guohua Jiang
- College of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Dabao Xu
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Benlong Shi
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Bolin Tang
- School of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Nanotechnology Research Institute, G60 STI Valley Industry & Innovation Institute, Jiaxing University, Jiaxing, 314001, China.
| |
Collapse
|
9
|
Guo L, Fu Z, Li H, Wei R, Guo J, Wang H, Qi J. Smart hydrogel: A new platform for cancer therapy. Adv Colloid Interface Sci 2025; 340:103470. [PMID: 40086017 DOI: 10.1016/j.cis.2025.103470] [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: 07/25/2024] [Revised: 01/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Cancer is a significant contributor to mortality worldwide, posing a significant threat to human life and health. The unique bioactivity, ability to precisely control drug release, and minimally invasive properties of hydrogels are indispensable attributes that facilitate optimal performance in cancer therapy. However, conventional hydrogels lack the ability to dynamically respond to changes in the surrounding environment, withstand drastic changes in the microenvironment, and trigger drug release on demand. Therefore, this review focuses on smart-responsive hydrogels that are capable of adapting and responding to external stimuli. We comprehensively summarize the raw materials, preparation, and cross-linking mechanisms of smart hydrogels derived from natural and synthetic materials, elucidate the response principles of various smart-responsive hydrogels according to different stimulation sources. Further, we systematically illustrate the important role played by hydrogels in modern cancer therapies within the context of therapeutic principles. Meanwhile, the smart hydrogel that uses machine learning to design precise drug delivery has shown great prospects in cancer therapy. Finally, we present the outlook on future developments and make suggestions for future related work. It is anticipated that this review will promote the practical application of smart hydrogels in cancer therapy and contribute to the advancement of medical treatment.
Collapse
Affiliation(s)
- Li Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ziming Fu
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Haoran Li
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ruibo Wei
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Jing Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Haiwang Wang
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Jian Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
10
|
Zhang W, Liu H, Zhu B, Li W, Han X, Fu J, Luo R, Wang H, Wang J. Advances in Cytosolic Delivery of Proteins: Approaches, Challenges, and Emerging Technologies. Chem Biodivers 2025; 22:e202401713. [PMID: 39921680 PMCID: PMC12168193 DOI: 10.1002/cbdv.202401713] [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: 07/14/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Although therapeutic proteins have achieved recognized clinical success, they are inherently membrane impermeable, which limits them to acting only on extracellular or membrane-associated targets. Developing an efficient protein delivery method will provide a unique opportunity for intracellular target-related therapeutic proteins. In this review article, we summarize the different pathways by which cells take up proteins. These pathways fall into two main categories: One in which proteins are transported directly across the cell membrane and the other through endocytosis. At the same time, important features to ensure successful delivery through these pathways are highlighted. We then provide a comprehensive overview of the latest developments in the transduction of covalent protein modifications, such as coupling cell-penetrating motifs and supercharging, as well as the use of nanocarriers to mediate protein transport, such as liposomes, polymers, and inorganic nanoparticles. Finally, we emphasize the existing challenges of cytoplasmic protein delivery and provide an outlook for future progress.
Collapse
Affiliation(s)
- Wenyan Zhang
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | | | | | - Wen Li
- Gansu Provincial HospitalLanzhouGansuChina
| | - Xue Han
- Gansu Provincial HospitalLanzhouGansuChina
| | - Jiaojiao Fu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | - Renjie Luo
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | - Haiyan Wang
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | - Jinxia Wang
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| |
Collapse
|
11
|
Hu C, Ma J, Su Z, Wang J, Zhang X, Pang L, Qu Y, Shi J, Zhang J. Co-delivery of doxorubicin and glycyrrhetinic acid via acid/glutathione dual responsive nano-prodrug with sodium bicarbonate carry-on for advanced combinational cancer treatment. J Colloid Interface Sci 2025; 687:248-260. [PMID: 39954419 DOI: 10.1016/j.jcis.2025.02.045] [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: 10/29/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
The combination of doxorubicin (Dox) and glycyrrhetinic acid (GA) has been widely explored for treating various cancers, while the heterogeneous distribution, uncontrolled release and acid tumor microenvironment often hinder their synergistic effects. Herein, we introduce an acid/glutathione (GSH)-dual responsive nano-prodrug (AS1411@Dox+GA/NPs) enabling precise co-delivery of Dox and GA for enhanced combination therapy. The GSH-activated Dox prodrug [Poly(lactic-co-glycolic acid)-disulfide-Dox, PLGA-ss-Dox], GA, and sulfur-terminated D-α-tocopherol polyethylene glycol succinate (TPGS-SH) are combined to form a nanoemulsion with sodium bicarbonate (NaHCO3) in the aqueous phase. The AS1411 aptamer is modified on the surface for tumor-specific targeting. Upon reaching the tumor via enhanced penetration and retention effects, AS1411 aptamer medicates specific endocytosis of AS1411@Dox+GA/NPs into tumor cells. Intracellularly, the acidic endosomal environment promotes carbon dioxide (CO2) production from NaHCO3, disrupting the nanoemulsion and causing a burst release of GA and PLGA-ss-Dox prodrug. Concurrently, high levels of GSH in the cytoplasm triggers the cleavage of the disulfide linker, thereby releasing Dox. Notably, the released NaHCO3 consumed hydrogen ion (H+), amplifying the sensitivity of tumor cells to Dox. As a consequence, AS1411@Dox+GA/NPs exhibits remarkable synergistic therapeutic efficacy in mouse models of both liver cancer and breast cancer. This work presents an appealing approach utilizing stimuli-sensitive nano-prodrug for advanced combinational cancer treatment.
Collapse
Affiliation(s)
- Chuan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiaqi Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ziye Su
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Junyu Wang
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Xueqian Zhang
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Lin Pang
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yan Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinfeng Shi
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
12
|
Wang Y, Jan H, Zhong Z, Zhou L, Teng K, Chen Y, Xu J, Xie D, Chen D, Xu J, Qin L, Tuan RS, Li ZA. Multiscale metal-based nanocomposites for bone and joint disease therapies. Mater Today Bio 2025; 32:101773. [PMID: 40290898 PMCID: PMC12033929 DOI: 10.1016/j.mtbio.2025.101773] [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: 03/09/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Bone and joint diseases are debilitating conditions that can result in significant functional impairment or even permanent disability. Multiscale metal-based nanocomposites, which integrate hierarchical structures ranging from the nanoscale to the macroscale, have emerged as a promising solution to this challenge. These materials combine the unique properties of metal-based nanoparticles (MNPs), such as enzyme-like activities, stimuli responsiveness, and photothermal conversion, with advanced manufacturing techniques, such as 3D printing and biohybrid systems. The integration of MNPs within polymer or ceramic matrices offers a degree of control over the mechanical strength, antimicrobial efficacy, and the manner of drug delivery, whilst concomitantly promoting the processes of osteogenesis and chondrogenesis. This review highlights breakthroughs in stimulus-responsive MNPs (e.g., photo-, magnetically-, or pH-activated systems) for on-demand therapy and their integration with biocomposite hybrids containing cells or extracellular vesicles to mimic the native tissue microenvironment. The applications of these composites are extensive, ranging from bone defects, infections, tumors, to degenerative joint diseases. The review emphasizes the enhanced load-bearing capacity, bioactivity, and tissue integration that can be achieved through hierarchical designs. Notwithstanding the potential of these applications, significant barriers to progress persist, including challenges related to long-term biocompatibility, regulatory hurdles, and scalable manufacturing. Finally, we propose future directions, including machine learning-guided design and patient-specific biomanufacturing to accelerate clinical translation. Multiscale metal-based nanocomposites, which bridge nanoscale innovations with macroscale functionality, are a revolutionary force in the field of biomedical engineering, providing personalized regenerative solutions for bone and joint diseases.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Hasnain Jan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Zheng Zhong
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Kexin Teng
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Ye Chen
- Department of Chemistry, Faculty of Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Dexin Chen
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou, 510632, China
| | - Jiake Xu
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, NT, Hong Kong Special Administrative Region of China
| |
Collapse
|
13
|
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
|
14
|
Wang C, Ren K, Yang M, Li X, Li N, Li P, Yang H, Zhang G, Wei X. How Traditional Chinese Medicine Can Play a Role In Nanomedicine? A Comprehensive Review of the Literature. Int J Nanomedicine 2025; 20:6289-6315. [PMID: 40416728 PMCID: PMC12103218 DOI: 10.2147/ijn.s518610] [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: 01/21/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Traditional Chinese medicine (TCM), a time-honored practice rooted in natural therapeutics, has served as a cornerstone in safeguarding human health across millennia, aiding in disease mitigation and life vitality preservation. However, many TCM active ingredients suffer from poor solubility, low bioavailability, uncertain toxicity and weak targeting ability. Nanomedicine represents a modern scientific frontier, emerging from the precise engineering of unique nanoscale characteristics, with extensive applications encompassing targeted therapeutic delivery and diverse biomedical fields. Although TCM and nanomedicine diverge fundamentally in historical origins and disciplinary foundations, growing investigations demonstrate their synergistic potential. In this review, nanosized TCM has been revealed as an innovative therapeutic strategy with significant clinical value. Based on the biological activities and structural characteristics of TCM active ingredients, we classify them into two categories: natural nanostructured formulations for TCM and nano-drug delivery systems for TCM. A systematic and comprehensive analysis of preparations specific and functions to two classes of TCM nanomedicines is highlighted. Insights into the advantage of TCM nanomedicines are also introduced. Subsequently, the applications of TCM nanomedicines in the biomedical treatment, including anti-cancer, anti-inflammation and anti-bacterial are summarized. Finally, challenges and future research directions are emphasized, aiming to offer guidance for the modernization of TCM nanomedicines.
