1
|
Wu H, Diao J, Li X, Yue D, He G, Jiang X, Li P. Hydrogel-based 3D printing technology: From interfacial engineering to precision medicine. Adv Colloid Interface Sci 2025; 341:103481. [PMID: 40132296 DOI: 10.1016/j.cis.2025.103481] [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: 09/27/2024] [Revised: 01/03/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Advances in 3D printing technology and the development of hydrogel-based inks have significantly enhanced the potential of precision medicine, promoting progress in medical diagnosis and treatment. The development of 3D printing enables the fabrication of complex gradient structures that emulate natural tissue environments, while advancements in interface engineering facilitate the precise control of interface properties, thereby enhancing the performance of hydrogels in biomedical applications. This review focuses on the latest advancements in three critical 3D printing application areas: efficient real-time detection, drug delivery systems, and regenerative medicine. The application of 3D printing technology enhances nucleic acid-based molecular diagnostic platforms and wearable biosensors for real-time monitoring of physiological parameters, thereby providing robust support for early disease diagnosis. Additionally, it facilitates the development of targeted and controlled drug delivery systems, which offer promising methods for efficient drug utilization, and enables the construction of complex tissue and organ structures with bioactivity and functionality, providing new solutions for regenerative medicine. Collectively, these advancements propel the ongoing progress and development of precision medicine. Furthermore, the challenges associated with 3D printing technology in these three major applications are discussed along with an outlook on prospects.
Collapse
Affiliation(s)
- Haojie Wu
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China; State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Jibo Diao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xinrong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Dongmei Yue
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China
| | - Gaohong He
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xiaobin Jiang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China.
| | - Peipei Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China.
| |
Collapse
|
2
|
Liu Z, Ye P, Shi L, Zhao Z, Zhou J, Zhou Y, Li F, Qu JH, Wang Q, Jiang Z. Dual-filler mixed matrix membrane with covalent-organic framework and nano TiO 2/polyether sulfone for efficient antibody purification. J Chromatogr A 2025; 1751:465940. [PMID: 40203634 DOI: 10.1016/j.chroma.2025.465940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
With the rapid expansion of antibody drug market, most biopharmaceutical industries urgently need to optimize their downstream purification processes to reduce production costs and improve market competitiveness. In this study, a dual-filler polyether sulfone (PES) mixed matrix membrane (MMM) that combines covalent-organic framework (COF) with nano TiO2 was developed to overcome the drawbacks of conventional protein A based purification methods. Firstly, COF@TiO2 dual-filler was prepared by Schiff base reaction. The proposed dual-filler MMM was fabricated via nonsolvent-induced phase separation (NIPS), followed by functionalization with a Fab-specific affinity peptide (m-EDPW) of trastuzumab through atom-transfer radical-polymerization method. The resulting m-EDPW@COF@TiO2/PES affinity membrane effectively integrates the merits of COF and TiO2 and show synergistic effects, demonstrating satisfactory hydrophilicity, anti-fouling ability (BSA rejection rate: 97.7 %), enrichment recovery (90.8 %), binding capacity for trastuzumab (386.6 mg/g), and long-term stability (∼ 21 days). Particularly, this affinity membrane showed good selectivity and specificity, enabling the successful purification of trastuzumab from spiked HCC1937 cancer cell culture medium with satisfactory purity (~ 97.4 %) and preservation of the antibody secondary structure. This study not only developed a novel affinity membrane with satisfactory antibody separation performance but also opened a new route for developing dual-filler or multi-filler MMM for highly efficient downstream protein purification.
Collapse
Affiliation(s)
- Zhenhu Liu
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Peijun Ye
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Leying Shi
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Zheng Zhao
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Jingwei Zhou
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Ying Zhou
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Feng Li
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China
| | - Jia-Huan Qu
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China.
| | - Qiqin Wang
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China.
| | - Zhengjin Jiang
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
3
|
Li Y, Chen Y, Tang Y, Yang T, Zhou P, Miao L, Chen H, Deng Y. Breaking the barriers in effective and safe Toll-like receptor stimulation via nano-immunomodulators for potent cancer immunotherapy. J Control Release 2025; 382:113667. [PMID: 40157608 DOI: 10.1016/j.jconrel.2025.113667] [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: 12/02/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy is an emerging strategy that awakens the intrinsic immune system for cancer treatment. Generally, successful immunotherapy of malignant tumours relies on the effective production of tumour-associated antigens and their lymph node delivery, antigen processing and presentation for T-cell activation, and the dismantling of the immunosuppressive tumour microenvironment. Toll-like receptor (TLR) agonists are potent stimulants in cancer immunotherapy, which can directly activate antigen-presenting cells (APCs) and further induce T cell activation for antitumour immune response and convert immunosuppressive tumour microenvironment to an immunogenic one for cooperative tumour ablation. However, TLR agonists for effective cancer immunotherapy have encountered essential challenges, such as insufficient immune activation and systemic side effects. In recent years, nano-immunomodulators with TLR agonists have been employed for tumour- and/or lymph node-targeted immune activation to improve the antitumour immune response and alleviate their systemic toxicities, providing a promising strategy for enhanced cancer immunotherapy. Herein, we introduce the recent progress in developing various TLR nano-immunomodulators for cancer immunotherapy via APC activation and tumour microenvironment remodelling. Upon elucidating the rational design principles of nano-immunomodulators, we elucidate the advancement of TLR nanoagonists to break the barriers in effective and safe Toll-like receptor stimulation for potent cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoqi Li
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou 215006, China.
| | - Huabing Chen
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
4
|
Zhang H, Xie D, Chen M, Yin Q, Shi G, He J, Yu X, Lu Q, Wang Z, Dong J, Hao L. pH/GSH Dual-Responsive Janus-Type Au@H-MP@DOX MR Molecular Imaging Nanomotor for Combined Photothermal/Chemotherapeutic Treatment of Pancreatic Cancer. Mol Pharm 2025. [PMID: 40394882 DOI: 10.1021/acs.molpharmaceut.5c00542] [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: 05/22/2025]
Abstract
Chemotherapy is a widely used cancer treatment modality, while the complex tumor microenvironment (TME) significantly impedes drug delivery and deep tissue penetration. An MR molecular imaging drug-loaded nanomotor has been developed to achieve deep tumor tissue penetration and imaging-guided drug delivery, enabling combined photothermal and chemotherapeutic treatment of pancreatic cancer. A Janus-type nanomotor (Au@H-MP NMs) was fabricated via magnetron sputtering for application in photothermal therapy. Doxorubicin (DOX) was loaded onto one hemisphere of the nanomotor, achieving combined photothermal and chemotherapeutic treatment. Additionally, the nanomotor exhibits dual responsiveness to pH and glutathione (GSH), facilitating the controlled release of DOX within deep tumor tissues. Studies confirmed the nanomotors excellent biosafety, strong photothermal conversion capability, and effective induction of apoptosis. Tumor tissue penetration was validated through in vitro migration and infiltration assays, while in vivo experiments demonstrated significant tumor suppression and enhanced drug accumulation. Moreover, MR imaging technology enables real-time monitoring of nanomotor dynamics. These findings suggest that the synthesized Janus-type MR molecular imaging nanomotor offers a promising strategy for multimodal treatment of pancreatic cancer with significant clinical potential.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Dan Xie
- Department of Ultrasound, The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150000, China
| | - Meng Chen
- Department of Equipment Management Section, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar 161041, China
| | - Qiangqiang Yin
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Guangyue Shi
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Jialong He
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Xiaoyang Yu
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Qian Lu
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Zhengji Wang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Jing Dong
- Department of Cell Biology, Basic Medical College, Qiqihar Medical University, Qiqihar 161006, China
| | - Liguo Hao
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| |
Collapse
|
5
|
Hu X, Wang Z, Zhu Y, Li Z, Yan H, Zhao X, Wang Q. Advancements in molecular imaging for the diagnosis and treatment of pancreatic ductal adenocarcinoma. NANOSCALE ADVANCES 2025; 7:2887-2903. [PMID: 40270837 PMCID: PMC12012634 DOI: 10.1039/d4na01080a] [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: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor characterized by poor overall patient survival and prognosis, largely due to challenges in early diagnosis, limited surgical options, and a high propensity for therapy resistance. The integration of various imaging modalities through molecular imaging techniques, particularly multimodal molecular imaging, offers the potential to provide more precise and comprehensive information about the lesion. With advances in nanomedicine, new imaging and drug delivery approaches that allow the development of multifunctional theranostic agents offer opportunities for improving pancreatic cancer treatment using precision oncology. Herein, we review the diagnostic and therapeutic applications of molecular imaging for PDAC and discuss the adoption of multimodal imaging approaches that combine the strengths of different imaging techniques to enhance diagnostic accuracy and therapeutic efficacy. We emphasize the significant role of nanomedicine technology in advancing multimodal molecular imaging and theranostics, and their potential impact on PDAC management. This comprehensive review aims to serve as a valuable reference for researchers and clinicians, offering insights into the current state of molecular imaging in PDAC and outlining future directions for improving early diagnosis, combination therapies, and prognostic evaluations.
Collapse
Affiliation(s)
- Xun Hu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zihua Wang
- School of Basic Medical Sciences, Fujian Medical University Fuzhou 350122 Fujian Province China
| | - Yuting Zhu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital Shenzhen Guangdong 518036 China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School/Tsinghua University Shenzhen 518055 China
| | - Xinming Zhao
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| |
Collapse
|
6
|
Song K, Ming J, Tao B, Zhao F, Huang S, Wu W, Jiang C, Li X. Emerging glucose oxidase-delivering nanomedicines for enhanced tumor therapy. J Control Release 2025; 381:113580. [PMID: 40024341 DOI: 10.1016/j.jconrel.2025.02.076] [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: 08/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Abnormalities in glucose metabolism have been shown to characterize malignant tumors. Glucose depletion by glucose oxidase (GOD) has shown great potential in tumor therapy by causing tumor starvation. Since 2017, nanomedicines have been designed and utilized to deliver GOD for more precise and effective glucose modulation, which can overcome intrinsic limitations of different cancer therapeutic modalities by remodeling the tumor microenvironment to enhance antitumor therapy. To date, the topic of GOD-delivering nanomedicines for enhancing tumor therapy has not been comprehensively summarized. Herein, this review aims to provide an overview and discuss in detail recent advances in GOD delivery and directly involved starvation therapy strategies, GOD-sensitized various tumor therapy strategies, and GOD-mediated multimodal antitumor strategies. Finally, the challenges and outlooks for the future progress of the emerging tumor therapeutic nanomedicines are discussed. This review provides intuitive and specific insights to a broad audience in the fields of nanomedicines, biomaterials, and cancer therapy.
