1
|
Bing J, Zhou B, Chen M, Shen Y, Zhou M, Lin H, Wu W, Shi J. Nanomedicine-enabled concurrent regulations of ROS generation and copper metabolism for sonodynamic-amplified tumor therapy. Biomaterials 2025; 318:123137. [PMID: 39884132 DOI: 10.1016/j.biomaterials.2025.123137] [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: 09/26/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Sonodynamic therapy (SDT) shows substantial potentials in cancer treatment thanks to the deep tissue penetration of ultrasound. However, its clinical translation suffers from the potential damages to healthy tissues and the resistance of tumors, particularly from cancer stem-like cells (CSCs), to the ultrasound. To address these challenges, we designed a novel glutathione (GSH)-activated nanomedicine to simultaneously enhance the safety and efficacy of SDT by in situ regulating the generation of reactive oxygen species (ROS) and copper metabolism. This nanomedicine, Es@CuTCPP, was created by loading elesclomol (Es) onto CuTCPP nanosheets. By accumulating this nanomedicine in tumors, the Cu(II)-TCPP is reduced to the highly sonosensitive Cu(I)-TCPP by the intra-tumoral-overexpressed GSH, leading to the production of abundant ROS upon ultrasound exposure, which effectively kills large amounts of tumor cells. Concurrently, the released copper ions react with co-released Es to form a CuEs complex, which induces cuproptosis of CSCs surviving the ROS attack by disrupting cellular copper metabolism, evidently amplifying the effectiveness of SDT. This work presents the first paradigm of a GSH-activated and cuproptosis-enhanced SDT approach, offering a promising novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Jinhong Bing
- State Key Laboratory of High-performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, PR China
| | - Bangguo Zhou
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Minqi Chen
- Digestive Endoscopy Center, Shanghai Fourth People's Hospital to Tongji University, Shanghai, 200081, PR China
| | - Yucui Shen
- Digestive Endoscopy Center, Shanghai Fourth People's Hospital to Tongji University, Shanghai, 200081, PR China
| | - Min Zhou
- Digestive Endoscopy Center, Shanghai Fourth People's Hospital to Tongji University, Shanghai, 200081, PR China
| | - Han Lin
- State Key Laboratory of High-performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, PR China
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, PR China.
| | - Jianlin Shi
- State Key Laboratory of High-performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, PR China.
| |
Collapse
|
2
|
Tao W, Lai Y, Zhou X, Yang G, Wu P, Yuan L. A narrative review: Ultrasound-Assisted drug delivery: Improving treatments via multiple mechanisms. ULTRASONICS 2025; 151:107611. [PMID: 40068411 DOI: 10.1016/j.ultras.2025.107611] [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: 05/07/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 04/02/2025]
Abstract
Safe and efficient drug delivery is as important as drug development. Biological barriers, such as cell membranes, present significant challenges in drug delivery, especially for newly developed protein-, nucleic acid-, and cell-based drugs. Ultrasound-mediated drug delivery systems offer a promising strategy to overcome these challenges. Ultrasound, a mechanical wave with energy, produces thermal effects, cavitation, acoustic radiation, and other biophysical effects. Used alone or in combination with microbubbles or sonosensitizers, it breaks biological barriers, enhances targeted drug delivery, reduces adverse reactions, controls drug release, switches on/off drug functions, and ultimately improves therapeutic efficiency. Various ultrasound-mediated drug delivery methods, including transdermal drug delivery, nebulization, targeted microbubble destruction, and sonodynamic therapy, are being actively explored for the treatment of various diseases. This review article introduces the principles, advantages, and applications of ultrasound-mediated drug delivery methods for improved therapeutic outcomes and discusses future prospects in this field.
Collapse
Affiliation(s)
- Wenxin Tao
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Yubo Lai
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Xueying Zhou
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Shaanxi 710032, China
| | - Pengying Wu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China.
| |
Collapse
|
3
|
Wang S, Tan J, Zhang H, Guan S, Zeng Y, Nie X, Zhu H, Qian S, Liu X. Metastructure and strain-defect engineered Cu-doped TiO x coating to enhance antibacterial sonodynamic therapy. Bioact Mater 2025; 48:458-473. [PMID: 40093306 PMCID: PMC11910374 DOI: 10.1016/j.bioactmat.2025.02.028] [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: 11/28/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Sonodynamic therapy (SDT) has attracted widespread attention in treatment of implant-associated infections, one of the key factors leading to implant failure. Nevertheless, constructing efficient ultrasound-triggered coatings on implant surfaces remains a challenge. Herein, an acoustic metastructure Cu-doped defective titanium oxide coating (Cu-TiO x ) with lattice strain was constructed in situ on titanium implant to realize effective sonocatalysis. The redistribution of Cu atoms broke the pristine lattice of TiO2 during the thermal reduction treatment to regulate its energy structure, which favored separation of electron-hole pairs generated by ultrasound radiation to enhance the sonocatalytic generation of reactive oxygen species. In addition, the acoustic metastructure enhanced the absorption of ultrasound by Cu-TiO x metastructure coating, which further promoted its sonocatalytic effect. Thus, Cu-TiO x metastructure coating could efficiently eliminate Staphylococcus aureus and Escherichia coli infections under ultrasonic irradiation in 10 min. Besides, the osteogenic property of implant was significantly improved after infection clearance in vivo. This work provides a fresh perspective on the design of SDT biosurfaces based on metastructure and strain-defect engineering.
Collapse
Affiliation(s)
- Songsong Wang
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ji Tan
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Haifeng Zhang
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Shiwei Guan
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Yibo Zeng
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoshuang Nie
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongqin Zhu
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Shi Qian
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Xuanyong Liu
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Science, Hangzhou, 310024, China
| |
Collapse
|
4
|
Xu J, Pei Z, Wang Y, Jiang N, Gong Y, Gong F, Ni C, Cheng L. Bioactive microspheres to enhance sonodynamic-embolization-metalloimmune therapy for orthotopic liver cancer. Biomaterials 2025; 317:123063. [PMID: 39753085 DOI: 10.1016/j.biomaterials.2024.123063] [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: 05/17/2024] [Revised: 12/07/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
The development of novel microspheres for the combination of sonodynamic therapy (SDT) with transarterial embolization (TAE) therapy to amplify their efficacy has received increasing attention. Herein, a novel strategy for encapsulating sonosensitizers (e.g., oxygen-deficient manganese tungstate (MnWOX) nanodots) with gelatin microspheres was proposed. The obtained MnWOX-encapsulated microspheres (abbr. Mn-GMSs) facilitated efficient sonodynamic-embolization-metalloimmune therapy via the immune effects of metal ions on orthotopic liver cancer tumor after transarterial embolization (TAE). Due to the strong cavitation effect caused by the porous structure, Mn-GMSs exhibited a greater reactive oxygen species (ROS) generation rate than the free MnWOX nanodots under US irradiation. Efficient SDT revealed robust cell-killing effects and triggered strong immunogenic cell death (ICD). Moreover, the Mn ions released from the bioactive Mn-GMSs further stimulated the dendritic cells (DCs) maturation and triggered the activation of the cGAS/STING pathway to enhance the immunological effect. Thus, Mn-GMSs achieved significant SDT therapeutic outcomes in H22 tumors in mice, and the combination of the Mn-GMSs triggered SDT with programmed cell death ligand 1 (PD-L1) antibodies could further enhance therapeutic outcomes. The Mn-GMSs exhibited high ROS generation efficacy under US irradiation, significant immune activation, good efficacy in combination with immune checkpoint inhibitor, and great potential for artery embolization-assisted drug delivery, thus enabling effective destruction of liver tumors in rats and rabbits. Therefore, this work provides a strategy for applying SDT in deep tumors and highlights a promising sonodynamic-embolization therapy for combating liver cancers.
Collapse
Affiliation(s)
- Jiachen Xu
- Department of Vascular Surgery and Interventional Radiology, The Forth Affiliated Hospital of Soochow University, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215125, China; Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Nan Jiang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yuehan Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
5
|
Chen Z, Sang L, Qixi Z, Li X, Liu Y, Bai Z. Ultrasound-responsive nanoparticles for imaging and therapy of brain tumors. Mater Today Bio 2025; 32:101661. [PMID: 40206140 PMCID: PMC11979416 DOI: 10.1016/j.mtbio.2025.101661] [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: 10/23/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Central nervous system (CNS) cancers, particularly glioblastoma (GBM), are associated with high mortality and disability rates. Despite aggressive surgical resection, radiotherapy, and chemotherapy, patient survival remains poor. The blood-brain barrier (BBB) significantly impedes therapeutic efficacy, making BBB penetration a critical focus of research. Focused ultrasound (FUS) combined with microbubbles (MBs) can transiently open the BBB through mechanisms such as cavitation, modulation of tight junction protein expression, and enhanced vesicular transport in endothelial cells. This review highlights precision delivery and personalized treatment strategies under ultrasound visualization, including precise control of ultrasound parameters and modulation of the immune microenvironment. We discuss the applications of ultrasound-responsive nanoparticles in brain tumor therapy, including enhanced radiotherapy, gene delivery, immunotherapy, and sonodynamic therapy (SDT), with a particular emphasis on piezoelectric catalytic immunotherapy. Finally, we provide insights into the clinical translation potential of ultrasound-responsive nanoparticles for personalized and precision treatment of brain tumors.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Zhai Qixi
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | | | | | | |
Collapse
|
6
|
Song Y, Li H, Yuan Y, Zhang D, Wang Z, Qi B, Jiang P, Yu A. Synergistic photothermal-sonodynamic therapy for antibacterial and immune reprogramming in chronic osteomyelitis. J Control Release 2025; 381:113612. [PMID: 40073945 DOI: 10.1016/j.jconrel.2025.113612] [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: 01/03/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The development of antibiotic resistance and inadequate immune response in chronic inflammation pose significant challenges in treating chronic osteomyelitis. As accepted non-antibiotic antimicrobial therapies, sonodynamic therapy (SDT) and photothermal therapy (PTT) are recognized for their effectiveness in eliminating bacteria and promoting tissue repair, rendering them promising therapeutic strategies for treating bacterial infections and preventing the emergence of drug-resistant bacteria. However, the antimicrobial action and efficacy in promoting tissue repair depend on the activation status of the host immune system. In this study, by encapsulating horseradish peroxidase (HRP)-loaded gold/polydopamine (PDA) nanoparticles within DOTAP/DOPE cationic liposomes (DLPs), a novel multifunctional nanocatalyst, Au/PDA/HRP@DLP (APH@DLP), was developed to achieve antimicrobial effects and immunological reprogramming of chronic osteomyelitis through synergistic SDT and PTT. The impact on immune activation was investigated by assessing the anti-infective and healing effects in osteomyelitis rat models. The release of bacterial-associated antigens during treatment serves as an in situ vaccine, activating antigen-presenting cells and further stimulating adaptive immunity, while also inducing immune memory that significantly reduces the risk of recurrence. Additionally, macrophage phenotypic transformation during SDT and PTT facilitates tissue repair. This study highlights the role of immune activation in SDT/PTT-based antimicrobial therapy and suggests new strategies for treating chronic osteomyelitis.