Collapse
Affiliation(s)
- Chi Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Kaixiang Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Mei Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Xiang Li
- Department of Ophthalmology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Ningxi Li
- Mianyang Key Laboratory of Anesthesia and Neuroregulation, Department of Anesthesiology, Mianyang Central Hospital, Mianyang, 621000, People’s Republic of China
| | - Peng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People’s Republic of China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Xiaodan Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| |
Collapse
|
15
|
Zhang W, Tian X, Xu S, Wu B, Jiang T. Supramolecular Prodrug Hydrogel for One-Week Protection Against Thrombosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500193. [PMID: 40388659 DOI: 10.1002/smll.202500193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/26/2025] [Indexed: 05/21/2025]
Abstract
In clinical anticoagulant therapy, the drug Bivalirudin (Biva) presents a lower incidence of adverse events and more predictable pharmacokinetics in comparison to heparin. However, its short half-life of ≈20 min leads to poor patient compliance and increased medical burden. Here, a long-acting anticoagulant hydrogel based on Biva for antithrombotic treatment is described. The fusion peptide (d-RADA)8-B that integrates Biva, a D-type self-assembly motif, and an activated factor X (FXa)-responsive motif exhibits both supramolecular reservoir and prodrug-like properties. After subcutaneous injection, the anticoagulant peptide forms a semi-solid depot with protease-degradation resistance and slowly disassembles to release prodrug (d-RADA)8-B into the bloodstream. The circulating prodrug acts as an inert sentinel, which can be activated to release Biva to inhibit thrombus formation when exposed to the thrombus-related protease FXa. One week after the administration of (d-RADA)8-B, significant embolism suppression is observed in animal models of carotid artery thrombosis and pulmonary embolism without increasing hemorrhagic side effects. This study demonstrates a concise strategy to engineer a supramolecular anticoagulant hydrogel with long-term, high drug loading, and on-demand antithrombotic activation.
Collapse
Affiliation(s)
- Wenjing Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Xue Tian
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Sheng Xu
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Bin Wu
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
16
|
Dong H, Fa J, Yan M, Tan Y, Cheng W, Huang J, Ling Z. Development and analysis of chitin/cellulose reinforced galactomannan fluidic hydrogel for drug delivery application. J Colloid Interface Sci 2025; 686:701-710. [PMID: 39919515 DOI: 10.1016/j.jcis.2025.02.005] [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: 11/16/2024] [Revised: 01/23/2025] [Accepted: 02/01/2025] [Indexed: 02/09/2025]
Abstract
Rising demand for advanced materials in biomedical applications has made the development of effective hydrogels a critical focus, especially for drug delivery. Herein, a novel fluidic hydrogel with high drug-loading (>95 %) capacity was developed for controlled release of acetylsalicylic acid. Nature-derived galactomannan (GM) was served as the hydrogel matrix, and chitin nanocrystals (ChNC) and cellulose nanocrystals (CNC) were used to enhance structure and antimicrobial properties. Physical crosslinking of the components were tuned to optimize the porosity, flowability, and mechanical strength of the materials, as well as improving rheology and enabling rapid self-healing within 60 s. Co-culturing experiments demonstrate excellent biocompatibility of both ChNC@GM and CNC@GM hydrogels. Moreover, ChNC@GM shows superior 82 % antimicrobial activity, and the hydrogel exhibits pH-responsive drug release. Therefore, the proposed fluidic hydrogel may act as promising material for applications fields of wound healing, drug delivery, and biomedical engineering, etc.
Collapse
Affiliation(s)
- Hanqi Dong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Jingjing Fa
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengxing Yan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Yang Tan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Wenbo Cheng
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianfeng Huang
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhe Ling
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China.
| |
Collapse
|
17
|
Li S, Fan Z, Zheng K, Wu Y, Zhong G, Xu X. Engineered Probiotics with Low Oxygen Targeting Porphyromonas gingivalis and Gingival Fibroblasts for the Treatment of Periodontitis. ACS Biomater Sci Eng 2025; 11:2753-2767. [PMID: 40286317 DOI: 10.1021/acsbiomaterials.5c00111] [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: 04/29/2025]
Abstract
The overuse of antibiotics has increased the prevalence of drug-resistant bacteria in periodontitis. "Sentinel" gingival fibroblasts, stimulated by pathogenic bacteria, continue to release signaling factors that affect stem cell repair and recruit immune cells, resulting in persistent inflammation in periodontal tissues, eventually leading to the loosening and loss of teeth. Periodontal pathogenic bacteria cause surface hypoxia, and gingival fibroblasts in the inflammatory microenvironment express HIF-1α, promoting hypoxic areas in periodontal pockets. No drug delivery system is available for the hypoxic region of periodontal pockets. We synthesized BI NPs via berberine (BBR) and indocyanine green (ICG) and formed BIP NPs by wrapping BI NPs with polydopamine (PDA), and the BIP NPs were delivered to the hypoxic region of the periodontal pocket by hitchhiking with the anaerobic probiotic Bifidobacterium bifidum (Bif). The BIP NPs released berberin (BBR) under near-infrared (NIR) irradiation, which inhibited the sulfur metabolism of Porphyromonas gingivalis via mild photothermal action and BBR-targeted serine acetyltransferase, resulting in a decrease in resistance to oxidative stress, thus exerting a nonantibiotic bacteriostatic effect. This mild photothermal effect facilitated the uptake of BIP NPs bygingival fibroblasts. Moreover, BBR targeted nuclear factor-erythroid 2-related factor 2 (NRF2) to reduce ferroptosis, and the gingival fibroblast supernatant modulated macrophage polarization through the NF-κB pathway. In the periodontitis rat model, Bif@BIP+NIR treatment carried the drug to deep periodontal pockets, decreasing local gingival ferroptosis and alleviating periodontitis symptoms. To summarize, engineered probiotics target low-oxygen periodontal pockets for drug delivery, P. gingivalis for nonantibiotic bacterial inhibition, and gingival fibroblasts to mitigate ferroptosis, thus alleviating periodontitis to reduce periodontitis.
Collapse
Affiliation(s)
- Shenghong Li
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhibo Fan
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Kaijun Zheng
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yujie Wu
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Guannan Zhong
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaomei Xu
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
18
|
Gomez NA, Blumel D, Dueñas D, Young B, Hazel M, Yu M. Influence of experimental conditions on the adsorption of disease biomarker proteins to InP/ZnS quantum dots. Anal Biochem 2025; 704:115903. [PMID: 40368225 DOI: 10.1016/j.ab.2025.115903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/25/2025] [Accepted: 05/11/2025] [Indexed: 05/16/2025]
Abstract
The spontaneous formation of quantum dot (QD)-protein assemblies in the physiological environment exhibits challenges or benefits for nanomedicine applications. In this study, we investigated the QD-protein assemblies spontaneously formed with the greener water soluble InP/ZnS-COOH QDs and isolated disease biomarker proteins under various environmental conditions, including QDs size, solution pH, incubation time, ionic strength, different salts, as well as the lowest concentrations of the proteins that started the formation of detectable assemblies. It was shown that higher ionic strength or valence charge disrupted the assembly's formation. The basic pH 8.5 facilitated the formation to a greater extent than the pH 7.4 did. The heat shock protein 90-alpha (HSP90α) adsorbed on QDs surface more readily than cytochrome C (CytoC) and lysozyme (Lyz) in the basic environment. Among the three-sized QDs compared, the medium-sized QDs were the most effective in promoting the assemblies' formation. The detectable assemblies started at as low as 0.4 ng/mL of CytoC, 1.0 ng/mL of HSP90α, or 1.8 ng/mL of Lyz, respectively. The findings add insights into how the biomarker proteins interacted with the QDs under different environmental conditions, which promotes the understanding of QD-protein assemblies' collaborative behaviors when they facilitate bioimaging and biomedicine applications.
Collapse
Affiliation(s)
- Nathaniel A Gomez
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058
| | - Daniel Blumel
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058
| | - Davies Dueñas
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058
| | - Bronson Young
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058
| | - Matt Hazel
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058
| | - Ming Yu
- Department of Chemistry, Utah Valley University, Orem, UT, USA, 84058.
| |
Collapse
|
19
|
Shinde S, Shah S, Famta P, Wagh S, Pandey G, Sharma A, Vambhurkar G, Jain A, Srivastava S. Next-Generation Transformable Nanomedicines: Revolutionizing Cancer Drug Delivery and Theranostics. Mol Pharm 2025. [PMID: 40317253 DOI: 10.1021/acs.molpharmaceut.4c01495] [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: 05/07/2025]
Abstract
Nanomedicine has significantly advanced the treatment of various cancer phenotypes, addressing numerous challenges associated with conventional therapies. Researchers have extensively investigated the physicochemical properties of nanocarriers, such as charge, morphology, and surface chemistry, to optimize drug delivery systems. In the context of transformable nanomedicine, these properties are particularly critical for overcoming existing limitations, including suboptimal blood circulation times, sequestration by the reticuloendothelial system and mononuclear phagocyte system, and inefficient targeting of the tumor microenvironment (TME). Alterations in nanocarrier geometry, surface charge, and hydrophilicity have shown potential in mitigating these barriers, offering improved therapeutic outcomes and enhanced biomedical applications. This review explores controlled modulation of these properties in the context of anticancer therapy, offering an in-depth exploration of transformable strategies activated by both internal and external stimuli. We analyze the implications of these tunable characteristics on pharmacokinetics, biodistribution, and targeted delivery to the TME. Additionally, we address the current challenges in the clinical translation of these advanced nanocarriers and propose strategies to overcome these obstacles to enhance the clinical feasibility of nanomedicine-based cancer therapies.
