1
|
Yang L, Wang P, Zhang Y, Zhou J, Bi X, Qian Z, Hou S, Li L, Fan Y. Hybrid cell membrane coating orchestrates foreign-body reactions, anti-adhesion, and pro-regeneration in abdominal wall reconstruction. Biomaterials 2025; 321:123289. [PMID: 40154120 DOI: 10.1016/j.biomaterials.2025.123289] [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: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Tension-free synthetic meshes are the clinical standard for hernia repair, but they often trigger immune response-mediated complications such as severe foreign-body reactions (FBR), visceral adhesions, and fibrotic healing, increasing the risk of recurrence. Herein, we developed a hybrid cell membrane coating for macroscale mesh fibers that acts as an immune orchestrator, capable of balancing immune responses with tissue regeneration. Cell membranes derived from red blood cells (RBCs) and platelets (PLTs) were covalently bonded to fiber surfaces using functionalized-liposomes and click chemistry. The fusion of clickable liposomes with cell membranes significantly improved coating efficiency, coverage uniformity, and in vivo stability. Histological and flow cytometric analyses of subcutaneous implantation in rats and mice demonstrated significant biofunctional heterogeneity among various cell membrane coatings in FBR. Specifically, the RBC-PLT-liposome hybrid cell membrane coating markedly mitigated FBR, facilitated host cell infiltration, and promoted M2-type macrophage polarization. Importantly, experimental results of abdominal wall defect repairs in rats indicate that the hybrid cell membrane coating effectively prevented visceral adhesions, promoted muscle regenerative healing, and enhanced the recruitment of Pax7+/MyoD+ muscle satellite cells. Our findings suggest that the clickable hybrid cell membrane coating offers a promising approach to enhance clinical outcomes of hernia mesh in abdominal wall reconstruction.
Collapse
Affiliation(s)
- Lingbing Yang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Pu Wang
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yilin Zhang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Jin Zhou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Xuewei Bi
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhiyong Qian
- Department of Anatomy, Basic Medicine College, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Sen Hou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Linhao Li
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| |
Collapse
|
2
|
Liu S, Ren Z, Yan M, Ye W, Hu Y. Strategies to enhance the penetration of nanomedicine in solid tumors. Biomaterials 2025; 321:123315. [PMID: 40185056 DOI: 10.1016/j.biomaterials.2025.123315] [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: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Nanomedicine was previously regarded as a promising solution in the battle against cancer. Over the past few decades, extensive research has been conducted to exploit nanomedicine for overcoming tumors. Unfortunately, despite these efforts, nanomedicine has not yet demonstrated its ability to cure tumors, and the research on nanomedicine has reached a bottleneck. For a significant period of time, drug delivery strategies have primarily focused on targeting nanomedicine delivery to tumors while neglecting its redistribution within solid tumors. The uneven distribution of nanomedicine within solid tumors results in limited therapeutic effects on most tumor cells and significantly hampers the efficiency of drug delivery and treatment outcomes. Therefore, this review discusses the challenges faced by nanomedicine in penetrating solid tumors and provides an overview of current nanotechnology strategies (alleviating penetration resistance, size regulation, tumor cell transport, and nanomotors) that facilitate enhanced penetration of nanomedicine into solid tumors. Additionally, we discussed the potential role of nanobionics in promoting effective penetration of nanomedicine.
Collapse
Affiliation(s)
- Sen Liu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Zhendong Ren
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Manqi Yan
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wei Ye
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
3
|
Ismail M, Liu J, Wang N, Zhang D, Qin C, Shi B, Zheng M. Advanced nanoparticle engineering for precision therapeutics of brain diseases. Biomaterials 2025; 318:123138. [PMID: 39914193 DOI: 10.1016/j.biomaterials.2025.123138] [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: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Despite the increasing global prevalence of neurological disorders, the development of nanoparticle (NP) technologies for brain-targeted therapies confronts considerable challenges. One of the key obstacles in treating brain diseases is the blood-brain barrier (BBB), which restricts the penetration of NP-based therapies into the brain. To address this issue, NPs can be installed with specific ligands or bioengineered to boost their precision and efficacy in targeting brain-diseased cells by navigating across the BBB, ultimately improving patient treatment outcomes. At the outset of this review, we highlighted the critical role of ligand-functionalized or bioengineered NPs in treating brain diseases from a clinical perspective. We then identified the key obstacles and challenges NPs encounter during brain delivery, including immune clearance, capture by the reticuloendothelial system (RES), the BBB, and the complex post-BBB microenvironment. Following this, we overviewed the recent progress in NPs engineering, focusing on ligand-functionalization or bionic designs to enable active BBB transcytosis and targeted delivery to brain-diseased cells. Lastly, we summarized the critical challenges hindering clinical translation, including scalability issues and off-target effects, while outlining future opportunities for designing cutting-edge brain delivery technologies.
Collapse
Affiliation(s)
- Muhammad Ismail
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ningyang Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Changjiang Qin
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meng Zheng
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
4
|
Hao H, Sun S, Fu Y, Wen S, Wen Y, Yi Y, Peng Z, Fang Y, Tang J, Wang T, Wu M. Magnesium peroxide-based biomimetic nanoigniter degrades extracellular matrix to awake T cell-mediated cancer immunotherapy. Biomaterials 2025; 317:123043. [PMID: 39754969 DOI: 10.1016/j.biomaterials.2024.123043] [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: 08/20/2024] [Revised: 11/25/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
As the elite force of our immune system, T cells play a determining role in the effectiveness of cancer immunotherapy. However, the clever tumor cells construct a strong immunosuppressive tumor microenvironment (TME) fortress to resist the attack of T cells. Herein, a magnesium peroxide (MP)-based biomimetic nanoigniter loaded with doxorubicin (DOX) and metformin (MET) is rationally designed (D/M-MP@LM) to awake T cell-mediated cancer immunotherapy via comprehensively destroying the strong TME fortress. The nanoigniter not only effectively initiate CD8+ T cell-mediated immune response by promoting the presentation of tumor antigens, but also greatly facilitate the infiltration of T cells by degrading rigid extracellular matrix (ECM). More importantly, the nanoigniter significantly augment the effector functions of infiltrated CD8+ T cells by Mg2+-mediated metalloimmunotherapy and avoid the exhaustion of CD8+ T cells by improving the acidic TME. Thus, the nanoigniter comprehensively awakes T cells and achieves remarkable tumor inhibition efficacy.
Collapse
Affiliation(s)
- Huisong Hao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yanan Fu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Simin Wen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yingfei Wen
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yunfei Yi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jia Tang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Tianqi Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
5
|
Obeid MA, Alyamani H, Alenaizat A, Tunç T, Aljabali AAA, Alsaadi MM. Nanomaterial-based drug delivery systems in overcoming bacterial resistance: Current review. Microb Pathog 2025; 203:107455. [PMID: 40057006 DOI: 10.1016/j.micpath.2025.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Antimicrobial resistance is one of the most serious contemporary global health concerns, threatening the effectiveness of existing antibiotics and resulting in morbidity, mortality, and economic burdens. This review examines the contribution of nanomaterial-based drug delivery systems to solving the problems associated with bacterial resistance and provides a thorough overview of their mechanisms of action, efficiency, and perspectives for the future. Owing to their unique physicochemical properties, nanomaterials reveal new ways of passing through the traditional mechanisms of bacterial defence connected to the permeability barrier of membranes, efflux pumps, and biofilm formation. This review addresses the different types of nanomaterials, including metallic nanoparticles, liposomes, and polymeric nanoparticles, in terms of their antimicrobial properties and modes of action. More emphasis has been placed on the critical discussion of recent studies on such active systems. Both in vitro and in vivo models are discussed, with particular attention paid to multidrug-resistant bacteria. This review begins by reviewing the urgency for antimicrobial resistance (AMR) by citing recent statistics, which indicate that the number of deaths and reasons for financial losses continue to increase. A background is then provided on the limitations of existing antibiotic therapies and the pressing need to develop innovative approaches. Nanomaterial-based drug delivery systems have been proposed as promising solutions because of their potential to improve drug solubility, stability, and targeted delivery, although side effects can also be mitigated. In addition to established knowledge, this review also covers ongoing debates on the continuous risks associated with the use of nanomaterials, such as toxicity and environmental impact. This discussion emphasizes the optimization of nanomaterial design to target specific bacteria, and rigorous clinical trials to establish safety and efficacy in humans. It concludes with reflections on the future directions of nanomaterial-based drug delivery systems in fighting AMR, underlining the need for an interdisciplinary approach, along with continuous research efforts to translate these promising technologies into clinical practice. As the fight against bacterial resistance reaches its peak, nanomaterials may be the key to developing next-generation antimicrobial therapies.
Collapse
Affiliation(s)
- Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan.
| | - Hanin Alyamani
- William Harvey Research Institute, Center for Microvascular Research, Queen Mary University of London, London, United Kingdom
| | | | - Tutku Tunç
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan
| | - Manal M Alsaadi
- Department of Industrial Pharmacy, Faculty of Pharmacy, University of Tripoli, PO Box, Tripoli, 13645, Libya
| |
Collapse
|
6
|
Soman S, Kulkarni S, Kulkarni J, Dhas N, Roy AA, Pokale R, Mukharya A, Mutalik S. Metal-organic frameworks: a biomimetic odyssey in cancer theranostics. NANOSCALE 2025; 17:12620-12647. [PMID: 40336407 DOI: 10.1039/d5nr00794a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
This review comprehensively explores biomimetic metal-organic frameworks (MOFs) and their significant applications in cancer theranostics. Although MOFs have promising features such as adjustable porosity, improved surface area, and multifunctionality, they are limited by factors like low biocompatibility and specificity. Biomimetic strategies involving biological membranes and materials are proposed to address these challenges. The review begins by examining the unique characteristics and preparation methods of biomimetic carriers used in MOF-based nanoplatforms, with a comparative analysis of each method. It then delves into the various biomedical applications of biomimetic MOFs, including biosensing, bioimaging, immunotherapy, gene therapy, and multimodal therapies. The review also discusses the bio-interaction of these nanoplatforms, including their immunogenicity and interactions with fluids and tissues. Toxicity perspectives are also critically assessed. Overall, the article emphasizes the need for continued research into biomimetic MOFs, highlighting their potential to overcome current obstacles and provide safe, effective, and targeted therapeutic options.
