1
|
Wang X, Song B, Wu M, Qin L, Liang W. Immune cell targeting-mediated cytomimetic drug delivery system for BBB-penetrating and precise therapy of in situ glioma. Mater Today Bio 2025; 32:101694. [PMID: 40225137 PMCID: PMC11986483 DOI: 10.1016/j.mtbio.2025.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas are a group of highly malignant tumors that are prone to recurrence after surgery. Due to the limitation of the blood-brain barrier (BBB), most antitumor drugs cannot cross it. Therefore, improving the delivery efficiency of antitumor drugs in their treatment remains a significant challenge. Herein, we report a unique cellular biomimetic drug delivery system (CTP@RAW) that benefits from the exceptional immune homing and long-term tracking ability of RAW 264.7 cells to specifically penetrate BBB and target tumor sites. The drug (TMZ) is encapsulated in RAW264.7 to avoid being cleared or degraded by the blood, improve bioavailability and reduce systemic toxicity. And that, owning to polydopamine (PDA) coating on the quantum dots-drug nanoparticles, which can endogenously and controllably release TMZ in response to certain tumor microenvironment (high GSH and low pH). This delivery system can also achieve precise localization and real-time visualization of tumors via fluorescence imaging. The released drugs effectively inhibit tumor growth by regulating cytokine expression levels, including GFAP, Ki67, Caspase-3, and TNF-α. Our study demonstrates that this drug delivery system can cross BBB, improve drug delivery efficiency, and has excellent potential for visualization and precision treatment of in situ gliomas.
Collapse
Affiliation(s)
- Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, China
- State Laboratory of Advanced Drug Delivery and Control Release System, Shandong First Medical University, China
| | - Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, China
| | - Mengru Wu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, China
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, China
- State Laboratory of Advanced Drug Delivery and Control Release System, Shandong First Medical University, China
- Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong Academy of Medical Sciences, China
| |
Collapse
|
2
|
Zhou M, Wang Y, Li C, Li J, Dou R, Jiang H, Zhang Y, Chen D, Zhang J, Zhu S. Neutrophil hitchhiking liposomal drugs for starvation therapy in endometriosis. Theranostics 2025; 15:4848-4860. [PMID: 40225582 PMCID: PMC11984412 DOI: 10.7150/thno.107758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Endometriosis, characterized by the presence of ectopic endometrial-like tissue, is a common chronic inflammatory disorder in gynecology. However, current treatments, including surgery and hormone therapy, often yield suboptimal outcomes and significant adverse effects. The lack of a drug delivery system specifically targeting ectopic lesions remains a major barrier to the development of more effective treatments. Methods: To exploit the continuous recruitment of neutrophils and elevated reactive oxygen species (ROS) levels within endometriotic lesions, we designed a ROS-responsive liposome (cLipo) modified with neutrophil-targeting peptides on its surface. A minimally invasive mouse model of endometriosis was used to assess the neutrophil-mediated delivery of cLipo. Additionally, we co-loaded the glucose analog 2-deoxy-D-glucose (2-DG) and the autophagy inhibitor chloroquine (CQ) into the liposomes, resulting in the formulation termed cLipo-DC. The therapeutic efficacy of cLipo-DC was evaluated using human endometriotic cells (12Z), endometrial cancer cells (Ishikawa), and an endometriosis mouse model. Results: Although neutrophil hitchhiking strategies are rarely reported in chronic inflammatory diseases, we found that cLipo specifically bound to neutrophils and accumulated in ectopic lesions following intraperitoneal injection in the endometriosis mouse model. Subsequently, in vitro experiments showed that cLipo-DC effectively inhibited glycolysis and autophagy in both 12Z and Ishikawa cells, resulting in cell death. Furthermore, in vivo administration of cLipo-DC in the endometriosis mouse model exerted a significant anti-endometriosis effect, with no detectable side effects. Conclusions: This study provides a novel neutrophil hitchhiking platform for non-hormonal treatment of endometriosis.