Collapse
Affiliation(s)
- Yuchen Song
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haimei Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China
| | - Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dong Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Baiwen Qi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Peng Jiang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
7
|
Song K, Ming J, Tao B, Zhao F, Huang S, Wu W, Jiang C, Li X. Emerging glucose oxidase-delivering nanomedicines for enhanced tumor therapy. J Control Release 2025; 381:113580. [PMID: 40024341 DOI: 10.1016/j.jconrel.2025.02.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Abnormalities in glucose metabolism have been shown to characterize malignant tumors. Glucose depletion by glucose oxidase (GOD) has shown great potential in tumor therapy by causing tumor starvation. Since 2017, nanomedicines have been designed and utilized to deliver GOD for more precise and effective glucose modulation, which can overcome intrinsic limitations of different cancer therapeutic modalities by remodeling the tumor microenvironment to enhance antitumor therapy. To date, the topic of GOD-delivering nanomedicines for enhancing tumor therapy has not been comprehensively summarized. Herein, this review aims to provide an overview and discuss in detail recent advances in GOD delivery and directly involved starvation therapy strategies, GOD-sensitized various tumor therapy strategies, and GOD-mediated multimodal antitumor strategies. Finally, the challenges and outlooks for the future progress of the emerging tumor therapeutic nanomedicines are discussed. This review provides intuitive and specific insights to a broad audience in the fields of nanomedicines, biomaterials, and cancer therapy.
Collapse
Affiliation(s)
- Kaiyue Song
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhao
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China.
| | - Xianglong Li
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
8
|
Tian S, Liu Y, Tan Y, Cui X, Liu R, Liu C, Zhao Y, Xu K, Zhou J. Necroptosis-inducing nanobubbles for effective oxygen delivery and enhanced sonodynamic immunotherapy of breast cancer via UTND. Eur J Pharm Biopharm 2025; 210:114675. [PMID: 39993510 DOI: 10.1016/j.ejpb.2025.114675] [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: 09/07/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
PURPOSE Breast cancer (BC) is a global threat to female health. Sonodynamic therapy (SDT) has been shown to induce apoptosis in tumor cells and trigger immunogenic cell death, leading to the activation of antitumor immunity. However, the immunogenicity of this process may be compromised by oxidative stress and proteolysis. Necroptosis caused by ultrasound-targeted nanobubble destruction (UTND) could boost immunity. Therefore, we tested if necroptosis-inducible nanobubbles (NB) could enhance sonodynamic immunotherapy for BC. We also assessed whether O2-filled NB could address tumor hypoxia and enhance SDT efficacy. METHODS A novel multifunctional nano-system, comprising NB for UTND encapsulating chlorin e6 (Ce6) for SDT and O2 for hypoxia alleviation was established. Ce6-O2NB cytocompatibility and intracellular uptake was studied in vitro, as well as whether Ce6-O2NB could generated reactive oxygen species when exposed to ultrasound irradiation in order to induce apoptosis in tumor cells. In vivo pharmacokinetics, therapeutic efficacy, and immune activation after Ce6-O2NB treatment were studied in 4T1 tumor-bearing mice. RESULTS Ce6-O2NB had a well-designed core-shell structure and desirable biocompatibility and safe therapeutic effects. Ce6-O2NB was able to load both ce6 and oxygen to increase ce6 and oxygen accumulation in tumors. After triggering by ultrasound, Ce6-O2NB generated reactive oxygen species (ROS) and acted as sonosensitizers for SDT, promoting tumor cell death through apoptotic and/or necrotic mechanisms. Furthermore, antitumor immunity was activated by stimulation of spleen lymphocyte proliferation and cytotoxicity, and increasing cytotoxic T lymphocyte numbers. Combination of oxygen with SDT ultimately strengthened its antitumor effects. In addition, Ce6-O2NB alleviated tumor hypoxia, induced increased ROS generation, and improved immune responses and therapeutic efficacy of SDT. Ce6-O2NB also facilitated fluorescence and contrast-enhanced ultrasound imaging. CONCLUSIONS Ce6-O2NB can mitigate tumor hypoxia, enhance SDT, and activate antitumor immunity by inducing simultaneous immunogenic apoptosis and necroptosis, ultimately activating antitumor immunity and inhibiting breast tumor growth in mice.
Collapse
Affiliation(s)
- Shun Tian
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Yun Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yandi Tan
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China
| | - Chaoqi Liu
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yun Zhao
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Kui Xu
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Jun Zhou
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China.
| |
Collapse
|
9
|
Wang T, Du M, Chen Z. Sonosensitizers for Sonodynamic Therapy: Current Progress and Future Perspectives. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:727-734. [PMID: 39909788 DOI: 10.1016/j.ultrasmedbio.2025.01.011] [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: 05/22/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
Sonodynamic therapy (SDT) is a novel non-invasive treatment method that combines low-intensity ultrasound and sonosensitizers. Compared with photodynamic therapy, SDT has the advantages of deeper tissue penetration, higher accuracy and fewer adverse reactions. Sonosensitizers are essential for the efficacy of SDT. Sonosensitizers have the advantages of clear structure, easy monitoring, evaluation of drug metabolism and clinical transformation, etc. Notably, biochemical techniques can be used in the field of sonosensitizers and SDT to overcome inherent barriers and achieve sustainable innovation. This article first summarizes the molecular mechanism of SDT, focusing on organic sonosensitizers, inorganic nano-sonosensitizers and multi-functional drug delivery systems with targeting, penetration and imaging functions after a series of modifications. This review provides ideas and references for the design of sonosensitizers and SDT and promotes their future transformation into clinical applications.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China; Department of Medical Imaging, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China.
| |
Collapse
|
10
|
Yu D, Liu M, Ding Q, Wu Y, Wang T, Song L, Li X, Qian K, Cheng Z, Gu M, Li Z. Molecular imaging-guided diagnosis and treatment integration for brain diseases. Biomaterials 2025; 316:123021. [PMID: 39705925 DOI: 10.1016/j.biomaterials.2024.123021] [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: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In practical clinical scenarios, improved diagnostic methods have been developed for the precise visualization of molecular targets using molecular imaging in brain diseases. Recently, the introduction of innovative molecular imaging modalities across both macroscopic and mesoscopic dimensions, with remarkable specificity and spatial resolution, has expanded the scope of applications beyond diagnostic testing, with the potential to guide therapeutic interventions, offering real-time feedback in the context of brain therapy. The molecular imaging-guided integration of diagnosis and treatment holds the potential to revolutionize disease management by enabling the real-time monitoring of treatment responses and therapy adjustments. Given the vibrant and ever-evolving nature of this field, this review provides an integrated picture on molecular image-guided diagnosis and treatment integration for brain diseases involving the basic concepts, significant breakthroughs, and recent trends. In addition, based on the current achievements, some critical challenges are also discussed.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Menghao Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| | - Youxian Wu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianqing Wang
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Litong Song
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyu Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Meijia Gu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
11
|
Zhu Y, Wang D, Du C, Wu T, Wei P, Zheng H, Li G, Zheng S, Su L, Yan L, Hu Y, Wang H, Lin L, Ding C, Chen X. Ruthenium Single-Atom Nanozyme Driven Sonosensitizer with Oxygen Vacancies Enhances Electron-Hole Separation Efficacy and Remodels Tumor Microenvironment for Sonodynamic-Amplified Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416997. [PMID: 40279631 DOI: 10.1002/advs.202416997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Indexed: 04/27/2025]
Abstract
Sonodynamic therapy (SDT) has emerged as a promising noninvasive approach for tumor therapy. However, the effectiveness of traditional inorganic semiconductor sonosensitizers is hindered by rapid electron (e-) and hole (h+) recombination under ultrasonic (US) stimulation, as well as the hypoxic and reductive conditions of tumor microenvironment (TME), which limit the generation of reactive oxygen species (ROS). Herein, a ruthenium (Ru) single-atom nanozyme-driven superimposition-enhanced titanium dioxide-based sonosensitizer (Ru/TiO2-x SAE) is presented that features sufficient oxygen vacancies and high e-/h+ separation efficiency. Through synchrotron radiation-based X-ray absorption spectroscopy and extended X-ray absorption fine structure analysis it is confirmed that oxygen vacancies in TiO2-x nanoparticles promote the immobilization of single-atomic Ru, forming Ru-O₄ active sites. Density functional theory calculations demonstrate that oxygen vacancies alter the electronic structure of nanosensitizer, enhanced e-/h+ separation, increasing oxygen adsorption, and accelerating reaction kinetics under US stimulation, ultimately improving ROS production. Moreover, Ru/TiO2-x SAE boosts sonodynamic efficacy by mitigating the hypoxic and reductive TME. This is attributed to its catalase- and glutathione peroxidase 4-like activities, which facilitate the generation of ROS and trigger lipid peroxidation-mediated ferroptosis. These findings highlight the innovative role of single-atom Ru in optimizing sonosensitizers for SDT-induced ferroptosis, demonstrating its potential for advancing cancer therapy.
Collapse
Affiliation(s)
- Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Dengliang Wang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Chengzhong Du
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Tiantian Wu
- School of Pharmaceutical Sciences/NHC key laboratory of tropical disease control/School of Tropical Medicine, Hainan Medical University, Haikou, 571199, P. R. China
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Hongjia Zheng
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Guanting Li
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - ShunZhe Zheng
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Lichao Su
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Lingjun Yan
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Yongrui Hu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Huimin Wang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Lisen Lin
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Chenyu Ding
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350209, P. R. China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
12
|
Liang M, Kang X, Liu H, Zhang L, Wang T, Ye M, Li W, Qi J. Ultrasound-Energized OX40L-Expressing Biohybrid for Multidimensional Mobilization of Sustained T Cell-Mediated Antitumor Immunity and Potent Sono-Immunotherapy. J Am Chem Soc 2025; 147:13833-13850. [PMID: 40200836 DOI: 10.1021/jacs.5c02025] [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: 04/10/2025]
Abstract
Harnessing immunostimulation to reinvigorate antitumor effector immune cells represents a promising strategy for tumor eradication. However, achieving durable clinical outcomes necessitates multidimensional activation to sustain robust immune responses. Here, we present an ultrasound-empowered living biohybrid that strategically mobilizes T-cell-mediated immunity for potent tumor sono-immunotherapy. Through synthetic biology, we engineer bacteria to express a fusion protein encoding the costimulatory OX40 ligand (OX40L), and further functionalize them with a high-performance polymer sonosensitizer tethered via a reactive oxygen species-cleavable linker. Upon ultrasound irradiation, the sono-activated nanocargoes detach from the bacterial surface, facilitating cellular entry and exposing immune-stimulating OX40L. The potent sonodynamic effects, coupled with the native immunogenicity of bacteria, promotes tumor-associated antigen release, fosters a proinflammatory microenvironment, and drives dendritic cell maturation, thereby priming cytotoxic T-cell activation. The OX40L expressed by the engineered bacteria amplifies and sustains T-cell activity, orchestrating a robust and durable antitumor response. This cascade-amplified immune activation effectively suppresses tumor growth, induces long-lasting immune memory, and provides protection against tumor metastasis and recurrence, significantly enhancing survival outcomes. By integrating ultrasound-energized nanoadjuvants with costimulatory immune boosters, this hybrid living biotherapeutic platform offers a versatile and powerful strategy for multidimensional immune activation, advancing the frontier of cancer sono-immunotherapy.