Collapse
Affiliation(s)
- Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Jahnavi Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Rahul Pokale
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Anoushka Mukharya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
7
|
Chen Q, Yu Y, Tong L, Weiss RM, Wei SG. Targeted delivery of TAPI-1 via biomimetic nanoparticles ameliorates post-infarct left ventricle function and remodelling. Cardiovasc Res 2025; 121:760-774. [PMID: 40038918 PMCID: PMC12101320 DOI: 10.1093/cvr/cvaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/21/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
AIMS The potential of nanoparticles as effective drug delivery tools for treating failing hearts in heart failure remains a challenge. Leveraging the rapid infiltration of neutrophils into infarcted hearts after myocardial infarction (MI), we developed a nanoparticle platform engineered with neutrophil membrane proteins for the targeted delivery of TAPI-1, a TACE/ADAM17 inhibitor, to the inflamed myocardium, aiming to treat cardiac dysfunction and remodelling in rats with MI. METHODS AND RESULTS Neutrophil-mimic liposomal nanoparticles (Neu-LNPs) were constructed by integrating synthesized liposomal nanoparticles with LPS-stimulated neutrophil membrane fragments and then loaded with TAPI-1. MI rats were treated with TAPI-1 delivered via Neu-LNPs for 4 weeks. Left ventricular function was assessed by echocardiography and cardiac fibrosis was evaluated post-treatment. The novel Neu-LNPs maintained typical nanoparticle features, but with increased biocompatibility. Neu-LNPs demonstrated improved targeting ability and cellular internalization, facilitated by LFA1/Mac1/ICAM-1 interaction. Neu-LNPs displayed higher accumulation and cellular uptake by macrophages and cardiomyocytes in infarcted hearts post-MI, with a sustained duration. Treatments with TAPI-1-Neu-LNPs demonstrated greater protection against myocardial injury and cardiac dysfunction in MI rats compared to untargeted TAPI-1, along with reduced cardiac collagen deposition and expression of fibrosis biomarkers as well as altered immune cell compositions within the hearts. CONCLUSIONS Targeted treatment with TACE/ADAM17 inhibitor delivered via biomimetic nanoparticles exhibited pronounced advantages in improving left ventricle function, mitigating cardiac remodelling, and reducing inflammatory responses within the infarcted hearts. This study underscores the effectiveness of Neu-LNPs as a drug delivery strategy to enhance therapeutic efficacy in clinical settings.
Collapse
Affiliation(s)
- Qing Chen
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Yang Yu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Lei Tong
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
| | - Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
- Iowa Neuroscience Institute, University of Iowa, 169 Newton Road, Iowa City, IA 52242, USA
| |
Collapse
|
8
|
Lin YH, Chen CW, Chen MY, Xu L, Tian X, Cheung SH, Wu YL, Siriwon N, Wu SH, Mou KY. The Bacterial Outer Membrane Vesicle-Cloaked Immunostimulatory Nanoplatform Reinvigorates T Cell Function and Reprograms Tumor Immunity. ACS NANO 2025. [PMID: 40392526 DOI: 10.1021/acsnano.5c02541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Bacterial outer membrane vesicles (OMVs) represent powerful immunoadjuvant nanocarriers with the capacity to reprogram the tumor microenvironment (TME) and activate immune responses. Here, we investigate a nanotherapeutic platform integrating immunostimulatory cytosine-phosphate-guanine oligodeoxynucleotides (CpG-ODNs, hereafter termed CpG) into mesoporous silica nanoparticles cloaked with OMVs (CpG@MSN-PEG/PEI@OMVs) for cancer immunotherapy. Systemic administration of these nanohybrids facilitates precise tumor targeting, induces antitumor cytokines such as IFNγ, and suppresses immunosuppressive cytokine TGF-β, reshaping the TME. Additionally, CpG@MSN-PEG/PEI@OMVs promote M1 macrophage polarization, dendritic cell maturation, and the generation of durable tumor-specific immune memory, resulting in pronounced tumor regression with minimal systemic toxicity. The platform demonstrates efficacy against metastatic and solid tumor models including 4T1 breast and MC38 colorectal cancers. Transcriptomic analyses reveal that CpG@MSN-PEG/PEI@OMVs enhance mitochondrial oxidative phosphorylation in T cells within tumor-draining lymph nodes, mitigating T cell exhaustion and restoring metabolic fitness. These results support the potential of CpG@MSN-PEG/PEI@OMVs as a modular nanoplatform to modulate innate and adaptive immunity in cancer immunotherapy.
Collapse
Affiliation(s)
- Yu-Han Lin
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Wei Chen
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Mei-Yi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Li Xu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Xuejiao Tian
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
- Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 11529, Taiwan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Siu-Hung Cheung
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yen-Ling Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Natnaree Siriwon
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn 10540, Thailand
| | - Si-Han Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Kurt Yun Mou
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
9
|
Wen X, Hao Y. The combined application of exosomes/exosome-based drug preparations and ultrasound. J Mater Chem B 2025. [PMID: 40390561 DOI: 10.1039/d4tb01530d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Exosomes are small extracellular vesicles with a diameter of 30-150 nm, secreted by a variety of cells and containing various active substances such as nucleic acids, proteins and lipids. The use of exosomes as drug carriers for targeted delivery of therapeutics has been studied for a long time. Ultrasound is recognized as a non-invasive diagnostic and therapeutic method for assisting drug loading and targeted delivery, cellular uptake and therapy. In this review, we summarize the applications of ultrasound in assisting drug loading into exosomes, targeted delivery of exosome-based drug formulations, cellular uptake, and therapy, and explore the prospects for the combined application of exosomes/exosome-based drug formulations and ultrasound.
Collapse
Affiliation(s)
- Xiuli Wen
- Department of Ultrasound, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China.
| | - Yi Hao
- Department of Ultrasound, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China.
| |
Collapse
|
10
|
Fu P, Yang XF, Deng WW, Yu JN, Xu XM. Advances in cerebral edema research and targeted drug delivery systems. Eur J Pharmacol 2025; 1000:177744. [PMID: 40389128 DOI: 10.1016/j.ejphar.2025.177744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 05/06/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Cerebral edema, marked by excessive brain fluid accumulation, hinders stroke recovery and impacts survival, highlighting the need for effective therapies. This review examines the glymphatic system's role in post-stroke edema pathogenesis, explores edema formation mechanisms, and identifies therapeutic targets. While small molecule drugs show promise, their limited solubility and brain targeting necessitate advanced delivery approaches. Nanodrug delivery systems, capable of crossing the blood-brain barrier (BBB) and targeting cells via ligands, offer a compelling solution. We discuss the application of novel nanodrugs to enhance post-stroke edema treatment, aiming to improve survival and neurological recovery. This review seeks to guide future research in post-stroke edema management.
Collapse
Affiliation(s)
- Peng Fu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Xiu-Fen Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Wen-Wen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| | - Jiang-Nan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| | - Xi-Ming Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
11
|
Feng Y, Li Z, Song L, Zhang S, Chen B, Wang G, Yang K, Lu Y, Zhu R. A ZIF-8-based dual-modal smart responsive nanoplatform for overcoming radiotherapy resistance in advanced tumors. NANOSCALE 2025; 17:12134-12148. [PMID: 40298942 DOI: 10.1039/d5nr01093d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Radiation therapy is one of the core means of tumor treatment, playing an irreplaceable role in local control and radical treatment. However, radiotherapy resistance is one of the major challenges in current clinical practice. Tumor cells have a strong ability to repair DNA damage, which can effectively resist DNA double-strand breaks caused by X-rays, thus weakening the killing effect of radiotherapy. In addition, the complexity of the tumor microenvironment (TME) will further reduce the sensitivity of radiotherapy, leading to poor treatment results. With the rapid development of nanotechnology, the use of multi-modal combined therapy nanoplatforms has gradually become a new strategy to overcome radiotherapy resistance. These nanoplatforms achieve synergies by integrating multiple therapeutic approaches, such as radiation sensitization, photothermal therapy and chemotherapy. In this study, we utilized ZIF-8, a type of metal-organic framework, to simultaneously load ICG and rapamycin for X-ray sensitization and combined photothermal therapy. In this formulation, rapamycin enhances tumor cells' sensitivity to radiotherapy by inhibiting the mTOR signaling pathway, increasing DNA damage, regulating the cell cycle, and stimulating the STING pathway, which amplifies the tumor immune response. Meanwhile, ICG, as a photosensitizer, effectively converts light energy into heat, achieving tumor photothermal ablation. The modified drug-delivery system becomes a tumor-microenvironment-responsive smart carrier, increasing tumor cell uptake, prolonging retention at the tumor site, and achieving targeted drug delivery. It releases drugs in the specific tumor microenvironment, enhancing photothermal and radiotherapy sensitization effects. The results show that the smart dual-loaded nanoplatform effectively combines photothermal therapy and external-beam sensitization, reshapes the immunosuppressive tumor microenvironment and significantly inhibits tumor proliferation in the HepG2 subcutaneous xenograft model, demonstrating marked antitumor activity and good biosafety.
Collapse
Affiliation(s)
- Yi Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Zijing Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Luqi Song
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Shu Zhang
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radio-Toxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China
| | - Bin Chen
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Yan Lu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Ran Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
12
|
Gao F, Zhou R, He Y, Zhang Y, Bao C, Feng G. Bio-Mimicking Nanoparticle System Facilitates Sonodynamic-Mediated Clearance of Extensively Drug-Resistant Bacteria. ACS Biomater Sci Eng 2025; 11:2988-3002. [PMID: 40294106 DOI: 10.1021/acsbiomaterials.4c02455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The increasing prevalence of carbapenem-resistant and extensively drug-resistant Acinetobacter baumannii (XDR-Ab) poses a critical challenge in treating hospital-acquired pulmonary infections. In this study, we developed a biomimetic neutrophil membrane-coated nanoparticle system, NM@PCN-TIG, for the targeted delivery of tigecycline (TIG). The system utilizes the porphyrin-based metal-organic framework (MOF) PCN-224 as the core of the nanoparticle, encapsulating TIG and coated with a neutrophil membrane (NM) to enhance immune evasion and targeting of infection sites. Its loading efficiency, controlled release properties, cytotoxicity, and bactericidal activity under ultrasound mediation were systematically evaluated in vitro and in vivo. Our results demonstrated that NM@PCN-TIG significantly enhanced the bactericidal efficacy of TIG, increased reactive oxygen species (ROS) production, and promoted macrophage polarization toward an anti-inflammatory phenotype. This innovative biomimetic TIG nanosystem shows great potential as a platform for addressing XDR-Ab-induced pneumonia.
Collapse
Affiliation(s)
- Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Runlu Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Cui Bao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
13
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
14
|
Ye Z, Zhu M, Li S, Zhang F, Ran Y, Liu C, Xu X, Liu S, Xie X, Wang Y, Yao L. Multifunctional nanoparticles for immune regulation and oxidative stress alleviation in myocarditis. J Control Release 2025; 381:113607. [PMID: 40054631 DOI: 10.1016/j.jconrel.2025.113607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Cardiac autoimmune injury and oxidative stress play critical roles in the development of myocarditis. Promising approaches for treating this condition include suppressing excessive immune responses and reducing oxidative stress in the myocardium. The programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) axis is known to regulate immune responses and prevent damage caused by T-cell overactivation, while elevated reactive oxygen species (ROS) contribute to the progression of myocarditis. In this study, we developed multifunctional nanoparticles (PMN@EDR) that overexpress PD-L1 and are loaded with edaravone (EDR). The PMN@EDR NPs were successfully synthesized and comprehensively characterized. PMN@EDR effectively targeted inflammation-stimulated CD4+ T cells and damaged myocardial cells, inhibiting CD4+ T-cell proliferation, activation, and the release of pro-inflammatory cytokines via the PD-1/PD-L1 pathway. Additionally, PMN@EDR further suppressed CD4+ T-cell activation and alleviated HL-1 cardiomyocyte damage by releasing EDR to eliminate free radicals. For the in vivo treatment of myocarditis, compared to traditional single-target anti-inflammatory and antioxidant drugs, PMN@EDR not only reduced inflammation and the release of inflammatory mediators but also decreased ROS levels, thereby minimizing cardiomyocyte apoptosis and improving cardiac function. In conclusion, the PMN@EDR-based modulation of immune responses and oxidative stress offers a promising therapeutic strategy for myocarditis.