Collapse
Affiliation(s)
- Mengni Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ying Wang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
| | - Caihua Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jinfu Li
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
| | - Renbin Dou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Huanhuan Jiang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yunjiao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 510006, China
| | - Dawei Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jiqian Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
| | - Shasha Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| |
Collapse
|
3
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [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: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University 75123 Uppsala, Sweden.
| |
Collapse
|
4
|
Zhou M, Tang Y, Lu Y, Zhang T, Zhang S, Cai X, Lin Y. Framework Nucleic Acid-Based and Neutrophil-Based Nanoplatform Loading Baicalin with Targeted Drug Delivery for Anti-Inflammation Treatment. ACS NANO 2025; 19:3455-3469. [PMID: 39817852 DOI: 10.1021/acsnano.4c12917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Targeted drug delivery is a promising strategy for treating inflammatory diseases, with recent research focusing on the combination of neutrophils and nanomaterials. In this study, a targeted nanodrug delivery platform (Ac-PGP-tFNA, APT) was developed using tetrahedral framework nucleic acid (tFNA) along with a neutrophil hitchhiking mechanism to achieve precise delivery and anti-inflammatory effects. The tFNA structure, known for its excellent drug-loading capacity and cellular uptake efficiency, was used to carry a therapeutic agent─baicalin. The results demonstrate that the development of this drug delivery platform not only considerably enhances the bioavailability and effective concentration of the drug (baicalin) but also promotes the polarization of pro-inflammatory M1 macrophages to anti-inflammatory M2 macrophages by modulating the interactions between the neutrophils and macrophages. This targeted therapeutic method effectively treats inflammatory conditions such as sepsis and introduces a strategy for managing inflammatory diseases characterized by neutrophil infiltration.
Collapse
Affiliation(s)
- Mi Zhou
- 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, Sichuan 610041, China
| | - Yuanlin Tang
- 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, Sichuan 610041, China
| | - Yifei Lu
- 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, Sichuan 610041, China
| | - Tianxu Zhang
- 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, Sichuan 610041, China
| | - Shunhao Zhang
- 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, Sichuan 610041, China
| | - Xiaoxiao Cai
- 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, Sichuan 610041, China
| | - Yunfeng Lin
- 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, Sichuan 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
5
|
He J, Li J, Li M, He Z, Ye Y, Li J, Rao J, Zhao X, Li M, He Q. Platelet backpacking nanoparticles based on bacterial outer membrane vesicles enhanced photothermal-immune anti-tumor therapy. NANOSCALE 2025; 17:1510-1523. [PMID: 39621096 DOI: 10.1039/d4nr02757d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Bacterial outer membrane vesicles (OMVs), produced by Gram-negative bacteria, retain the immunostimulatory capacity of parental bacteria. OMVs have been recognized as potent natural immune adjuvants and drug delivery vehicles. Photothermal therapy that triggers immunogenic cell death further stimulates the immune system by releasing damage-associated molecular patterns. This therapeutic effect can be synergized with OMVs to achieve enhanced anti-tumor outcomes. We also observed that tumors can recruit platelets. Leveraging the phenomenon, we have innovatively employed platelets as "couriers" to boost the tumor-targeting delivery efficiency of both OMVs and photothermal agents. In detail, based on OMVs, we meticulously engineered nanoparticles (IR780-SLN@O-P) with platelet-binding capacity. These "courier" platelets carry "cargo" IR780-SLN@O-P NPs to tumor sites via P-selectin, ensuring targeted delivery. Under laser irradiation, the photothermal agents generate significant photothermal effects, which combined with the immune-stimulating properties of OMVs, creating a robust anti-tumor immune response. For "cold" tumors such as triple-negative breast cancer (TNBC), our IR780-SLN@O-P NPs not only prolonged the survival of mice bearing 4T1 orthotopic tumors, but also significantly suppressed tumor growth. Moreover, they facilitated dendritic cell maturation and the infiltration of CD8+ T cells to ameliorate the immunosuppressive tumor environment. Our research aims to highlight the unique advantages of OMVs and explore the potential of the tumor-targeting strategy that synergizes photothermal therapy with immunotherapy. We hope that our findings can offer insights into TNBC clinical treatments.
