1
|
Zhu S, Diao S, Liu X, Zhang Z, Liu F, Chen W, Lu X, Luo H, Cheng X, Liao Q, Li Z, Chen J. Biomaterial-based strategies: a new era in spinal cord injury treatment. Neural Regen Res 2025; 20:3476-3500. [PMID: 40095657 PMCID: PMC11974648 DOI: 10.4103/nrr.nrr-d-24-00844] [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: 07/29/2024] [Revised: 09/02/2024] [Accepted: 12/16/2024] [Indexed: 03/19/2025] Open
Abstract
Enhancing neurological recovery and improving the prognosis of spinal cord injury have gained research attention recently. Spinal cord injury is associated with a complex molecular and cellular microenvironment. This complexity has prompted researchers to elucidate the underlying pathophysiological mechanisms and changes and to identify effective treatment strategies. Traditional approaches for spinal cord injury repair include surgery, oral or intravenous medications, and administration of neurotrophic factors; however, the efficacy of these approaches remains inconclusive, and serious adverse reactions continue to be a concern. With advancements in tissue engineering and regenerative medicine, emerging strategies for spinal cord injury repair now involve nanoparticle-based nanodelivery systems, scaffolds, and functional recovery techniques that incorporate biomaterials, bioengineering, stem cell, and growth factors as well as three-dimensional bioprinting. Ideal biomaterial scaffolds should not only provide structural support for neuron migration, adhesion, proliferation, and differentiation but also mimic the mechanical properties of natural spinal cord tissue. Additionally, these scaffolds should facilitate axon growth and neurogenesis by offering adjustable topography and a range of physical and biochemical cues. The three-dimensionally interconnected porous structure and appropriate physicochemical properties enabled by three-dimensional biomimetic printing technology can maximize the potential of biomaterials used for treating spinal cord injury. Therefore, correct selection and application of scaffolds, coupled with successful clinical translation, represent promising clinical objectives to enhance the treatment efficacy for and prognosis of spinal cord injury. This review elucidates the key mechanisms underlying the occurrence of spinal cord injury and regeneration post-injury, including neuroinflammation, oxidative stress, axon regeneration, and angiogenesis. This review also briefly discusses the critical role of nanodelivery systems used for repair and regeneration of injured spinal cord, highlighting the influence of nanoparticles and the factors that affect delivery efficiency. Finally, this review highlights tissue engineering strategies and the application of biomaterial scaffolds for the treatment of spinal cord injury. It discusses various types of scaffolds, their integrations with stem cells or growth factors, and approaches for optimization of scaffold design.
Collapse
Affiliation(s)
- Shihong Zhu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Sijun Diao
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhujun Zhang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Fujun Liu
- Department of Ophthalmology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiyue Lu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huiyang Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Liao
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhongyu Li
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
3
|
Guo L, Fu Z, Li H, Wei R, Guo J, Wang H, Qi J. Smart hydrogel: A new platform for cancer therapy. Adv Colloid Interface Sci 2025; 340:103470. [PMID: 40086017 DOI: 10.1016/j.cis.2025.103470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Cancer is a significant contributor to mortality worldwide, posing a significant threat to human life and health. The unique bioactivity, ability to precisely control drug release, and minimally invasive properties of hydrogels are indispensable attributes that facilitate optimal performance in cancer therapy. However, conventional hydrogels lack the ability to dynamically respond to changes in the surrounding environment, withstand drastic changes in the microenvironment, and trigger drug release on demand. Therefore, this review focuses on smart-responsive hydrogels that are capable of adapting and responding to external stimuli. We comprehensively summarize the raw materials, preparation, and cross-linking mechanisms of smart hydrogels derived from natural and synthetic materials, elucidate the response principles of various smart-responsive hydrogels according to different stimulation sources. Further, we systematically illustrate the important role played by hydrogels in modern cancer therapies within the context of therapeutic principles. Meanwhile, the smart hydrogel that uses machine learning to design precise drug delivery has shown great prospects in cancer therapy. Finally, we present the outlook on future developments and make suggestions for future related work. It is anticipated that this review will promote the practical application of smart hydrogels in cancer therapy and contribute to the advancement of medical treatment.
Collapse
Affiliation(s)
- Li Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ziming Fu
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Haoran Li
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ruibo Wei
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Jing Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Haiwang Wang
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Jian Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Luo Z, Li X, Zhu D, Fu W, Liu Y, Zheng L, Chen P, Gong C, Liu X. Implantable Immunostimulant Microneedle Patch for Post-Surgical Prevention of Cancer Recurrence and Distant Tumor Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22362-22374. [PMID: 40194999 DOI: 10.1021/acsami.5c01155] [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: 04/09/2025]
Abstract
Cancer recurrence after surgical resection remains a grand challenge in achieving long-term eradication. Here, we develop a biocompatible and implantable immunostimulant microneedle patch designed to suppress local tumor recurrence after surgery. The patch, fabricated using methacrylate-modified hyaluronic acid, incorporates 2'3'-cGAMP, a STING agonist, and IL-2, a cytokine approved for clinical cancer immunotherapy that expands T cells. The patch enables controlled release of cGAMP to induce dendritic cell maturation, antitumor macrophage polarization (M1 macrophage), and T cell priming and activation. Simultaneously, localized IL-2 activates CD8+ T cells and recruits immune cells to the tumor microenvironment. When combined with an anti-CTLA-4 antibody, an immune checkpoint blockade, the hybrid microneedle patch significantly reduces Treg cells at the surgery sites, enhancing immune responses and effectively inhibiting the progression of distant tumors in both prophylactic and therapeutic models. Compared with traditional postsurgical chemotherapy and radiotherapy, this patch-mediated immunotherapy demonstrates superior efficacy in mitigating tumor relapse while offering higher biocompatibility. Our findings suggest that this immunotherapeutic patch has potential as a translational tool to prevent cancer recurrence in patients with resectable tumors.
Collapse
Affiliation(s)
- Zichao Luo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031 China
- NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031 China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200031 China
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Xinchao Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dandan Zhu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Wangxian Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxia Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Lewen Zheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
- Skin Research Institute of Singapore, 308232 Singapore
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921 Singapore
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaogang Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 138634, Singapore
| |
Collapse
|
5
|
Meng Q, Ding B, Ma P, Lin J. Inorganic Nanobiomaterials Boost Tumor Immunotherapy: Strategies and Applications. Acc Chem Res 2025; 58:1210-1223. [PMID: 40179239 DOI: 10.1021/acs.accounts.4c00843] [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: 04/05/2025]
Abstract
ConspectusTumor immunotherapy, as a new antitumor method to fight cancer by activating or enhancing the body's own immune system, has been extensively studied and applied in clinical practice. However, as an extremely complex system, tumor heterogeneity and complex immunosuppressive tumor microenvironment (TME) lead to poor immune response rate or secondary drug resistance. The advent of nanotechnology has ushered in a new era for immunotherapy. In particular, inorganic nanomaterials, with their unique physicochemical properties and excellent biocompatibility, are becoming an important tool for enhancing immunotherapy. Inorganic nanomaterials can be used as carriers for immune agents, improving drug delivery efficiency and thereby reducing systemic immunotoxicity and enhancing immune responses. Inorganic nanomaterials also trigger tumor immunogenic cell death (ICD), stimulate antitumor immune responses, and alleviate immunosuppressive TME by increasing oxygen levels, modulating metabolic pathways, and altering the secretion of immunosuppressive cytokines. The synergistic integration of inorganic nanomaterials with immunotherapy adeptly navigates around the constraints of conventional treatments, reducing side effects while concurrently augmenting therapeutic efficacy. In this review, we summarize our recent efforts in the design and synthesis of inorganic nanobiomaterials to enhance the efficacy of tumor immunotherapy. These nanomaterials achieve the desired immune efficacy mainly through four strategies, including inducing ICD, developing tumor nanovaccines, activating pyroptosis, and regulating tumor metabolism, providing beneficial implications for tumor immunotherapy. For one thing, due to the deficiency of ICD effect in single therapy, we mainly developed nanocatalysts that integrate multiple therapeutic functions to play a catalytic role in TME, converting tumor substances or metabolites into therapeutic products in situ, and further enhancing ICD. For another, in order to solve the problems of low antigen loading and therapeutic efficiency of existing adjuvants, several novel multifunctional nanoadjuvants were prepared, which combine high antigen loading and multimode therapeutic function in one, and achieve efficient immune activation. Moreover, to attain strong inflammatory responses and immunogenicity, we engineer pyroptosis adjuvants that selectively induce tumor cell pyroptosis by enhancing intracellular oxidative stress or ion overload. Finally, to reverse the immunosuppressive microenvironment, we developed nanoplatforms that target tumor metabolism, altering the levels of nutrients and metabolites in tumor such as glucose, lactic acid, citric acid, and tryptophan to effectively alter the TME, thereby activating and enhancing the body's immune response. The implementation of these strategies not only improves the therapeutic effect but also reduces the side effects and provides valuable insights and references for the development of novel nanomaterials to assist immunotherapy.
Collapse
Affiliation(s)
- Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
6
|
Ake B, Yang H, Yang H, Liu H, Gui X, Liu T, Chen J, Liu J, Zhou W, Qu B, Zeng Z, Zhou C. Ultrasound-responsive smart biomaterials for bone tissue engineering. J Mater Chem B 2025; 13:4527-4543. [PMID: 40111085 DOI: 10.1039/d5tb00109a] [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: 03/22/2025]
Abstract
Bone defects resulting from trauma, tumors, or other injuries significantly impact human health and quality of life. However, current treatments for bone defects are constrained by donor shortages and immune rejection. Bone tissue engineering has partially alleviated the limitations of traditional bone repair methods. The development of smart biomaterials that can respond to external stimuli to modulate the biofunctions has become a prominent area of research. Ultrasound technology is regarded as an optimal "remote controller" and "trigger" for bone repair biomaterials. This review reports the comprehensive and systematic overview of ultrasound-responsive bone repair smart biomaterials. It presents the fundamental theories of bone repair, the definition of ultrasound, and its applications. Furthermore, the review summarizes the ultrasound effect mechanisms of biomaterials and their roles in bone repair, including detailed studies on anti-inflammation, immunomodulation, and cell therapy. Finally, the advantages of ultrasound-responsive smart biomaterials and their future prospects in this field are discussed.
Collapse
Affiliation(s)
- Bicheng Ake
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hongsheng Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hao Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hao Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| | - Taoyu Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| | - Jie Chen
- Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jia Liu
- The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Wenzheng Zhou
- The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Bo Qu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Zhimou Zeng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| |
Collapse
|
7
|
Jin S, Chen Y, Li F, Yan P, Guo G, Xu G, Song W, Zhong W. Photodynamic and Photothermal Co-Induced Efficient Anti-Tumor Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20833-20848. [PMID: 40152855 DOI: 10.1021/acsami.5c00184] [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: 03/29/2025]
Abstract
Currently, immunotherapy based on photothermal and the application of photodynamic therapy in anti-tumor treatment is showing great potential. Its uniqueness lies in the critical role of small molecule immunomodulators in promoting effective immune responses against tumors, and the use of laser-activated biophysical mechanisms to precisely trigger the swift demise of cancer cells, avoiding damage to surrounding normal tissues. However, the use of photodynamic therapy (PDT) alone is hampered by the tumors' hypoxic environment, resulting in poor antitumor effects, while photothermal therapy (PTT) alone cannot arouse enough antigen presentation. It is of great significance to design photosensitizers (PSs) that possess both PDT and PTT effects. Herein, a series of PSs with both PDT and PTT efficacy are reported, ultimately selecting Cy7-Naph as the star molecule due to its best overall phototherapeutic effect. Upon reactive oxygen species (ROS) production and thermogenesis in tumor cells, Cy7-Naph induced significant apoptosis and eventually boosted the release of damage-associated molecular patterns (DAMPs) under near-infrared (NIR) light irradiation. By combining Cy7-Naph with the Toll-like receptor agonist Resiquimod (R848), a synergistic treatment for bilateral tumor-bearing mice is achieved. This combination promotes dendritic cell (DC) maturation and increases the infiltration of cytotoxic T lymphocytes (CTLs), leading to significant inhibition of both primary and distant tumors.