Collapse
Affiliation(s)
- Kaiyue Song
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhao
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China.
| | - Xianglong Li
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
7
|
Wan G, Gu L, Chen Y, Wang Y, Sun Y, Li Z, Ma W, Bao X, Wang R. Nanobiotechnologies for stroke treatment. Nanomedicine (Lond) 2025:1-21. [PMID: 40327588 DOI: 10.1080/17435889.2025.2501514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Stroke has brought about a poor quality of life for patients and a substantial societal burden with high morbidity and mortality. Thus, the efficient stroke treatment has always been the hot topic in the research of medicine. In the past decades, nanobiotechnologies, including natural exosomes and artificial nanomaterials, have been a focus of attention for stroke treatment due to their inherent advantages, such as facile blood - brain barrier traversal and high drug encapsulation efficiency. Recently, thanks to the rapid development of nanobiotechnologies, more and more efforts have been made to study the therapeutic effects of exosomes and artificial nanomaterials as well as relevant mechanisms in stroke treatment. Herein, from recent studies and articles, the application of natural exosomes and artificial nanomaterials in stroke treatment are summarized. And their prospects of clinical translation and future development are also discussed in further detail.
Collapse
Affiliation(s)
- Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lingui Gu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangyang Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiqing Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Sun
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenwei Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Cheng Q, Huang Y, Duan W, Liu L. A pillar[5]arene-based hyaluronic acid-decorated amorphous bimetallic metal-organic framework for multimodal synergistic cancer therapy. Int J Biol Macromol 2025; 309:142994. [PMID: 40210065 DOI: 10.1016/j.ijbiomac.2025.142994] [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: 02/15/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Current antitumor monotherapies have many limitations, and developing novel synergistic anticancer strategies with low side effects and high antitumor efficiency remains a significant challenge. Herein, we developed a pH and GSH dual-responsive pillar[5]arene-based amorphous bimetallic metal-organic framework (DOX@Fe/CuP5H) for synergistic antitumor therapy involving ferroptosis, cuproptosis and apoptosis. The hydrazide-functionalized pillar[5]arene derivatives were coordinated with Cu2+ to form irregular nanoparticles, which were subsequently etched and surface-coordinated using Fe3+. Finally, doxorubicin (DOX) was loaded onto the structures, followed by surface decoration with hyaluronic acid (HA) to yield the multifunctional DOX@Fe/CuP5H. The porous structure and amorphous nature of Fe/CuP5, and the specific binding of HA to CD44 overexpressed in cancer cells endowed the DOX@Fe/CuP5H with a high drug-loading capacity and effective targeting ability, while simultaneously reducing its toxicity to normal cells. DOX@Fe/CuP5H can dissociate in the tumor microenvironment, rapidly releasing DOX to induce apoptosis. Excess Fe3+ and Cu2+ deplete intracellular GSH, leading to a redox imbalance. The accumulation of Fe2+ further promotes the production of reactive oxygen species (ROS) and lipid peroxide (LPO), triggering ferroptosis. Additionally, FDX1 regulates cellular protein lipoylation, while Cu+ binds to lipoylated proteins, causing acute proteotoxic stress and inducing cellular cuproptosis. Therefore, the rationally designed pillar[5]arene-based amorphous bimetallic metal-organic framework provides a safe and high-performance platform for enhancing the efficacy of multimodal synergistic anticancer therapies.
Collapse
Affiliation(s)
- Qi Cheng
- Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guang xi University, Nanning 530004, China
| | - Yan Huang
- Guang xi Key Laboratory of Traditional Chinese Medicine Quality Standards, Guang xi Institute of Chinese Traditional Medical & Pharmaceutical Science, Nanning, China.
| | - Wengui Duan
- Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guang xi University, Nanning 530004, China
| | - Luzhi Liu
- Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guang xi University, Nanning 530004, China.
| |
Collapse
|
9
|
Zhang R, Mao D, Fu Y, Ju R, Wei G. A self-assembled and H 2O 2-activatable hybrid nanoprodrug for lung infection and wound healing therapy. Theranostics 2025; 15:5953-5968. [PMID: 40365280 PMCID: PMC12068296 DOI: 10.7150/thno.114344] [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: 03/24/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Background: The pursuit of effective antibacterial strategies aimed at mitigating pathogenic bacterial infections while minimising drug resistance remains of paramount importance. A combinational therapeutic strategy that integrates distinct treatment components can enhance overall efficacy and mitigate undesired effects, thereby exhibiting considerable promise in combating bacterial infections. Methods: In this study, a meticulously engineered self-assembling hybrid nanoprodrug (CPBP NPs) has been devised, functioning as a hybrid prodrug of Ciprofloxacin (Cip) and hydroxybenzyl alcohol (HBA). Results: CPBP molecules can generate nanoassemblies via self-assembly and subsequently undergo decomposition to synchronously release Cip and HBA upon hydrogen peroxide (H2O2) exposure. The CPBP NPs exert antibacterial and anti-inflammatory properties through the controlled release of Cip and HBA, while also facilitating the scavenging of reactive oxygen species. These CPBP NPs exhibit broad-spectrum antibacterial activity against both Gram-negative bacteria (E. coli, 98.4%) and Gram-positive bacteria (S. aureus, 98.5%). Notably, CPBP NPs not only accumulate in the lungs to facilitate organ-specific infection treatment but also expedite the healing process of infected wounds. Conclusions: Consequently, this H2O2-activatable hybrid nanoprodrug, possessing excellent biocompatibility, holds substantial promise for advancing clinical applications in managing bacterial infections.
Collapse
Affiliation(s)
| | | | | | | | - Guoqing Wei
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
10
|
Ye R, Guo J, Yang Z, Wang Z, Chen Y, Huang J, Dong Y. Somatostatin and Mannooligosaccharide Modified Selenium Nanoparticles with Dual-Targeting for Ulcerative Colitis Treatment. ACS NANO 2025; 19:14914-14930. [PMID: 40214514 DOI: 10.1021/acsnano.5c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Inflammatory bowel disease (IBD) is a prevalent condition worldwide, characterized by complex etiologies, limited efficacy of clinical drug treatments, and potential adverse effects. In this study, we designed 269 nm selenium nanoparticles with double-cell targeting for ulcerative colitis treatment. Somatostatin (SST) and mannooligosaccharide (MOS) were employed to functionalize an Eucommia ulmoides polysaccharide selenium nanoparticle (EUP-SeNP), resulting in the formulation of SST/MOS@EUP-SeNP. Nanoparticles were engineered to target intestinal epithelial cells and macrophages through specific cell surface receptors, enabling dual-targeted treatment. In addition, sodium alginate (SA) microspheres incorporating SST/MOS@EUP-SeNP were prepared for oral administration, protecting the nanoparticles from gastric fluid. The results showed that SA/SST/MOS@EUP-SeNP could preferentially target the inflamed colon tissue and adhere to the colon, enhance the intestinal barrier function, regulate the level of colon inflammation, enhance antioxidant capacity, and regulate the composition of intestinal microbes to effectively relieve the colitis induced by sodium glucan sulfate (DSS). Meanwhile, SA/SST/MOS@EUP-SeNP had excellent biocompatibility both in vivo and in vitro. To some extent, this study can provide a reference for the treatment of IBD.
Collapse
Affiliation(s)
- Ruihua Ye
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jianying Guo
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zhongjin Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaqiang Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
11
|
Zheng X, Cui Y, Rong J, Chen S, Qu X, Hu X. GSH/pH-sensitive Förster resonance energy transfer nanoparticles for synergistic chemotherapy and chemodynamic therapy. J Mater Chem B 2025; 13:4705-4712. [PMID: 40145409 DOI: 10.1039/d5tb00243e] [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: 03/28/2025]
Abstract
Stimulus-responsive polymers have attracted significant attention as intelligent and advanced drug delivery systems. In this work, a glutathione-responsive polymer was synthesized by reversible addition-fragmentation chain transfer polymerization of natural biological molecules lipoic acid and tetraphenylene (TPE)-containing vinyl monomers. The poly(disulfide) block ensures rapid degradation of carriers and drug release under specific conditions. In addition, the introduction of pendant carboxyl groups enables Fe3+ incorporating capacity and the hydrophobic TPE block significantly boosts drug loading and aggregation induced emission (AIE) for visualization of assembly. Fe3+ and doxorubicin (DOX) loaded nanoparticles (DOX@Fe NPs) were obtained via coordination and hydrophobic interactions for synergistic chemodynamic therapy and chemotherapy. Especially, the Förster resonance energy transfer (FRET) between TPE and DOX further enables visualization of DOX release via a fluorescence signal. The in vitro release experiment results demonstrated that under the conditions of pH 5.5 and 5 mM GSH, the release efficiency of DOX reached 79.2% in 12 hours. In the cellular experiment, the viability of 4T1 cells co-incubated with DOX@Fe NPs for 48 hours was only 2.5%, verifying that DOX@Fe NPs possess potent tumor-killing capability.
Collapse
Affiliation(s)
- Xiaodong Zheng
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Yingjian Cui
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Jianxin Rong
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Shengli Chen
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Xiongwei Qu
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Xiuli Hu
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China.
| |
Collapse
|
12
|
Vicente-da-Silva J, Pereira JOSL, do Carmo FA, Patricio BFDC. Skin and Wound Healing: Conventional Dosage versus Nanobased Emulsions Forms. ACS OMEGA 2025; 10:12837-12855. [PMID: 40224422 PMCID: PMC11983225 DOI: 10.1021/acsomega.5c00455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
The skin plays a crucial role in the body's homeostasis through its thermoregulation functions, metabolic activity, and, mainly, its barrier function. Once this system has its homeostasis disturbed, through the promotion of tissue discontinuity, an injury happens and a restoration process starts. Different products can be used to promote, accelerate, or stimulate the healing process, such as hydrogels, emulsions, and ointments (main conventional formulations). Despite the historical use and wide market and consumer acceptance, new systems emerged for wound management with the main challenge to overcome conventional form limitations, in which nanosystems are found, mainly nanobased emulsion forms (nano- and microemulsions, NE and ME). Here, we discuss the skin function and wound healing process, highlighting the cellular and molecular processes, the different wound classifications, and factors that affect physiological healing. We also investigated the recent patents (2012-2023) filed at the United States Patent and Trademark Office, where we found few patents for conventional forms (hydrogels = 5; emulsions = 4; ointments = 6) but a larger number of patents for nanobased emulsions filed in this time (NE = 638; ME = 4,072). Furthermore, we address the use of nanobased emulsions (NE and ME) and their particularities, differences, and application in wound treatment. This work also discusses the challenges, bottlenecks, and regulatory framework for nanosystems, industrial, academic, and government interest in nanotechnology, and future perspectives about this key factor for the nanosystems market and consumer acceptance.