Collapse
Affiliation(s)
- Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jiayu Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Min Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhidi He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Yunxia Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jingdong Rao
- Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Jin C, Xue L, Zhang L, Yu L, Wu P, Qian H. Engineered Nanoparticles for Theranostic Applications in Kidney Repair. Adv Healthc Mater 2025; 14:e2402480. [PMID: 39617999 DOI: 10.1002/adhm.202402480] [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: 07/06/2024] [Revised: 11/05/2024] [Indexed: 01/03/2025]
Abstract
Kidney diseases are characterized by their intricate nature and complexity, posing significant challenges in their treatment and diagnosis. Nanoparticles (NPs), which can be further classified as synthetic and biomimetic NPs, have emerged as promising candidates for treating various diseases. In recent years, the development of engineered nanotherapeutics has focused on targeting damaged tissues and serving as drug delivery vehicles. Additionally, these NPs have shown superior sensitivity and specificity in diagnosis and imaging, thus providing valuable insights for the early detection of diseases. This review aims to focus on the application of engineered synthetic and biomimetic NPs in kidney diseases in the aspects of treatment, diagnosis, and imaging. Notably, the current perspectives and challenges are evaluated, which provide inspiration for future research directions, and encourage the clinical application of NPs in this field.
Collapse
Affiliation(s)
- Can Jin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Lingling Xue
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Leilei Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Lixia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Peipei Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
7
|
Song X, Wei J, Cai X, Liu Y, Wu F, Tong S, Li S, Yao Q, Xie J, Yang H. Organotropic Engineering of Luminescent Gold Nanoclusters for In Vivo Imaging of Lung Orthotopic Tumors. ACS NANO 2024; 18:33555-33565. [PMID: 39587391 DOI: 10.1021/acsnano.4c11509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Gold nanoclusters (AuNCs) are emerging as promising functional probes for bioapplications. However, because of rapid renal clearance, it is a challenge to tailor their biofate and improve their disease-targeting ability in vivo. Herein, we report an efficient strategy to tailor their organotropic actions by rationally designing AuNC assemblies. The nanocluster assembly is established based on the moderate electrostatic interaction or strong coordination between AuNCs, enabled by solely chitosan (CS) or the coadded chelating metal ions (e.g., Gd3+). We show that AuNCs-CS is rapidly excreted into urine, while further coordination of Gd3+ confers assemblies with liver and lung accumulation capabilities, dependent on Gd3+ contents. The organotropic actions are unraveled to result from their tunable stability in vivo and binding capability to cells/proteins. We also demonstrate that lung-targeting assemblies can enable specific NIR-II luminescence imaging of lung orthotopic tumors, which cannot be realized by employing discrete AuNCs. We anticipate that these findings will offer insights into the design principles of metal nanocluster probes and related bioapplications.
Collapse
Affiliation(s)
- Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Jing Wei
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Xiyang Cai
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yizhuo Liu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Fengbo Wu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Shufen Tong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Shihua Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Qiaofeng Yao
- Key Laboratory of Organic Integrated Circuits, Ministry of Education, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| |
Collapse
|
8
|
Tian H, Yao J, Ba Q, Meng Y, Cui Y, Quan L, Gong W, Wang Y, Yang Y, Yang M, Gao C. Cerebral biomimetic nano-drug delivery systems: A frontier strategy for immunotherapy. J Control Release 2024; 376:1039-1067. [PMID: 39505218 DOI: 10.1016/j.jconrel.2024.10.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Brain diseases are a significant threat to human health, especially in the elderly, and this problem is growing as the aging population increases. Efficient brain-targeted drug delivery has been the greatest challenge in treating brain disorders due to the unique immune environment of the brain, including the blood-brain barrier (BBB). Recently, cerebral biomimetic nano-drug delivery systems (CBNDSs) have provided a promising strategy for brain targeting by mimicking natural biological materials. Herein, this review explores the latest understanding of the immune microenvironment of the brain, emphasizing the immune mechanisms of the occurrence and progression of brain disease. Several brain targeting systems are summarized, including cell-based, exosome-based, protein-based, and microbe-based CBNDSs, and their immunological mechanisms are highlighted. Moreover, given the rise of immunotherapy, the latest applications of CBNDSs in immunotherapy are also discussed. This review provides a comprehensive understanding of CBNDSs and serves as a guideline for immunotherapy in treating brain diseases. In addition, it provides inspiration for the future of CBNDSs.