Collapse
Affiliation(s)
- Shanshan Jin
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Yongkang Chen
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Fahui Li
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Ping Yan
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Guanhong Guo
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Guangzhao Xu
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Weiguo Song
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Wenda Zhong
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| |
Collapse
|
8
|
Barik P, Mondal S. Immunomodulatory effects of metal nanoparticles: current trends and future prospects. NANOSCALE 2025. [PMID: 40202489 DOI: 10.1039/d5nr01030f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The advent of nanotechnology has steered into a new era of medical advancements, with metal nanoparticles (MNPs) emerging as potent agents for precise regulation of the immune system. This review provides a comprehensive overview of the immunomodulatory roles of MNPs, including gold, silver, and metal oxide nanoparticles, in regulating innate and adaptive immunity. Additionally, we discuss the immunological effects of metal ions and metal complexes, offering a comparative analysis with nanoparticulate systems. We analyse cutting-edge strategies utilising MNPs to optimise vaccine efficacy, achieve targeted delivery to immune cells, and orchestrate inflammatory responses. Additionally, we discuss the therapeutic potential of MNPs in combating autoimmune diseases, cancers, and infectious agents, which is evaluated within the framework of precision medicine. Furthermore, we critically assess challenges such as biocompatibility, potential toxicity, and regulatory hurdles. Finally, we propose future directions for integrating MNPs with advanced delivery systems and other nanomaterials to propel the frontiers of immunotherapy. This review aims to provide a foundational understanding of MNP-mediated immunomodulation, inspiring further research and development in this burgeoning field.
Collapse
Affiliation(s)
- Puspendu Barik
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
- Department of Physics, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
| | - Samiran Mondal
- Department of Chemistry, Rammohan College (University of Calcutta), 102/1-Raja Rammohan Sarani, Kolkata 700009, West Bengal, India.
| |
Collapse
|
9
|
Ou M, Cao J, Luo R, Zhu B, Miao R, Yu L, Wang X, Li W, Fu Y, Zhang J, Zhang F, Wang Q, Mei L. Drug-loaded microneedle patches containing regulatory T cell-derived exosomes for psoriasis treatment. Acta Biomater 2025:S1742-7061(25)00256-9. [PMID: 40210183 DOI: 10.1016/j.actbio.2025.04.015] [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: 02/09/2025] [Revised: 03/17/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by epidermal hyperplasia, skin inflammation, and immune dysregulation. These factors contribute to the persistent progression of the disease. While addressing excessive keratinocyte proliferation or inhibiting inflammation may provide temporary therapeutic relief, unresolved immune dysregulation often exacerbates the condition. Therefore, comprehensive treatments that alleviate skin symptoms and regulate immune tolerance are urgently required. An ideal treatment would target multiple factors, including keratinocyte proliferation, inflammation, and immune tolerance, while minimizing systemic side effects. In this study, we developed a dissolvable hyaluronic acid microneedle patch containing regulatory T cell (Treg) exosomes loaded with dimethyl fumarate (DMF) (rExo@DMF MNs). DMF acts as an inhibitor of keratinocyte proliferation and an anti-inflammatory agent through NF-κB suppression and Nrf2 activation, inhibiting the production of pro-inflammatory cytokines and the activation of inflammatory cells. Delivering DMF via Treg exosomes enhances its retention at the lesion site. This system inhibits keratinocyte proliferation and migration, reduces pro-inflammatory cytokine release, and alleviates epidermal hyperplasia and inflammation in an imiquimod-induced psoriasis mouse model. Additionally, Treg exosomes modulate immune responses to promote tolerance. rExo@DMF MNs demonstrate immunomodulatory effects by inhibiting T helper 17 (Th17) cells and inducing regulatory immune cells such as Tregs and tolerogenic dendritic cells (tDCs) differentiation. rExo@DMF MNs alleviate skin symptoms and regulate immune cells in the skin, spleen, and lymph nodes, demonstrating both local and systemic immunoregulation with promising therapeutic potential for psoriasis. STATEMENT OF SIGNIFICANCE: Novel therapies are urgently needed to alleviate skin symptoms and regulate immunity, as current psoriasis treatments focus on symptom relief while neglecting the underlying immune dysfunction, resulting in limited efficacy. Moreover, systemic immunosuppression often leads to severe side effects. This study introduces a hybrid microneedle system (rExo@DMF MNs) that alleviates psoriasis symptoms and modulates immune responses locally and systemically. In addition, rExo@DMF MNs penetrate hyperkeratotic skin, ensuring targeted rExo@DMF release while minimizing systemic exposure and side effects. All components of the system, including hyaluronic acid (a key component of the skin matrix), regulatory T cell-derived exosomes, and DMF (a clinically validated drug), exhibit biocompatibility. This comprehensive approach addresses multiple pathogenic factors, promising an effective and safe psoriasis treatment.
Collapse
Affiliation(s)
- Meitong Ou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jiahui Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Ran Luo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Baisong Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Rourou Miao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Liu Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Wen Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Yiqiu Fu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jinxie Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Qiangsong Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
10
|
He L, Ren W, Cheng W, Chen J, Lai J, Wu Y, Wu Z, Bao D, Wei Y, Piao JG. Arsenene-Vanadene nanodots co-activate Apoptosis/Ferroptosis for enhanced chemo-immunotherapy. Acta Biomater 2025; 196:453-470. [PMID: 40032219 DOI: 10.1016/j.actbio.2025.02.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Triple-Negative Breast Cancer (TNBC) represents a highly aggressive subtype of breast cancer with an unfavorable prognosis, characterized by minimal immune infiltration and pronounced immune suppression, resulting in a limited response to immunotherapy. In this study, a multifunctional Arsenene-Vanadene nanodot (AsV) drug delivery system is introduced, which responds to the tumor microenvironment by releasing arsenic and vanadium. Arsenic undergoes oxidation to generate highly toxic trivalent arsenic, which induces apoptosis in tumor cells while utilizing apoptotic cell debris to transiently activate the immune system. Additionally, arsenic binds to cysteine, indirectly facilitating ferroptosis. Concurrently, vanadium's redox cycling properties are harnessed to trigger a Fenton-like reaction, promoting lipid peroxidation. Furthermore, ferroptosis is enhanced through the depletion of glutathione and inactivation of glutathione peroxidase 4 (GPX4), leading to the release of damage-associated molecular patterns and thereby amplifying the anti-tumor immune response. This study represents the first instance of integrating arsenene's apoptosis-inducing properties with vanadium's ferroptosis-enhancing effects, providing a synergistic approach to improving the immunotherapeutic response and offering a potential strategy for enhancing TNBC prognosis. STATEMENT OF SIGNIFICANCE: Triple-negative breast cancer (TNBC) exhibits resistance to immunotherapy due to its highly immunosuppressive tumor microenvironment. In this study, tumour-responsive Arsenene-Vanadene nanodots (AsV) were developed to induce a synergistic effect by triggering apoptosis and ferroptosis through microenvironment-specific mechanisms. The arsenic component generates cytotoxic trivalent arsenic, promoting apoptosis while binding to cysteine, thereby reducing GSH synthesis. Simultaneously, vanadium initiates lipid peroxidation through Fenton-like reactions and disruption of the glutathione/GPX4 axis, further amplifying ferroptotic cell death. This dual-action system transforms tumor cell debris into immune-stimulating signals while circumventing conventional immunotherapy limitations. As the first strategy integrating arsenic-induced apoptosis with vanadium-enhanced ferroptosis, this approach provides a mechanistic framework to overcome TNBC immunosuppression through coordinated cell death pathways, demonstrating potential for precision nanomedicine applications.
Collapse
Affiliation(s)
- Li He
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - WeiYe Ren
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - WeiYi Cheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - JingQuan Chen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Jianjun Lai
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China
| | - Yajun Wu
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China
| | - Zhibing Wu
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China; Department of Radiation Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China.
| | - Dandan Bao
- Department of Dermatology & Cosmetology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, PR China.
| | - Yinghui Wei
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China; Jinhua Academy of Zhejiang Chinese Medical University.
| | - Ji-Gang Piao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China.
| |
Collapse
|
11
|
Cai XY, Zheng CX, Guo H, Fan SY, Huang XY, Chen J, Liu JX, Gao YR, Liu AQ, Liu JN, Zhang XH, Ma C, Wang H, Fu F, Peng P, Xu HK, Sui BD, Xuan K, Jin Y. Inflammation-triggered Gli1 + stem cells engage with extracellular vesicles to prime aberrant neutrophils to exacerbate periodontal immunopathology. Cell Mol Immunol 2025; 22:371-389. [PMID: 40016585 PMCID: PMC11955562 DOI: 10.1038/s41423-025-01271-0] [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: 09/05/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
Periodontitis is a prevalent and progressive detrimental disease characterized by chronic inflammation, and the immunopathological mechanisms are not yet fully understood. Mesenchymal stem cells (MSCs) play crucial roles as immunoregulators and maintain tissue homeostasis and regeneration, but their in vivo function in immunopathology and periodontal tissue deterioration is still unclear. Here, we utilized multiple transgenic mouse models to specifically mark, ablate and modulate Gli1+ cells, a critical and representative subset of MSCs in the periodontium, to explore their specific role in periodontal immunopathology. We revealed that Gli1+ cells, upon challenge with an inflammatory microenvironment, significantly induce rapid trafficking and aberrant activation of neutrophils, thus exacerbating alveolar bone destruction. Mechanistically, extracellular vesicles (EVs) released by Gli1+ cells act as crucial immune regulators in periodontal tissue, mediating the recruitment and activation of neutrophils through increased neutrophil generation of reactive oxygen species and stimulation of nuclear factor kappa-B signaling. Furthermore, we discovered that CXC motif chemokine ligand 1 (CXCL1) is exposed on the surface of EVs derived from inflammation-challenged Gli1+ cells to prime aberrant neutrophils via the CXCL1-CXC motif chemokine receptor 2 (CXCR2) axis. Importantly, specific inhibition of EV release from Gli1+ cells or pharmacological therapy with GANT61 ameliorates periodontal inflammation and alveolar bone loss. Collectively, our findings identify previously unrecognized roles of Gli1+ cells in orchestrating infiltration and promoting aberrant activation of neutrophils under inflammation, which provides pathological insights and potential therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Xin-Yue Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Si-Yuan Fan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiao-Yao Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Oral Implantology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jie-Xi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yu-Ru Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - An-Qi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jia-Ning Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiao-Hui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chao Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Fei Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Peng Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao-Kun Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Kun Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi key Laboratory of Stomatology, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Yan Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
12
|
Wang F, Lou J, Lou X, Wu F, Gao X, Yao X, Wan J, Duan X, Deng W, Ma L, Zhang L, He G, Wang M, Ni C, Lei N, Qin Z. A Spleen-Targeted Tolerogenic mRNA-LNPs Vaccine for the Treatment of Experimental Asthma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412543. [PMID: 39921498 PMCID: PMC11967843 DOI: 10.1002/advs.202412543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/27/2024] [Indexed: 02/10/2025]
Abstract
Lipid nanoparticles (LNPs)-based mRNA vaccines have witnessed their great advantages in the fight against infectious diseases. However, the pro-inflammatory properties of mRNA-LNPs vaccines may hinder the induction of antigen-specific tolerogenic immune responses. Here, it is demonstrated that stearic acid-doped LNPs co-loaded with nucleoside-modified mRNA and celastrol selectively target spleen, convert their adjuvanticity and promote a tolerogenic rather than immunogenic DCs phenotype. Furthermore, the tolerogenic mRNA vaccine also invokes the generation of antigen-specific regulatory T cells (Tregs) in the spleen and migration of the induced Tregs to the lung. In a mouse model of allergic asthma, immunization with the tolerogenic mRNA vaccine significantly alleviated symptom induction, reducing eosinophilic granulocyte accumulation and mucus secretion. In conclusion, this spleen-targeted mRNA-LNPs vaccine platform induces tolerogenic immune responses, offering promise for the development of therapeutics against allergic asthma and other conditions requiring immune tolerance modulation.