Collapse
Affiliation(s)
- Swapnil Shinde
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Abhishek Sharma
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Akshita Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| |
Collapse
|
20
|
Bakir M, Dawalibi A, Mufti MA, Behiery A, Mohammad KS. Nano-Drug Delivery Systems for Bone Metastases: Targeting the Tumor-Bone Microenvironment. Pharmaceutics 2025; 17:603. [PMID: 40430894 PMCID: PMC12115183 DOI: 10.3390/pharmaceutics17050603] [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: 04/02/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Bone metastases are a prevalent and debilitating consequence of various cancers, including breast and prostate carcinomas, which significantly compromise patient quality of life due to pain, fractures, and other skeletal-related events (SREs). This review examines the pathophysiology of bone metastases, emphasizing the role of the bone microenvironment in tumor progression through mechanisms such as osteotropism and the dysregulated bone remodeling cycle. The primary focus is on the emerging nano-drug delivery systems (DDS) designed to target the bone microenvironment and improve the therapeutic index of anticancer agents. Current treatments, mainly comprising bisphosphonates and radiotherapy, provide palliative benefits but often have limited efficacy and significant side effects. Innovative strategies, such as bisphosphonate-conjugated nanoparticles and targeted therapies that utilize the unique bone marrow niche, are explored for their potential to enhance drug accumulation at metastatic sites while minimizing systemic toxicity. These approaches include the use of liposomes, polymeric nanoparticles, and inorganic nanoparticles, which can be functionalized to exploit the biological barriers within the bone microenvironment. This review also discusses the challenges and future directions for nano-DDS in clinical settings, emphasizing the need for multidisciplinary research to effectively integrate these technologies into standard care protocols.
Collapse
Affiliation(s)
- Mohamad Bakir
- Department of Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.B.); (M.A.M.)
| | - Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.D.); (A.B.)
| | - Mohammad Alaa Mufti
- Department of Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.B.); (M.A.M.)
| | - Ayman Behiery
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.D.); (A.B.)
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.D.); (A.B.)
| |
Collapse
|
21
|
Li M, Chu K, Zhou Q, Wang H, Zhang W, Zhang Y, Lv J, Zhou H, An J, Wu Z, Li S. Dual-drug loaded hyaluronic acid conjugates coated polydopamine nanodrugs for synergistic chemo-photothermal therapy in triple negative breast cancer. Int J Biol Macromol 2025; 308:142559. [PMID: 40154698 DOI: 10.1016/j.ijbiomac.2025.142559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Although combination of chemotherapy and photothermal therapy (PTT) holds significant promise for treating triple-negative breast cancer, the existing delivery systems for achieving synergistic antitumor activity remains unsatisfactory. Herein, we developed of dual-drug loaded hyaluronic acid (HA) nanodrugs, which exhibited pH, glutathione (GSH), and thermal triple-responsiveness and CD44-targeting capabilities for chemo-PTT synergistic therapy in breast cancer. Gemcitabine (GCB) and metformin (MET) were conjugated to HA via amide and disulfide bonds to form dual-drug loaded prodrugs (HSGM), which were then coated onto the surface of polydopamine nanoparticles (PDA NPs) to self-assemble into HSGM/PDA NPs. These NPs selectively accumulated at the tumor site through HA receptors and released GCB and MET in response to low pH and high GSH concentrations. The NPs demonstrated excellent photothermal performance, with heat generated from near-infrared (NIR)-laser irradiation accelerating drug release within tumor. Additionally, MET inhibited the production of heat shock protein 70 (HSP 70), mitigating thermotolerance induced by PTT, thereby enhancing the PTT effect. The combination of chemotherapy and PTT synergistically improved anti-tumor efficacy (tumor inhibition ratio: 99.11 %) while showing negligible systemic toxicity, effectively preventing tumor metastasis and recurrence. This integrated approach offers valuable insights for the clinical treatment of breast cancer and other malignant tumors.
Collapse
Affiliation(s)
- Min Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Kaile Chu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Qin Zhou
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Hongliang Wang
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Wenjun Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, Liaoning Province, PR China; School of Chemical Engineering, Dalian University of Technology, Panjin 124221, Liaoning Province, PR China
| | - Yaqiong Zhang
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Junping Lv
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Haitao Zhou
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Jie An
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| | - Zhifang Wu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| | - Sijin Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| |
Collapse
|
22
|
Huang J, Yang J, Yang Y, Lu X, Xu J, Lu S, Pan H, Zhou W, Li W, Chen S. Mitigating Doxorubicin-Induced Cardiotoxicity and Enhancing Anti-Tumor Efficacy with a Metformin-Integrated Self-Assembled Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415227. [PMID: 40052211 PMCID: PMC12061326 DOI: 10.1002/advs.202415227] [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: 11/19/2024] [Revised: 01/06/2025] [Indexed: 05/10/2025]
Abstract
Doxorubicin (Dox) is a potent chemotherapeutic agent commonly used in cancer treatment. However, cardiotoxicity severely limited its clinical application. To address this challenge, a novel self-assembled nanomedicine platform, PMDDH, is developed for the co-delivery of Dox and metformin, an antidiabetic drug with cardioprotective and anti-tumor properties. PMDDH integrates metformin into a polyethyleneimine-based bioactive excipient (PMet), with Dox intercalated into double-stranded DNA and a hyaluronic acid (HA) coating to enhance tumor targeting. The PMDDH significantly improves the pharmacokinetics and tumor-targeting capabilities of Dox, while metformin enhances the drug's anti-tumor activity by downregulating programmed cell death ligand 1 (PD-L1) and activating the AMP-activated protein kinase (AMPK) signaling pathway. Additionally, the DNA component stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which synergizes with Dox-induced immunogenic cell death (ICD) to promote a robust anti-tumor immune response. PMDDH markedly reduces Dox-induced cardiotoxicity by preserving mitochondrial function, reducing reactive oxygen species (ROS) production, and inducing protective autophagy in cardiomyocytes. These findings position PMDDH as a promising dual-function nanomedicine that enhances the anti-tumor efficacy of Dox while minimizing its systemic toxicity, offering a safer and more effective alternative for cancer therapy.
Collapse
Affiliation(s)
- Jiaxin Huang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jieru Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yuanying Yang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Xiaofeng Lu
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
- Department of CardiologyShanghai General Hospital Jiuquan HospitalNo. 22, West StJiuquanGansu735000China
| | - Juan Xu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| | - Shan Lu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Hong Pan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical PreparationsSchool of Pharmaceutical ScienceChangsha Medical UniversityChangshaHunan410219China
| | - Wenqun Li
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Songwen Chen
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| |
Collapse
|
23
|
Liu H, Shen Y. Environmental stimuli-responsive hydrogels in endodontics: Advances and perspectives. Int Endod J 2025; 58:674-684. [PMID: 39915932 PMCID: PMC11979316 DOI: 10.1111/iej.14208] [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: 08/29/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 04/10/2025]
Abstract
Stimuli-responsive hydrogels are smart and functional materials that respond to various environmental stimuli, including temperature, light, magnetic field, pH, redox, enzymes and glucose. This responsiveness allows for the controlled release of therapeutic agents encapsulated within the hydrogels, enhancing treatment precision, improving therapeutic outcomes and minimizing side effects. Such hydrogels show great potential in root canal disinfection, management of dental pulp inflammation and pulp regeneration, making them promising candidates for more personalized and effective endodontic treatments. This article provides an overview of the latest advancements in the design and application of stimuli-responsive hydrogels in endodontics, emphasizing their potential to revolutionize endodontic treatments. It also addresses current challenges and explores future directions in the field, aiming to inspire and motivate researchers to further engage in or intensify their efforts within this promising area of research.
Collapse
Affiliation(s)
- He Liu
- Division of Endodontics, Department of Oral Biological and Medical Sciences, Faculty of DentistryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ya Shen
- Division of Endodontics, Department of Oral Biological and Medical Sciences, Faculty of DentistryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
24
|
Sun C, Xie F, Zhang H, Feng L, Wang Y, Huang C, Cui Z, Luo C, Zhang L, Wang Q. Paclitaxel/Luteolin Coloaded Dual-Functional Liposomes for Esophageal Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411930. [PMID: 40265971 PMCID: PMC12120766 DOI: 10.1002/advs.202411930] [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: 09/26/2024] [Revised: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Combination therapy integrating chemotherapeutic agents with natural bioactive ingredients represents an attractive strategy for esophageal squamous cell carcinoma (ESCC) treatment, yet achieving tumor-specific co-delivery remains a critical challenge. Herein, we report that the combination of luteolin (LUT) and paclitaxel (PTX) exerts a remarkable synergy in ESCC treatment, while concurrently alleviating PTX-induced hepatotoxicity; EA2 aptamer has been identified for its exceptional specificity and strong affinity toward Catenin Alpha 1 protein (CTNNA1) in ESCC cells. Leveraging this specificity, nanosized EA2-modified pH-sensitive liposomes (EA2-PSL-PTX/LUT) are successfully developed with effective co-loading, controlled release, and good biostability. EA2-PSL-PTX/LUT exhibits stimuli-triggered release in the acidic tumor microenvironment and facilitates specific cellular uptake and endosomal escape in ESCC cells. In vivo imaging confirms precise tumor localization, deep tumor penetration, and prolonged retention of the nanocarrier. In vitro and in vivo findings validate that the nanocarrier potentiates synergistic inhibitions of PTX and LUT. Notably, EA2-PSL-PTX/LUT significantly activates the tumor microenvironment by promoting dendritic cell maturation and T cell infiltration. And the immunosuppressive microenvironment has been remodeled by decreasing myeloid-derived suppressor cells and regulatory T cell accumulation. This study provides a strategy for precise delivery of combinational chemotherapeutic drugs for ESCC targeted therapy.