Collapse
Affiliation(s)
- Zhou Ye
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Manman Zhu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Shaojie Li
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Fan Zhang
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yingqi Ran
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Cong Liu
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Xiangchang Xu
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Shujiao Liu
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Xiang Xie
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yingchen Wang
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Lan Yao
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| |
Collapse
|
15
|
Xiao H, Raza F, Li K, Song J, Zafar H, Yang S, Su J, Qiu M. Cell membrane derived biomimetic nanomedicine for precision delivery of traditional Chinese medicine in cancer therapy. J Control Release 2025; 383:113829. [PMID: 40355044 DOI: 10.1016/j.jconrel.2025.113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
The rapidly developing modern nanotechnology has brought new vitality to the application of traditional Chinese medicine (TCM), improving the pharmacokinetics and bioavailability of unmodified natural drugs. However, synthetic materials inevitably introduce incompatibilities. This has led to focusing on biomimetic drug delivery systems (DDS) based on biologically derived cell membranes. This "top-down" approach to nanomedicine preparation is simple and effective, as the inherited cell membranes and cell surface substances can mimic nature when delivering drugs back into the body, interacting similarly to the source cells at the biological interface. The concept of biologically derived TCM and biomimetic membranes aligns well with nature, the human body, and medicine, thereby enhancing the in vivo compatibility of TCM. This review focused on the recent progress using biomimetic membranes for TCM in cancer therapy, emphasizing the effective integration of biomimetic nanomedicine and TCM in applications such as cancer diagnosis, imaging, precision treatment, and immunotherapy. The review also provided potential suggestions on the challenges and prospects in this field.
Collapse
Affiliation(s)
- Hang Xiao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinpu Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
16
|
Lee S, Kim B, Lee MJ, Kim D, Park S, Kim J, Arai Y, Lee SH. Curcumin-PLGA NPs coated with targeting biomimetic personalized stem cell membranes for osteoarthritis therapy. J Control Release 2025; 381:113625. [PMID: 40081740 DOI: 10.1016/j.jconrel.2025.113625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Traditional drug delivery systems for OA treatments face limitations due to rapid clearance within the joint and low biocompatibility. Moreover, the inflammation associated with OA exacerbates tissue damage and delays the regenerative capacity of therapeutics. To overcome these limitations, an OA-specific drug delivery system designated dCOL2-CM-Cur-PNPs is developed herein to target OA cartilage for anti-inflammatory and cartilage regeneration purposes. This system is constructed using cell membranes obtained from induced pluripotent stem cell -derived mesenchymal stem cells (iMSC-CMs), poly(D,l-lactide-co-glycolide) (PLGA) nanoparticles loaded with the well-known anti-inflammatory and cartilage-regenerating agent curcumin (Cur-PNPs), and damaged type II collagen (dCOL2)-targeting phospholipids. Coating the Cur-PNPs with iMSC-CMs enhances the sustained release of curcumin and improves its cellular uptake by OA-induced chondrocytes. The dCOL2-CM-Cur-PNPs restores the chondrogenic properties of the OA-induced chondrocytes, inhibit the pro-inflammatory function of M1 macrophages, and promote the anti-inflammatory function of M2 macrophages. The dCOL2-targeting phospholipids integrated on the surface of the iMSC-CMs facilitate specific binding to OA cartilage, as validated by in-vitro and in-vivo experiments. Additionally, the dCOL2-CM-Cur-PNPs alleviate OA progression in a DMM rat model. This drug delivery system based on iMSC-CMs modified with dCOL2-targeting phospholipids demonstrates significant potential as a next-generation platform for promoting cartilage regeneration through OA-specific therapy.
Collapse
Affiliation(s)
- Sunjun Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Min-Ju Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsik Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
17
|
Pereira-Silva M, Veiga F, Paiva-Santos AC, Concheiro A, Alvarez-Lorenzo C. Biomimetic nanosystems for pancreatic cancer therapy: A review. J Control Release 2025; 383:113824. [PMID: 40348133 DOI: 10.1016/j.jconrel.2025.113824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer (PC) is a highly lethal and aggressive malignancy, currently one of the leading causes of cancer-related deaths worldwide, in both women and men. PC is highly resistant to standard chemotherapy (CT) because its immunosuppressive and hypoxic tumor microenvironment and a dense desmoplastic stroma compartment extensively limit drug accessibility and perfusion. Although CT is one of the main therapeutic strategies for PC management contributing to tumor eradication through a cytotoxic effect, CT is associated with a poor pharmacokinetic profile and provokes deleterious systemic toxicity. This low efficacy-poor safety scenario urgently calls for innovative and highly specific therapeutic strategies to counteract this urgent clinical challenge. Nanotechnology-based precision materials for cancer may help improve drug stability and minimize the systemic cytotoxic effects by increasing drug tumor accumulation and also enabling controlled release, but several drawbacks still persist, such as the poor targeting efficiency. In the last few years increased attention has been paid to bioinspired nanosystems that can mimic either partially or totally biological systems, including lipid layers as suitable stealth coatings resembling the composition of cell membranes, lipoprotein- and blood protein-based nanosystems, and cell membrane-derived systems, such as extracellular vesicles, cell membrane nanovesicles and cell membrane-coated nanosystems, which display intrinsic cancer-targeting abilities, enhanced biocompatibility, decreased immunogenicity, and prolonged blood circulation profile. This review covers the recent breakthroughs on advanced biomimetic PC-targeted nanosystems, focusing on their design, properties and applications as innovative, multifunctional and versatile tools paving the way to improved PC diagnosis and treatment.
Collapse
Affiliation(s)
- Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
18
|
Zhang L, Mao Z, Li W, Chi S, Cheng H, Wang Z, Wang C, Liu Z. Promoting Drug Delivery to the Brain by Modulating the Transcytosis Process across the Blood-Brain Barrier. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27030-27048. [PMID: 40179275 DOI: 10.1021/acsami.5c02767] [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: 04/05/2025]
Abstract
The blood-brain barrier (BBB) presents a major challenge in the theranostics of brain diseases by impeding the delivery of drugs to the brain. Currently the most common strategy for transferring substances across the BBB is receptor-mediated transcytosis, which is restricted by several key factors, including insufficient endocytosis by brain microvessel endothelial cells (BMECs) due to underexpressed pinocytotic vesicles, lysosomal retention, and limited exocytosis to the brain parenchyma. We report a hybrid cell membrane (HCM)-coated and 2-methacryloyloxyethyl phosphorylcholine (MPC)-modified nanocarrier to promote drug delivery across the BBB by modulating the transcytosis process. The HCM incorporates a brain metastatic tumor cell membrane for recognition of BMECs and a GFP-293-S cell membrane expressing Spike protein to facilitate membrane fusion between the nanocarrier and BMECs, thereby bypassing vesicle-dependent endocytosis and enhancing cellular uptake. Membrane fusion reduces the chance of lysosomal retention, and MPC modification enhances exocytosis into the brain parenchyma via the interaction of MPC with transporters expressed on the abluminal endothelial membrane. The nanocarrier achieves significantly improved delivery of CuS, a photothermal agent, to the brain and thus enables highly efficient therapy of brain glioma.
Collapse
Affiliation(s)
- Li Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zhennan Mao
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Weibin Li
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Siyu Chi
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Hemei Cheng
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zijun Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Caixia Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zhihong Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
19
|
Yan X, Dong H, Gao L, Liu M, Wang C. Mechanism of selenium-doped black phosphorus nanosheets wrapped with biomimetic tumor cell membrane for prostate cancer immunotherapy. BIOMATERIALS ADVANCES 2025; 176:214339. [PMID: 40393102 DOI: 10.1016/j.bioadv.2025.214339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Prostate cancer (PCa) is commonly considered a "cold tumor" due to its immunosuppressive microenvironment. Cold tumors are typically identified by the absence of T-cell infiltration within the tumor, while other immune populations and myeloid cells can be observed in these tumors. To achieve light-heat combined immunotherapy checkpoint inhibitor treatment for castration-resistant prostate cancer, we aimed to transforming "cold tumors" into "hot tumors". We designed and synthesized a two-dimensional material, selenium-doped black phosphorus (BP), to enhance the photothermal conversion efficiency, and formed Se@BPNSs by liquid-phase exfoliation. To address the issue of enhanced permeability and retention effect, and to achieve efficient targeting, we coated the Se@BPNSs with RM-1 cell membrane derived from mouse prostate cancer cells. By injecting a certain dose of Se@BPNSs into the tumor and irradiating with a 808 nm laser, the Se@BPNSs converted light energy into heat to kill tumor cells at high temperatures while releasing antigens captured by dendritic cells. In addition, we combined the immunotherapy checkpoint inhibitor anti-PD1 to enhance the immune response and promote immune cell infiltration. The successful preparation of Se@BPNSs was verified through material characterization, cell-level and animal-level experiments, and the antitumor effect was meanwhile verified, which further provided guidance for prostate cancer treatment by photothermal synergistic immunotherapy.
Collapse
Affiliation(s)
- Xingjian Yan
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China.
| | - Han Dong
- Department of Geriatric, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China
| | - Liyin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China
| | - Mengqi Liu
- Key Laboratory of Bionic Engineering, Ministry of Education, College of Biological and Agricultural Engineering, Jilin University, Changchun City, Jilin Province 130022, China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China.
| |
Collapse
|
20
|
Wang R, Wei X, He X, Wang L, Zhou M, Tang J, Che X, Zhou G, Liu H. ML228-loaded nanoparticles with platelet membrane coating promote endothelialization of vascular grafts by enhancing HIF-1α expression. Colloids Surf B Biointerfaces 2025; 253:114756. [PMID: 40334473 DOI: 10.1016/j.colsurfb.2025.114756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Small-diameter vascular grafts (SDVGs) often struggle to maintain long-term patency due to thrombus formation, intimal hyperplasia, and inflammation. Endothelialization emerges as a pivotal strategy for addressing these concerns. As a representative activator of the hypoxia-inducible factor (HIF) pathway, ML228 can stimulate the expression of downstream target genes like vascular endothelial growth factor (VEGF) to induce angiogenesis, yet it requires encapsulation by nanoparticles for optimal delivery and efficacy. However, the immune system often recognizes nanoparticles as foreign entities, posing a significant risk of clearance. In this study, we developed ML228-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles and coated them with platelet membranes, thereby enhancing their biocompatibility and enabling immune escape. The ML228-loaded PLGA nanoparticles coated with platelet membranes (MPNP) were immobilized onto electrospinning SDVGs made of silk fibroin (SF) and polycaprolactone (PCL) to obtain MPNP-coated grafts (SF/PCL@MPNP) with the ability to promote endothelialization. In vitro biological activity studies demonstrated that SF/PCL@MPNP activated the HIF pathway, upregulating the downstream target gene VEGF, which facilitated endothelial cells migration and angiogenesis. In vivo implantation in a rat abdominal aorta model revealed that SF/PCL@MPNP promoted endothelialization, supported the regeneration of contractile smooth muscle cells, and modulated inflammatory responses. Overall, this study presents a strategy for constructing SDVGs using ML228-loaded nanoparticles with platelet membrane coating, highlighting the promises of using ML228 to activate the HIF pathway and membrane-coated nanoparticles to improve endothelialization in vascular graft applications.