Collapse
Affiliation(s)
- Fazhan Wang
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Jia Lou
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
- Department of Pain and RehabilitationSecond Affiliated HospitalArmy Medical UniversityChongqing400038China
| | - Xiaohan Lou
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Fang Wu
- Department of Microbiology and ImmunologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenan450001China
| | - Xiaoke Gao
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Xiaohan Yao
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Jiajia Wan
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Xixi Duan
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Wenjing Deng
- Department of Neuro‐Intensive Care UnitThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Lixia Ma
- Department of Neuro‐Intensive Care UnitThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Lijing Zhang
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Guangjie He
- Xinxiang Key Laboratory of Forensic Science EvidenceSchool of Forensic MedicineXinxiang Medical UniversityXinxiangHenan453003China
| | - Ming Wang
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Chen Ni
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| | - Ningjing Lei
- Department of Microbiology and ImmunologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenan450001China
| | - Zhihai Qin
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450052China
| |
Collapse
|
13
|
Zhou M, Zhou C, Geng H, Huang Z, Lin Z, Wang Y, Zhu Y, Shi J, Tan J, Guo L, Zhao Y, Zhang Y, Peng Q, Yu H, Dai W, Lv H, Lin Z. EGCG-enabled Deep Tumor Penetration of Phosphatase and Acidity Dual-responsive Nanotherapeutics for Combinatory Therapy of Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406245. [PMID: 39558766 DOI: 10.1002/smll.202406245] [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: 07/23/2024] [Revised: 10/23/2024] [Indexed: 11/20/2024]
Abstract
The presence of dense collagen fibers is a typical characteristic of triple-negative breast cancer (TNBC). Although these fibers hinder drug penetration and reduce treatment efficacy, the depletion of the collagen matrix is associated with tumor metastasis. To address this issue, epigallocatechin-3-gallate (EGCG) is first exploited for disrupting the dense collagenous stroma and alleviate fibrosis by specifically blocking the TGF-β/Smad pathway in fibroblasts and tumor cells when intraperitoneally administrated in TNBC tumor-bearing mice. A methotrexate (MTX)-loaded dual phosphate- and pH-responsive nanodrug (pHA@MOF-Au/MTX) is next engineered by integrating Fe-based metal-organic frameworks and gold nanoparticles for improved chemo/chemodynamic therapy of TNBC. Surface modification with pH (low)-insertion peptide substantially enhanced the binding of the nanodrug to 4T1 cells owing to tumor stroma remodeling by EGCG. High-concentration EGCG inhibited glutathione peroxidase by regulating mitochondrial glutamine metabolism, thus facilitating tumor cell ferroptosis. Furthermore, sequential EGCG and pHA@MOF-Au/MTX treatment showed remarkable anti-tumor effects in a mouse model of TNBC, with a tumor growth inhibition rate of 79.9%, and a pulmonary metastasis rate of 96.8%. Altogether, the combination strategy developed in this study can improve the efficacy of chemo/chemodynamic therapy in TNBC and represents an innovative application of EGCG.
Collapse
Affiliation(s)
- Mengxue Zhou
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Chuang Zhou
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, P. R. China
| | - Huan Geng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, P. R. China
| | - Zhiwei Huang
- MOE, Frontiers Science Center for Rare Isotopes, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Zhiyuan Lin
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Ying Wang
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Yin Zhu
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Jiang Shi
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Junfeng Tan
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Li Guo
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Yanni Zhao
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, P. R. China
| | - Yue Zhang
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Qunhua Peng
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Weidong Dai
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Haipeng Lv
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| | - Zhi Lin
- Key Laboratory of Tea Biology and Resource Utilization of Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, P. R. China
| |
Collapse
|
14
|
Shi X, Wang Z, Bi Z, Liu J, Zhang P. Enhancing Immunomodulation and Osseointegration of Bone Implants via Thrombin-Activated Platelet-Rich Plasma Self-Assembly. Adv Healthc Mater 2025; 14:e2404295. [PMID: 39887661 DOI: 10.1002/adhm.202404295] [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: 11/01/2024] [Revised: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Platelet-rich plasma (PRP) is characterized by elevated concentrations of growth factors that facilitate bone repair. Nonetheless, the effective integration of PRP with bone implants and the sustained release of its active constituents pose significant challenges. In this study, thrombin is grafted onto the surface of polyetheretherketone (PEEK) via an N,N'-Disuccinimidyl Carbonate (DSC) linker and the retained enzymatic activity of thrombin enables the controlled activation of PRP self-assembly, resulting in the formation of a functional bio-gel layer. The optimal thrombin concentration to be 100 U/ mL-1 is determined, at which point both the grafting amount and enzymatic activity of thrombin reaches their peak, with no further increases observed at higher concentrations. PRP solutions with varying platelet enrichment ratios are subsequently activated on the thrombin-grafted PEEK surface, yielding self-assembled bio-gels capable of sustained growth factor release for up to 16 days. The thrombin-activated PRP bio-gel on PEEK surface not only enhances in vitro cell adhesion, proliferation, osteogenic differentiation, vascularization and specific polarization of macrophages, but also effectively facilitates in vivo angiogenesis, immunomodulation and bone formation in a platelet dose-dependent manner. Consequently, the thrombin-activated PRP gel presents a promising strategy for the biological functionalization of PEEK implants in orthopedic applications.
Collapse
Affiliation(s)
- Xiaotong Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Department of Orthopaedics, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, 100050, China
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zhiguo Bi
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jianguo Liu
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
15
|
Fu G, Zhao Y, Mao C, Liu Y. Enhancing nano-immunotherapy of cancer through cGAS-STING pathway modulation. Biomater Sci 2025. [PMID: 40111213 DOI: 10.1039/d4bm01532k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a critical role in cancer immunotherapy due to the secretion of multiple pro-inflammatory cytokines and chemokines. Numerous cGAS-STING agonists have been developed for preclinical and clinical trials in tumor immunity. However, several obstacles, such as agonist molecules requiring multiple doses, rapid degradation and poor targeting, weaken STING activation at the tumor site. The advancement of nanotechnology provides an optimized platform for the clinical application of STING agonists. In this review, we summarize events of cGAS-STING pathway activation, the dilemma of delivering STING agonists, and recent advances in the nano-delivery of cGAS-STING agonist formulations for enhancing tumor immunity. Furthermore, we address the future challenges associated with STING-based therapies and offer insights to guide subsequent clinical applications.
Collapse
Affiliation(s)
- Gaohong Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Yanan Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Chengqiong Mao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, P. R. China
| | - Yang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
16
|
Chu H, Shan Y, Liu Z, Sun M, Zhao W, Xie X, Wang K, Yang C, Fang X, Shen N, Tang Z. Rejuvenation of Tumor-Specific T Cells via Ultrahigh DAR Antibody-Polymeric Imidazoquinoline Complexes: Coordinated Targeting of PDL1 and Efficient TLR7/8 Activation in Intratumoral Dendritic Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2412974. [PMID: 40091265 DOI: 10.1002/adma.202412974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Intratumoral dendritic cells (DCs) are pivotal in tumor treatment due to their immature and pro-tumoral state induced by the tumor microenvironment. Clinically, these immature DCs correlate with disease progression and recurrence, adversely affecting prognosis. Activation of DCs by the TLR7/8 agonist imidazoquinoline (IMDQ) has yielded promising results, but they are limited by systemic inflammation risks, and high programmed death ligand 1 (PDL1) expression on DCs impedes CD8+ T cell activity. Thus, the study introduces an antibody-polymeric IMDQ complex (αPDL1-PLG-IMDQ) with an ultrahigh drug-to-antibody ratio, where αPDL1 is conjugated to Fc-binding peptides on polymeric IMDQ. This complex targets high PDL1-expressing intratumoral DCs with high probability, inducing PDL1-mediated endocytosis to deliver IMDQ to TLR7/8 within endosomes, effectively activating DCs (CD11c+MHC II+: 2.33% versus 1.09%, CD11c+CD86+: 2.49% versus 1.00% on tumors compared to phosphate-buffered saline treatment) and priming T cells. It also blocks PDL1/PD1 interactions, enhancing tumor-specific T-cell activation and memory. Notably, αPDL1-PLG-IMDQ achieved a 97% tumor inhibition rate, prevented tumor regrowth in rechallenge experiments, and reduced lung metastases of tumors by 83%. These findings underscore its potential for intratumoral DC-targeted immunotherapy and novel systemic IMDQ and checkpoint inhibitor combinations.
Collapse
Affiliation(s)
- Hongyu Chu
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yuezhan Shan
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zongyu Liu
- The second hospital of Jilin University, Changchun, 130041, China
| | - Mengmeng Sun
- The second hospital of Jilin University, Changchun, 130041, China
| | - Weidong Zhao
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xiao Xie
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Kun Wang
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Chenguang Yang
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Na Shen
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zhaohui Tang
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
17
|
Zheng F, Wan X, Zhang Y, Yue Y, Li Q, Zhang Z, Li S, Xu H, Su Q, Chen X, Tong L, Zhao L, Cao J, Tang X, Yang X, Wu J, Li J, Lv X, Zhou Z, Wang D. A multimodal defect-rich nanoreactor triggers sono-piezoelectric tandem catalysis and iron metabolism disruption for implant infections. SCIENCE ADVANCES 2025; 11:eads8694. [PMID: 40085709 PMCID: PMC11908489 DOI: 10.1126/sciadv.ads8694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/21/2025] [Indexed: 03/16/2025]
Abstract
Tracking and eradicating drug-resistant bacteria are critical for combating implant-associated infections, yet effective antibacterial therapies remain elusive. Herein, we propose an oxygen vacancy-rich (BiFe)0.9(BaTi)0.1O3-x nanoreactor as a piezoelectric sonosensitizer by spatiotemporal ultrasound-driven sono- and chemodynamic tandem catalysis to amplify antibacterial efficacy. The piezoelectric charge carriers under a built-in electric field synchronize the reaction of O2 and H2O, efficiently generating H2O2. The electron-rich oxygen vacancies modulate the local electronic structure of an Fe site. It facilitates reactive oxygen species generation by piezoelectric electrons and accelerates valence state cycles of Fe(III)/Fe(II) to achieve the sustained maintenance of hydroxyl radicals via H2O2/Fe(II)-catalyzed chemodynamic reactions, which lead to bacterial membrane damage. Transcriptomics analysis revealed that intracellular Fe overload induced by excessive Fe(II)-mediated dysregulation of the two-component system disrupts bacterial metabolism, triggering bacterial ferroptosis-like death. Thus, the porous titanium scaffold, engineered with a piezoelectric nanoreactor, demonstrates superior antibacterial efficacy under ultrasound and facilitates osteogenesis via piezoelectric immunomodulation-activated therapy.
Collapse
Affiliation(s)
- Fuyuan Zheng
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xufeng Wan
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yangming Zhang
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Yue
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiaochu Li
- Department of Orthopedics, First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhuang Zhang
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuoyuan Li
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Xu
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Su
- Department of Orthopedics, Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang 621000, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Le Tong
- Department of Emergency Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
- Disaster Medical Center, Sichuan University, Chengdu 610041, China
| | - Long Zhao
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Cao
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Tang
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jiagang Wu
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jian Li
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiang Lv
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zongke Zhou
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Duan Wang
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Zhao Y, An Y, Wu F, Liu L, Tay FR, Jiao Y, Wang J. Regulation of immune microenvironments by polyetheretherketone surface topography for improving osseointegration. J Nanobiotechnology 2025; 23:199. [PMID: 40069791 PMCID: PMC11895393 DOI: 10.1186/s12951-025-03272-7] [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: 10/13/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Optimizing the immune microenvironment is essential for successful implant osseointegration. In this study, four different nano/microstructures were fabricated on polyetheretherketone (PEEK) substrates by varying the agitation speed during sulfonation to influence osteoimmunomodulation and implant integration. The results indicate that nano/microstructures with minimal dimensions (SP450) inhibit actin polymerization by reducing calcium influx through PIEZO1, activating the anti-inflammatory M2 macrophage phenotype. Among the tested specimens, SP450 exhibited the lowest expression levels of tumor necrosis factor-α and interleukin-1β while releasing the highest levels of anti-inflammatory factors, including interleukin-4 and interleukin-10. This optimized immune environment promotes the osteogenesis of MC3T3-E1 pre-osteoblasts and enhances the osseointegration of PEEK implants. Transcriptomic analysis and validation experiment further revealed that SP450 inhibits osteoclastic differentiation by down-regulating transforming growth factor-β2 and suppressing the NF-κB signaling pathway. These findings suggest that manipulating the surface topography of PEEK implants is an effective strategy for enhancing osseointegration with promising clinical applications.