Collapse
Affiliation(s)
- João
Vitor Vicente-da-Silva
- PostGraduate
Program in Molecular and Cellular Biology, Department of Physiological
Sciences − Pharmacology, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
- Pharmaceutical
and Technological Innovation Laboratory, Department of Physiological
Sciences − Pharmacology, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
| | - Juliana Oliveira
da Silva Lopes Pereira
- Pharmaceutical
and Technological Innovation Laboratory, Department of Physiological
Sciences − Pharmacology, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
| | - Flávia Almada do Carmo
- Laboratory
of Pharmaceutical Industrial Technology, Department of Drugs and Pharmaceutics,
Faculty of Pharmacy, Federal University
of Rio de Janeiro, Rio de Janeiro 21941-971, Brazil
- PostGraduate
Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro 21941-971, Brazil
| | - Beatriz Ferreira de Carvalho Patricio
- PostGraduate
Program in Molecular and Cellular Biology, Department of Physiological
Sciences − Pharmacology, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
- Pharmaceutical
and Technological Innovation Laboratory, Department of Physiological
Sciences − Pharmacology, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
| |
Collapse
|
13
|
Li YJ, Hua X, Zhao YQ, Mo H, Liu S, Chen X, Sun Z, Wang W, Zhao Q, Cui Z, An T, Song J. An Injectable Multifunctional Nanosweeper Eliminates Cardiac Mitochondrial DNA to Reduce Inflammation. Adv Healthc Mater 2025; 14:e2404068. [PMID: 39811901 DOI: 10.1002/adhm.202404068] [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: 11/15/2024] [Revised: 01/05/2025] [Indexed: 01/16/2025]
Abstract
Myocarditis, a leading cause of sudden cardiac death and heart transplantation, poses significant treatment challenges. The study of clinical samples from myocarditis patients reveals a correlation between the pathogenesis of myocarditis and cardiomyocyte mitochondrial DNA (mtDNA). During inflammation, the concentration of mtDNA in cardiomyocytes increases. Hence, it is hypothesized that the combined clearance of mtDNA and its downstream STING pathway can treat myocarditis. However, clearing mtDNA is problematic. An innovative mtDNA scavenger is introduced, Nanosweeper (NS), which utilizes its nanostructure to facilitate the transport of NS-mtDNA co-assemblies for degradation, achieving mtDNA clearance. The fluorescent mtDNA probe on NS, bound to functional peptides, enhances the stability of NS. NS also exhibits robust stability in human plasma with a half-life of up to 10 hours. In a murine myocarditis model, NS serves as a drug delivery vehicle, targeting the delivery of the STING pathway inhibitor C-176 to the myocardium. This approach synergistically modulates the cGAS-STING axis with NS, effectively attenuating myocarditis- associated inflammatory cascade. This evaluation of NS in porcine models corroborated its superior biosafety profile and cardiac targeting capability. This strategic approach of targeted mtDNA clearance couple with STING pathway inhibition, significantly augments therapeutic efficacy against myocarditis, outperforming the conventional drug C-176, indicating its clinical potential.
Collapse
Affiliation(s)
- Yi-Jing Li
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650102, China
| | - Yi-Qi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Han Mo
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Shun Liu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiao Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650102, China
| | - Zhe Sun
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Weiteng Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Qian Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zeyu Cui
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Tao An
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650102, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| |
Collapse
|
14
|
Yang S, Fang Y, Ma Y, Wang F, Wang Y, Jia J, Yang Y, Sun W, Zhou Q, Li Z. Angiogenesis and targeted therapy in the tumour microenvironment: From basic to clinical practice. Clin Transl Med 2025; 15:e70313. [PMID: 40268524 PMCID: PMC12017902 DOI: 10.1002/ctm2.70313] [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: 11/16/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Angiogenesis, as a core marker of cancer survival and growth, is integral to the processes of tumour growth, invasion and metastasis. In recent years, targeted angiogenesis treatment strategies have gradually become an important direction in cancer treatment. Single-cell sequencing technology can provide new insights into targeted angiogenesis by providing a deeper understanding of the heterogeneity of tumour endothelial cells and exploring the interactions between endothelial cells and surrounding cells in the tumour microenvironment. Here, we systematically review the research progress in endothelial cell pathophysiology and its endothelial‒mesenchymal transition and illustrate the heterogeneity of endothelial cells from a single-cell perspective. Finally, we examine the contributions of different cell types within the tumour microenvironment in relation to tumour angiogenesis, as well as the latest progress and strategies in targeted angiogenesis therapy, hoping to provide useful insights into the clinical application of antiangiogenic treatment. Furthermore, a summary of the present progress in the development of potential angiogenesis inhibitors and the ongoing clinical trials for combination therapies is provided. KEY POINTS: Angiogenesis plays a key role in tumour progression, invasion and metastasis, so strategies targeting angiogenesis are gradually becoming an important direction in cancer therapy. Interactions between endothelial cells and stromal cells and immune cells in the tumour microenvironment are significant in angiogenesis. The application of antiangiogenic immunotherapy and nanotechnology in antiangiogenic therapy provides a vital strategy for prolonging the survival of cancer patients.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingshuai Fang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yangcheng Ma
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fuqi Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhang Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiachi Jia
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yabing Yang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Weipeng Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhen Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
15
|
Lin J, Yang L, Liu T, Zhao H, Liu Y, Shu F, Huang H, Liu W, Zhang W, Jiang L, Xiao S, Zheng Y, Xia Z. Mannose-modified exosomes loaded with MiR-23b-3p target alveolar macrophages to alleviate acute lung injury in Sepsis. J Control Release 2025; 379:832-847. [PMID: 39870316 DOI: 10.1016/j.jconrel.2025.01.073] [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: 09/13/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
The anti-inflammatory role of miR-23b-3p (miR-23b) is known in autoimmune diseases like multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis. However, its role in sepsis-related acute lung injury (ALI) and its effect on macrophages in ALI remain unexplored. This investigation aimed to evaluate miR-23b's therapeutic potential in macrophages in the context of ALI. The study found reduced miR-23b expression in macrophages within ALI tissue. Intratracheal delivery of miR-23b mimics alleviated ALI by partially inhibiting M1 macrophage activation through the Lpar1-NF-κB pathway. Effective delivery systems are crucial for prolonging miR-23b activity in the lungs, reducing dosage, and minimizing side effects by specifically targeting macrophages. However, current vector systems for nucleic acid delivery, including viral, lipid-based, polymer-based, and peptide-based vectors, face limitations due to eliciting immune responses. Exosomes have garnered significant attention as a leading gene delivery system due to the safety, effectivity and low immunogenicity. We further isolated exosomes from bone marrow-derived mesenchymal stem cells, modified exosomes with mannosylated ligands to enhance the targeted delivery of miR-23b to macrophage. This approach represents a promising novel therapeutic strategy for treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Jiezhi Lin
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Department of Burn Surgery, the 963rd Hospital of Joint Logistics Support Force of PLA, Jiamusi, Heilongjiang 154007, China
| | - Lu Yang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Tianyi Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Hui Zhao
- Department of Burn Surgery, the 963rd Hospital of Joint Logistics Support Force of PLA, Jiamusi, Heilongjiang 154007, China
| | - Yingying Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Futing Shu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Hongchao Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Wenzhang Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shichu Xiao
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
16
|
Li X, Liu Y, Wu X, Huang R, Chen S, Yuan K. Ultrasound meets nanomedicine: towards disease treatment and medical imaging. Mikrochim Acta 2025; 192:215. [PMID: 40053162 DOI: 10.1007/s00604-025-07042-y] [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: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 03/18/2025]
Abstract
As a kind of mechanical wave, ultrasound has been widely employed in the biomedical field due to its superiors of deep tissue penetration, non-destructiveness and non-toxicity. In this review, we highlight current progress and prospects in using ultrasound as a powerful tool for disease treatment and medical imaging, including (1) ultrasound as energy input for driving nano/micromotor in drug delivery, this part first introduces the synthesis and motion behavior of nano/micromotors, then reviews the small molecular and macromolecular compounds that the nano/micromotors are delivering; (2) sonosensitive nanomaterials for disease therapy, the sonodynamic, sonopiezoelectric, sonothermal, and sonomechanical therapy will all be covered; (3) ultrasound as a non-invasive technique for nano/micromotor tracking or medical imaging; (4) the sonoporation of nano/microbubble. Future challenges in using ultrasound for disease treatment or medical imaging will also be described in the conclusion part.
Collapse
Affiliation(s)
- Xiaochun Li
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yanting Liu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Xuewan Wu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Rui Huang
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Shaoqi Chen
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Kaisong Yuan
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
17
|
Sotirova Y, Ivanova N, Ermenlieva N, Vilhelmova-Ilieva N, Simeonova L, Metodiev M, Gugleva V, Andonova V. Antimicrobial and Antiherpetic Properties of Nanoencapsulated Hypericum perforatum Extract. Pharmaceuticals (Basel) 2025; 18:366. [PMID: 40143142 PMCID: PMC11944447 DOI: 10.3390/ph18030366] [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: 02/06/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: This study aims to gain insights into the antimicrobial and antiherpetic activity of hyperforin-rich Hypericum perforatum L. (HP) extract using nanostructured lipid carriers (NLCs) as delivery platforms. Methods: Two established NLC specimens, comprising glyceryl behenate and almond oil or borage oil, and their extract-loaded counterparts (HP-NLCs) were utilized. Their minimal bactericidal/fungicidal concentrations (MBC; MFC) were investigated against Escherichia coli ATCC 25922, Staphylococcus aureus ATCC 25923, Pseudomonas aeruginosa ATCC 10145, Klebsiella pneumoniae ATCC 10031, and Candida albicans ATCC 10231. The anti-herpesvirus (HSV-1) potential was evaluated concerning antiviral and virucidal activity and impact on viral adsorption. Results: The borage oil-based extract-loaded nanodispersion (HP-NLC2) exhibited pronounced microbicidal activity against S. aureus (MBC 6.3 mg/mL), K. pneumoniae (MBC 97.7 µg/mL), and C. albicans (MFC < 48.8 µg/mL), unlike the almond oil-containing sample (HP-NLC1), which showed only weak inhibition of the fungal growth. HP-NLC2 was found to be less cytotoxic and to suppress HSV-1 replication slightly more than HP-NLC1, but generally, the effects were weak. Neither the empty lipid nanoparticles nor the HP extract-loaded carriers expressed activity against E. coli, P. aeruginosa, the HSV-1 extracellular virions, or viral adhesion. Conclusions: It could be concluded that both HP-NLC samples revealed only minor antiherpetic potential of the hyperforin-rich extract, but HP-NLC2 demonstrated significant antibacterial and antimycotic activity. Therefore, the latter was featured as a more convenient HP-carrier system for nano-designed dermal pharmaceutical formulations. Such a thorough investigation of hyperforin-determined anti-HSV-1 effects and antibacterial and antimycotic properties, being the first of its kind, contributes to the fundamental knowledge of HP and reveals new perspectives for the utilization, limitations, and therapeutic designation of its non-polar components.