Collapse
Affiliation(s)
- Ruichen Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinbo Wei
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xi He
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Li Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, P. R. China
| | - Moyan Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Jiarui Tang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xin Che
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Gang Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Haifeng Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
21
|
Zhong X, Deng X, Yang Y, Xie X, Li B, Peng X. Immuno-engineered macrophage membrane-coated nanodrug to restore immune balance for rheumatoid arthritis treatment. Acta Biomater 2025; 197:374-385. [PMID: 40097125 DOI: 10.1016/j.actbio.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Current immunosuppressive therapies for rheumatoid arthritis (RA) lack disease specificity, primarily targeting inflammation while causing debilitating side effects. To address this limitation, we developed a biomimetic nanodrug MP@NEs/CT to induce antigen-specific immune tolerance for precise, effective and safe RA immunotherapy. MP@NEs/CT features a core of multiepitope citrullinated peptide (CitP) and triptolide (TPL) co-loaded nanoemulsion and coated with a macrophage membrane harvested from IFN-γ treated RAW264.7 cells. CitP, an RA autoantigen, specifically targets the immune response, while TPL acts as an immunosuppressant by inhibiting dendritic cells (DCs) maturation. IFN-γ treatment upregulates programmed death-ligand 1 (PD-L1) expression, facilitating MP@NEs/CT accumulation within inflamed tissues via programmed death-1 (PD-1) binding following intravenous administration. Additionally, the immune-engineered macrophage membrane sequesters proinflammatory cytokines, further dampening local inflammation. A significant reduction of CII-specific IgG levels in collagen-induced arthritis (CIA) mice model provides the evidence of CitP in restoring antigen-specific immune tolerance. Importantly, a low dose of TPL within MP@NEs/CT promotes tolerogenic DCs and generation of anti-inflammatory cytokines, ultimately leading to upregulation of antigen-specific regulatory T cells (Tregs) and B cells (Bregs) and a reduction in pro-inflammatory cytokine levels. Consequently, the nanodrug demonstrates synergistic and effective anti-inflammatory and immunosuppressive effects, alleviating autoimmune damage in a CIA mice model. STATEMENT OF SIGNIFICANCE: : Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by dysregulated immune responses, leading to synovial hyperplasia, tissue destruction, and irreversible disability. Early work in RA therapy mainly applying anti-inflammatory drugs which focuses on delaying joint deformity, but have no effects on the aberrant immune response. However, these drugs often require high doses and long-term administration, leading to potential adverse effects. In this work, we reported a therapeutic system that co-delivery of autoantigens with immune modulators promotes antigen-specific tolerance for effective and safe RA immunotherapy.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/therapy
- Mice
- Macrophages/immunology
- RAW 264.7 Cells
- Nanoparticles/chemistry
- Diterpenes/pharmacology
- Diterpenes/chemistry
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/drug therapy
- Cell Membrane/chemistry
- Cell Membrane/immunology
- Phenanthrenes/pharmacology
- Phenanthrenes/chemistry
- Mice, Inbred DBA
- Immunotherapy
- Coated Materials, Biocompatible/pharmacology
- Coated Materials, Biocompatible/chemistry
- Epoxy Compounds
Collapse
Affiliation(s)
- Xiaofang Zhong
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Xiaoyu Deng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Yongqing Yang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Xin Xie
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Bowen Li
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore.
| | - Xinsheng Peng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China.
| |
Collapse
|
22
|
Luo X, Zhou Y, Rao K, Xiang J, Ning S, Zhu D, Li G, Chen H. Biomimetic Cascade Nanozyme Catalytic System for the Treatment of Lymph Node Metastasis in Gastric Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411576. [PMID: 40123244 DOI: 10.1002/smll.202411576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/18/2025] [Indexed: 03/25/2025]
Abstract
Lymphatic metastasis of gastric cancer is a challenging issue in clinical practice. Recently, copper single-atom nanozymes (SAZ) have gained tremendous attention due to its superior peroxidase (POD) activity that has good nonocatalytic tumor therapy (NCT) capabilities, and photothermal properties. Therefore, using a high-expressing P-selectin platelet membrane (PM) to encapsulate SAZ and cisplatin is proposed, forming PSC nanoparticles. Due to their exquisite nanoscale size and the unique structure of lymphatic vessels, PSC can highly target cancer cells in invasive primary tumors and metastatic lymph nodes that both highly express CD44. It is noteworthy that cisplatin can simultaneously perform chemotherapy and generate H₂O₂ under the action of NADPH oxidases (NOXs) that further enhance the catalytic activity of SAZ and increase intracellular reactive oxygen species (ROS) production. Both in vitro and vivo experiments have demonstrated the superior targeting and elimination capability of the PCS system in primary and metastatic tumor cells. In addition, transcriptomic analysis reveals that PSC + NIR induced apoptosis in MFC cells. This marks the first proposal of combining single-atom nanozymes and chemotherapy drugs for dual-targeting in gastric cancer and lymphatic metastasis, providing new insights into a challenging clinical issue in the treatment of gastric cancer lymphatic metastasis.
Collapse
Affiliation(s)
- Xi Luo
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yingguang Zhou
- Department of Joint Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, P. R. China
| | - Kexiang Rao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jingfeng Xiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, P. R. China
| | - Daoming Zhu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
- Cancer Center of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, P. R. China
| | - Hao Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
23
|
Lin S, Xu Z, Liu Y, Yang G, Qi X, Huang Y, Zhou M, Jiang X. Engineered Macrophage Membrane-Camouflaged Nanodecoys Reshape the Infectious Microenvironment for Efficient Periodontitis Treatment. ACS NANO 2025; 19:15345-15362. [PMID: 40228155 DOI: 10.1021/acsnano.4c14305] [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: 04/16/2025]
Abstract
A vicious cycle between microbiota dysbiosis and hyperactivated inflammation, hardly disrupted by conventional therapies, remains a significant clinical challenge for periodontitis treatment. Herein, by cloaking a cascade catalysis system in an engineered macrophage membrane, a nanodecoy-based strategy, with targeted bacteria-killing and immunomodulatory abilities, is proposed for reshaping the hostile periodontitis microenvironment. Specifically, recombinant human antimicrobial peptide, LL-37, is anchored to a Toll-like receptor-enriched macrophage membrane via genetic engineering, which facilitates the specific bacteria elimination and efficient tissue retention of the nanodecoys. Moreover, the cascade catalysis system integrates L-amino acid oxidase (LAAO) with hollowed manganese dioxide (hMnO2) by reciprocal elevation of the catalytic efficiency of hMnO2 and LAAO, leading to accelerated O2 generation under a hypoxic microenvironment and disrupted metabolism of periodontopathogenic bacteria. Notably, the nanodecoys trigger the nuclear translocation of NF-E2-related factor-2 (NRF2) to reduce oxidative stress response and rewire the polarization of macrophages, thereby boosting the osteogenic differentiation of osteoblasts. Furthermore, the alveolar bone regeneration therapeutically benefits from the nanodecoys in vivo. Altogether, these results highlight the attractive functions of engineered macrophage membrane-cloaked nanodecoys for effective periodontitis treatment.
Collapse
Affiliation(s)
- Sihan Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Zeqian Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Yulan Liu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Xuanyu Qi
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Yijia Huang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, People's Republic of China
- National Center for Stomatology, Shanghai 200011, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, People's Republic of China
- Shanghai Stomatological Hospital, Fudan University, Shanghai 201102, People's Republic of China
| |
Collapse
|
24
|
Chen H, Zhang Y, Aikebaier M, Du Y, Liu Y, Zha Q, Zheng L, Shan S, Wang Y, Chen J, Li Y, Yang K, Yang Y, Cui W. Decursin-Loaded Nanovesicles Target Macrophages Driven by the Pathological Process of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417489. [PMID: 40285666 DOI: 10.1002/advs.202417489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Atherosclerosis (AS) is a major pathological factor contributing to the mortality associated with ischemic heart disease and is driven primarily by macrophage-mediated lipid accumulation and inflammatory processes. Conventional cardiovascular pharmacotherapies address these pathological mechanisms but often show limited efficacy, highlighting the need for innovative agents capable of effectively reducing lipid accumulation and inflammation with minimal toxicity. In this study, decursin, a monomer derived from traditional Chinese medicine, is shown to inhibit both lipid accumulation and inflammatory responses in macrophages through direct interaction with protein kinase Cδ (PKCδ), resulting in low cytotoxicity in vitro and negligible toxicity in vivo. To address the short half-life of decursin, a targeted cascade drug delivery system (ALD@EM), which is specifically designed to target AS pathophysiology, is developed. This system employs ICAM-1 and VCAM-1 antibodies for plaque localization and incorporates low-density lipoproteins (LDLs) to facilitate chemotaxis to lesion sites, with an inner layer of apoptotic endothelial cell membranes to increase macrophage internalization and drug release. As a result, ALD@EM nanovesicles significantly increased the accumulation and therapeutic efficacy of decursin within plaques, substantially reducing lipid deposition and plaque inflammation, thereby offering a novel strategy for targeted AS treatment.
Collapse
Affiliation(s)
- Hui Chen
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mirenuer Aikebaier
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200002, China
| | - Yan Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing Zha
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuyao Shan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanping Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiawei Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yiping Li
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Wenguo Cui
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
25
|
Li L, Sun C, Cai S, Hang Z, Gao X, Hou L, Li L, Wu Y, Xing C, Du H. Biomimetic Nanoparticles Enhance Recovery of Movement Disorders in Parkinson's Disease by Improving Microglial Mitochondrial Homeostasis and Suppressing Neuroinflammation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23536-23552. [PMID: 40202159 DOI: 10.1021/acsami.4c22181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Neuroinflammation is a key risk factor for cognitive impairment, and microglia are the main drivers. Metformin has been shown to suppress inflammation and reduce microglial activation, protecting neurons from damage. However, its clinical efficacy is limited by low bioavailability and metabolic challenges, especially in terms of precise delivery to specific targets. To overcome this problem, we developed biomimetic microglial nanoparticles (MePN@BM) to enhance the targeted delivery and bioavailability of metformin. Through homologous targeting, the delivery efficiency of drugs in the inflammatory site of Parkinson's disease was enhanced to improve the therapeutic effect. The results showed that MePN@BM effectively delivers metformin to the brain, promotes autophagy, restores mitochondrial membrane potential, and reduces oxidative stress. In a Parkinson's disease (PD) mouse model, MePN@BM improved motor function, repaired dopaminergic neurons, and cleared α-synuclein aggregates. Notably, transcriptome analysis revealed enriched inflammation-related pathways, and immunofluorescence showed that PD mice treated with MePN@BM had higher levels of anti-inflammatory factors and lower levels of pro-inflammatory factors. Therefore, it provides a promising strategy for the treatment of inflammation-mediated motor dysfunction.