Collapse
Affiliation(s)
- Hao Tian
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Jiaxin Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Qi Ba
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Yuanyuan Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liangzhu Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
9
|
He A, Huang Y, Cao C, Li X. Advances in drug delivery systems utilizing blood cells and their membrane-derived microvesicles. Drug Deliv 2024; 31:2425156. [PMID: 39520082 PMCID: PMC11552282 DOI: 10.1080/10717544.2024.2425156] [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: 08/01/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The advancement of drug delivery systems (DDSs) in recent decades has demonstrated significant potential in enhancing the efficacy of pharmacological agents. Despite the approval of certain DDSs for clinical use, challenges such as rapid clearance from circulation, toxic accumulation in the body, and ineffective targeted delivery persist as obstacles to successful clinical application. Blood cell-based DDSs have emerged as a popular strategy for drug administration, offering enhanced biocompatibility, stability, and prolonged circulation. These DDSs are well-suited for systemic drug delivery and have played a crucial role in formulating optimal drug combinations for treating a variety of diseases in both preclinical studies and clinical trials. This review focuses on recent advancements and applications of DDSs utilizing blood cells and their membrane-derived microvesicles. It addresses the current therapeutic applications of blood cell-based DDSs at the organ and tissue levels, highlighting their successful deployment at the cellular level. Furthermore, it explores the mechanisms of cellular uptake of drug delivery vectors at the subcellular level. Additionally, the review discusses the opportunities and challenges associated with these DDSs.
Collapse
Affiliation(s)
- Andong He
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Yuye Huang
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Liu C, Gao J, Cheng Y, Zhang S, Fu C. Homologous-adhering/targeting cell membrane- and cell-mediated delivery systems: a cancer-catch-cancer strategy in cancer therapy. Regen Biomater 2024; 12:rbae135. [PMID: 39811105 PMCID: PMC11729729 DOI: 10.1093/rb/rbae135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Low tumor enrichment remains a serious and urgent problem for drug delivery in cancer therapy. Accurate targeting of tumor sites is still a critical aim in cancer therapy. Though there have been a variety of delivery strategies to improve the tumor targeting and enrichment, biological barriers still cause most delivered guests to fail or be excreted before they work. Recently, cell membrane-based systems have attracted a huge amount of attention due to their advantages such as easy access, good biocompatibility and immune escape, which contribute to their biomimetic structures and specific surface proteins. Furthermore, cancer cell membrane-based delivery systems are referred to as homologous-targeting function in which they exhibit significantly high adhesion and internalization to homologous-type tumor sites or cells even though the exact mechanism is not entirely revealed. Here, we summarize the sources and characterizations of cancer cell membrane systems, including reconstructed single or hybrid membrane-based nano-/microcarriers, as well as engineered cancer cells. Additionally, advanced applications of these cancer cell membrane systems in cancer therapy are categorized and summarized according to the components of membranes. The potential factors related to homologous targeting of cancer cell membrane-based systems are also discussed. By discussing the applications, challenges and opportunities, we expect the cancer cell membrane-based homologous-targeting systems to have a far-reaching development in preclinic or clinics.
Collapse
Affiliation(s)
- Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Jingjie Gao
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Yuying Cheng
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shanshan Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Caiyun Fu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
11
|
Moulton C, Baroni A, Quagliarini E, Leone L, Digiacomo L, Morotti M, Caracciolo G, Podda MV, Tasciotti E. Navigating the nano-bio immune interface: advancements and challenges in CNS nanotherapeutics. Front Immunol 2024; 15:1447567. [PMID: 39600701 PMCID: PMC11588692 DOI: 10.3389/fimmu.2024.1447567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
In recent years, significant advancements have been made in utilizing nanoparticles (NPs) to modulate immune responses within the central nervous system (CNS), offering new opportunities for nanotherapeutic interventions in neurological disorders. NPs can serve as carriers for immunomodulatory agents or platforms for delivering nucleic acid-based therapeutics to regulate gene expression and modulate immune responses. Several studies have demonstrated the efficacy of NP-mediated immune modulation in preclinical models of neurological diseases, including multiple sclerosis, stroke, Alzheimer's disease, and Parkinson's disease. While challenges remain, advancements in NPs engineering and design have led to the development of NPs using diverse strategies to overcome these challenges. The nano-bio interface with the immune system is key in the conceptualization of NPs to efficiently act as nanotherapeutics in the CNS. The biomolecular corona plays a pivotal role in dictating NPs behavior and immune recognition within the CNS, giving researchers the opportunity to optimize NPs design and surface modifications to minimize immunogenicity and enhance biocompatibility. Here, we review how NPs interact with the CNS immune system, focusing on immunosurveillance of NPs, NP-induced immune reprogramming and the impact of the biomolecular corona on NPs behavior in CNS immune responses. The integration of NPs into CNS nanotherapeutics offers promising opportunities for addressing the complex challenges of acute and chronic neurological conditions and pathologies, also in the context of preventive and rehabilitative medicine. By harnessing the nano-bio immune interface and understanding the significance of the biomolecular corona, researchers can develop targeted, safe, and effective nanotherapeutic interventions for a wide range of CNS disorders to improve treatment and rehabilitation. These advancements have the potential to revolutionize the treatment landscape of neurological diseases, offering promising solutions for improved patient care and quality of life in the future.