Collapse
Affiliation(s)
- Mengyun Liang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoying Kang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hanwen Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lu Zhang
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Tianjiao Wang
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Mengjie Ye
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
14
|
Makkar S, Rana N, Priyadarshi N, Bajaj G, Kumar S, Singhal NK. Unravelling the therapeutic properties of aptamer-modified exosome nanocomposite. Adv Colloid Interface Sci 2025; 342:103517. [PMID: 40245577 DOI: 10.1016/j.cis.2025.103517] [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: 12/30/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Exosomes are naturally occurring nanocarriers derived from various cells. In recent years, they have attained significant attention for their potential in precise drug delivery and therapeutic applications. Exosomes exhibit several advantages, remarkably improved stability, bioavailability, and delivery efficiency, which are further augmented by integration with nanomaterials. Functionalizing the aptamer and nanomaterial on the exosomal surface significantly improves the binding affinity and specificity. Here in this review, we examine the synergistic therapeutic effect of exosome-nanomaterial-aptamer conjugate with particular attention to their uses in cancer therapy, bone fracture regeneration, wound healing, etc. Recent advances in the field demonstrated that the amalgamation of different nanomaterials, aptamers, and exosomes has proven to be a transformative approach in the field of therapeutics. Here in the nanocomposite, the aptamer is exclusively used as a recognition molecule to provide specificity to the target cells. Exosomes serve as biocompatible nanocarriers, and different nanomaterials (AuNPs, AuNRs, SiNPs, Graphene, etc.) complement the therapeutic efficiency by PTT/PDT/ROS generation/SO generation, etc. Briefly, the above-mentioned nanocomposite serves as the perfect therapeutic agent by utilizing the exosome's biocompatibility, aptamer's high affinity and nanomaterial's multifunctionality. Furthermore, the challenges and limitations of this nanocomposite have been discussed, along with its prospects in clinical practices.
Collapse
Affiliation(s)
- Simran Makkar
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Niket Rana
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Nitesh Priyadarshi
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Geetika Bajaj
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Sandeep Kumar
- Department of Physics, Punjab Engineering College (Deemed to be University), Chandigarh 160012, India
| | - Nitin Kumar Singhal
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India.
| |
Collapse
|
15
|
Yang M, Lu Z, Liu B, Liu G, Shi M, Wang P. Low-intensity pulsed ultrasound affects proliferation and migration of human hepatocellular carcinoma cells. J Cancer Res Clin Oncol 2025; 151:136. [PMID: 40208346 PMCID: PMC11985662 DOI: 10.1007/s00432-025-06183-0] [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: 11/03/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Low-intensity pulsed ultrasound (LIPUS) is an effective ancillary treatment modality for various malignancies. However, the mechanisms underlying the role of LIPUS in cancer treatment have not been fully elucidated. We investigated the effects and underlying mechanism of LIPUS on the proliferation, apoptosis, migration, and invasion of hepatocellular carcinoma (HCC) cells. METHODS The HCC cell lines SMMC7721 and HCCLM3 were exposed to 1 MHz LIPUS at intensities of 0.5, 1.0, 1.5 W/cm2 for 60 s. Cell morphology, viability, apoptosis, colony formation, migration, and invasion were assessed. Intracellular reactive oxygen species (ROS) levels and mitochondrial membrane potential were evaluated using a ROS assay kit and a JC-1 staining kit. Western blotting was performed to quantify changes in matrix metallopeptidases and epithelial-mesenchymal transition-related proteins. Orthotopic Hep3B-Luc tumor-bearing mice were treated with LIPUS at 1.5 W/cm2 or 0 W/cm2 and growth trend was measured. RESULTS The results showed that different intensities of ultrasound affected cellular activity, inhibited cell proliferation and cloning, facilitated intracellular cytoskeletal protein reorganization, and induced cell apoptosis, particularly at the intensity of 1.5 W/cm2, through the ROS/mitochondria pathway. LIPUS enhanced SMCC7721 and HCCLM3 cell migration and invasion in a dose-dependent manner by regulating matrix metallopeptidases and epithelial-mesenchymal transition-related proteins. In vivo experiments confirmed the inhibitory effect of LIPUS at 1.5 W/cm2 on tumor growth. CONCLUSIONS Although LIPUS induced cell apoptosis and inhibited cell proliferation, it also promoted the invasion and metastasis of HCC cells under certain conditions, which was related to the regulation of matrix metallopeptidases and epithelial-mesenchymal transition-related proteins.
Collapse
Affiliation(s)
- Mingzhen Yang
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Rehabilitation with Integrated Western and Chinese Traditional Medicine, Shanghai, 200032, China
| | - Zhihui Lu
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bangzhong Liu
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guanghua Liu
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mingfang Shi
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Ping Wang
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
16
|
Liao H, Chen M, Liao Z, Luo Y, Chen S, Wang L, Wang Z, Niu C. MnO 2-based nanoparticles remodeling tumor micro-environment to augment sonodynamic immunotherapy against breast cancer. Biomater Sci 2025. [PMID: 40202432 DOI: 10.1039/d5bm00189g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) is characterized by a complex array of factors, including aerobic conditions, high glutathione (GSH) levels, acidic pH, and elevated hydrogen peroxide (H2O2) content, all of which promote cancer progression and contribute to poor prognosis. Fortunately, these challenges can be addressed using MnO2-based nanomaterials. In this study, we have designed and synthesized a Curcumin/MnO2@PLGA@4T1 cell membrane (CMP@4T1m) system aimed at remodelling the TME and enhancing sonodynamic immunotherapy for breast cancer. Through the homologous targeting ability of 4T1m, CMP@4T1m efficiently accumulates at the tumor site. Upon ultrasound irradiation, curcumin (Cur) acts as a sonosensitizer, generating cytotoxic reactive oxygen species (ROS) that induce immunogenic cell death (ICD), activate T-cell responses, and repolarize protumoral M2-like macrophages to antitumoral M1-like macrophages. In the TME, which is mildly acidic and enriched with GSH and H2O2, MnO2 not only oxidizes GSH to glutathione disulfide (GSSG) but also reacts with H2O2 and H+ to produce oxygen, alleviating hypoxia and significantly enhancing the sonodynamic immunotherapy effect. Additionally, Mn2+ generated during this process converts H2O2 into cytotoxic hydroxyl radicals (˙OH). This study thus lays the foundation for advancing cancer nanomedicine, offering a novel approach that integrates TME remodelling with sonodynamic immunotherapy.
Collapse
Affiliation(s)
- Haiqin Liao
- Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Mingyu Chen
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Zhipeng Liao
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Yi Luo
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Sijie Chen
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
| | - Zhigang Wang
- Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Chengcheng Niu
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| |
Collapse
|
17
|
Liu Q, Wang Y, Huang J, Liu X, Mao C, Wang C, Zheng Y, Liu H, Lai KW, Li Z, Zhu S, Jiang H, Cui Z, Wu S. Interfacial Engineering Enhancing Electron Transfer of CaF 2 Nanoparticles for Periodontitis Treatment of Sonotherapy and Bone Resorption. ACS NANO 2025; 19:13341-13355. [PMID: 40145746 DOI: 10.1021/acsnano.5c01112] [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: 03/28/2025]
Abstract
Conventional periodontitis treatments can be superseded by sonodynamic therapy. In this study, a Zn(OH)F/CaF2 heterojunction was synthesized via a hydrothermal method, which exhibited good biocompatibility and antibacterial properties. The band gap decreased significantly after the formation of the heterojunction, thereby enhancing the internal electron transfer. In addition, after the bacteria contact with the material, the electrons of the electron transport chain turn to Zn(OH)F/CaF2 and affect its steady state. Ultimately, H2O2 in an inflammatory environment was used to react with Zn(OH)F/CaF2 to facilitate electron transfer under the influence of ultrasound, ultimately enhancing the catalytic activity. Zn(OH)F/CaF2 exhibits significant antibacterial efficacy against Porphyromonas gingivalis and Staphylococcus aureus. Trace elements Ca and Zn can promote tissue repair and osteogenic differentiation, resulting in less bone destruction and intact gingival tissue after periodontitis treatment. This sonodynamic therapy provides a rapid antibacterial, effective, and deep therapeutic method for treating periodontitis.
Collapse
Affiliation(s)
- Qiuyu Liu
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Yi Wang
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Jin Huang
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Xiangmei Liu
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Congyang Mao
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Chaofeng Wang
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Beijing 100871, China
| | - Hanpeng Liu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Khin Wee Lai
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 57000, Malaysia
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shuilin Wu
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
- School of Materials Science & Engineering, Peking University, Beijing 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| |
Collapse
|
18
|
Yu J, Hu JR, Tian Y, Lei YM, Hu HM, Lei BS, Zhang G, Sun Y, Ye HR. Nanosensitizer-assisted sonodynamic therapy for breast cancer. J Nanobiotechnology 2025; 23:281. [PMID: 40197318 PMCID: PMC11978163 DOI: 10.1186/s12951-025-03311-3] [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: 01/05/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Despite advancements in therapeutic modalities, its prognosis remains poor owing to complex clinical, pathological, and molecular characteristics. Sonodynamic therapy (SDT) is a promising approach for tumor elimination, using sonosensitizers that preferentially accumulate in tumor tissues and are activated by low-intensity ultrasound to produce reactive oxygen species. However, the clinical translation of SDT faces challenges, including the limited efficiency of sonosensitizers and resistance posed by the tumor microenvironment. The emergence of nanomedicine offers innovative strategies to address these obstacles. This review discusses strategies for enhancing the efficacy of SDT using sonosensitizers, including rational structural modifications, improved tumor-targeted enrichment, tumor microenvironment remodeling, and imaging-guided therapy. Additionally, SDT-based multimodal therapies, such as sono-chemotherapy, sono-immunotherapy, and sono-photodynamic therapy, and their potential applications in breast cancer treatment are summarized. The underlying mechanisms of SDT in breast cancer are briefly outlined. Finally, this review highlights current challenges and prospects for the clinical translation of SDT, providing insights into future advancements that may improve therapeutic outcomes for breast cancer.