Collapse
Affiliation(s)
- Liang Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Chunbin Sun
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Shanglin Cai
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xiaoyu Gao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liangxuan Hou
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Luping Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yawen Wu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
26
|
Bahlol HS, Zhang K, Deng J, Zhang W, Ma Z, Zhang J, Han H. Biomimetic Copper-Based Nanoplatform for Enhanced Tumor Targeting and Effective Melanoma Therapy. ACS APPLIED BIO MATERIALS 2025; 8:3290-3299. [PMID: 40186581 DOI: 10.1021/acsabm.5c00074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Designing advanced biomimetic nanoplatforms that combine photothermal therapy (PTT) and immune activation represents a modern approach to addressing the challenges of cancer therapy. This study presents a nanobiomimetic hollow copper-sulfide (HCuS) platform for precise homotypic tumor targeting and melanoma treatment. The HCuS@OVA@CM (COC) nanoplatform-encapsulated ovalbumin (OVA) antigen protein within HCuS nanoparticles and was coated with melanoma cell membranes (B16F10). Importantly, this design facilitates specific tumor accumulation and achieves 16.0% photothermal conversion efficiency under 1064 nm NIR-II irradiation, which is a key factor for therapeutic success. In vitro studies have demonstrated that this nanoplatform induces immunogenic cell death (ICD), enhances antigen presentation, and stimulates dendritic cell (DCs) maturation. In vivo experiments confirmed that COC-mediated NIR-II photothermal treatment significantly suppressed tumor growth without notable body weight loss. This biomimetic nanoplatform approach offers a targeted, enhanced, and effective immune response for tumor photothermal immunotherapy, making it a promising candidate for advanced melanoma treatment and anticancer therapy.
Collapse
Affiliation(s)
- Hagar Shendy Bahlol
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, China
- Department of Biochemistry, Faculty of Agriculture, Benha University, Moshtohor, Toukh 13736, Egypt
| | - Kai Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Jiamin Deng
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, China
| | - Weiyun Zhang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhaoyu Ma
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Jin Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Heyou Han
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
27
|
Lei H, Cui H, Xia Y, Sun F, Zhang W. Illuminating Hope for Tumors: The Progress of Light-Activated Nanomaterials in Skin Cancer. Int J Nanomedicine 2025; 20:5081-5118. [PMID: 40264819 PMCID: PMC12013650 DOI: 10.2147/ijn.s506000] [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: 11/12/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Skin cancer is a common malignant tumor that poses significant global health and economic burdens. The main clinical types include malignant melanoma and non-melanoma. Complications such as post-surgical recurrence, wound formation, or disfigurement can severely impact the patient's mental well-being. Traditional treatments such as surgery, chemotherapy, radiation therapy, and immunotherapy often face limitations. These challenges not only reduce the effectiveness of treatments but also negatively impact patients' quality of life. Phototherapy, a widely used and long-standing method in dermatology, presents a promising alternative for skin cancer treatment. Light-triggered nanomaterials further enhance the potential of phototherapy by offering advantages such as improved therapeutic precision, controlled drug release, minimal invasiveness, and reduced damage to surrounding healthy tissues. This review summarizes the application of light-triggered nanomaterials in skin cancer treatment, focusing on the principles, advantages, and design strategies of photodynamic therapy (PDT), photothermal therapy (PTT), and photoacoustic therapy (PAT). In this manuscript we have an in-depth discussion on overcoming translational barriers, including strategies to enhance light penetration, mitigate toxicity, reduce production costs, and optimize delivery systems. Additionally, we discuss the challenges associated with their clinical translation, including limited light penetration in deep tissues, potential toxicity, high production costs, and the need for advanced delivery systems.
Collapse
Affiliation(s)
- Huaqing Lei
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| | - Hengqing Cui
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Institute of Aesthetic Plastic Surgery and Medicine, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yu Xia
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Fujia Sun
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
28
|
Xu C, Liu Y, Pan Y, Zhang H, Sun Y, Li J, Wu A, Bian L. Neutrophil-like cell membrane-coated molybdenum-based nanoclusters for reduced oxidative stress and enhanced neurological recovery after intracerebral hemorrhage. Acta Biomater 2025:S1742-7061(25)00286-7. [PMID: 40254230 DOI: 10.1016/j.actbio.2025.04.035] [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: 11/09/2024] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Excessive reactive oxygen species (ROS) are detrimental to the brain that can result in neurological impairment and inhibiting neurological functionals recovery after intracerebral hemorrhage (ICH). However, there is still a lack of effective treatment for ICH, either with medicine or neurosurgery. Nanozymes with excellent superoxide dismutase and catalase properties can scavenge ROS and may provide therapeutic opportunities for ICH patients. However, the ability of nanozymes to non-invasively target cerebral hemorrhage lesions and further antioxidation effect are still unknown. Herein, neutrophile membrane-disguised molybdenum-based polyoxometalate nanozymes (POM@Mem) were developed to alleviate oxidative stress after ICH. Coating with neutrophil membrane allowed POM to target the hemorrhage sites and further inhibit ROS generation. POM@Mem can improve neuroinflammatory microenvironment and promote behavioral improvement of ICH mouse. Combining neutrophile membrane and nanozymes for targeting brain hemorrhage sites provides an effective strategy for the treatment of ICH. STATEMENT OF SIGNIFICANCE: Excessive reactive oxygen species (ROS) are detrimental to the brain and can lead to neurological impairment, hindering the recovery of neurological functions after intracerebral hemorrhage (ICH). Despite this, effective treatments for ICH, whether pharmaceuticals or neurosurgery, remain scarce. In this study, we developed neutrophil membrane-disguised molybdenum-based polyoxometalate nanozymes (POM@Mem) as a novel approach to alleviate oxidative stress following ICH. The neutrophil membrane coating enabled the POM nanozymes to specifically target hemorrhagic sites, thereby inhibiting ROS production. Additionally, POM@Mem improved the neuroinflammatory microenvironment and facilitated behavioral recovery in ICH mice. The combination of neutrophil membranes and nanozymes for targeted delivery to brain hemorrhage sites offers a promising strategy for the treatment of ICH.
Collapse
Affiliation(s)
- Canxin Xu
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China; Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Yikui Liu
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yuanbo Pan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Hongchi Zhang
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yuhao Sun
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Juan Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Ningbo Cixi Institute of Biomedical Engineering, Cixi 315300, China.
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Ningbo Cixi Institute of Biomedical Engineering, Cixi 315300, China.
| | - Liuguan Bian
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| |
Collapse
|
29
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
30
|
Zhang Z, Ma T, Liu Q, Nan J, Liu G, Yang Y, Hu Y, Xie J. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells Encapsulated in M2 Macrophage Cell Membrane Targeted to Inhibit Joint Periprosthetic Inflammation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22279-22292. [PMID: 40168527 DOI: 10.1021/acsami.4c22304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Periprosthetic osteolysis (PPOL) is a serious complication following total joint replacement surgery, and exploring treatments for this complication is of significant societal importance. Exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos, Exos) have diverse cellular functions, such as inhibiting osteoclast formation, suppressing inflammation progression, and promoting M2 macrophage polarization. However, standalone Exosomes are easily recognized and phagocytosed by the immune system, have a short half-life, and lack specificity. This study is based on the homing effect possessed by M2 macrophages under the regulation of various factors. By combining this with cell membrane encapsulation technology and embedding BMSC-Exos within the membrane of M2 macrophages (M2M-Exos), the aim is to inhibit inflammation and treat PPOL. It was found that M2M-Exos can target the PPOL area, enhancing the therapeutic effects of the BMSC-Exos and reducing wear particle-induced cranial osteolysis. Additionally, M2M-Exos provide immune camouflage through the cell membrane, allowing the BMSC-Exos to evade clearance by the mononuclear macrophage system in the body. Therefore, the study demonstrates the targeting ability of M2M-Exos and their unique role in preventing PPOL. These biomimetic nanoparticles establish a targeted nanodrug delivery system for PPOL treatment.
Collapse
Affiliation(s)
- Zheyu Zhang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Tianliang Ma
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Qimeng Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jiangyu Nan
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Guanzhi Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yute Yang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jie Xie
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
31
|
Yan D, Li X, Wang H, Li B, Wang W, Liao Y, Tang BZ, Wang D. NIR-II aggregation-induced emission nanoparticles camouflaged with preactivated macrophage membranes for phototheranostics of pulmonary tuberculosis. Nat Protoc 2025:10.1038/s41596-025-01146-8. [PMID: 40210746 DOI: 10.1038/s41596-025-01146-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/10/2025] [Indexed: 04/12/2025]
Abstract
Phototheranostics, which allows simultaneous diagnosis and therapy, offers notable advantages in terms of noninvasiveness, controllability and negligible drug resistance, presenting a promising approach for disease treatment. By integrating second near-infrared (NIR-II, 1,000-1,700 nm) phototheranostic agents characterized by aggregation-induced emission (AIE) and cell membranes with specific targeting capacity, we have developed a versatile type of biomimetic nanoparticle (NP) for precise phototheranostics of pulmonary tuberculosis (TB). Coating the phototheranostic agents with preactivated macrophage membranes results in the formation of biomimetic NPs, which exhibit specific binding to TB through a lesion-pathogen dual-targeting strategy, allowing the accurate detection of Mycobacterium tuberculosis via NIR-II fluorescence imaging and precise photothermal therapy using the irradiation of a 1,064 nm laser. In comparison with traditional treatments, small individual granulomas (0.2 mm in diameter) in TB-infected mice are visualized, and improved antibacterial effects are achieved upon NP administration. Here we present a standardized workflow for the synthesis of the NIR-II AIE agents, their use for the fabrication of the biomimetic NPs and their in vitro and in vivo applications as phototheranostics against M. tuberculosis. The preparation and characterization of the NIR-II AIE agents requires ~8 d, the synthesis and characterization of the phototheranostic NPs requires ~8 d, the validation of in vitro targeting capacity and photothermal eradication requires ~26 d, and the in vivo NIR-II fluorescence imaging and imaging-guided photothermal therapy requires ~74 d. All procedures are straightforward and suitable for clinicians or researchers with prior training in organic synthesis and biomedical engineering.