Collapse
Affiliation(s)
| | - Anna Baroni
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
| | - Erica Quagliarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Leone
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Morotti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Vittoria Podda
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ennio Tasciotti
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, Università telematica San Raffaele, Rome, Italy
| |
Collapse
|
12
|
Pashirova T, Shaihutdinova Z, Tatarinov D, Titova A, Malanyeva A, Vasileva O, Gabdurakhmanov K, Dudnikov S, Schopfer LM, Lockridge O, Masson P. Pharmacokinetics and fate of free and encapsulated IRD800CW-labelled human BChE intravenously administered in mice. Int J Biol Macromol 2024; 282:137305. [PMID: 39515732 DOI: 10.1016/j.ijbiomac.2024.137305] [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: 08/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Human butyrylcholinesterase (BChE) is an efficient bioscavenger of toxicants. Highly purified BChE was labelled with the near infrared fluorescent IRDye800CW. The goal was to determine the pharmacokinetics and fate of enzyme in mice. BChE-IRDye800CW was encapsulated in polyethylene glycol-polypropylene sulfide-based spherical polymersome nanoreactors with the following characteristics: 140 nm diameter, ξ = -6 mV, PDI ≤ 0.2, 1 year stability. Encapsulation did not alter the functional properties of BChE. Free and encapsulated enzyme were injected intravenously to CD-1 mice (single dose of enzyme 1.5 mg/kg and PEG-PPS polymersomes 25 mg/kg) and were analyzed for 8 days using an in vivo imaging system. Results showed that the pharmacokinetic distribution α-phase of encapsulated BChE (t1/2 = 17.6 h) was longer than for free enzyme (t1/2 = 6.6 h). The mean half-time for elimination β-phase was 2-time longer for encapsulated enzyme than for free enzyme (150 vs 72 h). Transient changes in infrared fluorescence in organs showed that BChE is eliminated from liver. However, free and encapsulated enzymes were cleared via different pathways. This first study of pharmacokinetics and fate of BChE encapsulated in polymersomes initiates research of new formulations of bioscavengers aimed at increasing the residence time of enzymes in the blood stream.
Collapse
Affiliation(s)
- Tatiana Pashirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation.
| | - Zukhra Shaihutdinova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Dmitry Tatarinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Angelina Titova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Albina Malanyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Olga Vasileva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Kamil Gabdurakhmanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Sergei Dudnikov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | | | - Oksana Lockridge
- University of Nebraska Medical Center, Eppley Institute, Omaha, NE, USA
| | - Patrick Masson
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation.