Collapse
Affiliation(s)
- Jing Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Jun-Rui Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi Tian
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yu-Meng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Hai-Man Hu
- Department of Electrical and Electronic Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Bing-Song Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticides, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Hua-Rong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| |
Collapse
|
19
|
Yan W, Wang Y, Li J, Li L, Liang Q, Huang S, Yang C, Li Z, Yao H. Near-infrared-II-driven Z-scheme heterojunction Polyglycolated MoS 2/CoFe 2O 4 amplified edge potential for dual-mode imaging guided tumor synergistic therapy. J Colloid Interface Sci 2025; 683:793-806. [PMID: 39752929 DOI: 10.1016/j.jcis.2024.12.218] [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: 08/08/2024] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
Targeting the peculiarities of tumor tissue microenvironment different from normal tissue, such as lower pH and overexpression of hydrogen peroxide is the key to effective treatment. In this study, acid-responsive Z-scheme heterojunctions polyglycolated MoS2/CoFe2O4 (MoS2 = molybdenum disulfide, CoFe2O4 = cobalt ferrite) was synthesized using a two-step hydrothermal method, designated as MSCO-PEG, guided by dual modes of photoacoustic imagine (PAI) and nuclear magnetic imaging (MRI). MSCO-PEG (PEG = polyethylene glycol) responded to the acidic environment of tumor tissues and overexpression of hydrogen peroxide to turn on multimodal synergistic treatment of tumor cells under near-infrared-II (NIR-II) illumination. In particular, MSCO-PEG amplified the oxidizing ability of edge valence band holes (h+) and the reducing ability of conduction band electrons (e-) under NIR-II illumination through a "step-like" charge transfer mechanism, promoting the conversion of H2O to oxygen (O2) and the generation of superoxide radicals (O2-). In addition, the outstanding light absorption and photothermal conversion ability of MSCO made it have excellent photodynamic therapy (PDT) and photothermal therapy (PTT) effects. Meanwhile, the abundant multivalent metals endowed MSCO-PEG with the ability to generate chemodynamic therapy (CDT). MSCO-PEG's ability to clear glutathione (GSH) promotes tumor oxidative stress, increases reactive oxygen species (ROS) production, and enhances the synergistic therapeutic effect. This work provides a promising approach to advancing the clinical application of nanomaterials for anticancer therapy targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Weisong Yan
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Yuqing Wang
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Jiang Li
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Lingjun Li
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Qiulong Liang
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Shan Huang
- College of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Changyi Yang
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Zhaojun Li
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China
| | - Huiqin Yao
- School of Basic Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
20
|
Zhu A, Ren H, Li X, Yang W, Han X, Hou X, Zhang S, Li S, Xie Y, Yu M, Chen Y, Xu H. Transdermal STING nano-agonists enhance multifaced functions of antigen-specific T cells triggered by sonodynamic cancer vaccination. NANO TODAY 2025; 61:102590. [DOI: 10.1016/j.nantod.2024.102590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
21
|
Nguyen VN, Nguyen Cao TG, Jeong H, Truong Hoang Q, Pham BTT, Bang J, Koh CW, Kang JH, Lee JH, Wu X, Rhee WJ, Ko YT, Swamy KMK, Park S, Park J, Shim MS, Yoon J. Tumor-Targeted Exosome-Based Heavy Atom-Free Nanosensitizers With Long-Lived Excited States for Safe and Effective Sono-Photodynamic Therapy of Solid Tumors. Adv Healthc Mater 2025:e2500927. [PMID: 40165690 DOI: 10.1002/adhm.202500927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Theranostic nanosensitizers with combined near-infrared (NIR) fluorescence imaging and sono-photodynamic effects have great potential for use in the personalized treatment of deep-seated tumors. However, developing effective nanosensitizers for NIR fluorescence image-guided sono-photodynamic therapy remains a considerable challenge, including the low generation efficacy of reactive oxygen species (ROS), poor photostability, and the absence of cancer specificity. Herein, a novel heavy atom-free nanosensitizer is developed, which exhibits intense NIR fluorescence, high ROS generation efficiency, and improved aqueous stability. By conjugating a bulky and electron-rich group, 4-(1,2,2-triphenylvinyl)-1,1'-biphenyl (TPE), to the IR820 backbone, the resulting IR820 bearing TPE (IR820-TPE) effectively generates ROS via type I and II photochemical mechanisms under 808 nm laser irradiation. Moreover, TPE conjugation considerably increases the sono-photodynamic performance of IR820. To improve the intracellular delivery and tumor-targeting ability of IR820-TPE, biotin-conjugated exosome (B-Exo) is used as a natural nanocarrier. In vitro studies demonstrate the outstanding therapeutic performance of IR820-TPE-loaded B-Exo (IR820-TPE@B-Exo) in synergistic sono-photodynamic cancer therapy. In vivo studies reveal that IR820-TPE@B-Exo shows enhanced tumor accumulation, strong fluorescence signals, and effective sono-photodynamic therapeutic activity with high biosafety. This work demonstrates that IR820-TPE@B-Exo is a promising sono-phototheranostic agent for safe and targeted cancer therapy and NIR fluorescence imaging.
Collapse
Affiliation(s)
- Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Department of Chemistry, School of Chemistry and Life Sciences, Hanoi University of Science and Technology, Ha Noi, 100000, Vietnam
| | - Thuy Giang Nguyen Cao
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hyunsun Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Quan Truong Hoang
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Binh T T Pham
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Jieun Bang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Chang Woo Koh
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hee Kang
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Jeong Hyun Lee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Xiaofeng Wu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon, 22012, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - K M K Swamy
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sungnam Park
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - JaeHong Park
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| |
Collapse
|
22
|
Li W, Yang Z, Yang C, Guo W. Novel hollow ultrasound-triggered ZnFe 2O 4-Bi 2MoO 6 S-scheme heterojunction for efficient ferroptosis-based tumor therapy. J Colloid Interface Sci 2025; 683:132-146. [PMID: 39673926 DOI: 10.1016/j.jcis.2024.12.063] [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: 09/21/2024] [Revised: 11/24/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
This study addresses the challenge of enhancing ferroptosis efficacy for tumor therapy, particularly the limited therapeutic efficiency of current inducers due to tumor microenvironment constraints. Herein, we developed a hollow ultrasound-triggered ZnFe2O4-Bi2MoO6 (ZB) S-scheme heterojunction loaded with artesunate (ART) to overcome these limitations. The ZB heterojunction with a particle size of ∼250 nm efficiently separates electron-hole pairs under ultrasound (US), promoting the generation of reactive oxygen species (ROS). The photodynamic effect of ZB further boosts ROS production, while ART, controlled-released by phase change materials under laser/US stimulation, enhances ROS production via Fe2+-mediated decomposition. This triple-enhanced strategy accumulates lipid peroxidation (LPO), significantly improving ferroptosis effects with a tumor suppression rate of 94.3 %. Moreover, ZB enables multimodal imaging and stimulates antitumor immunity, demonstrating its potential as a diagnostic and therapeutic agent. Our findings demonstrate the potential of this ZB@ART system in advancing ferroptosis-based tumor therapies, inspiring future designs of efficient ferroptosis inducers.
Collapse
Affiliation(s)
- Wenting Li
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Zhuoran Yang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Chunyu Yang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China.
| | - Wei Guo
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China.
| |
Collapse
|
23
|
Ran H, Yang Y, Han W, Liang R, Zhu D, Yuan B, Xu C, Li D, Ren J, Pan H, Liu L, Ma T, Ma A, Cai L. Programmable ultrasound-mediated swarms manipulation of bacteria-red blood cell microrobots for tumor-specific thrombosis and robust photothermal therapy. Trends Biotechnol 2025; 43:868-892. [PMID: 39709244 DOI: 10.1016/j.tibtech.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Despite the excellent advantages of biomicrorobots, such as autonomous navigation and targeting actuation, effective penetration and retention to deep lesion sites for effective therapy remains a longstanding challenge. Here, we present dual-engine cell microrobots, which we refer to as PR-robots, created by conjugating photosynthetic bacteria (PSB) with red blood cells (RBCs). The robots penetrate the tumor interior in swarms through combined hypoxic traction and ultrasound actuation (UA). The hypoxia-targeting ability of PSB induced PR-robot accumulation in the tumor region. Subsequently, programmable UA trapped the PR-robots to form bioswarms and traverse tissue obstacles, penetrating the tumor interior. The substantial influx of PR-robots into the tumor tissue promoted the formation of tumor-specific thrombus (TST). Finally, the PSB and TST synergistically improved the effect of photothermal therapy. Thus, these advantages of remote ultrasound control technology pave the way for various new therapies in practical biomedicine.
Collapse
Affiliation(s)
- Hui Ran
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China
| | - Ye Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Weijing Han
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Denghui Zhu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Dan Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China
| | - Jian Ren
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Teng Ma
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Aiqing Ma
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; Sino-Euro Center of Biomedicine and Health, Shenzhen 518024, PR China.
| |
Collapse
|
24
|
Li M, Liu Q, Xie S, Weng D, He J, Yang X, Liu Y, You J, Liao J, Wang P, Lu X, Zhao J. Transformable Tumor Microenvironment-Responsive Oxygen Vacancy-Rich MnO 2@Hydroxyapatite Nanospheres for Highly Efficient Cancer Sonodynamic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414162. [PMID: 39960349 PMCID: PMC11984894 DOI: 10.1002/advs.202414162] [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: 11/02/2024] [Revised: 12/31/2024] [Indexed: 04/12/2025]
Abstract
Despite the promise of sonodynamic therapy (SDT)-mediated immunotherapy, the anticancer efficacy of current sonosensitizers is greatly limited by the immunosuppressive tumor microenvironment (TME) and their inability to selectively respond to it. Herein, oxygen vacancy-rich MnO2@hydroxyapatite (Ca10(PO4)6(OH)2) core-shell nanospheres (denoted as Ov-MO@CPO) as an advanced TME-responsive sonosensitizer for sonodynamic immunotherapy is demonstrated. The Ov-MO@CPO maintains its structural integrity under neutral conditions but dissolves the pH-sensitive hydroxyapatite shell under acidic TME to release active oxygen vacancy-rich MnO2 core, which reinvigorates H2O2 consumption and hypoxia alleviation due to its catalase-like activity. Furthermore, the introduced oxygen vacancies optimize the electronic structure of Ov-MO@CPO, with active electronic states near the Fermi level and higher d-band center. It results in accelerated electron-hole pair separation and lower catalytic energy barriers to boost ultrasound (US)-initiated ROS production. These multimodal synergistic effects effectively reverse the immunosuppressive tumor microenvironment, inhibiting tumor growth and metastasis in 4T1 tumor-bearing mice. No evident toxic effects are observed in normal mouse tissues. Additionally, when combined with an immune checkpoint inhibitor, Ov-MO@CPO-mediated SDT further improves the effectiveness of immunotherapy. This work affords a new avenue for developing TME-dependent sonosensitizers for SDT-mediated immunotherapy.
Collapse
Affiliation(s)
- Minxing Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Qiyu Liu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Songzuo Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Desheng Weng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinjun He
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Xinyi Yang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Yuanyuan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinqi You
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinghao Liao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Peng Wang
- Department of Emergency MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Xihong Lu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Jingjing Zhao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
25
|
Li M, Zhou Y, Chen X, Chen W, Yang Y, Qian C, Yang L, Zhang Y, Zhang Z, Wu W, Yin Y. Engineered Nanocatalyst-Enabled Cheolesterol Depletion for Enhanced Tumor Piezocatalytic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500967. [PMID: 39965056 PMCID: PMC11984833 DOI: 10.1002/advs.202500967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/06/2025] [Indexed: 02/20/2025]
Abstract
The inadequate generation of reactive oxygen species (ROS) and metastasis of malignant tumors are critical factors that limit the efficacy of conventional sonodynamic therapy in cancer treatment. Herein, an engineered piezocatalyst: cholesterol oxidase (CHO)-loaded Pt-ZnO nanoparticles (Pt-ZnO/CHO) that can explosively generate large amounts of ROS and block the metastasis of tumor, is developed for improving piezocatalytic tumor therapy. In this process, Pt-ZnO can substantially generate ROS via initiating ultrasound (US)-triggered piezocatalytic reactions. In situ-grown Pt nanoparticles not only optimize piezocatalytic activities but also facilitate oxygen (O2) production, thereby synergistically boosting ROS generation. Moreover, O2 produced by Pt-ZnO can accelerate the depletion of excess cholesterol in tumor cells under CHO catalysis to disrupt the integrity of lipid rafts and inhibit the formation of lamellipodia, significantly suppressing the proliferation and metastasis of tumor cells. This strategy by promoting ROS generation and blocking the metastatic pathway of cancer cells offers a new idea for enhanced efficacy-oriented cancer therapeutic strategies.
Collapse
Affiliation(s)
- Mengdan Li
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Yajun Zhou
- Department of UltrasoundThe Fourth Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210029P. R. China
| | - Xiaoyang Chen
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Weiwei Chen
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Yifei Yang
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Cheng Qian
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Lei Yang
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Yaohan Zhang
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| | - Zheng Zhang
- Department of Medical UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiangJiangsu212000P. R. China
| | - Wencheng Wu
- Central Laboratory and Department of Medical UltrasoundSichuan Academy of Medical SciencesSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072P. R. China
| | - Yifei Yin
- Department of Medical UltrasoundAffiliated Hospital of Nantong University, Medical School of NanTong UniversityNantongJiangsu226001P. R. China
| |
Collapse
|
26
|
Cao X, Mao L, Tian Y, Yan L, Geng B, Zhou Y, Zhu J. In situ construction of heterojunctions to regulate the biodegradation behavior of copper carriers for tumor-specific cuproptosis-enhanced sono-immunotherapy. J Nanobiotechnology 2025; 23:246. [PMID: 40128745 PMCID: PMC11934600 DOI: 10.1186/s12951-025-03334-w] [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: 02/09/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Cuproptosis, a novel approach utilizing copper carriers to trigger programmed cell death, exhibits promise for enhancing traditional therapies and activating robust adaptive immune responses. However, the uncontrolled release of Cu ions risks triggering cuproptosis in healthy tissues, potentially causing irreversible damage. To address this, we report on the use of a Cu-MOF (copper metal-organic framework) protective layer to regulate the biodegradation of copper-based nanomaterials. In situ formation of Cu-MOF on Cu2O nanocubes not only stabilizes the material under physiological conditions but also enhances its sonodynamic therapy (SDT) capabilities by establishing a Z-Scheme heterojunction. Upon SDT activation, the targeted Cu ion release at the tumor site triggers a cascade of reactions, generating reactive oxygen species (ROS) via Fenton-like processes and depleting glutathione (GSH). This ROS surge, combined with effective cuproptosis, modulates the immunosuppressive tumor microenvironment, inducing immunogenic cell death to eliminate primary tumors and inhibit metastasis. This study offers a new paradigm for the controlled integration of SDT, chemodynamic therapy (CDT), cuproptosis, and immunotherapy, achieving precise tumor-targeted treatment via controlled copper nanomaterial degradation.