Collapse
Affiliation(s)
- Yuqing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Yanxin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, PR China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Lipeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, PR China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China.
| |
Collapse
|
19
|
Ai X, Yu P, Hou Y, Shu B, Han B, Yang M, Fan X, Wang J. Research progress in the extraction, purification, structural features, biological activities, and structure-activity relationships from Prunella vulgaris polysaccharides. Int J Biol Macromol 2025; 307:141957. [PMID: 40074139 DOI: 10.1016/j.ijbiomac.2025.141957] [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: 01/08/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Prunella vulgaris L., a globally recognized medicinal and edible plant, has been extensively used in China for over 2000 years, possessing the efficacy of clearing liver-fire, brightening the eyes, dispersing the knot, and reducing swelling. Phytochemical studies have shown that Prunella vulgaris contains various biologically active compounds, including flavonoids, triterpenes, polysaccharides, and phenolic acids. Increasing evidence has demonstrated that Prunella vulgaris polysaccharides (PVPs) are one of representative macromolecular components with various bioactivities that are beneficial to health, such as immunomodulatory, antiviral, antitumor, antioxidant, anti-inflammatory, and hepatoprotective effects, and others. However, the current lack of an understanding of PVPs has restricted their development and utilization. This review emphatically summarized recent progress in the extraction and purification, structural characterization, biological functions, structure-activity relationship, toxicity, and potential applications of PVPs. Additionally, this article also provides a comprehensive understanding, update information and new valuable insights for further research and development of PVPs as therapeutic agents and functional foods.
Collapse
Affiliation(s)
- Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Peiling Yu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Bo Shu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Bin Han
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Xingyue Fan
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China.
| | - Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, PR China; School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China.
| |
Collapse
|
20
|
Yuan X, Kang Y, Li R, Niu G, Shi J, Yang Y, Fan Y, Ye J, Han J, Pei Z, Zhang Z, Ji X. Magnetically triggered thermoelectric heterojunctions with an efficient magnetic-thermo-electric energy cascade conversion for synergistic cancer therapy. Nat Commun 2025; 16:2369. [PMID: 40064895 PMCID: PMC11894112 DOI: 10.1038/s41467-025-57672-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Thermoelectric therapy has been emerging as a promising and versatile strategy for targeting malignant tumors treatment. However, the lack of effective time-space controlled triggering of thermoelectric effect in vivo limits the application of thermoelectric therapy. Here a magnetically triggered thermoelectric heterojunction (CuFe2O4/SrTiO3, CFO/STO) for synergistic thermoelectric/chemodynamic/immuno-therapy is developed. The efficient magnetothermal nanoagent (CFO) is synthesized using the hydrothermal method, and thermoelectric nanomaterials (STO) are grown on its surface to create the heterojunction. To enhance oral delivery efficiency, a fusion membrane (M) of Staphylococcus aureus and macrophage cell membranes are coated the CFO/STO heterojunction, enabling effective targeting of orthotopic colorectal cancer. Once the CFO/STO@M reaches the tumor region, in vitro alternating magnetic field (AMF) stimulation activates the catalytic treatment through a magnetic-thermo-electric energy cascade conversion effect. Additionally, the immunogenic death of tumor cells, down-regulating vascular endothelial growth factor and heat shock protein HSP70, increasing expression of endothelial cell adhesion molecule (ICAM-1/VCAM-1), and M1 polarization of macrophages contribute to tumor immunotherapy. Overall, the magnetically triggered thermoelectric heterojunction based on CFO/STO@M shows remarkable antitumor capability in female mice, offering a promising approach to broaden both the scope of application and the effectiveness of catalytic therapy.
Collapse
Affiliation(s)
- Xue Yuan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yong Kang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Gaoli Niu
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jiacheng Shi
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yiwen Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jiamin Ye
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jingwen Han
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhengcun Pei
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China.
- Medical College, Linyi University, Linyi, China.
| |
Collapse
|
21
|
Zhang N, Ping W, Xiang J, Chu S, Li D, Ning S, Zhu D, Zeng W, Xu Q. Biomimetic Single-Atom Nanozyme for Dual Starvation-Enhanced Breast Cancer Immunotherapy. Adv Healthc Mater 2025; 14:e2401362. [PMID: 39363640 DOI: 10.1002/adhm.202401362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/08/2024] [Indexed: 10/05/2024]
Abstract
Cold exposure (CE) therapy is an innovative and cost-efficient cancer treatment that activates brown adipose tissue to compete for glucose uptake, leading to metabolic starvation in tumors. Exploring the combined antitumor mechanisms of CE and traditional therapies (such as nanocatalysis) is exciting and promising. In this study, a platelet membrane biomimetic single-atom nanozyme (SAEs) nanodrug (PFB) carrying bis-2-(5-phenylacetamido-1, 2, 4-thiadiazol-2-yl) ethyl sulfide (BPTES) is developed for use in cancer CE therapy. Owing to the platelet membrane modification, PFB can effectively target tumors. Upon entering cancer cells, the dual starvation effect induced by CE treatment and BPTES can significantly diminish intracellular glucose and ATP levels, resulting in a substantial reduction in cellular (glutathione) GSH, which can enhance the cytotoxic efficacy of reactive oxygen species generated by SAEs. This strategy not only boosts ROS production in tumors, but also strengthens immune responses, particularly by increasing memory T-cell abundance and suppressing distant tumor growth and tumor metastasis. Compared with SAEs therapy alone, this combined approach offers superior benefits for tumor immunotherapy. This study achieves a combination of CE and nanomedicines for the first time, providing new ideas for future nanomedicine combination therapy modalities.
Collapse
Affiliation(s)
- Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wei Ping
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, 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, China
| | - Sitong Chu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Dan Li
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530003, China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530003, 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, China
| | - Wen Zeng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qingyong Xu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| |
Collapse
|
22
|
Wang Y, Ding L, Feng J, Lin Z, Yao H, You X, Zhang X, Sun W, Liu Y, Wang P. Mesoporous cerium oxide nanoenzyme for Efficacious impeding tumor and metastasis via Conferring resistance to anoikis. Biomaterials 2025; 314:122876. [PMID: 39383776 DOI: 10.1016/j.biomaterials.2024.122876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Tumor cells can survive when detached from the extracellular matrix or lose cell-to-cell connections, leading to a phenomenon known as anoikis resistance (AR). AR is closely associated with the metastasis and proliferation of tumor cells, enabling them to disseminate, migrate, and invade after detachment. Here, we have investigated a novel composite nanoenzyme comprising mesoporous silica/nano-cerium oxide (MSN-Ce@SP/PEG). This nanoenzyme exhibited satisfactory catalase (CAT) activity, efficiently converting high levels of H2O2 within tumor cells into O2, effectively alleviating tumor hypoxia. Furthermore, MSN-Ce@SP/PEG nanoenzyme demonstrated high peroxidase (POD) activity, elevating reactive oxygen species (ROS) levels and attenuating AR in hepatocellular carcinoma (HCC) cells. The MSN-Ce@SP/PEG nanoenzyme exhibited satisfactory dual bioactivity in CAT and POD and was significantly enhanced under favorable photothermal conditions. Through the synergistic effects of these capabilities, the nanoenzyme disrupted the epithelial-mesenchymal transition (EMT) process in detached HCC cells, ultimately inhibiting the recurrence and metastasis potential of anoikis-resistant HCC cells. This study represents the first report of a novel nanoenzyme based on mesoporous silica/nano-cerium oxide for treating AR in HCC cells, thereby suppressing HCC recurrence and metastasis. The findings of this work offer a pioneering perspective for the development of innovative strategies to prevent the recurrence and metastasis of HCC.
Collapse
Affiliation(s)
- Yunhao Wang
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Lei Ding
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China; School of Rare Earths, University of Science and Technology of China, Hefei, 230026, PR China
| | - Juan Feng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Ziguo Lin
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Hanlin Yao
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Xinyu You
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Xiaolong Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China.
| | - Yang Liu
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | - Peiyuan Wang
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China.
| |
Collapse
|
23
|
Ye J, Wang H, Zheng J, Ning S, Zhu D, Shi J, Shi R. Cold Exposure Therapy Enhances Single-Atom Nanozyme-Mediated Cancer Vaccine Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11752-11763. [PMID: 39945542 DOI: 10.1021/acsami.4c20487] [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: 02/28/2025]
Abstract
Single-atom nanozymes are highly effective in the preparation of tumor vaccines (TV) due to their superior peroxidase (POD) activity and excellent biocompatibility. However, the immunosuppressive environment within tumors can diminish the efficacy of these vaccines. Cold exposure (CE) therapy, a noninvasive and straightforward antitumor method, not only suppresses tumor metabolism but also ameliorates the immunosuppressive tumor milieu. In this study, we developed personalized TV using copper single-atom nanozyme (Cu SAZ) and enhanced their long-term antitumor efficacy by introducing CE. We initially synthesized the Cu SAZ via high-temperature carbonization, which demonstrated robust POD activity and photothermal characteristics. Upon exposure to 808 nm laser irradiation, the nanozyme generated reactive oxygen species (ROS) and heat, inducing immunogenic cell death in 4T1 breast cancer cells or CT26 colon cancer cells and facilitating TV production. In our in vivo tumor prevention and treatment model, we noted that CE significantly boosted the efficacy of the TV. The primary mechanism involves CE's ability to lower the ratio of myeloid-derived suppressor cells (MDSCs), decrease glucose metabolism in tumor cells, and increase the proportions of CD8+ T cells and memory T cells. Collectively, our findings offer promising avenues for designing innovative TV systems.
Collapse
Affiliation(s)
- Jinjun Ye
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Shenzhen 518100, China
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongwei Wang
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Shenzhen 518100, China
| | - Jingzhi Zheng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, 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, China
| | - Jing Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Runze Shi
- The Second Ward of Breast Surgery, Cancer Hospital Affiliated to Harbin Medical University, Harbin 150086, China
| |
Collapse
|
24
|
Jiramonai L, Liang XJ, Zhu M. Extracellular Vesicle-Based Strategies for Tumor Immunotherapy. Pharmaceutics 2025; 17:257. [PMID: 40006624 PMCID: PMC11859549 DOI: 10.3390/pharmaceutics17020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Immunotherapy is one of the most promising approaches for cancer management, as it utilizes the intrinsic immune response to target cancer cells. Normally, the human body uses its immune system as a defense mechanism to detect and eliminate foreign objects, including cancer cells. However, cancers develop a 'switch off' mechanism, known as immune checkpoint proteins, to evade immune surveillance and suppress immune activation. Therefore, significant efforts have been made to develop the strategies for stimulating immune responses against cancers. Among these, the use of extracellular vesicles (EVs) to enhance the anti-tumor immune response has emerged as a particularly promising approach in cancer management. EVs possess several unique properties that elevate the potency in modulating immune responses. This review article provides a comprehensive overview of recent advances in this field, focusing on the strategic usage of EVs to overcome tumor-induced immune tolerance. We discuss the biogenesis and characteristics of EVs, as well as their potential applications in medical contexts. The immune mechanisms within the tumor microenvironment and the strategies employed by cancers to evade immune detection are explored. The roles of EVs in regulating the tumor microenvironment and enhancing immune responses for immunotherapy are also highlighted. Additionally, this article addresses the challenges and future directions for the development of EV-based nanomedicine approaches, aiming to improve cancer immunotherapy outcomes with greater precision and efficacy while minimizing off-target effects.