| |
Collapse
|
13
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024; 21:5430-5454. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [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: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
14
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
15
|
Trivedi S, Belgamwar V. Fabrication and optimization of chitosan-g-m-PEG-NH 2 copolymer for advanced glioblastoma therapy using surface engineered lentinan loaded nanovesicles for nasal delivery. Int J Biol Macromol 2024; 273:133125. [PMID: 38897498 DOI: 10.1016/j.ijbiomac.2024.133125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
Glioblastoma multiforme (GBM) exhibits a high mortality with an incidence rate of 3-5 per 100,000 each year, which demands existence of newer approach for its treatment. The current study focuses on synthesis of novel lipidic nanovesicles (LNs) loaded with highly potent macromolecule Lentinan (LNT) and surface modified with methoxy poly (ethylene glycol; PEG) amine (m-PEG-NH2)-grafted-chitosan (CS) for intranasal delivery. The grafting procedure was optimized using Box Behnken design (BBD) to limit the use of organic solvents. The fabricated polymer showed enhanced aqueous solubility, biodegradability and mucoadhesion, resulting in higher nasal mucosa permeation (z = 53.52 μm). The presence of PEG enabled the sustained release of LNT till 48 h and assisted in achieving higher accumulation of LNT in CSF (41.7 ± 3.1 μg/mL) and a higher brain targeting potential of 96.3 ± 2.31 % (p < 0.05). In-vitro cellular studies showed the enhanced anti-GBM effect of LNT on U87 MG cells by reducing the cell viability (~2 times reduction in IC50 value) accompanied with large number of cells undergoing late apoptosis and death (p < 0.05) because of the higher cellular uptake (63.22 ± 3.01 ng/100 cells) of novel formulation. The copolymer comprising LNs were biocompatible, stable and can be used as an effective tool in the management of GBM.
Collapse
Affiliation(s)
- Sagar Trivedi
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra 440033, India.
| | - Veena Belgamwar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra 440033, India.
| |
Collapse
|
16
|
Yu H, Chen Q, Zheng M, Wang R, Wang H, Cheng L, Hu Y, Dai M, Du C, Luo W, Tan M, Cao Y, Guo Y, Ran H. Combination of MHI148 Targeted Photodynamic Therapy and STING Activation Inhibits Tumor Metastasis and Recurrence. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29672-29685. [PMID: 38813586 DOI: 10.1021/acsami.4c02528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Metastasis and recurrence are notable contributors to mortality associated with breast cancer. Although immunotherapy has shown promise in mitigating these risks after conventional treatments, its effectiveness remains constrained by significant challenges, such as impaired antigen presentation by dendritic cells (DCs) and inadequate T cell infiltration into tumor tissues. To address these limitations, we developed a multifunctional nanoparticle platform, termed GM@P, which consisted of a hydrophobic shell encapsulating the photosensitizer MHI148 and a hydrophilic core containing the STING agonist 2'3'-cGAMP. This design elicited robust type I interferon responses to activate antitumor immunity. The GM@P nanoparticles loaded with MHI148 specifically targeted breast cancer cells. Upon exposure to 808 nm laser irradiation, the MHI148-loaded nanoparticles produced toxic reactive oxygen species (ROS) to eradicate tumor cells through photodynamic therapy (PDT). Notably, PDT stimulated immunogenic cell death (ICD) to foster the potency of antitumor immune responses. Furthermore, the superior photoacoustic imaging (PAI) capabilities of MHI148 enabled the simultaneous visualization of diagnostic and therapeutic procedures. Collectively, our findings uncovered that the combination of PDT and STING activation facilitated a more conducive immune microenvironment, characterized by enhanced DC maturation, infiltration of CD8+ T cells, and proinflammatory cytokine release. This strategy stimulated local immune responses to augment systemic antitumor effects, offering a promising approach to suppress tumor growth, inhibit metastasis, and prevent recurrence.
Collapse
Affiliation(s)
- Huilin Yu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Qiaoqi Chen
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Min Zheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Ruoyao Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Haiyang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
- Department of Abdominal Wall, Hernia and Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Long Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yaqin Hu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Mingyuan Dai
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Chier Du
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Wenpei Luo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Mixiao Tan
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yuan Guo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| |
Collapse
|
17
|
Ming L, Wu H, Fan Q, Dong Z, Huang J, Xiao Z, Xiao N, Huang H, Liu H, Li Z. Bio-inspired drug delivery systems: A new attempt from bioinspiration to biomedical applications. Int J Pharm 2024; 658:124221. [PMID: 38750980 DOI: 10.1016/j.ijpharm.2024.124221] [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: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Natural organisms have evolved sophisticated and multiscale hierarchical structures over time to enable survival. Currently, bionic design is revolutionizing drug delivery systems (DDS), drawing inspiration from the structure and properties of natural organisms that offer new possibilities to overcome the challenges of traditional drug delivery systems. Bionic drug delivery has contributed to a significant improvement in therapeutic outcomes, providing personalized regimens for patients with various diseases and enhancing both their quality of life and drug efficacy. Therefore, it is important to summarize the progress made so far and to discuss the challenges and opportunities for future development. Herein, we review the recent advances in bio-inspired materials, bio-inspired drug vehicles, and drug-loading platforms of biomimetic structures and properties, emphasizing the importance of adapting the structure and function of organisms to meet the needs of drug delivery systems. Finally, we highlight the delivery strategies of bionics in DDS to provide new perspectives and insights into the research and exploration of bionics in DDS. Hopefully, this review will provide future insights into utilizing biologically active vehicles, bio-structures, and bio-functions, leading to better clinical outcomes.