Collapse
Affiliation(s)
- 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, China
| | - Xue Li
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, China
| | - Huanhuan Wang
- Institute for Engineering Medicine, Kunming Medical University, Kunming, China
| | - Bin Li
- Institute for Engineering Medicine, Kunming Medical University, Kunming, China
| | - Wei Wang
- Institute for Engineering Medicine, Kunming Medical University, Kunming, China
| | - Yuhui Liao
- Institute for Engineering Medicine, Kunming Medical University, Kunming, China.
| | - Ben Zhong Tang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering, Shenzhen University, Shenzhen, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 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, China.
| |
Collapse
|
32
|
Li X, Wu F, Yu D, Su X, Wang K, Huang Z, Lu Z. Archaea-inspired deoxyribonuclease I liposomes prevent multiple organ dysfunction in sepsis. J Control Release 2025; 380:1109-1126. [PMID: 39986474 DOI: 10.1016/j.jconrel.2025.02.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Neutrophil extracellular traps (NETs) and circulating cell-free DNA (cfDNA) are pivotal in driving excessive inflammation and organ damage during sepsis, with their levels correlating positively with sepsis severity in both patients and murine models. Despite the ability of deoxyribonuclease I (DNase I) to degrade NETs and cfDNA, its short half-life and rapid degradation limit its therapeutic effectiveness. To address this challenge, we developed a methyl-branched liposome fused with a red blood cell membrane for the systemic delivery of DNase I (DNase I/Rm-Lipo). The efficacy of DNase I/Rm-Lipo was evaluated in the stimulated immune cells and septic model. The data confirmed that DNase I/Rm-Lipo efficiently removed excess NETs and cfDNA in activated neutrophils. Following injection, DNase I/Rm-Lipo exhibited an extended circulation time, effectively suppressing neutrophil activation and regulating macrophage polarization to mitigate inflammation and prevent organ dysfunction in septic mice. These findings highlight the therapeutic potential of DNase I/Rm-Lipo as a promising candidate for sepsis management by targeting the degradation of NETs and cfDNA.
Collapse
Affiliation(s)
- Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Fan Wu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Dedong Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiayi Su
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Kaikai Wang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Zhiwei Huang
- Central Laboratory, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China.
| |
Collapse
|
33
|
Krishnan N, Zhou J, Mohapatra A, Ventura CJ, Duan Y, Lee J, Hoenig JJ, Nizet O, Gao W, Fang RH, Zhang L. Universal Prophylactic Antitumor Vaccination Using Stem Cell Membrane-Coated Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501616. [PMID: 39989146 PMCID: PMC11983251 DOI: 10.1002/adma.202501616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Cancer vaccines are a promising immunotherapeutic modality that function by training the immune system to recognize and destroy malignant cells. As tumor-specific and tumor-associated antigens generally cannot be identified until after a tumor has already been established, these vaccines must be applied therapeutically when strong immunosuppressive mechanisms are already in place. Building upon previous work using cell membrane coating nanotechnology, the development of a broad-spectrum prophylactic cancer nanovaccine that consists of induced pluripotent stem cell (iPSC) membrane coated around an adjuvant-loaded nanoparticle core is shown. The resulting nanostructure is capable of presenting iPSC-derived oncofetal antigens, which are oftentimes re-expressed on cancer cells but lowly present on normal adult tissues. When administered in vivo, the iPSC membrane-coated nanoparticles are highly immunostimulatory and elicit strong antitumor immunity that can successfully inhibit the growth of multiple tumor types, including five different murine tumor models and in a bilaterial heterogeneous tumor model. Overall, this work demonstrates an effective approach for engineering iPSC-based nanovaccines that can be applied broadly to prevent cancer before it occurs.
Collapse
Affiliation(s)
- Nishta Krishnan
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Jiarong Zhou
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Animesh Mohapatra
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Christian John Ventura
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Yaou Duan
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Junyong Lee
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Jacob J. Hoenig
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Oliver Nizet
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Ronnie H. Fang
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
- Division of Host‐Microbe Systems and TherapeuticsDepartment of PediatricsUniversity of California San DiegoLa JollaCA92093USA
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department of Chemical and Nano EngineeringUniversity of California San DiegoLa JollaCA92093USA
| |
Collapse
|
34
|
An Y, Ji C, Zhang H, Jiang Q, Maitz MF, Pan J, Luo R, Wang Y. Engineered Cell Membrane Coating Technologies for Biomedical Applications: From Nanoscale to Macroscale. ACS NANO 2025; 19:11517-11546. [PMID: 40126356 DOI: 10.1021/acsnano.4c16280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Cell membrane coating has emerged as a promising strategy for the surface modification of biomaterials with biological membranes, serving as a cloak that can carry more functions. The cloaked biomaterials inherit diverse intrinsic biofunctions derived from different cell sources, including enhanced biocompatibility, immunity evasion, specific targeting capacity, and immune regulation of the regenerative microenvironment. The intrinsic characteristics of biomimicry and biointerfacing have demonstrated the versatility of cell membrane coating technology on a variety of biomaterials, thus, furthering the research into a wide range of biomedical applications and clinical translation. Here, the preparation of cell membrane coatings is emphasized, and different sizes of coated biomaterials from nanoscale to macroscale as well as the engineering strategies to introduce additional biofunctions are summarized. Subsequently, the utilization of biomimetic membrane-cloaked biomaterials in biomedical applications is discussed, including drug delivery, imaging and phototherapy, cancer immunotherapy, anti-infection and detoxification, and implant modification. In conclusion, the latest advancements in clinical and preclinical studies, along with the multiple benefits of cell membrane-coated nanoparticles (NPs) in biomimetic systems, are elucidated.
Collapse
Affiliation(s)
- Yongqi An
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cheng Ji
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Hao Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
| | - Junqiang Pan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an 710003, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Research Unit. of Minimally Invasive Treatment of Structural Heart-Disease, Chinese Academy of Medical Sciences (2021RU013), Chengdu, 610065, China
| |
Collapse
|
35
|
Ni C, Li X, Jiang H, Gui S, Yin H, Nie X. A targeted and synergetic nano-delivery system against Pseudomonas aeruginosa infection for promoting wound healing. Mater Today Bio 2025; 31:101470. [PMID: 39882550 PMCID: PMC11772151 DOI: 10.1016/j.mtbio.2025.101470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Purpose Pseudomonas aeruginosa infection is the most common pathogen in burn wound infections, causing delayed wound healing and progression to chronic wounds. Therefore, there is an urgent need to develop antimicrobial agents that can promote wound healing for effectively treating infected wounds. Patients and methods Using magnetic stirring and ultrasound to synthesize Apt-pM@UCNPmSiO2-Cur-CAZ. The nanosystems were characterized using transmission electron microscopy (TEM), dynamic light scattering (DLS), and ultraviolet-visible spectrophotometry (UV-Vis). Flow cytometry, bacterial LIVE/DEAD staining and scanning electron microscopy were performed to assess the in vitro antibacterial and anti-biofilm effects of the nanosystems. The wound healing potential and in vivo toxicity of the nanosystems were evaluated in a mouse skin wound model. Results The Apt-pM@UCNPmSiO2-Cur-CAZ synthesized exhibited uniform circular shape with a Zeta potential of -0.8 mV. In vitro, Apt-pM@UCNPmSiO2-Cur-CAZ demonstrated superior antibacterial effects compared to standalone antibiotics. Bacteria treated with Apt-pM@UCNPmSiO2-Cur-CAZ showed varying degrees of deformation and shrinkage, resulting in severe damage to the bacterial cells. Additionally, Apt-pM@UCNPmSiO2-Cur-CAZ can inhibit and eradicate bacterial biofilms, while also targeting bacteria for enhanced antibacterial efficacy. Interestingly, the NIR light could enhance the antibacterial and anti-biofilm effects of Apt-pM@UCNPmSiO2-Cur-CAZ due to the photodynamic action. In a mouse skin wound infection model, the nanosystem effectively eliminated wound bacteria, promoting the healing of Pseudomonas aeruginosa-infected wounds without significant toxic effects. Conclusion Apt-pM@UCNPmSiO2-Cur-CAZ is a novel targeted nano-delivery system with promising potential in combating Pseudomonas aeruginosa infections, and it may serve as a new therapeutic approach for treating skin wound infections.
Collapse
Affiliation(s)
| | | | - Haiye Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Shumin Gui
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Heng Yin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Xinmin Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| |
Collapse
|
36
|
Li J, Yang R, Zhang C, Lovell JF, Zhang Y. Manganese-Driven Plasmid Nanofibers Formed In Situ for Cancer Gene Delivery and Metalloimmunotherapy. J Am Chem Soc 2025; 147:10504-10518. [PMID: 40065259 DOI: 10.1021/jacs.4c18511] [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: 03/27/2025]
Abstract
While nucleic-acid-based cancer vaccines hold therapeutic potential, their limited immunogenicity remains a challenge due in part to the low efficiency of cytoplasmic delivery caused by lysosomal entrapment. In this work, we found that plasmids encoding both an antigen and a STING agonist protein adjuvant can self-assemble into coordination nanofibers, triggered by manganese ions. We developed a strategy to construct a DNA vaccine, termed MnO2-OVA-CDA-mem, formed by the coencapsulation of manganese dioxide (MnO2), an antigen-expressing plasmid (encoding ovalbumin, OVA), and an adjuvant enzyme-expressing plasmid (encoding STING agonist, CDA) within dendritic cell (DC) membranes. Upon uptake into acidic lysosomes, Mn2+ released from MnO2 triggered the nucleic acids to undergo a morphological change from nanospheres (∼180 nm diameter) to nanofibers (∼1 μm length), resulting in an increase in mechanical strength by about 9-fold and consequently lysosomal membrane disruption. The antigen OVA and adjuvants Mn2+ and CDA in the cytoplasm triggered strong DC activation and antigen-specific CD8+ T cell metalloimmune responses, significantly inhibiting the growth of B16-OVA tumors and inducing long-term immune memory. Altogether, MnO2-OVA-CDA-mem holds potential as a platform for nucleic acid antigen and adjuvant delivery using an in situ self-assembly strategy in a metal-driven, stimulus-responsive, and programmable manner for cancer metalloimmunotherapy.
Collapse
Affiliation(s)
- Jiexin Li
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Ruiqi Yang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Chen Zhang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Jonathan F Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| |
Collapse
|
37
|
Jiang T, Guo C, Zhang Z, Li C, Xiang C, Xiang J, Yang X, Liu Y, Cai L, Gong P, Hu Y, Li C. Biomimetic NIR-II aggregation-induced emission nanoparticles for targeted photothermal therapy of ovarian cancer. J Mater Chem B 2025; 13:4094-4102. [PMID: 40047675 DOI: 10.1039/d4tb02855d] [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: 03/27/2025]
Abstract
Photothermal therapy (PTT) is a cutting-edge technique that harnesses light energy and converts it into heat for precise tumor ablation. By employing photothermal agents to selectively generate heat and target cancer cells, PTT has emerged as a promising cancer treatment strategy. Notably, therapies conducted in the second near-infrared (NIR-II) window exhibit superior therapeutic outcomes, owing to deeper tissue penetration and reduced light scattering. In this study, we developed biomimetic NIR-II aggregation-induced emission (AIE) nanoparticles (2TB-NPs@TM) for high-efficiency NIR-II imaging and targeted phototherapy of ovarian cancer. The core nanoparticle aggregates (2TB-NPs) display strong NIR-II fluorescence and high photothermal conversion efficiency, while the outer tumor cell membrane coating facilitates active targeting and precise recognition of tumor tissues. This design imparts excellent biocompatibility and enhances drug delivery efficiency, leading to potent synergistic therapeutic effects. Our findings open new avenues for advancing targeted, high-performance phototherapy diagnostics in cancer treatment.