Collapse
Affiliation(s)
- Luksika Jiramonai
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
25
|
Li Y, Du B, Yu L, Luo H, Rong H, Gao X, Yin J. Strategies and challenges of cytosolic delivery of proteins. J Drug Target 2025:1-16. [PMID: 39862226 DOI: 10.1080/1061186x.2025.2458616] [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: 12/09/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The cytosolic delivery of therapeutic proteins represents a promising strategy for addressing diseases caused by protein dysfunction. Despite significant advances, efficient delivery remains challenging due to barriers such as cell membrane impermeability, endosomal sequestration and protein instability. This review summarises recent progress in protein delivery systems, including physical, chemical and biological approaches, with a particular focus on strategies that enhance endosomal escape and targeting specificity. We further discuss the clinical translatability of these approaches and propose future directions for improving delivery efficiency and safety, ultimately unlocking the therapeutic potential of intracellular proteins.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Baojie Du
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lichao Yu
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Luo
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Xiangdong Gao
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
26
|
Yang J, Shang N, Li Z, Xu J, Zhou X, Zhou H, Luo W, Xu P, Zhou Y, Sheng X, Zhu Z, Zhang M, Ma X, Tan M, Wu H. Oral Lactoferrin-Responsive Formulation Anchoring around Inflammatory Bowel Region for IBD Therapy. Adv Healthc Mater 2025; 14:e2402731. [PMID: 39722174 DOI: 10.1002/adhm.202402731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/11/2024] [Indexed: 12/28/2024]
Abstract
Oral formulation is the ideal treatment method for inflammatory bowel disease (IBD) therapy, but the mucosal damage and diarrhea symptoms impede the drug retention around the inflammatory region, severely limiting IBD therapeutic efficacy. To address this, an oral astaxanthin (Ast) precise delivery formulation is developed with the selective Ast anchoring around the inflammatory region by the novel lactoferrin (LF)-responsive flocculation. This formulation also heightens the apparent solubility of Ast with the minimized edible safety risks for the edible raw materials. For in vivo IBD therapy, the precise delivery formulation exhibits remarkable outcomes, including a significant increase in colon length and a 100% survival rate. Furthermore, it is verified that the mechanism of treatment is primarily attributed to the improved immunoregulation, epithelial repair, and gut microbiota remodeling after the LF-responsive flocculation. This effective inflammatory-responsive delivery design is instructive and valuable to develop more precise delivery systems for IBD therapy.
Collapse
Affiliation(s)
- Jinfan Yang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ning Shang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhengqing Li
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ji Xu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xin Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hui Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wen Luo
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Peng Xu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yucheng Zhou
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xueru Sheng
- Liaoning Key Lab of Lignocellulose Chemistry and BioMaterials, College of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, Liaoning, 116034, China
| | - Hao Wu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
27
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
28
|
Su F, Zhang C, Zhang Q, Shen Y, Li S, Shi J, Zhu Y, Lin H, He B. Multifaceted Immunomodulatory Nanocomplexes Target Neutrophilic-ROS Inflammation in Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411823. [PMID: 39737874 PMCID: PMC11848588 DOI: 10.1002/advs.202411823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/18/2024] [Indexed: 01/01/2025]
Abstract
The sepsis-induced acute lung injury (ALI) still represents one of the leading causes of death in critically ill patients, underscoring the need for novel therapies. Excessive activation of immune cells and damage of reactive oxygen species (ROS) are the main factors that exacerbate lung injury. Here, the multifaceted immunomodulatory nanocomplexes targeting the proinflammatory neutrophilic activation and ROS damage are established. The S100A8/9 inhibitor, ABR2575, is loaded in the nanocomplexes, which effectively blocks the neutrophils-S100A8/A9- toll-like receptors (TLRS)-Inflammasome signaling in ALI. Synergically, the SiH nanosheets are encapsulated together with ABR2575 into the core of poly(lactic-co-glycolic acid) (PLGA) nanosponges, to achieve sustainable hydrogen release for the alleviation of ROS-induced lung tissue injury, and also promote the M2 polarization of macrophages. This novel combination strategy is proven to significantly suppress the infiltration of neutrophils and pro-inflammatory macrophages into the lungs, decrease the activation of neutrophils and pro-inflammatory monocytes in the blood, facilitate the anti-inflammatory polarization of macrophages and monocytes, and reduce the expression of pro-inflammatory cytokines in both the lung and blood circulation, all of which alleviate the lung injuries in preclinical murine ALI models. The current investigations offer a novel nanomedicine for the treatment of ALI with great potential in clinical invention.
Collapse
Affiliation(s)
- Fan Su
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Chong Zhang
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Qianyun Zhang
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Yi Shen
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Saiqi Li
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jianlin Shi
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of MedicineTongji UniversityShanghai200072P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
| | - Ya‐Xuan Zhu
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of MedicineTongji UniversityShanghai200072P. R. China
| | - Han Lin
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of MedicineTongji UniversityShanghai200072P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
| | - Bin He
- Department of Critical Care Medicine and Emergency, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030P. R. China
| |
Collapse
|
29
|
Ding Z, Bao X, Chen T, Zhang J, Xu C, Tang N, Hu M, Liu Z. Biocompatible Metal-Organic Framework-Based Fabric Composite as an Efficient Personal Protective Equipment for Particulate Matter-Induced Pulmonary Injury. Adv Healthc Mater 2025; 14:e2403061. [PMID: 39470050 DOI: 10.1002/adhm.202403061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/06/2024] [Indexed: 10/30/2024]
Abstract
Efficient personal protection has emerged as a crucial approach for reducing pulmonary injury induced by particulate matter (PM). However, current personal protective equipments usually lack essential biosafety concerns and fail to own adsorbing/antioxidant/antibacterial function together, making it a challenge to develop an integrated platform with the above characteristics. Herein, a facile oxygen-free hydrothermal strategy is proposed to synthesize new copper-based metal-organic frameworks, Cu-HHTPs, (HHTP: 2,3,6,7,10,11-hexahydroxytriphenylene), with great adsorbing/antioxidant/antibacterial activity and high biosafety. The Cu-HHTPs can serve as an efficient additive incorporated with various fabrics including cellulose acetate (CA) membrane to achieve novel fabric composites, such as CA@Cu-HHTPs, with ideal scavenging outcome for the main components of PM. Evidenced by the animal experiments, CA@Cu-HHTPs can highly mitigate PM-induced adverse effects via adsorbing PM, scavenging ROS, and killing bacteria, leading to a significant reduction in lung permeability, inflammation and oxidative stress, and pulmonary infection. Last but not least, a two-week exposure of CA@Cu-HHTPs exhibits no obvious damage toward the animals by examining their long-term toxicity. Collectively, this study not only highlights the potential of Cu-HHTPs as attractive additives for the preparation of fabric composites, but also lays out a new concept toward the development of new-generation multifunctional personal protective equipment against PM.
Collapse
Affiliation(s)
- Zhen Ding
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Xingfu Bao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Tianyan Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Jinming Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Chengjing Xu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Nan Tang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Min Hu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Zhen Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
30
|
Yu Q, Sun S, Yang N, Pei Z, Chen Y, Nie J, Lei H, Wang L, Gong F, Cheng L. Self-Cascaded Pyroptosis-STING Initiators for Catalytic Metalloimmunotherapy. J Am Chem Soc 2025; 147:3161-3173. [PMID: 39818788 DOI: 10.1021/jacs.4c12552] [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: 01/19/2025]
Abstract
Gasdermin (GSDM)-mediated pyroptosis involves the induction of mitochondrial damage and the subsequent release of mitochondrial DNA (mtDNA), which is anticipated to activate the cGAS-STING pathway, thereby augmenting the antitumor immune response. However, challenges lie in effectively triggering pyroptosis in cancer cells and subsequently enhancing the cGAS-STING activation with specificity. Herein, we developed intelligent self-cascaded pyroptosis-STING initiators of cobalt fluoride (CoF2) nanocatalysts for catalytic metalloimmunotherapy. CoF2 nanocatalysts with a semiconductor structure and enzyme-like activity generated a substantial amount of reactive oxygen species (ROS) under stimulation by endogenous H2O2 and exogenous ultrasound. Importantly, we discovered that Co-based nanomaterials themselves induce pyroptosis in cancer cells. Therefore, CoF2 nanocatalysts initially acted as pyroptosis inducers, triggering caspase-1/GSDMD-dependent pyroptosis in cancer cells via Co2+ and ROS, leading to mtDNA release. Subsequently, CoF2 nanocatalysts were further utilized as intelligent STING agonists that were specifically capable of detecting mtDNA and augmenting the activation of the cGAS-STING pathway. These cascade events triggered a robust immune response, effectively modulating the immunosuppressive tumor microenvironment into an immune-supportive state, thereby providing favorable support for antitumor therapy. This innovative strategy not only significantly impeded the growth of the primary tumor but also elicited an immune response to further augment the efficacy of immune checkpoint inhibitors in preventing distant tumor progression. Overall, this study proposed a self-cascade strategy for activating and amplifying the cGAS-STING pathway with specificity mediated by pyroptosis, representing a valuable avenue for future cancer catalytic metalloimmunotherapy.