Collapse
Affiliation(s)
- Yoana Sotirova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria; (Y.S.); (V.G.); (V.A.)
| | - Nadezhda Ivanova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria; (Y.S.); (V.G.); (V.A.)
| | - Neli Ermenlieva
- Department of Microbiology and Virology, Faculty of Medicine, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Neli Vilhelmova-Ilieva
- Department of Virology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 G. Bonchev Str., 1113 Sofia, Bulgaria; (N.V.-I.); (L.S.); (M.M.)
| | - Lora Simeonova
- Department of Virology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 G. Bonchev Str., 1113 Sofia, Bulgaria; (N.V.-I.); (L.S.); (M.M.)
| | - Miroslav Metodiev
- Department of Virology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 G. Bonchev Str., 1113 Sofia, Bulgaria; (N.V.-I.); (L.S.); (M.M.)
| | - Viliana Gugleva
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria; (Y.S.); (V.G.); (V.A.)
| | - Velichka Andonova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria; (Y.S.); (V.G.); (V.A.)
| |
Collapse
|
18
|
Chen H, Hu P, Wang Y, Liu H, Zheng J, Huang Z, Zhang X, Liu Y, Zhou T. From quorum sensing inhibition to antimicrobial defense: The dual role of eugenol-gold nanoparticles against carbapenem-resistant Pseudomonas aeruginosa. Colloids Surf B Biointerfaces 2025; 247:114415. [PMID: 39622152 DOI: 10.1016/j.colsurfb.2024.114415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/22/2025]
Abstract
To address the pressing challenge of antibiotic resistance, particularly the robust defense mechanisms of Pseudomonas aeruginosa (P. aeruginosa) against conventional antibiotics, this study employs nanotechnology to enhance antimicrobial efficacy while ensuring good biocompatibility with the host. In this study, gold nanoparticles were chemically decorated with eugenol, a phenol-rich natural compound, using a one-pot synthesis method. The successful synthesis and functionalization of eugenol-decorated gold nanoparticles (Eugenol_Au NPs) were validated by comprehensive physicochemical analyses, demonstrating their stability and biocompatibility. These nanoparticles exhibited potent antimicrobial activity against both planktonic and biofilm-embedded carbapenem-resistant P. aeruginosa strains. Eugenol_Au NPs disrupted the bacterial quorum sensing system and stimulated intracellular reactive oxygen species production, which enhance their antibacterial effects. This dual mechanism of action has promising clinical implications for the treatment of infections associated with antibiotic-resistant P. aeruginosa. In vivo assessments in a murine peritoneal infection model showed that Eugenol_Au NPs significantly reduced bacterial loads and mitigated inflammatory responses, thereby improving survival rates. The study highlights the potential of Eugenol_Au NPs as an alternative strategy for refractory infections caused by carbapenem-resistant P. aeruginosa, and underscores the feasibility and promise of further clinical research and development of new therapeutic approaches targeting this resistant pathogen.
Collapse
Affiliation(s)
- Huale Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China; Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Panjie Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yaran Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Haifeng Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Junyuan Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zeyu Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xiaotuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
19
|
Zheng Y, Duan XY, Wang X, Wang XF, Liu B. Insight into the effect of ZIF-8 on the interaction between drugs and protein/cell. Int J Biol Macromol 2025; 294:139530. [PMID: 39761886 DOI: 10.1016/j.ijbiomac.2025.139530] [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: 09/03/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025]
Abstract
Understanding the impact of nanomaterials on drug-protein/cell interactions is crucial for comprehending their in vivo biological effects. We investigated the impact of zeolitic imidazolate framework (ZIF)-8 on the interaction between curcumin (Cur) and human serum albumin (HSA) using various spectroscopic techniques and molecular docking. Additionally, we examined its effect on drug-cell interaction using HepG2 cells and Escherichia coli (E. coli). The UV-vis spectra and fluorescence results demonstrated the occurrence of an interaction between Cur-HSA and ZIF-8, potentially resulting in the formation of ground-state complexes. ZIF-8 did not alter the static quenching mechanism, interaction force type, and binding stoichiometry between Cur and HSA, but it induced subtle changes in the secondary structure and esterase activity of HSA. Cur predominantly binds in the site I of HSA. Molecular docking analysis confirmed the results. The incorporation of ZIF-8 enhanced the antitumor activity of Cur-HSA and the antibacterial efficacy of ciprofloxacin (CIP)-HSA, while concurrently enhancing the uptake of CIP by E. coli. These results indicate that the influence of ZIF-8 on drug-protein interactions may consequently exert a significant impact on drug efficacy.
Collapse
Affiliation(s)
- Ying Zheng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xin-Yue Duan
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Liaoning University, Shenyang 110036, China
| | - Xiao-Fang Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China.
| | - Bin Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China.
| |
Collapse
|
20
|
Dai XJ, Li WJ, Xie DD, Liu BX, Gong L, Han HH. Stimuli-Responsive Nano Drug Delivery Systems for the Treatment of Neurological Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410030. [PMID: 39840482 DOI: 10.1002/smll.202410030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/07/2025] [Indexed: 01/23/2025]
Abstract
Nanomaterials with unparalleled physical and chemical attributes have become a cornerstone in the field of nanomedicine delivery. These materials can be engineered into various functionalized nanocarriers, which have become the focus of research. Stimulus-responsive nanodrug delivery systems (SRDDS) stand out as a sophisticated class of nanocarriers that can release drugs in response to environmental cues. Due to the complex pathogenesis and the multifaceted pathological environment of the nervous system, developing accurate and effective drug therapy with low side-effects is a formidable task. In recent years, SRDDS have been widely used in the treatment of neurological diseases. By customizing SRDDS to align with the specific microenvironment of the nervous system tissues or external stimulation, the efficacy of drug delivery can be enhanced. This review provides an in-depth look at the characteristics of the microenvironment of neurological diseases and highlights case studies of SRDDS tailored to treat these disorders based on the unique stimulation criteria of nervous system tissues or external triggers. Additionally, this review provides a comprehensive overview of the progress and future prospects of SRDDS technology in the treatment of neurological diseases, providing valuable guidance for its transition from fundamental research to clinical application.
Collapse
Affiliation(s)
- Xi-Jian Dai
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, 330006, P. R. China
| | - Wen-Jia Li
- Molecular Imaging Center, National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, P. R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Dong-Dong Xie
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, P. R. China
| | - Bi-Xia Liu
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, 330006, P. R. China
| | - Lianggeng Gong
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, 330006, P. R. China
| | - Hai-Hao Han
- Molecular Imaging Center, National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, P. R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P. R. China
| |
Collapse
|
21
|
Kadhim SH, Dehghan G, Mahdavi M. Ciprofloxacin-encapsulated solid lipid nanoparticles: a comprehensive biochemical analysis of cytotoxic effects, proliferation inhibition, and apoptotic induction in KG1-a leukemia cells. Med Oncol 2025; 42:75. [PMID: 39945900 DOI: 10.1007/s12032-025-02619-2] [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: 11/21/2024] [Accepted: 01/29/2025] [Indexed: 05/09/2025]
Abstract
As a fundamental approach to the treatment of acute myeloid leukemia (AML), chemotherapeutic agents face significant clinical challenges, including poor solubility and low bioavailability. In this context, solid lipid nanoparticles (SLNs) have emerged as a promising drug delivery system in oncologic therapies, owing to their advantageous characteristics, such as enhanced physical stability and controlled drug-release profiles. This study focuses on the synthesis of ciprofloxacin (CP)-loaded SLNs, aiming to enhance the anticancer efficacy of CP, an antibiotic recognized for its potential anticancer properties, while simultaneously reducing its associated side effects. Characterization of blank SLN and CP-SLN was conducted using dynamic light scattering (DLS), atomic force microscopy (AFM), UV-Vis spectrophotometry, and Fourier transform infrared spectroscopy (FTIR). In vitro release was carried out using dialysis bag method in isotonic phosphate buffer (pH = 7.4). The anticancer and pro-apoptotic effects of the CP-SLN formulation were assessed through cell viability assays, Hoechst staining, and Annexin V/PI flow cytometry. Additionally, expression levels of Bax, Bcl2, and p53 were analyzed via Real-Time PCR. The synthesized CP-SLN formulation exhibited optimal characteristics, including a particle size of 340-360 nm, a polydispersity index (PDI) of 0.4, and an entrapment efficiency of 90%. The in vitro drug release showed an initial burst release in the time points 4-10 h. Both CP and the CP-SLN formulations demonstrated significant anti-proliferative and pro-apoptotic effects on KG1-a cells, as indicated by the upregulation of the Bax/Bcl2 ratio and p53, resulting in G0/G1 cell cycle arrest and apoptosis induction. The results suggest that encapsulating CP in SLN enhances its anticancer and pro-apoptotic effects in KG1-a stem-like leukemia cells. Thus, CP-SLN may serve as a promising formulation for leukemia treatment and could improve the efficacy of other therapeutic agents.
Collapse
Affiliation(s)
- Salma Hussein Kadhim
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Majid Mahdavi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
22
|
Shi C, Chen T, Li Y, Li W, Shen Y, Cai K, Wang M, Chen Y. Encapsulation of individual mammalian cells as a cell-based drug delivery carrier for lung cancer treatment. J Control Release 2025; 378:209-220. [PMID: 39662681 DOI: 10.1016/j.jconrel.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Cell-based delivery holds great promise for advancing cell therapy, but it often faces challenges such as low cell survival rates and immunological rejection during cell injection and circulation. In this study, we develop an alternative approach aimed at engineering cell membranes to produce encapsulated individual mammalian (EIM) cells. The encapsulation shell was formed by catalyzing the reaction of H2O2 with hyaluronic acid-dopamine (HA-DA) on the cell surface using horseradish peroxidase (HRP) enzymes. This protective shell not only protects live mammalian cells from physical stress but also from biological assaults. The individual cell encapsulation system enables the loading of cells with chemotherapy drugs, enhancing their targeting ability towards tumor sites and resulting in over 5.1-fold cell enrichment at metastatic tumors, thereby improving tumor-killing efficacy and reducing metastasis. Overall, the individual cell encapsulation system demonstrates potential for targeted drug delivery to the lungs in the treatment of lung cancer by reducing side effects and enhancing treatment outcomes.
Collapse
Affiliation(s)
- Chaochen Shi
- Medical 3D Printing Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yingying Li
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Wenshuai Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yuan Shen
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Kehan Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Ming Wang
- Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yazhou Chen
- Medical 3D Printing Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China.
| |
Collapse
|
23
|
Li X, Xu J, Yao S, Zhang N, Zhang B, Zhang Z. Targeting Drug Delivery System to Skeletal Muscles: A Comprehensive Review of Different Approaches. J Cachexia Sarcopenia Muscle 2025; 16:e13691. [PMID: 39910928 PMCID: PMC11799587 DOI: 10.1002/jcsm.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/18/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
The skeletal muscle is one of the largest organs in the body and is responsible for the mechanical activity required for posture, movement and breathing. The effects of current pharmaceutical therapies for skeletal muscle diseases are far from satisfactory; approximately 24% of Duchenne muscular dystrophy (DMD) trials have been terminated because of unsatisfactory outcomes. The lack of a skeletal muscle-targeting strategy is a major reason for these unsuccessful trials, contributing to low efficiency and severe side effects. The development of targeting strategies for skeletal muscle-specific drug delivery has shown the potential for increasing drug concentrations in the skeletal muscle, minimising off-target effects, and thereby improving the therapeutic effects of drugs. Over the past few decades, novel methods for specifically delivering cargo to skeletal muscles have been developed. In this review, we categorise targeting methods into four types: peptides, antibodies, small molecules and aptamers. Most research has focused on peptide and antibody ligands, and there are several well-established drugs in this category; however, drawbacks such as protease degradation and immunogenicity limit their use. Aptamers and small molecules have low immunogenicity and are simple to chemically produce. However, small molecule ligands generally exhibit lower affinity because of their small size and high mobility. Aptamers are promising ligands for skeletal muscle-targeting delivery systems. Additionally, if the active site of the cargo is located inside the cell, an internalisation pathway becomes necessary. The order of internalisation ligands and targeting ligands in the complex is a crucial factor, because an inappropriate order could lead to much lower targeting and internalisation efficiencies. Moreover, ligand density also merits consideration, as increasing the density of the targeting ligands may result in steric hindrance, which could impact the accessibility of the receptor and cause enlargement of the targeted ligands. More efforts are required to optimise drug delivery systems that specifically recognise skeletal muscle, with the aim of enhancing quality of life and promoting patient well-being.