Collapse
Affiliation(s)
- Congyong Sun
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Fei Xie
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Huiyun Zhang
- Department of Pharmaceutical EngineeringSchool of Chemistry and Chemical EngineeringYancheng Institute of TechnologyYanchengJiangsu224003China
| | - Lulu Feng
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Yuting Wang
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Chaofan Huang
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Zhizhen Cui
- Department of Acute Infectious Disease Control and PreventionHuai'an Center for Disease Control and PreventionHuai'anJiangsu223003China
| | - Chao Luo
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Li Zhang
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Qilong Wang
- The Comprehensive Cancer CenterDepartment of Central LaboratoryThe Affiliated Huai'an No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| |
Collapse
|
25
|
Zhao N, Shi Y, Liu P, Lv C. pH-responsive carbohydrate polymer-based nanoparticles in cancer therapy. Int J Biol Macromol 2025; 306:141236. [PMID: 39978518 DOI: 10.1016/j.ijbiomac.2025.141236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/01/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Using the specific features of the tumor microenvironment (TME) for the development and design of novel nanomaterials can improve the capacity in tumor suppression. One of the prominent features of the TME is the mild acidic pH. Therefore, the development of pH-responsive nanoparticles can lead to the release of cargo and therapeutics at the tumor site, improving the selectivity and specificity. The materials used for the development of nanoparticles should possess a number of unique features including biocompatibility and anti-cancer activity. Hence, a special attention has been directed towards the use of carbohydrate polymers in the development of nanoparticles. The carbohydrate polymers can develop smart nanoparticles respond to the pH in TME to increase targeting ability and provide controlled drug release. Such approach is also beneficial in decreasing the side effects of systemic chemotherapy. The pH-responsive nanoparticles developed from carbohydrate polymers can be also used for the combination chemotherapy/immunotherapy/phototherapy of cancer. Furthermore, these nanoparticles demonstrate theranostic application capable of cancer diagnosis and therapy. Further attention to the large-scale production, biocompatibility and long-term safety of carbohydrate polymer-based pH-responsive nanoparticles should be directed to improve the clinical translation in the treatment of cancer patients.
Collapse
Affiliation(s)
- Nanxi Zhao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yang Shi
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Pai Liu
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chengzhou Lv
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
26
|
Li Z, Xing J. Role of sirtuins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic potential. Int J Biol Macromol 2025; 310:143591. [PMID: 40300682 DOI: 10.1016/j.ijbiomac.2025.143591] [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: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
The high incidence and mortality rate of cardiac arrest (CA) establishes it as a critical clinical challenge in emergency medicine globally. Despite continuous advances in advanced life support (ALS) technology, the prognosis for patients experiencing cardiac arrest remains poor, with cerebral ischemia and reperfusion injury (CIRI) being a significant determinant of adverse neurological outcomes and increased mortality. Sirtuins (SIRTs) are a class of highly evolutionarily conserved NAD+-dependent histone deacylenzymes capable of regulating the expression of various cytoprotective genes to play a neuroprotective role in CIRI. SIRTs mainly regulate the levels of downstream proteins such as PGC 1-α, Nrf 2, NLRP 3, FoxOs, and PINK 1 to inhibit inflammatory response, attenuate oxidative stress, improve mitochondrial dysfunction, promote angiogenesis, and inhibit apoptosis while reducing CIRI. Natural active ingredients are widely used in regulating the protein level of SIRTs in the body because of their multi-components, multi-pathway, multi-target, and minimal toxic side effects. However, these naturally active ingredients still face many challenges related to drug targeting, pharmacokinetic properties, and drug delivery. The emergence and vigorous development of new drug delivery systems, such as nanoparticles, micromilk, and exosomes, provide strong support for solving the above problems. In the context of the rapid development of molecular biology technology, non-coding RNA (NcRNA), represented by miRNA and LncRNA, offers great potential for achieving gene-level precision medicine. In the context of multidisciplinary integration, combining SIRTs proteins with biotechnology, omics technologies, artificial intelligence, and material science will strongly promote the deepening of their basic research and expand their clinical application. This review describes the major signaling pathways of targeting SIRTs to mitigate CIRI, as well as the current research status of Chinese and Western medicine and medical means for the intervention level of SIRTs. Meanwhile, the challenges and possible solutions in the clinical application of targeted drugs are summarized. In the context of medical and industrial crossover, the development direction of SIRTs in the future is discussed to provide valuable reference for basic medical researchers and clinicians to improve the clinical diagnosis and treatment effects of CIRI.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
27
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
28
|
Du Z, Wei W, Lu S, Wang H, Feng C, Li Y, Cui X, Zhe J, Sun K, Liu K, Fan Q, Sun D, Bao W. PCL-PEtOx-based Crystalline-core Micelles for the Targeted Delivery of Paclitaxel and Trabectedin in Ovarian Cancer Therapy. Acta Biomater 2025:S1742-7061(25)00303-4. [PMID: 40306394 DOI: 10.1016/j.actbio.2025.04.050] [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: 01/20/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Ovarian cancer (OC), which primarily metastasizes through ascites, is both invasive and fatal. Despite its toxicity and drug resistance, the platinum-based chemotherapy Taxol®+Carboplatin has been the first-line standard treatment for decades. Trabectedin (TBD) is a recently developed, highly effective antitumor drug that is also capable of regulating tumor-associated macrophages (TAMs), however, its severe side-effects hinder further clinical application. Here, we developed safe and efficient pH-responsive crystalline-core micelles for the combined treatment of OCs, exploiting parallel delivery of paclitaxel (PTX) and TBD. PCL-PEtOx-COOH was selected as the optimal carrier to encapsulate PTX or TBD, which self-assemble into micelles with internal crystalline cores. The carboxyl group exposed on the surface of the micelles was utilized to react with the amines of Herceptin and hyaluronic acid cross-linked polymer (Herceptin-HA) to form PTX(Target). Similarly, TBD(Target) was formed by reaction with the CD206-targeted peptide mUNO. The low critical micelle concentrations of PTX(Target) and TBD(Target) stabilize the micelles in the bloodstream and normal tissues to prevent drug release. In an acidic microenvironment, the tertiary amide group on PEtOx chain of micelles ionizes, causing disassembly and pH-responsive release. Compared with Taxol®+Carboplatin, the combination therapy displayed dramatically improved safety and efficacy, as evidenced by the elimination of peritoneal tumor spheroids and reduced expression of NOX4, a gene that is overexpressed in most OC tissues. Furthermore, in human tissues, the ROS-response gene NOX4 is linked to the development of M2-type TAMs. Collectively, this study provides a safe and effective non-platinum-based chemotherapy for OC, offering an alternative to traditional Taxol®+Carboplatin. STATEMENT OF SIGNIFICANCE: (1) Significance: This work reports a new approach for ovarian cancer (OC) treatment. We utilized trabectedin (TBD) which a recently developed, highly effective antitumor drug that is also capable of regulating tumor associated macrophages (TAMs) combined with paclitaxel (PTX) to replace platinum-based chemotherapy Taxol®+Carboplatin (TC regimen). Compared to the clinical formulations, Yondelis® and Taxol®, pH-responsive PCL-PEtOx-based crystalline-core micelles were utilized for targeted independent delivery of TBD and PTX to TAMs and tumor cells, which maintained safe and efficient transport, overcoming the challenges posed by TAMs and carboplatin resistance. The system capabilities have also been confirmed in organoid and PDX models. (2) This is the first report demonstrating that this approach simultaneously overcomes the abdominal metastasis and carboplatin resistance of OC.
Collapse
Affiliation(s)
- Zixiu Du
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Wei Wei
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuli Lu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Chenxu Feng
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yinuo Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Xinyi Cui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jianan Zhe
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China
| | - Kuo Sun
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 1st Minde Road, Nanchang, Jiangxi, 330006, China
| | - Kuai Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qiong Fan
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China.
| | - Donglei Sun
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Shanghai Key Laboratory for Antibody-Drug Conjugates with Innovative Target, Shanghai, 200240, China.
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
29
|
Cao C, Wang W, Zhu S, Huang S, Fan J, Li L, Pang X, Liu L. Robust Acid-Responsive AILE Luminescence Effect Nanoparticle for Drug Release Monitoring and Induction of Apoptosis in Cancer Cells. ACS APPLIED BIO MATERIALS 2025; 8:3135-3143. [PMID: 40067751 DOI: 10.1021/acsabm.4c02003] [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: 04/22/2025]
Abstract
Through the PFOEP-SO3(-) + multidrug molecules constructed nanoparticle (NP) experiments and validated by molecular simulation docking experiments, we propose a molecular interaction principle for inducing aggregation-induced locally excited emission (AILE) luminescence from fluorenone (FO)-based conjugated polymers (CPs). Based on this molecular interaction mechanism, we constructed a NP built by π-π stacking. The NPs demonstrate facile synthesis, robust stability, and high drug-loading efficiency, enabling tumor-specific drug release in acidic lysosomal environments (pH 3.8-4.7) to minimize off-target toxicity. Concurrently, the PFOEPA NPs exhibit pH-dependent fluorescence enhancement: drug incorporation induces structural reorganization into a "sandwich" configuration, amplifying fluorescence with a blue shift under neutral/alkaline conditions, while acidic-triggered protonation collapse disrupts NPs. Moreover, it can be used as an indicator for monitoring drug release, as it is accompanied by changes in fluorescence during the drug release process. This NP possesses multiple functions and is expected to serve as an effective pH-responsive drug delivery system.