Collapse
Affiliation(s)
- Xiqian Cao
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang Province, 316004, China
| | - Lingwei Mao
- Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Yijun Tian
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Lang Yan
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, 200433, China
| | - Bijiang Geng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Yingtang Zhou
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang Province, 316004, China.
| | - Jiangbo Zhu
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
27
|
Zhou X, Xie J, Zhou X, Ma T, Lu Y, Yang Y, Xie Z, Ling H, Xu R, Wu M, Wang J, Wang W, Kong D, Xu P, Wan X, Wu H, Tong P, Xia H. Single-atom Zr doped heterojunction enhanced piezocatalysis for implant infection therapy through synergistic metal immunotherapy with sonodynamic and physical puncture. J Nanobiotechnology 2025; 23:243. [PMID: 40128749 PMCID: PMC11931772 DOI: 10.1186/s12951-025-03309-x] [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: 09/26/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
Recent common clinical treatments for implant bacterial infections involve replacing inert implants and using antibiotics. However, these methods remain limited in their effectiveness for pathogen clearance, immune regulation, and osteogenesis. In this study, we developed a Zr-doped heterointerface of SrTiO3 and Hap (SrTiZrO3/Hap) heterojunction coating with single-atom Zr doping and heterogeneous interfaces designed for ultrasound-responsive antimicrobial activity and bone formation. Under ultrasound, the mechanical force exerted by SrTiZrO3/Hap enhances its physical puncture and sonodynamic activity, synergizing with the metalloimmunotherapy effect of Zr4+ for efficient antimicrobial activity. The primary mechanism enhancing sonodynamic activity involves local interfacial polarization from single-atom Zr doping, achieving piezoelectric catalysis in conjunction with electronic polarization from the built-in electric field. SrTiZrO3/Hap achieved a 99.3% antibacterial rate against S. aureus and 99.7% against E. coli under ultrasound. Additionally, SrTiZrO3/Hap promoted osteogenic differentiation after ultrasound irradiation by activating the PI3K/Akt pathway via its piezoelectric, needle-like topological surface and the release of functional ions, thus accelerating bone repair.
Collapse
Affiliation(s)
- Xing Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Shangcheng District, Hangzhou, 310006, China
| | - Jingbo Xie
- Department of Orthopaedics, The People's Hospital of Fengcheng City, Fengcheng, Jiangxi, China
- The Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Xingchen Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tianyou Ma
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yichen Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiwen Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhefei Xie
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Houfu Ling
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rui Xu
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Mo Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jinglei Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Weixiang Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Derong Kong
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, P.R. China
| | - Pengchao Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xuan Wan
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- University Medicine Rostock, University of Rostock, Parkstr. 6, 18057, Rostock, Germany
| | - Hongbo Wu
- Department of Rehabilitation Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China.
| | - Peijian Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Hanting Xia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China.
| |
Collapse
|
28
|
Peng H, Wang D, Huang S, Yu A. Dual-targeting Aggregation-induced emission polymer micelles mediate immunogenic sonodynamic therapy for Tumor cell growth inhibition and macrophage reprogramming. Acta Biomater 2025; 195:321-337. [PMID: 39900272 DOI: 10.1016/j.actbio.2025.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Sonodynamic therapy (SDT) is a promising cancer treatment known for its deep tumor penetration and high efficacy. However, developing highly efficient sonosensitizers remains a significant challenge. Reports on SDT using aggregation-induced emission luminogens (AIEgens) are rare, highlighting the urgent need for novel AIE-active sonosensitizers. For the first time, we have developed tumor- and macrophage-targeting nano micelles, AIE/Biotin/Mannose-M (ABM-M), utilizing aggregation-induced emission polymers. The ABM-M mediate immunogenic cell death through SDT. By reprogramming tumor-associated macrophages (TAMs), they promote the conversion of M2 macrophages into M1 macrophages, reversing the tumor's immunosuppressive environment. We optimized the ratio of functional molecules to achieve maximum fluorescence intensity and reactive oxygen species (ROS) generation. The multi-targeting nature of ABM-M enables them to bind to relevant antibodies or other molecules, enhancing the capture and presentation of tumor antigens. This, in turn, activates the immune responses of dendritic cells and T cells while inhibiting angiogenesis, creating a more favorable microenvironment for antitumor therapy. Furthermore, ABM-M can be combined with immune checkpoint inhibitors, such as anti-PD-L1 antibodies, to achieve promising outcomes in cancer immunotherapy. The ABM-M nanomaterials offer multi-layered and multi-targeting immune regulation. This study provides a blueprint for developing next-generation cancer diagnostic and therapeutic strategies. STATEMENT OF SIGNIFICANCE: Our research pioneers the use of nanomicelles to simultaneously target both tumor cells and tumor-associated macrophages (TAMs), integrated with sonodynamic therapy. Through precise ratio adjustments, we engineered nanomicelles capable of multi-target regulation. These micelles uniquely induce immunogenic cell death (ICD) and repolarize macrophages from an immunosuppressive M2 phenotype to an immunostimulatory M1 phenotype, reversing the tumor's immunosuppressive microenvironment. This dual mechanism can be enhanced by combining with immune checkpoint inhibitors, such as anti-PD-L1 antibodies, offering a promising strategy to treat refractory cancers. Extensive in vitro and in vivo validation confirms their therapeutic potential, providing a solid foundation for clinical application. This innovative approach shows significant promise for revolutionizing cancer treatment and improving patient outcomes.
Collapse
Affiliation(s)
- Haiheng Peng
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China
| | - Dandan Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Shiwen Huang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China; Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China.
| |
Collapse
|
29
|
Li X, Liu Y, Wu X, Huang R, Chen S, Yuan K. Ultrasound meets nanomedicine: towards disease treatment and medical imaging. Mikrochim Acta 2025; 192:215. [PMID: 40053162 DOI: 10.1007/s00604-025-07042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 03/18/2025]
Abstract
As a kind of mechanical wave, ultrasound has been widely employed in the biomedical field due to its superiors of deep tissue penetration, non-destructiveness and non-toxicity. In this review, we highlight current progress and prospects in using ultrasound as a powerful tool for disease treatment and medical imaging, including (1) ultrasound as energy input for driving nano/micromotor in drug delivery, this part first introduces the synthesis and motion behavior of nano/micromotors, then reviews the small molecular and macromolecular compounds that the nano/micromotors are delivering; (2) sonosensitive nanomaterials for disease therapy, the sonodynamic, sonopiezoelectric, sonothermal, and sonomechanical therapy will all be covered; (3) ultrasound as a non-invasive technique for nano/micromotor tracking or medical imaging; (4) the sonoporation of nano/microbubble. Future challenges in using ultrasound for disease treatment or medical imaging will also be described in the conclusion part.
Collapse
Affiliation(s)
- Xiaochun Li
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yanting Liu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Xuewan Wu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Rui Huang
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Shaoqi Chen
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Kaisong Yuan
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
30
|
Liu S, Meng Q, Liu Z, Wang J, Li J, Ma X, Hu Y, Wang Z, Ma P, Lin J. Engineered Metal-Organic Framework with Stereotactic Anchoring and Spatial Separation of Porphyrins for Amplified Ultrasound-Mediated Pyroptosis and Cancer Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202421402. [PMID: 39573847 DOI: 10.1002/anie.202421402] [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: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Ultrasound-mediated reactive oxygen species (ROS) generation is pivotal in specifically inducing pyroptosis of tumor cells. However, the effectiveness of pyroptosis is generally hindered by the constraints of ROS generation efficiency. Herein, a new porphyrin-based metal-organic framework (Fe(TCPP)-MOF) was rationally designed via an innovative dual-solvent strategy to amplify ROS generation for ultrasound-controlled pyroptosis. The crystal structure of Fe(TCPP)-MOF was elucidated by continuous rotation electron diffraction technique, revealing its regular and rigid conformation. The porphyrin molecules were precisely oriented and firmly confined within the scaffold, effectively restricting intramolecular motion. The ample distance of 6.8 Å between two porphyrin molecules, combined with the interaction region indicator visualization, confirmed the absence of π-π stacking interactions in the Fe(TCPP)-MOF framework, thereby avoiding the aggregation-caused quenching effect. Furthermore, the permanent porosity and expansive surface area of Fe(TCPP)-MOF enhanced its interaction with oxygen. These ingenious structural features endowed Fe(TCPP)-MOF with a unique ability to generate a large amount of singlet oxygen under ultrasound activation. Meanwhile, the impetus of ultrasound also accelerated the rate of the Fenton reaction catalyzed by iron ions, significantly boosting the generation of hydroxyl radicals. Benefiting from the dual amplification of ROS, Fe(TCPP)-MOF could efficiently induce tumor cells pyroptosis under ultrasound stimulation, thereby intensifying the potency of cancer immunotherapy.
Collapse
Affiliation(s)
- Sainan Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Zhendong Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Jiwei Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Xinyu Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Yarui Hu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Zhanfeng Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, Jilin, China
| |
Collapse
|
31
|
Chen M, Cen J, Shi Q, Shao B, Tan J, Ye X, He Z, Liu Y, Zhang G, Hu J, Bao J, Liu S. Ultrasound-Enhanced Spleen-Targeted mRNA Delivery via Fluorinated PEGylated Lipid Nanoparticles for Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202500878. [PMID: 39878170 DOI: 10.1002/anie.202500878] [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: 01/11/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 01/31/2025]
Abstract
Lipid nanoparticles (LNPs) based messenger RNA (mRNA) therapeutics hold immense promise for treating a wide array of diseases, while their nonhepatic organs targeting and insufficient endosomal escape efficiency remain challenges. For LNPs, polyethylene glycol (PEG) lipids have a crucial role in stabilizing them in aqueous medium, but they severely hinder cellular uptake and reduce transfection efficiency. In this study, we designed ultrasound (US)-assisted fluorinated PEGylated LNPs (F-LNPs) to enhance spleen-targeted mRNA delivery and transfection. Through liquid-to-gas phase transition, we enabled the controlled shedding of fluorinated PEG lipids from F-LNPs, facilitating cellular uptake, membrane fusion, and mRNA release. In vivo results demonstrated that US-assisted F-LNPs increased mRNA transfection approximately 4.0-fold in the spleen following intravenous administration. Notably, the F-LNPs achieved effective mRNA delivery to antigen-presenting cell subsets, such as dendritic cells, macrophages, and B cells. The targeted delivery of full-length ovalbumin-encoding mRNA vaccine induced significant CD8+ T cell response and exhibited excellent therapeutic effect against the ovalbumin-transduced B16F10 tumor model. These findings establish a novel strategy for spleen-specific mRNA delivery through the combination of fluorinated PEG lipids and US treatment, which holds substantial promise for enhancing the efficacy of immunotherapy, potentially broadening the scope of clinical applications for mRNA-based therapy.