Collapse
Affiliation(s)
- Xiaofang Li
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Jintao Xu
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Shanshan Yao
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Ning Zhang
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Bao‐Ting Zhang
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Zong‐Kang Zhang
- Faculty of MedicineSchool of Chinese MedicineThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
24
|
Quan Z, Wang S, Xie H, Zhang J, Duan R, Li M, Zhang J. ROS Regulation in CNS Disorder Therapy: Unveiling the Dual Roles of Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410031. [PMID: 39676433 DOI: 10.1002/smll.202410031] [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: 10/26/2024] [Revised: 12/01/2024] [Indexed: 12/17/2024]
Abstract
The treatment of brain diseases has always been the focus of attention. Due to the presence of the blood-brain barrier (BBB), most small molecule drugs are difficult to reach the brain, leading to undesirable therapeutic outcomes. Recently, nanomedicines that can cross the BBB and precisely target lesion sites have emerged as thrilling tools to enhance the early diagnosis and treat various intractable brain disorders. Extensive research has shown that reactive oxygen species (ROS) play a crucial role in the occurrence and progression of brain diseases, including brain tumors and neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease, stroke, or traumatic brain injury, making ROS a potential therapeutic target. In this review, on the structure and function of BBB as well as the mechanisms are first elaborated through which nanomedicine traverses it. Then, recent studies on ROS production are summarized through photodynamic therapy (PDT), chemodynamic therapy (CDT), and sonodynamic therapy (SDT) for treating brain tumors, and ROS depletion for treating NDDs. This provides valuable guidance for the future design of ROS-targeted nanomedicines for brain disease treatment. The ongoing challenges and future perspectives in developing nanomedicine-based ROS management for brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Zhengyang Quan
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Sa Wang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Huanhuan Xie
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jiayi Zhang
- International department, Beijing 101 Middle School, Beijing, 100091, P. R. China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Menglin Li
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jinfeng Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
25
|
Zhu J, Tian L, Yan D, Wang D, Tang BZ. Mechanistic Insights Into NIR-II AIEgens Boosted Multimodal Phototheranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410441. [PMID: 39937448 DOI: 10.1002/smll.202410441] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Exploiting single molecular species synchronously affording powerful second near-infrared (NIR-II) fluorescence, superior photoacoustic output, prominent reactive oxygen species generation, and satisfactory photothermal conversion is supremely appealing for phototheranostics, yet remains formidably challenging. In this work, electron donor/π-bridge engineering is implemented on the basis of 6,7-di(thiophen-2-yl)-[1,2,5]thiadiazolo[3,4-g]quinoxaline moiety. The optimal molecule, namely TPATO-TTQ, is demonstrates to exhibit those notable features requested by exceptional phototheranostics, which are systematically elucidated through the depictions of excited-state energy dissipation pathways and the influence of intramolecular motion on the photophysical properties, with assistances of quantum chemical calculation and molecular dynamic simulation. By utilizing TPATO-TTQ nanoparticles, unprecedented performance on NIR-II fluorescence-photoacoustic-photothermal trimodal imaging-navigated type I photodynamic-photothermal synergistic therapy to orthotopic breast cancer is authenticates by the precise tumor diagnosis and complete tumor ablation.
Collapse
Affiliation(s)
- Jun Zhu
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Leyuan Tian
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Dingyuan Yan
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Dong Wang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, China
| |
Collapse
|
26
|
Lu Z, Li H, Song N, Wang Z, Wang H, Rehman A, Han L, Zeng KW. Therapeutic Potential of Carbon Dots Derived from Phytochemicals as Nanozymes Exhibiting Superoxide Dismutase Activity for Anemia. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4562-4578. [PMID: 39792367 DOI: 10.1021/acsami.4c17885] [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: 01/12/2025]
Abstract
Anemia is a potentially life-threatening blood disorder caused by an insufficient erythroblast volume in the circulatory system. Self-renewal failure of erythroblast progenitors is one of the key pathological factors leading to erythroblast deficiency. However, there are currently no effective drugs that selectively target this process. In this work, we present a carbon dot (CP-CDs) derived from phytochemicals that significantly promotes the self-renewal of erythroblast progenitors for anemia therapy. As a superoxide dismutase (SOD)-like nanozyme, CP-CDs significantly activate the hypoxia response and JAK/STAT3 pathways in erythroid cells by reprogramming the oxidative stress state. This results in unique erythropoiesis-enhancing properties by promoting the generation of erythroid progenitor cells. Moreover, CP-CDs protect mature red blood cells by inhibiting oxidative stress-induced damage and improving the immune-inflammatory microenvironment. In vivo, CP-CDs showed a promising therapeutic effect in mouse and zebrafish models of anemia with minimal adverse effects, indicating significant translational medical value. Collectively, this study not only illustrates a successful approach for nanomedicine-enhanced anemia therapy but also enhances our understanding of the interaction between nanomedicine and the self-renewal of erythroblast progenitors.
Collapse
Affiliation(s)
- Zhiyuan Lu
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Haojia Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Nannan Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Zhiwei Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Hua Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Asma Rehman
- National Institute for Biotechnology & Genetic Engineering College Pakistan Institute of Engineering & Applied Sciences (NIBGE-C, PIEAS), Faisalabad 38000, Pakistan
| | - Liwen Han
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
27
|
Wang L, Wu X, Wang X, Dong M, Zhang H, Zhao P. Targeting CHEK1: Ginsenosides-Rh2 and Cu2O@G-Rh2 nanoparticles in thyroid cancer. Cell Biol Toxicol 2025; 41:30. [PMID: 39808342 PMCID: PMC11732901 DOI: 10.1007/s10565-024-09961-7] [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: 06/21/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Thyroid cancer (THCA) is an increasingly common malignant tumor of the endocrine system, with its incidence rising steadily in recent years. For patients who experience recurrence or metastasis, treatment options are relatively limited, and the prognosis is poor. Therefore, exploring new therapeutic strategies has become particularly urgent. This study confirmed that effective suppression of THCA cell proliferation and stimulation of apoptosis can be achieved through the application of Ginsenosides-Rh2. Through network pharmacology screening, the molecular target of Ginsenosides-Rh2 in THCA was identified as CHEK1, and its inhibitory effect was confirmed by downregulating CHEK1 protein expression. Furthermore, demonstrations conducted both in vitro and in vivo showcased that delivering Ginsenosides-Rh2 using nanoparticle carriers significantly reduced cell viability by approximately 50%, regulated DNA damage levels, apoptosis-related protein expression, and cell cycle control. The IC50 of the nanoparticle formulation was determined (B-CPAP IC50 = 88.24 μM), TPC IC50 = 79.52 μM). This study confirmed that Cu2O@G-Rh2 is effective in suppressing tumors and exhibits a significant inhibitory effect on tumor recurrence and metastasis while maintaining good safety. Cu2O@G-Rh2 nanoparticles possess excellent stability and anti-tumor efficacy. This research offers new perspectives for the treatment of THCA and demonstrates potential clinical applications.
Collapse
Affiliation(s)
- Lidong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Xin Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - XinLu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Meng Dong
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Hao Zhang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
28
|
Xiong H, Du C, Ye J, Zhang H, Qin Y, Zeng F, Song R, Shi C, Guo H, Chen J, Shen H, Cui Y, Zhou Z. Therapeutic co-assemblies for synergistic NSCLC treatment through dual topoisomerase I and tubulin inhibitors. J Control Release 2025; 377:485-494. [PMID: 39592024 DOI: 10.1016/j.jconrel.2024.11.054] [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: 07/14/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024]
Abstract
Camptothecin (CPT) and podophyllotoxin (PPT) function as topoisomerase (TOP) I and tubulin inhibitors, respectively, with potent anticancer effects in a variety of cancers. Despite its promise, the clinical applicability of the combination of CPT and PPT faces challenges, including potential side effect and limited therapeutic efficacy. In this study, we designed co-assembly nanomedicines with the different weight (w/w) ratios of amphiphilic Evans blue conjugated CPT prodrug (EB-ss-CPT) and PPT molecules, denoted as ECT Nano. The co-assembly of EB-ss-CPT and PPT without other excipients has nearly 100% drug loading efficiency and high drug loading content of PPT of up to 74.29 ± 0.90 wt%. Notably, the ECT Nano (1:2) equipped with the ability to inhibit TOP I activity and tubulin polymerization, which provided a highly efficient strategy to improve synergistic efficacy and decrease side toxicity in non-small cell lung cancer mouse model. This work represents a step forward to the development of practical applications for dual TOP I and tubulin inhibitors and especially hopeful to the rational design of combination nanomedicine for therapeutic purposes.
Collapse
Affiliation(s)
- Hehe Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Chao Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Jinmin Ye
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Heng Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Yatong Qin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Fantian Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Ruirui Song
- Department of Radiology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Changrong Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Huifeng Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Jiang Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Huaxiang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China.
| | - Zijian Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
29
|
Nikolić I, Đoković J, Mehn D, Guerrini G, Savić S, Jordan O, Borchard G. When conventional approach in toxicity assays falls short for nanomedicines: a case study with nanoemulsions. Drug Deliv Transl Res 2025:10.1007/s13346-024-01776-7. [PMID: 39779651 DOI: 10.1007/s13346-024-01776-7] [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] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
The aim of this study was to assess the critical quality attributes of parenteral nanoemulsion formulations by measuring several physicochemical parameters and linking them to their in vitro performance, illustrating how simplistic and routinely used approaches are insufficient for understanding a potential nanomedicine. Physicochemical characterization should encompass size and size distribution through at least two orthogonal techniques, such as dynamic light scattering (DLS) and electron microscopy, with added value from analytical ultracentrifugation. In vitro toxicity assessment was performed using three different assays to determine mitochondrial activity (WST-1), membrane integrity (lactate dehydrogenase release (LDH) assay), and cell viability (propidium iodide (PI) staining). Special focus was placed on estimating appropriate incubation times for relevant results in biological investigations. All formulations had an average diameter of around 100 nm. Conclusions regarding in vitro safety were assay-dependent: LDH and PI-based assays showed good correlation, while the WST-1 assay indicated that the non-PEGylated formulation altered mitochondrial activity more significantly compared to the PEGylated ones. The study underlined that the selection of appropriate cytotoxicity assays should be based on the possible mechanism of cellular perturbation. Alternatively, different aspects of cellular toxicity should be tested. Additionally, there is a need for well-designed controls to overcome nanoparticle scattering effects and avoid potentially false high toxicity results, which was demonstrated. Combining orthogonal, well-designed physicochemical and biological assays in a standardized manner as an initial step in the reliable preclinical characterization of nanomedicines is suggested. This represents a key aspect of new methodologies in nanomedicine characterization.