Collapse
Affiliation(s)
- Liangshan Ming
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Hailian Wu
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Qimeng Fan
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Zishu Dong
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Jia Huang
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Zijian Xiao
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Nan Xiao
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical, University, Jiangxi, Ganzhou 341000, China.
| | - Hongning Liu
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China.
| | - Zhe Li
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China.
| |
Collapse
|
18
|
Liu J, Yang F, Hu J, Zhang X. Nanoparticles for efficient drug delivery and drug resistance in glioma: New perspectives. CNS Neurosci Ther 2024; 30:e14715. [PMID: 38708806 PMCID: PMC11071172 DOI: 10.1111/cns.14715] [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: 12/30/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/07/2024] Open
Abstract
Gliomas are the most common primary tumors of the central nervous system, with glioblastoma multiforme (GBM) having the highest incidence, and their therapeutic efficacy depends primarily on the extent of surgical resection and the efficacy of postoperative chemotherapy. The role of the intracranial blood-brain barrier and the occurrence of the drug-resistant gene O6-methylguanine-DNA methyltransferase have greatly limited the efficacy of chemotherapeutic agents in patients with GBM and made it difficult to achieve the expected clinical response. In recent years, the rapid development of nanotechnology has brought new hope for the treatment of tumors. Nanoparticles (NPs) have shown great potential in tumor therapy due to their unique properties such as light, heat, electromagnetic effects, and passive targeting. Furthermore, NPs can effectively load chemotherapeutic drugs, significantly reduce the side effects of chemotherapeutic drugs, and improve chemotherapeutic efficacy, showing great potential in the chemotherapy of glioma. In this article, we reviewed the mechanisms of glioma drug resistance, the physicochemical properties of NPs, and recent advances in NPs in glioma chemotherapy resistance. We aimed to provide new perspectives on the clinical treatment of glioma.
Collapse
Affiliation(s)
- Jiyuan Liu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Fan Yang
- Department of Cardiologythe Fourth Affiliated Hospital of China Medical UniversityShenyangChina
| | - Jinqu Hu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Xiuchun Zhang
- Department of Neurologythe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
19
|
Zhang SS, Li RQ, Chen Z, Wang XY, Dumont AS, Fan X. Immune cells: potential carriers or agents for drug delivery to the central nervous system. Mil Med Res 2024; 11:19. [PMID: 38549161 PMCID: PMC10979586 DOI: 10.1186/s40779-024-00521-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Drug delivery systems (DDS) have recently emerged as a promising approach for the unique advantages of drug protection and targeted delivery. However, the access of nanoparticles/drugs to the central nervous system (CNS) remains a challenge mainly due to the obstruction from brain barriers. Immune cells infiltrating the CNS in the pathological state have inspired the development of strategies for CNS foundation drug delivery. Herein, we outline the three major brain barriers in the CNS and the mechanisms by which immune cells migrate across the blood-brain barrier. We subsequently review biomimetic strategies utilizing immune cell-based nanoparticles for the delivery of nanoparticles/drugs to the CNS, as well as recent progress in rationally engineering immune cell-based DDS for CNS diseases. Finally, we discuss the challenges and opportunities of immune cell-based DDS in CNS diseases to promote their clinical development.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Ruo-Qi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Zhong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiao-Ying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
20
|