Collapse
Affiliation(s)
- Ting Jiang
- Peking University Shenzhen Hospital, No. 1120, Lianhua Road, 518036 Shenzhen, People's Republic of China.
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlei Guo
- Peking University Shenzhen Hospital, No. 1120, Lianhua Road, 518036 Shenzhen, People's Republic of China.
| | - Zhiwei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Li
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunbai Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing Yang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yan Hu
- Peking University Shenzhen Hospital, No. 1120, Lianhua Road, 518036 Shenzhen, People's Republic of China.
| | - Changzhong Li
- Peking University Shenzhen Hospital, No. 1120, Lianhua Road, 518036 Shenzhen, People's Republic of China.
| |
Collapse
|
38
|
Zhang Q, Shen Y, Zhang C, Zhang H, Li X, Yang S, Dai C, Yu X, Lou J, Feng J, Hu C, Lin Z, Li X, Zhou X. Immunoengineered mitochondria for efficient therapy of acute organ injuries via modulation of inflammation and cell repair. SCIENCE ADVANCES 2025; 11:eadj1896. [PMID: 40106554 DOI: 10.1126/sciadv.adj1896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
Acute organ injuries represent a major public health concern, driven by inflammation and mitochondrial dysfunction, leading to cell damage and organ failure. In this study, we engineered neutrophil membrane-fused mitochondria (nMITO), which combine the injury-targeting and anti-inflammatory properties of neutrophil membrane proteins with the cell repairing function of mitochondria. nMITO effectively blocked inflammatory cascades and restored mitochondrial function, targeting both key mechanisms in acute organ injuries. In addition, nMITO selectively targeted damaged endothelial cells via β-integrins and were delivered to injured tissues through tunneling nanotubes, enhancing their regulatory effects on inflammation and cell damage. In mouse models of acute myocardial injury, liver injury, and pancreatitis, nMITO notably reduced inflammatory responses and repaired tissue damage. These findings suggest that nMITO is a promising therapeutic strategy for managing acute organ injuries.
Collapse
Affiliation(s)
- Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, PR China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Chengyuan Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
| | - Hanyi Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xuemei Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Shengqian Yang
- Institute of Materia Medica College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Chen Dai
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xiuyan Yu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Jinwei Feng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Chenglu Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Zhihua Lin
- Chongqing University of Chinese Medicine, Chongqing 402760, PR China
| | - Xiaohui Li
- Institute of Materia Medica College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
- School of Pharmaceutical Sciences & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, PR China
| |
Collapse
|
39
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
40
|
Zhao YY, Meng QF, Cui H, Liu W, Chen Y, Lan Z, Chen H, Ma DD, Rao L, Yu GT. Injectable cellular vesicle-based bone meal for inflammatory bone defect repair through restoring immune homeostasis. Theranostics 2025; 15:4465-4480. [PMID: 40225579 PMCID: PMC11984383 DOI: 10.7150/thno.110795] [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: 01/20/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Immune homeostasis microenvironment of bone regeneration is especially important for inflammatory-derived bone defect repair. The two key influencing factors for achieving ideal bone regeneration are the balance between inflammatory cells represented by T cells and anti-inflammatory cells represented by MDSCs, and the dynamic balance between osteoblasts and osteoclasts. Methods: Herein, an injectable thermosensitive bone meal was designed with Pluronic F127 hydrogel loading myeloid-derived suppressive cells (MDSCs) membrane vesicles coated nano-hydroxyapatite (F127/nHA/MDSCs-MV, abbreviated as F127/nHAM) for periodontitis-derived bone defect repair. Results: The proteomics revealed F127/nHAM were able to catalyze the production of adenosine from ATP depend on CD73 and CD39. In vitro and in vivo assays further showed that F127/nHAM inhibited the proliferation and function of T cells by component MDSCs-MV, exerting an anti-inflammatory role. Subsequently, the RNA-sequencing and confirmation experiments revealed that F127/nHAM inhibiting the differentiation of macrophages into osteoclasts through down-regulating the secretion of CCL2 and CCL5. In the periodontal bone defect rat model, the results of micro-CT and histological staining demonstrated that F127/nHAM had an outstanding anti-inflammatory and bone regeneration promoting properties, restoring immune homeostasis. Conclusion: This biomimetic and multifunctional bone meal opens new avenues for inflammatory-derived bone defect repair and future clinical application.
Collapse
Affiliation(s)
- Yu-Yue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Qian-Fang Meng
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yan Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Zhou Lan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Hao Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Dan-Dan Ma
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Lang Rao
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Guang-Tao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
41
|
Migliavacca M, Cedrún-Morales M, Ceballos M, Soprano E, Polo E, Pelaz B, Pino PD. Engineered cell membrane-cloaked metal-organic framework nanocrystals for intracellular cargo delivery. J Colloid Interface Sci 2025; 682:31-40. [PMID: 39612761 DOI: 10.1016/j.jcis.2024.11.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/07/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
This investigation demonstrates the development and functionality of cell membrane-cloaked UiO-67 nanosized metal-organic frameworks (NMOFs), which are engineered for precise intracellular delivery of encapsulated cargoes. Utilizing the robust and porous nature of UiO-67, we enveloped these NMOFs with fusogenic cell membrane-derived nanovesicles (FCSMs) sourced from adenocarcinomic human alveolar basal epithelial (A549) cells. This biomimetic coating enhances biocompatibility and leverages the homotypic targeting capabilities of the cell-derived coatings, facilitating direct cytoplasmic delivery and avoiding endolysosomal entrapment. Our nanoplatform exhibited high efficiency in loading and releasing two model cargoes: cresyl violet (CV), a staining agent, and carboplatin (CbPt), a chemotherapeutic agent. This approach demonstrated substantial potential in drug delivery in both 2D cell cultures and 3D spheroids. This study underscores the enhanced cellular uptake facilitated by the engineered biomimetic shell and highlights the system's capacity for sustained release, offering a promising strategy for targeted drug delivery.
Collapse
Affiliation(s)
- Martina Migliavacca
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Manuela Cedrún-Morales
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Manuel Ceballos
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Enrica Soprano
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica y Biología Molecular, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain.
| |
Collapse
|
42
|
Dai J, Hu Y, Liu W, Liu H, Wang S, Xia F, Lou X. Cell-Sensing Analogue Nanopore for Rapid Detection of Protein-Related Targets. Angew Chem Int Ed Engl 2025; 64:e202421721. [PMID: 39592429 DOI: 10.1002/anie.202421721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024]
Abstract
Nanopores offer significant advantages in biosensing. Conventional nanopore sensors require probe modification within the pore, and there are two major obstacles: inhomogeneity of probe modification within the pore, and distortion of the detection signal due to the uncontrollable dynamics of the target within the pore. Here, we constructed a cell-sensing analogue nanopore (CeSa-nanopore), by coating the outer surface of the nanopore with cell membrane. The inhomogeneity of probe modification and the uncontrollable kinetics of target-probe binding were also addressed. Specific cells are selected to prepare CeSa-nanopore, for example, cells with high expression of angiotensin-converting enzyme 2 (ACE2) are used to achieve the detection of SARS-CoV-2. When SARS-CoV-2 binds to CeSa-nanopore the surface potential changes, causing a change in the ionic current, thus enabling its detection with a detection rate of 100 %. In addition, the detection of different proteins, such as follicle-stimulating hormone (FSH), can be achieved by changing the cell membrane coating. The identification of cancer cells in ascites can also be achieved by utilizing homologous targeting between cancer cells. Importantly, the use of CeSa-nanopores for the detection of these targets eliminates the need for pre-processing and significantly reduces detection time.
Collapse
Affiliation(s)
- Jun Dai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuxin Hu
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Liu
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| |
Collapse
|
43
|
Wang X, Fang H, Hu W, Feng Y, Zhou Z, Hu M, Jiang D, Zhang Y, Lan X. Oxygen-delivery nanoparticles enhanced immunotherapy efficacy monitored by granzyme B PET imaging in malignant tumors. J Nanobiotechnology 2025; 23:186. [PMID: 40050894 PMCID: PMC11887188 DOI: 10.1186/s12951-025-03257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
Limited treatment response and inadequate monitoring methods stand firmly before successful immunotherapy. Recruiting and activating immune cells in the hypoxic tumor microenvironment is the key to reversing immune suppression and improving immunotherapy efficacy. In this study, biomimetic oxygen-delivering nanoparticles (CmPF) are engineered for homologous targeting and hypoxia alleviation within the tumor environment. CmPF targets the tumor microenvironment and delivers oxygen to reduce hypoxia, thereby promoting immune cell activity at the tumor site. In addition, granzyme B-targeted positron emission tomography (PET) imaging is employed to monitor immune cell activity changes in response to immunotherapy efficacy in vivo. The combination of CmPF with carboplatin and PD-1 inhibitors significantly suppresses tumor growth by 2.4-fold, exhibiting the potential of CmPF to enhance the efficacy of immunotherapy. Immunohistochemistry further confirms increased expression of key immune markers, highlighting the reprogramming of the tumor microenvironment. This study demonstrates that hypoxia alleviation enhances tumor immunotherapy efficacy and introduces a non-invasive PET imaging method for dynamic, real-time assessment of therapeutic response.
Collapse
Affiliation(s)
- Xingyi Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Wenzhu Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yuan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Zhangyongxue Zhou
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Mengyan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
44
|
Wu J, Cheng Y, Qian K, Yang P, Zhou L, Xu M, Sheng D, Wang T, Li Y, Yang X, Wei Y, Zhang Q. siRNA-Encapsulated Biomimetic Liposomes Effectively Inhibit Tumor Cells' Hexosamine Biosynthesis Pathway for Attenuating Hyaluronan Barriers to Pancreatic Cancer Chemotherapy. ACS NANO 2025; 19:7928-7947. [PMID: 39978787 DOI: 10.1021/acsnano.4c14969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses significant therapeutic challenges due to excessive hyaluronic acid (HA) accumulation, which impedes drug delivery. Here, we present a targeted approach to reduce HA production by specifically silencing glutamine-fructose-6-phosphate aminotransferase 1 (GFAT1), a key enzyme of the hexosamine biosynthesis pathway (HBP) in pancreatic cancer cells. An engineered liposomal system for siGFAT1 delivery, PMLip@siGFAT1, characterized by macrophage membrane camouflage, LFC131 peptide-mediated targeting, and calcium phosphate (CaP) as the core, was designed to ensure prolonged circulation, enhanced inflamed vascular endothelial penetration, and subsequent effective tumor cell uptake and endosomal escape. Consequently, PMLip@siGFAT1 markedly downregulated the HA level in the PDAC microenvironment, decompressing the tumor vasculature and weakening the stromal barrier, which in turn improved the permeability of chemotherapeutics. In combination with Doxil, PMLip@siGFAT1 demonstrated potent antitumor efficacy with minimal systemic toxicity. Importantly, unlike PEGPH20 (hyaluronidase), PMLip@siGFAT1 reduced tumor invasiveness, while preserving skeletal muscle integrity. These findings highlight that PMLip@siGFAT1 holds great potential to revitalize HA downregulation strategies in pancreatic cancer for enhanced drug delivery and efficacy.