Collapse
Affiliation(s)
- Ines Nikolić
- Faculty of Science, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology, University of Belgrade, Belgrade, Serbia
| | - Jelena Đoković
- Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology, University of Belgrade, Belgrade, Serbia
| | - Dora Mehn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Snežana Savić
- Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology, University of Belgrade, Belgrade, Serbia
| | - Olivier Jordan
- Faculty of Science, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Gerrit Borchard
- Faculty of Science, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
30
|
Liew KB, Koh EV, Kong XE, Ismail NA, Abu Bakar RA, Kee PE, Khalid SH, Phang HC. Recent Advancements in Nanopharmaceuticals for Novel Drug Delivery Systems. Pharm Nanotechnol 2025; 13:271-286. [PMID: 39323341 DOI: 10.2174/0122117385324246240826042254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 09/27/2024]
Abstract
Nanoparticles have found applications across diverse sectors, including agriculture, food, cosmetics, chemicals, mechanical engineering, automotive, and oil and gas industries. In the medical field, nanoparticles have garnered considerable attention due to their great surface area, high solubility, rapid dissolution, and enhanced bioavailability. Nanopharmaceuticals are specifically designed to precisely deliver drug substances to targeted tissues and cells, aiming to optimize therapeutic efficacy while minimizing potential adverse effects. Furthermore, nanopharmaceuticals offer advantages, such as expedited therapeutic onset, reduced dosages, minimized variability between fed and fasted states, and enhanced patient compliance. The increasing interest in nanopharmaceuticals research among scientists and industry stakeholders highlights their potential for various medical applications from disease management to cancer treatment. This review examines the distinctive characteristics of ideal nanoparticles for efficient drug delivery, explores the current types of nanoparticles utilized in medicine, and delves into the applications of nanopharmaceuticals, including drug and gene delivery, as well as transdermal drug administration. This review provides insights into the nanopharmaceuticals field, contributing to the development of novel drug delivery systems and enhancing the potential of nanotechnology in healthcare.
Collapse
Affiliation(s)
- Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Ee Va Koh
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Xue Er Kong
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Nurdina Aleyah Ismail
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | | | - Phei Er Kee
- Biorefinery and Bioprocessing Engineering Laboratory, Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan, 320, Taiwan
| | - Syed Haroon Khalid
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi Mara, Puncak Alam, Selangor, 42300, Malaysia
| | - Hiu Ching Phang
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| |
Collapse
|
31
|
Luo Y, Jia X, Wu X, Diao L, Zhao Y, Liu X, Peng Y, Zhong W, Xing M, Lyu G. Bacteria-activated macrophage membrane coated ROS-responsive nanoparticle for targeted delivery of antibiotics to infected wounds. J Nanobiotechnology 2024; 22:781. [PMID: 39702152 DOI: 10.1186/s12951-024-03056-5] [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: 09/15/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
Bacterial infections and antibiotic resistance represent significant global public health challenges, necessitating the development of innovative antibacterial agents with targeted delivery capabilities. Our study utilized macrophages' natural ability to recognize bacteria and the increased reactive oxygen species (ROS) at infection sites to develop a novel nanoparticle for targeted delivery and controlled release. We prepared bacteria-activated macrophage membranes triggered by Staphylococcus aureus (Sa-MMs), which showed significantly higher expression of Toll-like receptors (TLRs), compared to normal macrophage membranes (MMs). These Sa-MMs were then used to coat vancomycin-loaded amphiphilic nanoparticles with ROS responsiveness (Van-NPs), resulting in the novel targeted delivery system Sa-MM@Van-NPs. Studies both In vitro and in vivo demonstrated that biocompatible Sa-MM@Van-NPs efficiently targeted infected sites and released vancomycin to eliminate bacteria, facilitating faster wound healing. By combining targeted delivery to infected sites and ROS-responsive antibiotic release, this approach might represent a robust strategy for precise infection eradication and enhanced wound healing.
Collapse
Affiliation(s)
- Ying Luo
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaoli Jia
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaozhuo Wu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Ling Diao
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Yawei Zhao
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Xing Liu
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yi Peng
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Wen Zhong
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Guozhong Lyu
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214000, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.
- National Research Center for Emergency Medicine, Beijing, 100000, China.
| |
Collapse
|
32
|
Ren H, Jin Y, Huang H, Wu W, Dai X, Fang W, Qin J, Li H, Zhao P. In vivo engineering chimeric antigen receptor immune cells with emerging nanotechnologies. NANO TODAY 2024; 59:102517. [DOI: 10.1016/j.nantod.2024.102517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Guan P, Yuan C, Li J, Yu K, Xie R, Hu E, Ding W, Lan G, Lu F. Delivering urokinase-type plasminogen activator using mulberry leaf exosomes enables thrombolysis and remodeling of venous microenvironments. Int J Biol Macromol 2024; 282:136866. [PMID: 39454913 DOI: 10.1016/j.ijbiomac.2024.136866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
In the treatment of thrombosis, conventional nanocarriers inevitably have problems, such as adverse reactions and difficulties in synthesis. Inspired by the concept of 'medicine food homology,' we extracted and purified natural exosomes from mulberry leaves as carriers for the delivery of urokinase-type plasminogen activator (uPA) for targeted therapy. The obtained mulberry leaf exosomes (MLE) possessed a desirable hydrodynamic particle size (119.4 nm), a uniform size distribution (polydispersity index = 0.174), and a negative surface charge (-23.3 mv). Before loading with uPA, MLE were grafted with cyclic RGD (cRGD) to selectively bind activated platelets for thrombus targeting. The cytometry studies revealed that MLE@cRGD has a high thrombus targeting ability about 74.3 %. Animal tests demonstrated that the delivered uPA could dissolve clots almost completely in femoral vein thrombosis models. In addition, MLE could remodel venous microenvironments by effectively eliminating reactive oxygen species (ROS) and promoting the phenotypic transformation of macrophages from M1 to M2 for venous tissue repair.
Collapse
Affiliation(s)
- Pengpeng Guan
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Caijie Yuan
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Jiali Li
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Kun Yu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Ruiqi Xie
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Enling Hu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Weiwei Ding
- Division of Trauma and Surgical Intensive Care Unit, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Guangqian Lan
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Fei Lu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China.
| |
Collapse
|
34
|
Xie B, Liu Y, Li X, Yang P, He W. Solubilization techniques used for poorly water-soluble drugs. Acta Pharm Sin B 2024; 14:4683-4716. [PMID: 39664427 PMCID: PMC11628819 DOI: 10.1016/j.apsb.2024.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/28/2024] [Accepted: 08/14/2024] [Indexed: 12/13/2024] Open
Abstract
About 40% of approved drugs and nearly 90% of drug candidates are poorly water-soluble drugs. Low solubility reduces the drugability. Effectively improving the solubility and bioavailability of poorly water-soluble drugs is a critical issue that needs to be urgently addressed in drug development and application. This review briefly introduces the conventional solubilization techniques such as solubilizers, hydrotropes, cosolvents, prodrugs, salt modification, micronization, cyclodextrin inclusion, solid dispersions, and details the crystallization strategies, ionic liquids, and polymer-based, lipid-based, and inorganic-based carriers in improving solubility and bioavailability. Some of the most commonly used approved carrier materials for solubilization techniques are presented. Several approved poorly water-soluble drugs using solubilization techniques are summarized. Furthermore, this review summarizes the solubilization mechanism of each solubilization technique, reviews the latest research advances and challenges, and evaluates the potential for clinical translation. This review could guide the selection of a solubilization approach, dosage form, and administration route for poorly water-soluble drugs. Moreover, we discuss several promising solubilization techniques attracting increasing attention worldwide.
Collapse
Affiliation(s)
- Bing Xie
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
35
|
Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol 2024; 15:1451989. [PMID: 39483479 PMCID: PMC11524880 DOI: 10.3389/fimmu.2024.1451989] [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: 06/20/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
In recent decades, nanotechnology has significantly advanced drug delivery systems, particularly in targeting subcellular organelles, thus opening new avenues for disease treatment. Mitochondria, critical for cellular energy and health, when dysfunctional, contribute to cancer, neurodegenerative diseases, and metabolic disorders. This has propelled the development of nanomedicines aimed at precise mitochondrial targeting to modulate their function, marking a research hotspot. This review delves into the recent advancements in mitochondrial-targeted nanotherapeutics, with a comprehensive focus on targeting strategies, nanocarrier designs, and their therapeutic applications. It emphasizes nanotechnology's role in enhancing drug delivery by overcoming biological barriers and optimizing drug design for specific mitochondrial targeting. Strategies exploiting mitochondrial membrane potential differences and specific targeting ligands improve the delivery and mitochondrial accumulation of nanomedicines. The use of diverse nanocarriers, including liposomes, polymer nanoparticles, and inorganic nanoparticles, tailored for effective mitochondrial targeting, shows promise in anti-tumor and neurodegenerative treatments. The review addresses the challenges and future directions in mitochondrial targeting nanotherapy, highlighting the need for precision, reduced toxicity, and clinical validation. Mitochondrial targeting nanotherapy stands at the forefront of therapeutic strategies, offering innovative treatment perspectives. Ongoing innovation and research are crucial for developing more precise and effective treatment modalities.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiao-meng Li
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Li-si Wei
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Cong X, Zhang Z, Li H, Yang YG, Zhang Y, Sun T. Nanocarriers for targeted drug delivery in the vascular system: focus on endothelium. J Nanobiotechnology 2024; 22:620. [PMID: 39396002 PMCID: PMC11470712 DOI: 10.1186/s12951-024-02892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - Zebin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, 130012, Jilin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China.
| |
Collapse
|
37
|
Tikhonov A, Kachanov A, Yudaeva A, Danilik O, Ponomareva N, Karandashov I, Kostyusheva A, Zamyatnin AA, Parodi A, Chulanov V, Brezgin S, Kostyushev D. Biomimetic Nanoparticles for Basic Drug Delivery. Pharmaceutics 2024; 16:1306. [PMID: 39458635 PMCID: PMC11510494 DOI: 10.3390/pharmaceutics16101306] [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: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Biomimetic nanoparticles (BMNPs) are innovative nanovehicles that replicate the properties of naturally occurring extracellular vesicles, facilitating highly efficient drug delivery across biological barriers to target organs and tissues while ensuring maximal biocompatibility and minimal-to-no toxicity. BMNPs can be utilized for the delivery of therapeutic payloads and for imparting novel properties to other nanotechnologies based on organic and inorganic materials. The application of specifically modified biological membranes for coating organic and inorganic nanoparticles has the potential to enhance their therapeutic efficacy and biocompatibility, presenting a promising pathway for the advancement of drug delivery technologies. This manuscript is grounded in the fundamentals of biomimetic technologies, offering a comprehensive overview and analytical perspective on the preparation and functionalization of BMNPs, which include cell membrane-coated nanoparticles (CMCNPs), artificial cell-derived vesicles (ACDVs), and fully synthetic vesicles (fSVs). This review examines both "top-down" and "bottom-up" approaches for nanoparticle preparation, with a particular focus on techniques such as cell membrane coating, cargo loading, and microfluidic fabrication. Additionally, it addresses the technological challenges and potential solutions associated with the large-scale production and clinical application of BMNPs and related technologies.