Collapse
Affiliation(s)
- Chang Cao
- School of Stomatology, Southern Medical University, Guangzhou 510515, China
| | - Wen Wang
- School of Materials and Chemistry, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shuo Zhu
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518038, China
| | - Shouhui Huang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518038, China
| | - JiYe Fan
- Hebei Chemical & Pharmaceutical College, No. 88, Fangxing Road, Shijiazhuang City, Hebei Province 050026, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou 234000, China
| | - Xinlong Pang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518038, China
| | - Lisi Liu
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, China
| |
Collapse
|
30
|
Banoo T, Ghosh A, Mishra P, Roy S, Nagarajan S. Biocompatible glycolipid derived from bhilawanol as an antibiofilm agent and a promising platform for drug delivery. RSC Med Chem 2025; 16:1715-1728. [PMID: 39925733 PMCID: PMC11799929 DOI: 10.1039/d4md00828f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Stimuli-responsive smart materials for biomedical applications have gained significant attention because of their potential for selectivity and sensitivity in biological systems. Even though ample stimuli-responsive materials are available, the use of traditional Ayurvedic compounds in the fabrication of pharmaceuticals is limited. Among various materials, gels are one of the essential classes because of their molecular-level tunability with little effort from the environment. In this study, we report a simple synthesis method for multifunctional glycolipids using a starting material derived from biologically significant natural molecules and carbohydrates in good yields. The synthesized glycolipids were prone to form a hydrogel by creating a 3D fibrous architecture. The mechanism of bottom-up assembly involving the molecular-level interaction was studied in detail using SEM, XRD, FTIR, and NMR spectroscopy. The stability, processability, and thixotropic behavior of the hydrogel were investigated through rheological measurements, and it was identified to be more suitable for biomedical applications. To evaluate the potential application of the self-assembled hydrogel in the field of medicine, we encapsulated a natural drug, curcumin, into a gel and studied its pH as a stimuli-responsive release profile. Interestingly, the encapsulated drug was released both in acidic and basic pH levels at a different rate, as identified using UV-vis spectroscopy. It is worth mentioning that the gelator used for fabricating smart soft materials displays significant potential in selectively compacting the biofilm formed by Streptococcus pneumoniae. We believe that the reported multifunctional hydrogel derived from bhilawanol-based glycolipid holds great promise in medicine.
Collapse
Affiliation(s)
- Tohira Banoo
- Assembled Organic and Hybrid Material Lab, Department of Chemistry, National Institute of Technology Warangal Hanumakonda-506004 Telangana India
| | - Abhijit Ghosh
- Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute Hyderabad - 500034 Telangana India
| | - Priyasha Mishra
- Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute Hyderabad - 500034 Telangana India
| | - Sanhita Roy
- Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute Hyderabad - 500034 Telangana India
| | - Subbiah Nagarajan
- Assembled Organic and Hybrid Material Lab, Department of Chemistry, National Institute of Technology Warangal Hanumakonda-506004 Telangana India
| |
Collapse
|
31
|
Yao W, Lin Y, Xu N, Xi Q, Liu Y, Li L. Laminarin-coated Genexol-PM pH sensitive nanomicelles targeting miR-620/IRF2BP2 axis for inhibition of cell proliferation and induction of apoptosis in Invitro thyroid carcinoma. Int J Biol Macromol 2025; 310:143198. [PMID: 40246117 DOI: 10.1016/j.ijbiomac.2025.143198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
This study explores the efficacy of Laminarin-coated Genexol-PM pH-sensitive nanomicelles targeting the miR-620/IRF2BP2 axis for cancer therapy using Dextran (BP), BP@PLGA, and BP@PLGA/PLA drug delivery systems. Among these, BP@PLGA/PLA demonstrated the highest cytotoxic potential in AGS cells, as confirmed by MTT assays, due to its advanced dual-polymer composition, which enhances drug encapsulation, stability, and targeted release in acidic tumor microenvironments. AO-EB and DAPI nuclear staining further validated these findings, showing significant apoptotic activity in BP@PLGA/PLA-treated cells, characterized by chromatin condensation, nuclear fragmentation, and apoptotic body formation. Additionally, ROS detection using carboxy-H2DCFDA staining indicated that BP@PLGA/PLA induced the highest oxidative stress levels, further driving apoptosis and disrupting cancer cell viability. In contrast, Dextran (BP) exhibited minimal cytotoxicity, and BP@PLGA showed moderate effectiveness, highlighting the superior therapeutic efficacy of BP@PLGA/PLA. The pH-sensitive nature of Laminarin-coated Genexol-PM micelles further enhanced the targeted inhibition of the miR-620/IRF2BP2 axis, improving specificity while minimizing off-target effects. By leveraging both oxidative stress mechanisms and apoptosis induction, BP@PLGA/PLA offers a promising approach for overcoming limitations in conventional chemotherapy. These findings underscore the potential of pH-responsive nanomicelles in precision oncology, offering improved drug delivery, enhanced therapeutic index, and a more effective strategy for combating drug-resistant cancers.
Collapse
Affiliation(s)
- Wei Yao
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| | - Yuhe Lin
- Department of Oncology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| | - Nan Xu
- Department of Plastic Surgery, 1st. Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Qi Xi
- The First Hospital of China Medical University, Department of Pain Medicine Shenyang, Liaoning, 110001, China
| | - Yan Liu
- The First Hospital of China Medical University, Department of Pain Medicine Shenyang, Liaoning, 110001, China
| | - Li Li
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| |
Collapse
|
32
|
Gao K, Xu K. Advancements and Prospects of pH-Responsive Hydrogels in Biomedicine. Gels 2025; 11:293. [PMID: 40277729 PMCID: PMC12026617 DOI: 10.3390/gels11040293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
As an intelligent polymer material, pH-sensitive hydrogels exhibit the capability to dynamically sense alterations in ambient pH levels and subsequently initiate corresponding physical or chemical responses, including swelling, contraction, degradation, or ion exchange. Given the significant pH variations inherent in human pathophysiological microenvironments, particularly in tumor tissues, inflammatory lesions, and the gastrointestinal system, these smart materials demonstrate remarkable application potential across diverse domains such as targeted drug delivery systems, regenerative medicine engineering, biosensing, and disease diagnostics. Recent breakthroughs in nanotechnology and precision medicine have substantially propelled advancements in the design and application of pH-responsive hydrogels. This review systematically elaborates on the current research progress and future challenges regarding pH-responsive hydrogels in biomedical applications, with particular emphasis on their stimulus-response mechanisms, fabrication methodologies, multifunctional integration strategies, and application scenarios.
Collapse
Affiliation(s)
- Ke Gao
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China;
| | - Ke Xu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China;
- Key Laboratory of Advanced Textile Materials & Manufacturing Technology, Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
33
|
Xia Q, Zhou S, Zhou J, Zhao X, Saif MS, Wang J, Hasan M, Zhao M, Liu Q. Recent Advances and Challenges for Biological Materials in Micro/Nanocarrier Synthesis for Bone Infection and Tissue Engineering. ACS Biomater Sci Eng 2025; 11:1945-1969. [PMID: 40067283 DOI: 10.1021/acsbiomaterials.4c02118] [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: 04/15/2025]
Abstract
Roughly 1.71 billion people worldwide suffer from large bone abnormalities, which are the primary cause of disability. Traditional bone grafting procedures have several drawbacks that impair their therapeutic efficacy and restrict their use in clinical settings. A great deal of work has been done to create fresh, more potent strategies. Under these circumstances, a crucial technique for the regeneration of major lesions has emerged: bone tissue engineering (BTE). BTE involves the use of biomaterials that can imitate the natural design of bone. To yet, no biological material has been able to fully meet the parameters of the perfect implantable material, even though several varieties have been created and investigated for bone regeneration. Against this backdrop, researchers have focused a great deal of interest over the past few years on the subject of nanotechnology and the use of nanostructures in regenerative medicine. The ability to create nanoengineered particles that can overcome the current constraints in regenerative strategies─such as decreased cell proliferation and differentiation, insufficient mechanical strength in biological materials, and insufficient production of extrinsic factors required for effective osteogenesis has revolutionized the field of bone and tissue engineering. The effects of nanoparticles on cell characteristics and the application of biological materials for bone regeneration are the main topics of our review, which summarizes the most recent in vitro and in vivo research on the application of nanotechnology in the context of BTE.