Collapse
Affiliation(s)
- Minglong Chen
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jie Cen
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Qiangqiang Shi
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Bing Shao
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jiajia Tan
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xianjun Ye
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Zhihua He
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Yang Liu
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Guoying Zhang
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jianqiang Bao
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of the University of Science and Technology of China, State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China
| |
Collapse
|
32
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 PMCID: PMC11875835 DOI: 10.1016/j.ultsonch.2025.107270] [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: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
33
|
Chen Z, Sang L, Liu Y, Bai Z. Sono-Piezo Dynamic Therapy: Utilizing Piezoelectric Materials as Sonosensitizer for Sonodynamic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417439. [PMID: 39921482 PMCID: PMC11948011 DOI: 10.1002/advs.202417439] [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: 12/28/2024] [Indexed: 02/10/2025]
Abstract
Sonodynamic therapy (SDT) represents a promising approach for cancer treatment. Compared to photodynamic therapy, SDT offers increased penetration depth and higher precision. However, the practical application of SDT is constrained by the low water solubility, poor tumor specificity, and metabolic susceptibility of most sonosensitizers. Recent research has explored the use of piezoelectric materials as sonosensitizers in cancer treatment and inhibition of bacterial growth. Upon ultrasound excitation, the separation of electron-hole (e--h+) pairs occurs within the piezoelectric material. By improving the crystal structure of the material or incorporating other nanoparticles to prevent rapid recombination of e--h+ pairs, the piezoelectric material accumulates charges in the conduction band and valence band, achieving the redox potential of O2/·O2 -. This enables the piezoelectric material to serve as a sonosensitizer, leading to the concept termed Sono-Piezo Dynamic Therapy (SPDT). This review aims to define the concept of SPDT, provide a systematic overview of the historical development of piezoelectric materials in the application of SDT, and elucidate the potential mechanisms by which piezoelectric materials act as sonosensitizers. Importantly, various piezoelectric materials will be discussed in terms of their feasibility, advantages, and disadvantages as sonosensitizers, offering new perspectives for identifying potential sonosensitizers.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of UltrasoundThe First Hospital of China Medical UniversityNo. 155, Nanjing North Street, Heping DistrictShenyangLiaoning110001China
| | - Liang Sang
- Department of UltrasoundThe First Hospital of China Medical UniversityNo. 155, Nanjing North Street, Heping DistrictShenyangLiaoning110001China
| | - Yanjun Liu
- Department of UltrasoundThe First Hospital of China Medical UniversityNo. 155, Nanjing North Street, Heping DistrictShenyangLiaoning110001China
| | - ZhiQun Bai
- Department of UltrasoundThe First Hospital of China Medical UniversityNo. 155, Nanjing North Street, Heping DistrictShenyangLiaoning110001China
| |
Collapse
|
34
|
Guo Q, Tang Y, Wang S, Xia X. Applications and enhancement strategies of ROS-based non-invasive therapies in cancer treatment. Redox Biol 2025; 80:103515. [PMID: 39904189 PMCID: PMC11847112 DOI: 10.1016/j.redox.2025.103515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Reactive oxygen species (ROS) play a crucial role in the pathogenesis of cancer. Non-invasive therapies that promote intracellular ROS generation, including photodynamic therapy (PDT), sonodynamic therapy (SDT), and chemodynamic therapy (CDT), have emerged as novel approaches for cancer treatment. These therapies directly kill tumor cells by generating ROS, and although they show great promise in tumor treatment, many challenges remain to be addressed in practical applications. Firstly, the inherent complexity of the tumor microenvironment (TME), such as hypoxia and elevated glutathione (GSH) levels, hinders ROS generation, thereby significantly diminishing the efficacy of ROS-based therapies. In addition, these therapies are influenced by their intrinsic mechanisms. To overcome these limitations, various nanoparticle (NP) systems have been developed to improve the therapeutic efficacy of non-invasive therapies against tumors. This review first summarizes the mechanisms of ROS generation for each non-invasive therapy and their current limitations, with a particular focus on the enhancement strategies for each therapy based on NP systems. Additionally, various strategies to modulate the TME are highlighted. These strategies aim to amplify ROS generation in non-invasive therapies and enhance their anti-tumor efficiency. Finally, the current challenges and possible solutions for the clinical translation of ROS-based non-invasive therapies are also discussed.
Collapse
Affiliation(s)
- Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yingnan Tang
- School of Pharmacy, Hunan Vocational College of Science And Technology, Changsha, Hunan, 410208, China
| | - Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
35
|
Zhang M, Jia H, Zhuang L, Xu Y, Zhang T, Gu J, He S, Li D. Ultrathin high-entropy hydrotalcites-based injectable hydrogel with programmed bactericidal and anti-inflammatory effects to accelerate drug-resistant bacterial infected wound healing. Colloids Surf B Biointerfaces 2025; 247:114450. [PMID: 39671734 DOI: 10.1016/j.colsurfb.2024.114450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Drug-resistant bacteria infected wounds often bring high risks of delayed healing process and even death. Sonodynamic therapy (SDT) can efficiently kill drug-resistant bacteria. However, superabundant reactive oxygen species (ROS) generated during SDT inevitably trigger significant inflammatory responses, hindering tissue remodeling. Herein, we develop intelligent ultrathin high-entropy hydrotalcites (UHE-HTs)-based injectable thermal-responsive hydrogel loaded with nicotinamide mononucleotide (UHE-HTs/PFN), aiming to achieve programmed antibacterial and anti-inflammatory effects. In the early infection stage, sonosensitive UHE-HTs/PFN hydrogel simultaneously can trigger rapid production of singlet oxygen (1O2) under ultrasound and efficient MDR bacterial sterilization. After halting ultrasonic irradiation, oxidoreductase-mimicking catalysis and nicotinamide mononucleotide release of UHE-HTs/PFN hydrogel effectively reduce ROS levels at wound sites, dampening the NF-κB inflammatory pathway. Such inhibited NF-κB expression can not only reduce the production of pro-inflammatory cytokines and inflammatory responses, but also significantly down-regulate the pyroptosis pathways (NLRP3/ASC/Casp-1) and inhibit pyroptosis that leads to inflammation. Moreover, significantly reduced ROS levels and synergistic release of Mg2+ reverse pro-inflammatory immune microenvironment. Both in vitro and in vivo assays demonstrate that UHE-HTs/PFN hydrogel can transform the adverse infected wound environment into a regenerative one by eradicating drug-resistant bacteria, scavenging ROS, and synergistic anti-inflammation. Therefore, this work develop an intelligent UHE-HTs/PFN hydrogel act as a "lever" that effectively achieve a balance between ROS generation and annihilation, rebuilding harmonious bactericidal and anti-inflammatory effects to remedy drug-resistant bacteria infected wound.
Collapse
Affiliation(s)
- Mingming Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Huaping Jia
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Liang Zhuang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing 100048, China
| | - Yongjie Xu
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Ting Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Jianwen Gu
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China.
| | - Shan He
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing 100048, China.
| | - Dawei Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China.
| |
Collapse
|
36
|
Wu YL, Zhao RR, Wu X, Liu CL, Liu CZ. Carrier-free nanodrug based on small molecule drug and sonosensitizer for enhanced the ferroptosis-driven multimodal synergistic therapy of prostate cancer. Eur J Pharm Biopharm 2025; 208:114656. [PMID: 39909320 DOI: 10.1016/j.ejpb.2025.114656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Ferroptosis plays a significant role in overcoming the therapeutic resistance of cancer cells. Herein, a carrier-free nanoparticle based on small molecule drug sorafenib (SRF) and sonosensitizer rose bengal (RB) was constructed (named SR NPs) for ferroptosis-driven chemotherapy and sonodynamic synergistic therapy (SDT) of prostate cancer (PCa). SR NPs could be enriched in tumor cells and efficiently inhibit tumor cell proliferation. These nanodrugs could significantly reduce glutathione (GSH) synthesis, and inhibit glutathione peroxidase 4 (GPX4) expression by inhibiting the glutamate/cysteine antiporter system (System Xc-) pathway of ferroptosis. Moreover, SR NPs-mediated ferroptosis significantly improved the amount of reactive oxygen species (ROS) generated by SDT, inhibited cell migration and adhesion, enhanced the accumulation of lipid peroxides (LPO) and augmented chemo-sonodynamic therapy. Notably, in vivo studies demonstrated that SR NPs enhanced tumor accumulation, exhibited good biocompatibility, and showed high anti-tumor efficacy in PC-3 tumor-bearing mice. This work offered a new strategy to enhance the treatment efficacy of prostate cancer (PCa) through ferroptosis-chemotherapy-sonodynamic synergistic therapy.
Collapse
Affiliation(s)
- Yao-Lin Wu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Rui-Rui Zhao
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China.
| | - Xiao Wu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chun-Lei Liu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chun-Zhao Liu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
37
|
Liang Z, Sun R, Zhang X, Luan S, Xu H, Wang R, Song L, Shi H, Wang L. Ultrasound-Controllable Release of Carbon Monoxide in Multifunctional Polymer Coating for Synergetic Treatment of Catheter-Related Infections. Adv Healthc Mater 2025; 14:e2403597. [PMID: 39744785 DOI: 10.1002/adhm.202403597] [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: 09/24/2024] [Revised: 12/23/2024] [Indexed: 03/04/2025]
Abstract
Medical catheters are susceptible to biological contamination and pathogen invasion, leading to infection and inflammatory complications. The development of antimicrobial coatings for medical devices has emerged as a promising strategy. However, limited biological functionality and the incompatibility between bactericidal properties and biosafety remain great challenges. Herein, a multifunctional polymer coating (CPB-Ac) is created, incorporating an ultrasonic-responsive carbon monoxide release unit (CORM-Ac) and antifouling unit to treat catheter-related complications. As-synthesized CPB-Ac polymer can be stably anchored to various medical devices with arbitrary shapes and compositions via facile UV treatment. Both in vivo and vitro experiments demonstrated that this multi-functional coating exhibits anti-fouling, anti-inflammatory, and broad-spectrum antibacterial activities as well as good biosafety. During the initial implantation phase, the antifouling units of CPB-Ac coating effectively inhibit the attachment of biological contaminants, significantly reducing the risk of thrombosis and bacterial infection. Once bacterial infection occurs, ultrasonic irradiation can activate CPB-Ac coating to release CO with a much higher amount of 55.3 µm than non-ultrasound controls, therefore rapidly eliminating bacteria and alleviating inflammatory response. It is believed that the work may provide an effective method for the development of next-generation intelligent medical coatings against catheter-related complications.
Collapse
Affiliation(s)
- Ziqing Liang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Rui Sun
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xu Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Shifang Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Hong Xu
- College of Food Science and Light Industry, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, P. R. China
| | - Rui Wang
- College of Food Science and Light Industry, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, P. R. China
| | - Lingjie Song
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, P. R. China
| | - Hengchong Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Lei Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
38
|
Yang Z, Yuan M, Liu B, Ma Z, Ma J, Ma X, Li K, Ma P, Cheng Z, Lin J. Dual-Defect Regulated G-C 3N 4 for Piezoelectric Catalytic Tumor Therapy with Enhanced Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412069. [PMID: 39906915 DOI: 10.1002/adma.202412069] [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: 08/15/2024] [Revised: 12/23/2024] [Indexed: 02/06/2025]
Abstract
Piezoelectric catalysis for tumor treatment is an emerging method for generating reactive oxygen species (ROS). However, the development and optimization of piezoelectric catalytic nanomaterials remain the major challenge. Herein, by regulating the internal and surface defects of graphene phase carbon nitride (defect-engineered g-C3N4), its piezoelectricity and sonocatalytic performance is enhanced, thus achieving efficient tumor treatment. By reducing bulk defects, the charges excited by ultrasound (US) within the defect-engineered g-C3N4 can migrate more rapidly to the material surface, thereby enhancing their participation in redox reactions. Increasing surface defects not only introduce more active sites on the surface of defect-engineered g-C3N4 but also enhance the asymmetry of the defect-engineered g-C3N4 structure, resulting in excellent piezoelectric properties. This defect-engineered g-C3N4 nanosheet can effectively generate ROS in tumor cells and induce tumor cell apoptosis under US stimulation. This work not only introduces a method to enhance the piezoelectric catalytic performance of g-C3N4 but also expands the potential application of defect-engineered piezoelectric materials to tumor treatment.