Collapse
Affiliation(s)
- Andrey Tikhonov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Oleg Danilik
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Ivan Karandashov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
38
|
Ling J, Wu J, Cao Y, Zhang T, Cao X, Ge X, Liu Y, Wang M, Ren B, Lu J. Advances in nano-preparations for improving tetrandrine solubility and bioavailability. Arch Pharm (Weinheim) 2024; 357:e2400274. [PMID: 39031554 DOI: 10.1002/ardp.202400274] [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: 04/12/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/22/2024]
Abstract
Tetrandrine (TET) is a natural bis-benzylisoquinoline alkaloid isolated from Stephania species with a wide range of biological and pharmacologic activities; it mainly serves as an anti-inflammatory agent or antitumor adjuvant in clinical applications. However, limitations such as prominent hydrophobicity, severe off-target toxicity, and low absorption result in suboptimal therapeutic outcomes preventing its widespread adoption. Nanoparticles have proven to be efficient devices for targeted drug delivery since drug-carrying nanoparticles can be passively transported to the tumor site by the enhanced permeability and retention (EPR) effects, thus securing a niche in cancer therapies. Great progress has been made in nanocarrier construction for TET delivery due to their outstanding advantages such as increased water-solubility, improved biodistribution and blood circulation, reduced off-target irritation, and combinational therapy. Herein, we systematically reviewed the latest advancements in TET-loaded nanoparticles and their respective features with the expectation of providing perspective and guidelines for future research and potential applications of TET.
Collapse
Affiliation(s)
- Jie Ling
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingping Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuening Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingting Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiujun Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Ge
- School of Marxism, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yilan Liu
- Hematology Department, The General Hospital of the Western Theater Command PLA, Chengdu, China
| | - Maolin Wang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Bo Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
39
|
Gao Y, Cao Q, Xiao Y, Wu Y, Ding L, Huang H, Li Y, Yang J, Meng L. The progress and future of the treatment of Candida albicans infections based on nanotechnology. J Nanobiotechnology 2024; 22:568. [PMID: 39285480 PMCID: PMC11406819 DOI: 10.1186/s12951-024-02841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024] Open
Abstract
Systemic infection with Candida albicans poses a significant risk for people with weakened immune systems and carries a mortality rate of up to 60%. However, current therapeutic options have several limitations, including increasing drug tolerance, notable off-target effects, and severe adverse reactions. Over the past four decades, the progress in developing drugs to treat Candida albicans infections has been sluggish. This comprehensive review addresses the limitations of existing drugs and summarizes the efforts made toward redesigning and innovating existing or novel drugs through nanotechnology. The discussion explores the potential applications of nanomedicine in Candida albicans infections from four perspectives: nano-preparations for anti-biofilm therapy, innovative formulations of "old drugs" targeting the cell membrane and cell wall, reverse drug resistance therapy targeting subcellular organelles, and virulence deprivation therapy leveraging the unique polymorphism of Candida albicans. These therapeutic approaches are promising to address the above challenges and enhance the efficiency of drug development for Candida albicans infections. By harnessing nano-preparation technology to transform existing and preclinical drugs, novel therapeutic targets will be uncovered, providing effective solutions and broader horizons to improve patient survival rates.
Collapse
Affiliation(s)
- Yang Gao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qinyan Cao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuyang Xiao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yue Wu
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Liang Ding
- Nanjing Stomatological Hospital, Nanjing, 210008, China
| | - He Huang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yanan Li
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Jingpeng Yang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Lingtong Meng
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
40
|
Tu L, Wang J, Sun Y, Wan Y. Fabrication of Luteolin Nanoemulsion by Box-Behnken Design to Enhance its Oral Absorption Via Lymphatic Transport. AAPS PharmSciTech 2024; 25:206. [PMID: 39237659 DOI: 10.1208/s12249-024-02898-4] [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] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Intestinal lymphatic transport offers an alternative and effective way to deliver drugs, such as avoiding first-pass metabolism, enhancing oral bioavailability, and facilitating the treatment of targeted lymphoid-related diseases. However, the clinical use of luteolin (LUT) is limited by its poor water solubility and low bioavailability, and enhancing lymphatic transport by nanoemulsion may be an efficient way to enhance its oral bioavailability. The objective of this work is to prepare the luteolin nanoemulsions (LUT NEs), optimized its preparation parameters by using Box-Behnken design optimization (BBD) and evaluated it in vitro and in vivo. An Caco-2 / Raji B cell co-incubation monolayer model was established to simulate the M-cell pathway, and the differences in the transmembrane transport of LUT and NEs were compared. Cycloheximide (CHX) was utilized to establish rat chylomicron (CM) blocking model, and for investigating the influence of pharmacokinetic parameters in rats thereafter. The results showed that LUT NEs have good stability, the particle sizes were about 23.87 ± 0.57 nm. Compared with LUT suspension, The Papp of LUT NEs was enhanced for 3.5-folds, the oral bioavailability was increased by about 2.97-folds. In addition, after binding with chylomicron, the oral bioavailability of LUT NEs was decreased for about 30% (AUC 0-∞ (μg/L*h): 5.356 ± 1.144 vs 3.753 ± 0.188). These results demonstrated that NEs could enhance the oral absorption of luteolin via lymphatic transport routes.
Collapse
Affiliation(s)
- Liangxing Tu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Ju Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Yongbing Sun
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
41
|
An J, Zhang Z, Zhang J, Zhang L, Liang G. Research progress in tumor therapy of carrier-free nanodrug. Biomed Pharmacother 2024; 178:117258. [PMID: 39111083 DOI: 10.1016/j.biopha.2024.117258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/25/2024] Open
Abstract
Carrier-free nanodrugs are a novel type of drug constructed by the self-assembly of drug molecules without carrier involvement. They have the characteristics of small particle size, easy penetration of various barriers, targeting tumors, and efficient release. In recent years, carrier-free nanodrugs have become a hot topic in tumor therapy as they solve the problems of low drug loading, poor biocompatibility, and low uptake efficiency of carrier nanodrugs. A series of recent studies have shown that carrier-free nanodrugs play a vital role in the treatment of various tumors, with similar or better effects than carrier nanodrugs. Based on the literature published in the past decades, this paper first summarizes the recent progress in the assembly modes of carrier-free nanodrugs, then describes common therapeutic modalities of carrier-free nanodrugs in tumor therapy, and finally depicts the existing challenges along with future trends of carrier-free nanodrugs. We hope that this review can guide the design and application of carrier-free nanodrugs in the future.
Collapse
Affiliation(s)
- Junling An
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Zequn Zhang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Jinrui Zhang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Lingyang Zhang
- Institute of Biomedical Research, Henan Academy of Sciences, Zhengzhou, Henan, People's Republic of China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China; Institute of Biomedical Research, Henan Academy of Sciences, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
42
|
Zhang Z, Liu C, Lu Y, Zhao W, Zhu Q, He H, Chen Z, Wu W. In vivo fluorescence imaging of nanocarriers in near-infrared window II based on aggregation-caused quenching. J Nanobiotechnology 2024; 22:488. [PMID: 39143492 PMCID: PMC11323397 DOI: 10.1186/s12951-024-02761-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
Accurate fluorescence imaging of nanocarriers in vivo remains a challenge owing to interference derived mainly from biological tissues and free probes. To address both issues, the current study explored fluorophores in the near-infrared (NIR)-II window with aggregation-caused quenching (ACQ) properties to improve imaging accuracy. Candidate fluorophores with NIR-II emission, ACQ984 (λem = 984 nm) and IR-1060 (λem = 1060 nm), from the aza-BODIPY and cyanine families, respectively, were compared with the commercial fluorophore ICG with NIR-II tail emission and the NIR-I fluorophore P2 from the aza-BODIPY family. ACQ984 demonstrates high water sensitivity with complete fluorescence quenching at a water fraction greater than 50%. Physically embedding the fluorophores illuminates various nanocarriers, while free fluorophores cause negligible interference owing to the ACQ effect. Imaging based on ACQ984 revealed fine structures in the vascular system at high resolution. Moreover, good in vivo and ex vivo correlations in the monitoring of blood nanocarriers can be established, enabling real-time noninvasive in situ investigation of blood pharmacokinetics and dynamic distribution in various tissues. IR-1060 also has a good ACQ effect, but the lack of sufficient photostability and steady post-labeling fluorescence undermines its potential for nanocarrier bioimaging. P2 has an excellent ACQ effect, but its NIR-I emission only provides nondiscriminative ambiguous images. The failure of the non-ACQ probe ICG to display the biodistribution details serves as counterevidence for the improved imaging accuracy by NIR-II ACQ probes. Taken together, it is concluded that fluorescence imaging of nanocarriers based on NIR-II ACQ probes enables accurate in vivo bioimaging and real-time in situ pharmacokinetic analysis.
Collapse
Affiliation(s)
- Zichen Zhang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Chang Liu
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Yi Lu
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
- Fudan Zhangjiang Institute, Shanghai, 201203, China
| | - Weili Zhao
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Haisheng He
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China.
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| | - Wei Wu
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China.
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
- Fudan Zhangjiang Institute, Shanghai, 201203, China.
| |
Collapse
|
43
|
Li Y, Wu C, Yang R, Tang J, Li Z, Yi X, Fan Z. Application and Development of Cell Membrane Functionalized Biomimetic Nanoparticles in the Treatment of Acute Ischemic Stroke. Int J Mol Sci 2024; 25:8539. [PMID: 39126107 PMCID: PMC11313357 DOI: 10.3390/ijms25158539] [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: 05/25/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Ischemic stroke is a serious neurological disease involving multiple complex physiological processes, including vascular obstruction, brain tissue ischemia, impaired energy metabolism, cell death, impaired ion pump function, and inflammatory response. In recent years, there has been significant interest in cell membrane-functionalized biomimetic nanoparticles as a novel therapeutic approach. This review comprehensively explores the mechanisms and importance of using these nanoparticles to treat acute ischemic stroke with a special emphasis on their potential for actively targeting therapies through cell membranes. We provide an overview of the pathophysiology of ischemic stroke and present advances in the study of biomimetic nanoparticles, emphasizing their potential for drug delivery and precision-targeted therapy. This paper focuses on bio-nanoparticles encapsulated in bionic cell membranes to target ischemic stroke treatment. It highlights the mechanism of action and research progress regarding different types of cell membrane-functionalized bi-onic nanoparticles such as erythrocytes, neutrophils, platelets, exosomes, macrophages, and neural stem cells in treating ischemic stroke while emphasizing their potential to improve brain tissue's ischemic state and attenuate neurological damage and dysfunction. Through an in-depth exploration of the potential benefits provided by cell membrane-functionalized biomimetic nanoparticles to improve brain tissue's ischemic state while reducing neurological injury and dysfunction, this study also provides comprehensive research on neural stem cells' potential along with that of cell membrane-functionalized biomimetic nanoparticles to ameliorate neurological injury and dysfunction. However, it is undeniable that there are still some challenges and limitations in terms of biocompatibility, safety, and practical applications for clinical translation.