Collapse
Affiliation(s)
- Qiao Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jihu Nie
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| |
Collapse
|
31
|
Li Y, Dong Y, Shen D, Guo Y, Cao Y, Zhang K, Li X, Zhu R, Yi J, Yao X, Dang X, Li R, Zhang Z, Qin Z, Yang W. Personalized Nanovaccine Based on STING-Activating Nanocarrier for Robust Cancer Immunotherapy. ACS NANO 2025; 19:3226-3239. [PMID: 39817337 DOI: 10.1021/acsnano.4c11014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Tumor-specific T cells play a vital role in potent antitumor immunity. However, their efficacy is severely affected by the spatiotemporal orchestration of antigen-presentation as well as the innate immune response in dendritic cells (DCs). Herein, we develop a minimalist nanovaccine that exploits a dual immunofunctional polymeric nanoplatform (DIPNP) to encapsulate ovalbumin (OVA) via electrostatic interaction when the nanocarrier serves as both STING agonist and immune adjuvant in DCs. In vitro results reveal that the nanocarrier induces STING activation via facilitating interferon regulatory factor 3 phosphorylation by block poly 18-crown-6-yl methacrylate (P18C6MA) mediated K+ perturbation cascade with endoplasmic reticulum stress, and stimulates DC maturation via the Toll-like receptor 4 activation by primary amine. In vivo studies indicate that the smart nanovaccine dramatically inhibits tumor growth with a long-term immune memory response in both the B16-OVA and EG7-OVA tumor models. After combination with programmed death ligand-1 antibody (aPD-L1), mice survival rate is notably prolonged. In addition, DIPNP forms a personalized nanovaccine after resected autologous primary tumor cell membranes decoration with a high antitumor activity in a homologous distant tumor model. The rational design provides inspiration for personalized nanovaccine construction via immunofunctional nanocarriers.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ya Dong
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Danyang Shen
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yichen Guo
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kaixin Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rongrong Zhu
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaohan Yao
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaowei Dang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rui Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Weijing Yang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
32
|
Cheng W, Duan Z, Chen H, Wang Y, Wang C, Pan Y, Wu J, Wang N, Qu H, Xue X. Macrophage membrane-camouflaged pure-drug nanomedicine for synergistic chemo- and interstitial photodynamic therapy against glioblastoma. Acta Biomater 2025; 193:392-405. [PMID: 39800099 DOI: 10.1016/j.actbio.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Glioblastoma (GBM) persists as a highly fatal malignancy, with current clinical treatments showing minimal progress over years. Interstitial photodynamic therapy (iPDT) holds promise due to its minimally invasive nature and low toxicity but is impeded by poor photosensitizer penetration and inadequate GBM targeting. Here, we developed a biomimetic pure-drug nanomedicine (MM@CT), which co-assembles the photosensitizer chlorin e6 (Ce6) and the first-line chemotherapeutic drug (temozolomide, TMZ) for GBM, then camouflaged with macrophage membranes. This design eliminates the need for traditional excipients, ensuring formulation safety and achieving exceptionally high drug loading with 73.2 %. By leveraging the biomimetic properties of macrophage membranes, MM@CT evades clearance by the mononuclear phagocyte system and can overcome blood circulatory barriers to target intracranial GBM tumors due to its inherent tumor-homing ability. Consequently, this targeted strategy enables precise delivery of TMZ to the tumor site while significantly enhancing Ce6 accumulation within the tumor tissue. Upon intra-tumoral irradiation using an optical fiber, activated Ce6 synergizes with TMZ to exert both cytotoxic effects from chemotherapy and unique advantages from iPDT simultaneously attacking GBM tumors in a dual manner. In subcutaneous and intracranial GBM mouse models, MM@CT exhibits remarkable anti-tumor efficacy with minimal systemic toxicity, emerging as a promising GBM treatment strategy. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) remains a formidable brain cancer, posing significant therapeutic challenges due to the presence of the blood-brain barrier (BBB) and tumor heterogeneity. To overcome these obstacles, we have developed MM@CT, a biomimetic nanomedicine with exceptional drug loading efficiency of 73.2 %. MM@CT incorporates the photosensitizer Ce6 and chemotherapy agent TMZ, encapsulated within nanoparticles and camouflaged with macrophage membranes. This innovative design enables efficient BBB penetration, precise tumor targeting, and synergistic application of chemotherapy and photodynamic therapy. Encouragingly, preclinical evaluations have demonstrated substantial antitumor activity with minimal systemic toxicity, positioning MM@CT as a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjun Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
33
|
Nie J, Yang N, Sun S, Wang L, Pei Z, Wu J, Yu Q, Han Z, Chen Y, Cheng L. Antimony Component Schottky Nanoheterojunctions as Ultrasound-Heightened Pyroptosis Initiators for Sonocatalytic Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202416426. [PMID: 39305135 DOI: 10.1002/anie.202416426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Indexed: 11/08/2024]
Abstract
Pyroptosis, an inflammatory modality of programmed cell death associated with the immune response, can be initiated by bioactive ions and reactive oxygen species (ROS). However, bioactive ion-induced pyroptosis lacks specificity, and further exploration of other ions that can induce pyroptosis in cancer cells is needed. Sonocatalytic therapy (SCT) holds promise due to its exceptional penetration depth; however, the rapid recombination of electron-hole (e--h+) pairs and the complex tumor microenvironment (TME) impede its broader application. Herein, we discovered that antimony (Sb)-based nanomaterials induced pyroptosis in cancer cells. Therefore, Schottky heterojunctions containing Sb component (Sb2Se3@Pt) were effectively designed and constructed via in situ growth of platinum (Pt) nanoparticles (NPs) on Sb2Se3 semiconductor with narrow band gaps, which were utilized as US-heightened pyroptosis initiators to induce highly effective pyroptosis in cancer cells to boost SCT-immunotherapy. Under US irradiation, excited electrons were transferred from Sb2Se3 nanorods (NRs) to the co-catalyst Pt via Schottky junctions, and band bending effectively prevented electron backflow and achieved efficient ROS generation. Moreover, the pores oxidized and depleted the overexpressed GSH in the TME, potentially amplifying ROS generation. The biological effects of the Sb2Se3@Pt nanoheterojunction itself combined with the sonocatalytic amplification of oxidative stress significantly induced Caspase-1/GSDMD-dependent pyroptosis in cancer cells. Therefore, SCT treatment with Sb2Se3@Pt not only effectively restrained tumor proliferation but also induced potent immune memory responses and suppressed tumor recurrence. Furthermore, the integration of this innovative strategy with immune checkpoint blockade (ICB) treatment elicited a systemic immune response, effectively augmenting therapeutic effects and impeding the growth of abscopal tumors. Overall, this study provides further opportunities to explore pyroptosis-mediated SCT-immunotherapy.
Collapse
Affiliation(s)
- Jihu Nie
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078, Macau SAR, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qiao Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078, Macau SAR, China
| |
Collapse
|
34
|
Du FY, Zhou F, Zhao N, Bao L, Hu CB, Lei J, Liu AQ, Gao YF, Bao LH, Ni H, Yu XR, Chen J, Sui BD. YAP1 mediates the dimensional and chemical coordination of immunoregulation and therapy in extensively passaged mesenchymal stem cells. Theranostics 2025; 15:1930-1948. [PMID: 39897564 PMCID: PMC11780522 DOI: 10.7150/thno.103314] [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: 09/05/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Mesenchymal stem cells (MSCs) possess potent immunomodulatory capability, but occasionally, clinical application of MSCs is hindered by compromised cell functionality and insufficient therapeutic efficacy. Methods: Here, well-established mouse models of dextran sulfate sodium (DSS)-induced colitis and streptozotocin (STZ)-induced type 1 diabetes (T1D) were used to evaluate therapeutic immunomodulatory effects of human umbilical cord-derived MSCs. MSCs were examined at the fifth (P5) and the fifteenth (P15) passages, and three-dimensional (3D) culture was conducted by Matrigel incorporation. A series of biochemical, histopathological and cellular assays were performed to investigate the MSC function and therapeutic performance, and immunoregulation was evaluated by in vitro co-culture with T cells and in vivo analyses of T-cell infiltration into target tissues. RNA sequencing (RNA-seq) analysis followed by immunofluorescence staining, gene expression analyses and chemical regulation were used to investigate the molecular targets. Results: MSCs lose therapeutic immunomodulatory effects after extensive expansion to P15 when cell senescence occurs. Intriguingly, 3D preconditioning of MSCs in Matrigel promotes diminished immunoregulatory capability despite extensive passages, which benefits function of P15-MSCs to modulate T-cell subsets in co-culture, suppress infiltration of pro-inflammatory T cells in the colon and pancreas tissues after infusion, ameliorate systemic inflammation, and alleviate colitis and T1D in mice. Mechanistically, 3D culture provokes transcriptomic reprogramming of MSCs toward a Yes-associated protein 1 (YAP1)-marked, Hippo signaling pathway-upregulated state with promoted release of the anti-inflammatory cytokine, transforming growth factor-beta1 (TGF-β1). Moreover, chemical regulation of YAP1 by clinically relevant drugs, verteporfin (VP) and prostaglandin E2 (PGE2), affects TGF-β1 expression and the immunomodulatory capability of MSCs during dimensional culture. Conclusions: Taken together, these findings unravel YAP1-based dimensional and chemical coordination of expanded MSC immunoregulation, which will shed light on precisely controlled translational application.
Collapse
Affiliation(s)
- Fang-Ying Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Na Zhao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Lei Bao
- Department of Obstetrics and Gynecology, Xi'an No. 4 Hospital, Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Cheng-Biao Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Lei
- Department of Obstetrics and Gynecology, Xi'an No. 4 Hospital, Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - An-Qi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ying-Feng Gao
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Li-Hui Bao
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Hua Ni
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Xiao-Rui Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Department of Oral Implantology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
35
|
Yin X, Lai Y, Zhang X, Zhang T, Tian J, Du Y, Li Z, Gao J. Targeted Sonodynamic Therapy Platform for Holistic Integrative Helicobacter pylori Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408583. [PMID: 39535366 PMCID: PMC11727135 DOI: 10.1002/advs.202408583] [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: 07/25/2024] [Revised: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Helicobacter pylori (H. pylori) is a primary pathogen associated with gastrointestinal diseases, including gastric cancer. The increase in resistance to antibiotics, along with the adverse effects caused by complicated medication protocols, has made the eradication of H. pylori a more formidable challenge, necessitating alternative therapeutics. Herein, a targeted nanoplatform is reported based on sonodynamic therapy, the chitosan-conjugated fucose loaded with indocyanine green (ICG@FCS). It penetrates the gastric mucosa and homes in on H. pylori through dual targeting mechanisms: molecular via fucose and physical via ultrasound. Upon ultrasound activation, it generates singlet oxygen, effectively attacking planktonic bacteria, disrupting biofilms, and facilitating the clearance of intracellular bacteria by promoting autophagy, including multidrug-resistant strains. The ICG@FCS nanoplatform minimally affects the gut microbiota and aids in gastric mucosa repair. a holistic integrative H. pylori therapy strategy is proposed that targets eradication while preserving gastrointestinal health. This strategy emphasizes the importance of maintaining patient health while eradicating the pathogen. This advancement is set to refine the comprehensive antibacterial approach, offering a promising horizon in the ongoing battle against antibiotic resistance and more effective gastric cancer prevention strategies.
Collapse
Affiliation(s)
- Xiaojing Yin
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
| | - Yongkang Lai
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- Department of GastroenterologyGanzhou People's Hospital Affiliated to Nanchang UniversityGanzhou341000China
| | - Xinyuan Zhang
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghai200093China
| | - Tingling Zhang
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Jing Tian
- Department of PharmacyShanghai Changhai Hospitalthe First Affiliated Hospital of Navy Medical UniversityShanghai200433China
| | - Yiqi Du
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Zhaoshen Li
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Jie Gao
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical DevicesShanghai200433China
| |
Collapse
|
36
|
Song Q, Gu L, Kong L. Structural Characterization and Anti-Inflammatory Activities of a Purified Polysaccharide from Veronica persica Poir. Chem Biodivers 2025; 22:e202401566. [PMID: 39301582 DOI: 10.1002/cbdv.202401566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
In this manuscript, the polysaccharide (VPP-I) from Veronica persica Poir., was characterized in detail by high performance gel permeation chromatography (HPGPC), high performance liquid chromatography (HPLC), Fourier transform infrared spectroscopy (FT-IR) and nuclear magnetic resonance spectroscopy (NMR). In addition, lipopolysaccharide (LPS) induced inflammation model of RAW264.7 cells was used to evaluate the in vitro anti-inflammatory activity of VPP-I. The results showed that the relative molecular weight of VPP-I was 2.355 KDa, which was mainly composed of mannose (Man), glucose (Glc) and galactose (Gal) in a ratio of 1 : 32.46 : 28.76. Moreover, the VPP-I contained sugar alcohol derivatives of T-DGlcp(1→, →4)-D-Galp(1→, →3,6)-D-Manp(1→, →4)-D-Glcp(1→, →6)-D-Galp(1→ and →6)-D-Glcp(1→. In vitro anti-inflammatory results showed that VPP-I could inhibit the secretion of IL-β, IL-6 and TNF-α in RAW264.7 cells induced by LPS. Moreover, compared to the LPS group, the mRNA expression levels of iNOS, COX-2, IL-β, IL-6 and TNF-α produced by RAW264.7 were significantly decreased after treatment with VPP-I (P<0.05). In addition, VPP-I could increase the SOD and GSH-Px enzymes activity and decrease the content of MDA in LPS-induced RAW264.7 cells (P<0.05). In summary, this paper laid theoretical foundation for the application of Veronica persica Poir. in the field of medicine.