Collapse
Affiliation(s)
- Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
- Institute of Traditional Chinese Medicine, & Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
45
|
Qiu P, Wen M, Zhuang Z, Niu S, Tao C, Yu N, Chen Z. Biomimetic polymeric nanoreactors with photooxidation-initiated therapies and reinvigoration of antigen-dependent and antigen-free immunity. Biomaterials 2025; 314:122884. [PMID: 39405823 DOI: 10.1016/j.biomaterials.2024.122884] [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: 07/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/10/2024]
Abstract
Immune cell-mediated anticancer modalities usually suffer from immune cell exhaustion and limited efficacy in solid tumors. Herein, the oxygen-carrying biomimetic nanoreactors (BNR2(O2)) have been developed with photooxidation-driven therapies and antigen-dependent/antigen-free immune reinvigoration against xenograft tumors. The BNR2(O2) composes polymeric nanoreactors camouflaged with cancer cell membranes can efficiently target homotypic tumors. It continuously releases O2 to boost intracellular reactive oxygen species (ROS) to oxide diselenide bonds, which controllably releases seleninic acids and anti-folate Pemetrexed compared to hydrogen peroxide and glutathione incubation. The O2-rich microenvironment sensitizes Pemetrexed and blocks programmed cell-death ligand 1 (PD-L1) to reverse T cell immunosuppression. The ROS and Pemetrexed upregulate pro-apoptosis proteins and inhibit folate-related enzymes, which cause significant apoptosis and immunogenic cell death to stimulate dendritic cell maturation for improved secretion of cytokines, expanding antigen-dependent T cell immunity. Furthermore, by regulating the release of seleninic acids, the checkpoint receptor human leukocyte antigen E of tumor cells can be blocked to reinvigorate antigen-free natural killer cell immunity. This work offers an advanced antitumor strategy by bridging biomimetic nanoreactors and modulation of multiple immune cells.
Collapse
Affiliation(s)
- Pu Qiu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Mei Wen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Zixuan Zhuang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Shining Niu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Cheng Tao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Nuo Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| | - Zhigang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
46
|
Sari BR, Yesilot S, Ozmen O, Aydin Acar C. Superior In Vivo Wound-Healing Activity of Biosynthesized Silver Nanoparticles with Nepeta cataria (Catnip) on Excision Wound Model in Rat. Biol Trace Elem Res 2025; 203:1502-1517. [PMID: 38865065 PMCID: PMC11872767 DOI: 10.1007/s12011-024-04268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Silver nanoparticles were biosynthesized with Nepeta cataria plant extract. It was determined that the synthesized Nc-AgNPs gave a strong absorbance peak at 438 nm wavelength in the UV-vis spectrophotometer. SEM and TEM analyses of Nc-AgNPs showed that the synthesized nanoparticles had a spherical morphology. Based on XRD analysis, the average crystallite size of Nc-AgNPs was calculated at 15.74 nm. At the same time, EDS spectrum analysis exhibited dominant emission energy at 3 keV, indicative of Nc-AgNPs. Nc-AgNPs showed an inhibition zone of 12 nm in gram-negative Escherichia coli, 10 nm in gram-positive Enterococcus faecalis, and 11 nm in Staphylococcus aureus. Nc-AgNPs showed high antioxidant properties, with 63% at 5000 μg/mL. The wound-healing properties of Nc-AgNPs were evaluated in vivo in wound models created in a total of 20 Wistar albino male rats, divided into four groups. After 10 days of treatment, the highest wound closure rate was seen in the Nc-AgNP + Vaseline (Group IV) treatment group, at 94%. It was observed that Nc-AgNP + Vaseline nanoformulation significantly increased wound healing, similar to Silverdin®, and Vaseline alone supported healing but did not result in complete closure. Histopathological examination revealed an increase in mature Type 1 collagen in Group IV and positive control (Group II), with better collagen maturation in vehicle control (Group III) compared to negative control (Group I). Immunohistochemical analysis showed complete epithelialization in Group IV and Group II, with distinct cytokeratin expressions, while Group III exhibited mild expressions.
Collapse
Affiliation(s)
- Berfin Rumeysa Sari
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Sukriye Yesilot
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
- Department of Nursing, Bucak School of Health, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ozlem Ozmen
- Veterinary Faculty, Department of Pathology, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Cigdem Aydin Acar
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey.
- Department of Nursing, Bucak School of Health, Burdur Mehmet Akif Ersoy University, Burdur, Turkey.
| |
Collapse
|
47
|
Yang W, Liu H, Liu X, Cao Y, Wang X, Wang X, Cao W, Guo C, Chen D. Biomimetic fucoidan nanoparticles with regulation of macrophage polarization for targeted therapy of acute lung injury. Carbohydr Polym 2025; 351:122967. [PMID: 39778982 DOI: 10.1016/j.carbpol.2024.122967] [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: 07/15/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 01/11/2025]
Abstract
Acute lung injury (ALI) is a complex acute respiratory illness with a high mortality rate. Reactive oxygen species (ROS) play a pivotal role in ALI, inducing cellular damage, inflammation, and oxidative stress, thereby exacerbating the severity of the injury. In this study, inspired by the "subtractive" strategy, we developed a fucoidan-based macrophage membrane bio-nanosystem, abbreviated as MF@CB, designed as an anti-inflammatory and antioxidant agent to alleviate lipopolysaccharide (LPS)-induced inflammation in ALI. MF@CB coated with macrophage membrane for effective targeting and accumulation in ALI lesions. In addition, MF@CB activates Nrf2 transcriptional activity in macrophages, inhibiting ROS synthesis at its origin while effectively removing ROS already present in the ALI. This dual-pronged approach demonstrates robust antioxidant properties and restores the macrophage antioxidant defense barrier. In the LPS-induced ALI mouse model, MF@CB significantly mitigated lung inflammatory damage by modulating lung macrophage polarization and inhibiting the over-secretion of pro-inflammatory cytokines by activated immune cells. More importantly, unlike most surface modification strategies because it remove the molecule, this approach is easier to apply and potentially safer and may provide useful insights into the development of more effective therapeutic strategies for ALI.
Collapse
Affiliation(s)
- Weili Yang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Hui Liu
- Department of Pharmacy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, PR China
| | - Xiaowei Liu
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Yuxin Cao
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Xuemei Wang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Xinxin Wang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Wenwen Cao
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, 5# Yushan 10 Road, Qingdao 266003, PR China.
| | - Daquan Chen
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China.
| |
Collapse
|
48
|
Ge Y, Zhou Q, Pan F, Wang R. Utilizing Nanoparticles to Overcome Anti-PD-1/PD-L1 Immunotherapy Resistance in Non-Small Cell Lung cancer: A Potential Strategy. Int J Nanomedicine 2025; 20:2371-2394. [PMID: 40027868 PMCID: PMC11871910 DOI: 10.2147/ijn.s505539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, with non-small cell lung cancer (NSCLC) constituting 85% of cases. Immune checkpoint inhibitors (ICIs) represented by anti-programmed cell death protein 1 (PD-1)/ programmed cell death ligand 1 (PD-L1) have emerged as a promising frontier in cancer treatment, effectively extending the survival of patients with NSCLC. However, the efficacy of ICIs exhibits significant variability across diverse patient populations, with a substantial proportion showing poor responsiveness and acquired resistance in those initially responsive to ICIs treatments. With the advancement of nanotechnology, nanoparticles offer unique advantages in tumor immunotherapy, including high permeability and prolonged retention(EPR) effects, enhanced drug delivery and stability, and modulation of the inflammatory tumor microenvironment(TME). This review summarizes the mechanisms of resistance to ICIs in NSCLC, focusing on tumor antigens loss and defective antigen processing and presentation, failure T cell priming, impaired T cell migration and infiltration, immunosuppressive TME, and genetic mutations. Furthermore, we discuss how nanoparticles, through their intrinsic properties such as the EPR effect, active targeting effect, shielding effect, self-regulatory effect, and synergistic effect, can potentiate the efficacy of ICIs and reverse resistance. In conclusion, nanoparticles serve as a robust platform for ICIs-based NSCLC therapy, aiding in overcoming resistance challenges.
Collapse
Affiliation(s)
- Yuli Ge
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Qiong Zhou
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Fan Pan
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Rui Wang
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| |
Collapse
|
49
|
Yang H, Li J, Song C, Li H, Luo Q, Chen M. Emerging Gene Therapy Based on Nanocarriers: A Promising Therapeutic Alternative for Cardiovascular Diseases and a Novel Strategy in Valvular Heart Disease. Int J Mol Sci 2025; 26:1743. [PMID: 40004206 PMCID: PMC11855571 DOI: 10.3390/ijms26041743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular disease remains a leading cause of global mortality, with many unresolved issues in current clinical treatment strategies despite years of extensive research. Due to the great progress in nanotechnology and gene therapy in recent years, the emerging gene therapy based on nanocarriers has provided a promising therapeutic alternative for cardiovascular diseases. This review outlines the status of nanocarriers as vectors in gene therapy for cardiovascular diseases, including coronary heart disease, pulmonary hypertension, hypertension, and valvular heart disease. It discusses challenges and future prospects, aiming to support emerging clinical treatments. This review is the first to summarize gene therapy using nanocarriers for valvular heart disease, highlighting their potential in targeting challenging tissues.
Collapse
Affiliation(s)
- Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
50
|
Bi S, Yang R, Ju H, Liu Y. Dynamic Nanostructure-Based DNA Logic Gates for Cancer Diagnosis and Therapy. Chembiochem 2025; 26:e202400754. [PMID: 39429047 DOI: 10.1002/cbic.202400754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
DNA logic gates with dynamic nanostructures have made a profound impact on cancer diagnosis and treatment. Through programming the dynamic structure changes of DNA nanodevices, precise molecular recognition with signal amplification and smart therapeutic strategies have been reported. This enhances the specificity and sensitivity of cancer theranostics, and improves diagnosis precision and treatment outcomes. This review explores the basic components of dynamic DNA nanostructures and corresponding DNA logic gates, as well as their applications for cancer diagnosis and therapies. The dynamic DNA nanostructures would contribute to cancer early detection and personalized treatment.
Collapse
Affiliation(s)
- Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Ruowen Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|