Collapse
Affiliation(s)
- Zhuang Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Meng Yuan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Bin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Zhizi Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jie Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Xinyu Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Kai Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Ziyong Cheng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
39
|
Xu J, Liu Y. Nanomaterials for liver cancer targeting: research progress and future prospects. Front Immunol 2025; 16:1496498. [PMID: 40092984 PMCID: PMC11906451 DOI: 10.3389/fimmu.2025.1496498] [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: 09/14/2024] [Accepted: 01/07/2025] [Indexed: 03/19/2025] Open
Abstract
The incidence and mortality rates of liver cancer in China remain elevated. Although early-stage liver cancer is amenable to surgical resection, a significant proportion of patients are diagnosed at advanced stages. Currently, in addition to surgical resection for hepatocellular carcinoma, the primary treatment modalities predominantly include chemotherapy. The widespread use of chemotherapy, which non-selectively targets both malignant and healthy cells, often results in substantial immunosuppression. Simultaneously, the accumulation of chemotherapeutic agents can readily induce drug resistance upon reaching the physiological threshold, thereby diminishing the efficacy of these treatments. Besides chemotherapy, there exist targeted therapy, immunotherapy and other therapeutic approaches. Nevertheless, the development of drug resistance remains an inevitable challenge. To address these challenges, we turn to nanomedicine, an emerging and widely utilized discipline that significantly influences medical imaging, antimicrobial strategies, drug delivery systems, and other related areas. Stable and safe nanomaterials serve as effective carriers for delivering anticancer drugs. They enhance the precision of drug targeting, improve bioavailability, and minimize damage to healthy cells. This review focuses on common nanomaterial carriers used in hepatocellular carcinoma (HCC) treatment over the past five years. The following is a summary of the three drugs: Sorafenib, Gefitinib, and lenvatinib. Each drug employs distinct nanomaterial delivery systems, which result in varying levels of bioavailability, drug release rates, and therapeutic efficacy.
Collapse
Affiliation(s)
- Jiahong Xu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yefu Liu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
40
|
Li W, Lin Z, Liu J, Zhang J, Li Y, Liu Y, Yuan X, Li H, Shen H. Pt(IV) prodrug as a potent nanosonosensitizer self-cyclically amplifies sonodynamic-chemotherapy with dually reversing cisplatin resistance. J Mater Chem B 2025; 13:3186-3197. [PMID: 39905853 DOI: 10.1039/d4tb02615b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Although sonodynamic therapy (SDT) has shown promising advancements in combination with chemotherapy, it frequently necessitates the requirement of conventional sonosensitizers and chemotherapeutic agents, engendering intricate systems and potential drug resistance. Herein, we fabricated a potent Pt(IV)-poly(amino acid) coordination nanosonosensitizer (PHPt) with dual reversal of cisplatin resistance, producing abundant 1O2 and ˙OH upon ultrasound irradiation without the use of any external sonosensitizers. The Pt(IV) prodrug in PHPt efficiently reduced to cisplatin through SDT-induced ˙H and glutathione (GSH), inducing ˙OH accumulation and CDDP release, which further amplified the oxidative stress on SDT. Moreover, the high GSH depletion performance of PHPt and administration of aspirin effectively inhibited cisplatin detoxification and activation of the nuclear factor-kappa B pathway, respectively. This cooperative action between the Pt(IV) prodrug and SDT in the tumor microenvironment promoted self-cyclic amplification of sonodynamic-chemotherapy, achieving a significant tumor inhibition rate of 99.4%. Thus, this study offers novel perspectives on the sonosensitizer development and cisplatin application in SDT.
Collapse
Affiliation(s)
- Wenxin Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ziyi Lin
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Jiahui Liu
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Jiarui Zhang
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yuxuan Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yian Liu
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xinru Yuan
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Huimin Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Heyun Shen
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
41
|
Nguyen VN, Nguyen MV, Pham Thi H, Vu AT, Nguyen TX. Recent advances in near-infrared organic photosensitizers for photodynamic cancer therapy. Biomater Sci 2025; 13:1179-1188. [PMID: 39868556 DOI: 10.1039/d4bm01457j] [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: 01/28/2025]
Abstract
With the advancement of photodynamic therapy, various photosensitizers have been developed to enhance the efficacy of cancer treatment while minimizing side effects. Recently, near-infrared organic fluorophores have gained significant attention as promising photodynamic agents for cancer therapy due to their tunable photophysical properties, structural versatility, good biocompatibility, high biosafety, and synthetic flexibility. In particular, near-infrared organic photosensitizers offer several notable advantages, including deep tissue penetration, a low fluorescence background for bioimaging, and reduced damage to biological tissues compared to traditional visible-spectrum photosensitizers. In this minireview, we will discuss the current developments in near-infrared organic photosensitizers for photodynamic cancer therapy. Furthermore, we will briefly highlight the challenges and prospects in this field. This minireview aims to encourage more researchers to develop advanced near-infrared organic photosensitizers and facilitate their transition from laboratory research to preclinical studies and ultimately to clinical use.
Collapse
Affiliation(s)
- Van-Nghia Nguyen
- School of Chemistry and Life Sciences, Hanoi University of Science and Technology, 1 Dai Co Viet Road, Ha Noi, Vietnam.
| | - Minh Viet Nguyen
- VNU-Key Laboratory of Advanced Materials for Green Growth, Faculty of Chemistry, University of Science, Vietnam National University, Hanoi, Vietnam.
| | - Huong Pham Thi
- Laboratory of Environmental Science and Climate Change, Institute for Computation Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, Vietnam.
- Faculty of Environment, School of Technology, Van Lang University, Ho Chi Minh City, Vietnam
| | - Anh-Tuan Vu
- School of Chemistry and Life Sciences, Hanoi University of Science and Technology, 1 Dai Co Viet Road, Ha Noi, Vietnam.
| | - Truong Xuan Nguyen
- School of Chemistry and Life Sciences, Hanoi University of Science and Technology, 1 Dai Co Viet Road, Ha Noi, Vietnam.
| |
Collapse
|
42
|
Yang M, Wang X, Peng M, Wang F, Hou S, Xing R, Chen A. Nanomaterials Enhanced Sonodynamic Therapy for Multiple Tumor Treatment. NANO-MICRO LETTERS 2025; 17:157. [PMID: 39992547 PMCID: PMC11850698 DOI: 10.1007/s40820-025-01666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025]
Abstract
Sonodynamic therapy (SDT) as an emerging modality for malignant tumors mainly involves in sonosensitizers and low-intensity ultrasound (US), which can safely penetrate the tissue without significant attenuation. SDT not only has the advantages including high precision, non-invasiveness, and minimal side effects, but also overcomes the limitation of low penetration of light to deep tumors. The cytotoxic reactive oxygen species can be produced by the utilization of sonosensitizers combined with US and kill tumor cells. However, the underlying mechanism of SDT has not been elucidated, and its unsatisfactory efficiency retards its further clinical application. Herein, we shed light on the main mechanisms of SDT and the types of sonosensitizers, including organic sonosensitizers and inorganic sonosensitizers. Due to the development of nanotechnology, many novel nanoplatforms are utilized in this arisen field to solve the barriers of sonosensitizers and enable continuous innovation. This review also highlights the potential advantages of nanosonosensitizers and focus on the enhanced efficiency of SDT based on nanosonosensitizers with monotherapy or synergistic therapy for deep tumors that are difficult to reach by traditional treatment, especially orthotopic cancers.
Collapse
Affiliation(s)
- Mengyao Yang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Xin Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Mengke Peng
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Fei Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Senlin Hou
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China.
| | - Ruirui Xing
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
| | - Aibing Chen
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
43
|
Malla P, Wang YM, Su CH. New horizons for the therapeutic application of nanozymes in cancer treatment. J Nanobiotechnology 2025; 23:130. [PMID: 39979897 PMCID: PMC11844087 DOI: 10.1186/s12951-025-03185-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/01/2025] [Indexed: 02/22/2025] Open
Abstract
The advent of nanozymes has revolutionized approaches to cancer diagnosis and therapy, introducing innovative strategies that address the limitations of conventional treatments. Nanozyme nanostructures with enzyme-mimicking catalytic abilities exhibit exceptional stability, biocompatibility, and customizable functions, positioning them as promising tools for cancer theranostics. By emulating natural enzyme reactions, nanozymes can selectively target and eradicate cancer cells, minimizing harm to adjacent healthy tissues. Nanozymes can also be functionalized with specific targeting ligands, allowing for the precise delivery and regulated release of therapeutic agents, improving treatment effectiveness and reducing adverse effects. However, issues such as biocompatibility, selectivity, and regulatory compliance remain critical challenges for the clinical application of nanozymes. This review provides an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments. The strategic oncological deployment of nanozymes could profoundly impact future advancements in personalized medicine, highlighting recent progress and prospective directions in enzyme-mimetic approaches for cancer treatment. This review summarizes an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments.
Collapse
Affiliation(s)
- Pravanjan Malla
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Ming Wang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
| | - Chia-Hao Su
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
44
|
Zhang Z, Zeng W, Guo N, Ran M, Gan H, Wu Q, Xu J, Wang H, Han S, Liu Y. A nanodrug loading indocyanine green and metformin dually alleviating tumor hypoxia for enhanced chemodynamic/sonodynamic therapy. J Colloid Interface Sci 2025; 680:341-355. [PMID: 39571354 DOI: 10.1016/j.jcis.2024.11.133] [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: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 12/16/2024]
Abstract
As an emerging therapeutic method, the application of sonodynamic therapy (SDT) is hindered by its intrinsic unsatisfactory efficiency, the tumor hypoxia and low tumor specificity. Here, we reported the design of a tumor-targeting multifunctional nanodrug for O2-generation/O2-economization dually enhanced SDT/chemodynamic therapy (CDT) combination therapy. After the co-encapsulation of sonosensitizer indocyanine green (ICG) and oxidative phosphorylation inhibitor metformin (Met) into hollow MnO2 (H-MnO2) nanoparticles, ICG/Met@H-MnO2@MPN-FA (IMMMF) was conveniently prepared through the formation of metal-phenolic networks (MPNs) between Fe3+ and folic acid (FA) immobilized tannic acid (TA, TA-FA) onto its surface. In vitro experiments indicated its selective uptake by 4T1 cells via the specific folate receptors-FA interactions. Responding to glutathione (GSH) and the acidic environment, the decomposition of IMMMF led to the release of Mn2+ and Fe2+ for enhanced CDT, and ICG for SDT. Furthermore, Met was continuously released to reduce O2 consumption for enhanced SDT. More importantly, IMMMF catalyzed the endogenous H2O2 into O2 for further enhanced SDT. Expectedly, both in vitro and in vivo antitumor assays confirmed its satisfactory therapeutic efficiency via CDT/SDT synergistic therapy. Hence, this intelligent sonocatalytic nanoagent emerges as a promising candidate for CDT-enhanced SDT, which also provides a novel strategy for dually alleviating tumor hypoxia with better therapy.