Liu Y, He J, Li M, Ren K, Zhao Z. Inflammation-Driven Nanohitchhiker Enhances Postoperative Immunotherapy by Alleviating Prostaglandin E2-Mediated Immunosuppression. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6879-6893. [PMID: 38300288 DOI: 10.1021/acsami.3c17357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Inflammation contributes to the immunosuppressive microenvironment and leads to the recurrence of surgically resected tumors. The COX-2/PGE2 axis is considered a key player in shaping the immunosuppression microenvironment. However, targeted modulation of the postoperative tumor microenvironment is challenging. To specifically curb the inflammation and alleviate immunosuppression, here, we developed a PGE2 inhibitor celecoxib (CXB)-loaded bionic nanoparticle (CP@CM) coated with activated murine vascular endothelial cell (C166 cells) membrane to target postoperative melanoma and inhibit its recurrence. CP@CM adhered to inflammatory white blood cells (WBCs) through the adhesion molecules, including ICAM-1, VCAM-1, E-selectin, and P-selection, expressed on the surface of C166 cells. Leveraging the natural tropism of the WBC to the inflammatory postoperative tumor site, CP@CM efficiently targeted postoperative tumors. In melanoma postoperative recurrence models, CXB significantly reduced PGE2 secretion and the recruitment of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) by inhibiting the activity of COX-2. This was followed by an increase in the infiltration of CD8+ T cells and CD4+ T cells in tumor tissues. Additionally, the immune responses were further enhanced by combining a PD-L1 monoclonal antibody. Ultimately, this immunotherapeutic strategy reversed the tumor immunosuppressive microenvironment and inhibited tumor recurrence, demonstrating a promising potential for postoperative immunotherapy for melanoma.
Collapse
Affiliation(s)
- Yingke Liu
- 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, Sichuan China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Kebai Ren
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- 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, Sichuan China
| |
Collapse
|
21
|
Wan T, Zhang FS, Qin MY, Jiang HR, Zhang M, Qu Y, Wang YL, Zhang PX. Growth factors: Bioactive macromolecular drugs for peripheral nerve injury treatment - Molecular mechanisms and delivery platforms. Biomed Pharmacother 2024; 170:116024. [PMID: 38113623 DOI: 10.1016/j.biopha.2023.116024] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Bioactive macromolecular drugs known as Growth Factors (GFs), approved by the Food and Drug Administration (FDA), have found successful application in clinical practice. They hold significant promise for addressing peripheral nerve injuries (PNIs). Peripheral nerve guidance conduits (NGCs) loaded with GFs, in the context of tissue engineering, can ensure sustained and efficient release of these bioactive compounds. This, in turn, maintains a stable, long-term, and effective GF concentration essential for treating damaged peripheral nerves. Peripheral nerve regeneration is a complex process that entails the secretion of various GFs. Following PNI, GFs play a pivotal role in promoting nerve cell growth and survival, axon and myelin sheath regeneration, cell differentiation, and angiogenesis. They also regulate the regenerative microenvironment, stimulate plasticity changes post-nerve injury, and, consequently, expedite nerve structure and function repair. Both exogenous and endogenous GFs, including NGF, BDNF, NT-3, GDNF, IGF-1, bFGF, and VEGF, have been successfully loaded onto NGCs using techniques like physical adsorption, blend doping, chemical covalent binding, and engineered transfection. These approaches have effectively promoted the repair of peripheral nerves. Numerous studies have demonstrated similar tissue functional therapeutic outcomes compared to autologous nerve transplantation. This evidence underscores the substantial clinical application potential of GFs in the domain of peripheral nerve repair. In this article, we provide an overview of GFs in the context of peripheral nerve regeneration and drug delivery systems utilizing NGCs. Looking ahead, commercial materials for peripheral nerve repair hold the potential to facilitate the effective regeneration of damaged peripheral nerves and maintain the functionality of distant target organs through the sustained release of GFs.
Collapse
Affiliation(s)
- Teng Wan
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Feng-Shi Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Ming-Yu Qin
- Suzhou Medical College, Soochow University, Suzhou 215026, China
| | - Hao-Ran Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Meng Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Yang Qu
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Yi-Lin Wang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China.
| | - Pei-Xun Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China; Peking University People's Hospital Qingdao Hospital, Qingdao 266000, China.
| |
Collapse
|