Collapse
Affiliation(s)
- Ying Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Chuang Wu
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Rui Yang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Jiannan Tang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhanqing Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Xue Yi
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhongxiong Fan
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
44
|
Li J, Zhang Y, Yang YG, Sun T. Advancing mRNA Therapeutics: The Role and Future of Nanoparticle Delivery Systems. Mol Pharm 2024; 21:3743-3763. [PMID: 38953708 DOI: 10.1021/acs.molpharmaceut.4c00276] [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: 07/04/2024]
Abstract
The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
45
|
Guérin M, Lepeltier E. Nanomedicines via the pulmonary route: a promising strategy to reach the target? Drug Deliv Transl Res 2024; 14:2276-2297. [PMID: 38587757 DOI: 10.1007/s13346-024-01590-1] [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] [Accepted: 03/22/2024] [Indexed: 04/09/2024]
Abstract
Over the past decades, research on nanomedicines as innovative tools in combating complex pathologies has increased tenfold, spanning fields from infectiology and ophthalmology to oncology. This process has further accelerated since the introduction of SARS-CoV-2 vaccines. When it comes to human health, nano-objects are designed to protect, transport, and improve the solubility of compounds to allow the delivery of active ingredients on their targets. Nanomedicines can be administered by different routes, such as intravenous, oral, intramuscular, or pulmonary routes. In the latter route, nanomedicines can be aerosolized or nebulized to reach the deep lung. This review summarizes existing nanomedicines proposed for inhalation administration, from their synthesis to their potential clinical use. It also outlines the respiratory organs, their structure, and particularities, with a specific emphasis on how these factors impact the administration of nanomedicines. Furthermore, the review addresses the organs accessible through pulmonary administration, along with various pathologies such as infections, genetic diseases, or cancer that can be addressed through inhaled nanotherapeutics. Finally, it examines the existing devices suitable for the aerosolization of nanomedicines and the range of nanomedicines in clinical development.
Collapse
Affiliation(s)
- Mélina Guérin
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, 49000, Angers, France
| | - Elise Lepeltier
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, 49000, Angers, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
46
|
He Y, Chen Z, Liu Q, Li Z, Wen D, Zhang H, Zhang M, Jiang D, Li H, Wen L, Chen G. Reversible opening of the blood-labyrinth barrier by low-pressure pulsed ultrasound and microbubbles for the treatment of inner ear diseases. J Control Release 2024; 372:318-330. [PMID: 38906419 DOI: 10.1016/j.jconrel.2024.06.043] [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: 01/10/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Systemic drug administration provides convenience and non-invasive benefits for preventing and treating inner ear diseases. However, the blood-labyrinth barrier (BLB) restricts the transport of drugs to inner ear tissues. Ultrasound can stimulate specific areas and penetrate tissues, with the potential to overcome physiological barriers. We present a novel strategy based on low-pressure pulsed ultrasound assisted by microbubbles (USMB) to transiently open the BLB and deliver therapeutics into the inner ear. A pulsed ultrasound device with adjustable pressure was established; the generated ultrasound was transmitted through the external auditory canal into the guinea pig's inner ear. We observed that the application of microbubbles allowed the use of safe and efficient ultrasound conditions to penetrate the BLB. We found that USMB-mediated BLB opening seemed to be associated with a reduced expression of the tight junction proteins zonula occludens-1 and occludin. Following intravenous administration, hydrophilic dexamethasone sodium phosphate (DSP), hydrophobic curcumin (CUR), as well as drug-loaded nanoparticles (Fe3O4@CUR NPs) could be efficiently delivered into the inner ear. We observed better drug accumulation in the perilymph of the inner ear, resulting in less drug (cisplatin)-induced ototoxicity. Furthermore, physiological, hematological, and histological studies showed that the modulation of the BLB by low-pressure USMB was a safe process without significant adverse effects. We conclude that USMB could become a promising strategy for the systematic delivery of therapeutics in the treatment of inner ear diseases.
Collapse
Affiliation(s)
- Yuanwei He
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ziyu Chen
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qinglang Liu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhiyang Li
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dingsheng Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hong Zhang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ming Zhang
- Guangdong Sunho Pharmaceutical Co. Ltd, Zhongshan 528437, China
| | - Di Jiang
- Department of Otolaryngology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523000, China
| | - Huaan Li
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lu Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Gang Chen
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
47
|
Jiang Y, Nie D, Hu Z, Zhang C, Chang L, Li Y, Li Z, Hu W, Li H, Li S, Xu C, Liu S, Yang F, Wen W, Han D, Zhang K, Qin W. Macrophage-Derived Nanosponges Adsorb Cytokines and Modulate Macrophage Polarization for Renal Cell Carcinoma Immunotherapy. Adv Healthc Mater 2024; 13:e2400303. [PMID: 38647150 DOI: 10.1002/adhm.202400303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/07/2024] [Indexed: 04/25/2024]
Abstract
Renal cell carcinoma (RCC) is a hot tumor infiltrated by large numbers of CD8+ T cells and is highly sensitive to immunotherapy. However, tumor-associated macrophages (TAMs), mainly M2 macrophages, tend to undermine the efficacy of immunotherapy and promote the progression of RCC. Here, macrophage-derived nanosponges are fabricated by M2 macrophage membrane-coated poly(lactic-co-glycolic acid)(PLGA), which could chemotaxis to the CXC and CC chemokine subfamily-enriched RCC microenvironment via corresponding membrane chemokine receptors. Subsequently, the nanosponges act like cytokine decoys to adsorb and neutralize broad-spectrum immunosuppressive cytokines such as colony stimulating factor-1(CSF-1), transforming growth factor-β(TGF-β), and Lnterleukin-10(IL-10), thereby reversing the polarization of M2-TAMs toward the pro-inflammatory M1 phenotype, and enhancing the anti-tumor effect of CD8+ T cells. To further enhance the polarization reprogramming efficiency of TAMs, DSPE-PEG-M2pep is conjugated on the surface of macrophage-derived nanosponges for specific recognition of M2-TAMs, and the toll like receptors 7/8(TLR7/8) agonist, R848, is encapsulated in these nanosponges to induce M1 polarization, which result in significant efficacy against RCC. In addition, these nanosponges exhibit undetectable biotoxicity, making them suitable for clinical applications. In summary, a promising and facile strategy is provided for immunomodulatory therapies, which are expected to be used in the treatment of tumors, autoimmune diseases, and inflammatory diseases.
Collapse
Affiliation(s)
- Yao Jiang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of Urology, Air Force 986 Hospital, Xi'an, 710054, China
| | - Disen Nie
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhihao Hu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chao Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingdi Chang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhengxuan Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Hu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Hongji Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Sikai Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
48
|
Chota A, Abrahamse H, George BP. Green synthesis and characterization of AgNPs, liposomal loaded AgNPs and ZnPcS 4 photosensitizer for enhanced photodynamic therapy effects in MCF-7 breast cancer cells. Photodiagnosis Photodyn Ther 2024; 48:104252. [PMID: 38901719 DOI: 10.1016/j.pdpdt.2024.104252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Breast cancer remains a formidable challenge in oncology despite significant advancements in treatment modalities. Conventional therapies such as surgery, chemotherapy, radiation therapy, and hormonal therapy have been the mainstay in managing breast cancer for decades. However, a subset of patient's experiences treatment failure, leading to disease recurrence and progression. Therefore, this study investigates the therapeutic potential of green-synthesized silver nanoparticles (AgNPs) using an African medicinal plant (Dicoma anomala methanol root extract) as a reducing agent for combating breast cancer. AgNPs were synthesized using the bottom-up approach and later modified with liposomes (Lip) loaded with photosensitizer (PS) zinc phthalocyanine tetrasulfonate (Lip@ZnPcS4) using thin film hydration method. The successful formation and Lip modification of AgNPs, alongside ZnPcS4, were confirmed through various analytical techniques including UV-Vis spectroscopy, Fourier-transform infrared spectroscopy (FT-IR), high-resolution transmission electron microscopy (HR-TEM), scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDS). Following a 24 h treatment period, MCF-7 cells were assessed for viability using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT viability assay), cell death analysis using mitochondrial membrane potential (MMP) (ΔΨm), Annexin V-fluorescein isothiocyanate (FITC)-propidium iodide (PI) kit, and caspase- 3, 8 and 9 activities. The experiments were repeated four times (n = 4), and the results were analyzed using SPSS statistical software version 27, with a confidence interval set at 0.95. The synthesized nanoparticles and nanocomplex, including AgNPs, AgNPs-Lip, Lip@ZnPcS4, and AgNPs-Lip@ZnPcS4, exhibited notable cytotoxicity and therapeutic efficacy against MCF-7 breast cancer cells. Notably, the induction of apoptosis, governed by the upregulation of apoptotic proteins i.e., caspase 8 and 9 activities. In addition, caspase 3 was not expressed by MCF-7 cells in both control and experimental groups. Given the challenging prognosis associated with breast cancer, the findings underscore the promise of liposomal nanoformulations in cancer photodynamic therapy (PDT), thus warranting further exploration in clinical settings.
Collapse
Affiliation(s)
- Alexander Chota
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa.
| |
Collapse
|
49
|
Feng X, Jia P, Zhang D. Nanocarrier drug delivery system: promising platform for targeted depression therapy. Front Pharmacol 2024; 15:1435133. [PMID: 39119603 PMCID: PMC11306052 DOI: 10.3389/fphar.2024.1435133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Depression is a chronic mental disorder characterized by persistent low mood and loss of interest. Treatments for depression are varied but may not be sufficient cure. Drug-based treatment regimens have drawbacks such as slow onset of action, low bioavailability, and drug side effects. Nanocarrier Drug Delivery Systems (NDDS) has received increasing attention for brain drug delivery since it assists the drug through the blood-brain barrier and improves bioavailability, which may be beneficial for treating depression. Due to the particle size and physicochemical properties of nanocarriers, it presents a promise to improve the stability and solubility of antidepressants, thereby enhancing the drug concentration. Moreover, ligand-modified nanocarriers can be taken as a target direct medicines release system and reduce drug side effects. The purpose of the present review is to provide an up-to-date understanding of the Nanocarrier drug delivery system and relevant antidepressants in different routes of ingestion, to lay a foundation for the treatment of patients with depression.
Collapse
Affiliation(s)
- Xiaoying Feng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Jia
- Department of Neurosurgery Nursing, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Genetic Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
50
|
Li H, Gong Q, Luo K. Biomarker-driven molecular imaging probes in radiotherapy. Theranostics 2024; 14:4127-4146. [PMID: 38994026 PMCID: PMC11234278 DOI: 10.7150/thno.97768] [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: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Background: Biomarker-driven molecular imaging has emerged as an integral part of cancer precision radiotherapy. The use of molecular imaging probes, including nanoprobes, have been explored in radiotherapy imaging to precisely and noninvasively monitor spatiotemporal distribution of biomarkers, potentially revealing tumor-killing mechanisms and therapy-induced adverse effects during radiation treatment. Methods: We summarized literature reports from preclinical studies and clinical trials, which cover two main parts: 1) Clinically-investigated and emerging imaging biomarkers associated with radiotherapy, and 2) instrumental roles, functions, and activatable mechanisms of molecular imaging probes in the radiotherapy workflow. In addition, reflection and future perspectives are proposed. Results: Numerous imaging biomarkers have been continuously explored in decades, while few of them have been successfully validated for their correlation with radiotherapeutic outcomes and/or radiation-induced toxicities. Meanwhile, activatable molecular imaging probes towards the emerging biomarkers have exhibited to be promising in animal or small-scale human studies for precision radiotherapy. Conclusion: Biomarker-driven molecular imaging probes are essential for precision radiotherapy. Despite very inspiring preliminary results, validation of imaging biomarkers and rational design strategies of probes await robust and extensive investigations. Especially, the correlation between imaging biomarkers and radiotherapeutic outcomes/toxicities should be established through multi-center collaboration involving a large cohort of patients.
Collapse
Affiliation(s)
- Haonan 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, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, 699 Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, China
| | - 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, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|