Collapse
Affiliation(s)
- Qipeng Xia
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Shuyan Zhou
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, PR China
| | - Jingya Zhou
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, PR China
| | - Xia Zhao
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| | - Muhammad Saqib Saif
- Department of Biochemistry, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Jianping Wang
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Murtaza Hasan
- Department of Biotechnology, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Min Zhao
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Qiang Liu
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| |
Collapse
|
34
|
Rumon MMH. Advances in cellulose-based hydrogels: tunable swelling dynamics and their versatile real-time applications. RSC Adv 2025; 15:11688-11729. [PMID: 40236573 PMCID: PMC11997669 DOI: 10.1039/d5ra00521c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
Cellulose-derived hydrogels have emerged as game-changing materials in biomedical research, offering an exceptional combination of water absorption capacity, mechanical resilience, and innate biocompatibility. This review explores the intricate mechanisms that drive their swelling behaviour, unravelling how molecular interactions and network architectures work synergistically to enable efficient water retention and adaptability. Their mechanical properties are explored in depth, with a focus on innovative chemical modifications and cross-linking techniques that enhance strength, elasticity, and functional versatility. The versatility of cellulose-based hydrogels shines in applications such as wound healing, precision drug delivery, and tissue engineering, where their biodegradability, biocompatibility, and adaptability meet the demands of cutting-edge healthcare solutions. By weaving together recent breakthroughs in their development and application, this review highlights their transformative potential to redefine regenerative medicine and other biomedical fields. Ultimately, it emphasizes the urgent need for continued research to unlock the untapped capabilities of these extraordinary biomaterials, paving the way for new frontiers in healthcare innovation.
Collapse
Affiliation(s)
- Md Mahamudul Hasan Rumon
- Department of Mathematics and Natural Sciences, Brac University 66 Mohakhali Dhaka 1212 Bangladesh
| |
Collapse
|
35
|
Xu Z, He X, Gui Y, Tang L, Zhao Y, Song L, Xu T, Chen M, Zhao Y, Du P, Wang X, Chen S, Luo Y, Luo F, Meng H, Hu J, Zhuo W, Jing J, Shi H. Intrapleural pressure-controlled piezo-catalytic nanozyme for the inhibition of malignant pleural effusion. Nat Commun 2025; 16:3194. [PMID: 40180981 PMCID: PMC11968801 DOI: 10.1038/s41467-025-58354-9] [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: 09/21/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
Malignant pleural effusion (MPE), persistently generated by thorax tumor cells at the advanced stage, remains a major challenge for cancer therapy. Herein, we develop an ultra-sensitive piezoelectric nano-system by doping ytterbium in metal-organic framework (O3P@LPYU), which can be triggered by physiological intrapleural pressure during breath. Under the gently alterative pressure, the piezoelectric nanoparticles with notable peroxidase-like activity effectively produce a burst of reactive oxygen species and induce immunogenic cell death by catalysis of carried ozone as well as peroxide in interstitial fluid. A clear and sustained biodistribution is observed in thorax effusion and tumors upon intrapleural administration of particle. Remarkably, due to the abundant substrates in oxygen-rich environment of pleural cavity, O3P@LPYU particle provides a potent reduction of MPE volume and durable inhibition of tumor growth in thorax. Our work not only develops a bio-responsive piezoelectric nano-system, but also provides a strategy for persistent suppression of MPE in clinics.
Collapse
Affiliation(s)
- Zihan Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiujing He
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yu Gui
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lingkai Tang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Yuxin Zhao
- Department of Ultrasound, the Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu City, Chengdu, Sichuan, China
| | - Linlin Song
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Tianyue Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Meixu Chen
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yujie Zhao
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Peixin Du
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Xin Wang
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Siyi Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yong Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Yingshan Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Jianping Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Wei Zhuo
- Department of Colorectal Surgery and Oncology, Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Cell Biology, Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jing Jing
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.
| | - Hubing Shi
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.
| |
Collapse
|
36
|
Li Z, Lei H, Hu J, Zhou T, Yuan S, Ma X, Zhu Y, Liu C, Wang D, Wu Y, Xu S. Inhaled pH-Responsive polymyxin B-loaded albumin nanoparticles against pneumonia caused by carbapenem resistant Klebsiella pneumoniae. Mater Today Bio 2025; 31:101590. [PMID: 40104651 PMCID: PMC11919421 DOI: 10.1016/j.mtbio.2025.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The pneumonia induced by carbapenem resistant Klebsiella pneumoniae (CRKP) has high morbidity and mortality. Among the antibiotics currently available, polymyxin B (PMB) is considered to be the last line of defense. Routine intravenous administration of PMB has many problems, such as severe neurotoxicity and nephrotoxicity. In this study, a novel inhaled PMB-loaded albumin nanoparticles (PEG-pHSA@PMB) capable of penetrating airway mucus and responding to the infection microenvironment is constructed. An acid-responsive functional molecule (PEBA) and NH2-PEG-SH are linked to the surface of human serum albumin (HSA) via the conjugation reaction. Subsequently, PMB is loaded through electrostatic interactions to yield PEG-pHSA@PMB. The sulfhydryl groups of PEG-pHSA@PMB interact with mucins to help penetrate mucus after inhaled. In an acidic environment, the protonation of the tertiary amino groups within PEG-pHSA@PMB causes the charge alteration, which leads to the release of PMB. It demonstrated excellent mucus permeability, potent bactericidal activity, and superior bacteriostatic effects compared to sole PMB. Inhalation of PEG-pHSA@PMB significantly reduced the bacterial load in the lungs of mice with CRKP pneumonia, alleviating inflammatory response. Moreover, PEG-pHSA@PMB exhibited good cytocompatibility and biosafety. The novel strategy of the inhalation drug delivery system is promising for the treatment of pneumonia caused by drug-resistant bacteria.
Collapse
Affiliation(s)
- Ziling Li
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Huiling Lei
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - Jiannan Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Tong Zhou
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - Shuaiqi Yuan
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - Xinyue Ma
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Yunfei Zhu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Chao Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Decai Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| | - Yuzhou Wu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - Shuyun Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Key Laboratory of Respiratory Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, Hubei Province, China
| |
Collapse
|
37
|
Fang B, Pan F, Shan T, Chen H, Peng W, Tian W, Huang F, Mao Z, Ding Y. An Integrated Virtual Screening Platform to Identify Potent Co-Assembled Nanodrugs for Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414154. [PMID: 39988868 DOI: 10.1002/adma.202414154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/18/2025] [Indexed: 02/25/2025]
Abstract
Co-assembled nanodrugs provide significant advantages in cancer treatment and drug delivery, yet effective screening methods to identify molecular combinations for co-assembly are lacking. This study presents a screening strategy integrating ligand-based virtual screening (LBVS) and density functional theory (DFT) calculations to explore new molecular combinations with co-assembly capabilities. The accuracy of this screening was validated by synthesizing various co-assembled nanodrugs under mild conditions. Vinpocetine (Vin) and lenvatinib (Len) are representative co-assembly combinations that can directly co-assemble into nanoparticles (NPs) through hydrogen bonding, van der Waals forces, and π-π interactions. These NPs were further functionalized with polyethylene glycol (PEG), resulting in PEG-L/V NPs that exhibited enhanced stability and biocompatibility. In addition, PEG-L/V NPs can respond to acidic conditions and release Vin and Len, working synergistically to induce cell cycle arrest and apoptosis in tumor cells in vitro while also inhibiting xenograft tumor growth in vivo. RNA sequencing (RNA-seq) analysis revealed that the co-assembled nanodrugs exhibited mechanisms that are distinct from those of single drugs. This study demonstrates the feasibility of utilizing a computational approach combining LBVS and DFT to identify small molecules with co-assembly capabilities, leading to innovative anticancer strategies.
Collapse
Affiliation(s)
- Bo Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Fei Pan
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Tianyu Shan
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Hualei Chen
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Wenjun Peng
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Wenli Tian
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, 310009, China
- Center for Medical Research and Innovation in Digestive System Tumors Ministry of Education, China, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- ZJU-Pujian Research and Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, 310058, China
| |
Collapse
|
38
|
Hegelmann M, Cokoja M. Two-Phase Epoxidations with Micellar Catalysts: Insights, Limitations, and Perspectives. Chempluschem 2025:e2500122. [PMID: 40168424 DOI: 10.1002/cplu.202500122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/03/2025]
Abstract
Biphasic molecular catalysis is a promising strategy for combining catalyst recycling with the synthesis of advanced chemical products. The anchoring of catalysts to surfactants in water allows for both catalyst solubility in aqueous media and a simple separation from the organic product. In biphasic epoxidations, this approach allows the use of environmentally benign hydrogen peroxide as oxidant. However, challenges remain due to mass transport limitations between the aqueous and organic phase, incompatibilities in the multicomponent system, and side reactions in the acidic medium. Hence, the development of surface-active catalysts that enable controlled phase separation from all other components is highlighted in this concept article.
Collapse
Affiliation(s)
- Markus Hegelmann
- Department of Chemistry and Catalysis Research Center, School of Natural Sciences, Technical University of Munich, Ernst-Otto-Fischer Straße 1, D-85748, Garching bei München, Germany
| | - Mirza Cokoja
- Department of Chemistry and Catalysis Research Center, School of Natural Sciences, Technical University of Munich, Ernst-Otto-Fischer Straße 1, D-85748, Garching bei München, Germany
| |
Collapse
|
39
|
Chen Z, Zheng X, Mu Z, Lu W, Zhang H, Yan J. Intelligent Nanomaterials Design for Osteoarthritis Managements. SMALL METHODS 2025:e2402263. [PMID: 40159773 DOI: 10.1002/smtd.202402263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disorder, characterized by progressive joint degradation, pain, and diminished mobility, all of which collectively impair patients' quality of life and escalate healthcare expenditures. Current treatment options are often inadequate due to limited efficacy, adverse side effects, and temporary symptom relief, underscoring the urgent need for more effective therapeutic strategies. Recent advancements in nanomaterials and nanomedicines offer promising solutions by improving drug bioavailability, reducing side effects and providing targeted therapeutic benefits. This review critically examines the pathogenesis of OA, highlights the limitations of existing treatments, and explores the latest innovations in intelligent nanomaterials design for OA therapy, with an emphasis on their engineered properties, therapeutic mechanisms, and translational potential in clinical application. By compiling recent findings, this work aims to inspire further exploration and innovation in nanomedicine, ultimately advancing the development of more effective and personalized OA therapies.