Collapse
Affiliation(s)
- Qiaoying Song
- College of Biotechnology and Food Science, Anyang Institute of Technology, Huanghe Road, Anyang, 455000, China
| | - Lingbiao Gu
- College of Biotechnology and Food Science, Anyang Institute of Technology, Huanghe Road, Anyang, 455000, China
| | - Lingqi Kong
- College of Biotechnology and Food Science, Anyang Institute of Technology, Huanghe Road, Anyang, 455000, China
| |
Collapse
|
37
|
Oluwole SA, Weldu WD, Jayaraman K, Barnard KA, Agatemor C. Design Principles for Immunomodulatory Biomaterials. ACS APPLIED BIO MATERIALS 2024; 7:8059-8075. [PMID: 38922334 DOI: 10.1021/acsabm.4c00537] [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: 06/27/2024]
Abstract
The immune system is imperative to the survival of all biological organisms. A functional immune system protects the organism by detecting and eliminating foreign and host aberrant molecules. Conversely, a dysfunctional immune system characterized by an overactive or weakened immune system causes life-threatening autoimmune or immunodeficiency diseases. Therefore, a critical need exists to develop technologies that regulate the immune system to ensure homeostasis or treat several diseases. Accumulating evidence shows that biomaterials─artificial materials (polymers, metals, ceramics, or engineered cells and tissues) that interact with biological systems─can trigger immune responses, offering a materials science-based strategy to modulate the immune system. This Review discusses the expanding frontiers of biomaterial-based immunomodulation, focusing on principles for designing these materials. This Review also presents examples of immunomodulatory biomaterials, which include polymers and metal- and carbon-based nanomaterials, capable of regulating the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Samuel Abidemi Oluwole
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Welday Desta Weldu
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Keerthana Jayaraman
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Kelsie Amanda Barnard
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Christian Agatemor
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
- Department of Biology, University of Miami, Coral Gables, Florida 33124, United States
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida 33136, United States
| |
Collapse
|
38
|
Liu W, Yao C, Wang D, Du G, Ji Y, Li Q. Dynamic Double-Networked Hydrogels by Hybridizing PVA and Herbal Polysaccharides: Improved Mechanical Properties and Selective Antibacterial Activity. Gels 2024; 10:821. [PMID: 39727579 DOI: 10.3390/gels10120821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Chinese herbal medicine has offered an enormous source for developing novel bio-soft materials. In this research, the natural polysaccharide isolated from the Chinese herbal medicine Dendrobium was employed as the secondary building block to fabricate a "hybrid" hydrogel with synthetic poly (vinyl alcohol) (PVA) polymers. Thanks to the presence of mannose units that contain cis-diol motifs on the chain of the Dendrobium polysaccharides, efficient crosslinking with the borax is allowed and reversible covalent borate ester bonds are formed. Eventually, highly dynamic and double-networked hydrogels were successfully prepared by the integration of Dendrobium polysaccharides and PVA. Interestingly, the introduction of polysaccharides has given rise to more robust and dynamic hydrogel networks, leading to enhanced thermal stability, mechanical strength, and tensile capacity (>1000%) as well as the rapid self-healing ability (<5 s) of the "hybrid" hydrogels compared with the PVA/borax single networked hydrogel. Moreover, the polysaccharides/PVA double network hydrogel showed selective antibacterial activity towards S. aureus. The reported polysaccharides/PVA double networked hydrogel would provide a scaffold to hybridize bioactive natural polysaccharides and synthetic polymers for developing robust but dynamic multiple networked hydrogels that are tailorable for biomedical applications.
Collapse
Affiliation(s)
- Weidong Liu
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chuying Yao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Daohang Wang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Guangyan Du
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yutian Ji
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-Constructed by the Province and Ministry, Ministry-of-Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Quan Li
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
39
|
Liu W, Nie F, Jiang H, Zhao Y, Zhang Y, Zhang Z, Zhang J, Xu J, Guo Y. Preparation of pH-Sensitive Polysaccharide-Small Molecule Nanoparticles and Their Applications for Tumor Chemo- and Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68437-68452. [PMID: 39586061 DOI: 10.1021/acsami.4c16504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Hydrophobic chemotherapy drugs face significant challenges in cancer treatment, including low bioavailability, unavoidable toxic side effects, and the development of drug resistance. To address these issues, a multifunctional nanoplatform was developed for cancer therapy, aimed at achieving effective drug delivery and enhancing antitumor efficacy. Poria cocos polysaccharide (PCP), a natural polymer known for its immunomodulatory properties, was utilized as an immunoreactive vector for drug delivery after being cross-linked with 1,4-phenylenebisboronic acid (BDBA). Subsequently, a small-molecule chemotherapy drug, esculetin (EL), was confirmed through density functional theory (DFT) simulations to be encapsulated within the PCP-BDBA nanoparticles via weak interactions. The results demonstrated that the synthesized nanoparticles were spherical, with an average particle size of 162.0 nm. In addition to exhibiting excellent stability, the nanoparticles also displayed pH-responsive drug release properties. In vivo experiments indicated that EL@PCP-BDBA NPs exhibited antitumor effects. Furthermore, EL@PCP-BDBA NPs showed superior in vitro antitumor activity compared to EL at the cellular level. Additionally, EL@PCP-BDBA NPs were found to increase intracellular reactive oxygen species (ROS) levels, induce cell apoptosis, and suppress cell migration to combat cancer. Meanwhile, EL@PCP-BDBA NPs enhanced immune function in vivo. In summary, this study developed a nano-pharmaceutical that combined chemotherapy and immunotherapy functions, which was considered a promising tool for cancer therapy.
Collapse
Affiliation(s)
- Wenhui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Fan Nie
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Haojing Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yinan Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yan Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, People's Republic of China
| | - Zheng Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, People's Republic of China
| | - Jie Zhang
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi, Xinjiang 832003, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| |
Collapse
|
40
|
Jia W, Chandra J, Teoh SM, Tolley L, Yang H, Tse BWC, Sokolowski KA, Liang X, Crawford DHG, Steel JC, Yu C, Roberts MS, Frazer IH, Chen Y, Wang H, Yu M. STING Nanoagonist Boosts Antitumor Immunity of Therapeutic DNA Vaccines. NANO LETTERS 2024; 24:15588-15597. [PMID: 39621441 DOI: 10.1021/acs.nanolett.4c03846] [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: 12/12/2024]
Abstract
Therapeutic DNA cancer vaccines can stimulate specific immune responses against cancer antigens but often induce suboptimal therapeutic responses. Here, we demonstrate that a manganese-doped silica nanoparticle STING agonist (MSNA) enhances the immune response of plasmid DNA vaccines, promoting the activation and migration of distinct subsets of dendritic cell (DC) and improving antitumor immunity in three animal models. MSNA coadministered with an α-fetoprotein (AFP) encoded plasmid DNA (AFP-DNA) elicited significantly higher AFP-specific CD8 T cell responses than free AFP-DNA. Animals immunized with MSNA-AFP-DNA remained tumor-free in an AFP expressing hepatocellular carcinoma challenge model. MSNA combined with a DNA plasmid encoding the human papillomavirus type 16 oncoproteins E6 and E7 induced potent E7-specific CD8 T cell responses, preventing the growth of E7-expressing solid TC-1 tumors and promoting the shrinkage of E7-expressing skin grafts. These findings together demonstrate that coadministration of MSNA can improve the efficacy of therapeutic DNA vaccines targeting cancer-specific antigens.
Collapse
Affiliation(s)
- Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Janin Chandra
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Siok Min Teoh
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Lynn Tolley
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Haotian Yang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, Queensland 4120 Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4006 Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Xiaowen Liang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, Queensland 4120 Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4006 Australia
| | - Darrell H G Crawford
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, Queensland 4120 Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4006 Australia
| | - Jason C Steel
- School of Health, Medical and Applied Sciences, CQUniversity, North Rockhampton, Queensland 4702, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michael S Roberts
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Ian H Frazer
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Haolu Wang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
- Internal Medicine Services, The Prince Charles Hospital, Chermside, Queensland 4032, Australia
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
41
|
He S, Ma L, Zheng Q, Wang Z, Chen W, Yu Z, Yan X, Fan K. Peptide nanozymes: An emerging direction for functional enzyme mimics. Bioact Mater 2024; 42:284-298. [PMID: 39285914 PMCID: PMC11403911 DOI: 10.1016/j.bioactmat.2024.08.033] [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: 06/17/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
The abundance of molecules on early Earth likely enabled a wide range of prebiotic chemistry, with peptides playing a key role in the development of early life forms and the evolution of metabolic pathways. Among peptides, those with enzyme-like activities occupy a unique position between peptides and enzymes, combining both structural flexibility and catalytic functionality. However, their full potential remains largely untapped. Further exploration of these enzyme-like peptides at the nanoscale could provide valuable insights into modern nanotechnology, biomedicine, and even the origins of life. Hence, this review introduces the groundbreaking concept of "peptide nanozymes (PepNzymes)", which includes single peptides exhibiting enzyme-like activities, peptide-based nanostructures with enzyme-like activities, and peptide-based nanozymes, thus enabling the investigation of biological phenomena at nanoscale dimensions. Through the rational design of enzyme-like peptides or their assembly with nanostructures and nanozymes, researchers have found or created PepNzymes capable of catalyzing a wide range of reactions. By scrutinizing the interactions between the structures and enzyme-like activities of PepNzymes, we have gained valuable insights into the underlying mechanisms governing enzyme-like activities. Generally, PepNzymes play a crucial role in biological processes by facilitating small-scale enzyme-like reactions, speeding up molecular oxidation-reduction, cleavage, and synthesis reactions, leveraging the functional properties of peptides, and creating a stable microenvironment, among other functions. These discoveries make PepNzymes useful for diagnostics, cellular imaging, antimicrobial therapy, tissue engineering, anti-tumor treatments, and more while pointing out opportunities. Overall, this research provides a significant journey of PepNzymes' potential in various biomedical applications, pushing them towards new advancements.
Collapse
Affiliation(s)
- Shaobin He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, 350004, China
| | - Long Ma
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qionghua Zheng
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, 350004, China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Chen
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, 350004, China
| | - Zihang Yu
- Department of Biomedical Engineering, Hajim School of Engineering & Applied Sciences, University of Rochester, Rochester, 14627, USA
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| |
Collapse
|
42
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [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: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
43
|
Li C, Xu W, Li L, Zhou Y, Yao G, Chen G, Xu L, Yang N, Yan Z, Zhu C, Fang S, Qiao Y, Bai J, Li M. Concrete-Inspired Bionic Bone Glue Repairs Osteoporotic Bone Defects by Gluing and Remodeling Aging Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408044. [PMID: 39455287 DOI: 10.1002/advs.202408044] [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: 07/15/2024] [Revised: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Osteoporotic fractures are characterized by abnormal inflammation, deterioration of the bone microenvironment, weakened mechanical properties, and difficulties in osteogenic differentiation. The chronic inflammatory state characterized by aging macrophages leads to delayed or non-healing of the fracture or even the formation of bone defects. The current bottleneck in clinical treatment is to achieve strong fixation of the comminuted bone fragments and effective regulation of the complex microenvironment of aging macrophages. Inspired by cement and gravel in concrete infrastructure, a biomimetic bone glue with poly(lactic-co-glycolic acid) microspheres is developed and levodopa/oxidized chitosan hydrogel stabilized on an organic-inorganic framework of nanohydroxyapatite, named DOPM. DOPM is characterized via morphological and mechanical characterization techniques, in vitro experiments with bone marrow mesenchymal stromal cells, and in vivo experiments with an aged SD rat model exhibiting osteoporotic bone defects. DOPM exhibited excellent adhesion properties, good biocompatibility, and significant osteogenic differentiation. Transcriptomic analysis revealed that DOPM improved the inflammatory microenvironment by inhibiting the NF-κB signaling pathway and promoting aging macrophage polarization toward M2 macrophages, thus significantly accelerating bone defect repair and regeneration. This biomimetic bone glue, which enhances osteointegration and reestablishes the homeostasis of aging macrophages, has potential applications in the treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Chong Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
- Department of Orthopedics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230022, China
| | - Wei Xu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Lei Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Yonghui Zhou
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Gang Yao
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Guang Chen
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Lei Xu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Ning Yang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Zhanjun Yan
- Department of Orthopedics, The Ninth People's Hospital of Suzhou, Suzhou, Jiangsu, 215006, China
| | - Chen Zhu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Shiyuan Fang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
- Department of Orthopedics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| | - Meng Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, China
| |
Collapse
|
44
|
Hu Y, Chen LL, Ye Z, Li LZ, Qian HZ, Wu MQ, Wang J, Qin KH, Ye QB. Indigo naturalis as a potential drug in the treatment of ulcerative colitis: a comprehensive review of current evidence. PHARMACEUTICAL BIOLOGY 2024; 62:818-832. [PMID: 39475104 PMCID: PMC11533244 DOI: 10.1080/13880209.2024.2415652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/25/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024]
Abstract
CONTEXT Ulcerative colitis (UC) is an intractable inflammatory bowel disease that threatens the health of patients. The limited availability of therapeutic strategies makes it imperative to explore more efficient and safer drugs. Indigo naturalis (IN) is a traditional Chinese medicine that possesses many pharmacological activities, including anti-inflammatory, antioxidant, and immunomodulatory activities. The treatment potential of IN for UC has been proven by numerous preclinical and clinical studies in recent years. OBJECTIVE This article provides a comprehensive review of the utility and potential of IN in the treatment of UC. METHODS 'Indigo naturalis' 'Qing dai' 'Qingdai' 'Ulcerative colitis' and 'UC' are used as the keywords, and the relevant literature is collected from online databases (Elsevier, PubMed, and Web of Science). RESULTS AND CONCLUSION Indirubin, indigo, isatin, tryptanthrin, and β-sitosterol are considered the key components in the treatment of UC with IN. Both preclinical and clinical studies support the efficacy of IN for UC, especially in severe UC or in those who do not respond to or have poor efficacy with existing therapies. The mechanisms of IN for UC are associated with the aryl hydrocarbon receptor pathway activation, immune regulation, oxidative stress inhibition, and intestinal microbial modulation. However, the clinical use of IN has the risks of adverse events such as pulmonary hypertension, which suggests the necessity for its rational application. As a potential therapeutic agent for UC that is currently receiving more attention, the clinical value of IN has been initially demonstrated and warrants further evaluation.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liu-lin Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin-zhen Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan-zhu Qian
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming-quan Wu
- Department of Pharmacy, Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Juan Wang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai-hua Qin
- Health Preservation and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiao-bo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
45
|
Feng Y, Tang Q, Wang B, Yang Q, Zhang Y, Lei L, Li S. Targeting the tumor microenvironment with biomaterials for enhanced immunotherapeutic efficacy. J Nanobiotechnology 2024; 22:737. [PMID: 39605063 PMCID: PMC11603847 DOI: 10.1186/s12951-024-03005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) is a complex system characterized by low oxygen, low pH, high pressure, and numerous growth factors and protein hydrolases that regulate a wide range of biological behaviors in the tumor and have a profound impact on cancer progression. Immunotherapy is an innovative approach to cancer treatment that activates the immune system, resulting in the spontaneous killing of tumor cells. However, the therapeutic efficacy of these clinically approved cancer immunotherapies (e.g., immune checkpoint blocker (ICB) therapies and chimeric antigen receptor (CAR) T-cell therapies) is far from satisfactory due to the presence of immunosuppressive TMEs created in part by tumor hypoxia, acidity, high levels of reactive oxygen species (ROS), and a dense extracellular matrix (ECM). With continuous advances in materials science and drug-delivery technologies, biomaterials hold considerable potential for targeting the TME. This article reviews the advances in biomaterial-based targeting of the TME to advance our current understanding on the role of biomaterials in enhancing tumor immunity. In addition, the strategies for remodeling the TME offer enticing advantages; however, the represent a double-edged sword. In the process of reshaping the TME, the risk of tumor growth, infiltration, and distant metastasis may increase.