Collapse
Affiliation(s)
- Ziying Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China
| | - Weishen Zeng
- Dongguan Children's Hospital, Dongguan 523000, China
| | - Ning Guo
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China
| | - Mengnan Ran
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Department of Nuclear Medicine, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, Guangdong, China
| | - Huixuan Gan
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China
| | - Quanxin Wu
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China
| | - Jiehua Xu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Department of Nuclear Medicine, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, Guangdong, China
| | - Hao Wang
- Dongguan Children's Hospital, Dongguan 523000, China.
| | - Shisong Han
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Department of Nuclear Medicine, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, Guangdong, China.
| | - Yun Liu
- School of Pharmacy, Guangdong Medical University, Dongguan 523000, China.
| |
Collapse
|
45
|
Yang C, Wang W, Gao Y, Yin L, Pan K, Chen D, Yang F, Xing N. Sonodynamic Therapy by Reactive Oxygen Species Generation-Responsive Pseudo-Semiconducting Polymer Nanoparticles Combined with a Fibroblast Growth Factor Receptor Inhibitor for Enhancing Immunotherapy in Bladder Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:9125-9139. [PMID: 39883874 DOI: 10.1021/acsami.4c20545] [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: 02/01/2025]
Abstract
Sonodynamic therapy, a treatment modality recently widely used, is capable of disrupting the tumor microenvironment by inducing immunogenic cell death (ICD) and enhancing antitumor immunity during immunotherapy. Erdafitinib, an inhibitor of the fibroblast growth factor receptor, has demonstrated potential benefits for treating bladder cancer. However, Erdafitinib shows effectiveness in only a small number of patients, and the majority of patients responding positively to the medication have "immune-cold" tumors. To increase the therapeutic efficacy of Erdafitinib, we have herein developed a biodegradable pseudoconjugate polymer (PSP) with sonodynamic capabilities. Erdafitinib could be efficiently encapsulated in nanoparticles (NP-PE) prepared through the self-assembly of PSP with an oxidation-sensitive polymer (P1). Under ultrasound conditions, NP-PE effectively induced cytotoxicity by producing reactive oxygen species and further triggering ICD. Compared with Erdafitinib, NP-PE inhibited the expression of FGFR3 to a higher extent. In animal models with bladder cancer, NP-PE inhibited tumor growth, stimulated antitumor immunity, and synergized with antiprogrammed cell death-ligand 1 (aPD-L1), offering a novel approach for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Chao Yang
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wenkuan Wang
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yunhao Gao
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lu Yin
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Kehao Pan
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong Chen
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Feiya Yang
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nianzeng Xing
- Department of Urology/State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
46
|
Sun Z, Li X. A promising mesoporous silica carrier material for the diagnosis and treatment of liver diseases: recent research advances. J Mater Chem B 2025; 13:1935-1960. [PMID: 39801308 DOI: 10.1039/d4tb01822b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The therapeutic diagnosis of liver diseases has garnered significant interest within the medical community. In recent years, mesoporous silica nanoparticles (MSNs) have emerged as crucial nanocarriers for the treatment of liver ailments. Their remarkable diagnostic capabilities enable them to be used in techniques such as high-throughput mass spectrometry (MS), magnetic resonance imaging (MRI), near-infrared (NIR) fluorescence imaging, photoacoustic imaging (PAI), and ultrasonography (US), attracting considerable attention. Furthermore, the introduction of amino and carboxyl group modifications in MSNs has facilitated their use as drug delivery carriers for treating liver diseases, including hepatocellular carcinoma. This paper reviews the preparation methods, in vitro diagnostic capabilities, and in vivo therapeutic delivery systems of MSNs for liver disease treatment. It also summarizes relevant toxicity studies, aiming to provide a comprehensive overview of the diagnostic and therapeutic applications of MSNs in the treatment of liver diseases, particularly hepatocellular carcinoma. Through this review, we seek to offer theoretical insights into the potential of MSNs for diagnostic and therapeutic applications in liver disease treatment.
Collapse
Affiliation(s)
- Zihao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
47
|
Guo J, Qiu Y, Hu C, Cao Y, Li D, Du Y. Enhancing Antituberculosis Treatment Nanoparticles Encapsulated with Catalase and Levofloxacin Under Ultrasound Stimulation: A 3D Spheroid Study. Mol Pharm 2025; 22:747-759. [PMID: 39772621 DOI: 10.1021/acs.molpharmaceut.4c00748] [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: 01/11/2025]
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (MTB). Tuberculous granuloma is the central and key pathological structure of tuberculosis and is characterized by tissue hypoxia and ineffective drug delivery. To address these issues, this study fabricated a composite nanoparticle loaded with catalase (CAT) and levofloxacin (LEV) (CAT@LEV-NPs) and then combined it with ultrasound (US) to investigate the bactericidal effect and underlying mechanisms using TB spheroids. The TB spheroids were constructed using attenuated Bacillus Calmette-Guérin (BCG) instead of MTB to facilitate operation under general experimental conditions. This study examined the physical properties and oxygen production efficiency of CAT@LEV-NPs. Subsequently, we treated TB spheroids with nanoparticles alone or in combination with US and found that ultrasound significantly increased drug permeability and activated CAT@LEV-NPs to produce a large number of reactive oxygen species (ROS). The combined treatment showed excellent antibacterial effects, resulting in more severe damage to the bacterial structure than other treatments. Additionally, the combined treatment induced a higher M1 polarization of macrophages, increased the apoptosis rate, and improved the anoxic microenvironment in TB spheroids. These factors may be closely related to the enhanced bactericidal effects of combined treatment. In conclusion, our study suggests that US combined with CAT@LEV-NPs could serve as a novel, noninvasive, safe, and effective treatment modality for intractable MTB infections.
Collapse
Affiliation(s)
- Jiajun Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yan Qiu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Can Hu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yuchao Cao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
48
|
Zhang Y, Li Z, Zhang C, Shao C, Duan Y, Zheng G, Cai Y, Ge M, Xu J. Recent advances of photodiagnosis and treatment for head and neck squamous cell carcinoma. Neoplasia 2025; 60:101118. [PMID: 39721461 PMCID: PMC11732236 DOI: 10.1016/j.neo.2024.101118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) are the most common type of head and neck tumor that severely threatens human health due to its highly aggressive nature and susceptibility to distant metastasis. The diagnosis of HNSCC currently relies on biopsy and histopathological examination of suspicious lesions. However, the early mucosal changes are subtle and difficult to detect by conventional oral examination. As for treatment, surgery is still the primary treatment modality. Due to the complex anatomy and the lack of intraoperative modalities to accurately determine the incision margins, surgeons are in a dilemma between extensive tumor removal and improving the quality of patient survival. As more knowledge is gained about HNSCC, the increasing recognition of the value of optical imaging has been emphasized. Optical technology offers distinctive possibilities for early preoperative diagnosis, intraoperative real-time visualization of tumor margins, sentinel lymph node biopsies, phototherapy. Fluorescence imaging, narrow-band imaging, Raman spectroscopy, optical coherence tomography, hyperspectral imaging, and photoacoustic imaging have been reported for imaging HNSCC. This article provides a comprehensive overview of the fundamental principles and clinical applications of optical imaging in the diagnosis and treatment of HNSCC, focusing on identifying its strengths and limitations to facilitate advancements in this field.
Collapse
Affiliation(s)
- Yining Zhang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhenfang Li
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Chengchi Zhang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Zhejiang University of Technology, Hangzhou 310023, China
| | - Chengying Shao
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yanting Duan
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Guowan Zheng
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Yu Cai
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China.
| | - Jiajie Xu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China.
| |
Collapse
|
49
|
Wu F, Kuang X, Deng S, Qi S, Xiong J, Zhao B, Li C, Tan S, Kang Q, Xiao H, Tan X, Wu GL, Yang Q, Chen G. Conversion therapy strategy: A novel GPC3-targeted multimodal organic phototheranostics platform for mid-late-stage hepatocellular carcinoma. Mater Today Bio 2025; 30:101442. [PMID: 39866786 PMCID: PMC11762635 DOI: 10.1016/j.mtbio.2024.101442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is typically diagnosed at intermediate to advanced stage, making surgical treatment unfeasible. Conversion therapy aims to reduce tumor stage, improve hepatic resection feasibility, and lower recurrence rates. Since traditional therapies are often accompanied by uncertainty of efficacy, there is an urgent need to explore new treatment strategies. Near-infrared phototheranostics technology provides a new way for HCC diagnosis and treatment by its excellent optical properties. However, complex preparation and poor biocompatibility of phototheranostics probes limit clinical application. In this study, we developed a fluorescence/magnetic resonance dual-modality imaging (FLI/MRI) as well as photothermal/photodynamic therapy (PTT/PDT) GPC3-targeted multifunctional phototheranostics probe, IR820-GPC3-Gd NPs (IGD NPs), to improve the efficiency of conversion therapy for HCC. The IGD NPs were simply prepared with the IR820 as the core. Conjugating the HCC-specific targeting molecule GPC3 peptide and the MRI agent DOTA-Gd through click chemistry. IGD NPs target HCC cells through GPC3, releasing heat and reactive oxygen species (ROS) under noninvasive 808 nm laser irradiation to reduce tumor size and achieve downstaging. High-sensitivity FLI/MRI precisely delineates tumor boundaries, providing real-time surgical navigation and prognosis assessment. This novel probe offers a feasible and effective treatment option for advanced HCC.
Collapse
Affiliation(s)
- Fan Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xin Kuang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Sanlin Deng
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuo Qi
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jian Xiong
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bibo Zhao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Chuanfu Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Senyou Tan
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qiang Kang
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hao Xiao
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaofeng Tan
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Gui-long Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qinglai Yang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Center for Molecular Imaging Probe Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Engineering Research Center for Early Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of General Surgery, Turpan City People's Hospital, Tulufan, 838000, China
| |
Collapse
|
50
|
Wang X, Guo X, Ren H, Song X, Chen L, Yu L, Ren J, Chen Y. An "Outer Piezoelectric and Inner Epigenetic" Logic-Gated PANoptosis for Osteosarcoma Sono-Immunotherapy and Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415814. [PMID: 39726343 DOI: 10.1002/adma.202415814] [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: 10/16/2024] [Indexed: 12/28/2024]
Abstract
The precise manipulation of PANoptosis, a newly defined cell death pathway encompassing pyroptosis, apoptosis, and necroptosis, is highly desired to achieve safer cancer immunotherapy with tumor-specific inflammatory responses and minimal side effects. Nonetheless, this objective remains a formidable challenge. Herein, an "AND" logic-gated strategy for accurately localized PANoptosis activation, utilizing composite 3D-printed bioactive glasses scaffolds integrated with epigenetic regulator-loaded porous piezoelectric SrTiO3 nanoparticles is proposed. The "logic-gated" strategy is co-programmed by an "outer" input signal of exogenous ultrasound irradiation to produce reactive oxygen species and an "inner" input signal of acid tumor microenvironment to ensure the epigenetic demethylation regulation, guaranteeing the tumor-specific PANoptosis. Specifically, immunogenic PANoptosis triggers dendritic cell maturation and cytotoxic T cell activation, amplifying antitumor immune responses and significantly suppressing osteosarcoma growth, with a suppression rate of ≈73.47 ± 5.2%. In addition, the well-known bioactivities of Sr-doped scaffolds expedite osteogenic differentiation and reinforce bone regeneration. Therefore, this work provides a paradigm of logic-gated sono-piezoelectric biomaterial platform with concurrently exogenous/endogenous activated PANoptosis for controlled sono-immunotherapy of osteosarcoma, and related bone defects repair.
Collapse
Affiliation(s)
- Xiaoting Wang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Xun Guo
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Luodan Yu
- Department of Radiology, Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jianli Ren
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, 400010, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|