Collapse
Affiliation(s)
- Zhihao Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xuan Zheng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, 130022, China
| | - Weijie Lu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- Department of Orthopedics, Yanjiang Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyuan Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiao Yan
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| |
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
|
Wang J, Wang H, Zou F, Gu J, Deng S, Cao Y, Cai K. The Role of Inorganic Nanomaterials in Overcoming Challenges in Colorectal Cancer Diagnosis and Therapy. Pharmaceutics 2025; 17:409. [PMID: 40284405 PMCID: PMC12030334 DOI: 10.3390/pharmaceutics17040409] [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: 02/15/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Colorectal cancer poses a significant threat to human health due to its high aggressiveness and poor prognosis. Key factors impacting patient outcomes include post-surgical recurrence, chemotherapeutic drug resistance, and insensitivity to immunotherapy. Consequently, early diagnosis and the development of effective targeted therapies are essential for improving prevention and treatment strategies. Inorganic nanomaterials have gained prominence in the diagnosis and treatment of colorectal cancer owing to their unique size, advantageous properties, and high modifiability. Various types of inorganic nanomaterials-such as metal-based, metal oxide, quantum dots, magnetic nanoparticles, carbon-based, and rare-earth nanomaterials-have demonstrated significant potential in enhancing multimodal imaging, drug delivery, and synergistic therapies. These advancements underscore their critical role in improving therapeutic outcomes. This review highlights the properties and development of inorganic nanomaterials, summarizes their recent applications and progress in colorectal cancer diagnosis and treatment, and discusses the challenges in translating these materials into clinical use. It aims to provide valuable insights for future research and the clinical application of inorganic nanomaterials in colorectal cancer management.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Hanwenchen Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| | - Falong Zou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| | - Junnan Gu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Shenghe Deng
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Yinghao Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| |
Collapse
|
42
|
Ding H, Su L, Xie Z, Castano AD, Li Y, Perez LR, Chen J, Luo K, Tian X, Battaglia G. Morphological insights in oxidative sensitive nanocarrier pharmacokinetics, targeting, and photodynamic therapy. J Mater Chem B 2025; 13:3852-3863. [PMID: 39946164 DOI: 10.1039/d4tb02194k] [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: 03/17/2025]
Abstract
Nanoparticle (NP) morphology holds significant importance in nanomedicine, particularly concerning its implications for biological responses. This study investigates the impact of synthesizing polymers with varying degrees of methionine (MET) polymerization on three distinct drug delivery systems: spherical micelles, worm-like micelles, and vesicles, all loaded with the photosensitizer chlorin e6 (Ce6). We analyzed their distribution at both cellular and animal levels, revealing how NP morphology influences cellular uptake, subcellular localization, penetration of multicellular spheroids, blood half-life, and biodistributions across major organs. Employing a physiologically based pharmacokinetic (PBPK) model enabled us to simulate diverse distribution patterns and quantify the targeting efficiency of NPs toward tumors. Our investigation elucidates that spherical micelles exhibit lower accumulation levels within the reticuloendothelial system, potentially mitigating adverse side effects despite their higher glomerular filtration rate. This nuanced understanding underscores the complex interplay between NP morphology and biological responses, providing valuable insights into optimizing therapeutic efficacy while minimizing undesirable effects. We thus report the integration of experimental analyses with PBPK modeling to elucidate the topological characteristics of NP, thereby shedding light on their distribution patterns, therapeutic efficacy, and potential side effects.
Collapse
Affiliation(s)
- Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Liping Su
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Zhendong Xie
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Electronic and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - Aroa Duro Castano
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Lorena Ruiz Perez
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Applied Physics, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - Junyang Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Xiaohe Tian
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Giuseppe Battaglia
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Electronic and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluís Companys, 23, 08010, Barcelona, Spain
| |
Collapse
|
43
|
Fu Z, Wang S, Zhou X, Ouyang L, Chen Z, Deng G. Harnessing the Power of Traditional Chinese Medicine in Cancer Treatment: The Role of Nanocarriers. Int J Nanomedicine 2025; 20:3147-3174. [PMID: 40103746 PMCID: PMC11913986 DOI: 10.2147/ijn.s502104] [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: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
For centuries, traditional Chinese medicine (TCM) has had certain advantages in the treatment of tumors. However, due to their poor water solubility, low bioavailability and potential toxicity, their effective delivery to target sites can be a major challenge. Nanocarriers based on the active ingredients of TCM, such as liposomes, polymer nanoparticles, inorganic nanoparticles, and organic/inorganic nanohybrids, are a promising strategy to improve the delivery of TCM, resulting in higher therapeutic outcomes and fewer side effects. Therefore, this article intends to review the application of Chinese medicine nano preparation in tumor. Firstly, we introduce the classification and synthesis of nanometer preparations of Chinese medicine. The second part mainly introduces the different responses of TCM nano-preparations in the course of treatment to introduce how TCM nano-preparations play a role in anti-tumor therapy. The third part focuses on Different response modes of Chinese medicine nano preparations in tumor therapy. The fourth part elucidates the application of Chinese medicine nano preparations in the treatment of cancer. Finally, the research direction to be explored in related fields is put forward.
Collapse
Affiliation(s)
- Ziyu Fu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Xin Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Linqi Ouyang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Zhen Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
- The second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410005, People’s Republic of China
| |
Collapse
|
44
|
Guo H, Wang H, Zhang Y, Deng H, Tung CH, Zhang W. Iron-Doxorubicin Hyaluronan Nanogel as an Effective Targeted Chemotherapy with Modulated Cardiotoxicity. Biomacromolecules 2025; 26:1614-1626. [PMID: 39908570 DOI: 10.1021/acs.biomac.4c01397] [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: 02/07/2025]
Abstract
The complex of doxorubicin (Dox) and intracellular iron in cardiomyocytes generates reactive oxygen species (ROS), contributing to commonly observed cardiotoxicity. To enhance the anticancer potency and minimize the cardiotoxicity, here Dox was formulated into a hyaluronan (HA) nanogel using ferric ion (Fe3+) coordination to control the intracellular distribution and release of Dox. Taking advantage of the paramagnetic properties of iron and the fluorescence of Dox, we conveniently monitored the targeted delivery of the HA@Dox nanogel in murine breast tumors through both T1-weighted magnetic resonance imaging and fluorescence imaging. Compared with free Dox, HA@Dox nanogel affords a CD44-targeted delivery, lysosomal distribution, pH-responsive release, and significant tumor inhibition. Mechanistically, the lysosome-enriched HA@Dox produces ROS, causing lysosomal membrane permeabilization, which further promotes the intracellular Dox distribution. This HA@Dox nanogel not only provides a facile cancer-targeted delivery but also successfully relieves Dox toxicity, representing a potent delivery system for Dox.
Collapse
Affiliation(s)
- Hua Guo
- State Key Laboratory of Molecular Oncology and Department of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P. R. China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P. R. China
| | - Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P. R. China
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10065, United States
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P. R. China
| |
Collapse
|
45
|
Dai W, Song X, Wang R, He W, Yin J, Nie S. Mechanism exploration of intestinal mucus penetration of nano-Se: regulated by polysaccharides with different functional groups and molecular weights. J Control Release 2025; 379:524-536. [PMID: 39809422 DOI: 10.1016/j.jconrel.2025.01.023] [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: 09/23/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Selenium deficiency associated with a high risk of many diseases remains a global challenge. Owing to the narrow margin between "nutrition-toxicity" doses of selenium, it is imperative to achieve accurate selenium supplement. Nano‑selenium (SeNPs) is a novel form of selenium supplement with low toxicity, but it could be trapped and removed by intestinal mucus, thus limiting its oral delivery. The mucus penetration of SeNPs is highly associated with interactions between SeNPs and mucin (the structural component of mucus). In this study, we selected four polysaccharides with different functional groups and molecular weights, i.e. chitosan oligosaccharide (COS), chitosan (CS), chitosan quaternary ammonium salt (HACC), and carboxymethyl cellulose (CMC) as templates to modify SeNPs. Then we systematically explored the non-covalent interactions between polysaccharides stabilized nano-Se (PS-SeNPs) and mucin, determined and examined mucus penetration behavior and mechanism of different PS-SeNPs by coarse-grained molecular dynamics simulations, both in vitro and in vivo. It could be observed that penetration of PS-SeNPs depends on their distinct surface properties and mucus pH conditions. COS-SeNPs with short oligosaccharide chains accumulated and bridged with mucin, hindering its mucus penetration at pH 7.4. While HACC-SeNPs with NH3+ and N+ exhibited high binding affinity with mucin, inducing its mucus penetration. The negatively charged CMC-SeNPs diffused freely in mucus due to their electrostatic-repelled interaction and hydrophobic interaction with mucin. This study establishes a theoretical foundation for precise application of SeNPs in oral administration and offers valuable insights into the precise utilization of polysaccharides as tailored carriers of nanoparticles in mucus-covered tissues.