Collapse
Affiliation(s)
- Yekai Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
46
|
Wang F, Li Z. Engineered extracellular vesicles as "supply vehicles" to alleviate type 1 diabetes. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:618-621. [PMID: 39811729 PMCID: PMC11725422 DOI: 10.20517/evcna.2024.61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 01/16/2025]
Abstract
Recent findings have indicated that the deficiency of inhibitory programmed cell death ligand 1 (PD-L1) and galectin-9 (Gal-9) in pancreatic β-cells is associated with the progression of type 1 diabetes (T1D). This suggests that exogenous PD-L1 and Gal-9 may have promising potential as therapeutics for the treatment of T1D. In light of these reports, a recent work investigated the potential of artificial extracellular vesicles (aEVs) with the presentation of PD-L1 and Gal-9 ligands (PD-L1-Gal-9 aEVs) as a treatment for T1D, with the findings published in Diabetes. Notably, the PD-L1-Gal-9 aEVs demonstrated the capacity to induce apoptosis of T cells and the formation of regulatory T (Treg) cells, thereby maintaining immune tolerance. Furthermore, the in vivo administration of PD-L1-Gal-9 aEVs resulted in a reduction in T cell infiltration in the pancreas, an increase in β-cell integrity protection, a significant decrease in blood glucose levels, and a delay in the progression of T1D. In conclusion, this study proposed an innovative approach to the treatment of T1D progression through the use of immunosuppressive EVs. This highlight provides a comprehensive analysis and discussion of the pivotal findings of this study.
Collapse
Affiliation(s)
- Fei Wang
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523059, Guangdong, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhenhua Li
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523059, Guangdong, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
47
|
Wei X, Sun L, Deng J, Yang Q, Zhao J, Zhou S. A Multifunctional Exosome with Dual Homeostasis Disruption Augments cGAS-STING-Mediated Tumor Immunotherapy by Boosting Ferroptosis. NANO LETTERS 2024; 24:14263-14272. [PMID: 39475013 DOI: 10.1021/acs.nanolett.4c03862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Ferroptosis has shown great potential in activating antitumor immunity. However, the cunning tumor cells can evade ferroptosis by increasing the efflux of iron and promoting the production of the reductant glutathione to mitigate oxidative stress. Herein, a multifunctional exosome loaded with manganese-doped iron oxide nanoparticles (MnIO), GW4869, and l-buthionine sulfoximine (BSO) is developed to disrupt the iron metabolism homeostasis and redox homeostasis to enhance tumor immunotherapy. The efficient transport of MnIO by exosomes and the inhibition of iron exocytosis by GW4869 led to a high retention of up to 29.57% ID/g for iron in the tumors. Such a high retention of iron, in combination with the BSO-induced disruption of the redox homeostasis, effectively promotes the ferroptosis of tumor cells. Consequently, the multifunctional exosomes that noticeably enhance ferroptosis by dual homeostasis disruption provoke the cGAS-STING-based antitumor immune response and effectively suppress tumor growth and lung metastasis in orthotopic breast cancer.
Collapse
Affiliation(s)
- Xiaoqing Wei
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Ling Sun
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Junzhen Deng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Qingping Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jingya Zhao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
48
|
Ma J, Hua L, Zhu Y, Mao G, Fu C, Qin S. Photo-Thermally Controllable Tumor Metabolic Modulation to Assist T Cell Activation for Boosting Immunotherapy. Int J Nanomedicine 2024; 19:11181-11194. [PMID: 39513087 PMCID: PMC11542477 DOI: 10.2147/ijn.s483815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Background Glycolysis is crucial for tumor cell proliferation, supporting their energy needs and influencing the tumor microenvironment (TME). On one hand, increased lactate levels produced by glycolysis acidifies the TME, inhibiting T cell activity. On the other hand, glycolysis promotes the expression of PD-L1 through various mechanisms, facilitating immune evasion. Therefore, controlled modulation of glycolysis in tumor cells to subsequently improve the immune tumor microenvironment holds significant implications for clinical cancer treatment and immune regulation. Methods To reverse the immunosuppressive microenvironment caused by tumor glycolysis and reduce tumor immune escape, we developed a photo-thermal-controlled precision drug delivery platform to regulate tumor metabolism and aid in the activation of T cells, thereby enhancing immunotherapy. First, hollow mesoporous Prussian blue (HPB) was prepared, and the glycolysis inhibitor 3-bromopyruvate (3-BrPA) was encapsulated within HPB using the phase-change material 1-tetradecanol, resulting in B/T-H. This product was then modified with tumor cell membranes to obtain a photo-thermal controllable regulator (B/T-H@Membrane, B/T-HM). Results Due to the excellent drug loading and photo-thermal properties of HPB, upon reaching the tumor, B/T-HM can rapidly heat under 808 nm irradiation, causing the 1-tetradecanol to transition to a liquid phase and release 3-BrPA, which effectively inhibits tumor glycolysis through the HK2 pathway, thereby reducing tumor cell proliferation, decreasing lactate production, and downregulating tumor PD-L1 expression. In synergy with photo-thermal and αPD-1, this photo-thermally controllable metabolic-immune therapy effectively activates T cells to eliminate tumor. Conclusion In response to the changes in immune microenvironment caused by tumor metabolism, a photo-thermal precision-controlled drug delivery platform was successfully developed. This platform reshapes the tumor immunosuppressive microenvironment, providing a new approach for T cell-based tumor immunotherapy. It also opens new avenues for photo-thermal controllable metabolic-immune therapy.
Collapse
Affiliation(s)
- Jun Ma
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Lixin Hua
- Department of General Surgery, Affiliated Huishan Hospital of Xinglin College, Nantong University, Wuxi Huishan District People’s Hospital, Wuxi, People’s Republic of China
| | - Yinxing Zhu
- Department of Traditional Chinese Medicines, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, 225300, People’s Republic of China
| | - Guangyao Mao
- Institute of Clinical Medicine, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| | - Chunsheng Fu
- Institute of Clinical Medicine, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| | - Shiyue Qin
- Department of Ophthalmology, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| |
Collapse
|
49
|
Yuan K, Zhang C, Pan X, Hu B, Zhang J, Yang G. Immunomodulatory metal-based biomaterials for cancer immunotherapy. J Control Release 2024; 375:249-268. [PMID: 39260573 DOI: 10.1016/j.jconrel.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Cancer immunotherapy, as an emerging cancer treatment approach, harnesses the patient's own immune system to effectively prevent tumor recurrence or metastasis. However, its clinical application has been significantly hindered by relatively low immune response rates. In recent years, metal-based biomaterials have been extensively studied as effective immunomodulators and potential tools for enhancing anti-tumor immune responses, enabling the reversal of immune suppression without inducing toxic side effects. This review introduces the classification of bioactive metal elements and summarizes their immune regulatory mechanisms. In addition, we discuss the immunomodulatory roles of biomaterials constructed from various metals, including aluminum, manganese, gold, calcium, zinc, iron, magnesium, and copper. More importantly, a systematic overview of their applications in enhancing immunotherapy is provided. Finally, the prospects and challenges of metal-based biomaterials with immunomodulatory functions in cancer immunotherapy are outlined.
Collapse
Affiliation(s)
- Kangzhi Yuan
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Xinlu Pan
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Bin Hu
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, Jiangsu 215004, China.
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School 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, Jiangsu 215123, China.
| |
Collapse
|
50
|
Li S, Yue Y, Wang W, Han M, Wan X, Li Q, Chen X, Cao J, Zhang Y, Li J, Li J, Cheng L, Yang J, Wang D, Zhou Z. Ultrasound-Activated Probiotics Vesicles Coating for Titanium Implant Infections Through Bacterial Cuproptosis-Like Death and Immunoregulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405953. [PMID: 39101293 DOI: 10.1002/adma.202405953] [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: 04/25/2024] [Revised: 07/09/2024] [Indexed: 08/06/2024]
Abstract
Implant-associated infections (IAIs) are the main cause of prosthetic implant failure. Bacterial biofilms prevent antibiotic penetration, and the unique metabolic conditions in hypoxic biofilm microenvironment may limit the efficacy of conventional antibiotic treatment. Escaping survival bacteria may not be continually eradicated, resulting in the recurrence of IAIs. Herein, a sonosensitive metal-organic framework of Cu-TCPP (tetrakis(4-carboxyphenyl) porphyrin) nanosheets and tinidazole doped probiotic-derived membrane vesicles (OMVs) with high-penetration sonodynamic therapy (SDT), bacterial metabolic state interference, and bacterial cuproptosis-like death to eradicate IAIs is proposed. The Cu-TCPP can convert O2 to toxic 1O2 through SDT in the normoxic conditions, enhancing the hypoxic microenvironment and activating the antibacterial activity of tinidazole. The released Cu(II) under ultrasound can be converted to Cu(I) by exogenous poly(tannic acid) (pTA) and endogenous glutathione. The disruption of the bacterial membrane by SDT can enhance the Cu(I) transporter activity. Transcriptomics indicate that the SDT-enhanced Cu(I) overload and hypoxia-activated therapy hinder the tricarboxylic acid cycle (TCA), leading to bacterial cuproptosis-like death. Moreover, the OMVs-activated therapy can polarize macrophages to a M2-like phenotype and facilitate bone repair. The sonodynamic biofilm microenvironment modulation strategy, whereby the hypoxia-enhanced microenvironment is potentiated to synergize SDT with OMVs-activated therapy, provides an effective strategy for antibacterial and osteogenesis performance.
Collapse
Affiliation(s)
- Shuoyuan Li
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Yue
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenqi Wang
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingyue Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xufeng Wan
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiaochu Li
- Department of orthopedics, the First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Cao
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangming Zhang
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jianshu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Duan Wang
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|