Collapse
Affiliation(s)
- Wanting Dai
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Xiaoxiao Song
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Rui Wang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Weiwei He
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Junyi Yin
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| |
Collapse
|
46
|
Zeng H, Zeng P, Baek J, Kim B, Müllner M. Self-Assembly of Amorphous 2D Polymer Nanodiscs with Tuneable Size, pH-Responsive Degradation and Controlled Drug Release. Angew Chem Int Ed Engl 2025; 64:e202424269. [PMID: 39791445 PMCID: PMC11878338 DOI: 10.1002/anie.202424269] [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: 12/11/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
Amphiphilic bottlebrush block copolymers (BBCs) with tadpole-like, coil-rod architecture can be used to self-assemble into functional polymer nanodiscs directly in water. The hydrophobic segments of the BBC were tuned via the ratio of ethoxy-ethyl glycidyl ether (EE) to tetrahydropyranyl glycidyl ether (TP) within the grafted polymer sidechains. In turn, this variation controlled the sizes, pH-responsiveness, and drug loading capacity of the self-assembled nanodiscs. Notably, as EE exhibited faster hydrolysis than TP, the nanodiscs featured variable degradation rates under mild acidic conditions, aiding small molecule release and time-dependent and complete degradation of discs into fully water-soluble copolymers. The nanodiscs demonstrated biocompatibility and cellular uptake by breast cancer cells, underscoring their potential development into drug delivery systems.
Collapse
Affiliation(s)
- Haoxiang Zeng
- Key Centre for Polymers and ColloidsSchool of ChemistryThe University of SydneySydney2006NSWAustralia
| | - Ping Zeng
- Key Centre for Polymers and ColloidsSchool of ChemistryThe University of SydneySydney2006NSWAustralia
| | - Jinsu Baek
- Department of ChemistryYonsei UniversitySeoul03722Korea
| | - Byeong‐Su Kim
- Department of ChemistryYonsei UniversitySeoul03722Korea
| | - Markus Müllner
- Key Centre for Polymers and ColloidsSchool of ChemistryThe University of SydneySydney2006NSWAustralia
- The University of Sydney Nano Institute (Sydney Nano)The University of SydneySydney2006NSWAustralia
| |
Collapse
|
47
|
Li J, Mao K, Meng X, Wang J, Zhao M, Lv Y, Xin Y, Sun H, Zhang Y, Yang YG, Sun T. Injectable hydrogel-assisted local lipopolysaccharide delivery improves immune checkpoint blockade therapy. Acta Biomater 2025; 194:153-168. [PMID: 39827003 DOI: 10.1016/j.actbio.2025.01.021] [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: 07/17/2024] [Revised: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Tumor-associated macrophages (TAMs) significantly influence the clinical outcomes of immune checkpoint blockade (ICB) therapy. Strategies aimed at reprogramming TAMs from the immunosuppressive M2 phenotype to the pro-inflammatory M1 phenotype hold promise for enhancing ICB efficacy. Lipopolysaccharide (LPS), a potent Toll-like receptor 4 (TLR4) ligand, can reprogram TAMs toward an M1 phenotype. However, the systemic application of LPS is restricted due to its pronounced pro-inflammatory properties, which limit safe dosing in cancer treatment. To address this, thermosensitive hydrogels offer a viable solution by optimizing drug bioavailability and reducing systemic dissemination. In our study, carboxymethyl chitosan (CS) was incorporated into Pluronic F127 to extend the hydrogel's degradation period, facilitating the localized delivery and accumulation of LPS within tumor sites. Peritumoral injection of this hydrogel enhanced the tumor-inhibitory effects of anti-PD-1 antibodies, significantly improving the survival of 4T1 tumor-bearing mice. The GelF127CS-LPS hydrogel also increased the expression of the activation marker on tumor-infiltrating dendritic cells, promoted a higher M1/M2 TAM ratio, and enhanced CD8+ T cell infiltration into tumors-key indicators of T-cell-mediated anti-tumor immunity. Notably, no significant liver or hematological toxicity was observed with GelF127CS-LPS treatment, underscoring its favorable safety profile. These findings demonstrate the potential of GelF127CS-LPS as a TAMs-modulating agent and a promising combinatorial strategy to boost ICB therapy effectiveness. STATEMENT OF SIGNIFICANCE: LPS, a potent TLR4 ligand, can reprogram tumor-associated macrophages (TAMs) toward an M1 phenotype, thereby contributing to tumor inhibition. However, its anti-tumor application is constrained by the contradiction between high-dose toxicity and insufficient efficacy at low doses. To address this issue, we developed a thermosensitive hydrogel encapsulating LPS, GelF127CS-LPS, which allows localized LPS release within the tumor area. This hydrogel reprograms TAMs at a picogram level of LPS to achieve a favorable M1/M2 ratio and promotes the activation of T cell-mediated antitumor immunity without observable toxicity. Consequently, when combined with immune checkpoint blockade (ICB), the hydrogel can inhibit tumor growth and improve overall survival. This study provides an effective method for tumor-targeted therapeutic LPS delivery to enhance the efficacy of ICB.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Mengfei Zhao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Yue Lv
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Huating Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China; International Center of Future Science, Jilin University, Changchun, Jilin, PR China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China; International Center of Future Science, Jilin University, Changchun, Jilin, PR China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, PR China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, PR China; International Center of Future Science, Jilin University, Changchun, Jilin, PR China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, PR China.
| |
Collapse
|
48
|
Wang J, Gu X, Gao X, Chen J, Lv Z, Zhang S, Ni S, Shi F, Chen X, Cao L, Wang Z, Xiao W. Formulation and optimization of glycyrrhetinic acid-modified pH-sensitive curcumin liposomes for anti-hepatocellular carcinoma. Pharm Dev Technol 2025; 30:233-245. [PMID: 39935270 DOI: 10.1080/10837450.2025.2465549] [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: 07/26/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
In order to enhance the therapeutic value of curcumin in liver cancer treatment, glycyrrhetinic acid-modified pH-sensitive curcumin liposomes (GA-pH-Lip@Cur) was developed.GA-pH-Lip@Cur was prepared using a thin film dispersion ultrasonication method, and the optimal formulation process was selected through single-factor experiments and a Box-Behnken design-response surface methodology. The liposomes were evaluated for their morphological appearance, particle size, in vitro release at different pH levels, and biocompatibility. The anti-tumor effect of GA-pH-Lip@Cur was assessed using cell viability assays (CCK-8). The in vivo hepatic targeting and anti-liver tumor efficacy of GA-pH-Lip@Cur were evaluated through pharmacokinetic and pharmacological experiments. The results indicated that optimized GA-pH-Lip@Cur exhibited uniform particle size distribution, good stability, pH-sensitive in vitro release with sustained behavior. Compared to conventional liposomes, GA-pH-Lip@Cur showed prolonged average retention time in vivo and significantly increased curcumin distribution in liver tissues, indicating excellent liver targeting. Both in vitro and in vivo evaluations demonstrated the effectiveness of GA-pH-Lip@Cur in inhibiting liver cancer cell proliferation and suppressing liver tumor growth in tumor-bearing mice. In conclusion, GA-pH-Lip@Cur, by leveraging the acidic tumor microenvironment and overexpression of glycyrrhetinic acid receptors in liver cells, encapsulates curcumin to improve its bioavailability, and target its delivery to the liver tumor sites.
Collapse
Affiliation(s)
- Jie Wang
- Nanjing University of Chinese Medicine, Nanjing, China
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Xuemei Gu
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Xia Gao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Jing Chen
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Zhiyang Lv
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyu Zhang
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Siyu Ni
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Fei Shi
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Xialin Chen
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Liang Cao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Zhenzhong Wang
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Wei Xiao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| |
Collapse
|
49
|
Zhang C, Yuan Y, Xia Q, Wang J, Xu K, Gong Z, Lou J, Li G, Wang L, Zhou L, Liu Z, Luo K, Zhou X. Machine Learning-Driven Prediction, Preparation, and Evaluation of Functional Nanomedicines Via Drug-Drug Self-Assembly. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415902. [PMID: 39792782 PMCID: PMC11884566 DOI: 10.1002/advs.202415902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Indexed: 01/12/2025]
Abstract
Small molecules as nanomedicine carriers offer advantages in drug loading and preparation. Selecting effective small molecules for stable nanomedicines is challenging. This study used artificial intelligence (AI) to screen drug combinations for self-assembling nanomedicines, employing physiochemical parameters to predict formation via machine learning. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are identified as effective carriers for antineoplastic drugs, with high drug loading. Nanomedicines, PEG-coated indomethacin/paclitaxel nanomedicine (PiPTX), and laminarin-modified indomethacin/paclitaxel nanomedicine (LiDOX), are developed with extended circulation and active targeting functions. Indomethacin/paclitaxel nanomedicine iDOX exhibits pH-responsive drug release in the tumor microenvironment. These nanomedicines enhance anti-tumor effects and reduce side effects, offering a rapid approach to clinical nanomedicine development.
Collapse
Affiliation(s)
- Chengyuan Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative MedicineKunming Medical UniversityKunming650500China
| | - Yuchuan Yuan
- School of MedicineNorthwest UniversityXi'an710068China
| | - Qiong Xia
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Junjie Wang
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Kangkang Xu
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Zhiwei Gong
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Jie Lou
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Gen Li
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Lu Wang
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Li Zhou
- Department of Biomedical EngineeringSchool of EngineeringChina Pharmaceutical UniversityNanjing210009China
| | - Zhirui Liu
- Department of PharmacyXinan HospitalArmy Medical UniversityChongqing400038China
| | - Kui Luo
- Department of Radiologyand Department of GeriatricsHuaxi MR Research Center (HMRRC)National Clinical Research Center for Geriatrics Frontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityNo. 37 Guoxue AlleyChengdu610041China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative MedicineKunming Medical UniversityKunming650500China
| |
Collapse
|
50
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [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: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|